Human/animal diseases

De AnimalZooFrance
Aller à la navigation Aller à la recherche

Ce texte relativement daté a largement été diffusé sur les premiers forums et newsgroups consacrés à la zoophilie. Il a accompagné la constitution de notre communauté.

Texte original

Human/animal diseases

(essay by Pony)

A little while ago I asked Pony to put together a few words for me addressing health concerns. I got this in the mail a couple of weeks later. Almost everything you could ever need on the subject. Hats off to Pony for excellent work. --- midnitecrow


ZOONOTIC DISEASES

If you are, or think you are sick, DO NOT rely on this information to self diagnose, or treat yourself!!! As far as I know, there is NOTHING which you may get after sharing an intimate moment with an animal, that you could not get innocently elsewhere, or from a human, so DO NOT be embarrassed, seek help, and go see your doctor.. Oh, and get your animal for periodic checkups…(although if you have recently been intimate with your animal, it may be wise to not schedule any general health check too soon after) J

Each year, we choose to share our lives, (and occasionally our beds) with animals. But how safe a relationship is that? Okay, so we know that our dog is able to bite us, and can imagine what damage may occur to us if that were to happen, but can those animals we love actually pose a significant hazard to our health? Could the family rabbit possibly put you in hospital? Or if we became intimate with a horse, is that likely to present a risk of, at the worst, more than a kick? The simple answer to this is…yes; it is possible for anyone to contract a disease from virtually any animal. In addition to this, it does not have to come via intimate or sexual contact in many cases. There have been cases reported (and quite frequently) of people who have suffered badly from pathogens (germs, spores etc.) which they have been exposed to just by kissing, or petting an animal. The risks to your health increase dramatically as the level of intimacy increases. This is not meant to be a scare story! Whilst this text does not claim to be complete in any way, what I wanted as far as possible to do, is to break down each and every zoonotic (contagious between animals and humans) disease that I knew something about. So that there is and can be, far less concern about such diseases or the possibility of contracting them.

Firstly, we live in a disease prevalent world, we are surrounded daily by the agents responsible for the most serious and deadly diseases that have ever ravaged the earth. Influenza, salmonella, legionnaire's disease, the common cold, there would not be enough space to even think about the number of contaminants that we encounter during the average day. So why are we not already sick or dead? The body has it's own immune system that usually keeps us safe from most things. This coupled with a clean and healthy lifestyle should keep us safe from harm. Occasionally, the body may encounter something which is stronger than our own defenses, or we ourselves are already unfit, or run down. This is when we get sick, and sometimes fatally so. But, as mentioned before, if we keep fit and healthy, there is little risk of harm. Okay, so this long list of animal to human diseases will look scary, but in reality, the risks to your health are no greater than crossing a road. I have extensively searched both the Internet, and my own personal textbooks on this subject, and am greatly indebted to all those who have researched this subject, and publicized their findings. Without such research, the human race would doubtless be in a much poorer state than it already is. My thanks also goes to the many educational and university sites, which have greatly assisted me with this piece. As far as I can ascertain, this (as a compilation of scientific data and general information) is not copywritten. I have done my best to acknowledge all contributors and authors. If you feel that you need greater credit for the information contained within, then please contact me. I have broken the information down, so that it is simple to understand, and clear about the risks to you and your health. I apologize for the many long technical terms used within this page, but these are the REAL terms, and any attempt by me to alter or to simplify these, will result in some or all of the actual definition being lost. There are, however, a few things that you should generally bear in mind, when considering ANY relationship with animals, and these should keep you much safer…and hopefully a little more comfortable with the situation.

Animals that are sick, or have been badly kept, are run down, previously straying, or even living wild have a MUCH greater chance of infecting you with a disease. Also, if you are sick, or recovering from any illness, the chance of contracting a disease is greater. Be especially wary of an animal that shows any sign of illness Eg. Parasites, sores, abbrasions etc.

As long as you and the animal are in reasonable health, there are FAR FEWER diseases that you can catch from an animal, than you can contract from a fellow human. So, if you tell Fido to leave the bed…you also have to tell your (should you have one) human partner too!!

Anyone with any other input for this, please contact me at [email protected]

…and best wishes to you….and those you love.

BRUCELLOSIS

(In humans: Mediterranean fever, undulant fever, Malta fever. In animals: contagious abortion, epizootic abortion, and Bang's disease)

  • AGENT:

Brucella abortus: cattle, sheep Brucella canis: dogs Brucella melitensis: sheep, goats Brucella suis: swine

  • RESERVOIR AND INCIDENCE

Of the above species, Brucella canis is most likely zoonotic agent of the above list, which the zoophile may encounter, due to the popularity of dogs in zoophilic relationships. Prevalence: 1 to 10% in dogs, throughout the U.S. B. canis is well adapted to dogs, and is not the subject of a large scale eradication program in the general dog population, as Brucella has been in other animals. Human brucellosis due to B. canis is uncommon but can be acquired from dogs; most cases resulted from contact with aborting bitches. In 1988, the CDC noted 96 cases of brucellosis reported in the U.S.: 22 from Texas and 20 from Calif.

  • TRANSMISSION:

Ingestion of unpasteurized milk Poorly defined transmission cycle in zoonotic diseases: contact with infected animals especially aborted fetuses, fluids or membranes, or urine. Possibly airborne.

  • DISEASE IN ANIMALS:

Abortions are followed by immunity, though carrier state persists especially with secretions from the udder. Infertility, testicular abnormalities, poor semen quality in dogs, inapparent infection may be common, as indicated by seropositivity.

  • DISEASE IN MAN:

Lymphadenopathy, splenomegaly, fever, headache, chills, orchitis, weakness, nausea, weight loss. The chronic form may assume an undulant nature, with periods of normal temperature between acute attacks; symptoms may persist for years, either continuously or intermittently. Antibiotics can effect a cure within one year in about 80% of cases. Case fatality if untreated is less than 2%.

  • DIAGNOSIS:

Rapid slide agglutination test is available. Blood culture and additional serologic tests used to confirm slide test results.

  • TREATMENT:

Single-drug regimens are not recommended because the relapse rate may be as high as 50%. Combination regimens of two or three drugs are more effective. (1) Doxycycline plus rifampin or streptomycin (or both) (2) trimethoprim-sulfamethoxazole plus rifampin or streptomycin (or both) are effective in doses for 21 days. Longer courses of therapy may be required to cure relapses, osteomyelitis, or meningitis.

  • PREVENTION\CONTROL:

Be EXTREMELY careful if you have a bitch which aborts a pregnancy, this is an indication that something is not right. Protect yourself and the animal. Use disposable gloves, chlorine based bleach, or iodine to disinfect any contaminated surfaces. Then contact your vet to arrange for a brucellosis test, and general health checkup for your bitch.

SALMONELLOSIS

(Salmonella food poisoning, enteric paratyphosis) A common bacterial cause of food poisoning worldwide. Over 1800 food-poisoning serotypes of salmonella (bacterium) exist. The prevalence of individual serotypes constantly changes. In the U.S., 5 million cases are diagnosed annually. S. typhi, the cause of Typhoid Fever, rarely occurs in the U.S. and is not discussed here.

  • RESERVOIR AND MODE OF TRANSMISSION:

Salmonellas are common commensals of all animals and birds and are excreted in feces. Host-adapted strains may cause serious illness (e.g., S. dublin in cattle, S. pullorum in chickens), but most human food-poisoning salmonellas do not cause clinical signs in animals. The main reservoirs for human infection are poultry, cattle, sheep and pigs. Infection in animals is maintained by recycling slaughterhouse waste as animal feed, fecal oral spread and fecal contamination of hatching eggs. Transmission occurs when organisms, introduced into the kitchen in poultry carcasses, meat or unpasteurized milk, multiply in food owing to inadequate cooking, cross-contamination of cooked foods and inadequate storage. Person-to-person spread is common in institutions such as hospitals. The organism inhabits the intestinal tract of many animals including birds, cattle, sheep, pigs, and rats, mice, hamsters, guinea pigs, nonhuman primates and humans. Salmonella occurs worldwide. *The house mouse may also be a reservoir of the infection and may play a role in human and animal salmonellosis. Humans, rarely, and animals may be carriers and asymptomatic shedders of the organism. *Salmonella prevalence in the U.S. canine population may be 10% or more. *Prevalence data from 8 studies conducted worldwide indicated that a wide range (0.6-27%) of cats were culture-positive for Salmonella. *Salmonella carriers in newly imported Rhesus and Cynomolgus monkeys exceeded 20% in some shipments. *Birds, reptiles, and turtles are especially dangerous sources of Salmonellosis. 94% of all reptiles harbor Salmonella. Turtles alone in 1970 may have caused 280,000 human cases of Salmonellosis. *In 1975 the FDA ruled it illegal to sell a. Viable Turtle eggs b. Live turtles with a carapace length < 10.2cm (4 inches) c. Exceptions - Educational & scientific institutions and marine turtles. d. Marine turtles have not been shown to be a reservoir of Salmonella

  • TRANSMISSION:

Indirect transmission via contaminated food and water are the most common sources but transmission may also be by direct contact. It is a common contaminant of sewage. Found in many environmental water sources. Environmental contamination continues to be a potential source of infection for lab animals and secondarily for personnel handling those animals. *Animal feed containing animal by products continues to be a source of Salmonella contamination, especially if the diets consist of raw meal and have not undergone the pelleting process.

  • INCUBATION PERIOD:

Humans. 12-72 hours. Animals. 1-5 days.

  • CLINICAL FEATURES:

Humans. The presence and severity of symptoms depends on the infecting dose. Typically there is watery diarrhea for about ten days, possibly leading to dehydration, with abdominal pain and low-grade fever. Septicemia and abscess formations are rare. Animals. Subclinical infection is common and many animals may be intermittent or persistent carriers. However, cows may suffer with fever, diarrhea and abortion. Calves undergo epizootic outbreaks of diarrhea with high mortality. In pigs, fever and diarrhea are less common than in cattle. Infected sheep, goats and poultry usually show no signs of infection.

  • PATHOLOGY:

Humans. Enteritis is a feature. Extraintestinal infection may cause abscesses. Animals. Penetration of the infection into the mucosa is followed by inflammation, especially ileitis, progressing to inflamed mesenteric lymph nodes in the mesentery, possibly progressing to septicemia and pneumonia especially in calves. Dehydration and rapid loss of weight are due to stimulation of chloride excretion and inhibition of sodium absorption. Abortion in cattle is caused by massive proliferation of salmonella in the placenta leading to placental necrosis.

  • DIAGNOSIS:

Humans. Isolate salmonella from feces and suspected foods using selective media followed by serotyping and, if appropriate, phage typing. Animals. Culture feces, postmortem tissues and foods of animal origin. Serological tests are of limited value as many noninfected animals have titers from past infections. Humans. Usually only a self-limiting illness occurs. Deaths from dehydration or septicemia are rare and occur usually in infants, or debilitated or elderly patients. Animals. There is abortion in cattle and endometritis with temporary infertility. In calves, dehydration and septicemia may lead to death.

  • PREVENTION:

Humans. Educate food handlers in good kitchen hygiene. Ensure thorough cooking of meat, refrigerate cooked foods and prevent cross-contamination. Pasteurize all milk. Ensure personal hygiene. Reduce contamination of poultry carcasses at abattoirs. Irradiation of meat and other foods before purchase will reduce contamination. Animals. This is difficult and often impractical because there are many sources of infection. Principles of control include the following: maintain closed herds and flocks; keep animals in small groups; purchase replacements direct from the farm of origin; avoid mixing animals from different sources; sterilize ingredients of animal feed; provide mains drinking water for grazing livestock; prevent access of wild birds and rodents to animal houses; completely de-stock animals and thoroughly cleanse and disinfect housing between batches; monitor poultry breeding stock and remove excreters; disinfect hatching eggs and fumigate incubators.

  • TREATMENT:

Humans. Treatment of uncomplicated enterocolitis is symptomatic only. Young, malnourished, or immuno-compromised infants, severely ill patients, those with sickle cell disease, and those with suspected bacteremia should be treated for 3-5 days with trimethoprim-sulfamethoxazole (one double-strength tablet twice a day), ampicillin (100 mg/kg IV or orally), or ciprofloxacin 9750 mg twice daily). Animals. Treatment with antibiotics and sulfonamides immediately diarrhea and fever occur reduces mortality but is contraindicated in healthy carriers in which treatment may prolong the carrier state.

  • VACCINATION:

Humans. None. Animals. Vaccines are available against S. dublin and S. typhimurium in calves. A live vaccine prepared from a rough strain of S. dublin gives good protection in calves against both S. dublin and S. typhimurium.

  • LEGISLATION:

Humans. The disease is notifiable specifically in the USA, Australia, New Zealand and several European countries, or as food poisoning as in the UK. Animals. Notification of infection in food animals is obligatory in some countries, including the UK, with statutory sampling of animal protein for animal feed. Heat treatment of waste food applies in the UK. A slaughter policy is claimed in Luxembourg, Germany and Czechoslovakia.

SHIGELLOSIS

SYNONYM: Bacillary dysentery.

  • ETIOLOGY:

The type species is Shigella dysenteriae, other agents are S. flexneri, S. boydii, and S. sonnei. The first three species are subdivided into serotypes.

  • GEOGRAPHIC DISTRIBUTION:

Worldwide.

  • THE DISEASE IN MAN:

It is seen most often in preschool-age children. A new serotype introduced into tropical areas where the population is undernourished provokes disease in all age groups, particularly children, the elderly, and debilitated individuals. Generally, the incubation period is less than 4 days. The disease begins with fever and abdominal pains, followed by diarrhea and dehydration for 1 to 3 days. A second phase of the symptomatology can last for several weeks. The main symptom is tenesmus; in serious cases, stools contain blood, mucus, and pus. The symptomatology is usually variable. In many countries, strains of Shigella resistant to sulfonamides and to several antibiotics have been observed.

  • THE DISEASE IN ANIMALS:

A clinical picture similar to that in man occurs in monkeys.

  • SOURCE OF INFECTION AND MODE OF TRANSMISSION:

The principal reservoir of the infection for man is other humans that are sick or carriers. The sources of the infection are feces and contaminated objects. The most common mode of transmission is the fecal-oral route. Outbreaks comprising numerous cases have had their origin in a common source of infection, such as foods contaminated by hands or feces of carrier individuals. Insects, particularly flies, can also play a role as mechanical vectors. Bacillary dysentery is a serious disease with high mortality in nonhuman primates in captivity, but there is doubt that monkeys can harbor the etiologic agent in their natural habitat. Monkeys probably contract the infection by contact with infected humans. The infection spreads rapidly in nonhuman primate colonies because the monkeys defecate on the cage floor and also often throw their food there.

  • ROLE OF ANIMALS IN THE EPIDEMIOLOGY OF THE DISEASE:

Of little significance. Cases of human bacillary dysentery contracted from nonhuman primates are known. The victims are mainly children. In highly endemic areas, dogs may shed Shigella temporarily. The etiologic agent has also been isolated from horses, bats, and rattlesnakes. Nevertheless, animals other than nonhuman primates play an insignificant role.

  • DIAGNOSIS:

Definitive diagnosis depends on isolation of the etiologic agent by culture of fecal material on selective media. Serologic identification and typing are important from the epidemiologic viewpoint.

  • TREATMENT:

In humans, treatment of dehydration and hypotension is lifesaving in severe cases. The current antimicrobial treatment of choice is trimethoprim-sulfamethoxazole (one double-strength tablet twice a day), or ciprofloxacin (750 mg twice a day; contraindicated in children and pregnant women). Parental hydration and correction of acidosis and electrolyte disturbances are of primary importance. Antispasmodics (e.g., tincture of belladonna) are helpful when cramps are severe. Drugs that inhibit intestinal peristalsis (paregoric, diphenoxylate with atropine) may ameliorate symptoms but prolong fever, diarrhea, and excretion of Shigella in feces. Appropriate precautions should be taken both in the hospital and in the home to limit spread of infection.

  • CONTROL:

In man, control methods include a) environmental hygiene, especially disposal of human waste and provision for potable water; b) personal hygiene; c) education of the public and of food handlers about the sources of infection and methods of transmission; d) sanitary supervision of the production, preparation, and preservation of foods: e) control of flies; f) reporting and isolation of cases and sanitary disposal of feces; and g) search for contacts and the source of infection. A live, streptomycin-dependent vaccine, administered orally in three or four doses has given good protection against the clinical disease for 6 to 12 months. Its use is indicated in institutions where shigellosis is endemic. Indiscriminate use of antibiotics must be avoided in order to prevent the emergence of multi-resistant strains and to ensure that these medications remain available for use in severe cases. In animals, control consists of a) isolation and treatment of sick or carrier monkeys: b) careful cleaning and sterilization of cages; c) prevention of crowding in cages: and d) prompt disposal of wastes and control of insects.

YERSINIA

Animals are susceptible to three Yersinia species that are potentially zoonotic: AGENT: Gram negative, non spore forming rods

1. YERSINIA PSEUDOTUBERCULOSIS AND ENTEROCOLITICA (Pseudotuberculosis)
  • RESERVOIR AND INCIDENCE

Ubiquitous in nature, isolated from dust, soil, water, and milk. Natural infections occur in man, birds, rodents, rabbits , guinea pigs, mice, cats, nonhuman primates, sheep, swine, goats.

  • TRANSMISSION:

direct contact, or fecal contaminated food or water cause most transmission from animals to man. Human cases of have been reported in association with disease in household pets, particularly sick puppies and kittens. The most important source of Y. enterocolitica infection may be pork, as the pharynx of pigs may be heavily colonized.

  • DISEASE IN ANIMALS:

guinea pigs, rabbits, and hamsters exhibit poor condition and enlarged lymph nodes. Subacute clinical signs are common, with diarrhea and weight loss, possibly death within 2 weeks to 3 months. Chinchillas are very susceptible to infection with Y. enterocolitica. In sheep, abortions, epididymitis and orchitis occur with high mortality. In cattle, abortion and pneumonia occur. Nonhuman primates exhibit an ulcerative colitis. Histopathological lesions include acute inflammation of the terminal ileum with mesenteric lymphadenitis occurs. Sometimes abscesses develop in the liver, spleen, and lungs. Usually self-limiting, but there is a fatality rate of 5-7%.

  • DISEASE IN MAN:

acute watery diarrhea, mesenteric lymphadenitis which can be confused with appendicitis, fever, headache, pharyngitis, anorexia, vomiting erythema nodosum (in about 10% of adults), post-infectious arthritis, iritis, cutaneous ulceration, hepatosplenic abscesses, osteomyelitis and septicemia.

  • DIAGNOSIS:

fecal culture using cold enrichment technique. Serologic diagnosis can be made by an agglutination test or by ELISA.

  • TREATMENT:

usually resistant to penicillin and its derivatives. Agents of choice are the aminoglycosides and cotrimoxazole. Both are usually sensitive to the tetracyclines.

  • PREVENTION/CONTROL:

Control rodents and prevent contamination of food and water by rodents and birds. Pasteurize milk. Cook pork thoroughly. Personal hygiene is important.

2. YERSINIA PESTIS

(Plague, Pest, black death, pestilential fever) The second pandemic of plague, known then as the "Black Death," originated in Mesopotamia about the middle of the 11th century, attained its height in the 14th century and did not disappear until the close of the 17th century. It is thought that the Crusaders, returning from the Holy Land in the 12th and 13th centuries, were instrumental in hastening the spread of the disease. Again the land along trade routes was primarily involved and from them the infections spread east, west, and north. During the course of the disease, 25,000,000 people perished, a fourth of the population of the world.

  • AGENT:

a gram negative coccobacillus

  • RESERVOIR AND INCIDENCE

Endemic in wild rodents in Southwestern U.S., as well as in Africa and Asia. Most important reservoirs worldwide are the domestic rat, Rattus rattus, and the urban rat, Rattus norvegicus. Human infections have increased since 1965 and usually result from contact with infected fleas or rodents. The disease is also associated with cats, goats, camels, rabbits, dogs and coyotes. Dogs and cats may serve as passive transporters of infected rodent fleas into the home.

  • TRANSMISSION:

Contact with infected rodent fleas or rodents. Fleas may remain infected for months. Note: a protein secreted by the Yersinia is a coagulase that causes blood ingested by the flea to clot in the proventriculus. The bacillus proliferates in the proventriculus, and thousands of organisms are regurgitated by obstructed fleas and inoculated intradermally into the skin. This coagulase is inactive at high temperatures and is thought to explain the cessation of plague transmission during very hot weather. Pulmonary form spread by airborne or droplet infection. Human infections from non-rodent species usually result from direct contact with infected tissues, by scratch or bite injuries, and handling of infected animals. Several recent reports have detailed human plague associated with exposure to domestic cats. Exposure can be from inhalation of respiratory secretions of cats with pneumonic plague or by contaminating mucous membranes or skin wounds with secretins or exudates.

  • DISEASE IN ANIMALS:

dogs usually have a brief self-limiting illness cats usually exhibit severe and often fatal infection, with fever, lymphadenopathy, hemorrhagic pneumonia, and encephalitis. rodents may carry the disease asymptomatically or develop fatal disease. infected rats and squirrels frequently die unless they are from an enzootic area and have acquired immunity.

  • DISEASE IN MAN:

Incubation period 2 to 6 days. In humans the disease is called Bubonic, Septicemic, or Pneumonic plague depending on the pattern of distribution of the infection. Bubonic is the most common form causing fever and swollen, tender lymph nodes (called Buboes). Pneumonic plague is systemic plague with lung involvement. Mortality may exceed 50%. Plague is also called the "black death" because disseminated intravascular coagulation takes place and areas of skin undergo necrosis.

  • DIAGNOSIS:

Impression smears of aspirates or blood stained with gram or Giemsa. Organisms have a typical "safety pin" appearance culture of the organisms can be performed, by reference lab FA of smear is confirmatory. Serology via Complement fixation, passive hemagglutination, and immunofluorescence (IFA)

  • TREATMENT:

streptomycin with tetracycline or chloramphenicol.

  • PREVENTION\CONTROL:

Wild rodents should be controlled and fleas should be eliminated. It is important to control rodents and fleas for outdoor housed animals. Sentinel animal programs used in endemic areas. Endemic areas of the U.S. include California, Nevada, Arizona, and New Mexico. Masks gowns, and gloves should be worn when handling cats suspected to be infected and all contaminated surfaces disinfected. Notify Health Department of suspected cases Vaccines available for high-risk personnel.

TUBERCULOSIS

(Consumption)

  • AGENT, RESERVOIR AND INCIDENCE

TB is caused by the gram positive, acid fast, aerobic, bacillus of the Mycobacterium genera. The most common species of mycobacteria are: 1. M. bovis (cattle, dogs, swine) 2. M. avium (birds, swine, sheep) 3. M. tuberculosis (man, nonhuman primates, cattle, dogs, swine, psitticines). 4. M. marinum, fortuitum, platypolcitis (fish) Note: Atypical mycobacterium, M. scrofulaceum, M. kansasii, and M. intracellulare have been reported in NHP's and are also present in soil and water. They can cause pulmonary disease refractory to treatment in man, and are most often seen in immunocompromised people. Specific reagents can be used to skin test for these bacteria. TB continues to be a major cause of morbidity and mortality throughout the world. One billion people are infected with the tubercle bacillus, and there are 8 million new cases and 3 million deaths annually. Cases of active tuberculosis are increasing in the United States after years of decline. The end of 1990 reported a provisional total of 23,720 cases. Case rates increased 4.4 percent between 1988 and 1989, and perhaps another 1 percent in 1990. Overall rate was 9.5 per 100,000 in 1989, with the Middle Atlantic and Pacific regions reporting the highest rates, 36 percent and 34 percent, respectively, above the U.S. total. The lowest rates were in the West, North, Central, and Mountain regions. Rates continue to be higher among urbanites, minorities, the poor, and the homeless, substance abusers and persons infected with HIV. All three types are capable of causing disease in man although M. tuberculosis (variety hominis) is by far the most common. Nonhuman primates can carry all three types but most infections are caused by M. tuberculosis variety hominis. While most nonhuman primates are capable of contracting TB, Old World species appear to be more susceptible to the disease than New World species and great apes. Most cases of TB in monkeys are thought to arise from human contact. Animals may be imported from areas of the world where the incidence of the disease is high and where contact between humans and simians is frequent. In close confinement the disease can spread rapidly.

  • TRANSMISSION:

Mycobacterium bacilli are transmitted from infected animals or infected tissue primarily via the aerosol route. May also be contracted via ingestion or cutaneous inoculation of the bacilli. Personnel caring for infected animals as well as those performing necropsies on infected animals are at risk for contracting the disease. Exposure to dusty bedding of infected animals, coughing of infected animals, and aerosolization of the organism during sanitation procedures may also be sources of the disease in the lab environment. Once within the body the organism may spread throughout the lungs, lymphatics, blood vascular system, and many visceral organs.

  • DISEASE IN NONHUMAN PRIMATES:

The signs of TB may be insidious with only slight behavioral changes noticed, followed by anorexia and lethargy. Often animals die suddenly while appearing to be in good condition. Other signs that might be seen include diarrhea, suppuration of lymph nodes, ulceration of the skin, and palpable splenomegaly and hepatomegaly. The organ of predilection is the lung but lesions may also be seen in the pleura, intestines, lymph nodes, liver, kidney, spleen, and peritoneum. Under the surface of these tissues are yellowish-white to gray nodules filled with caseous material which may rupture and produce cavitation. Although skeletal involvement in primates is rare, tuberculosis of the spine may cause paralysis of the hindlimbs (Pott's disease).

  • DISEASE IN FISH:

In infected fish, granulomatous lesions are usually observed.

  • DISEASE IN MAN:

In humans the clinical signs depend on the organ system involved. The most familiar signs related to pulmonary TB are cough, sputum production, and hemoptysis. The patient may be asymptomatic for years. General signs may include anorexia, weight loss, lassitude, fatigue, fever, chills and cachexia. Skin lesions are characterized by ulcers or by papular lesions progressing to dark suppurative lesions. TB may affect virtually every other organ system with signs or symptoms relating to the individual system. Miliary TB is most often seen in the very young and old people.

  • DIAGNOSIS:

The diagnosis of TB is often difficult. Four tests are commonly used for presumptive diagnosis: 1. Intradermal TB test - Mammalian tuberculin 2. Radiography 3. Acid fast stained sputum smear 4. ELISA Confirmation by culture, histopath, or animal inoculation.

  • TREATMENT:

Regimens currently accepted in the USA include isoniazid combined with rifampin, with or without pyrazinamide.

  • PREVENTION\CONTROL:

Multifaceted and includes: personnel education wearing of protective clothing when handling nonhuman primates a regular health surveillance program for humans and nonhuman primates isolation and quarantine of suspect animals rapid euthanasia and careful disposal of infected animals Vaccine - A vaccine, BCG, is available (Bacille Calmette-Guerin, strain of M. Bovis) a. Used in humans quite often in G. Britain b. Used in high risk groups c. Effective, but it causes the patient to have a positive TB test. Personnel working with NHP's who convert to a positive skin test should be referred for appropriate medical treatment and follow up and should not work with animals until shown to be noninfectious.

LEPROSY

SYNONYM: Hansen's disease.

  • ETIOLOGY:

Mycobacterium leprae, a polymorphic acid-alcohol-fast bacillus. M. leprae is hard to distinguish from other unculturable mycobacteria naturally infecting animals. The failure of attempts to culture M. leprae in vitro constitutes a great barrier to better determining its biochemical characteristics for identification purposes as well as for therapeutic and immunologic studies. In part, this difficulty has been overcome, first, by in vivo culture on mouse foot pads and, lately, by the discovery that the leprosy organism can infect the nine-banded armadillo (Dasypus novemcinctus). At present, the latter serves as a model for lepromatous leprosy and provides a large number of bacilli for research.

  • OCCURRENCE IN MAN:

An estimated 12 million people are affected by leprosy. The highest prevalence is in tropical and subtropical regions of Asia, Africa, Latin America, and Oceania. Leprosy is very prevalent in India, Southeast Asia, the Philippines, Korea, southern China, Papua New Guinea, and some pacific islands. Ninety percent of the cases reported in the Americas come from five countries: Argentina, Brazil, Colombia, Mexico, and Venezuela. Chile is the only South American country free of the infection. In the United States 2,500 cases are known, most of them in immigrants. Autochthonous cases arise in Hawaii, Puerto Rico, Texas, and Louisiana. The infection's prevalence is related to the socioeconomic level of the population. The fact that the disease has practically disappeared in Europe is attributed to the improved standard of living there. The proportion of total leprosy cases represented by lepromatous leprosy (see The Disease in Man) varies with the region. In Asia and the Americas this form makes up between 25 and 65% of all cases, while in Africa it accounts for only 6 to 20%.

  • THE DISEASE IN MAN:

The incubation period is usually 3 to 5 years, but it can vary from 6 months to 10 years or more. Clinical forms of leprosy cover a wide spectrum, ranging from mild self-healing lesions to a progressive and destructive chronic disease. The polar form at one end of the spectrum is tuberculoid leprosy, and at the other, lepromatous leprosy. Intermediate forms are also found. Tuberculoid leprosy is characterized by localized lesions of the skin and nerves, often asymptomatic. Basically, the lesions consist of a granulomatous, paucibacillary, inflammatory process. The bacilli are difficult to detect, and can be observed most frequently in the nerve endings of the skin. This form results from active destruction of the bacilli by the cellular immunity of the patient. On the other hand, serum antibody titers are generally low. Nerve destruction causes lowered conduction; heat sensibility is the most affected, tactile sensibility less so. Trophic and autonomic changes are common, especially ulcers on the sole and mutilation of body members. Lepromatous leprosy is characterized by numerous symmetrical skin lesions consisting of macules and diffuse infiltrations, plaques, and nodules of varying sizes (lepromas). There is involvement of the mucosa of the upper respiratory tract, of lymph nodes, liver, spleen, and testicles. Infiltrates are basically histiocytes with a few lymphocytes. Cellular immunity is absent (negative reaction to lepromin) and antibody titers are high. In this form of the disease, as in the borderline, erythema nodosum leprosum (ENL) often appears. The indeterminate form of leprosy has still not been adequately characterized from the clinical point of view; it is considered to be the initial stage of the disease. The first cutaneous lesions are flat, hypopigmented, and have ill-defined borders. If this form is not treated, it may develop into tuberculoid, borderline, or lepromatous leprosy. Bacilli are few, and it is difficult to confirm their presence. Finally, the borderline form occupies a position intermediate between the two polar forms (tuberculoid and lepromatous), and shares properties of both; it is unstable and may progress in either direction. Destruction of nerve trunks may be extensive. Bacilli are observed in scrapings taken from skin lesions. An estimated one-third of clinical cases become incapacitated. half of them completely. Nevertheless, these proportions are now changing, due to both prevention/control programs and early implementation of effective treatments. There is evidence that inapparent infection may occur with a certain frequency among persons, especially family members, in contact with patients.

  • THE DISEASE IN ANIMALS:

The disease in armadillos (Dasypus novemcinctus) is similar to the lepromatous form in man. Infection in these animals is characterized by macrophage infiltrates containing a large number of bacilli. Skin lesions vary from mild to severe. The small dermal nerves are invaded by the etiologic agent. Many bacilli are seen in the macrophages of the lymph tissue, in the pulp of the spleen, and in Kupffer's cells in the liver. M. leprae is known to prefer the cooler parts of the human or mouse body. For this reason, armadillos were used as experimental animals even before natural inaction was confirmed in them, since their body temperature is from 30 to 35oC. Experimental inoculation of armadillos with human leproma material reproduces the disease, characterized by broad dissemination of the agent, and involvement of lymph glands, liver, spleen, lungs, bone marrow, meninges, and other issues, in a more intense form than is usually observed in man. The disease in the chimpanzee appeared as a progressive chronic dermatitis with nodular thickening of the skin of the ears, eyebrow nose, and lips. Lesions of the nose, skin, and dermal nerves contained copious quantities of acid-fast bacteria. The case was histologically classified as borderline 12 months after the clinical symptoms were first observed, and as lepromatous after a later biopsy. In the case of the Cercocebus monkey, the initial lesion consisted of nodules on the face. Four months later, a massive infiltration and ulceration were seen on the face and nodules appeared on the ear and the forearms. Sixteen months after cutaneous lesions were first observed. The animal began to suffer deformities and paralysis of the extremities. Histopathologic findings indicated the subpolar or intermediate lepromatous form. The disease was progressive, with neuropathic deformation of feet and hands. It seemed to regress when specific treatment was administered. The animal apparently contracted the disease from a patient with active leprosy. Experimental infections carried out to date have indicated that these animals may experience a spectrum of different forms similar to those in man.

  • SOURCE OF INFECTION AND MODE OF TRANSMISSION:

Man is the principal reservoir of M. leprae. The method of transmission is still not well known due to the extended incubation period. Nevertheless, the principal source of infection is believed to be lepromatous patients, in whom the infection is multibacillary, skin lesions are often ulcerous, and a great number of bacilli are shed through the nose similarly. Bacilli are found in the mouth and pharynx. Consequently, transmission might be effected by contact with infected skin, especially through wounds or abrasions, and by aerosols, as is the case in tuberculosis. Lately, more importance has been attributed to aerosol transmission. Oral transmission and transmission by hematophagous arthropods are not discounted, but they are assigned less epidemiologic importance. Until recently, leprosy was believed to be an exclusively human disease. But research in recent years has demonstrated that the infection and the disease also occur naturally in wild animals. Even though some researchers have expressed doubt that the animal infection is identical to the human, at present an accumulation of evidence indicates that the etiologic agent is the same. The origin of infection in animals is unknown. It is believed that armadillos contracted the infection from a human source, perhaps from multibacillary patients before the era of sulfones. In this regard, it should be pointed out that leprosy bacilli may remain viable for a week in dried nasal secretions and that armadillos are in close contact with the soil. The high disease prevalence in some localities would indicate armadillos can transmit the infection to one another, either by inhalation or direct contact. Another possible transmission vehicle is maternal milk, in which the agent has been detected. It is difficult to demonstrate that armadillos are a source infection for man because of the long incubation period in humans and the impossibility of excluding a human source in an endemic area. In Texas, a case of human leprosy was attributed to a patient's practice of capturing armadillos and eating their meat. Subsequently, another five cases with hand lesions were detected in natives of the same state who habitually hunted and cleaned armadillos but had no known contact with human cases. The prevalence of leprosy in armadillos in Louisiana and Texas suggests that these animals could serve as a reservoir of M. leprae; however, nothing is known about the frequency of infection in nonhuman primates and the role they may play in transmission of the disease. The sources of the cases of leprosy in these animals were probably people with lepromatous leprosy.

  • DIAGNOSIS:

Laboratory confirmation of leprosy requires the demonstration of acid-fast bacilli in scrapings from slit skin smears or the nasal septum. Biopsy of skin or of a thickened involved nerve also gives a typical histologic picture. M. leprae does not grow in artificial media.

  • CONTROL:

Control is based on early detection and chemotherapy. In the face of multiple confirmed cases of resistance to dapsone, combination of this medication with rifampicin is presently recommended for paucibacillary leprosy, and the same two medications in combination with clofazimine for multibacillary leprosy. Rifampicin has a rapid bactericidal effect and eliminates contagion in patients in 1 to 2 weeks. The isolation of patients in leprosariums is no longer necessary, since the chemotherapy effectively eliminates infectiousness and thereby interrupts transmission of the disease.


VIBRIOSIS

Vibrios other than Vibrio cholerae that cause human disease are Vibrio parahaemolyticus, Vibrio vulnificus and Vibrio alginolyticus. All are halophilic marine organisms. Infection is acquired by exposure to organisms in contaminated, undercooked, or raw crustaceans or shellfish and warm ( 20o C) ocean waters and estuaries. Infections are more common during the summer months from regions along the Atlantic coast and the Gulf of Mexico in the United States and from tropical waters around the world. Oysters are implicated in up to 90% of food-related cases. V. parahaemolyticus causes an acute watery diarrhea with crampy abdominal pain and fever, typically occurring within 24 hours after ingestion of contaminated shellfish. The disease is self-limited, and antimicrobial therapy is usually not necessary. V. parahaemolyticus may also cause cellulitis and sepsis, though these findings are more characteristic of V. vulnificus infection. V. vulnificus and V. alginolyticus-neither of which is associated with diarrheal illness-are important causes of cellulitis and primary bacteremia, which may follow ingestion of contaminated shellfish or exposure to sea water. Cellulitis with or without sepsis may be accompanied by bulla formation and necrosis with extensive soft tissue destruction, at times requiring debridement and amputation. The infection can be rapidly progressive and is particularly severe in immunocompromised individuals-especially those with cirrhosis-with death rates as high as 50%. Patients with chronic liver disease and those who are immunocompromised should be cautioned to avoid eating raw oysters. Tetracycline at a dose of 500 mg four times a day is the drug of choice for treatment of suspected or documented primary bacteremia or cellulitis caused by Vibrio species. V. vulnificus is susceptible in vitro to penicillin, ampicillin, cephalosporins, chloramphenicol, aminoglycosides, and fluoroquinolones, and these agents may also be effective. V. parahaemolyticus and V. alginolyticus produce betalactamase and therefore are resistant to penicillin and ampicillin, but susceptibilities otherwise are similar to those listed for V. vulnificus.

LISTERIOSIS

(Circling disease)

  • AGENT

- Listeria monocytogenes, gram positive, pleomorphic rod

  • RESERVOIR AND INCIDENCE

Isolated from fish, birds, swine, horses, ruminants, guinea pigs, ferrets, gerbils, rabbits, and chinchillas. The principle reservoir of the organism is in forage, water, mud, and silage. The seasonal use of silage as fodder is frequently followed by an increased incidence of listeriosis in animals.

  • TRANSMISSION:

outbreaks have been reported associated with ingestion of unpasteurized milk and cheese and contaminated vegetables; some sporadic cases may also be due to foodborne transmission. Refrigeration of foods may provide selective growth of Listeria. Papular lesions on hands and arms may occur from direct contact with infectious material or soil contaminated with infected animal feces. In neonatal infections, the organism may have been transmitted from mother to fetus in utero or during passage through the infected birth canal. Person-to-person transmission through venereal contact is possible, as is infection from inhalation of the organism.

  • DISEASE IN ANIMALS:

Two forms exist, the meningoencephalitic and visceral. The former involves neurological signs with dullness and somnolence. Drooling and lack of interest in food and mastication soon follow. There is lateral deviation of the head with a tendency to circle. Paralysis then sets in with recumbency and death from respiratory failure. The visceral from involves abortion, with retained placenta. Microabscesses occur throughout the brain. Visceral lesions involve multiple foci of necrosis in the liver, spleen and heart. Placental lesions are characteristic with yellow necrotic foci and multiple granulomas in the fetal liver. Abscess formation in the eye can lead to blindness. Fatality is very high, approaching 3-30% in outbreaks.

  • DISEASE IN MAN:

Symptomless fecal carriage is common. Fever, headache, nausea, vomiting, endocarditis, granulomatous lesions in multiple organs, cutaneous involvement, coryza, conjunctivitis, metritis with abortion, sepsis, & meningitis. Granulomatous lesions and abscesses occur in the liver and other organs and beneath the skin. Focal necrosis in the placenta with mononuclear infiltration is seen. Fatality rates may exceed 20%.

  • DIAGNOSIS:

culture and isolation (special media required). Serologic tests are unreliable because of cross reactions with other bacterial species.

  • TREATMENT:

ampicillin plus an aminoglycoside or Trimethoprim-sulfamethoxazole.

  • PREVENTION\CONTROL:

caution and protective clothing when handling infected tissues. Pregnant women and immunocompromised individuals should avoid contact with potentially infective materials such as aborted animal fetuses and known infected persons; they should eat only properly cooked meats and pasteurized dairy products.


LEPTOSPIROSIS

[Weil's disease, Hemorrhagic jaundice (Leptospira icterohaemorrhagiae), canicola fever (L. canicola), dairy worker fever (L. hardjo)]

  • AGENT
Spirochete, Leptospira. Pathogenic leptospires belong to the species Leptospira interrogans, which is subdivided into more than 200 serovars. The main natural reservoirs for human infection vary with serovar: L. canicola in dogs, L. hardjo in cattle, L. pomona in swine, and L. icterohaemorrhagiae in rats.
  • RESERVOIR AND INCIDENCE

Rats, mice, field moles, guinea pigs, gerbils, squirrels, rabbits, hamsters, reptiles, nonhuman primates, livestock, and dogs. In one study, 40 % of stray dogs were seropositive. Rats and mice are common animal hosts for L. ballum. Infection in mice is inapparent and can persist for the animal's lifetime. *Rodents are the only major animal species that can shed leptospires throughout their life-span without clinical manifestations. Active shedding by lab animals can go unrecognized until personnel handling the animals become clinically ill.

  • TRANSMISSION:

Handling affected animals, contaminating hands, or abrasions with urine, or aerosol exposure during cage cleaning are most common. The organism is often transmitted to humans by the urine of the reservoir host. The organism may also enter through minor skin lesions and probably via the conjunctiva. Many infections have followed bathing or swimming in infected waters.

  • DISEASE IN ANIMALS:

In cattle, fever and anorexia occur with rapid decline in milk yield and atypical mastitis. Pregnant cows abort with retention of the placenta. Also, mild jaundice and severe anemia occurs with enlarged and friable liver and swollen kidneys. In pigs subclinical infection is common, though it can cause abortion and birth of weak piglets. In dogs and cats, gastroenteritis, jaundice, and nephritis may occur.

  • DISEASE IN MAN:

Ranges from inapparent infection to severe infection and death. Biphasic Illness a. Weakness, headache, myalgia, malaise, chills, & fever. b. Leukocytosis, painful orchitis (testes not usually enlarged), conjunctival effusion, and rash. Icteric leptospirosis (Weil's syndrome-usually caused by L. icterohaemorrhagiae) is the most severe form of the disease, characterized by impaired renal and hepatic function, abnormal mental status, hypotension, and a 5-10% mortality rate. Signs and symptoms are continuous and not biphasic.

  • DIAGNOSIS:

Early in the disease, the organism may be identified by darkfield examination of the patient's blood or by culture on a semisolid medium. Culture is difficult and requires several weeks. A rapid diagnosis is made with the DOT-ELISA test. *Leptospires can be recovered only from mature mice even though antibodies can be detected from infected mice of all ages.

  • TREATMENT:

Penicillins or tetracyclines. Can eliminate L. ballum from a colony (mice) with 1000 gm chlortetracycline HCL/Ton of feed for ten days.

  • PREVENTION\CONTROL:

Vaccination in cattle, swine, and dogs Avoid swimming in or drinking from potentially contaminated water. Protect workers by providing boots and gloves. Rodent control. Drain wet ground. Doxycycline chemoprophylaxis for persons at high exposure.

  • BORRELIOSIS

(Relapsing fever, tick-borne relapsing fever, spirochetal fever, vagabond fever, famine fever) A widely distributed bacterial infection spread from wild rodents by ticks or lice, with high fatality Tick-borne relapsing fever occurs in Africa, the Americas, Asia and possibly parts of Europe. The causative agents are Borrelia recurrentis and several other borrelia strains (bacterium). There is no vaccine.

  • RESERVOIR AND MODE OF TRANSMISSION:

Epidemic louse-borne infection is not considered zoonotic. Endemic tick-borne relapsing fever is transmitted from the natural wild rodent reservoir by tick bites to humans and dogs. Transovarial transmission in ticks occurs. Blood-borne person-to-person and intrauterine transmission have been reported.

  • INCUBATION PERIOD:

Humans: 1-15 days. Animals.Unknown.

  • CLINICAL FEATURES:

Humans. Sudden onset of fever lasting for 3-5 days ends with a crisis. Then a febrile period of 2-4 days is followed by one to ten or more recurrences of fever accompanied by severe headaches, nausea, vomiting, diarrhea, jaundice and sometimes a macular rash with bleeding due to thrombocytopenia. Meningitis and cranial nerve involvement are possible. Animals. Arthritis and fever predominate in infected dogs. The arthritis recurs and may progress to chronic deformity.

  • PATHOLOGY:

Humans. Many lesions occur, including enlarged, soft, infarcted spleen, hepatomegaly, hemorrhages in bone marrow and skin, myocarditis, bronchopneumonia, and meningitis. Animals. Arthritis, especially of the phalangeal joints, occurs with the possibility of progression to fibrosis of the joint capsule and ankylosis.

  • DIAGNOSIS:

Humans. Identify borrelia in thick blood smears. Otherwise isolate the pathogen by inoculation of blood into susceptible animals if possible. Animals. Inoculate blood or tissues into rats or mice.

  • PROGNOSIS:

Humans. The fatality rate is up to 40 per cent. Animals. Although fatality is uncommon, the lesions tend to be progressive.

  • PREVENTION:

Humans and animals. Control tick vectors and prevent tick bites.

  • TREATMENT:

Humans. A single dose of tetracycline or erythromycin, 0.5 g orally, or a single dose of procaine penicillin G, 600,000 units intramuscularly, probably constitutes adequate treatment for louse-borne relapsing fevers. Because of higher relapse rates, tick-borne disease is treated with 0.5 g of tetracycline or erythromycin given 4 times daily for 5-10 days. Jarisch-Herxheimer reactions may occur and respond to aspirin given every 4 hours. Pretreatment with steroids is not effective in preventing this reaction. Animals. Tetracycline, penicillin, erythromycin, and ceftriaxone at standard dosages for 21-28 days.

  • LEGISLATION:

Humans.Louse-borne relapsing fever is notifiable to the World Health organization. Tick-borne infection may be notifiable in some countries (e.g. the UK). Animals.None.

LYME DISEASE

(Lyme arthritis, Bannworth's syndrome, tick-borne meningopolyneuritis, erythema chronicum migrans [ECM], Steere's disease)

  • AGENT:

spirochete, Borrelia burgdorferi

  • RESERVOIR AND INCIDENCE

First implicated in 1982 as agent in a 1975 epidemic of juvenile inflammatory arthropathy in Old Lyme Connecticut. Cases have been reported from 46 states and the annual number of Lyme disease cases has increased 18 fold from 497 to 8803. It is now the most common tick transmitted disease in the USA. Also seen in Europe, England, Soviet Union, China, Japan, Southeast Asia, South Africa, Australia, and Canada.

  • TRANSMISSION:

Transmitted mostly by Ixodes dammini and other ixodid ticks (three host tick with a two to three year life cycle). Ixodes dammini has a broad range of hosts; adults prefer white tailed deer but will also parasitize dogs, horses, and humans. Larvae feed primarily on rodents, especially mice. Nymphs feed on all hosts and appears to be primarily responsible for transmission of the disease to people. Birds are an important reservoir and means of dispersal. Also found in Dermacentor, Rhipicephalus and Amblyomma and other ticks and biting insects, including mosquitoes, fleas, and biting flies. Because of lack of any proof to the contrary it is generally believed at this time that any potential increased risk to human beings from infected animals is attributable to animals bringing ticks into areas of human habitation rather than any pet transmission. Dogs appear to be at greater risk than humans.

  • DISEASE IN ANIMALS:

Serologic evidence has been reported in the dog, cat, horse, and ruminants. However, correlation with disease is lacking except in the dog. The dog exhibits the same symptoms as noted below for humans. Expanding skin lesions have been noted in mice and rabbits.

  • DISEASE IN MAN:

Multisystemic disease which may have chronic sequelae; an annular rash known as erythema chronicum migrans (ECM) develops in 60-80% of patients in the area of the tick bite and is considered pathognomonic. Also flu like symptoms, which resolve in about three weeks. 8-10% of people develop cardiac involvement several weeks later. Manifestations include atrioventricular block, cardiomyopathy, heart failure, myocarditis, and pancarditis. 15% develop neurologic disorders such as facial nerve palsies which usually resolve. Other manifestations include meningitis, cranial neuritis, radiculoneuritis, neuropathy, and encephalopathy. 60% develop the most common sequelae, arthritis. Disease may remain latent with symptoms developing 4 years after seroconversion.

  • DIAGNOSIS:

Most common test is detecting antibody titers by IFA or ELISA (on blood, CSF or synovial fluid). Culture is definitive but is difficult and requires special media such as Barbour-Stoener-Kelly media. Histologically with Dieterle Silver Stain or immunoperoxidase stains, but is often unrewarding.

  • TREATMENT:

A positive serology is no grounds for treatment when no clinical signs are present. Borrelia burgdorferi is sensitive to tetracycline and moderately sensitive to penicillin. amoxicillin, ceftriaxone, and imipenem are also highly active.

  • PREVENTION\CONTROL:

Tick control care when removing ticks or when handling potentially infective materials a vaccine against Lyme Disease tested in hamsters has been found effective. More research is needed but in the future vaccination may be beneficial for those at constant risk of exposure.




ENTERIC INFECTIONS

CAMPYLOBACTERIOSIS

(Vibriosis, vibrionic abortion)

  • AGENT:

Campylobacter (Vibrio) fetus ss. jejuni, a gram negative, microaerophilic, curved, motile rod that is worldwide in distribution.

  • RESERVOIR AND INCIDENCE

isolated from laboratory animals including dog, cat, hamsters, ferrets (>60 % in one study), nonhuman primates, rabbits, swine, sheep, cattle, and birds Although most cases of human campylobacteriosis are of unknown origin, infection after contact with sick animals has been well documented. *In most reports of pet to human transmission of C. jejuni, diarrheic puppies or kittens from pounds have been the source of infection. Pet birds, chickens, and kittens are implicated in other reports. A lab animal technician developed Campylobacter enteritis after feeding and cleaning up after a recently imported nonhuman primate. The organism was first isolated from nonhuman primates from Macaca fascicularis in 1979 and has since been reported in baboons, rhesus, patas, and marmosets. Can be shed for long periods of time in stool by asymptomatic carriers. Younger animals seem more likely to acquire the infection and hence may more commonly shed the organism.

  • TRANSMISSION:

Transmission is thought to occur by the fecal-oral route, through contamination of food or water, or by direct contact with infected fecal material. The organism has also been isolated from houseflies. At 40 C the organism is viable for three weeks in feces and milk, four weeks in water, and five weeks in urine. Campylobacter is shed in the feces for at least six weeks after infection. Infected children may transmit infection to puppies or kittens, which may then expose other children. Poultry and cattle are the main reservoirs for human infection, which is acquired by ingesting contaminated raw milk, undercooked chicken or other food contaminated in the kitchen.

  • DISEASE IN NONHUMAN PRIMATES:

Variable. the majority are asymptomatic carriers. Mild to severe enteritis may be seen accompanied by fever, vomiting, and mucus and blood in the feces. Bacteremia may occur complicated by meningitis or abortion. Most signs appear 1 to 7 days after exposure and affect primarily the jejunum, ileum, and colon.

  • DISEASE IN FERRETS:

Asymptomatic to proliferative colitis. Shed organisms for long period of time (> 16 weeks).

  • DISEASE IN OTHER ANIMALS:

Has also been shown to cause hepatitis in poultry, proliferative ileitis in hamsters, and abortion in ruminants. In all animals, it may be associated with diarrhea, especially when acting secondarily to virus infection.

  • DISEASE IN MAN:

Acute gastrointestinal illness, diarrhea with or without blood, abdominal pain, and fever. It may cause pseudoappendicitis and, rarely, septicemia and arthritis. Usually a brief, self-limiting disease. In humans the asymptomatic carrier state is rare. Reinfection is possible in both animals and man.

  • DIAGNOSIS:

1. Rapid diagnosis is done with dark field or phase contrast microscopy of fecal material. 2. This is confirmed by stool culture which requires a special selective growth media(CAMPY-BAP) and incubation at 43oC with 10% CO2, 5% O2 and 85% Nitrogen. 3. Warthin Starry stain and histo 4. Various techniques are being used to detect seroconversion to the antigens of Campylobacter.

  • TREATMENT:

Animals can be treated based on culture and sensitivity. Currently erythromycin is the drug of choice, but does not eliminate the carrier state. Tetracycline or ciprofloxacin are alternatives.

  • PREVENTION\CONTROL:

Vaccines provide partial protection of short duration and routine use is not recommended. Control is aimed at isolation of affected individuals and personal hygiene. An increased awareness of the potential of infection due to Campylobacter is of primary importance. Thoroughly cook all foodstuffs derived from animal sources, particularly poultry. Recognize, prevent, and control Campylobacter infections among domestic animals and pets. Wash hands after handling poultry and animal feces.

COLIBACILLOSIS

(Colibacteriosis, colitoxemia, white scours, gut edema of swine)

  • AGENT:

Escherichia coli are gram-negative, aerobic, and facultatively anaerobic medium-sized rods.

  • RESERVOIR AND INCIDENCE

Worldwide; some endemic areas exist in developing countries.

  • TRANSMISSION:

Some serotypes are species-specific, others are not. Milk, milk products, and meat products can contain pathogenic serotypes. Foods of animal origin and contact with dogs and cats have been indicated as sources of infection for children.

  • DISEASE IN ANIMALS:

Calf diarrhea (white scours) is an acute disease causing mortality in calves less than 10 days old. It manifests itself as serious diarrhea, with whitish feces and rapid dehydration. Mastitis caused by E. coli appears especially in older cows with dilated milk ducts. A long-term study of horse fetuses and newborn colts found that close to 1% of abortions and 5% of deaths of newborns were due to E. coli. Neonatal enteritis caused by E. coli in suckling pigs begins 12 hours after birth with a profuse watery diarrhea, and may end with fatal dehydration. Edema in suckling pigs (gut edema) is an acute disease that generally attacks between 6 and 14 weeks of age. It is characterized by sudden onset, incoordination, and edema of the eyelids, the cardiac region of the stomach, and sometimes other parts of the body. During septicemic diseases of fowl, such as cases of salpingitis and pericarditis, pathogenic serotypes of E. coli have been isolated. A colibacillary etiology has also been attributed to Hjarre's disease (coligranuloma), which is a condition in adult fowl characterized by granulomatous lesions in the liver, cecum, spleen, bone marrow, and lungs.

  • DISEASE IN HUMANS:

The enterotoxigenic stains (ETEC) cause profuse and watery diarrhea, abdominal colic, vomiting, acidosis, and dehydration. Enteroinvasive strains cause a dysenteric syndrome with mucoid diarrhea, at times tinged with blood. E. coli is also an important agent of urogenital infections.

  • DIAGNOSIS:

Stool culture or immunoassays for enterotoxins.

  • TREATMENT:

Ciprofloxacin or trimethoprim-sulfa.

  • PREVENTION/CONTROL:

With respect to man, control measures include: a) personal cleanliness and hygienic practices, sanitary waste removal and b) protection of food products. Vaccines for swine and bovine have been developed.


SALMONELLOSIS

(Enteric paratyphosis)

  • AGENT:

Gram negative bacteria. Out of 1600 recognized serotypes of Salmonella, S. typhimurium & S. enteritidis have been associated most commonly with lab animal colony infections. In the U.S., 5 million cases are diagnosed annually. S. typhi, the cause of Typhoid Fever, rarely occurs in the U.S. and is not discussed here.

  • RESERVOIR AND INCIDENCE

The organism inhabits the intestinal tract of many animals including birds, cattle, sheep, pigs, lab. animals (rats, mice, hamsters, guinea pigs, nonhuman primates) and humans. Salmonella occurs worldwide. *The house mouse may also be a reservoir of the infection and may play a role in human and animal salmonellosis. Humans, rarely, and animals may be carriers and asymptomatic shedders of the organism. *Salmonella prevalence in the U.S. canine population may be 10% or more. *Prevalence data from 8 studies conducted worldwide indicated that a wide range (0.6-27%) of cats were culture-positive for Salmonella. *Salmonella carriers in newly imported Rhesus and Cynomolgus monkeys exceeded 20% in some shipments. *Birds, reptiles, and turtles are especially dangerous sources of Salmonellosis. 94% of all reptiles harbor Salmonella. Turtles alone in 1970 may have caused 280,000 human cases of Salmonellosis. *In 1975 the FDA ruled it illegal to sell a. Viable Turtle eggs b. Live turtles with a carapace length < 10.2cm (4 inches) c. Exceptions - Educational & scientific institutions and marine turtles. d. Marine turtles have not been shown to be a reservoir of Salmonella THERE WAS A 77% DECREASE IN TURTLE ASSOCIATED SALMONELLOSIS AFTER ENACTMENT OF THIS LAW.

  • TRANSMISSION:

Indirect transmission via contaminated food and water are the most common sources but transmission may also be by direct contact. It is a common contaminant of sewage. Found in many environmental water sources. Environmental contamination continues to be a potential source of infection for lab animals and secondarily for personnel handling those animals. *Animal feed containing animal by products continues to be a source of Salmonella contamination, especially if the diets consist of raw meal and have not undergone the pelleting process.

  • DISEASE IN ANIMALS:

Can be asymptomatic with clinical signs precipitated by stress; penetration of the infection into the mucosa is followed by inflammation, especially ileitis, progressing to inflamed mesenteric lymph nodes in the mesentery, possibly progressing to septicemia and pneumonia especially in calves. Calves undergo epizootic outbreaks of diarrhea with high mortality. Abortion in cattle is caused by massive proliferation of salmonella in the placenta leading to placental necrosis. High percentage of survivors become carriers. Infected sheep, goats, and poultry usually show no signs of infection.

  • DISEASE IN MAN:

Acute gastroenteritis with sudden onset of abdominal pain, diarrhea, nausea, and fever. May lead to septicemia. May be an inapparent infection.

  • DIAGNOSIS:

Fecal Culture with selective media. Can get false negatives, though because organism is shed intermittently. In the carrier state bacterium resides in the gall bladder (NHP).

  • TREATMENT:

Symptomatic. Severely ill patients are treated with trimethoprim-sulfamethoxazole, ampicillin, or ciprofloxacin.

  • PREVENTION\CONTROL:

Rapid detection and treatment (acute and chronic) in lab animals. Treatment based on culture and sensitivity. Cull carrier animals. Watch during quarantine period. Sanitation and hygiene, protective clothing, gloves Rodent, bird & wild animal control is important. Examine feed and bedding and pasteurize or autoclave, if necessary. Consider screening animal care personnel for inapparent Salmonella infection to prevent the introduction of Salmonella into the colony from infected workers. Thoroughly cook all foodstuffs derived from animal sources. Exclude animal care personnel with diarrhea.


STAPHYLOCOCCAL FOOD POISONING

(Staphylococcal Alimentary Toxicosis, Staphylococcal Gastroenteritis)

  • AGENT:

Coagulase-positive strains of Staphylococcus aureus, a gram-positive cocci.

  • RESERVOIR AND INCIDENCE

Worldwide. The principle reservoir is the human carrier. Infected cows, fowls, and dogs may give rise to and be a source of staphylococcal poisoning in man.

  • TRANSMISSION:

A high proportion of healthy humans (30-35%) have staphylococci in the nasopharynx and on the skin. Sneezing, coughing, expectorating can contaminate food. Similarly, he may contaminate foods handled if he has a skin lesion. Milk from cow udders infected can contaminate numerous milk products. Contaminated egg contents can also be a source of infection.

  • DISEASE IN ANIMALS:

Mastitis in cattle. Pyoderma, impetigo, folliculitis, and furunculosis in dogs. In fowl, staphylococcal infection can cause diseases ranging from pyoderma to septicemia with different locations (salpingitis, arthritis, and other disorders).

  • DISEASE IN HUMANS:

The major symptoms are nausea, vomiting, abdominal pains, and diarrhea. It is the cause of toxic shock syndrome in women.

  • DIAGNOSIS:

Culture of vomitus, feces, or a suspected food item.

  • TREATMENT:

Electrolyte and fluid replacement. Ciprofloxacin.

  • PREVENTION/CONTROL:

Reduce food handling time. Exclude persons with boils, abscesses, and other purulent lesions from handling food. Educate food handlers in strict food hygiene.


ANTHRAX

(Malignant pustule, woolsorter's disease, charbon, malignant edema, splenic fever)

  • AGENT:

Bacillus anthracis, a gram-positive spore-forming aerobic rod.

  • RESERVOIR AND INCIDENCE

Occurrence is worldwide. Most mammals susceptible; most commonly seen in cattle, sheep, horses, swine, goats (guinea pigs, rabbits, mice, experimentally)

  • TRANSMISSION:

Herbivorous animals infected by ingestion of spores which are viable in soil for years. Man is infected by handling contaminated carcasses, wool, hide, or hair. Also can be infected by ingestion or inhalation of spores or bacilli. Recent outbreaks have been related to prospective military use of the organism.

  • DISEASE IN ANIMALS:

Sudden, acute illness, with high fever, localized swellings, bleeding from body orifices, death in 1 to 3 days. Blood is non-clotting.

  • DISEASE IN MAN:

Cutaneous form ("malignant pustule"): most common, incubation 1 to 7 days, then an erythematous papule appears on an exposed area of skin and becomes vesicular, with a purple to black center. The center of the lesion finally forms a necrotic eschar and sloughs. Regional adenopathy, fever, malaise, headache, and nausea and vomiting may be present. After the eschar sloughs, hematogenous spread and sepsis may occur, resulting in shock, cyanosis, sweating, and collapse. Hemorrhagic meningitis may also occur. 20% fatality if not treated. Pulmonary form ("woolsorter's disease"): following the inhalation of spores from hides, bristles, or wool, incubation 1 to 5 days, fever, cough, dyspnea, respiratory failure and death in 24 hours. 100% fatality. Intestinal form: incubation 12 hours to 5 days, anorexia, vomiting, diarrhea, 50% fatality if untreated.

  • DIAGNOSIS:

o Gram or Giemsa stain of sputum, blood or tissue o culture

  • TREATMENT:

Penicillin G. For mild cases, tetracycline.

  • PREVENTION\CONTROL:

o Vaccine for livestock and high risk personnel. o avoid necropsy of suspect animals, o disinfect wool or hair from animals in endemic areas with 10% formalin, 5%lye


STAPHYLOCOCCAL FOOD POISONING

  • SYNONYMS:

Staphylococcal alimentary toxicosis, staphylococcal gastroenteritis.

  • ETIOLOGY:

Coagulase-positive strains of Staphylococcus aureus that produce enterotoxins. Very few coagulase-negative stains are enterotoxigenic. The toxin is preformed in the food involved. To date, six types of enterotoxins are known: A, B, C, D, E, and F; of these A is the most prevalent in outbreaks. Enterotoxin F is implicated in toxic shock syndrome (TSS). Some strains can produce two or even three different enterotoxins. The toxins are heat-resistant and can withstand a temperature of 100oC for 30 minutes.

  • GEOGRAPHIC DISTRIBUTION:

Worldwide.

  • THE DISEASE IN MAN:

The incubation period is short, generally 3 hours after ingestion of the food involved. The interval between consumption of the enterotoxin and the first symptoms can vary from 30 minutes to 8 hours, depending on the quantity of toxin ingested and the susceptibility of the individual. The major symptoms are nausea, vomiting, abdominal pains, and diarrhea. Some patients may show low pyrexia (up to 38oC). More serious cases manifest prostration, cephalalgia, abnormal temperature, and lowered blood pressure, as well as blood and mucus in the stool and vomit. The course of the disease is usually benign and the patient recovers without medication in 24 to 72 hours. Recently, a toxic shock syndrome has been described. Symptoms consist of vomiting, diarrhea, high fever, erythroderma, edema, renal insufficiency, and toxic shock. Most patients are women who become ill during their menstrual period. The above-described symptoms also are observed in association with abscesses and osteomyelitis caused by S. aureus. A staphylococcal enterotoxin designated F was isolated from 94% of these patients strains of S. aureus from nine patients with toxic shock were examined, and production of enterotoxin F was confirmed in eight of them; only 42% of 50 strains isolated from other hospitalized patients produced this toxin. Toxin F production was not found in 48 strains originating from animal clinical specimens. Of 24 strains from healthy human carriers, 25% produced the toxin.

  • SOURCE OF INFECTION AND MODE OF TRANSMISSION:

The principal reservoir of is S. aureus is the human carrier. A high proportion (from 30 to 35%) of healthy humans have staphylococci in the nasopharynx and on the skin. A carrier with a respiratory disease can contaminate foods by sneezing coughing, or expectorating. Similarly, he may contaminate foods he handles if he has a staphylococcal skin lesion. However. even if not sick himself, the carrier may spread the agent by handling food ingredients. utensils, and equipment. or the finished food product. According to different authors, the proportion of enterotoxin-producing S. aureus strains of human origin varies between 18 and 75%. The proportion of toxigenic strains isolated from various sources (human, animal, and food) is very high. Strains of human origin predominate in epidemics, but animals are also reservoirs of the infection. Milk from cow udders infected with staphylococci can contaminate numerous milk products. Many outbreaks have been produced by consumption of inadequately refrigerated raw milk or cheeses from cows whose udders harbored staphylococci. The largest outbreak affected at least 500 students in California between 1977 and 1981 and was traced to chocolate milk. In developing countries, where refrigeration after milking is often inadequate, milk and milk products may be an important source of staphylococcal intoxication. According to recent investigations, a high proportion of strains isolated from staphylococcal mastitis produce enterotoxin A, which causes many outbreaks in humans. In several investigations it was possible to isolate from skin lesions and cow's milk the S. aureus phage type 80/81, which is related to epidemic infections in man. One of the studies proved that phage type 80/81 produced interstitial mastitis in cows. The same phage type was found among animal caretakers, which indicates that the bacterium is intertransmissible between man and animals and that the latter may reinfect man. Infected fowl and dogs may also give rise to and be a source of staphylococcal poisoning in man. One subject that deserves special attention is the appearance of antibiotic-resistant strains in animals whose food includes antibiotics. Concern exists over the possible transmission of these strains to man. On several occasions, resistant stains have been found both in animals (cows, swine, and fowl) and in their caretakers, with the same antibiotic resistance. Moreover, "human" strains (phage typed) have on occasion been isolated from the nostrils and lesions of other species of domestic animals. A variety of foods and dishes may be vehicles of the toxin. If environmental conditions are favorable, S. aureus multiplies in the food and produces enterotoxins. Once made, the toxin is not destroyed even if the food is subjected to boiling while being cooked. Consequently, the toxin may be found in the food whereas staphylococci are not. An important causal factor in food-borne intoxications is holding food at room temperature, which permits multiplication of staphylococci. Lack of hygiene in food handling is another notable factor. Frequently, outbreaks of food poisoning may be traced to a single dish.

  • THE ROLE OF ANIMALS IN THE EPIDEMIOLOGY OF THE DISEASE:

Most outbreaks are caused by human strains, and to a lesser degree by strains from cattle and other domestic animals. Animal products -- such as meat, ham, milk, cheese, cream, and ice cream -usually constitute a good substrate for staphylococcal multiplication. Milk pasteurization offers no guarantee of safety if toxins were produced before heat treatment, as the toxins are heat-resistant. Outbreaks have been caused by reconstituted powdered milk, even when the dried product contained few or no staphylococci.

  • DIAGNOSIS:

The short incubation period between ingestion of contaminated food and appearance of symptoms is the most important clinical criterion. Laboratory confirmation, when possible, is based above all on demonstration of the presence of enterotoxin in the food. Biological methods (inoculation of cats with cultures of the suspect food, or of rhesus monkeys with the foodstuffs or cultures) are expensive and not always reliable. As substitutes, serologic methods such as immunodiffusion, immunofluorescence, hemagglutination inhibition, and, recently, ELISA are increasingly used. In febrile patients, blood cultures are indicated. Isolation of enterotoxigenic staphylococcal strains from foods and typing by phage or, more recently, by immunofluorescence have epidemiologic value. Quantitative examination of staphylococci in processed or cooked foods serves as an indicator of hygienic conditions in the processing plant and of personnel supervision.

  • TREATMENT:

In humans, treatment usually consists of replacement of fluids and electrolytes and, very rarely, management of hypovolemic shock and respiratory embarrassment. If botulism is suspected, polyvalent antitoxin must be administered. Historically, antimicrobial drugs have not been recommended unless a specific microbial agent producing progressive systemic involvement can be identified. Preliminary data now suggest that ciprofloxacin, 500 mg every 12 hours for 5 days, may shorten the duration of diarrhea and lead to a more rapid resolution of symptoms. Antimotility drugs may relieve cramping and decrease diarrhea in mild cases. Their use should be limited to patients without fever and without dysentery (bloody stools), and they should be used in low doses.

  • CONTROL:

It includes the following measures: a) education of persons who prepare food at home or commercially in proper personal hygiene; b) exclusion from handling food of individuals with abscesses or other skin lesions; and c) refrigeration of all foods to prevent bacterial multiplication and formation of toxins. Foods should be kept at room temperature as little time as possible. The veterinary milk inspection service should supervise dairy installations, ensuring that refrigeration units function correctly and are used immediately after milking, and that milk is refrigerated during transport to pasteurization plants. The veterinary meat inspection service should be responsible for enforcing hygienic regulations before and after slaughter as well as during handling and production of meat products. Control of hygienic conditions in meat retail establishments ts also important.


LEPROSY

(Hansen's disease)

  • AGENT:

Mycobacterium leprae, a polymorphic acid-fast bacillus.

  • RESERVOIR & INCIDENCE:

The world prevalence is estimated to be between 10 and 12 million. Newly recognized cases in the USA are diagnosed principally in California, Hawaii, Texas, Florida, Louisiana, and New York City, and in Puerto Rico. Most of these cases are in immigrants and refugees whose disease was acquired in their native country; however, the disease remains endemic in Hawaii, Texas, California, Louisiana and Puerto Rico. Man is the only significant reservoir. Armadillos, mangabey monkeys, and chimpanzees can acquire the disease from humans. Epidemiologic data on leprosy in the U.S. reveal very high ratios of native to foreign-born leprosy patients in Texas and Louisiana in comparison to all other states. These are the 2 states known to have the highest prevalences of leprosy in wild armadillos.

  • TRANSMISSION:

Respiratory and involves prolonged exposure in childhood. Only rarely have adults become infected.

  • DISEASE IN ANIMALS:

The disease in armadillos (Dasypus novemcinctus) is similar to the lepromatous form in man. Infection in these animals is characterized by macrophage infiltrates containing a large number of bacilli. M. leprae is known to prefer the coldest parts of the human body. For this reason, armadillos are used as experimental animals since their body temperature is 30-35oC. In chimpanzees, the disease appears as a progressive chronic dermatitis with nodular thickening of the skin of the ears, eyebrows, nose, and lips. Sooty mangabey monkeys develop lepromatous leprosy with neuropathic deformities of the extremities, including clawing of the digits.

  • DISEASE IN HUMANS:

In lepromatous leprosy, nodules, papules, macules and diffuse infiltrations are bilaterally symmetrical and usually numerous and extensive; involvement of the nasal mucosa may lead to crusting, obstructed breathing and epistaxis; ocular involvement leads to iritis and keratitis. In tuberculoid leprosy, skin lesions are single or few, sharply demarcated, anesthetic or hyperesthetic, and bilaterally asymmetrical; peripheral nerve involvement tends to be severe.

  • DIAGNOSIS:

Demonstration of acid-fast bacilli from skin or nasal septum scrapings.

  • TREATMENT:

Combination therapy is recommended since single-drug treatment is accompanied by emergence of resistance. Lepromatous leprosy is treated with dapsone, clofazimine, and rifampin. Tuberculoid leprosy is treated with dapsone and rifampin.

  • PREVENTION/CONTROL:

early detection and treatment. Contact isolation for lepromatous leprosy but not necessary for tuberculoid leprosy.


DERMATOPHILOSIS

(Streptotrichosis, Mycotic Dermatitis of Sheep)

  • AGENT:

Dermatophilus congolensis. An aerobic actinomycete with gram positive long branching filaments and coccoid bodies.

  • RESERVOIR AND INCIDENCE

Occurs in temperate regions worldwide. Natural disease described in horses, cattle, sheep, goats, cottontail rabbits, owl monkeys, lizards & humans.

  • TRANSMISSION:

The etiologic agent is an obligate parasite that has been isolated only from lesions in animals. Human cases have arisen from direct contact with infected animals. The most common means of transmission between animals is mechanical thru arthropod vectors. The infection may also be transmitted by means of objects, such as plant thorns or shears.

  • DISEASE IN ANIMALS:

Circumscribed areas of alopecia, elevated crusty papillomatous lesions, and exudative dermatitis. **Owl monkeys may have relapsing Dermatophilosis after apparently appropriate antibiotic regimens. Therefore organism may persist on pelage of animals after resolution of lesions. In cats, the lesions differ from those of other domestic animals by affecting deeper tissues. In cats, granulomatous lesions have been found on the tongue, bladder, and popliteal lymph nodes.

  • DISEASE IN MAN:

pustular desquamative dermatitis

  • DIAGNOSIS:

Microscopic exam of stained material from lesions and culture.

  • PREVENTION\CONTROL:

1. Treatment with antibiotics 2. Isolate affected animals 3. Protective clothing, gloves, personal hygiene 4. Tick control


ERYSIPELOID

(Rosenbach's erysipeloid, erythema migrans, erysipelotrichosis, rose disease in swine, diamond skin disease in swine, fish-handler's disease or fish rose in man)

  • AGENT:

Erysipelothrix rhusiopathiae (insidiosa). 22 different serotypes are recognized. *Discovered by ROBERT KOCH==He called it the Bacillus of Mouse Septicemia

  • RESERVOIR AND INCIDENCE

Saprophyte in soil, water, and decaying organic matter. Pathogen in swine, lambs, calves, poultry, fish, & wild and lab mice. *Pigs probably represent the most likely source of exposure in the zoophile community. (Natural disease or zoonotic transmission from rodents has NOT been reported.)

  • TRANSMISSION:

contamination of wounds while handling infected tissues

  • DISEASE IN ANIMALS:

Diamond skin disease in pigs. Arthritis in sheep and swine. Cyanosis and hemorrhages in turkeys. Can be septicemic disease in many species.

  • DISEASE IN MAN:

Disease in humans is called Erysipeloid, and is primarily occupation related. Inflammatory lesions of the skin, with elevated erythematous edge; spreads circumferentially. Septicemia is an infrequent complication.

  • DIAGNOSIS:

Culture from lesion or blood

  • PREVENTION\CONTROL:

Treatment with Penicillin Gloves when handling animals Vaccine for swine and turkeys


MELIOIDOSIS

(Pseudoglanders, Whitmore's disease)

  • AGENT:

Pseudomonas pseudomallei (Malleomyces pseudomallei, Actinobacillus pseudomallei ) --MOTILE, Gram negative rod

  • RESERVOIR AND INCIDENCE

Normal inhabitant of surface soil and water in Southeast Asia, and tropical areas. Recent studies have shown that the water of tanks in which exotic aquarium fishes were imported was contaminated. Occurs in wild rodents, goats, pigs, sheep. Also identified in Chimps, orangutans, and macaques. There is no evidence that animals are important reservoirs, except in the transfer of the agent to new foci. Rare in the U.S. except in drug users.

  • TRANSMISSION:

by inhalation from moist soil-water reservoir, by contact with contaminated soil or water thru overt or inapparent skin wounds, or by ingestion of contaminated feeds. Can be venereal in man.

  • DISEASE IN ANIMALS:

Signs include loss of weight, swelling of joints, fever, cough, and chest pain. Skin lesions with fistulous tracks can develop. Emaciation and multiple abscesses in lung, bone, viscera. A chronic draining purulent skin lesion in a primate is suspect. Incubation period can be 6 months to three years. Sheep seem especially susceptible- over 25% mortality can occur in outbreaks.

  • DISEASE IN MAN:

Clinical disease is not common in man but subclinical disease in endemic areas based on serology is common. It may simulate typhoid fever or TB including pulmonary cavitation, empyema, chronic abscesses and osteomyelitis. High case fatality rate (80%) in people who do develop clinical signs.

  • DIAGNOSIS:

Culture and isolation from lesions, a rising serological titer is confirmatory.

  • TREATMENT:

ceftazidime. Alternates: Chloramphenicol or Trimethoprim-sulfamethoxazole.

  • CONTROL:

Safe disposal of sputum and wound discharges.


GLANDERS

(Farcy)

  • AGENT:

Pseudomonas mallei (Actinobacillus mallei) NONMOTILE, gram negative rod

  • RESERVOIR AND INCIDENCE

disease of equidae and rarely man. Occasionally reported in dogs, cats, sheep, and goats. Mostly seen in Asia and Mediterranean areas, rare in North America.

  • TRANSMISSION:

spread by contamination by infectious discharges of wounds and mucus membranes and by ingestion.

  • DISEASE IN ANIMALS AND MAN:

pulmonary form: cough, nasal discharge. cutaneous form: multiple, purulent, cutaneous eruptions, often following lymphatics. Usually affects hind legs of horses. May have long periods of remission. The fatality rate in humans is 95% if left untreated. Horses usually suffer chronic and sometimes fatal illness. Asses and mules usually suffer acute disease which is often fatal.

  • DIAGNOSIS:

Cannot be differentiated from P. pseudomallei serologically. Specific diagnosis can be made only by characterization of the isolated organism.

  • TREATMENT:

Streptomycin + tetracycline or chloramphenicol + streptomycin

  • PREVENTION/CONTROL:

1. Treatment with antibiotics 2. Elimination of carrier animals 3. Gloves, protective clothing when handling infected animals.


TULAREMIA

(Francis' disease, deer-fly fever, rabbit fever, O'Hara disease)

  • AGENT

- Francisella tularensis, a small pleomorphic, gram-negative, nonmotile rod or coccobacillus that can survive several weeks in the external environment.

  • RESERVOIR AND INCIDENCE

Common often fatal septicemic disease of rabbits, squirrels, muskrats, deer, bull snakes, sheep, wild rodents, cats and dogs. Major reservoirs are RABBITS, TICKS, MUSKRATS. Has been reported in NHP's at an urban zoo.

  • TRANSMISSION:

handling tissue of infected animals (direct contact with UNBROKEN skin is sufficient). Reported human infections due to a cat bite and scratch and a NHP bite also reported. transmitted by biting insects inhalation, ingestion

  • DISEASE IN ANIMALS:

Clinical signs usually occur alongside heavy infestation with ticks, and include sudden high fever, anorexia and stiffness, eventually leading to prostration and death. In sheep, pregnant ewes may abort. Affected dogs have soft nodular swellings under the skin. Miliary foci of necrosis occur in the liver, spleen and lymph nodes. Severe lesions in the lung involve widespread consolidation with edema and pleurisy.

  • DISEASE IN MAN:

Fever, headache, and nausea begin suddenly, and a local lesion-a papule-develops and soon ulcerates. Regional lymph nodes may become enlarged and tender and may suppurate. The local lesion may be on the skin of an extremity (ulceroglandular disease) or in the eye. Pleuropulmonary disease may develop from hematogenous spread or may be primary after inhalation. Following ingestion of infected meat or water, an enteric (typhoidal) form may be manifested by enteritis, stupor, and delirium. In any type of involvement, the spleen may be enlarged and tender and there may be nonspecific rashes, myalgias, and prostration. A case fatality rate of 5-10% mainly from the typhoidal or pulmonary form exists.

  • DIAGNOSIS:

Culture (requires specialized laboratory and dangerous, therefore, not recommended) A positive agglutination test (>1:80) develops in the second week after infection and may persist for several years.

  • TREATMENT IN MAN:

streptomycin + tetracycline. Chloramphenicol may be substituted for tetracycline.

  • PREVENTION\CONTROL:

wear impervious gloves while handling animals or tissues cook the meat of wild rabbits and rodents thoroughly vaccine available for high risk personnel avoid bites of flies, mosquitos, and ticks and avoid drinking, bathing, swimming in untreated water in endemic areas.



STREPTOCOCCOSIS

  • AGENT:

The causative agents are various streptococci species, including Streptococcus suis and S. zooepidemicus.

  • RESERVOIR AND INCIDENCE

Pigs are the reservoir of S. suis.

  • TRANSMISSION:

Humans are infected with S. suis by handling infected meat. S. zooepidemicus has occurred in persons in direct contact with domestic animals and from drinking raw milk.

  • DISEASE IN ANIMALS:

S. suis epizootics may occur in pigs with high mortality, heralded by signs of meningitis including depression, fever, incoordination and paralysis. Suppurative arthritis may occur. More usually the disease is subclinical. S. zooepidemicus may cause mastitis in cattle.

  • DISEASE IN MAN:

S. suis causes fever and occasionally meningitis. S. zooepidemicus may cause upper respiratory tract symptoms, cervical adenitis, pneumonia, endocarditis and nephritis. A fatality rate of 8% has been reported for S. suis, with residual deafness in a high proportion of survivors.

  • DIAGNOSIS:

isolation and culture

  • TREATMENT:

Benzathine Penicillin G. For persons allergic to penicillin, erythromycin is an effective alternative. However, increasing reports of resistance from Europe threatens its clinical utility.

  • PREVENTION/CONTROL:

Exercise caution in handling pig meat. Dress all wounds to avoid contamination. Pasteurize milk.


RAT BITE FEVER

(Streptobacillary fever, Haverhill fever, epidemic arthritic erythema, sodoku)

  • AGENT:

Gram negative, pleomorphic bacillus. Two different agents can cause disease: 1. Streptobacillus moniliformis (Haverhill Fever) *Named after a 1926 outbreak in Haverhill, Mass. attributed to contaminated milk. 2. Spirillum minus (Sodoku)

  • RESERVOIR AND INCIDENCE

Present in the oral and respiratory passages of a large number of asymptomatic rodents, including Rats and Mice. Incidence of disease appears to be low. Historically, wild rat bites and subsequent illness (usually small children) relate to poor sanitation and overcrowding.

  • TRANSMISSION:

Man infected by bite of infected rodent or via contaminated milk or food

  • DISEASE IN ANIMALS:

Rats: inapparent infection Mice: acute, systemic, fatal disease in immunologically inexperienced mice. Surviving mice (or if endemic disease), exhibit suppurative polyarthritis, swelling and loss of digits or limbs.

  • DISEASE IN MAN:

Acute febrile disease following bite from a rodent. Can see inflammation, lymphadenopathy, and nonspecific signs. May exhibit rash on extremities, often soles and palms. May see arthritis with S. moniliformis. Incubation period variable: S. moniliformis: hours to 1 to 3 days S. minus: 1 to 6 weeks Symptoms usually resolve spontaneously. Complications, if not treated promptly, lead to pneumonia, hepatitis, enteritis, endocarditis with a 10% fatality rate.

  • DIAGNOSIS:

Culture: S. moniliformis requires 10 to 20% horse or rabbit serum and reduced oxygen tension. S. minus- won't grow in vitro. Must inoculate culture specimens into lab animals and use dark field microscopy.

  • TREATMENT:

Treat with procaine penicillin G or tetracycline HCl. Give supportive and symptomatic measures as indicated.

  • PREVENTION/CONTROL:

Bacteriologic monitoring Proper treatment of rodent bites


PASTEURELLOSIS

(Shipping or transport fever, hemorrhagic septicemia)

  • AGENT:

Pasteurella multocida, small, nonmotile, polymorphic, gram-positive bacilli

  • RESERVOIR AND INCIDENCE

inhabits the oral cavity and upper respiratory tract of many animals (Rabbits, rodents, dogs, cats, mice, birds, swine). Dogs and cats are frequently healthy carriers.

  • TRANSMISSION:

All animals and birds may be colonized by pasteurellas, and human infection occurs by wound infection from bites or scratches. Animal-to-animal transmission may occur by ingestion and inhalation. 1986 case report of meningitis in a woman who kissed her dog (cultured positive for organism) and also had dental caries which was considered to be the route of infection.

  • DISEASE IN ANIMALS:

can cause acute pneumonia or septicemic disease in many species. May cause chronic infection of upper respiratory and middle ear especially in the rabbit.

  • DISEASE IN MAN:

Local inflammation occurs around the bite or scratch, possibly leading to abscess formation with systemic symptoms.

  • TREATMENT:

Penicillin, tetracycline, or cephalosporin

  • PREVENTION/CONTROL:

Proper treatment of bite Protective clothing (mask,gloves) Euthanize aggressive dogs and cats. Vaccinate cattle and sheep.


ANTHRAX

(Malignant pustule, wool-sorters' disease, charbon, malignant edema, splenic fever) An acute bacterial infection of humans and animals which may be rapidly fatal. The disease occurs worldwide and is enzootic in certain African and Asian countries. It is an occupational hazard of persons such as wool-sorters, fellmongers, knackermen, farm workers and veterinarians in contact with infected animals or their products (e.g., blood, wool, hides and bones). The causative agent is Bacillus anthracis (bacterium).

  • RESERVOIR AND MODE OF TRANSMISSION:

All domestic, zoo and wild animals are potentially at risk of infection. Anthrax bacilli are released from infected carcasses and form resistant spores on exposure to air. These spores contaminate soil for many years. Humans are usually infected by inoculation from direct contact with infected animals, carcasses or animal products and contaminated soil. Inhalation or ingestion of spores may occur. Animals are infected from contaminated feed, forage, water or carcasses.

  • INCUBATION PERIOD:

Humans. Cutaneous 3-10 days inhalation 1-5 days gastrointestinal 2-5 days. Animals. 1-5 days.

  • CLINICAL FEATURES:

Humans. Various forms include: 1. Cutaneous anthrax; localized ulceration and scab with fever and headache which may be followed within a few days by septicemia and meningitis. 2. Inhalation anthrax; fulminating pneumonia. 3. Intestinal anthrax; acute gastroenteritis with bloody diarrhea. Animals. Peracute cases are found dead or moribund. Acute cases show fever, excitation followed by depression, incoordination, convulsion and death. Chronic cases show edema of throat, pharynx and brisket, especially in pigs.

  • PATHOLOGY:

Humans. Features include black scab (eschar) with edema, enlargement of regional lymph nodes and possibly septicemia; pneumonia and generalized hemorrhages. Animals. Carcasses should not be opened, hence necropsy is rarely carried out. Main features include failure of the blood to clot and hemorrhages throughout the body. The spleen is enlarged and softened. The subcutaneous swelling, mainly about the neck and throat of affected pigs and horses, contains gelatinous fluid. The blood contains very large numbers of B. anthracis.

  • DIAGNOSIS:

Humans. Identify B. anthracis in stained blood smears or by inoculation of animals. Culture swabs from wounds. Animals. As for humans. Specific antigen for anthrax may be found in animal products (e.g. hides) using a precipitin (Ascoli) test.

  • PROGNOSIS:
Humans. Untreated cutaneous anthrax has a fatality rate of 5-20 Per cent and gastrointestinal anthrax of 25-75 per cent. Pulmonary anthrax is usually fatal. Animals. The condition is usually fatal in cattle unless treated early. Pigs and horses are more resistant.
  • PREVENTION:

Humans. Prohibit contact with infected animals and their products. Establish environmental and personal hygiene (e.g., ventilation and protective clothing) where a special risk exists. Treat wounds promptly and disinfect imports of hairs and wool. Vaccination may protect those occupationally exposed to risk. Apply strict safety measures. Isolate infected patients, with concurrent disinfection. Animals. Sterilize, or avoid using, meat and bone meal from high-risk countries for animal feed. Vaccinate livestock grazing in enzootic area. Dispose of infected carcasses safely and fence off areas contaminated by inadequately buried carcasses.

  • TREATMENT:

Humans. The mortality rate is high despite proper therapy, especially in pulmonary disease. Penicillin G, 2 million units IV every 4 hours, is the therapy of choice. tetracycline, 500 mg orally every 6 hours, may be used for mild, localized cutaneous infection. Animals. Penicillin injection of all animals showing fever after the first case is confirmed. This involves checking temperatures twice daily.

  • VACCINATION:

Humans. Offered to workers at risk. Animals. Non-encapsulated Stern strain vaccine can be used in all species of domestic animal. Annual vaccination of grazing animals using spore or alum precipitated antigen vaccine in areas of high risk is recommended.

  • LEGISLATION:

Humans. The disease is notifiable in most countries. It is a recognized occupational disease in some countries, including the UK. Animals. Notifiable in many countries with mandatory disposal of infected carcasses by burning or deep burial under lime. Opening of moving suspect carcasses is prohibited.


CLOSTRIDIAL INFECTIONS

(Clostridial myositis: black leg, malignant edema, gas gangrene. Enterotoxemia: pulpy kidney, struck, lamb dysentery, braxy. Tetanus: lockjaw)

  • AGENT:

Clostridium tetani (horses)--tetanus Clostridium perfringens (cattle, sheep)--gas gangrene Clostridium septicum (cattle) malignant edema

  • RESERVOIR AND INCIDENCE

Clostridia are normal intestinal flora and also survive by spores in the soil.

  • TRANSMISSION:

Infection may be by contamination of deep, penetrating wounds to cause tetanus, by ingestion of preformed toxin or spores which vegetate in the digestive tract to cause enterotoxemia, or by ingestion of spores which are carried by the blood to muscles where they remain dormant until activated by trauma to produce necrotizing myositis. C. perfringens food poisoning is due to spore contamination of foods which survive heating to vegetate in unrefrigerated conditions. Neonatal tetanus in humans is frequently caused by contamination of the umbilicus.

  • DISEASE IN ANIMALS:

Tetanus: as in humans. In myositis (black leg) cases a limb is still and painful with crepitus on palpation. Signs of toxemia. Rapidly fatal. C. chauvoei, novyi, and septicum toxins produce massive muscle necrosis, often with edema and SC gas formation. C. perfringens causes a variety of profound toxemias with cloudy swelling of parenchymatous organs and excess fluids, often bloodstained in serous cavities.

  • DISEASE IN MAN:

Tetanus: painful toxic contractions of muscles and trismus. The case fatality rate for tetanus is 30-90% even when treated. Gas gangrene: fever, toxemia, painful edema spreading from the edges of wounds, interstitial emphysema, neck stiffness. Food poisoning: vomiting and diarrhea of a few days' duration.

  • TREATMENT:

Myositis: penicillin, adequate surgical debridement and exposure of infected areas. Tetanus: immune globulin, penicillin, mechanical ventilation. Spasms are controlled with chlorpromazine or diazepam combined with a sedative.

  • PREVENTION/CONTROL:

immunization with toxoids (good for 10 years), proper treatment of wounds. Good food hygiene is essential. In animals, prevent wound contamination during lambing, shearing, castration and docking.


CAPNOCYTOPHAGA

  • AGENT:

Capnocytophaga canimorsus (formerly Dysgonic fermenter-2), a recently described aerobic, gram negative bacillus with unusual fermentation pattern.

  • RESERVOIR AND INCIDENCE

Found as part of oral flora of normal dogs and cats. C. canimorsus has been isolated from the mouths of 24% and 17% of normal dogs and cats respectively. Serious infections in man are most commonly reported in splenectomized or immunocompromised people, alcoholics, or persons who have chronic respiratory disease. More than 40 cases reported, many fatal, since first reported in 1976.

  • TRANSMISSION:

Contact, bite or scratch from dog or cat

  • DISEASE IN MAN:

can lead to cellulitis and overwhelming bacteremia, meningitis, endocarditis, septic arthritis, and DIC. The organism appears to have an affinity for the eye, causing angular blepharitis and severe keratitis. Accidental corneal inoculation occurred during a tooth extraction in a Poodle causing severe refractory keratitis in a veterinarian. The predisposition of the cornea to infection may be due to its avascularity and to the low concentrations of immunoglobulins and complement components in the tissue. Most serious disease and fatalities have occurred in splenectomized people. Case fatality rates of 4-27% have been reported.

  • DIAGNOSIS:

History, clinical signs, and culture. ORGANISM IS SLOW GROWING. May require 8 days of incubation. Micro exam of blood smear or buffy coat with gram stain to detect organisms.

  • PREVENTION/CONTROL:

Awareness, especially of high risk individuals Treatment of bite wounds, Penicillin G. (Treatment of high risk people even without sign of infection recommended.)


PSITTACOSIS

(Ornithosis, Parrot Fever, Chlamydiosis)

  • AGENT:

Obligate,intracellular organism with a unique development cycle and worldwide distribution Genus Chlamydia has only four species, many strains 1. Chlamydia trachomatis- humans, mice (Zoonotic potential not known) 2. Chlamydia psittaci- BIRDS, Mice, g. pig, rabbits, cats, frogs, ruminants 3. Chlamydia pneumoniae- humans 4. Chlamydia pecorum- ruminants

  • RESERVOIR AND INCIDENCE

The mammalian strains appear to be a zoonotic problem only rarely. 2 cases of human conjunctivitis reported from close association with cats with chlamydial pneumonitis and conjunctivitis. Birds are the main reservoir of human infection, however, 25% of human cases have no history of avian contact. Ovine strains may infect pregnant women.

  • TRANSMISSION:

Inhalation; dry feces produce highly infective aerosols Direct contact with feces or respiratory secretions May survive in dust for several months.

  • DISEASE IN ANIMALS:

There are many strains of C. psittaci which produce a diverse disease spectrum in animals, e.g., conjunctivitis, air sacculitis, pericarditis, hepatitis, meningoencephalitis, enteritis, urethritis, arthritis, and endometritis with abortion. G.I. infection results in enteric shedding of the organism. Latency - Well recognized feature of Chlamydia infection, i.e., the organism can cause inapparent infection or fulminant infection in the same host. In clinically healthy birds, stress can precipitate clinical signs and shedding of the organism.

  • DISEASE IN MAN:

Asymptomatic or clinical disease after 1-2 week incubation period. Fever, chills, myalgia, anorexia, headache, nonproductive cough. Pneumonitis or atypical pneumonia may be present. May see a toxic or septic form with hepatosplenomegaly, hepatitis, meningoencephalitis and cardiac involvement with endocarditis. Ovine chlamydial infection in pregnant women is life-threatening, causing late abortion and neonatal death and disseminated intravascular coagulation in the mother.

  • DIAGNOSIS:

fecal culture (rarely successful) serology (CF, IFA) [Note: African Grey Parrot, cockatiel, and budgie may remain serologically negative despite active infection.] ELISA-based tests for antigen in feces has proven reliable.

  • TREATMENT:

Tetracycline or Erythromycin.

  • PREVENTION/CONTROL:

Treatment with tetracycline Introduce birds into colony from psittacosis-free flocks or use chlortetracycline chemoprophylaxis. Protective clothing (masks, gowns, gloves). Wild caught birds should be placed on chlortetracycline during quarantine. In sheep, keep flocks closed or vaccinate annually. Isolate aborting ewes until discharges cease.


BLASTOMYCOSIS

SYNONYMS: North American blastomycosis, Chicago disease, Gilchrist's disease.

  • ETIOLOGY:

Blastomyces dermatitidis, a dimorphic fungus existing in mycelial form in cultures and as a budding yeast in the tissues of infected mammals.

  • GEOGRAPHIC DISTRIBUTION:

The disease has been observed in the United States, eastern Canada, Zaire, Tanzania, South Africa, and Tunisia. Autochthonous cases may have occurred in some Latin American countries.

  • THE DISEASE IN MAN:

The incubation period is not well known; it possibly extends to several weeks or months. Blastomycosis is a chronic disease that principally affects the lungs. The respiratory symptomatology initially resembles influenzas purulent or bloody expectoration, weight loss, and cachexia, in addition to fever and cough, may develop later. If the infection remains localized, it can become asymptomatic. When it disseminates, it can cause subcutaneous abscesses as well as localized infections in several organs. Death frequently results in cases of untreated disseminated infection. The cutaneous form is commonly secondary to the pulmo-nary and is characterized by an irregular-shaped, scabby ulcer that has raised borders and contains minute abscesses. Lesions develop on exposed parts of the body.

  • THE DISEASE IN ANIMALS:

The highest incidence is observed in dogs around 2 years of age. The symptoms consist of weight loss, chronic cough, dyspnea, cutaneous abscesses, fever, anorexia, and sometimes blindness. The lesions localize in the lungs, lymph nodes, eyes, skin, and joints and bones. Of 47 clinical cases recently described, 72% occurred in large males. There were lesions of the respiratory tract in 85% of the cases.

  • SOURCE OF INFECTION AND MODE OF TRANSMISSION:

The reservoir is environmental, probably the soil, but the ecologic biotope has not been determined. Transmission to man and to animals is effected by aerosols; the fungal conidia are the infecting element. Persons at highest risk are those having the most contact with the soil. Dogs most frequently infected are sporting and hunting breeds.

  • ROLE OF ANIMALS IN THE EPIDEMIOLOGY OF THE DISEASE:

None. It is a disease common to man and animals. Cases of transmission from individual to individual (man or animal) are not known.

  • DIAGNOSIS:

Diagnosis is based on direct microscopic examination of sputum and material from lesions, on isolation of the agent in culture media, and on examination of histologic preparations. B. dermatitidis grows well in Sabouraud's culture medium or other adequate median it is most distinctive in its sprouting yeast form, and therefore the inoculated medium should be incubated at 37oC, since at ambient temperature the mycelial form of the fungus is obtained. B. dermatitidis in its yeast form (in tissues or cultures at 37oC) is characterized by a single bud attached to the parent cell by a wide base, from which it detaches when it has reached a size similar to the parent cell. In contrast, Paracoccidioides brasiliensis, the agent of paracoccidioidomycosis ("South American blastomycosis"), has multiple buds in the yeast phase. Serologic tests in use are complement fixation and gel immunodiffusion; the latter gives better results. It should be borne in mind that cross-reactions with Histoplasma and Coccidioides may occur. At present, the intradermal test is considered to have no diagnostic value.

  • TREATMENT:

Humans. Itraconazole, 100-200 mg/d orally, is now the therapy of choice for nonmeningeal disease, with a response rate of over 70%. Amphotericin B is given for treatment failures or cases with central nervous system involvement. Follow-up for relapse should be regularly made for several years so that therapy may be resumed or another drug instituted. Animals. Rare primary cutaneous disease may persist for months; these lesions should be removed surgically since blastomycosis responds poorly to therapy. Amphotericin B is considered the drug of choice, but treatment is of little avail once the disease is disseminated. The combination of amphotericin B and ketoconazole has been suggested to reduce the rate of relapse.

  • CONTROL:

As long as the ecologic biotope remains poorly defined, practical prevention methods cannot be established.


CAT SCRATCH DISEASE

(Cat Scratch Fever, Benign Lymphoreticulosis, Benign nonbacterial Lymphadenitis, Bacillary Angiomatosis, Bacillary Peliosis Hepatis)

  • AGENT:

Controversial, it is not currently possible to definitively name the causative agent responsible for CSD. Felt to be either Afipia felis, a gram-negative rod or Rochalimaea henselae and Rochalimaea quintana. Both are members of class Proteobacteria and both are intracellular parasitic bacteria.

  • RESERVOIR AND INCIDENCE

Associated with domestic cats throughout the USA, and worldwide. Over 6000 cases annually. Seen more often in men than in women . Have seen clusters of infection within families within a 2 to 3 week period, suggesting that shedding by cats may occur periodically. Other sources of infection have included scratches from other species including dogs, squirrels, and goats and from wounds induced by crab claws, barbed wire, and plant material.

  • TRANSMISSION:

90% of patients have been exposed to a cat. 75% of these have been bitten, scratched, or licked. Most affected individuals are <20 years of age. 75-80% of the cases of CSD are diagnosed between September and February with a peak incidence in December. 4 to 6% of the general population and 20% of veterinarians have positive skin test reactions to CSD antigen.

  • DISEASE IN ANIMALS:

Subclinical

  • DISEASE IN MAN:

Different distinct syndromes exist:

Typical CSD A primary lesion, most common on neck or extremities, will develop in 50% of the cases and appear approximately 10 days after a bite or scratch. A pustule persists for 1-2 weeks. 10-14 days after the lesion appears, lymphadenopathy develops and usually regresses within 6 weeks. 30-50% of the enlarged nodes become suppurative. Of the approximately 65% who develop systemic illness, fever and malaise are the symptoms most often noted. The disease is usually benign and most patients recover spontaneously without sequelae within 2-4 months. Many unrecognized cases probably occur. Disease appears to confer lifelong immunity.

Atypical CSD The atypical forms of CSD, which constitute 11% of all cases, are extremely varied. The most common, representing 6% of all cases, is Parinaud's oculoglandular syndrome (POGS), or granulomatous conjunctivitis with preauricular adenopathy. Other, atypical presentations include tonsillitis, encephalitis, cerebral arteritis, transverse myelitis, radiculitis, granulomatous hepatitis and/or splenitis, osteolysis, atypical pneumonia, hilar adenopathy, pleural effusion, erythema nodosum, erythema annulare, maculopapular rash, thrombocytopenic purpura, and breast tumor. Bacillary Angiomatosis Dermal BA presents in several ways. The commonest form is an enlarging red papule with some resemblance to a cranberry, often with a collarette of scale and sometimes with a suggestion of surrounding erythema. This type of lesion may be mistaken for pyogenic granuloma, unless fairly deep biopsy specimens are examined. These lesions begin as small papules and enlarge, occasionally becoming several centimeters in diameter and rarely ulcerating. They may be single or quite numerous. Another form of dermal BA is a deeper, subcutaneous nodule that appears flesh-colored and may be either fixed to subcutaneous tissues or freely mobile. Rarely BA may present as a dermal plaque. BA has been reported to occur in every organ system, including the brain, and is often difficult to differentiate from mycobacterial and fungal infections or malignancy without the use of biopsy. It is unclear if the personality changes, ranging from frank psychosis to depression, that have been described in association with BA represent CNS involvement or a neurotoxic product of this infection. Bacillary Peliosis Hepatis BPH, a vasoproliferative condition involving the liver of HIV-infected patients, is characterized by a proliferation of cystic blood-filled spaces surrounded by fibromyxoid stroma in which one can see bacteria similar to those seen in BA. Clinically these patients may or may not have visible bacillary angiomas. Their symptoms usually include fever, weight loss, and abdominal pain or fullness. Physical exam may reveal organomegaly. Studies usually demonstrate elevation of alkaline phosphatase and ç-glutamyltransferase levels out of proportion to those of aminotransferase and bilirubin.

  • DIAGNOSIS:

The sedimentation rate is elevated, the white blood cell count normal, and the pus from the nodes is sterile. ID skin testing with antigen prepared from the pus is positive. Excisional biopsy, usually performed to exclude lymphoma, confirms the diagnosis.

  • TREATMENT:

For CSD: Rifampin, ciprofloxacin, gentamycin, and trimethoprim-sulfa. Aspiration of suppurating nodes is recommended for relief of pain. Symptoms resolve without treatment in 2-4 months. BA and BPH respond to erythromycin, rifampin, or doxycycline. Therapy must be continue for 4-6 weeks to avoid relapse.

  • PREVENTION/CONTROL:

Education. Wash hands after handling cat. Wash cuts and scratches promptly and don't allow cat to lick open wound.

RICKETTSIAL DISEASES

Q-FEVER

(Query fever, Balkan influenza, Balkan grippe, pneumorickettsiosis, abattoir fever)

  • AGENT:

Coxiella burnetii Multiplies only in living cells. Stains red with Gimenez & Macchiavello stains and purple with Giemsa. Infections in lab workers have been recognized for many years. Serious hazard in research facilities where infected "asymptomatic" ewes are used for projects.

  • RESERVOIR AND INCIDENCE

Found worldwide in wild and domestic animals in two self perpetuating cycles: 1. Wild animals, with numerous tick hosts 2. Domestic animals - sheep, goats, cattle. Widespread in sheep in the U.S. Dogs, cats, and chickens can also be infected. Enzootic infection among domestic animals is the main reservoir of infection for humans.

  • TRANSMISSION:

Organism shed in urine, feces, milk, and especially birth products of domestic ungulates that generally do not show clinical disease (usually sheep and goats). Organism is resistant to drying and can persist for months while providing extensive environmental contamination. Aerosol is a major means of transmission. Contact with infected tissues: placenta of the infected ewe contains 109 organisms per gram of tissue. Amniotic and fetal tissues are highly infective. Soiled linen may infect personnel in the laundry. One organism is considered to be enough to cause infection in humans. Ingestion.

  • DISEASE IN MAN:

Two weeks to one month incubation. Febrile illness or subacute endocarditis. No skin eruption or rash, which distinguishes it from other Rickettsial species infections. Severe frontal headache with retro-orbital pain, profuse sweating, myalgia, and nausea. Pulmonary involvement in half the cases. Asymptomatic in many cases. Most cases resolve in two weeks but may be protracted or relapsing in the elderly. Chronic endocarditis, particularly in persons with preexisting valvular disease, is difficult to treat and the case fatality rate may be as high as 60%.

  • DIAGNOSIS:

Leukopenia with a diagnostic rise in specific CF antibodies to Coxiella phase 2. The Weil-Felix test (a test specific for typhus and other rickettsial diseases) is negative. Liver function tests are often abnormal. In Q fever endocarditis, there is a titer of 1:200 or more by CF or IFA with phase 1 antigen. Isolation of the organism from blood or sputum is rarely attempted due to zoonotic concerns.

  • TREATMENT:

Treatment with tetracyclines can suppress symptoms and shorten the clinical course but does not always eradicate the infection. Even in untreated patients, the mortality rate is usually low, except with endocarditis. Treatment of endocarditis consists of protracted (often for years) of antibiotic therapy; valves often need replacement.

  • PREVENTION/CONTROL:

Use male or nonpregnant female sheep for research, when possible. Q-Fever free sheep - limited practicality because requires intense surveillance program and frequent testing. Also, serologic status is not a useful indicator of whether the animal is shedding virus. Personnel education and control. Physical separation of infected animals from humans are current methods ofcontrol. Restrictions on movement of animals within thefacility (with considerations of air handling). Label all potentially infected material and sterilize or disinfect it. Protective clothing, masks, gloves, & shoe covers. Intensive medical surveillance and health education program. Treatment of acute disease in humans with tetracycline. Experimental vaccine for sheep has shown promise. Delayed hypersensitivity skin test is available for high risk personnel.

  • SUITABLE DISINFECTANTS FOR Q-FEVER:

1:100 dilution of chlorine bleach containing 5-25% hypochlorite. 5% hydrogen peroxide. 1:100 Lysol.


EHRLICHIOSIS

(Tick-borne fever)

  • AGENT:

an intraleukocytic rickettsia, E. canis (many species of Ehrlichia exist. Previously only E. sennetsu was known to infect man). Occurs intracytoplasmically, singly or in compact clusters (morulae) in circulating leukocytes.

  • RESERVOIR AND INCIDENCE

First recognized in dogs in 1935. Epizootic occurred in military working dogs in Vietnam 1968-1970. Now known to have worldwide distribution. 11 to 58% of dogs in U.S. are serologically positive. First reported case of E. canis in man in 1987. Several cases since then.

  • TRANSMISSION:

tick vector, Rhipicephalus sanguineus, Brown Dog Tick. It is presumably transmitted to humans by tick bite.

  • DISEASE IN DOGS:

Incubation period 10 to 14 days. Fever, lymphadenopathy, edema of legs and scrotum, epistaxis. Acute disease followed by a subclinical carrier stage.

  • DISEASE IN MAN:

similar to Rocky mountain spotted fever, but no rash. 12 to 14 day incubation period and prodrome consisting of malaise, back pain and nausea, the patient develops sudden fever, bradycardia, and headache. Leukopenia and absolute lymphopenia as well as thrombocytopenia occur frequently.

  • DIAGNOSIS:

Not easy to identify in peripheral blood smears but can attempt to identify organisms in leukocytes. An IFA assay that may be used to diagnose infection is available thru CDC and requires acute and convalescent sera.

  • TREATMENT:

Tetracycline

  • PREVENTION/CONTROL:

Control ticks


ROCKY MOUNTAIN SPOTTED FEVER

(American Tick Typhus, Tick-borne Typhus Fever)

  • AGENT:

Rickettsia rickettsii.

  • RESERVOIR AND INCIDENCE

Dogs, wild rodents and rabbits. Reported from most of continental U.S., highest incidence in S. Atlantic and South Central States. 2/3 of human cases are reported in children.

  • TRANSMISSION:

Ixodid ticks (especially Dermacentor) or their host species. Most rickettsias are obligate intracellular parasites of the gut cells of invertebrates and can only survive briefly outside living cells. Crushed ticks or mites and their feces may infect through broken skin. Transmission from tick bite occurs only after several hours of attachment.

  • DISEASE IN ANIMALS:

Subclinical only.

  • DISEASE IN MAN:

Fever has a sudden onset, with chills, headache, severe muscle pains, photophobia and meningism for four weeks. A red, morbilliform rash develops within 3-5 days of onset of fever and with hemorrhages spreading on limbs. Enlarged liver and spleen, myocarditis, renal tubular necrosis and bronchopneumonia occur. Damage to endothelial cells of blood vessels by invasion of rickettsias causes thrombi and hemorrhages. Focal liver necrosis, hemorrhages in genitalis and gangrene of the scrotum may occur. The case fatality rate in untreated cases is 15-20%, but with prompt treatment is about 5%.

  • DIAGNOSIS:

Rickettsiae can sometimes be isolated in special laboratories from blood obtained in the first few days of illness. A rise in antibody titer during the second week of illness can be detected by specific CF, IFA, and microhemagglutination tests or by the Weil-Felix test. Antibody response may be suppressed if antimicrobial drugs are given very early.

  • TREATMENT/PREVENTION/CONTROL:

Treatment of human disease with tetracycline or chloramphenicol. Control ticks on newly arrived animals.


RICKETTSIALPOX

(Vesicular Rickettsiosis, Kew Gardens Spotted Fever)

  • AGENT:

R. akari

  • RESERVOIR AND INCIDENCE

House mouse is reservoir host; most commonly seen in rodent infested urban dwellings ie New York City and other Eastern U.S. cities. Rats and moles can also harbor the organism. Not identified as a natural disease in domestic rodents.

  • TRANSMISSION:

Mite, Allodermanyssus sanguineus, transmits to mice or to man. Lab infections in humans via respiratory route have occurred but lab infections due to mite bite have not been reported.

  • DISEASE IN ANIMALS:

Not known in wild animals. In experimental mice death follows pneumonia.

  • DISEASE IN MAN:

Illness lasting about a week is associated with an eschar which develops at the site of the mite bite, regional lymphadenopathy and fever. A vesicular rash over the body and limbs develops within 1-4 days.

  • DIAGNOSIS:

Leukopenia and a rise in antibody titer with rickettsial antigen in CF tests. However, the Weil-Felix test is negative.

  • TREATMENT:

Tetracycline

  • PREVENTION/CONTROL:

Eliminate wild mice from animal facilities Control mites.


MURINE TYPHUS

(Flea-borne Typhus Fever, Endemic Typhus Fever, Urban Typhus)

  • AGENT:

Rickettsia typhi

  • RESERVOIR AND INCIDENCE

natural pathogen of rats and mice. Other mammals including cats, and their ectoparasites have been found infected. Outbreaks continue to occur in U.S., especially Texas. Natural lab infections have not been reported but lab acquired infections in people handling experimentally infected mice have been documented.

  • TRANSMISSION:

transmitted by flea or lice (Xenopsylla cheopis, Nosopsyllus fasciatus) to rodents or man. Humans are infected by contamination of flea bites, broken skin or conjunctiva by flea feces. Domestic animals may transport the flea vector to humans. Inhalation of contaminated dust may be a route of infection.

  • DISEASE IN ANIMALS:

The agent localizes in the brain and various organs but with no known lesions.

  • DISEASE IN MAN:

There is a gradual onset of fever with severe headache, rigors, generalized pains and dry cough (sometimes developing to bronchopneumonia) of about 2 weeks. A macular rash appears by about 5 days, first appearing on the trunk and lasting about six days. CNS manifestations are possible. Damage is caused to vascular endothelia by invasion of rickettsia, possibly leading to thrombosis and hemorrhage. In untreated case, the case fatality rate is 1-2%.

  • DIAGNOSIS:

CF or IFA.

  • TREATMENT:

Tetracycline or chloramphenicol.

  • PREVENTION/CONTROL:

control wild rodents. In endemic areas control fleas while exterminating rats.


FUNGAL INFECTIONS

DERMATOMYCOSES

(Ringworm, Dermatophytosis, Tinea, Trichophytosis, Microsporosis, Jock Itch, Athlete's Foot)

  • AGENT:

Organisms are subclassified into: 1. Geophilic - inhabit soil 2. Zoophilic - parasitic on animals 3. Anthropophilic - Primarily infects humans All can produce disease in humans. Grouped in three genera 1. Microsporum 2. Trichophyton 3. Epidermophyton

  • RESERVOIR AND INCIDENCE

Fungal spores remain viable for long periods on carrier animals and fomites. Exposure to reservoir hosts harboring different dermatophytes determines the type and incidence of infection in humans. Microsporum canis can be carried by up to 89% of nonsymptomatic cats. Up to 50% of people exposed to infected cats, both symptomatic and asymptomatic, have acquired infection. Pets may also acquire disease from humans. **T. mentagrophytes is most commonly transmitted to man from rodents, M. canis from dogs and cats, and T. verrucosum from cattle and horses.

  • TRANSMISSION:

Direct or indirect contact with asymptomatic animals or with skin lesions of infected animals Contaminated bedding Equipment Fungi in air, dust, or on surfaces of room (spores persist on contaminated surfaces)

  • DISEASE IN ANIMALS:

In rodents is often asymptomatic and not recognized until people are affected. In cats is often asymptomatic. Dogs often show classic skin lesions. Varying severity of dermatitis occurs with local loss of hair. Deeper invasion produces a mild inflammatory reaction which increases in severity with the development of hypersensitivity.

  • DISEASE IN MAN:

Often mild, self limiting; scaling, redness, and occasionally vesicles or fissures. Thickening & discoloring of nails. May show circular lesions which clear in the center forming a ring. Fungal infections in man are categorized as to the location on the body: 1. Tinea capitis - Scalp & hair 2. Tinea corporis - Body (extremities, arm and hand, are most often affected in infections acquired from lab animals.) 3. Tinea pedis - foot 4. Tinea unguium - Nails

  • DIAGNOSIS:

KOH mount of skin scrapings, Fungal culture

  • TREATMENT:

Macerated (moist softening and fissuring) stage- aluminum subacetate ("Domeboro") solution soaks with potassium permanganate for secondary infections. Athlete's feet may respond better to 30% aqueous aluminum chloride or the carbol-fuchsin paint than to antifungal agents. Broad spectrum antifungal creams and solutions containing imidazoles or ciclopirox) instead of tolnaftate and haloprogin help to combat diphtheroids and other gram-positive organisms present at this stage and alone may be adequate therapy. Dry and scaly stage- several topical creams, liquids, or lotions are recommended (miconazole, clotrimazole, ketoconazole, econazole, sulconazole, oxiconazole, ciclopirox or naftifine. Betamethasone dipropionate with clotrimazole is recommended for acutely inflamed tinea lesions. For severe cutaneous infections, griseofulvin or ketoconazole is recommended.

  • PREVENTION/CONTROL:

Screen newly received animals. Routine sanitization of contaminated environment, equipment, and caging. Gloves, protective clothing, wash hands after exposure.


SPOROTRICHOSIS

  • AGENT:

Sporothrix schenckii, dimorphic fungus

  • RESERVOIR AND INCIDENCE

distributed worldwide with sporadic outbreaks. Saprophyte that grows on soil or vegetation. Has been reported in horses, dogs, cats and many other species. 19 cases of Sporotrichosis have been reported in man following contact with feline abscesses or ulcers (12 of these were veterinarians).

  • TRANSMISSION:

Penetrating injury, or contact with plant or soil. Transmission from feline to man by direct contact with lesions (penetrating injury NOT required). Inhalation can lead to pulmonic disease.

  • DISEASE IN ANIMALS:

Lesions can be anywhere but are usually located on distal extremities, head, or base of tail. Appear as draining puncture wounds, cellulitis. Can ulcerate and become nodular with seropurulent exudate. May cavitate and expose muscle and bone. May lead to disseminated disease which is usually fatal

  • DISEASE IN MAN:

The most common clinical form is cutaneolymphatic; it begins with a nodule or pustule at the point where broken skin allowed inoculation. The infection may remain confined or may eventually spread and produce SC nodules along the enlarged lymph ducts. These nodules may ulcerate, and a gray or yellowish pus appears. Disseminated forms, which are rare, may give rise to localizations in different organs, especially the bones and joints, as well as in the mouth, nose, kidneys, or the SC tissue. Pulmonary sporotrichosis can be confused with TB.

  • DIAGNOSIS::

Biopsy and histo fungal culture cytology of impression smear of exudate. Organisms are numerous in cats but are difficult to detect in man, horses, and dogs. Organism can also be found in feces of infected cat. Antigen tests are used to diagnose disseminated disease.

  • TREATMENT:

Potassium iodide, Amphotericin B, or itraconazole.

  • PREVENTION/CONTROL:

Gloves when handling infected animals. Wash with iodine or chlorhexidine.


CRYPTOCOCCOSIS AND HISTOPLASMOSIS

Cryptococcosis and histoplasmosis are systemic fungal infections caused by the organisms Cryptococcus neoformans and Histoplasma capsulatum. Cases of infection are worldwide and sporadic. These organisms are found in soil, pigeons, and starlings and are spread in bird feces. These systemic fungi have a predilection for the lung and central nervous system. There is an increased susceptibility in humans on steroid therapy and with disorders of the reticuloendothelial system.

VIRAL DISEASES

Pox Viruses -Many poxvirus infections of lab animals are also pathogenic for humans. Humans usually exhibit proliferative cutaneous or subcutaneous self-limiting lesions. Occasionally see systemic disease, particularly when diseases of NHP's involved.


CONTAGIOUS ECTHYMA

(Orf, Contagious pustular dermatitis, Contagious Pustular Stomatitis, Ecthyma Contagiosum)

  • AGENT:

Genus Parapoxvirus of Family Poxviridae.

  • RESERVOIR AND INCIDENCE

- Sheep and goats worldwide.

  • TRANSMISSION:

- crusted areas on muzzle, eyelids, oral cavity, feet, or external genitalia are laden with virus. Transmitted easily from animals to man by contact. The virus is highly resistant to adverse environments and persists for many years.

  • DISEASE IN ANIMALS:

necrosis in the skin and mucous membranes of the gastrointestinal and urogenital tracts. Intense pain can interfere with eating.

  • DISEASE IN MAN:

Large painful nodules usually distributed on the hands. Weeping red surfaces. These resolve with minimum scarring 1-2 months later.

  • DIAGNOSIS:

Diagnosis is made by a history of contact with sheep, goats, or wild ungulates; by EM demonstration of the poxvirus in the lesion; cell culture; or serologically.

  • PREVENTION/CONTROL:

Wear rubber gloves when handling infected sheep and when working in an environment near infected sheep.


MONKEY POX

  • AGENT:

Orthopoxvirus Disease in humans is indistinguishable from smallpox, (Variola) i.e., serologic & clinical syndrome.

  • RESERVOIR AND INCIDENCE

Animals: Nine reported outbreaks in captive NHP's, primarily rhesus and cynomolgus. Has also been reported in languors, baboons, chimpanzees, orangutans, marmosets, gorillas, gibbons, and squirrel monkeys. The virus has been isolated from a wild squirrel. Man: The first human case of Monkey Pox was reported in 1970. Between 1970 and 1986, over 400 cases had been reported from tropical rain forested areas of West and Central Africa.

  • TRANSMISSION:

Transmission can be via direct contact, aerosol, ingestion, or parenteral administration. Person to person transmission can occur.

  • DISEASE IN NONHUMAN PRIMATES:

Usually exhibit a high morbidity and low mortality. Clinical signs may be inapparent or an animal may exhibit fever, lymphadenopathy, and cutaneous eruptions of the extremities, trunk, lips, or face. Cynos seem to be most severely affected. Death is uncommon except in infant monkeys.

  • DISEASE IN MAN:

Signs in man include fever, sore throat, headache, and a vesiculopustular rash of peripheral distribution which clears up in 5 to 25 days. Severe complications include bronchopneumonia, vomiting, and diarrhea. Case fatality rate 10-15%. Although the disease is not common in man it is important from the standpoint of differentiating it from smallpox.

  • DIAGNOSIS:

based on progression of lesions, histopathology and virus isolation. On histological examination epidermal cells contain eosinophilic cytoplasmic and intranuclear inclusions. ELISA

  • TREATMENT:

Symptomatic.

  • PREVENTION/CONTROL:

Sanitation, isolation. Vaccination with vaccinia virus is protective in both man and nonhuman primates.


YABAPOX

  • AGENT:

Poxvirus

  • RESERVOIR AND INCIDENCE

Affects mangabeys, rhesus, cynos, vervets, stumptails, and patas monkeys. Latent infection in African species that can infect Asian primates and U.S. born African primates.

  • TRANSMISSION:

Need further clarification of the epidemiology of this disease. Role of insect vectors has not been determined. Aerosol transmission has been proven experimentally. *Transmission to humans from monkeys has not been recorded. The virus can affect man usually after accidental skin puncture.

  • DISEASE IN NONHUMAN PRIMATES:

Subcutaneous benign tumors (Histiocytomas) that may reach several cm. in diameter. They usually regress spontaneously in 3 to 6 weeks.

  • DISEASE IN MAN:

Lesions similar to those seen in monkeys

  • PREVENTION/CONTROL:

Usual care should be exercised by animal handlers, including wearing of protective clothing.


TANAPOX

(Benign Epidermal Monkeypox, BEMP)

  • AGENT:

Tanapox virus.

  • RESERVOIR AND INCIDENCE

Monkeys. In 1966 23 human cases were reported in the U.S. among personnel who worked with monkeys affected at 3 primate centers. A serologic study carried out on 263 monkeys of Asian origin (Macaca) revealed a 15% rate; in 55 African Green Monkeys, the rate was 76%.

  • TRANSMISSION:

Aerosols or vectors. Human cases have resulted from contamination of abrasions or scratches.

  • DISEASE IN NONHUMAN PRIMATES:

Lesions occur primarily on the face, consisting of raised areas with a central scab. Papules ulcerate, scab and heal.

  • DISEASE IN HUMANS:

There is a fever for a few days, with headache and prostration and a single skin vesicle. Cytoplasmic inclusions are seen in skin lesions. Within 3 weeks of onset, the lesion spontaneously regresses.

  • DIAGNOSIS:

EM of skin scrapings or viral isolation.

  • TREATMENT:

Symptomatic.

  • PREVENTION/CONTROL:

Mosquito control. Asian and African monkeys should be housed separately. Wear protective clothes.


HERPESVIRUSES

There are more than 35 Herpesviruses of NHPs most of which are NOT zoonotic.


HERPES B

(Herpesvirus simiae, Simian B Disease)

  • AGENT:

Double-stranded DNA Virus. Direct zoonosis. For each Herpesvirus there exists a host for which the virus is almost uniformly fatal and reservoir hosts in which the virus exists in subclinical or latent infection. a. H. suis latent in swine, fatal in cattle. b. H. simplex I latent in man, fatal in Aotus, Gibbon, Marmoset (especially). c. H. saimiri I latent in Saimiri sciureus, fatal in Marmosets, Aotus (also known as Herpes T, Herpes M, Herpes Tamarinus). d. H. saimiri II latent in Saimiri sciureus, fatal in Marmosets, Aotus (this is an oncogenic virus, causing Malignant Melanoma of Reticulum Cell Type). e. H. simiae latent in rhesus and other macaques, fatal in man (also known as Herpes B Virus). f. Liverpool Vervet Monkey Virus: reservoir host ???, fatal in Cercopithecus aethiops. The Herpesviruses are serologically distinct but do share some antigenic properties. Herpes B Virus has been shown to have some antigenic relationship to Herpesvirus simplex by serum neutralization tests. However, antibody from Herpesvirus simplex does not confer immunity to Herpes B Virus.

  • RESERVOIR AND INCIDENCE

Incidence of infection of the reservoir host is normally high but you rarely see clinical disease. Clinical disease is usually confined to the very young or to the immunologically compromised. On occasion lethal infection occurs in the reservoir host: in humans, generalized Herpes simplex is often fatal in infants; in swine, Herpes suis is fatal in piglets. Natural infection does not result in elimination of the virus or in immunity but produces a latent infection or carrier, which always makes vaccination of reservoirs impractical. Herpesvirus B was first described in 1934 by Sabin/Wright. The virus was recovered from a laboratory worker who had been bitten 18 days previously by an apparently healthy rhesus. Since 1934, there have been 24 reported cases, all fatal, except four. Of the four, only one is reasonably free of severe neurological deficits. (Patients have remained in coma for as long as 40 months prior to succumbing to the disease.) Incubation is considered to be 10-20 days from exposure to the virus; however, in the Pensacola cases, clinical disease occurred within 4-5 days of exposure. Contact with macaques does not constitute exposure. The susceptibility of man to clinical B infection is low although mortality is high.

  • TRANSMISSION:

direct contact, including sexual transmission, aerosols, and fomites.

  • THE DISEASE IN NHP:

The reservoir hosts for Herpesvirus simiae are monkeys of the genus Macaca. Monkeys incriminated include M. mulatta (rhesus), M. fascicularis (cyno), M. fuscata (Japanese macaque), and M. arctoides (Stump-tail macaque). B virus may produce mild cold-sore type lesions, primarily at mucocutaneous junctions, mucous membranes, and tongue. But, it has been demonstrated that infection is not confined to the mouth but can also be found in the genitalia. These lesions are similar to those caused by Herpesvirus simplex I in man. Two factors have been associated with antibody increases with age; 80-100% of imported adult rhesus may have antibody compared to 20% imported juveniles. The second factor was type of caging. Animals housed together had significantly higher titers than individually-caged animals. Clinical disease may develop at the time of primary infection but it is not known if lesions invariably follow infection. However, periodic shedding of the virus may occur without the presence of visible lesions. Once a monkey is infected with B Virus, it should be considered infective for life.

  • THE DISEASE IN MAN:

Human infection is characterized by encephalitis with diplopia; nystagmus, patch paresthesia of head, neck and upper extremities. Acute abdominal pain, fever and diarrhea have also been observed prior to neurologic symptoms. Patients may also have a vesicular rash and/or keratoconjunctivitis. The histopathological changes resemble those of fatal, systemic Herpesvirus simplex in infants: encephalitis, myelitis and foci of necrosis in liver, spleen, lymph nodes and adrenals. The case fatality rate is 70%.

  • DIAGNOSIS:

ELISA and viral isolation.

  • TREATMENT:

acyclovir.

  • PREVENTION/CONTROL:

Control includes personal hygiene, protective clothing and common sense in handling monkeys. The virus is susceptible to oxidizing agents, soap, and water. Guidelines for prevention and treatment have been developed. The reader is referred to the reference by Holmes, GP, et al for further details. Emphasis can not be too strong concerning the use of protective clothing when entering a room with macaques. A perfectly healthy monkey may be lethal to you.


Arthropod borne ARBOVIRUSES:

GENERAL: In most man is an accidental host infected when arthropods feed on him. Therefore quarantine of wild caught animals and elimination of ectoparasites should prevent: Examples: TICK BORNE viruses: 1. Russian-Spring-Summer Encephalitis 2. Louping Ill MOSQUITO BORNE viruses, ie 1. DENGUE (Breakbone Fever, Dengue Hemorrhagic Fever)

  • AGENT:

Flavivirus, Flavidviridae RESERVOIR: nonhuman primates, occurs in Asia, Africa, Australia, the Caribbean including Puerto Rico, the Pacific Islands, S. Europe, S. America

  • TRANSMISSION:

mosquito vector (Aedes)

  • DISEASE IN NONHUMAN PRIMATES:

subclinical DISEASE IN HUMANS: The first phase of illness is usually mild fever, headache, myalgia, lymphadenitis, pharyngitis, rhinitis and cough lasting 1-5 days and is followed by 1-2 days of remission. The second peak of fever is accompanied by a morbilliform maculopapular rash. Severe hemorrhagic manifestations occur during the second phase especially in children.

  • DIAGNOSIS:

> HI, CF, ELISA, or virus isolation. TREATMENT: Treat shock by expanding circulating blood volume. Acetaminophen given for discomfort. Prolonged convalescence.

  • PREVENTION/CONTROL:

Mosquito control by screening and insect repellents. Examples:

  1. EQUINE ENCEPHALITIDES (EASTERN, WESTERN, AND VENEZUELAN). These are diseases of horses, mules, birds, humans, and other animals caused by the neurotropic viruses, Alphavirus and Flavivirus. Wild birds serve as a reservoir, and the virus requires an arthropod vector to transfer from reservoir host to other susceptible species. Mosquitoes, chicken mites and lice, ticks, and any other bloodsucking insects can serve as vectors. This group of encephalitis viruses is unique to the New World. Prevention is accomplished by adequate vector control and vaccination of horses. Pigeons, chickens, pheasants, prairie chickens, ducks, and geese are susceptible and, if infected when suitable vectors are present, may pose a potential source of disease for humans. There is a high mortality in humans infected with these viruses.
  2. CRIMEAN CONGO HEMORRHAGIC FEVER. Nairovirus, Bunyaviridae family. Causes epidemics in Bulgaria, USSR, and sporadic cases in Iraq, Pakistan and East Africa. Associated with tick bites, primarily Hyalomma genus. Also nosocomial outbreaks among hospital personnel.
  3. OMSK HEMORRHAGIC FEVER (rodents)
  4. KYASANUR FOREST DISEASE (monkeys) Togaviridae occurs in bonnet macaques, languors, and rodents in India spread by ticks lab workers in the US affected while working with infected primates (no vector) NHP exhibit fever, vomiting, diarrhea, epistaxis, and death. In humans, there is a sudden onset of fever which may be biphasic, with headache, generalized pains, prostration, conjunctivitis, diarrhea, and vomiting. Vesicles occur on the soft palate. Hemorrhagic manifestations may follow. Case fatality rate is 2-50%. Diagnosed thru serology or viral isolation. Treatment is supportive. Convalescent plasma with a high neutralizing antibody titer has been reported to be useful. Prevention is thru tick control.
  5. CHIKUNGUNYA: Maintained by vervets and baboons in Southern Africa. Transmitted by biting flies and mosquitoes
  6. RIFT VALLEY FEVER (Enzootic Hepatitis)
  • AGENT:

Phlebovirus, Bunyaviridae

  • RESERVOIR AND INCIDENCE

endemic in Africa and Egypt. Affects nonhuman primates, and man. Cattle, bats, and sheep may be reservoir hosts.

  • TRANSMISSION:

mosquito vector highly contagious among laboratory workers where transmission does not require vector.

  • DISEASE IN ANIMALS:

Rapid death after fever occurs in lambs. In cattle abortion and diarrhea occur. Liver lesions predominate histologically, with areas of necrosis. Widespread hemorrhages occur.

  • DISEASE IN MAN:

Fever (which may be biphasic) has a sudden onset, with severe headache, muscle and joint pains and photophobia. In a small proportion of cases there are hemorrhages, liver necrosis, encephalitis and retinitis.

  • DIAGNOSIS:

Serology and virus isolation.

  • PREVENTION/CONTROL:

Control mosquitos. Precaution in handling necropsy specimens. Arbovirus where the natural cycle of transmission DOES involve man:


ST. LOUIS ENCEPHALITIS

SYNONYMS: Type C lethargic encephalitis.

  • ETIOLOGY:

RNA virus belonging to the genus Flavivirus (formerly group B of the arboviruses) of the family Togaviridae; forms part of the virus complex including Murray Valley, West Nile, and Japanese B encephalitides. There are indications that strains of the virus isolated in different zones differ in their capacity to produce viremia in birds, virulence in 3-week-old mice, and neurovirulence in rhesus monkeys. By the technique of nucleotide mapping, considerable genetic variation among the strains of the virus was demonstrated, and it was proposed to name these geographic variants as topotypes.

  • GEOGRAPHIC DISTRIBUTION

The agent is distributed from Argentina to Canada. The disease is unknown outside the Americas.

  • THE DISEASE IN MAN:

The clinical infection presents a wide spectrum, from an undifferentiated febrile disease similar to influenza to severe encephalitis. Three syndromes can be distinguished: febrile disease, aseptic meningitis, and encephalitis. The febrile syndrome usually has a benign course, with fever and intense cephalalgia lasting several days, followed by complete recovery. The aseptic meningitis has a sudden onset, with fever, stiffness of the neck, and positive Kemig and Brudzinski signs, but without neurologic dysfunction. Pleocytosis is common. The disease characterized by encephalitis also begins suddenly, with fever and one or more signs of brain inflammation, such as personality changes, confusion. delirium, lethargy, paresis, convulsions, and others. The encephalitis syndrome is more frequent in elderly persons; its frequency increases from 56% in patients up to 20 years old to 87% in those over 60. Convalescence in these cases lasts several weeks. The incubation period is estimated at 4 to 21 days. In the United States, the case fatality rate in 2,261 confirmed clinical cases between 1955 and 1968 was 5 to 10%. The majority of deaths were in persons over 50 years old, among whom the fatality rate can be as high as 30% or more. During the 1962 epidemic in Tampa Bay, Florida, the highest fatality rate (36.3%) was recorded in patients 65 years of age and older in Pinellas County, where a large number of retired persons live. In that county the general mortality rate was 22.2%, while the rate was 9.8% in the remaining three counties in that area. In Central and South America, most of the small number of recorded patients did not exhibit impairment of the central nervous system.

  • THE DISEASE IN ANIMALS:

The infection is subclinical in animals. Experimental peripheral inoculation of the virus produces viremia without clinical symptoms in domestic and wild fowl and in various species of insectivorous bats. When the disease occurs in man, antibodies for SLE are generally found in horses and in some other mammals. In contrast to western, eastern, and Venezuelan equine encephalitides, St. Louis encephalitis does not cause clinical illness in equines. Some equines inoculated experimentally develop viremia.

  • SOURCE OF INFECTION AND MODE OF TRANSMISSION:

The basic cycle of the infection involves wild birds and omithophilic mosquitoes. Culex salinarius, from which the SLE virus has been isolated, could be the vector in the wild enzootic cycle. In the United States, two different epidemiologic situations are known, depending on the habits of the primary vector and other ecologic conditions. West of the Rocky Mountains the disease is rural and sporadic because the vector, C. tarsalis, is sparse and the widely scattered human population has a high rate of subclinical infection, protecting it against reinfection. Though the vector and birds reach high concentrations in areas flooded by irrigation water, human cases are not numerous for the reasons given. In the south-central and north-central states of the country, by contrast, the disease is urban-suburban in character, primarily because the vectors are the peridomestic and domestic mosquitoes C. quinquefasciatus and C. pipiens. These vectors proliferate where water contaminated with organic wastes collects, that is, in poorer urban and suburban areas deficient in environmental sanitation. The same conditions favor the proliferation of sparrows, pigeons, and other birds that feed among household wastes. Peridomestic birds and domestic fowl serve as amplifiers of the virus: that fact, together with the increased density of the human population, creates the conditions necessary for epidemics. During the 1964 epidemic in Houston, the virus was isolated from geese, domestic pigeons, and various other species of birds. In addition, antibodies were found in 20% of the birds, especially house sparrows (Passer domesticus), and in almost all poultry examined. How the virus gets into urban areas is not yet established, but it is suspected that migratory wild birds may introduce it. As is true for many other arboviruses, the mechanism that allows the virus to overwinter in temperate climates is not fully known. The virus has been isolated from hibernating adult female C. pipiens, which indicates that the virus can persist in the vector during the winter in temperate climates. It also has been proven experimentally that low-level transovarial transmission occurs in C. pipiens.

  • ROLE OF ANIMALS IN THE EPIDEMIOLOGY OF THE DISEASE:

Man is an accidental host of the virus and does not play any role in the natural maintenance cycle. The basic reservoir is wild birds and perhaps the vector mosquitoes; poultry and peridomestic and domestic birds act as amplifiers of the virus, which circulates from one host to another by means of mosquitoes. Wild and domestic mammals are not thought to play a role in the virus cycle because their viremia is low-level and transitory and virus strains isolated from them possess low virulence. In Panama, sloths inoculated with the SLE virus developed prolonged high-titer viremia, but the role of these animals under natural conditions has not been determined. Likewise, bats may be involved in the virus's overwintering within enzootic foci in temperate climates, as well as in its dissemination to epizootic foci, a subject that requires further study.

  • DIAGNOSIS:

SLE may be confused clinically with other febrile diseases or with encephalitides and aseptic meningitis caused by different agents. Laboratory confirmation is essential. Laboratory diagnosis is based primarily on serology. only on a few occasions has it been possible to isolate the etiologic agent from the blood of viremic patients. Most successful isolations have been made from the brain of patients who died a short time after contracting the disease.

  • DIAGNOSIS:

is based on demonstrating serologic conversion in the patient by comparing titers of serum samples taken during the acute phase with those taken during convalescence. The most widely used tests are complement fixation, neutralization (which is the most specific), and hemagglutination inhibition. Antibodies can be detected during the first week of the disease by the hemagglutination inhibition and neutralization tests, while the complement fixation antibodies appear during the second or third week. In Latin American and Caribbean countries, where infections due to several flaviviruses occur, tests must include all of the other viruses of the group known to be present in the area.

  • TREATMENT:

Vigorous supportive therapy. Such measures include reduction of intracranial pressure (mannitol), monitoring of intraventricular pressure, the control of convulsions, maintenance of the airway, administration of oxygen, and attention to adequate nutrition during periods of prolonged coma. No antiviral agent is effective for arboviral encephalitis.

  • CONTROL:

The only preventive measure available is control of the vector. Programs of epidemiologic surveillance and vector control have given satisfactory results in California against C. tarsalis, in Florida against C. nigripalpus, and in Texas against C. quinquefasciatus. An effective vaccine is not yet available.


YELLOW FEVER

(Black Vomit)

  • AGENT:

RNA virus, Family Togaviridae, (Group B Arbovirus)

  • RESERVOIR AND INCIDENCE

all primates susceptible; major Public Health problem in Central and S. America and Africa.

  • TRANSMISSION:

o mosquito vector: Aedes and Hemagogues

  • DISEASE IN NONHUMAN PRIMATES:

There is high fever and vomiting, with jaundice, oliguria, and generalized hemorrhages. Microglobular fatty degeneration of liver cells occurs with disruption of the hepatic lobule and necrosis of midzonal liver cells, producing so called "Councilman" bodies. Degeneration and necrosis of the kidney tubules occurs. There are hemorrhages in tissues.

  • DISEASE IN MAN:

Most cases have fever, severe headache and backache, jaundice and albuminuria, followed by full recovery with a week, but in severe cases there is a second episode of fever, prostration, jaundice, renal failure and generalized hemorrhages. Microglobular fatty degeneration of liver cells occurs with disruption of the hepatic lobule and necrosis of midzonal liver cells, producing so called "Councilman" bodies. Degeneration and necrosis of the kidney tubules occurs. There are hemorrhages in tissues. The case fatality rate among indigenous populations of endemic regions is <5%, but may exceed 50% among nonindigenous groups and in epidemics.

  • DIAGNOSIS:

Virus isolation or serology.

  • TREATMENT:

Consists of limiting food to high-carbohydrate, high-protein liquids, IV glucose and saline, analgesics and sedatives, and saline enemas.

  • PREVENTION/CONTROL:

Monkeys should originate from a yellow fever free area, or be maintained in a double-screened mosquito-proof enclosure, or have been immunized against yellow fever. For humans, mosquito control, vaccination, and adherence to PHS quarantine standards.


HANTAVIRUS PULMONARY SYNDROME

INTRODUCTION: On May 14, 1993, the New Mexico Department of Health was notified of 2 persons who had died within 5 days of each other. Their illnesses were characterized by abrupt onset of fever, myalgia, headache, and cough, followed by the rapid development of respiratory failure. Tests for Yersinia pestis and other bacterial and viral pathogens were negative. After additional persons who had recently died following a similar clinical course were reported by the Indian Health Service, the health services of Arizona, Colorado and Utah were contacted to seek other possible cases. Blood and tissue specimens were sent to the Centers for Disease Control and Prevention (CDC). The results were negative except for signals for the Puumala hantavirus. Relying on molecular and immunological research performed by the Army, the National Institutes of Health, and the CDC itself, by June 9th, the CDC was able to prove that a new hantavirus was the culprit (1). As of November 5th, laboratory evidence of acute hantavirus infection had been confirmed in 42 persons. Twenty-six (62%) of these persons have died. Most cases were in the Southwest but some have been reported as far afield as North Dakota and California (1). This paper presents a brief overview of hantavirus infections with primary focus on Hantavirus Pulmonary Syndrome (HPS) and recommended precautions to reduce the risk of accidental exposure. Other detailed reviews are available elsewhere (2-7).

  • AGENT:

Isolation of the first recognized hantavirus (Hantaan virus) was reported from the Republic of Korea in 1978. The genus Hantavirus is a member of the family Bunyaviridae. Hantaviruses are further divided into genotypes. Representative viruses in each genotype are the Hantaan virus, the Seoul virus, the Puumala virus, and the Prospect Hill virus. Additional groups exist. Hantaan, Puumala, and Seoul viruses are known human pathogens; Prospect Hill has not been associated with disease. The causative agent of HPS represents a previously unidentified genotype. Since the 1930s, epidemic and sporadic hantavirus-associated disease has been described throughout Eurasia, especially in Scandinavia and Northeastern Asia. In the 1950s, thousands of United Nations military personnel were infected with hantavirus during the Korean conflict; more recently, transmission has been documented among United States military personnel training in the Republic of Korea. Hantaviruses have been isolated from rodents in the United States, and serological studies have documented human infections with hantaviruses. However, acute disease associated with infection by pathogenic hantaviruses has not previously been reported in the Western Hemisphere (8). Previously called the Four Corners Virus and Muerto Canyon Virus, the causative genotype for HPS is now called Sin Nombre Virus.

  • DISEASE IN HUMANS:

The clinical manifestations previously associated with hantavirus infections have been characterized by hemorrhagic features and by renal involvement. In HPS, however, onset of illness has been characterized by a prodrome consisting of fever, myalgia, and variable respiratory symptoms followed by the abrupt onset of acute respiratory distress. Other symptoms reported during the early phase of illness have included headache and gastrointestinal complaints. Hemoconcentration and thrombocytopenia have developed in a majority of the cases. The hospital course has been characterized by bilateral pulmonary infiltration, fever, hypoxia, and hypotension; recovery in survivors has been without sequelae. It is important to note that no defined set of symptoms and signs reliably distinguishes HPS from other forms of noncardiogenic pulmonary edema or adult respiratory distress syndrome (9). Postmortem examination has routinely revealed serous pleural effusions and heavy edematous lungs. Microscopic findings have included interstitial infiltrates of mononuclear cells in the alveolar septa, congestion, septal and alveolar edema with or without mononuclear cell exudate, focal hyaline membranes, and occasional alveolar hemorrhage. Large mononuclear cells with the appearance of immunoblasts have been found in red and periarteriolar white pulp of the spleen, hepatic portal triads, and other sites. Hantavirus antigens, localized primarily in endothelial cells, have been detected in most organs, with marked accumulations in the lungs (9). The incubation period for the known pathogenic hantaviruses, although highly variable, generally range from 2 to 4 weeks (8). Based on reported cases, the incubation period for HPS appears to be one to three weeks (10).

  • RESERVOIRS:

Rodents are the primary reservoir hosts with each hantavirus appearing to have a preferential rodent host. The epidemiological characteristics of outbreaks of human disease and the severity for the infection are determined mainly by the rodent host. Available data strongly supports the deer mouse (Peromyscus maniculatus) as the primary reservoir of the newly recognized hantavirus (11). Serologic evidence of infection has also been found in pi¤on mice (P. truei) and the brush mice (P. boylii). Other rodent species that have tested positive so far include the house mouse (Mus musculus), the harvest mouse (Reithrodontomys sp.), the rock squirrel (Spermophalus variegatus), the white-throated wood rat (Neotoma albigula), and the western chipmunk (Tamias spp.). P. maniculatus is highly adaptable and is found in different habitats, including human residences in rural and semirural areas, but generally not in urban centers (12). The wood mouse or striped field mouse (Apodemus sp.) associated hantaviruses also cause severe human disease with mortality rates between 3 and 7%. Rattus associated disease is less severe and asymptomatic infections may be more common. The most benign form of hantaviral disease (HVD), also called Nephropathia epidemica and first described in Scandinavia, is caused by a hantavirus that infects voles (Clethrionomys species). Infected voles and human disease occur throughout western Europe (13). The hantaviruses have been identified in other animals. At the International Symposium on Hemorrhagic Fever with Renal Syndrome (HFRS), Leningrad, 5-10 May 1991, the presence of hantaviral antigen was reported in 13 species of birds in eastern parts of the former U.S.S.R. (13). The CDC is also investigating whether other animals, particularly those that prey on rodents, may carry the virus. The impetus for this research is a 1987 study suggesting that cats, which tested positive for two other hantaviruses-the Hantaan and Seoul types-may help transmit the virus to humans in China. As for the HPS virus, so far CDC scientists have identified one infected nonrodent species (aside from humans): the desert cottontail (Sylvilagus auduboni). But virologists think most nonrodents are "dead-end" hosts that shed little virus and are unlikely to infect people (14) Rats, which were a reservoir of hantavirus, have been responsible for several outbreaks of HVD among animal caretakers and laboratory workers at research institutions in Korea, China, the former Soviet Union, Japan, Scandinavia, the U.K., France, the Netherlands and Belgium (13). Transmission of Hantavirus from laboratory reared mice and rats has not been documented in the United States.

  • TRANSMISSION:

Susceptibility of rodents may vary depending on the combination of rodent species and virus strains; however, Hantaviruses do not cause apparent illness in their reservoir hosts (15). In rodents, the virus is detected primarily in the lung and kidney, where it persists despite the presence of serum antibodies. Infected rodents shed large quantities of virus in saliva, urine, and feces for many weeks, but the duration and period of maximum infectivity are unknown. Although the main route of transmission is aerosolization, the demonstrated presence of infectious virus in saliva of infected rodents and the marked sensitivity of these animals to hantaviruses following inoculation suggests that biting may also be an important mode of transmission among rodents (12). Arthropod vectors are not known to have a role in the transmission of hantaviruses. Domestic animals may bring infected rodents into contact with humans (12). Human infection may occur when infective saliva or excreta are inhaled as aerosols produced directly from the animal. Transmission may also occur when dried materials contaminated by rodent excreta are disturbed, directly introduced into broken skin, introduced onto the conjunctivae, or, possibly, ingested in contaminated food or water. Persons have also become infected after being bitten by rodents (12). Person-to-person transmission has not been associated with any of the previously identified hantaviruses nor with the recent outbreak in the Southwest (16). In the current epidemic, known hantavirus infections of humans have occurred primarily in adults and are associated with domestic, occupational, or leisure activities that bring humans into contact with infected rodents, usually in a rural setting. Cases have been epidemiologically associated with the following situations: Planting or harvesting field crops Occupying previously vacant cabins or other dwellings Cleaning barns and other outbuildings Disturbing rodent-infested areas while hiking or camping Inhabiting dwellings with indoor rodent populations Residing in or visiting areas in which the rodent population has shown an increase in density (12). In Europe, isolation of hantaviruses from immunocytomas and ascites tumors has highlighted additional risks from working with persistently infected rodents. Tumors, passaged over the years in hantavirus-infected laboratory rats, transfer the virus when implanted in hantavirus-free rats. Since, in rodents, hantaviruses are not transmitted vertically but horizontally, the use of caesarian section and foster mother techniques have been recommended for laboratories breeding rodent colonies. Before implantation, tumors should be checked for the presence of the hantaviruses (this precaution should be followed by laboratory workers in the U.S. importing tumors, organs, or live rodents from hantavirus endemic areas) (12).

  • DIAGNOSIS:

The CDC in consultation with the Council of State and Territorial Epidemiologists has developed screening criteria for HPS (9). Cases meeting the screening criteria should be reported to the CDC through state health departments. These criteria are: Potential case-patients must have one of the following: a febrile illness (temperature ò101 oF [ò38.3 oC]) occurring in a previously healthy person characterized by unexplained adult respiratory distress syndrome, or bilateral interstitial pulmonary infiltrates developing within 1 week of hospitalization with respiratory compromise requiring supplemental oxygen, OR an unexplained respiratory illness resulting in death in conjunction with an autopsy examination demonstrating noncardiogenic pulmonary edema without an identifiable specific cause of death. Potential case-patients are to be excluded if they have any of the following: a predisposing underlying medical condition (e.g., severe underlying pulmonary disease, solid tumors or hematologic malignancies, congenital or acquired immunodeficiency disorders, or medical conditions [e.g., rheumatoid arthritis or organ transplant recipients] requiring immunosuppressive drug therapy [e.g., steroids or cytotoxic chemotherapy]). an acute illness that provides a likely explanation for the respiratory illness (e.g., recent major trauma, burn, or surgery; recent seizures or history of aspiration; bacterial sepsis; another respiratory disorder such as respiratory syncytial virus in young children; influenza; or legionella pneumonia). Confirmed case-patients must have the following: at least one specimen (i.e., serum and/or tissue) available for laboratory testing for evidence of hantavirus infection. AND in a patient with a compatible clinical illness, diagnosis is confirmed when any of the following criteria are met: IgM antibodies to hantavirus antigens, fourfold or greater increase in immunoglobulin G titers to hantavirus antigens in paired serum specimens, a positive immunohistochemical stain for hantavirus antigen in tissues, or positive polymerase chain reaction (PCR) for hantavirus ribonucleic acid. Currently, diagnosis of the HPS strain of Hantavirus in animals is in its infancy. IFA based tests offered by national research laboratories may be used for screening; however, false negatives can occur depending on the antigen used. PCR remains the method of choice for strain identification. Presently, one laboratory (Rockefeller University Laboratory Animal Research Center, [212]327-8522) offers this service with more, hopefully, coming on line in the future (17).

  • TREATMENT:

Supportive care and meticulous monitoring of vital signs and fluid balance are the basis for therapy. Severe hypoxia and overhydration should be avoided or prevented. Pressors or cardiotonic drugs should be employed to maintain perfusion without excessive fluid administration (9). In one controlled study involving HFRS, intravenous administration of the antiviral drug ribavirin was effective in treating severe cases of hantavirus infection when administered early in the course of illness (8). The effectiveness of using ribavirin to treat HPS has not been established, yet.

  • PREVENTION AND CONTROL:

Hantaviruses have lipid envelopes that are susceptible to most disinfectants (e.g., dilute hypochlorite solutions, detergents, ethyl alcohol (70%), or most general-purpose household disinfectants). How long these viruses survive after being shed in the environment is uncertain (12). The reservoir hosts of the hantavirus in the southwestern United States also act as hosts for the bacterium Yersinia pestis, the etiology agent of plague. Although fleas and other ectoparasites are not known to play a role in hantavirus epidemiology, rodent fleas transmit plague. Control of rodents without concurrent control of fleas may increase the risk of human plague as the rodent fleas seek an alternative food source. Thus, eradicating the reservoir hosts of hantaviruses is neither feasible nor desirable. Once the virus has been cultured, it might be possible to develop a vaccine against the HPS strain. However, currently, the best available approach for disease control and prevention is risk reduction through environmental hygiene practices that deter rodents from colonizing the home and work environment (12). No restriction of travel to areas affected by this outbreak is considered necessary; however, activities that may disrupt rodent burrows or result in contact with rodents or aerosolization or rodent excreta should be avoided. People practicing universal precautions while processing routine clinical materials (such as blood, urine, and respiratory specimens) are not considered to be at increased risk for hantavirus infection. However, human-acquired infections have occurred among persons who handled infected wild or domesticated rodents. Therefore, exposure that may result in propagation of hantaviruses should be conducted in a special facility (biosafety level 3) (8). Recommendations for animal facilities housing wild-caught rodents include: Access to rooms should be restricted to only those individuals who have a legitimate need to be in the room. Colony should be serologically screened for the agent. Animals should be housed and handled under standard microisolation techniques. Biological safety cabinets should be used and not laminar flow workbenches. Until the status of the colony can be ascertained, individuals working with the rodent should: a. Obtain a baseline serum sample. The serum should be stored at -20oC. b. Insure that all persons involved are informed of the symptoms of the disease and given detailed guidance on prevention measures. c. Seek immediate medical attention if a febrile or respiratory illness develops within 45 days of the last potential exposure. The attending physician should be informed of the potential occupational risk of hantavirus infection. d. Wear an half-face air-purifying (or negative-pressure) respirator or powered air-purifying respirator with HEPA filter when handling rodents or their cages. Respirators are not considered protective if facial hair interferes with the face seal. Respirators should be fitted by trained personnel in accordance with OSHA standards. e. Wear rubber or plastic gloves when handling rodents or cages. Gloves should be washed and disinfected before removing them. f. Wear dedicated outer garments (disposable, if possible), rubber boots or disposable shoe covers and protective goggles. Personal protective gear should be decontaminated upon removal. If not disposable, they should be laundered on site using hot water and detergent. Machine-dry using a high setting. If no laundry facilities are available, non-disposable items should be immersed in liquid disinfectant until they can be washed. All potentially infective waste material (including respirator filters, bedding, caging, disposable protective garments, and used disposables such as syringes, gauze, etc.) should be placed in autoclavable plastic bags and sterilized. Needles, scalpels, pipettes, and other sharp materials should be placed in puncture proof containers and sterilized. Spread from feral rodents was postulated as the cause of one source of contamination (18). Therefore, facilities and individual rooms should be vermin-proof to prevent accidental egress and ingress of rodents. All openings greater than ¬ inch should be screened or sealed. Carcasses should be placed in a plastic bag and disposed as biohazard waste or incinerated. Since feral rodents may transmit the disease, it is recommended that Hantavirus testing be included in animal health monitoring programs. Why the Current Epidemic? Because of the rodent connection with this disease, medical investigators and public health officials sought ecological information on the deer mouse and other native rodent species. Anecdotal information from residents in the afflicted areas suggested that rodents were exceptionally abundant last winter, and scientists speculated that, if true, the increased potential for rodent-human contact and disease transmission might account for the sudden epidemic. Biologists with the Sevilleta, New Mexico Long-Term Ecological Research (LTER) site have long-term data on rodent communities in the region. At the request of the CDC and the New Mexico Health Department, LTER researchers provided detailed demographic analyses from 1989-1993 for the 22 rodent species inhabiting the area. The LTER data showed tenfold population increases in various Peromyscus species and wood rats (Neotoma spp.) during the spring of 1993. Population increases occurred simultaneously in grasslands, desert-shrublands, and woodlands. Comparisons of the rodent data to the region's climatological data indicated that rodent population dynamics is associated with above-average precipitation during the winter of 1992-93, in turn leading to abundant food sources (19).

References Marshall, E. 1993. Hantavirus outbreak yields to PCR. Science. 262:832-836. LeDuc, J. W., J. E. Childs, and G. E. Glass. 1992. The Hantaviruses, etiologic agents of hemorrhagic fever with renal syndrome. Annu Rev Public Health. 13:79-98. Niklasson, B. S. 1992. Hemorrhagic fever with renal syndrome, virological and epidemiological aspects. Pediatr Nephrol. 6(2):201-204. Cosgriff, T. M. and R. M. Lewis. 1991. Mechanisms of disease in hemorrhagic fever with renal syndrome. Kidney Int Suppl. 35:S72-79. Tkachenko E. A. and H. W. Lee. 1991. Etiology and epidemiology of hemorrhagic fever with renal syndrome. Kidney Int Suppl. 35:S54-61. Beaty, B. J. and C. H. Calisher. 1991. Bunyaviridae--natural history. Curr Top Microbiol Immunol. 169:27-78. Gonzalez-Scarano, F., M. J. Endres, and N. Nathanson. 1991. Bunyaviridae: Pathogenesis. Curr Top Microbiol Immunol. 169:217-249. 1993. Emerging infectious diseases. Outbreak of acute illness. Wkly Epidemiol Rec. 68(25):186-8. 1993. Emerging infectious diseases. Update: Hantavirus Disease. MMWR. 42(29, 31, and 42). Sands, L. 1993. Guidelines for

  • DIAGNOSIS:

and treatment of unexplained adult respiratory distress syndrome. Arizona Department of Health Services.

Nichol, S. T., C. F. Spiropoulou, S. Morzunov, et al. 1993. Genetic identification of a Hantavirus associated with an outbreak of acute respiratory illness. Science. 262:914-917. 1993. Hantavirus infection-Southwestern United States: interim recommendations for risk reduction. MMWR. 42(RR-11). McKenna, P., G. VanDerGroen, G. Hoofd, et al. 1992. Eradication of hantavirus infection among laboratory rats by application of caesarian section and a foster mother techniques. J Infect. 25:181-190. Stone, R. 1993. The mouse-pion nut connection. Science. 262:833. Kawamura, K., X. K. Zhang, J. Arikawa, et al. 1991. Susceptibility of laboratory and wild rodents to Rattus or Apodemus-type hantaviruses. Acta Virol. 35:54-63. Hughes, J. M., C. J. Peters, M. L. Cohen, et al. 1993. Hantavirus pulmonary syndrome: an emerging infectious disease. Science. 262:850-851. Morse, S. 1994. Personal communication. Wong, T. W., Y. C. Chan, E. H. Yap, et al. 1988. Serological evidence of Hantavirus infection in laboratory rats and personnel. Int J Epidemiol. 17(4):887-890. Dybas, C. 1993. NSF-funded researchers find rodent population explosion may be behind hantavirus epidemic in southwest. NSF Bulletin #93-59.



HEMORRHAGIC FEVER WITH RENAL SYNDROME [HFRS]

(Korean Hemorrhagic Fever, Nephropathia Epidemica, Epidemic Hemorrhagic Fever, Hemorrhagic Nephrosonephritis)

  • AGENT:

Bunyaviridae family, Hantaan virus genus

  • RESERVOIR AND INCIDENCE

Recognized originally in troops serving in the Korean war. Named Hantaan after river in endemic area of Korea. Isolated in 1978 in Apodemus agrarius, then adapted to tissue culture and laboratory rats. Serologic mapping indicates that Hantaviruses have infected large numbers of people in the region from Japan across central and north Asia to the Scandinavian Peninsula, and southward in Europe to the Balkans. Other Hantaviruses have been identified in urban rats captured in major Asian and Western cities, including the USA and Brazil. Hosts of hantavirus are wild Rodents Several antigenic subtypes exist, each associated with a single rodent species: Apodemus species - Striped field mouse - A. agrarius (Korea) associated with KHF. Domestic Rattus norvegicus and Rattus rattus in Korea and Domestic Rattus norvegicus in the U.S. had virus similar to prototype Hantaan virus yet distinct from it. Microtus pennsylvanicus: the meadow vole. Reservoir for Prospect Hill virus, most recently isolated Hantavirus. Isolated at NIH and named for Prospect Hill in Frederick, MD where the vole was captured that yielded the first isolate. No human disease associated with this so far but antibody has been identified among mammalogists in the U.S.(1982). Cletheronomys glareolus: the bank vole. Reservoir for Puumala virus, cause of Nephropathia epidemica (NE), a mild form of HFRS found in Scandinavia, Western Soviet Union and much of Europe.

  • TRANSMISSION:

Aerosol transmission from rodent excreta is presumed. Virus is present in urine, feces and saliva of persistently infected asymptomatic rodents; highest virus concentration is found in the lungs. Human to human transmission does not occur.

  • DISEASE IN RODENTS:

Chronic, ASYMPTOMATIC infection. Following infection, rodent is viremic for about 1 week when virus is disseminated throughout the body. After viremia antigen is usually abundant in the lungs, spleen and kidneys. Antibody is produced and persists but does not diminish the abundance of antigen expressed in organs.

  • DISEASE IN MAN:

Symptoms begin with the sudden onset of fever which lasts 1-2 weeks, accompanied by prostration, anorexia, generalized pains, conjunctivitis, proteinuria and hypotension, possibly followed by hemorrhages and hematuria with renal failure. Case fatality rate is 7%.

  • DIAGNOSIS:

Serology using indirect immunofluorescence or ELISA.

  • TREATMENT:

IV ribavirin

  • PREVENTION/CONTROL:

Although most US commercial animal breeders have eliminated these viruses through barrier breeding and caesarean derivation, small suppliers and producers of select inbred strains may be at risk of infection. Be aware of potentially infected animals when receiving shipments from Japan, Belgium or other countries which may have the agent present in lab. animals (where virus-free certification cannot be provided). Animals should be serologically tested in advance of shipment. Test all rodent tissues, tumors, cell lines received from source that cannot provide virus free certification. Education and awareness of potential problem. Exclude rodents from housing and other buildings in endemic areas.


OTHER HEMORRHAGIC FEVERS

(Arenaviruses) All have natural persistent infection in rodents with humans being accidental hosts. Route of transmission to humans is generally thought to occur thru contamination of food, water, or air by rodent feces or urine or by inoculation of skin abrasions. Humans are infected primarily through infected rodents invading human habitats. Contact with infected rodent feces has produced disease in laboratory personnel.

  • DIAGNOSIS:

is by serology or virus isolation. Control is to reduce opportunity for exposure to infected rodents. 1. JUNIN VIRUS: produces Argentinian hemorrhagic fever There is an illness of 1-2 weeks with insidious onset of fever, malaise, rigors, fatigue, headache, vomiting, constipation or diarrhea, conjunctival congestion, retro-orbital pain, epistaxis, petechial hemorrhages beneath skin, palate and gums. Edema of the upper body is possible. In severe cases hematemesis and melena, encephalopathy, bradycardia and hypertension occur. Case fatality rate 5-30%. Several hundred cases reported each year in Argentina. Associated with at least 3 different cricetine rodent species in Argentina 2. MACHUPO VIRUS: produces Bolivian Hemorrhagic fever Signs and case fatality rate like Junin virus. Case #s have been decreasing rapidly since initiation of rodent control programs in 1975. Associated with Calomys callosus (Bolivia). 3. LASSA FEVER: Serologically related to Lymphocytic Choriomeningitis, Machupo, and Junin virus. Fever has insidious onset over 2-3 days and may persist for up to 4 weeks, with malaise, headache and generalized aching and sore throat. Vomiting and diarrhea, possibly edema of face and neck, lymphadenopathy with hemorrhages and renal failure occurs in the second week. The prostration is out of proportion to fever. Often there is a maculopapular rash. Occurs in large areas in West Africa. Documented man to man transmission. Found in common rodent Mastomys natalensis, multimammate rat (West Africa).


LYMPHOCYTIC CHORIOMENINGITIS - LCM

  • AGENT:

Arenavirus Of many latent viruses present in mice, only LCM naturally infects humans. LCM can easily be transmitted from animals to humans. Isolated by Armstrong and Lillie during investigation of a St. Louis Encephalitis outbreak in 1933.

  • RESERVOIR AND INCIDENCE

Worldwide in wild mice (M. musculus). This disease is principally confined to the eastern seaboard and northeastern states in the U.S. Wild mice infect the lab mouse. Mouse and hamster are the only species in which long term, asymptomatic infection is known to exist. *LCM virus is present in experimental mouse tumors which is a second source of infection for humans. This was first recognized in a transplantable leukemia of C58 mice. The disease can also be transmitted to laboratory animals via inoculation of infected tissue culture cells. The infection also occurs in guinea pigs, rabbits, rats, canines, swine, and primates.

  • TRANSMISSION:

Infection in mice is maintained by congenital infection followed by lifelong carriage and excretion of virus in saliva, urine, and feces. Human infections are probably from contaminated food and dust, the handling of dead mice, and mouse bites. Bloodsucking arthropod vectors such as ticks, lice, and mosquitos may transmit the disease. Person to person transmission does not occur.

  • DISEASE IN ANIMALS:

The clinical signs of LCM depend on the host's resistance and age when infected, although the various categories of the disease are not always clearly delineated. Animals infected in utero or during the first 48 hours postpartum may develop a transient viremia but recover completely within a few weeks. Other animals similarly infected may develop a persistent tolerant infection (PTI) that continues asymptomatically for 6 or more months. Animals infected after the first few days, when the virus will be recognized as foreign, often overcome the infection completely, but an acute, usually fatal syndrome can develop. Signs of acute infection in mice continue for 1-2 weeks and include decreased growth, rough hair coat, hunched posture, blepharitis, weakness, photophobia, tremors, and convulsions. The terminal stage of the PTI, which occurs over several weeks to 5 to 12 month old mice, is characterized by weight loss, blepharitis, and impaired reproductive performance and runted litters. The important necropsy signs are microscopic. Visceral organs, including the liver, kidneys, lungs, pancreas, blood vessels, and meninges, are infiltrated by lymphocytes. A glomerulonephritis of probable immune complex origin is a characteristic feature of terminal PTI.

  • DISEASE IN MAN:

The features may include influenza-like illness for up to 2 weeks, possibly with orchitis. Sometimes meningitis, paralysis and coma follow. Joint pains occur during convalescence.

  • DIAGNOSIS:

CF or virus isolation.

  • PREVENTION/CONTROL:

serologic monitoring Infection can be eradicated by cesarean derivation prevent wild mice from entering facilities control ectoparasites and roaches Restrict flow of traffic into and out of LCM infected colonies Protective clothing and proper care when handling infected animals or tissues. Basic hygienic practices Screen tissue culture cell lines and murine tumor lines and animals periodic serologic testing of high risk personnel


CALIFORNIA ENCEPHALITIS/LA CROSSE ENCEPHALITIS

(USA) (Tahyna virus [Europe]) A mild, febrile, viral disease which occasionally causes severe encephalitis. It is transmitted by mosquitoes from small wild mammals, mainly in summer, to persons frequenting woodland areas of the USA and Canada, and certain European countries such as Yugoslavia and the USSR. The causative agents are the California encephalitis group of viruses (Bunyaviridae). There is no vaccine.

  • RESERVOIR AND MODE OF TRANSMISSION:

The virus cycles amongst small wild animals (e. chipmunks, squirrels, rabbits and hares) and a variety of mosquito species. The infection can be maintained independently over several years by transovarial transmission in the mosquito. Humans are accidental hosts infected by mosquito bite during occupational or recreational activities in wooded areas. Accidental infections from laboratory accidents have occurred.

  • INCUBATION PERIOD:

Humans. 5-15 days. Animals. Unknown.

  • CLINICAL FEATURES:

Humans. Symptoms lasting about 5-10 days range from fever and headache with nausea and vomiting to fits and signs of aseptic meningitis, encephalitis and neurological sequelae. Animals. Unknown but assumed subclinical.

  • PATHOLOGY:

Humans. Encephalitis. Animals. Unknown.

  • DIAGNOSIS:

Humans. The virus may sometimes be isolated from blood or rarely, from cerebrospinal fluid. Serologic tests of blood or cerebrospinal fluid may be diagnostic in specific types of encephalitis (by demonstrating virus-specific IgM or a fourfold change in complement-fixing or neutralizing antibodies). Animals. Impracticable.

  • PROGNOSIS:

Humans. In humans, fatality is rare but neurological defects may persist. Animals. Thought to be subclinical.

  • PREVENTION:

Humans. Prevent mosquito bites. Control the mosquito vector. Apply laboratory safety procedures. Animals. Impracticable.

  • TREATMENT:

Humans. Vigorous symptomatic therapy. Such measures include reduction of intracranial pressure (Mannitol), monitoring of intraventricular pressure, the control of convulsions, maintenance of the airway, administration of oxygen, and attention to adequate nutrition during periods of prolonged coma. Animals. Not applicable.

  • LEGISLATION:

Humans. Acute encephalitis is notifiable in many countries, including the USA and the UK. Animals.None.


MARBURG VIRUS

(African Hemorrhagic Fever, Green or Vervet Monkey Disease)

  • AGENT:

Agent is classified as a Filovirus. It is an RNA virus, superficially resembling rhabdoviruses but has bizarre branching and filamentous or tubular forms shared with no other known virus group on EM.

  • RESERVOIR AND INCIDENCE

An acute highly fatal disease first described in Marburg, Germany in 1967. Brought to Marburg in a shipment of infected African Green Monkeys from Uganda. 31 people were affected and 7 died in 1967. Exposure to tissue and blood from African Green monkeys (Cercopithecus aethiops) or secondary contact with infected humans led to the disease. No disease occurred in people who handled only intact animals or those who wore gloves and protective clothing when handling tissues. A second outbreak was reported in Africa in 1975 involving three people with no verified contact with monkeys. Third and fourth outbreaks in Kenya 1980 and 1987. Natural reservoir is unknown. Monkeys thought to be accidental hosts along with man. Antibodies have been found in African Green monkeys, baboons, and chimpanzees. 100% fatal in experimentally infected African Green Monkeys, Rhesus, squirrel monkeys, guinea pigs, and hamsters.

  • TRANSMISSION:

Direct contact with infected blood or tissues or close contact with infected patients. Virus has also been found in semen, saliva, and urine.

  • DISEASE IN NONHUMAN PRIMATES:

No clinical signs occur in green monkeys, but the disease is usually fatal after experimental infection of other primate species. Leukopenia and petechial hemorrhages throughout the body of experimentally infected monkeys, sometimes with GI hemorrhages.

  • DISEASE IN MAN:

5-7 day incubation period. Headache, fever, muscle pain, vomiting, diarrhea, hemorrhagic diathesis, Conjunctivitis, photophobia, skin rash, and jaundice. Leukopenia, thrombocytopenia, proteinuria. Shock and death in 25% of cases. Hemorrhages throughout the body on post mortem examination.

  • DIAGNOSIS:

IFA, ELISA, Western blot, EM, or virus isolation.

  • TREATMENT:

Supportive Possibly immune serum

  • PREVENTION/CONTROL:

Strict quarantine on newly imported, wild-caught primates. Naturally infected monkeys should become ill or die within several weeks. Hygiene, sanitation, and protective clothing Isolation of human patients with prevention of sexual intercourse until semen is free of virus.


EBOLA

(African Hemorrhagic Fever)

  • AGENT:

similar to Marburg morphologically similar but antigenically distinct. Both are RNA Filoviruses and have bizarre branching and filamentous or tubular forms shared with no other known virus group.

  • RESERVOIR AND INCIDENCE

Reservoirs unknown. Monkeys are probably accidental hosts, along with humans. First recognized in 1976 in Northern Zaire and Southern Sudan, 500 cases with 350 deaths reported.

  • TRANSMISSION:

Person-to-person transmission occurs by direct contact with infected blood, secretions, organs or semen. Nosocomial infections have been frequent; all Zaire cases acquired from contaminated syringes and needles died.

  • DISEASE IN MAN:

Fever, headache, malaise, followed by chest discomfort, diarrhea, and vomiting. Case fatality rate is 50-90%.

  • DIAGNOSIS:

IFA, ELISA, Western blot, EM, or virus isolation.

  • TREATMENT:

Supportive Possibly immune serum

  • PREVENTION/CONTROL:

Strict quarantine on newly imported, wild-caught primates. Naturally infected monkeys should become ill or die within several weeks. Hygiene ,sanitation, and protective clothing Isolation of human patients with prevention of sexual intercourse until semen is free of virus.



RABIES

(Hydrophobia, Lyssa)

AGENT: - Rhabdovirus which causes an acute almost invariably fatal disease.

RESERVOIR AND INCIDENCE Worldwide distribution (few countries are exceptions) Primary reservoirs vary geographically, eg. foxes, bats, raccoons, skunks, dogs, cats, cattle, and others. In the U.S. and Canada, wildlife rabies most frequently involves skunks, raccoons, and bats. There has been a progressive epizootic among raccoons in the eastern U.S. for over a decade. Involve wild and domestic species. Mostly wild species involved, only 10% of cases are in domestic animals 16 cases have been confirmed in nonhuman primates, including chimpanzees, cebus, cynos, and squirrel monkeys. All source countries of NHP's have endemic rabies. In Germany, Paarman described 25 Avian cases of Rabies involving 11 chickens, 2 geese, 1 duck, 1 sparrow, 1 owl, 1 crow, 3 hawks, 1 kite, 1 magpie, and 4 buzzards with Negri bodies observed in only three. In the U.S., the Great Horned Owl may shed the virus in its droppings after consuming an infected skunk. Rodents and lagomorphs are unlikely to have rabies. In the U.S., rabies has been reported 13 times in ferrets since 1958, most often in pet ferrets acquired from pet shops.

TRANSMISSION: Virus laden saliva via bite, scratch, or abrasion Tissues and fluids in the laboratory Rabid dogs shed virus in saliva 5-7 days before showing signs. Cat does so for only 3 days before signs. Aerosol transmission has been documented in the laboratory and in caves where bats roost (requires a high concentration of suspended viral particles). Animals showing signs of rabies are usually shedding large amounts of virus.

DISEASE IN ANIMALS: Rabid animals of all species exhibit typical signs of CNS disturbance, with minor variations peculiar to carnivores, ruminants, bats, and man. The clinical course, particularly in dogs, can be divided into 3 phases: the prodromal, the excitative, and the paralytic. The term "furious rabies" refers to animals in which the excitative phase is predominant, and "dumb or paralytic rabies" to those in which the excitative phase is extremely short or absent and the disease progresses quickly to the paralytic phase. In any animal, the first sign is a change in behavior, which may be indistinguishable from a GI disorder, injury, foreign body in the mouth, poisoning, or an early infectious disease. Temperature change is not significant, and driveling may or may not be noted. Animals usually stop eating and drinking and may seek solitude. Frequently, the urogenital tract is irritated or stimulated as evidenced by frequent urination, erection in the male, and sexual desire. After the prodromal period of 1-3 days, animals either show signs of paralysis or become vicious. Carnivora, pigs, and occasionally, horses and mules bite other animals or people at the slightest provocation. Cattle butt any moving object. The disease progresses rapidly after the onset of paralysis, and death is virtually certain within 10 days of the first signs. Rabid domestic cats and bobcats attack suddenly, biting and scratching viciously. Rabid foxes frequently invade yards or even houses, attacking dogs and people. The irrationality of behavior that can occur is demonstrated in the fox that attacks a porcupine; finding a fox with porcupine quills can, in most cases, support a

DIAGNOSIS: of rabies. Rabid foxes and skunks are responsible for most pasture cattle losses, and have attacked cattle in barns. The rabid raccoon is characterized by its loss of fear of man, its frequent aggression and incoordination, and its activity during the day, being predominantly a nocturnal animal. In urban areas, they often attack domestic dogs. Bats flying in the daytime are probably rabid.

DISEASE IN MAN: There is usually a history of animal bite. Pain appears at the site of the bite, followed by paresthesias. The skin is quite sensitive to changes of temperature, especially air currents. Attempts at drinking cause extremely painful laryngeal spasm, so that the patient refuses to drink (hydrophobia). The patient is restless and behaves in a peculiar manner. Muscle spasm, laryngospasm, and extreme excitability are present. Convulsions occur. Large amounts of thick tenacious saliva are present.

DIAGNOSIS: Consider Rabies as a possible problem in any wild caught or random-source laboratory animal of unknown vaccination history showing central nervous system signs or symptoms. Virus isolation from body fluid or tissue Fluorescent antibody (FA) staining of tissues, including cornea, frozen skin, mucosal scrapings, as well as brain. Highly specific & rapid. Can now detect different strains (ie skunk vs raccoon origin) via monoclonal antibody analysis which is specific for one antigenic focus on the viral particle. The identifiable strains correlate well with species and geographic distributions observed. This allows identification of source and is an important epidemiologic tool. (5 strains have been isolated from terrestrial animals; 2 skunk, 1 raccoon, 1 gray fox, 1 red fox, More than 5 have been isolated from bats.)

TREATMENT: This very severe illness with an almost universally fatal outcome requires skillful intensive care with attention to the airway, maintenance of oxygenation, and control of seizures.

PREVENTION/CONTROL: Virus is destroyed rapidly at greater than 50 C and survives no more than a few hours at room temperature (Can persist for years in frozen tissues) Vigorous first aid for bite wounds. Consult Health Authority if suspected exposure. Postexposure immunization: Up to 50% of human rabies immune globulin is infiltrated around the wound; the rest is administered IM. Human Diploid Cell Vaccine (HDCV) is given as 5 injections IM at days 0, 3, 7, 14, and 28. Control disease in domestic animals by vaccination and enforced animal control measures. Discourage keeping of wild animals as pets. Discourage the vaccination of wild animal pets for rabies. Vaccination of high risk personnel.


HEPATITIS A

(Infectious hepatitis, Epidemic hepatitis, Epidemic jaundice)

AGENT: Family Picornaviridae, Genus enterovirus

RESERVOIR AND INCIDENCE More than 200 cases of hepatitis A virus infection in humans have been associated with nonhuman primates, principally chimpanzees but also woolly and patas monkeys, Celebes apes, siamangs, and gorillas. Man, and, rarely captive chimpanzees are the reservoir host; less frequently, certain other nonhuman primates. An enzootic focus has been identified in Malaysia, but there is no suggestion of transmission to man.

TRANSMISSION: Although transmission of the virus may occur by contaminated needles, it is usually by the fecal-oral route. Nonhuman primates acquire the disease from man.

DISEASE IN NONHUMAN PRIMATES: Usually do not show clinical signs, although malaise, vomiting, and jaundice have been reported. Disease is detected by elevated liver enzymes or diagnostic liver biopsies. Usually a self-limiting disease.

DISEASE IN MAN: Fever, malaise, anorexia, headache, muscle pain, abdominal discomfort, and jaundice. Liver enzymes are elevated along with LDH, bilirubin, and alkaline phosphatase. Morbidity is variable and mortality is 0.6% Development of antibody confers lifelong immunity.

DIAGNOSIS: RIA or ELISA

TREATMENT: Bed rest, IV fluids if dehydrated.

PREVENTION/CONTROL: Strictly quarantine newly arrived chimps and allow only limited human contact for at least 45-60 days. Protective clothing (gown, gloves, & mask) Routine disinfection of equipment and personal hygiene. Administer immune serum to handlers of newly imported chimpanzees (0.02ml/kg) every 3 mos. or 0.06 ml/kg every 4 months. Vaccine being developed.


MEASLES

(Rubeola, Morbilli)

AGENT: Family Paramyxoviridae, Genus Morbillivirus. The same genus contains the viruses of Canine Distemper and Bovine Rinderpest.

RESERVOIR AND INCIDENCE Man is the only known reservoir. New world monkeys are more resistant than old world monkeys but exhibit high mortality when infected.

TRANSMISSION: Virus is excreted from the mucous membranes of the eye and pharynx and later from the respiratory and urinary tracts. Virus is shed in the prodromal phase and continues through the exanthematous phase. Highly contagious! Can spread from man to monkey, monkey to monkey, man to man, and monkey to man.

DISEASE IN NONHUMAN PRIMATES: Many infections occur subclinically. Rash, fever, facial edema, giant cell pneumonia, conjunctivitis, nasal discharge.

DISEASE IN MAN: Incubation period 10-14 days. Conjunctivitis Koplik spots - bluish white spots on buccal mucosa 2-3 days after onset Leukopenia Rash in mouth, cheeks, neck, chest, and body. Can be complicated by middle ear infection, bronchopneumonia, encephalitis Fetal risk if contracted during pregnancy. The mortality rate in children in the U.S. is 0.2%, but may be as high as 10% in developing countries.

DIAGNOSIS: clinical signs, serology, histopath.

TREATMENT: Bed rest, acetaminophen, saline eye sponges, nose drops. Vitamin A (400,000 IU/day) has been shown to reduce pediatric morbidity and mortality.

PREVENTION/CONTROL: Vaccinate personnel working with nonhuman primates (Live attenuated measles vaccine) if they do not have: 1. a titer to rubeola (HI >1:4 protective) 2. confirmed history of previous vaccination 3. confirmed prior disease Live attenuated measles vaccine can be given to macaques but causes disease and death in marmosets and owl monkeys.


CYTOMEGALOVIRUS DISEASE

Most cytomegalovirus infections in healthy individuals are asymptomatic, with the virus remaining (exact cells of latency are not known). However the virus is isolable from up to 25% of salivary glands, 10% of uterine cervices, and 1% of neonatal urine samples. Seroprevalence increases with age and the number of sexual partners; detectable antibody is present in the serum of most homosexual men. Transmission is sexual, congenital, through or transplantation, and person-to-person (e.g., day care centers). Severe disease occurs primarily in the immunocompromised, especially those with AIDS and transplant patients.

DISEASE IN MAN: A. Classification: There are three recognizable syndromes. 1. Perinatal disease and cytomegalovirus inclusion disease-Intrauterine infection of infants whose mothers had a primary infection during pregnancy results in a neonatal syndrome of jaundice, hepatosplenomegaly, thrombocytopenia, periventricular central nervous system calcifications, mental retardation, motor disability, and purpura.

DIAGNOSIS: confirmed by viruria within the first week after birth or serum IgM antibodies to cytomegalovirus. Hearing deficits occur in over 15% and mental retardation in up to 30%. Neonatally acquired disease may resemble mononucleosis; while it is often asymptomatic, neurologic deficits may ensue later in life. 2. Acute acquired cytomegalovirus infection--This syndrome, akin to EBV-associated infectious mononucleosis, is characterized by fever, malaise, myalgias and arthralgias (but not pharyngitis or respiratory symptoms), atypical lymphocytes, and abnormal liver function tests. Unlike EBV-associated infectious mononucleosis, the heterophil antibody is not found. Transmission can be by sexual contact, by milk, by respiratory droplets (probably) among nursery or day care center attendants, and by (usually massive) transfusions of blood. 3. Disease in immunocompromised hosts-Tissue and bone marrow transplant patients are at increased risk for CMV infection, especially in the first I 00 days after allograft transplantation. HIV-infected patients may have a variety of CMV manifestations. Cytomegalovirus is itself immunosuppressive and may worsen manifestations of HIV infection, including Pneumocystis carinii pneumonia. a. CMV retinitis-Retinitis due to CMV infection occurs primarily in AIDS patients. Screening for visual symptoms may be helpful, but ophthalmologic documentation of neovascular, proliferative lesions ("pizza-pie" retinopathy) is required for diagnosis. b. Gastrointestinal and hepatobiliary CMV-Serious gastrointestinal CMV disease occurs in AIDS and after organ transplantation, cancer chemotherapy, or steroid therapy. Esophagitis presents with odynophagia; small bowel disease may mimic inflammatory bowel disease or may present as ulceration or perforation. Colonic CMV disease causes diarrhea, hematochezia. abdominal pain, fever, and weight loss. Pancreatitis, when not due to pentamidine, didanosine, or (less often) zalcitabine is often due to cytomegalovirus; hepatobiliary involvement often includes other pathogens, including Cryptosporidium. Diagnosis is by mucosal biopsy that shows characteristic CMV histopathologic findings of intranuclear ("owl's eye") and intracytoplasmic inclusions. c. Pulmonary CMV-Pulmonary CMV infection occurs in about 15% of bone marrow transplant recipients: the mortality rate is 80-90% in this group. CMV seronegative blood products should be used in seronegative recipients of seronegative transplants. High-titer CMV immunoglobulins may be effective in preventing CMV pneumonia in the seronegative recipients. d. Neurologic CMV-Polyradiculopathy and encephalitis have been reported but are not common. When they do occur, there is often concomitant retinitis, which may be subclinical. Prolonged ganciclovir may be helpful, and treatment should be continued indefinitely.

DIAGNOSIS: Cytomegalovirus is isolable from urine, cervical secretions, semen, saliva, blood, and other tissues, but virus isolation is most useful when combined with pathologic findings, including large cells with intranuclear and intracytoplasmic inclusions that resemble owl's eyes: cultures alone are of little use in diagnosing AIDS-related cytomegalovirus infections. Retinitis among aids patients is diagnosed clinically. The acute mononucleosis syndrome is associated with a lymphocytosis, often 2 weeks after the fever. Serologic tests (IFA and the anticomplement immunofluorescent antibody [ACIF]) are useful primarily in seroepidemiologic studies. In AIDS patients, titers may be depressed, and seroconversions are seldom documented, with most seroconversions having occurred in the past. Antigen detection by virus technology (including the polymerase chain reaction technique) must be interpreted in the context of clinical and pathologic findings.

TREATMENT: Two antiviral agents with efficacy against cytomegalovirus are ganciclovir, given in a dosage of 5 mg/kg IV every 12 hours for 14-21 days (maintenance: 5-7 mg/kg/day for 5 days each week, with dose reduction for renal impairment); and foscarnet, given as a loading dose of 20 mg/kg IV and then 60 mg/kg every 8 hours over 2 weeks (maintenance: 120 mg/kg/day). The induction phase is essential for AIDS patients with cytomegalovirus disease involving critical parts of the retina; for less critical areas, maintenance therapy can be used from the outset. Both agents are effective in preventing progression of retinitis, and ganciclovir is useful in cytomegalovirus colitis; treatment is usually lifelong in patients with AIDS. Complications include neutropenia with ganciclovir (preventing concomitant zidovudine therapy) and renal impairment with foscarnet (often manageable with hydration).

CONTROL: Cytomegalovirus hyperimmune globulin given to seronegative bone marrow or renal transplant recipients may be prophylactic. Limiting transfusions, using products filtered to remove leukocytes, and selecting cytomegalovirus-seronegative donors are all important in reducing the rate of cytomegalovirus transmission.


INFLUENZA

(Swine and Equine Influenza, Fowl Plague)

AGENT: The causative agent is influenza type A (Orthomyxoviridae).

RESERVOIR AND INCIDENCE Humans, wild and domestic birds, horses and pigs are reservoirs of influenza viruses, which appear to be species specific. Convalescent carriers act as reservoir hosts between epidemics. Antigenic shift is probably required before animal virus becomes epidemic in humans. Sporadic human infections with swine and avian strains have been reported. Human cases of influenza have occurred from contamination by aerosols from infected ferrets and vice versa.

TRANSMISSION: Transmission is by inhalation of droplets produced by coughing and sneezing, especially in crowded, enclosed spaces.

DISEASE IN ANIMALS: Fever has a sudden onset followed by anorexia, coughing, respiratory distress and mucoid nasal discharge, with rapid recovery. There is consolidation in the lung. Copious mucopurulent exudate fills the bronchioles.

DISEASE IN HUMANS: Typical symptoms include fever, chills, headache, myalgia, malaise, coryza, pharyngitis and cough with full recovery within two weeks, although viral or secondary bacterial pneumonia may develop, especially in the elderly.

DIAGNOSIS: Virus isolation, CF or HI serology.

TREATMENT: Bed rest, analgesics, and cough mixtures. Amantadine decreases duration by 50%. Antibiotics are reserved for bacterial complications. Ribavirin aerosol has helped severely ill patients.

PREVENTION/CONTROL: Polyvalent human influenza vaccine provides partial immunity (about 85% efficacy) for a few months to one year. The vaccine's antigenic configuration changes yearly and is based on prevalent strains of the preceding year. Chemoprophylaxis with amantadine will markedly reduce the attack rate among exposed individuals if begun immediately and continued for 10 days. Vaccinate horses annually. Prohibit imports of live poultry and poultry meat from countries where fowl plague occurs.


NEWCASTLE DISEASE

(Pseudo Fowl Pest)

AGENT: Newcastle disease virus, family Paramyxoviridae

RESERVOIR AND INCIDENCE The natural reservoir is wild and domesticated birds.

TRANSMISSION: Transmission is by inhalation of infectious aerosols. Intensive conditions favor spread by contact, on inanimate objects and airborne between poultry houses. Human infections occur mainly amongst laboratory workers and those who work with infected chickens or who give live vaccine, especially by aerosol.

DISEASE IN ANIMALS: In poultry, respiratory and nervous signs occur, including gasping and coughing; also drooping of wings, twisting of the head and neck; inappetence and paralysis. Egg production ceases. Petechial hemorrhages are characteristic, especially in the preventricular mucosa. Necrosis of the intestinal mucosa give a "bran" like appearance. Congestion and mucoid exudate appears in the lungs and bronchi.

DISEASE IN HUMANS: Usually symptoms are confined to painful conjunctivitis lasting a few days, but fever and influenza-like symptoms for up to 3 weeks may follow.

DIAGNOSIS: Virus isolation or ELISA.

TREATMENT: Symptomatic.

PREVENTION/CONTROL: Hygienic precautions are needed when handling infected birds. Avoid inoculation injuries and ensure laboratory safety. Maintain strict hygiene in poultry sheds and quarantine of imported live birds, prohibit importation from infected countries, sterilization of, or prohibition on, waste food fed to poultry. Vaccination of poultry.

OTHER SPECULATIVE VIRAL ZOONOSES:


ROTAVIRUS:

Rotaviruses cause gastroenteritis in animals and humans. They are not strictly species-specific, and experimental cross-infections with human or animal rotaviruses in several animal species have been confirmed. The finding of serotypes common to man and various animal species might indicate that some serotypes can infect both. However, the natural occurrence of cross-infections between species and the possible role of animals in the epidemiology of the disease in humans are still unknown.


RETROVIRUSES:

The human immunodeficiency viruses (HIVs)-acquired immunodeficiency syndrome (AIDS) or HAIDS pandemic originated from lentiviruses of nonhuman primates (thus qualifying as a zoonosis) that moved into humans in Africa. The HAIDS patients eventually die of opportunistic infections, all potentially zoonotic. The HAIDS infection remained parochial, first endemically and then epidemically, until the African urbanization that occurred in each of the countries postindependence. The latter included wars and the massive movement of soldiers (virologically naive) from the American continent to Africa and back. The HAIDS viral ecology coincided with African swine fever (ASF) in the Americas. Haiti became the focal point for both infections. Some infected Haitians also became, together with some infected drug addicts in the United States, a source of contaminated human blood for transfusions and production of plasma derivatives.

BITES AND SCRATCHES

One to two million Americans are bitten by animals annually, and bites are responsible for 1% of emergency department visits. The estimated annual incidence of animal bites is as follows: dog bites, 1-2 million; cat bites, 400,000; snake bites, 45,000; and rats and mice, 43,000. An increased risk of infection in patients more than 50 years of age, those with wounds of the upper extremities, and those with puncture wounds has been noted. Prior splenectomy or mastectomy may increase the risk of severe infection. Wild rat bites present public health problems. Bites cause pain, anxiety, wound disfigurement, and wound infections. Many organisms are capable of infecting animal bite wounds including Pasteurella spp., Capnocytophaga canimorsus, Afipia felis, Rochalimaea henselae and quintana, Clostridium tetani, Streptobacillus moniliformis, Spirillum minus, Tularemia, and Rabies. Bites from NHP's infected with Herpes B-Virus could pose a serious threat to humans. Pit Bulls were reported to be responsible for 20% of dog bite-related fatalities in 1979/80. By 1987-88, the breed was responsible for 62% of such deaths.

TREATMENT OF BITE WOUNDS:

Maintain records of all bites and scratches in the animal facility. Notify physician. A lab animal program should have an occupational medicine service available that understands the problems associated with animal handling and is informed about zoonotic diseases. Cultures should be taken and clean wound properly. * All wounds should be liberally irrigated. * Edematous body parts should be elevated. * Apply antiseptic. * Tetanus must be current. * Assess Rabies or Herpes B risk (notify public health personnel). * Antibiotic therapy should be based on the wound culture.

ALLERGIC SENSITIVITIES

Allergic skin and respiratory reactions are quite common in personnel working with laboratory animals. Many animals are implicated including the cat, dog, horse, rabbit, rat, mouse, hamster, gerbil, guinea pig, and NHP. Up to 8% of clients with budgies are affected. Hypersensitivity reactions include: 1. Nasal congestion 2. Runny nose 3. Sneezing 4. Itching of eyes 5. Angioedema 6. Asthma 7. A variety of skin manifestations (localized urticaria and eczema) Maximal allergenic activity in humans resulted from the albumin fraction of pelt extracts from rats, mice, and rabbits. The fraction of G. pig extract with maximum allergic activity seems to be a prealbumin. Two major allergens in mouse serum, skin, and urine have been identified. *THE MAJOR URINARY PROTEIN COMPLEX IS ONE OF THE MOST RECENT ALLERGENS TO BE IDENTIFIED FROM MOUSE PELTS AND SKIN. Suggests that a possible cause of sensitization in lab personnel is dispersal of urinary protein from litter in mouse cages.

DIAGNOSIS: History Physical Exam Pulmonary Function Tests Skin Testing Laboratory Tests a. CBC b. Immunoglobulins and IgE antibody specific to one allergen as measured by the RAST (Radioallergosorbent Test) c. Nasal smears for eosinophilia d. Serum precipitants to specific allergens

TREATMENT AND PREVENTION: Pharmacologic treatment Allergen Immunotherapy Complete avoidance of the antigen Reducing exposure to offending antigen a. Reduction of direct animal contact time b. Increasing room ventilation c. Exhaust hoods d. Filter caps on cages e. Protective clothing, masks, and respirators

ARTHROPOD INFESTATIONS In addition to the mite that causes sarcoptic scabies, there are several other species that occasionally infest human skin and cause a temporary dermatitis, although they are incapable of establishing themselves on this aberrant host. These mites belong to the families Cheyletidae, Dermanyssidae, and Macronyssidae. Animals play an essential role in the life cycle of the flies whose larvae cause specific myiases. Man is only an accidental host of these larvae, and in some myiases, such as those due to Oestrus ovis or Gasterophilus spp., an aberrant host in which the larvae cannot complete their development. Larval invasions of human skin or natural cavities occur when there is a high incidence of animal myiasis. Pentasomes (Armillifer sp.) are wormlike arthropods that are almost exclusively parasites of the reptilian respiratory system. Snakes are the definitive hosts and many wild rodents, on which snakes feed are the intermediate hosts. The female parasite deposits eggs in the respiratory cavities of the reptiles. The eggs are expectorated or swallowed and then eliminated with the feces. Humans can be accidental hosts, by handling infected reptiles and placing contaminated hands to the mouth. In humans the infection is usually asymptomatic. The encapsulated larvae might be found during laparotomies or can be diagnosed by radiographic examination.

PREVENTION/CONTROL: Fleas, ticks, and mites should be controlled. Gloves and protective clothing should be worn when handling lab animals.

PROTOZOAN DISEASES


BABESIOSIS

(Piroplasmosis)

AGENT: Babesiosis in humans is a rare intraerythrocytic infection caused by Babesia divergens and microti.

RESERVOIR AND INCIDENCE Natural hosts for B. microti are various wild and domestic animals, particularly the white-footed mouse and white-tailed deer. With extensions of the deer's habitat, the range of human infection appears to be increasing. In the USA, the parasite has been found in coastal and island areas of the northeast and mid-Atlantic states as well as Wisconsin, Minnesota, and California. B. divergens occurs in Europe.

TRANSMISSION: Humans are infected as a result of Ixodes tick bites, but transmission from blood transfusion has also been reported. Splenectomized, elderly, or immunosuppressed persons are the most likely to have severe manifestations.

DISEASE IN ANIMALS: Many animals show only mild fever and recover spontaneously. Deaths, which occur commonly in cattle, are due to either anemic anoxia or pulmonary thrombosis. Other lesions stem from the hemolysis and include enlarged spleen, liver, and hemoglobinuric nephrosis.

DISEASE IN HUMANS: B. microti infection lasts a few weeks to a month; the illness is characterized by irregular fever, chills, headache, diaphoresis, myalgia, and fatigue but is without malaria-like periodicity of symptoms. Most patients have a moderate hemolytic anemia, and some have hepatosplenomegaly. The disease is self-limited and most patents recover without sequelae. Infection with B. divergens has only been reported in splenectomized patients and progresses rapidly with high fever, severe hemolytic anemia, jaundice, hemoglobinuria, and renal failure; death usually follows.

DIAGNOSIS: ID of the intraerythrocytic parasite on Giemsa-stained blood smears or serology.

TREATMENT: B. divergens: blood transfusions, renal dialysis, pentamidine plus trimethoprim-sulfa. B. microti: Treat symptomatically since most case are self-limiting. In splenectomized patients, quinine plus clindamycin and transfusions.

PREVENTION/CONTROL: Control rodents and ticks. Vaccinate livestock.


TOXOPLASMOSIS

AGENT: Toxoplasma gondii - 4 to 7 microns long. 2-4 wide - Subphylum Apicomplexa, Family Eimeriidae

RESERVOIR AND INCIDENCE Infection in humans and lower animals is widespread. An estimated 500 million humans have been infected with the organism. Serologic surveys in the United States using the SABIN-FELDMAN DYE TEST have demonstrated T. gondii infection in 30-80% of cats. Significance - Presumably all serologically positive cats have shed toxoplasma oocysts and could re-shed organisms during reinfection or reactivation. Life cycle consists of: 1. Definitive host (felids: intestinal infection with shedding of oocysts; only host in which sexual form develops.) - Domestic cat predominates as reservoir for zoonotic transmission in the home and laboratory environment. 2. Intermediate hosts - *Mice, rats, hamsters, G. pigs, rodents, rabbits, dogs, sheep, cattle, & NHP's. - These have not proved to be important in zoonotic infection in the laboratory animal environment (organism replicates asexually in extraintestinal sites only).

TRANSMISSION: Fecal-Oral: Ingestion of meat containing cysts or tachyzoites or ingestion of oocysts Oocysts become infective after sporulation - occurs in 2 to 3 days. Transmission to man occurs via a. Eating raw or undercooked meat containing cysts. b. Ingesting sporulated oocysts from cat feces. c. Transplacentally. 1/3 of US human population has serologic evidence of past infection.

DISEASE IN CATS: Most postnatally acquired infections in cats are ASYMPTOMATIC. Prepatent period variable - 3 days to several weeks. Shedding occurs for 1-2 weeks - PUBLIC HEALTH HAZARD. Oocyst shedding reactivated by induction of hypercorticism or superinfection with other feline microorganisms.

DISEASE IN MAN: The infection is very common in humans, but clinical disease is of low incidence and occurs only sporadically. Postnatal infection - Less severe disease and commonly presents as a generalized lymphadenopathy that may resolve without treatment in a few weeks. Congenital infection results in systemic disease often with severe neuropathological changes. Rarely, serious ocular or systemic toxoplasmosis can be acquired by older individuals or reactivated in immunocompromised individuals. Clinical Signs include fever, skin eruption, malaise, myalgia, arthralgia, cervical lymphadenopathy, pneumonia, myocarditis, meningoencephalitis, and chorioretinitis.

DIAGNOSIS: Serology, isolation, microscopic demonstration of organisms in smear or section. Paired serum samples taken one or more weeks apart. a. IFA: serial titers of suspected infections. b. Sabin- Feldman Dye Test: most sensitive test, but rarely used. c. ELISA. The demonstration of cysts does not establish a causal relationship to clinical illness, since cysts may be found in both acute and chronic infections. However, only finding tachyzoites in blood or body fluids confirms active infection.

TREATMENT IN MAN: The treatment of choice is pyrimethamine plus either trisulfapyrimidines or sulfadiazine. Folinic acid is given to avoid the hematologic effects of pyrimethamine-induced folate deficiency.

PREVENTION/CONTROL: Freezing of meat to -20oC (-4oF) for 2 days or heating to 60oC (140oF) kills cysts. Under appropriate environmental conditions, oocysts passed in cat feces can remain infective for a year or more. Thus, children's play areas should be protected from cat and dog feces. Cats a. Daily cleaning of litter pans (since oocysts not infective for 2 to 3 days) b. Wear gloves c. Wash hands before eating d. Should only be fed dry, canned, or cooked meats Pregnant women should have their serum examined for Toxoplasma antibody. If the IgM test is negative but an IgG titer is present and less than 1:1000, no further evaluation is necessary. Those with negative titers should take measures to prevent infection by avoiding contact with cat feces and avoid working in soil or gardens that could be contaminated by cats, etc. and by thoroughly cooking meat. Hands should be washed after handling raw meat and before eating or touching the face.


PLASMODIUM spp.

Cause of malaria. Both quartan and tertian types occur in NHP. There is a high incidence in newly imported animals. Requires anopheline mosquitoes for transmission. Therefore zoonotic potential exists anywhere mosquitoes have access to infected animals. Generally thought that these conditions prevail only in Southern climates where monkeys are housed outdoors, but mosquitoes are found in Northern as well as Southern U.S. In general, human malaria caused by plasmodia of simian origin resembles a mild and benign infection caused by human plasmodia. The disease is of short duration, parasitemias are low, and relapses are rare. Must control flying insects in all primate facilities.


AFRICAN TRYPANOSOMIASIS

(African Sleeping Sickness, Gambian Trypanosomiasis, Rhodesian Trypanosomiasis)

AGENT: Trypanosoma brucei gambiense and rhodesiense.

RESERVOIR AND INCIDENCE Many wild and domestic animals harbor infection. In Gambian trypanosomiasis, humans are the main reservoir and source of infection for the vector tsetse fly (Glossina palpalis, tachinoides, or fuscipes). In Rhodesian trypanosomiasis, animals, especially domestic cattle and pigs, play an important role as reservoirs.

TRANSMISSION: The tsetse fly is infected when it bites during the parasitemic phases and the trypanosome develops in the vector, culminating in infection of its saliva. Transmission is by the tsetse fly bite. In humans, intrauterine infection has been recorded.

DISEASE IN ANIMALS: Occasionally mild disease occurs in domestic animals with chronic nervous sequela in T. gambiense infection.

DISEASE IN HUMANS: The trypanosomal chancre: This a local pruritic, painful inflammatory reaction with regional lymphadenopathy that appears about 48 hours after the tsetse bite and lasts 2-4 weeks. The hemolymphatic stage: Usually absent or unnoticed in T. b. gambiense infections. Irregular fevers, headaches, joint pains, malaise, pruritus, papular skin rash, edemas. Patients may succumb to myocarditis before signs of central nervous system invasion appear. The meningoencephalitic stage: Insomnia, motor and sensory disorders, abnormal reflexes, somnolence to coma. Trypanosomes and increased white cells and protein in cerebrospinal fluid.

DIAGNOSIS: Definitive diagnosis requires identifying the organism in the bite lesion, blood, lymph node aspirate, or CSF. Serologic tests become positive after 12 days.

TREATMENT: Hemolymphatic stage: Suramin, eflornithine or pentamidine. Late disease: melarsoprol or eflornithine or tryparsamide plus suramin.

PREVENTION/CONTROL: Wear long sleeves and trousers in endemic areas. Avoid wearing dark-colored clothing, and use mosquito nets while sleeping. Repellents do not work on tsetse flies. Pentamidine is used as a chemoprophylaxis against the Gambian type.


AMERICAN TRYPANOSOMIASIS

(Chagas's Disease, Chagas-Mazza Disease, South American Trypanosomiasis)

AGENT: Trypanosoma cruzi

RESERVOIR AND INCIDENCE Dogs, cats, and guinea pigs are the main reservoirs for human infection. T. cruzi occurs only in the Americas; it is found from southern South America to northern Mexico, Texas, and the southwestern U.S. An estimated 12 million people are infected, mostly in rural areas, resulting in about 60,000 deaths yearly.

TRANSMISSION: Humans are infected when the insect's feces become rubbed into the wound caused by the bite of an infected bloodsucking insect (triatomid) or when the conjunctiva, mucous membranes or abrasions become contaminated. After invading local reticuloendothelial cells, the trypanosome multiplies in the blood. Adaption of triatomid vector to the human domestic environment allows transfer of infection between animals, from animals to humans or from human to human. Transmission by blood transfusions from infected persons, congenital infection, breast milk and laboratory accidents are possible.

DISEASE IN ANIMALS: Acute and inapparent infection occur in wild animals and chronic disease is seen in dogs. The acute form, which includes fever, enlarged liver and lymph nodes and heart irregularities, lasts 10-30 days before becoming chronic without further clinical signs, though sometimes myocarditis occurs. Lesions in dogs resemble those in humans.

DISEASE IN HUMANS: Acute illness usually occurs in children with a furuncle at the site of infection. Signs include fever, malaise, enlarged lymph nodes, liver and spleen. If the primary site of infection is the eye there is unilateral edema of eyelids and conjunctivitis. Rarely myocarditis and meningoencephalitis occur. Chronic symptoms in adults result from arrhythmias and dilation of the heart, esophagus and colon. Furuncles (chagoma) appear at the point of entry of the infection. Enlarged liver and spleen, myocarditis, grossly dilated heart, intestines, esophagus, ureter and bladder and meningoencephalitis occur.

DIAGNOSIS: Several serologic tests are available and are of presumptive value; when possible, more than one test should be used. In the acute stage, trypanosomes should be looked for by examination of anticoagulated fresh blood for motile organisms. In the chronic stage, the parasite can only be detected by culture or xenodiagnosis.

TREATMENT: Therapy is unsatisfactory; the drugs are toxic and often ineffective. In the acute phase, however, cure is usually possible. In the chronic phase, although parasitemia and xenodiagnosis become negative, treatment does not alter the serologic reaction, cardiac function, or progression of the disease. Nifurtimox or benznidazole is used. Ketoconazole shows promise also.

PREVENTION/CONTROL: Destroy the vector by insecticides. Use insect nets to prevent bites. Screen blood donors.


AMEBIASIS

(Amebic Dysentery, Amebiosis)

AGENT: Entamoeba histolytica.

RESERVOIR AND INCIDENCE The reservoir of E. histolytica is man. The infections is present worldwide but is most prevalent and severe in tropical areas, where rates may exceed 40% under conditions of crowding, poor sanitation, and poor nutrition. It is estimated that there are about 50 million case of invasive amebiasis and 40,000-100,000 deaths annually worldwide. In temperate areas, however, amebiasis tends to be asymptomatic or a mild, chronic infection that often remains undiagnosed. In the USA, seropositive rates up to 2-5% have been reported in some populations. Reported incidence of 0-31% in the feces of clinically normal Rhesus monkeys, 2-67% in Chimps, and up to 30% in other NHP.

TRANSMISSION: Transmission may be by ingestion of infective cysts, contaminated water or food, by flies, or fomites. Exists as resistant cysts or more fragile trophozoites CYSTS are the INFECTIOUS form found in the stool of asymptomatic carriers or patients with mild disease. The cysts remain viable, if moist and cool for 12 days. Remain viable for 30 days in water. Laboratory animal personnel are usually infected from fecal matter transferred to the skin or clothing.

DISEASE IN ANIMALS: In dogs, infection by E. histolytica is generally asymptomatic and frequently localized in the cecum. Occasionally, it can invade tissues and cause acute or chronic amebiasis. Rhesus monkeys are generally resistant and usually experience asymptomatic infection, but chronic, mild colitis can occur. In chimpanzees, the infection can persist for a long time, in most cases subclinically, but sometimes it invades the tissues causing ulcerative colitis and hepatic abscesses. New World monkeys are considered more susceptible to the disease than Asian or African monkeys. Wild rats can also harbor E. histolytica; the protozoan can be found in the large intestine as a commensal or it can invade the mucosa and cause amebic dysentery.

DISEASE IN HUMANS: Mild to moderate colitis: recurrent diarrhea and abdominal cramps, sometimes alternating with constipation; mucus may be present; blood is usually absent. Severe colitis: semiformed to liquid stools streaked with blood and mucus, fever, colic, prostration. In fulminant cases, ileus, perforation, peritonitis, and hemorrhage occur. Hepatic amebiasis: fever, hepatomegaly, pain, localized tenderness.

DIAGNOSIS: Use fresh fecal specimen to identify cysts or trophozoites. Sedimentation. Must measure to distinguish from other nonpathogenic amoebae. Indirect HI For hepatic amebiasis, ultrasonography can locate the cyst and fine needle aspiration is performed to find the organism.

TREATMENT: May require the concurrent or sequential use of several drugs. The tissue amebicides dehydroemetine and emetine act on organisms in the bowel wall and in other tissue but not in gut lumen. Chloroquine is active principally against amebas in the liver. The luminal amebicides diloxanide furoate, iodoquinol, and paromomycin act on organisms in the bowel lumen but are ineffective against amebas in the bowel wall or other tissue. Metronidazole is unique in that it is effective both in the lumen and in the wall and in other tissue. However, when used alone, it only cures 50% of the cases.

PREVENTION/CONTROL: Strict sanitation & personal hygiene, protective clothing and gloves. Fecal screening of NHP. Protect water supply from fecal contamination. Usual chlorine levels don't destroy cysts. *10ppm chlorine residual necessary to destroy cysts Heat to 50oC (122oF) kills cysts. Adequate cooking to destroy cysts. Protect food from fly contamination.


BALANTIDIASIS

(Balantidial dysentery)

AGENT: large ciliated protozoan, Balantidium coli. Trophozoite 50-70 microns by 40-50 microns.

RESERVOIR AND INCIDENCE Distributed worldwide especially in the tropics. Swine and possibly rats and NHP's are the reservoir hosts. Humans, great apes, & several monkey species may carry it. Incidence in NHP colonies - 0 to 63% Usually asymptomatic, but may see diarrhea.

TRANSMISSION: Ingestion of cysts or trophozoites from infected animal or human feces. Cyst is the infectious form. Contaminated water or food.

DISEASE IN ANIMALS AND MAN: Many infections are asymptomatic and probably need not be treated. Chronic recurrent diarrhea, alternating with constipation, is most common, but severe dysentery with bloody mucoid stools, tenesmus, and colic may occur intermittently.

DIAGNOSIS: Use fresh fecal samples to identify trophozoites or cysts Trophozoites in scrapings or biopsy of ulcers of the large bowel.

TREATMENT: tetracycline or iodoquinol

PREVENTION/CONTROL: Good sanitation & personal hygiene practices in NHP & Swine colonies. Protect water & food from fecal contamination Identify positive lab animals and treat.


CRYPTOSPORIDIOSIS

AGENT: Extracellular protozoal organisms - similar to coccidia. Genus: Cryptosporidium, it remains unsettled whether more than one species exists. Taxonomy of species somewhat controversial but considered to be infective across species lines.

RESERVOIR AND INCIDENCE Rodents, birds (particularly turkeys and chickens), ruminants, fish, reptiles, cats, dogs, rabbits, NHP's. Children over 2 years of age, animal handlers, travelers, homosexual men, and close personal contacts of infected individuals (families, health care and day-care workers) may be particularly likely to be infected. More than a dozen outbreaks have been reported in day-care centers around the world. Two major waterborne outbreaks have been documented. Cryptosporidium antibodies were detected in the serum of 20 of 23 cats (87%) suggesting that the exposure rate may be high.

TRANSMISSION: Fecal-oral transmission is from animals to humans or humans to humans; waterborne transmission is also important. Oocysts passed in stool are fully sporulated and infectious; infection occurs as a result of their ingestion. In humans and animals, the full life cycle occurs within a single host. The organisms attach to the microvillous borders of enterocytes of the small bowel and also are found free in mucosal crypts. The host cell membrane deteriorates, leaving the parasitic membrane in direct contact with epithelial cell cytoplasm. The organisms do not, however, invade the tissues.

DISEASE IN ANIMALS: Severe watery diarrhea in neonatal calves and lambs. In turkeys and chickens, the parasites are reported to occur in the sinuses, trachea, bronchi, cloaca, and bursa of Fabricius. The respiratory disease causes coughing, gasping, and airsacculitis. In reptiles, cryptosporidiosis is frequently reported in association with postprandial regurgitation. The organism affects the GI mucosa, resulting in marked thickening of the rugae and loss of segmented motility.

DISEASE IN MAN: In immunocompetent persons, infection varies from no symptoms to mild enteritis to marked watery diarrhea (up to 10 stools daily) without mucus or gross or microscopic blood. Low-grade fever, malaise, nausea, vomiting, abdominal cramps, anorexia and weight loss may occur. The infection is generally self-limited and lasts a few days to about 2 weeks. In immunologically deficient patients, the illness is characterized by profuse (up to 15L daily), cholera-like diarrhea and by fever, severe malabsorption, marked weight loss, and lymphadenopathy. In AIDS, infection may involve any part of the GI tract, and multisystemic involvement has been described, especially involving the respiratory tract.

DIAGNOSIS: Diagnosis is by detection of oocysts in stool by a variety of flotation or concentration methods or by mucosal biopsy, followed by special staining methods that use modifications of an acid-fast stain (routine fecal staining methods do not detect the organisms). Three stools should be examined over 5 days. A fluorescein-labeled IgG monoclonal antibody test has recently become available to detect oocysts.

TREATMENT: No successful treatment has been developed so far. Generally, no treatment other than supportive is needed in immunocompetent persons since it is self-limiting. In immunoincompetent persons, spiramycin, zidovudine (AZT), paromomycin, octreotide, and eflornithine have been reported of value.

PREVENTION/CONTROL: Personal hygiene. Careful handwashing by those in contact with any animals with scours.


GIARDIASIS

Most common intestinal protozoan parasite of people in the U.S.

AGENT: Giardia lamblia Has both a cyst (infective) and trophozoite form

RESERVOIR AND INCIDENCE The parasite occurs worldwide and is nearly universal in children in developing countries. Humans are the reservoir for Giardia, but dogs and beavers have been implicated as a zoonotic source of infection. In psittacines, the disease is commonly found in cockatiels and budgerigars. Giardiasis is a well-recognized problem in special groups including travelers, campers, male homosexuals, and persons with impaired immune states. However, Giardiasis does not appear to be an opportunistic infection in AIDS.

TRANSMISSION: Only the cyst form is infectious by the oral route; trophozoites are destroyed by gastric acidity. Most infections are sporadic, resulting from cysts transmitted as a result of fecal contamination of water or food, by person-to-person contact, or by anal-oral sexual contact. After the cysts are ingested, trophozoites emerge in the duodenum and jejunum. They can cause epithelial damage, atrophy of villi, hypertrophic crypts, and extensive cellular infiltration of the lamina propria by lymphocytes, plasma cells, and neutrophils.

DISEASE IN ANIMALS: Giardia infections in dogs and cats may be inapparent or produce weight loss and chronic diarrhea or steatorrhea, which can be continuous or intermittent, particularly in puppies and kittens. Calves with clinical giardiasis have been reported. Feces are usually soft, poorly formed, pale, and contain mucus. Gross intestinal lesions are seldom evident, although microscopic lesions, consisting of villous atrophy and cuboidal enterocytes, may be present.

DISEASE IN MAN: Most infections are asymptomatic. In some cases, acute or chronic diarrhea, mild to severe, with bulky, greasy, frothy, malodorous stools, free of pus and blood. Upper abdominal discomfort, cramps, distention, excessive flatus, and lassitude.

DIAGNOSIS: Diagnosis is by identifying cysts or trophozoites in feces or duodenal fluid. Unless they can be examined with an hour, specimens should be preserved immediately in a fixative. Three stool specimens should be examined at intervals of 2 days or longer. A stool ELISA test or IgM serology are available.

TREATMENT: Tinidazole, Metronidazole (FLAGYL), quinacrine, or furazolidone. Alternative drugs are Tinidazole or albendazole.

PREVENTION/CONTROL: Fecals to screen dogs and NHP's. Hygiene, protective clothing, when handling animals. Prevention requires safe water supplies, sanitary disposal of human feces, adequate cooking of foods to destroy cysts, protection of foods from fly contamination, washing hands after defecation and before preparing or eating foods, and, in endemic areas, avoidance of foods that cannot be cooked or peeled.


LEISHMANIASIS

[Cutaneous leishmaniasis: Chiclero ulcer, espundia, pianbols, uta, and buba (in the Americas); oriental sore, Aleppo boil (in the Old World); Bagdad boil, Delhi boil, Bauru ulcer (in the Middle East). Visceral leishmaniasis: kala-azar]

AGENT: The causative agents of cutaneous leishmaniasis are Leishmania mexicana and L. brasiliensis in the Americas, and L. tropica in the Old World; and of visceral leishmaniasis, L. donovani, L. infantum, and L. chagasi.

RESERVOIRS AND INCIDENCE: The geographic distribution of the cutaneous disease is Texas, Mexico, Central and South America, India, Pakistan, the Middle East, southern Russia, the Mediterranean coast and Africa. The distribution of visceral leishmaniasis is poorly reported, but foci probably occur in the Mediterranean basin, the Middle East, India, China, Mexico, Central and South America, and Africa. Wild animals, dogs and humans serve as reservoirs. Domestic dogs may be an important reservoir for humans. Humans are the only known reservoir in India.

TRANSMISSION: Sandfly vectors transmit cutaneous leishmaniasis. Person-to-person, congenital, and blood-borne transmission of visceral leishmaniasis are possible.

DISEASE IN ANIMALS: L. mexicana causes ulcers of the skin in rodents and other wild animals, usually at the base of the tail. L. braziliensis causes a systemic infection with few skin lesions in wild animals. No skin lesions have been found in dogs. Dogs infected by L. tropica may suffer form cutaneous lesions similar to those found in humans. L. donovani produces visceral lesions in dogs, with enlarged lymph nodes, liver and spleen.

DISEASE IN HUMANS: In the cutaneous disease, the primary lesion is a painful ulcer or nodule at the site of infection persisting for several months, with residual scarring. Further lesions may develop in skin and mucous membranes. Infiltration by inflammatory cells at the inoculation site supports the growth of the parasite. This progresses into a large area of chronically inflamed granulation tissue. The overlying skin undergoes hyperplasia and then necrosis with spreading ulceration. Metastatic lesions occur with a similar inflammatory reaction. The lesions may heal, become fibrosed or extend indefinitely to produce considerable disfigurement. In the visceral disease, intermittent irregular fever occurs with sweats, enlarged spleen, weight loss and anemia leading to ascites, edema, diarrhea and secondary infections. Dark pigmentation of the skin may occur. There is gross enlargement of liver and spleen. Without treatment, the case fatality rate is 90%.

DIAGNOSIS: Definitive diagnosis is achieved by finding the parasite-either the amastigote in stained smears or biopsies, or the motile promastigote in culture. Serologic and skin tests provide only indirect evidence of infection.

TREATMENT: Treatment remains inadequate because of drug toxicity, long courses required, and frequent need for hospitalization. The drug of choice is sodium antimony gluconate. Alternative drugs for some forms of infection are amphotericin B and pentamidine.

PREVENTION/CONTROL: Use insecticides in house and buildings to control the vector. Eliminate rubbish heaps which are breeding areas for sandflies. Avoid sandfly bites by using insect repellents and protective clothing. Keep dogs indoors after sundown and remove infected dogs.


MICROSPORIDIOSIS

Protozoa of the order Microsporidia (Phylum: Microspora) are represented by about 700 species parasitic in hosts of many invertebrate and vertebrate groups. These protozoa have only recently been recognized as "opportunistic" parasites in patients with AIDS. In veterinary medicine, Microsporidia are well known as causative agents of certain animal infections, such as nosemosis in bees, encephalitozoonosis in rabbits and of numerous fish diseases. Microsporidia are obligatory intracellular parasites (macrophages, histiocytes, endothelial cells, kidney tubular cells, etc.) with a characteristic spore stage and a unique mode of infecting host cells. Spores of Microsporidia species infecting mammals are ovoid or piriform, 1.5-5.0 þm long with mostly a thick wall consisting of 2 main layers (exospore, endospore). The spore contains a coiled, minute tube (polar tube or polar filament) connected with a complex extrusion apparatus and a nucleated, infective sporoplasm. After ingestion by a suitable host, the physiological conditions of the digestive tract stimulate the spores to evert with force the coiled polar filament. The tip of the filament penetrates the host cell membrane, the sporoplasm migrates through the tube and enters the cytoplasm where asexual multiplication (merogony) and spore formation (sporogony) occur. Sporoplasms usually infect the gut epithelium either to develop there or to be transferred, probably by the action of phagocytic cells, to the circulation and their preferred site of development in various organs. The spores, released to the environment from the intestinal or urinary tract, are ingested by a new host. All Microsporidia examined so far have a direct developmental cycle. Up to now 5 genera of Microsporidia have been identified as causative agents of human infections. ENCEPHALITOZOON CUNICULI has a worldwide distribution and is a common parasite of wild and domesticated rabbits, laboratory rodents and a wide spectrum of other mammals such as carnivores, ruminants, pigs, monkeys and man. Encephalitozoon parasites found in birds may belong to other species. E. cuniculi, widespread in conventional rabbit colonies in up to 76% of the stock, mostly causes latent infections and only rarely disease. Focal granulomatous encephalitis and nephritis are the main pathological changes. Natural transmission in rabbits is possible by the oral, tracheal and transplacental routes, but oral infection appears to be the most important way. In rabbits, spores of E. cuniculi are excreted in the urine in concentrations up to 106 spores/ml. The human cases of E. cuniculi infections include two cases of encephalitis with a favorable evolution in two immunocompetent children. In a 35-year old man with AIDS the E. cuniculi infection resulted in a granulomatous hepatitis. Recently, another case in a 45-year old patient with AIDS (Greek nationality) was described in Switzerland. An unusual peritonitis with a large granulomatous mass, containing developmental stages of E. cuniculi, were found at autopsy. In Sweden (S) and Great Britain (GB) high prevalences of serum antibodies against E. cuniculi antigen have been detected in persons with malaria (S: 38%, GB: 7%), Chagas disease (S: 15%), schistosomiasis (GB: 12%), filariases (S: 9%), neurological disorders (GB: 6%) and in travellers after a stay in the tropics (S: 12%) while non-exposed persons, animal dealers and some other groups were seronegative. The question is open whether E. cuniculi is more common in the tropics or whether tropical parasites cause immunodepression which supports microsporidian infections. Besides a systemic infection in a immunodefective child with Nosema conori and two cases of keratitis caused by Microsporidian species, the recent description of a myositis in an AIDS patient due to Pleistophora sp. is of special interest as parasite transmission from fish was suspected. Another species of this group is Enterocytozoon bieneusi. This parasite was first described in 1984 in France in a 29-year old AIDS patient who had suffered from heavy diarrhea. Microsporidia were found in enterocytes of the duodenum, jejunum and ileum. This parasite or similar forms were subsequently detected in at least 9 other patients with AIDS in the USA, Uganda and the Netherlands. The origin of this parasite is unknown. An infection similar to E. bieneusi in the enterocytes of a Callicebus monkey has been described.

DIAGNOSIS: Diagnosis is based on the direct detection of Microsporidia by histology after hematoxylin-eosin, Giemsa, Gram or other staining (spores are Gram-positive!) or by electron microscopy in autopsy or biopsy material. Serum antibody detection provides another tool for the in vivo diagnosis of encephalitozoonosis.

TREATMENT: Information on chemotherapy of microsporidian infections is scanty. In experimental infections chloroquine and oxytetracycline reduced harvests of E. cuniculi spores only by 69% and 58%, respectively. Treatment of an AIDS patient suggested that either metronidazole or ganciclovir may have inhibited the microsporidian infection.

HELMINTH INFECTIONS Many of the helminth parasites common to both animals and humans have an indirect life cycle that is interrupted in the laboratory environment and thus limits the zoonotic potential of these organisms. Laboratory housed primates are the most likely source of parasitic infection for animal handlers. Helminths are POTENTIALLY zoonotic in the laboratory environment, but they represent a significantly smaller problem than bacterial or viral diseases. Exceptions are those with a direct life cycle. In General: proper quarantine, surveillance and treatment to decrease endoparasitic burden, routine sanitation to eliminate parasitic ova before they can become infective, and education of personnel on standard hygiene practices are measures that can be taken to prevent transmission of these diseases.


NEMATODE ZOONOSES


ANISAKIASIS

(Herring worm disease) A common parasitic infection from fish. The parasites are widely distributed. Human disease occurs where people eat raw or lightly smoked or salted saltwater fish or squid (e.g., in Japan, the Netherlands, Scandinavia and Central America). The causative agents are Anisakis, Phocanema and Contracaecum (Nematoda). There is no vaccine.

RESERVOIR AND MODE OF TRANSMISSION: Definitive hosts are marine mammals such as dolphins or seals. These pass the parasite's eggs in their feces. The eggs hatch and produce larvae which infect the first intermediate host, usually a crustacean. A fish may be the second intermediate host. Humans are aberrant hosts infected by eating fish.

INCUBATION PERIOD: Humans and animals. A few hours to a few weeks.

CLINICAL FEATURES: Humans. There may be fever, abdominal pain, vomiting, hematemesis, coughing, pseudoappendicitis, and possibly symptoms associated with intestinal perforation. Animals. Fish fail to thrive if heavily infected.

PATHOLOGY: Humans.The larvae usually remain in the intestine causing few lesions. However, they sometimes invade the stomach wall causing hematemesis and may lodge in the mesenteric veins or in the viscera where they induce eosinophilic granulomas and abscesses. Larvae may migrate up the esophagus to the oropharynx. There is a low grade eosinophilia. Animals. In fish, atrophy of the liver occurs and sometimes fatal infection of the heart. Visceral adhesions and muscle damage can be severe.

DIAGNOSIS:

Humans. Stools may show occult blood. Mild leukocytosis and eosinophilia may be present. ELISA and RAST serologic tests may be tried but are not reliable in chronic disease. In acute infection, the larvae sometimes can be seen and removed endoscopically from the stomach. X-rays of the stomach may show a localized edematous, ulcerated area with an irregularly thickened wall, decreased peristalsis, and rigidity. Double contrast technique may show the threadlike larvae. Small bowel x-rays may show thickened mucosa and segments of stenosis with proximal dilation. Ultrasound examination of gastric and intestinal lesions may also be useful. In the chronic stage, x-rays and endoscopy of the stomach-but not of the bowels-may be helpful. The diagnosis is often made only at laparotomy with surgical removal of the parasite. Animals. Demonstrate the parasite in tissues of fish.

PROGNOSIS: Humans. The condition is rarely fatal. Animals. Wide distribution of the parasites and consequent disease results in difficulty in maintaining marine vertebrates in laboratories.

PREVENTION: Humans. Avoid raw or undercooked fish. Freezing fish kills larvae. Eviscerate fish immediately after catching. Animals. Impractical.

TREATMENT: Humans. Physical removal of larvae by gastroscopy or surgery. Animals. Not appropriate. Legislation Humans and animals. None.


TRICHINOSIS

AGENT: Trichinella spiralis, an intestinal nematode.

RESERVOIR AND INCIDENCE Swine, dogs, cats, rats and many wild animals. Worldwide. In the U.S., there has been a marked reduction in the prevalence of trichinosis both in humans and pigs; prevalence in commercial pork now ranges from nil to 0.7%. Fewer than 100 human cases are reported annually and usually have been as a result of eating homemade sausage and other meat products using pork, horse meat, or arctic mammals.

TRANSMISSION: In the natural cycle, larvae develop into adult worms in the intestines when a carnivore ingests parasitized muscle. Pigs generally become infected by feeding on uncooked scraps or, less often, by eating infected rats. In humans, infection occurs by eating insufficiently cooked meat. In the epithelium of the small intestine, larvae develop into adults. Gravid female worms then produce larvae, which penetrate the lymphatics or venules and are disseminated via the bloodstream throughout the body. The larvae become encapsulated in skeletal muscle.

DISEASE IN ANIMALS: Usually subclinical.

DISEASE IN HUMANS: Clinical disease in humans is highly variable and can range from inapparent infection to a fulminating, fatal disease depending on the number of larvae ingested. Sudden appearance of muscle soreness and pain, together with edema of upper eyelids are common early and characteristic signs. These are sometimes followed by subconjunctival, subungual and retinal hemorrhages, pain and photophobia. Thirst, profuse sweating, chills, weakness, prostration and rapidly increasing eosinophilia may follow. GI symptoms may also occur. Remittent fever, cardiac and neurologic complications may appear. Lastly, death due to myocardial failure may occur.

DIAGNOSIS: Serologic testing and muscle biopsy.

TREATMENT: Treatment is principally supportive, since in most cases recovery is spontaneous. Mebendazole, thiabendazole, or albendazole can be given.

PREVENTION/CONTROL: Cooking (at 77oC [171oF] or above) destroys the parasite. Freezing meat up to 15 cm at 5oF for 30 days or -13oF for 10 days will destroy the parasite. Thicker pieces need to be frozen at the lower temperature for at least 20 days. These temperatures will not kill the cold-resistant Arctic strains, however. Gamma irradiation will kill the parasite. Prevent pigs from gaining access to rats or uncooked offal.


ANGIOSTRONGYLIASIS

SYNONYMS: Angiostrongylosis, eosinophilic meningitis or meningoencephalitis (A. cantonensis), abdominal angiostrongylosis (A. costaricensis).

ETIOLOGY: Two metastrongylids, Angiostrongylus (Morerastrongylus) costaricensis and A. cantonensis, are the etiologic agents. The first species is responsible for abdominal angiostrongyliasis, and the second one for eosinophilic meningitis or meningoencephalitis.The definitive hosts of both species are rodents; man is an accidental host. Both species require mollusks as intermediate hosts for the completion of their life cycle. The main definitive host of A. costaricensis is the cotton rat, Sigmondon hispidus, in which the adult nematode lodges in the mesenteric arteries and their branches on the intestinal wall. The first-stage larva emerges from eggs laid in the arteries, penetrates the intestinal wall, and is then carried with the fecal matter to the exterior. In order to continue their development, the first-stage larvae have to be ingested by a slug. Vaginulus ameghini, in which they change successively into second- and third-stage larvae. When the infective third-stage larva is ingested by a rodent, it seeks the ileocecal region, where it penetrates the intestinal wall and locates in the lymphatic vessels (both inside and outside the abdominal lymph nodes). In this location the larvae undergo two molts before migrating to their final habitat, the mesenteric arteries of the cecal region. Oviposition begins after about 18 days. and the first-stage larvae appear in the feces 24 days after infection (prepatent period). In man, an accidental host. the parasite can reach sexual maturity and produce eggs, but the eggs usually degenerate, causing a granulomatous tissue reaction. The development cycle of A. cantonensis is similar to that of A. costaricensis. The intermediate hosts are various species of land snails, slugs, and freshwater snails. The definitive hosts can become infected by ingesting infected snails, or plants and water contaminated by them with the third larvae. In addition, infection can occur as a result of consuming transfer hosts (paratenic hosts), such as crustaceans, fish, amphibians, and reptiles, which in turn have eaten infected mollusks (primary intermediate hosts). The definitive hosts of A. cantonensis are primarily various species of the genus Rattus. When they enter a rat's body, the third-stage larvae (which developed in a mollusk) penetrate the intestine and are carried by the circulatory system to the brain, where they undergo two more molts and become young adult parasites. From the cerebral parenchyma they migrate to the surface of the brain. They remain for a time in the subarachnoid space and later migrate to the pulmonary arteries, where they reach sexual maturity and begin oviposition. The eggs hatch in the pulmonary arterioles, releasing the first larva, which migrates up the trachea, is swallowed, and is eliminated with the feces. Mollusks are infected by ingesting fecal matter of infected rodents. In man, who is an accidental host, the larvae and young adults of A. cantonensis generally die in the brain, meninges, or medulla oblongata. The nematode can occasionally be found in the lungs.

GEOGRAPHIC DISTRIBUTION AND OCCURRENCE: Abdominal angiostrongyliasis, caused by A. costaricensis, is a parasitosis described a few years ago in Costa Rica; it is one of the most recently recognized zoonoses. Human disease has also been confirmed in Honduras, El Salvador, and Brazil. Suspected clinical cases have occurred in Nicaragua and Venezuela. In Panama, the adult parasite was found in five species of rodents belonging to three different families. In the past few years, the parasite has been found in several specimens of Sigmodon hispidus in Texas, USA. Ozyomys caliginosus in Colombia; and slugs in Guayaquil, Ecuador. The parasitosis is probably much more widespread than is currently recognized. A. costaricensis has not been recorded outside the Americas. Human cases of parasitism by A. cantonensis have occurred in Thailand, Vietnam, Kampuchea, the Philippines, Indonesia, Taiwan, Japan, Australia, and several Pacific islands. The parasite is much more widely distributed, and its existence in rats has been confirmed in southern China, India, Malaysia, Sri Lanka, Madagascar, Mauritius, and Egypt. Until recently, the geographic distribution of A. cantonensis was thought to be limited to Asia, Australia, the Pacific islands, and Africa. However, in recent years its presence has been confirmed in Cuba, where infected rats (Rattus norvegicus) and mollusks have been found; likewise, five human cases of meningoencephalitis have been attributed to A. cantonensis in that country. It is believed that the parasite was introduced to the island some years ago by rats from a ship from Asia. In a study carried out on rat species (R. norvegicus, R. rattus, and R. exulans) on the Hawaiian and Society Islands, the parasite was found in more than 40% of the specimens captured. In Egypt, 32.7% of 55 specimens of R. norvegicus harbored the parasite. In the province of Havana, Cuba, 12 out of 30 captured R. norvegicus were infected. In view of the worldwide distribution of R. norvegicus and R. rattus, these rodents were examined for the parasite in Puerto Rico, London, and New Orleans, but the results were negative. Eosinophilic meningitis associated with infection by A. cantonensis has been recorded in several hundred patients in endemic areas.

THE DISEASE IN MAN: The clinical manifestations of abdominal angiostrongyliasis caused by A. cantonensis are moderate but prolonged fever, abdominal pain on the right side. and, frequently, anorexia, diarrhea, and vomiting. Leukocytosis is characteristic (20,000 to 50,000 per mm3), with marked eosinophilia (11 to 82%). Palpation sometimes reveals tumoral masses or abscesses. Rectal examination is painful, and a tumor can occasionally be palpated. Lesions are located primarily in the ileocecal region, the ascending colon. appendix, and regional lymph nodes, but they are also found in the small intestine. Granulomatous inflammation of the intestinal wall can cause partial or complete obstruction. Appendicitis was the preoperative

DIAGNOSIS: in 34 cases. All but two of the children survived and recovered. The highest prevalence (53%) was found in children 6 to 13 years old, and twice as many boys as girls were affected. Ectopic localizations may occur; when the liver was affected in some Costa Rican patients, the syndrome resembled visceral larva migrans. Serologic studies carried out in Australia, in human populations living in localities where the infection occurs in rats and those living in other places where it does not, indicate that many human infections are asymptomatic.

THE DISEASE IN ANIMALS: In rodents, A. costaricensis produces lesions that are located primarily in the cecum, as well as focal or diffuse edema of the subserosa, a reduction in mesenteric fat, and swelling of the regional lymph nodes. In highly parasitized animals, eggs and larvae may be found in various viscera of the body. No significant difference in weight between parasitized and nonparasitized animals has been confirmed. Rats infected by A. cantonensis may show consolidation and fibrosis in the lungs. However, the physical appearance of the animals does not reflect the degree of pathologic changes. For both parasites, the prevalence of the infection is greater in adult than in young rodents.

SOURCE OF INFECTION AND MODE OF TRANSMISSION: Several species of rodents are known to serve as definitive hosts of A. costaricensis: Sigmodon hispidus, Rattus rattus, Zygodontomys microtinus, Liomys adspersus, 0ryzomys fulvescens, and 0. caliginosus; also, natural infection has been found in a coati (Nasua narica) and marmosets (Saguinus mystax). In a study carried out in Panama, the highest prevalence of the infection was found in the cotton rat S. hispidus, which was also the most abundant rodent in the six localities studied. The cotton rat inhabits areas close to dwellings in both tropical and temperate America. it is omnivorous, feeding on both plants and small vertebrate and invertebrate animals, including slugs (V. ameghini). All these facts indicate that the cotton rat is a prime reservoir and that it plays an important role in the epidemiology of the parasitosis. Rodents are infected by ingesting infected mollusks. Another probable source of infection is plants contaminated with mollusk secretions "slime") containing third-stage infective larvae of the parasite. The manner in which man contracts the infection is not well known. Infection probably occurs by ingestion of poorly washed vegetables containing small slugs or their secretions. It is believed that children can become infected while playing in areas where slugs are abundant by transferring snail secretions found on vegetation to their mouths. An increase in cases in children occurs in Costa Rica during the rainy season, when slugs are most plentiful. Humidity is an important factor in the survival of both the first- and third-stage larvae in the environment, since they are susceptible to desiccation. The parasite species in the Far East (A. cantonensis) has been found in at least ten different species of the genus Rattus and in Bandicota indica and Melomys littoralis. These rodents, natural definitive hosts, are infected by consuming mollusks or paratenic hosts that harbor third-stage larvae. The infection rate of the mollusks is usually high; both the prevalence and the number of larvae an individual mollusk can harbor vary according to the species. Man, who is an accidental host, is infected by consuming raw mollusks and also paratenic hosts such as crustaceans or fish. The ecology of angiostrongyliasis is closely related to the plant community, since it ultimately supports the appropriate mollusks and rodents. The frequency of the human parasitosis depends on the abundance of these hosts and the degree to which they are infected, and, also, in the case of A. cantonensis, on eating habits (consumption of raw mollusks, crustaceans, and fish).

DIAGNOSIS: The spinal fluid characteristically shows elevated protein and an eosinophilic pleocytosis. Occasionally, the parasite can be recovered from spinal fluid. Peripheral eosinophilia with a low-grade leukocytosis is common. A serologic test is available from the Centers for Disease Control and Prevention; its sensitivity and specificity are not established. CT and MRI may show a central nervous system lesion.

TREATMENT: No specific treatment is available; however, levamisole, albendazole, thiabendazole (25 mg/kg three times daily for 3 days), or ivermectin can be tried. Symptomatic treatment with analgesics or corticosteroids may be necessary. The illness usually persists for weeks to months, the parasite dies, and the patient then recovers spontaneously, usually without sequelae. However, fatalities have been recorded.

CONTROL: At least theoretically, angiostrongyliasis could be controlled by reducing rodent and mollusk populations. Preventive measures at the individual level consist of washing vegetables thoroughly, washing hands after garden or field work, not eating raw or undercooked mollusks and crustaceans, and not drinking water that may be unhygienic.


ANCYLOSTOMIASIS

(Uncinariasis, Necatoriasis, Hookworm Disease)

AGENT: The causative agents are mainly Necator americanus and Ancylostoma duodenale; occasionally A. ceylanicum and A. caninum.

RESERVOIR AND INCIDENCE A common worm infection of humans and domestic dogs and cats in various tropical and subtropical countries where disposal of human feces is inadequate. The reservoirs are humans, dogs, and cats.

TRANSMISSION: Adult worms, living in the small intestines of humans, produce eggs which pass on to the ground in feces. The eggs hatch and go through three larval stages. Human infection results from the third-stage larvae which survive in soil for several weeks in moist and warm conditions. Animals and man become infected by contact with infected soil, the larvae penetrating through skin or mucosa of the digestive tract. The parasites then migrate through the blood capillaries to the lung, eventually to be coughed up and swallowed. They reach maturity and complete their cycle in the intestines.

DISEASE IN ANIMALS: Factors such as the weight of infection and nutritional state of the animal are important. Loss of blood together with malnutrition produce anemia. Severe enteritis causes hemorrhagic diarrhea and weight loss from intestinal malabsorption. Prenatal infection of the dog causes death of the fetal pups. Mild infections generally cause no clinical signs.

DISEASE IN MAN: The condition is often asymptomatic. Self-limiting vesicular/pustular skin eruptions may appear at the site of larval entry. A. caninum does not penetrate human skin beyond the epidermis. With other species chronic symptoms due to iron deficiency anemia may occur. Rarely there is tracheitis and coughing due to lung infiltration with parasites.

DIAGNOSIS: Fecal flotation

TREATMENT: Pyrantel pamoate, mebendazole, albendazole, levamisole, or tetrachloroethylene.

PREVENTION/CONTROL: Educate the public to the dangers of soil contamination by feces. Wear shoes! Screen feces from persons and animals from endemic areas. Keep kennel floors dry and avoid feeding animals on the ground.


CAPILLARIASIS

AGENT: The causative agents are Capillaria hepatica (hepatic form), C. philippinensis (intestinal form) and C. aerophila (respiratory form).

RESERVOIR AND INCIDENCE C. hepatica and C. aerophila are very rare infections with isolated cases reported from North, Central and South America, Asia, and Europe. C. philippinensis is endemic in certain areas of the Philippines and cases have been reported from Thailand and Japan. Humans are the reservoir for C. philippinensis. With C. hepatica, rodents are the reservoirs. Cats and dogs are the reservoir for C. aerophila. Peromyscus maniculatus and Cletheronomys gapperi are the major hosts in North America.

TRANSMISSION: Humans are infected by eating raw fish containing infective larvae. The worm parasite lives in the intestines of humans and autoinfection occurs. Human feces contain large numbers of ova which contaminate watercourses and infect freshwater fish. Humans may be infected by the ingestion of ova in the soil also.

DISEASE IN ANIMALS: Infection with C. aerophila causes coughing, sneezing, and nasal discharge in dogs, cats, and foxes. Worms of C. hepatica mature and deposit eggs in liver tissue causing a local chronic inflammatory response in mice and rats.

DISEASE IN MAN: C. hepatica: acute and subacute hepatitis. The liver lesions consist of enlargements with foci of granulation tissue containing worms and ova. C. philippinensis: progressive weight and protein loss due to diarrhea and malabsorption. The jejunal villi are obliterated with massive accumulations of worms and ova. C. aerophila: fever and coughing. Worms present in epithelial lining of respiratory tract causing pneumonitis.

DIAGNOSIS: Liver or intestinal biopsy, necropsy, clinical signs & symptoms. Fecal exam for ova, larva, or adults.

TREATMENT: Albendazole, Thiabendazole, Mebendazole.

PREVENTION/CONTROL: Do not eat uncooked fish and other aquatic animal life in endemic areas. Control rodents and improve hygiene. Prevent pica. Sanitary disposal of feces.


FILARIASIS

(Brugiasis)

AGENT: The commonest causative agent is Wuchereria bancrofti, which is not zoonotic. Brugia malayi is zoonotic. Dirofilaria immitis may occasionally infect humans.

RESERVOIR AND INCIDENCE B. malayi has been identified in Malaya and the Philippines. D. immitis occurs in dogs in North and South America, Australia, India, the Far East and Europe. Wild monkeys and felines are the reservoir for B. malayi.

TRANSMISSION: Transmission to humans is via the bite of a mosquito vector (Mansonia and Anopheles). These, on biting humans, release microfilariae on to skin which enter the body through the puncture wound and pass via the lymphatics to lymph nodes. The worms are viviparous, producing microfilaria. These appear in the blood and reinfect the biting insect.

DISEASE IN ANIMALS: D. immitis is found in the right ventricle of dogs and in the pulmonary artery. Mild infection causes no signs, but long continued infection leads to cardiac insufficiency with ascites and passive congestion.

DISEASE IN MAN: B. malayi: Repeated bouts of fever, lymphadenopathy, lymphangitis and abscesses occur. This leads to lymphatic obstruction and massive lymphedema followed by fibrosis (so-called elephantiasis) especially in the legs. D. immitis: Initial symptoms include cough, chest pain, or hemoptysis. However, the filariae die in the human pulmonary vasculature and consequently nodules have been found in the skin or as solitary 1-2 cm "coin" lesions in the periphery of the lungs.

DIAGNOSIS: Diagnosis is established by finding microfilariae in the blood. Serologic testing is available but false-positive and false-negative reactions occur.

TREATMENT: Diethylcarbamazine

PREVENTION/CONTROL: Diethylcarbamazine or Ivermectin can be given as a prophylaxis. Control vectors and avoid their bites. Prophylactic treatment of dogs.


VISCERAL LARVAL MIGRANS

(Toxocariasis)

AGENT: Most cases of visceral larval migrans (VLM) are due to Toxocara canis, an ascarid of dogs and other canids, but in a few cases Toxocara cati in domestic cats has been implicated and rarely Baylisascaris procyonis of raccoons.

RESERVOIR AND INCIDENCE The reservoir mechanism for T. canis is latent infections in female dogs which are reactivated during pregnancy. Transmission from mother to puppies is via the placenta and milk. The life cycle of T. cati is similar, but transplacental transmission does not occur. Human infections are sporadic and occur worldwide.

TRANSMISSION: Infection is generally in dirt-eating young children who ingest T. canis or T. cati eggs from soil or sand contaminated with animal feces, most often from puppies. Direct contact with infected animals does not produce infection, as the eggs require a 3 to 4 week extrinsic incubation period to become infective; thereafter, eggs in soil remain infective for months to years. In humans, hatched larvae are unable to mature and continue to migrate through the tissues for up to 6 months. Eventually they lodge in various organs, particularly the lungs and liver and less often the brain, eyes, and other tissues, where they produce eosinophilic granulomas up to 1 cm in diameter.

DISEASE IN ANIMALS: The first indication of infection in young animals is lack of growth and loss of condition. Infected animals have a dull coat and often are "potbellied". Worms may be vomited and are often voided in the feces. In the early stages, pulmonary damage due to migrating larvae may occur; this may be complicated by bacterial pneumonitis, so that respiratory distress of variable severity may supervene. Diarrhea with mucus may be evident. In severe infections of puppies, verminous pneumonia, ascites, fatty degeneration of the liver, and mucoid enteritis are common. Cortical kidney granulomas containing larvae are frequent in young dogs.

DISEASE IN MAN: Migrating larvae induce fever, cough, wheezing; hepatomegaly, and sometimes splenomegaly and lymphadenopathy are present. The acute phase may last 2-3 weeks, but resolution of all physical and laboratory findings may take up to 18 months. Leukocytosis is marked due to eosinophils. Hyperglobulinemia occurs when the liver is extensively invaded. Ocular toxocariasis results in a eosinophilic granuloma of the retina that may be mistaken for retinoblastoma.

DIAGNOSIS: ELISA, no parasitic forms can be found by fecal exam.

TREATMENT: Thiabendazole, Mebendazole, or Ivermectin. Corticosteroids, antibiotics, antihistamines, and analgesics are given for symptomatic relief. Treatment for Ocular Toxocariasis includes the above plus vitrectomy and laser photocoagulation.

PREVENTION/CONTROL: Disease in humans is best prevented by periodic treatment of puppies, kittens, and nursing dogs and cats. Children should be supervised to prevent pica; their hands should be washed after playing in soil and sand; and play areas should be protected from animal feces.


CUTANEOUS LARVAL MIGRANS

(Creeping Eruption)

AGENT: Caused by the larvae of the dog and cat hookworms, Ancylostoma braziliense and Ancylostoma caninum. A number of other animal hookworms, gnathostomiasis, and strongyloidiasis are rarely also causative agents.

RESERVOIR AND INCIDENCE Cutaneous Larval Migrans is prevalent throughout the tropic and subtropics. Human infection is common in SE U.S., particularly where people come in contact with moist sandy soil (e.g., beaches, children's sand piles) contaminated by dog or cat feces.

TRANSMISSION: direct skin contact with larvae. soil to skin contact. contamination with animal feces.

DISEASE IN ANIMALS: Same as Ancylostomiasis.

DISEASE IN MAN: At the site of larval entry, particularly on the hands or feet, up to several hundred minute, intensely pruritic erythematous papules appear. Two to 3 days later, serpiginous eruptions appear as the larvae migrate at a rate of several millimeters a day; the parasite lies slightly ahead of the advancing border. The process continues for weeks or up to a year, and the lesions may remain severely pruritic, vesiculate, and become encrusted and secondarily infected. Without treatment, the larvae eventually die and are absorbed.

DIAGNOSIS: Presumptive - Characteristic clinical manifestations. Etiologic - ID of agent by biopsy in skin section but this is usually very difficult to achieve. Most cases are really not confirmed. No valid serodiagnostic tests currently available.

TREATMENT: Simple transient cases require no treatment. Albendazole or thiabendazole. Antihistamines and antibiotic ointments.

PREVENTION/CONTROL: Minimize contact (e.g., wear shoes!). Decontaminate environment: 10 lbs/ 100 sq ft. sodium borate - gravel/clay dog run; 1% sodium hypochlorite solution - cement dog run. Prevent environmental contamination. Public health education.


OESOPHAGOSTOMIASIS

(Nodular Intestinal Worm Infection)

AGENT: The causative agents are Oesophagostomum stephanostomum, bifurcum, and aculeatum.

RESERVOIR AND INCIDENCE The parasite lives in the intestines of various primates and sometimes humans. These definitive hosts can sometimes serve as intermediate hosts. It occurs mainly in Africa, but occasionally in South America and Asia.

TRANSMISSION: Eggs passed in feces release larvae which infect the definitive host on ingestion. The parasite invades the intestinal wall to form nodules. The fourth stage larvae which develop in these nodules migrate to the lumen of the large intestine to form the adult worm and complete the life cycle.

DISEASE IN ANIMALS: Mild infection is subclinical. Abdominal pain follows more severe infection with diarrhea or even dysentery. Death in the NHP may ensue from perforation of the intestine and peritonitis.

DISEASE IN HUMANS: Mild infection goes unnoticed but sometimes abdominal pain, and GI bleeding and even peritonitis occur. Granulomatous nodules in the intestinal wall contain larvae and parasites. These may be secondarily infected and lead to abscesses.

DIAGNOSIS: Fecal flotation. Culture to obtain larvae for species ID.

TREATMENT: Anthelmintic therapy.

PREVENTION/CONTROL: Fecal screening of NHP's Wearing of protective clothing and gloves. Ensure good personal hygiene. Sanitary disposal of feces is important.


STRONGYLOIDIASIS

AGENT: Strongyloidiasis is caused by infection with Strongyloides stercoralis.

RESERVOIR AND INCIDENCE The condition is an infection of humans, but dogs, cats, and primates have been found naturally infected. The disease is endemic in tropical and subtropical regions; although the prevalence is generally low, in some areas disease rates exceed 25%. In the USA, the disease is endemic in southern wet areas.

TRANSMISSION: The parasite is uniquely capable of maintaining its life cycle both within the human host and in soil. Infection occurs when filariform larvae in soil penetrate the skin, enter the bloodstream, and are carried to the lungs, where they escape from capillaries into alveoli and ascend the bronchial tree to the glottis. The larvae are then swallowed and carried to the duodenum and upper jejunum, where maturation to the adult stage takes place. The parasitic female, generally held to be parthenogenetic, matures and lives embedded in the mucosa, where its eggs are laid and hatch. Rhabditiform larvae, which are noninfective, emerge, and most migrate into the intestinal lumen to leave the host via the feces. The life span of the adult worm may be as long as 5 years. In the soil, the rhabditiform larva metamorphose into the infective (filariform) larvae. However, the parasite also has a free-living cycle in soil, in which some rhabditiform larvae develop into adults that produce eggs from which rhabditiform larvae emerge to continue the life cycle. Internal autoinfection takes place in the lower bowel when some rhabditiform larvae, instead of passing with the feces, develop into filariform larvae that penetrate the intestinal mucosa, enter the intestinal lymphatic and portal circulation, are carried to the lungs, and return to the small bowel to complete the cycle.

DISEASE IN ANIMALS: Young dogs and cats have thin skins which allow massive infection to penetrate, giving severe dermatitis, inappetence, coughing and even bronchopneumonia. Vomiting occurs, as does severe dermatitis during the period of larval penetration.

DISEASE IN MAN: Pruritic dermatitis is seen at sites of larval penetration. Diarrhea, epigastric pain, nausea, malaise, weight loss, coughing, rales, transient pulmonary infiltrates are also seen. In the hyperinfection syndrome, autoinfection is greatly increased, resulting in a marked increase in the intestinal worm burden and in massive dissemination of filariform larvae to the lungs and most other tissues, where they can cause local inflammatory reactions and granuloma formation. Severe diarrhea, bronchopneumonia, and ileus can result.

DIAGNOSIS: Diagnosis requires finding the larval stages in feces or duodenal fluid. ELISA and IFA serologic tests are being developed.

TREATMENT: Thiabendazole, albendazole, mebendazole, cambendazole, ivermectin, or levamisole.

PREVENTION/CONTROL: Rigid attention to hygienic habits, including use of footwear in endemic areas. Sanitary disposal of feces. Fecal exams of monkeys, dogs, and cats in quarantine.


TRICHOSTRONGYLOSIS

(Trichostrongyliasis, Trichostrongylosis)

AGENT: Several species of Trichostrongylus including Trichostrongylus axei, T. affinis, T. colubriformis, and many others. RESERVOIRS AND INCIDENCE: The reservoirs of most of the species of Trichostrongylus are domestic and wild herbivores. Trichostrongylids are very common parasites in domestic ruminants and their distribution is worldwide.

TRANSMISSION: The sources of infection are soil and vegetation, in which the eggs deposited with the animal host's feces develop in a few days to the infective larval stage. Man and animals are infected orally. Man acquires the infection mainly by consuming raw vegetables contaminated with the infective larvae. Another important factor in transmission is the preparation and use of animal manure as fuel.

DISEASE IN ANIMALS: In horses, these worms produce a chronic catarrhal gastritis and may result in weight loss. The lesions comprise nodular areas of thickened mucosa surrounded by a zone of congestion and covered with a variable amount of mucus. The lesions may be rather small and irregularly circumscribed, or may coalesce and involve most or all of the glandular portion of the stomach, and erosions and ulcerations may be seen. In ruminants, gastritis with superficial erosion of the mucosa, hyperemia, and diarrhea may result. Protein loss from the damaged mucosa and anorexia cause hypoproteinemia and weight loss.

DISEASE IN MAN: Most cases asymptomatic. Severe infections - diarrhea, blood in stool, abdominal cramps, and emaciation.

DIAGNOSIS:

Flotation and fecal culture (identification of larvae used to distinguish between species).

TREATMENT: Pyrantel pamoate, mebendazole, or levamisole.

PREVENTION/CONTROL: Regular deworming of animals. Preventive measures for humans consist of improved food, environmental, and personal hygiene. In endemic areas, raw vegetables or other foods should not be eaten. Pasture management is important. Most of the larvae die in a field left ungrazed for a month.


ASCARIASIS

(Roundworm infection, ascaridiasis) A common, but usually mild, roundworm infection of both humans and animals, occurring worldwide. There is doubt whether the worms cross between species. The causative agent in humans is usually Ascaris lumbricoides. Ascaris suum in pigs is considered occasionally a zoonotic infection (Nematoda). There is no vaccine.

RESERVOIR AND MODE OF TRANSMISSION: A. lumbricoides has its reservoir in humans and in contaminated soil. A. suum occurs in pigs. Eggs are passed in feces and become infective after two weeks. Infection is by the ingestion of eggs in soil or undercooked food. Eggs hatch in the gut, penetrate its wall and reach the lungs via the bloodstream. Larvae develop in the lungs, ascend the trachea and are swallowed and mature in the gut. Egg-laying begins 45-60 days after initial infection.

INCUBATION PERIOD: Humans. About two months. Animals. Development of A. suum in the pig is said to be quicker than in the human.

CLINICAL FEATURES: Humans. The condition is usually asymptomatic but there may be fever with asthma, spasmodic coughing and possibly pneumonitis. Abdominal pain, and even bowel or bile duct obstruction, is possible. Occasionally migrating larvae cause symptoms referable to the brain, eyes, kidneys and liver. Animals. Migrating larvae cause irregular breathing and coughing. Heavy infection gives abdominal pain with diarrhea. Suckling pigs are most affected.

PATHOLOGY: Humans. Eosinophilia and pulmonary eosinophilic infiltration occur. Liver abscesses and cholangitis are sometimes seen. Animals. Larvae migrating through the lung cause pneumonia. Worms in the intestine give enteritis.

DIAGNOSIS: Humans. Identify eggs or adult worms in feces. Larvae in sputum or gastric washings are diagnostic. Animals. Examine feces for eggs, or monitor worms passed rectally or via mouth and nose. An egg count of more than 1OOO/g of feces indicates clinical disease.

PROGNOSIS:

Humans. The condition is rarely fatal. Aberrant larvae in brain, eyes and kidney can give severe symptoms. Animals.Rarely fatal.

PREVENTION: Humans. Sanitary disposal of feces coupled with good personal and food hygiene are essential. Animals. Worm and wash sows before farrowing. Rear pigs on concrete or avoid close confinement on soil.

TREATMENT: Humans and animals. Pyrantel pamoate is the treatment of choice. Stools should be rechecked at 2 weeks and patients retreated until all ascarids are removed.

LEGISLATION: Humans and animals. None.


CESTODE ZOONOSES


DIPHYLLOBOTHRIASIS

(Fish tapeworm infection) A benign tapeworm infection of the small intestines caused by eating raw fish. The causative agents are Diphyllobothrium latum and D. pacificum (Cestoda). There is no vaccine. D. latum is common in northern temperate regions where the fish are eaten raw (e.g., in the Baltic countries, Finland and Canada/Alaska). D. pacificum is common in coastal South America, especially Peru.

RESERVOIR AND MODE OF TRANSMISSION: The definitive hosts of D. latum include humans, dogs and cats. For D. pacificum the natural reservoir is seals. Two intermediate hosts include a plankton crustacean and a freshwater fish. Gravid proglottids pass in the feces of the definitive host. The eggs hatch in lakes and waterways and then infect the crustacean. Freshwater fish consume these and the larvae encyst in the musculature. The fish, in their turn, may be eaten by larger fish which can still transmit the infection. Humans acquire the parasite by eating raw infected fish.

INCUBATION PERIOD: Humans. 3-6 weeks from ingestion to adult tapeworm. Animals. Unknown but presumably as for humans, in dogs and cats.

CLINICAL FEATURES: Humans. The condition is usually asymptomatic. Some patients develop vitamin B12 deficiency anemia. Massive infection may cause diarrhea and intestinal obstruction. Animals. No clinical signs are seen in dogs and cats. Heavy infection with larvae can kill the fish intermediate host.

PATHOLOGY: Humans. The presence of the large tapeworm, 3 to 10 meters long, in the intestine can cause mechanical obstruction. Megaloblastic anemia occurs owing to vitamin B12 deficiency. Animals. Subclinical. In fish, myositis and possibly even death occur in heavy infestation.

DIAGNOSIS: Humans and animals. Identify characteristic eggs in feces.

PROGNOSIS:

Humans. Usually benign. Animals. Usually benign, but heavy infection may be fatal to fish.

PREVENTION: Humans and animals. Dispose of feces hygienically. Educate for proper cooking of fish. Freeze fish or salt cure before marketing.

TREATMENT: Humans and animals. Anthelmintics, especially niclosamide and praziquantel.

LEGISLATION: Humans and animals. None.


SPARGANOSIS

SYNONYM: Larval diphyllobothriasis.

ETIOLOGY: The second larval stage (plerocercoid or sparganum) of the pseudophyllidean cestode of the genus Spirometra (Diphyllobothrium, Lueheela). Several species of medical interest have been described: Spirometra mansoni, S. mansonoides, S. erinacei-europaei, S. theileri, and S. proliferum. These are the most commonly accepted species at the present time, but it should be noted that they are difficult to differentiate and that the taxonomy remains in doubt. The definitive hosts are mainly domestic and wild canids and felids. The development cycle requires two intermediate hosts. The first is a copepod (planktonic crustacean) of the genus Cyclops. which ingests coracidia (free, ciliated embryos) that develop from Spirometra eggs when they reach the water with the feces of dogs or cats (definitive hosts). In the tissues of the copepod, the coracidium turns into the first larva, or procercoid. When a second intermediate host ingests an infected copepod, the procercoid develops into a second larval form, the plerocercoid or sparganum. The plerocercoid larva can be harbored by many vertebrates, including amphibians, reptiles, birds, small mammals (rodents and insectivores), man, nonhuman primates, and swine. Fish do not become infected. Some researchers believe that the second intermediate host is usually an amphibian, but can vary according to region. Numerous species of vertebrates become infected with plerocercoids by feeding on amphibians, but they may also develop plerocercoids after ingesting water containing copepods infected by procercoids (first larva). Several animal species that are not definitive hosts function as paratenic or transport hosts, since the larvae they acquire by feeding on animals infected with plerocercoids encyst again after passing through the intestinal wall and migrating to other tissues. This transfer process is undoubtedly important in the life cycle; but the fact that many species of secondary hosts can be infected directly by ingestion of copepods containing procercoids is probably no less important. When the sparganum reaches the intestine of the definitive host, it attaches to the mucosa; in 10 to 30 days, it matures into an adult cestode, completing the cycle. The adult S. mansonoides reaches about 25 cm in length in the intestine of the definitive hosts (cat, dog). The sparganum found in the tissues of the secondary intermediate hosts and paratenic hosts, including man, varies from 4 to 10 cm in length.

GEOGRAPHIC DISTRIBUTION AND OCCURRENCE: Sparganosis is found throughout the world, but human infection is not common. Just over 450 cases are known, mostly from Japan, China, Korea, and Southeast Asia. In the United States, about 60 cases have been described# in Latin America and the Caribbean, the disease has been recorded in Uruguay, Ecuador, Colombia, Venezuela, Guyana, Belize, and Puerto Rico; about 30 cases have been diagnosed in Africa. Infections in the Far East are attributed to plerocercoid larvae of Spirometra mansoni: in the United States, to S. mansonoides; in Europe, to S. erinacei-europaei; and in Africa, to S. theileri. However, as has already been mentioned, species identification can be difficult and therefore uncertain.

THE DISEASE IN MAN: The incubation period, determined in a study of 10 patients who ate raw frog meat, lasts from 20 days to 14 months. The most common localizations of the sparganum are subcutaneous connective tissue and superficial muscles. The lesion is nodular, develops slowly, and can be found on any part of the body. The main symptom is pruritus, sometimes accompanied by urticaria. The lesion is painful when there is inflammation. The patient may feel discomfort when the larva migrates from one location to another. The subcutaneous lesion resembles a lipoma, fibroma, or sebaceous cyst. Ocular sparganosis occurs mainly in Vietnam, Thailand, and parts of China. Its main symptoms consist of a painful edema of the eyelids, with lacrimation and pruritus. A nodule measuring I to 3 cm forms after 3 to 5 months, usually on the upper eyelid. Migration of the sparganum to internal organs can give rise to the visceral form of the disease. The preferred localizations are the intestinal wall, perirenal fat, and mesentery; vital organs are rarely affected. When the plerocercoid invades the lymphatic system, it produces a clinical picture similar to that of Eosinophils are abundant in the was near the parasite; examination of blood samples reveals mild leukocytosis and increased eosinophilia. An infrequent but serious form is proliferative sparganosis caused by Spirometra proliferum. The sparganum of S. proliferum is pleomorphic, with irregular branches and proliferative buds that detach from the larva and migrate to different tissues in the host, where they repeat the process and invade other organs. The life cycle of S. proliferum is not known.

THE DISEASE IN ANIMALS: The adult cestode, which lodges in the intestine of the definitive host, generally does not affect the health of the animal. In cats, however, it may produce weight loss, irritability, and emaciation, together with an abnormal or exaggerated appetite. Infection by the larvae or spargana can be clinically apparent when their number is large and especially when they invade vital organs. In the intermediate host, the disease is almost always asymptomatic if the number of parasites is relatively small.

SOURCE OF INFECTION AND MODE OF TRANSMISSION: Sparganosis is maintained in nature primarily by contamination of natural or artificial bodies of water (lagoons, marshes, lakes, and others) with feces from felids and canids infected with Spirometra spp. Contamination of water with eggs of Spirometra spp. leads to the infection of copepods and, consequently, of the second intermediate hosts that ingest these crustaceans. An important means of infection is the transfer of the second larva (sparganum, plerocercoid) from one secondary host to another, which increases the number of animal species and individuals infected. The common route of infection is ingestion; various mammal and bird species become infected by feeding on parasitized frogs or snakes. The high rate of infection in wild pigs in Australia may be due to this mechanism, although it may also stem from ingestion of copepods with drinking-water from lagoons. In any case, contamination of the water by wild canids (definitive hosts) that share the habitat assures that the cycle is perpetuated. The infection rate in man is low, compared to the rate in other animals. Man acquires sparganosis mainly by ingesting larvae contained in raw or undercooked meat of animals infected with spargana, such as amphibians, reptiles, birds, and wild mammals. Another mode of infection, also by larval transfer, is by contact. In Vietnam and Thailand, frogs are popularly believe to have an antiphlogistic effect, and their muscles are applied as poultices. This custom is responsible for ocular sparganosis. It is also probable that man can acquire sparganosis via drinking-water by ingesting copepods infected with procercoids (first larvae). Man is an accidental host and does not usually play any role in the life cycle of the parasite. However, under ecologic conditions in some regions of central Africa, it is suspected that man acts as an intermediate host. In this region, hyenas are the definitive hosts of Spirometra, and man is apparently the only host infected with spargana. In these circumstances, the infection cycle is maintained as a result of tribal custom of letting hyenas devour human corpses.

DIAGNOSIS: Specific diagnosis can be made only by removing the nodular lesion and confirming the presence of the plerocercoid. Attempts have been made to identify the species of Spirometra larvae by infecting dogs and cats via the digestive route. For reasons already mentioned, differentiation of species has proven difficult. Diagnosis in definitive hosts infected with adult cestodes can be made by coprologic examination or autopsy.

CONTROL: Human sparganosis can be prevented by avoiding ingestion of contaminated water that has not been treated, and by making sure that meat that might contain spargana is sufficiently cooked. In the Far East, public health education should emphasize the danger of using the tissue of frogs or other cold-blooded animals for medicinal purposes.


BERTIELLIASIS

AGENT: o Bertiella studeri and mucronanta, anoplocephalid tapeworms. o 260-300mm length tapeworm with 10mm width. o Gravid segments shed off in groups of about 20.

RESERVOIRS AND INCIDENCE: NHP's.

TRANSMISSION: NHP's acquire the parasitosis by ingesting mites infected with cysticercoid larvae. Man can become infected accidentally by ingesting food containing the mites.

DISEASE IN NHP'S: asymptomatic.

DISEASE IN HUMANS: The infection is usually asymptomatic, but cases with recurrent abdominal pain, vomiting, anorexia, constipation, and intermittent diarrhea have been observed.

DIAGNOSIS: Diagnosis is based on microscopic observation of the proglottids eliminated in the feces. The egg possesses a characteristic pyriform apparatus.

TREATMENT: Dichlorophen

PREVENTION/CONTROL: Control mites.


DIPYLIDIASIS

AGENT: Dipylidium caninum - common intestinal cestode of dogs. Gravid proglottids are "pumpkinseed" shaped.

RESERVOIRS AND INCIDENCE: Dogs, cats, and their wild counterparts. Arthropods serve as intermediate hosts. These include the dog flea (Ctenocephalides canis), the cat flea (C. felis), and dog louse (Trichodectes canis). Cosmopolitan including the U.S.

TRANSMISSION: Humans, dogs, and cats are infected by ingestion of arthropod intermediate hosts which harbor the cysticercoid larvae.

DISEASE IN ANIMALS: Usually no severe pathology. Possibly mild digestive problems or perianal pruritus.

DISEASE IN MAN: Slight symptoms, if any. Mild weight loss, perianal itching, diarrhea, vague abdominal pain.

DIAGNOSIS: Recovery of gravid proglottids that are passed in the feces or that crawl out of the anus.

TREATMENT: Niclosamide or praziquantel.

PREVENTION/CONTROL: Screen animals. Treat infected animals. Eliminate ectoparasites. Teach proper handling of pets to children.


ECHINOCOCCOSIS

(Hydatidosis, Hydatid Disease)

AGENT: Echinococcus granulosis - causes "cystic" disease. Echinococcus multilocularis - causes "alveolar" disease. E. vogeli - causes polycystic disease.

RESERVOIR AND INCIDENCE The definitive host for E. granulosis is a carnivore (all of which, except for the lion, are Canidae) that harbors the adult tapeworm in the small intestine. Human infection with E. granulosus occurs principally where dogs are used to herd grazing animals, particularly sheep. The disease is common throughout southern S. America, the Mediterranean and Middle East, central Asia, and East Africa. Foci of endemicity are in eastern Europe, Russia, Australasia, India, and the UK. In North America, endemic foci have been reported from the western USA, the lower Mississippi Valley, Alaska, and northwestern Canada. The life cycle for E. multilocularis involves foxes as definitive host and microtine (e.g., voles and meadow mice) rodents as intermediate host. Domestic dogs and cats can also become infected with the adult tapeworm when they eat infected wild rodents. The disease in humans has been reported in parts of central Europe, much of Siberia, northwestern Canada, and western Alaska. One case has been reported in Minnesota. The principle definitive host for E. vogeli is the bush dog; the main intermediate hosts are the paca and spiny rat. Domestic hunting dogs are also definitive hosts, and serve as an important source of human infection. Cases have been reported in South America.

TRANSMISSION: E. granulosis: Human infection occurs when eggs passed in dog feces are accidentally swallowed. E. multilocularis: Human infection is by accidental ingestion of tapeworm eggs passed in fox or dog feces.

DISEASE IN ANIMALS: Usually no clinical signs except for enteritis in heavy infestations. In sheep, hydatid cysts cause considerable condemnation of meat and loss of production.

DISEASE IN MAN: E. granulosis - Cystic hydatid disease A liver cyst may remain silent for 10-20 years or more until it becomes large enough to be palpable, to be visible as an abdominal swelling, to produce pressure effects, or to produce symptoms due to leakage or rupture. There may be right upper quadrant pain, nausea, and vomiting. The effects of pressure may result in biliary obstruction. If a cyst ruptures, anaphylaxis and death may result. If fluid and hydatid particles escape slowly, allergic manifestations may result. Rupture can occur into the pleural, pericardial, or peritoneal space or into the duodenum, colon, or renal pelvis. Dissemination of germinal elements may be followed by the development of multiple secondary cysts. Pulmonary cysts cause no symptoms until they leak; become large enough to obstruct a bronchus, or erode a bronchus and rupture. Brain cysts produce symptoms earlier and may cause seizures. Cysts in the bone marrow may present as pain or spontaneous fracture. The bones most often affected are the vertebrae and paraplegia may develop due to compression of the spinal cord. 20% of patients have multiple cysts. 15% of untreated patients eventually die. E. multilocularis - Alveolar disease The primary localization of alveolar cysts is in the liver, where they may extend locally or metastasize to other tissues. The larval mass has poorly defined borders and behaves like a neoplasm; it infiltrates and proliferates indefinitely by exogenous budding of the germinative membrane, producing an alveolus-like pattern of microvesicles. 90% of untreated cases die within 10 years. E. vogeli - Polycystic form of human hydatid disease. Symptoms are variable according to cyst size and location. The polycystic hydatid is unique in that the germinal membrane proliferates externally to form new cysts and internally to form septi that divide the cavity into numerous microcysts. Brood capsules containing many protoscolices develop in the microcysts.

DIAGNOSIS: Immunoblot assay.

TREATMENT: Currently the definitive treatment is surgical removal of cysts. Newly available chemotherapy (albendazole or mebendazole) may alter this position.

PREVENTION/CONTROL: In endemic areas, prevention is by prophylactic treatment of pet dogs with praziquantel and prevention of feeding dogs offal.


HYMENOLEPIS DIMINUTA

(Rat Tapeworm Infection)

AGENT: Hymenolepis diminuta is the common tapeworm of rodents. Size varies from 100-600 mm.

RESERVOIR AND INCIDENCE Rodents, many arthropods (fleas, beetles, and cockroaches) serve as intermediate hosts.

TRANSMISSION: Rodents and humans are infected by accidentally swallowing the infected arthropods, usually in cereals or stored products.

DISEASE IN ANIMALS: Mild catarrhal enteritis with diarrhea occurs if the infection is heavy.

DISEASE IN MAN: Light infections are generally asymptomatic. Heavy infections may cause diarrhea, abdominal pain, anorexia, vomiting, weight loss, and irritability, particularly in young children.

DIAGNOSIS: Infections are diagnosed by finding characteristic eggs in feces; proglottids are usually not seen.

TREATMENT: Niclosamide or praziquantel.

PREVENTION/CONTROL: Eliminate rodents and insects in facilities.


HYMENOLEPIS NANA

AGENT: Dwarf tapeworm, Hymenolepis nana - measures 5-90 mm long.

RESERVOIRS AND INCIDENCE: The animal reservoir is the house mouse, but humans can be both definitive and intermediate hosts. Worldwide occurrence in warm climates.

TRANSMISSION: Gravid proglottids disintegrate and eggs pass in the feces and may be ingested by another human. Larvae then develop in the intestinal villi and pass to the lumen of the gut to become the adult forms. Dogs, cats and their fleas can be infected as well as grain beetles which can serve as intermediate hosts.

DISEASE IN ANIMALS: Same as H. diminuta.

DISEASE IN MAN: Same as H. diminuta.

DIAGNOSIS: Same as H. diminuta.

TREATMENT: Same as H. diminuta.

PREVENTION/CONTROL: Personal hygiene, protective clothing and gloves Vermin control. Protect stored grains and feeds from grain beetles.

TREMATODE ZOONOSIS


SCHISTOSOMIASIS

(Bilharzia, Bilharziasis)

AGENT: Schistosomiasis infects more than 200 million persons worldwide. The causative agents are Schistosoma mansoni, haematobium, and japonicum.

RESERVOIRS AND INCIDENCE: Humans are the reservoir for S. mansoni and haematobium. S. japonicum infects cattle, water buffalo, horses, dogs, cats, rodents and monkeys. Intermediate hosts are species of snails (Biomphalaria and Bulinus). S. mansoni occurs in Africa, South America and some Caribbean islands (including Puerto Rico); S. haematobium in Africa and the Middle East; and S. japonicum in China, Japan, the Philippines, and South East Asia.

TRANSMISSION: Cercariae in contaminated water penetrate human skin, especially in irrigated fields or rivers. In the body the parasite migrates via the liver to the superior mesenteric vein where maturation takes place in about 6 weeks. Eggs are disseminated throughout the body via the blood, released into the intestinal lumen and excreted. In water miracidia develop and penetrate the snail, which in turn excretes cercariae into the water.

DISEASE IN ANIMALS: Abdominal pain, diarrhea, anemia, and emaciation occur. Cattle have hematuria.

DISEASE IN HUMANS: Penetration of larvae through the skin causes an itchy rash. With heavy infection, penetration of the parasite through the skin gives rise to local dermatitis and pruritus followed by pneumonitis when the parasites reach the lung. The deposition of ova provokes the growth of small multiple granulomata throughout the body. Eventually intestinal and hepatic fibroses develop. With S. japonicum, acute symptoms include fever, abdominal pain, cough, weight loss, diarrhea and dysentery. Chronic infection may result in symptoms months to years later, with enlarged liver and spleen, cirrhosis, ascites, and fits due to cerebral involvement.

DIAGNOSIS: Definitive diagnosis is made by finding the characteristic eggs in excreta or by mucosal or liver biopsy. Screening for infection is possible by skin or serologic tests, but neither is sufficiently sensitive or specific to justify treatment.

TREATMENT: Praziquantel, metrifonate, or oxamniquine.

PREVENTION/CONTROL: Dispose of feces and urine so that viable eggs will not reach bodies of fresh water containing intermediate snail hosts. Reduce snail habitats by removing vegetation or by draining and filling. Treat snail-breeding sites with molluscicides. Prevent exposure to contaminated water (e.g., wear rubber boots and gloves). To minimize cercarial penetration, towel dry, vigorously and completely, skin surfaces wet with suspected water. Apply 70% alcohol immediately to the skin to kill surface cercariae. Provide water for drinking, bathing, and washing clothes from sources free of cercariae. Rapid treatment of patients to prevent disease progression and to reduce transmission by reducing egg passage.

REFERENCES

Acha, P.N. and Szyfres, B. 1987. Zoonoses and communicable diseases man and animals. Pan American Health Organization, Washington, DC. August, J.R. 1992. Zoonotic complications of bite and scratch wounds. Proceedings of the 10th ACVIM Forum. 665-667. Bell, J.C., Palmer, S.R., and Payne, J.M. 1988. The zoonoses: infections transmitted from animals to man. Edward Arnold Press, London, UK. Benenson, A.S. 1990. Control of Communicable Diseases in Man (15th ed.). American Public Health Association, Washington, DC. Cho, S.N. 1990-91. Contemporary approaches to the serology of tuberculosis. Bull. Int. Union Tuberc. Lung Dis. 66 Suppl:59-62 Chomel, B.B. 1992. Zoonoses of house pets other than dogs, cats, and birds. The Pediatric Infectious Disease Journal. 11(6):479-487. Fox, J.G. and Lipman, N.S. 1991. Infections transmitted by large and laboratory animals. Infectious Disease Clinic of North America. 5(1):131-161. Fox, J.G., Newcomer, C.E., and Rozmiarek, H. (1984). Selected zoonoses and Other Health Hazards. In "Laboratory Animal Medicine" (J.G. Fox et.al.), pp. 613-648. Academic Press, Orlando. Fraser, C.M., Bergeron, J.A., Mays, A., and Aiello, S.E. 1991. The Merck Veterinary Manual (7th ed.). Merck and Co., Rahway, NJ. Goldstein, E.J. 1989. Management of human and animal bite wounds. J Am Acad Dermatol. 21(6):1275-9. Goldstein, E.J. 1991. Household pets and human infections. Infectious Disease Clinics of North America. 5(1):117-130. Harris, J.M. 1991. Zoonotic diseases of birds. Veterinary Clinics of North America: Small Animal Practice. 21(6):1289-1298. Holmes, G.P., Chapman, L.E., Stewart, J.A., et al. 1995. Guidelines for the prevention and treatment of B-virus infections in exposed persons. Clinical Infectious Diseases. 20:421-439. Irving, G.W.(1974). Selected Topics in Laboratory Animal Medicine: Zoonoses of Primates. In "Aeromedical Review" (USAF School of Aerospace Medicine), PP. 1-75. AFSC, Brooks Air Base, TX. Lappin, M.R.. 1993. Feline zoonotic diseases. Veterinary Clinics of North America: Small Animal Practice. 23(1):57-79. Madigan, J.E. 1992. Lyme disease update. Proc. 10th ACVIM Forum. 692-8. Marx, M.B. 1991. Parasites, pets, and people. Primary Care. 18(1):153-165. Meslin, F.X. 1992. Surveillance and control of emerging zoonoses. World Health Stat Q. 45(2-3):200-207. 1991. Tuberculosis increasing in the United States. Stat Bull Metrop Insur Co. 72(3):10-18. 1992. Nonhuman sources of leprosy. International Journal of Leprosy. 60(3):477-480. Schnurrenberger, P.R., and Hubbert, W.T.(1981). An Outline of the Zoonoses, pp. 1-157. Iowa State University Press, Ames. Starke, J.R. 1989. Prevention of tuberculosis. Semin Respir Infect. 4(4):318-25. Steele, J.H. (1979). Handbook Series in Zoonoses. Section A: Bacterial, Rickettsial, and Mycotic Diseases (Vol I), CRC Press, Boca Raton. Steele, J.H.(1980). Handbook Series in Zoonoses. Section A: Bacterial, Rickettsial, and Mycotic Diseases (Vol II), CRC Press, Boca Raton. Viral Steele, J.H.(1981). Handbook Series in Zoonoses. Section B: Viral Zoonoses (Vol I), CRC Press, Boca Raton. Viral Steele, J.H.(1981). Handbook Series in Zoonoses. Section B: Viral Zoonoses (Vol II), CRC Press, Boca Raton. Steele, J.H.(1982). Handbook Series in Zoonoses. Section B: Parasitic Zoonoses (Vol I), CRC Press, Boca Raton. Steele, J.H.(1982). Handbook Series in Zoonoses. Section B: Parasitic Zoonoses (Vol II), CRC Press, Boca Raton. Steele, J.H.(1982). Handbook Series in Zoonoses. Section B: Parasitic Zoonoses (Vol III), CRC Press, Boca Raton. Tappero, J.W., Mohle-Boetani, J., Koehler, J.E., et al. 1993. The epidemiology of bacillary angiomatosis and bacillary peliosis. JAMA. 269(6): 770-775. Tierney, Jr., L.M., McPhee, S.J., Papadakis, M.A., and Schroeder, S.A. 1995. Current Medical Diagnosis and Treatment. Appleton and Lange, Norwalk, Connecticut. Torres-Anjel, M.J. 1992. Macroepidemiology of the HIVs-AIDS (HAIDS) pandemic. Ann NY Acad Sci. 653:257-73. Wood, D.H.(1973). Selected Topics in Laboratory Animal Medicine: Zoonoses of Nonprimates. In "Aeromedical Review" (USAF School of Aerospace Medicine), PP. 1-28. AFSC, Brooks Air Base, TX




References

Collins, CH. 1993. Laboratory-Acquired Infections. Elsevier Publishers, Boston, MA.

Chomme, BB. 1992. Zoonoses of house pets other than dogs, cats, and birds. The Pediatric Infectious Disease Journal. 11(6):479-87.

Cooper, JE. 1977. Diseases of lower vertebrates and biomedical research. Laboratory Animals. 11(2):119-23.

Guide for the Care and Use of Laboratory Animals. 1985. National Institutes of Health, Bethesda, MD.

Fox JG and NS Lipman. 1991. Infections transmitted by large and small laboratory animals. Infectious Disease Clinics of North America. 5(1):131-63.

Hubbert, WT, WF McCulloch, and PR Schnurrenberger. 1975. Diseases Transmitted From Animals to Man. Charles C. Thomas Publishers, Springfield, IL. Richmond, JY, CA Dykewicz, LM Aldeman, et al. Working safely with LCMV-infected immunocompromised animals. Lab Animal. May, 1994:25-28.

Schlossberg, D. 1994. Infections of Leisure. Springer-Verlag, New York, NY.

Telford, SR, RJ Pollack, and A Spielman. 1991. Emerging vector-borne infections. Infectious Disease Clinics of North America. 5(1):7-17.

Text Modified from Document Created by Michael S. Rand, DVM, ACLAM


Source

http://packman.ianszoolinks.com/ponyzoohealth.htm