Advances in Animal and Veterinary Sciences Review Article

Zoonotic Pathogens Transmitted from Equines: Diagnosis and Control 1* 2 3 Sandip Kumar Khurana , Kuldeep Dhama , Minakshi Prasad , Kumaragurubaran 4 5 Karthik , Ruchi Tiwari 1National Research Centre on Equines, Hisar, 125 001, Haryana, India; 2Division of Pathology, 4Divison of Bac- teriology and Mycology, Indian Veterinary Research Institute, Izatnagar, Bareilly, 243 122, Uttar Pradesh, In- dia; 3Department of Biotechnology, College of Veterinary Sciences, LUVAS, Hisar, 125 004, Haryana, India; 5De- partment of Veterinary Microbiology, College of Veterinary Science and Animal Husbandry, Uttar Pradesh Pandit Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwa Vidyalaya Evam Go-Anusandhan Sansthan (DUVASU), Mathura (UP) – 281001. Abstract | The diseases of equines and other animal species that are shared between animals and humans come under the category of zoonotic diseases, and always put threat to veterinarians, ani- mal handlers, animal health personnel and general public. These pose a greater threat to pregnant women, infants, children, immunocompromised and old persons, individuals with stress of antibiotic therapy, and other susceptible humans. Equines also play an important role in transmitting several zoonotic diseases causing human illnesses such as those caused by encephalitic alphaviruses, hen- dravirus, West Nile virus and equine rabies, , glanders, , methicillin resistant Staphylococcus aureus (MRSA) , and Rhodococcus equi , therefore acts as substantial global health threat. Among these zoonotic threat agents, anthrax and glanders are also potential biological weapons and at times have been used as bio-terroristic agents also. The emer- gence and re-emergence of equine zoonotic pathogens have been observed from time to time. Anti- biotic resistance is a hot topic around the globe at present and certain strains like MRSA, extended spectrum beta lactamase producing Enterobacteriacea can also be transmitted between horses and humans. These drug resistant strains pose greater threat to human beings. Rapid detection of the causative agents of , close attention to personal hygiene, identification of potential fomites and vectors, and the use of protective clothing, newer therapeutics and vaccines may contribute to reduce the risk of zoonoses. Equines are used for antivenom and antitoxin production against various antigens, therefore the serum being obtained from equines should be properly screened for various pathogens either by serological methods or molecular assays which are specific for detection of zo- onotic agents. The present review discusses several important aspects of zoonotic diseases of equines with special focus on the recent advances in their diagnosis and control. Keywords | Equine zoonotic diseases, Diagnosis, Control

Editor | Ruchi Tiwari, College of Veterinary Sciences, Department of Veterinary Microbiology and Immunology Uttar Pradesh Pandit Deen Dayal Upadhayay Pashu Chikitsa, Vigyan Vishvidhyalaya Evum Go-Anusandhan Sansthan (DUVASU), Mathura (U.P.) – 281001, India. Special Issue | 2 (2015) “Reviews on Trends and Advances in Safeguarding Terrestrial /Aquatic Animal Health and Production” Received | January 20, 2015; Revised | February 10, 2015; Accepted | February 12, 2015; Published | February 16, 2015 *Correspondence | Sandip Kumar Khurana, National Research Centre on Equines, Hisar, Haryana, India; Email: [email protected] Citation | Khurana SK, Dhama K, Prasad M, Karthik K, Tiwari R (2015). Zoonotic pathogens transmitted from equines: diagnosis and control. Adv. Anim. Vet. Sci. 3(2s): 32-53. DOI | http://dx.doi.org/10.14737/journal.aavs/2015/3.2s.32.53 ISSN (Online) | 2307-8316; ISSN (Print) | 2309-3331

Copyright © 2015 Khurana et al. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 32 Advances in Animal and Veterinary Sciences INTRODUCTION highly contagious infection and thus can be a suitable candidate of also under certain circum- oonotic diseases, naturally transmitted between stances. Veterinarians have a key role in revealing var- vertebrate animals and man, are highly variable ious emerging zoonotic infections due to their close Zon the basis of their severity and transmissibility. The involvement with both animals and owners. Growing “one medicine concept” involving a convergence of an- possibilities of exploring zoonotic pathogens as bio- imal, human and environmental science professionals terrorism agents is another worry which reflects the for prevention, control and eradication of cross-spe- main functioning of veterinarians in early detection of cies disease transmission is gaining momentum, where bioterrorism-associated outbreaks of zoonotic diseas- zoonoses has assumed central position (Dhama et al., es. Antivenom are commonly produced from equine 2013a; Mukarim et al., 2015; Plowright et al., 2015). serum, so critical attention should be given during The emergence and re-emergence of zoonotic diseas- this time as this can be a major source for the trans- es poses a greater threat to pregnant women, infants mission of dangerous pathogens to human (Lalloo and children, immunocompromised and old persons, and Theakston, 2003; Theakston et al., 2003). Infer- persons under antibiotic therapy stress, veterinarians, ence accredited to equine zoonoses depends on prev- animal handlers and animal health personnel (Stull et alence of particular disease along with its case-fatality al., 2012). rate in human population. Encephalitic alphaviruses, hendravirus, equine rabies, salmonellosis, glanders, Zoonoses constitute nearly 60% of all known human anthrax, MRSA infection, brucellosis and Rhodococcus infections and over 75% of all emerging pathogens. equi are among the very important zoonoses. Weese They are caused by a diverse group of microorganisms (2002) has reviewed the risk of zoonotic diseases to and infectious syndromes caused by zoonotic patho- veterinary practitioners with emphasis on occupation- gens are equally diverse. Classification of zoonoses is al aspects. The present review covers several important broadly based on the nature of the pathogen, animal diseases which may be transmitted through equines host, severity of disease and mode of transmission be- to man or vice-versa with emphasis on their epide- tween animals to humans. Zoonoses have major im- miology, diagnosis and control calling for emergent plications on economics, labour and health produc- need of collaborative approach of medical doctors, tivity globally. Preventing and controlling zoonoses horse breeders, veterinarians, other governmental and is even more critical today in the context of globali- non-governmental agencies for early preparedness; zation of international trade, changes in agricultural however there could be several others which have not practices and global warming (Dhama et al., 2013b). been described here. They are responsible for affecting productivity of both humans and animals severely, thus contributing to ag- ZOONOTIC PATHOGENS gravation of poverty. TRANSMITTED FROM EQUINES VIRAL PATHOGENS Equines are used for various purposes like riding, rac- ing, sports, draught, transport, ceremonies, antitox- Encephalitic Alphaviruses in/antibody production, etc., throughout the world Western equine encephalitis virus (WEEV), eastern (Burnouf et al., 2004). There remains a contact be- equine encephalitis virus (EEEV) and Venezuelan tween the human and these equines at various stages equine encephalitis virus (VEEE) are common zo- which pave way for the spread of various diseases of onotic encephalitic alphaviruses. These have been iso- equines to human and hence human beings may ac- lated from horses, humans, mosquitoes, birds, rodents quire equine zoonotic bacterial, viral or other infec- and some other animals also. These are vector borne tious diseases either directly or by indirect means. The and transmitted through blood sucking arthropods, number of diseases affecting horses and other mem- thus considered as arboviral infection and condition bers of the Equidae family carries zoonotic potential is referred as Arboviral Encephalitis including EEE, including viral, bacterial, rickettsial, anaplasma asso- WEE, VEE and WNV as a cause of encephalitis in ciated, fungal and parasitic infections, which will be horse (Strauss et al., 1995; Smith et al., 1997; Kapoor elaborated through this review. Zoonotic diseases may et al., 2010). Ordinarily arboviruses are not directly occur as mild transient infection to severe lethal or transmitted from horses to humans under usual cir- NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 33 Advances in Animal and Veterinary Sciences cumstances, though aerosolization could be one pos- WEEV infection is done by ELISA, haemagglutina- sible risk factor. As per the recommendations of the tion-inhibition and neutralization assay (Martin et Centers for Disease Control and Prevention (CDC), al., 2000). RT-PCR has also been developed for its suspected clinical specimens such as cerebrospinal flu- diagnosis (Linssen et al., 2000; Lambert et al., 2003). id and serum must be handled under level 2 biocon- Formalin inactivated vaccine is used in horses as dou- tainment facility equipped laboratories with limited ble vaccine with EEEV. entrance. Lundstrom (2014) has reviewed progress in alphavirus vector development and vaccine technolo- Venezuelan Equine Encephalitis gy which enabled clinical trials in humans. Virus (VEEV) Infection VEEV is propagated in nature in enzootic cycle Eastern Equine Encephalitis between rodents and mosquito vector (Taylor and Virus (EEEV) Infection Paessler, 2013; Carossino et al., 2014; Go et al., 2014). EEEV was first isolated from horses in 1933 (Gilt- Epidemics occur when mosquitoes transmit the vi- ner and Shahan, 1933; TenBroeck and Merrill, 1933). rus to humans and equids (Tigrett and Downs, 1962; The virus is propagated in nature between birds and Walton et al., 1973). Horses, donkeys and have mosquitos. The virus is considered to be most viru- sufficient viremia and are able to transmit the infec- lent among the alphaviruses causing encephalitis tion through mosquitoes (Young, 1972; Mackenzie et with case-fatality rate of up to 70% in human beings al., 1976). Some reports suggest that humans are also (Zacks and Paessler, 2010). EEEV infections has able to transmit the infection especially in urban set- greater significance American continent where it was tings (Watts et al., 1997, 1998; Morrison et al., 2008). responsible for more than 180 cases in human (Agu- Madsen et al. (2014) demonstrated that inhibition of ilar et al., 2007). Mosquito vectors have a role in the Ago2, an important component of RNA-induced si- transmission of the virus form horses from human. lencing complex (RISC), resulted in decreased repli- The incubation period is 4-10 days, fever, headache, cation of encephalitic alphaviruses and thus may be vomiting, respiratory distress, seizures and coma may a future therapeutic. A live attenuated vaccine TC83 occur in human encephalitis cases. The mortality rate is thought to available option for humans as well as in horses is higher than in WEEV infection. Diagno- horses (Engler et al., 1992). However, formalin inac- sis is done by ELISA, haemagglutination-inhibition, tivated whole viral vaccines are also available (Tay- neutralization assay and virus isolation. Formalin in- lor and Paessler, 2013). DNA based vaccines are also activated vaccine is used in horses as double vaccine available which can be produced rapidly and are cost with WEEV. Honnold et al. (2014) demonstrated effective (Dupuy et al., 2011; Tretyakova et al., 2013; that chimeric live-attenuated EEEV vaccine candi- Carossino et al., 2014). Encouraging results have also dates protest mice against a lethal aerosol challenge. been obtained with chimeric vaccines (Paessler et al., Trobaugh et al. (2014) have reviewed recent advanc- 2003; Paessler and Weaver, 2009; Carossino et al., es in alphavirus virulence mechanisms that could be 2014). used to design live-attenuated vaccine against EEEV/ alphaviruses. Japanese Encephalitis ( JE) Japanese encephalitis ( JE) is very important and Western Equine Encephalitis common mosquito borne flavivirus causing enceph- Virus (WEEV) Infection alitis and is regarded a major public health problem WEEV was first isolated from brain of a horse (Mey- in Asian countries (van-den Hurk et al., 2008, 2009; er et al., 1931). First human WEEV was confirmed Pawaiya et al., 2010a). The disease affects primarily in 1938. WEEV is naturally propagated in enzootic human beings, horses and pigs. The infection in hors- cycle between passerine birds and specific mosquito es is usually subclinical with signs of pyrexia, depres- vector. Some rodents and lagomorphs also act as res- sion, tremors and ataxia. Abortions and stillbirths are ervoir hosts (Pfeffer and Dobler, 2010). Horses and common manifestations in pigs. humans are dead end hosts (Go et al., 2014). WEEV infections generally have an incubation period of 2-7 JE virus was first isolated from a case of fatal human days with early non-specific symptoms like fever, an- encephalitis in Japan in 1935 (Lewis et al., 1947). In orexia, headache, nausea and vomiting. Diagnosis of human beings incubation period is 5 to 15 days with NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 34 Advances in Animal and Veterinary Sciences majority of cases being asymptomatic. Encephalitis is Go et al., 2014). Birds like crow are also affected by reported in about 0.04% cases only. Severe rigors, py- this viral infection (Mishra et al., 2012). Molaei et al. rexia and malaise are non-specific symptoms lasting (2010) have studied the vector and host interactions one to six days. Signs of encephalitis are neck rigidity, governing the epidemiology of WNV in Southern cachexia, hemiperesis, convulsions and pyrexia. There California. Most of the WNV infections are subclini- is only one serotype of JEV and four genotypes (Chen cal and only less than 1% humans develop neurologic et al., 1990, 1992; Tsarev et al., 2000). Urbanization, disease (Mostashari et al., 2001; Hayes et al., 2005; population spurt in tropical areas, increased transpor- Hayes and Gubler, 2006; Porter et al., 2011). tation and global warming are responsible for spread of infections to newer areas (Go et al., 2014). Yeh et al. (2010) developed a duplex reverse tran- scriptase PCR for rapid differential detection of West JEV is transmitted by Culex tritaeniorhynchus, C an- Nile and Japanese encephalitis viruses which is useful nulus, C annulirostris and Aedis mosquitoes (Rosen, both in humans and horses. Yeh et al. (2012) could se- 1986). Pigs and aquatic birds are amplifying hosts rologically differentiate West Nile from Japanese en- that have high titre viremia which acts as a source cephalitis by a diagnostic algorithm. Currently, there of infection for mosquitoes (Rosen, 1986). Humans is no suitable therapy for WNV infection (Paterson et and horse do not have sufficient viremia to transmit al., 2011). A number of vaccines have been explored the infection and thus are dead end hosts. Yeh et al. for horses including inactivated whole West Nile virus (2010) developed a duplex reverse transcriptase PCR (West Nile–Innovator®, Vetera® WNV vaccine) and for rapid differential detection of west nile and japa- chimeric recombinant canarypoxvirus - Recombitek® nese encephalitis viruses which is rapid, sensitive and Equine WNV Vaccine (De Filette et al., 2012; Gu- specific and is useful both in humans and horses.Yeh lati et al., 2014). DNA vaccine for WNV has been et al. (2012) developed a diagnostic algorithm to se- licensed in USA, and with the use of prime boost ap- rologically differentiate west nile virus from japanese proaches it may protect WNV (Kumaragurubarn and encephalitis virus infection and its validation in field Kaliaperumal, 2013), but such vaccine (West Nile– surveillance of horses. Innovator® DNA) has recently been discontinued by Pfizer (Brandler and Tangy, 2013; Gulati et al., 2014). Earlier inactivated vaccines were used. A purified vaccine from vero-cell adapted SA 14-14-2 strain Hendra Virus (HeV) Infection has been developed (Tauber et al., 2007). Another Hendra virus (HeV) was first isolated in 1994 from chimeric vaccine containing pr M and E proteins of an outbreak among humans and horses (Selvey et al., JEV has also been developed and found to show high 1995; Murray, 1996). This is one of deadliest human level of immunogenicity (Guy et al., 2010; Halstead and veterinary pathogen causing respiratory and en- and Thomas, 2011). Singh et al. (2015) demonstrat- cephalitic illness in humans with high mortality rate ed JENVAC, a Vero-cell derived vaccine with a long which may exceed 70% (Dhama et al., 2010b; Croser lasting, broadly protective immunity. and Marsh, 2013). Hendra virus (HeV) is a zoonotic paramyxovirus in the genus Henipavirus. There had West Nile Virus (WNV) Infection been 48 outbreaks with increasing number of out- West Nile virus (WNV), a member of the Flavivirus breaks with each passing year since it was first reported genus of the Flaviviridae family, is one of the most (Aljofan, 2013). Horses acquire infection from flying widely distributed mosquito–transmitted arbovirus foxes. Clinical signs in horses include fever, anorexia having potential global public health concerns (Gula- followed by respiratory signs that include frothy nasal ti et al., 2014). WNV was first isolated in Uganda in discharge. Human beings get infection through di- 1937 (Smithburn et al., 1940). There have been out- rect contact with secretions from infected horses. No breaks in Africa, the Middle East, Asia, and Austral- evidence of human to human, human to horse and ia before spread to USA and Canada (Weaver and flying fox to human has been reported (Selvey et al., Barrett, 2004). The infection is maintained in nature 1996), however, Williamson et al. (1998) have shown between Culex mosquitoes and birds, whereas horses, the role of fruit bats, horses and cats in transmission humans and other mammals are dead end hosts (Blit- of Hendra virus. Flying fox bats acts as reservoirs and vich, 2008; Dhama et al., 2010a; Beck et al., 2013; horses may encounter the infection from contaminat

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 35 Advances in Animal and Veterinary Sciences

Figure 1: An overview on equine zoonoses ed secretions or excretions through environment. Vet- virus through their saliva. Inspite of low incidences in erinarian get the disease while physical examination of horses, literature reveal documented reports of equine oral cavity of horses. Human have flu like symptoms rabies in past years as 82 reports of rabies in 1998, 65 with mainly respiratory signs. The diagnosis of this in- cases in 1999 and 52 reports of equine rabies in 2000 fection is based on ELISA (IgG and IgM), RT-PCR have been from United States. Rabies in horse’s do and isolation of virus. There are no effective therapeu- not have the typical symptomps as in dog but the ani- tics against HeV infections and protective measures mal seems alert, lack of muscular in-coordination and while examining horses are the only way to minimize seizures are common (Hudson et al., 1996). Though as the risk of its spread (Mahalingam et al., 2012). Hu- compared to small animal practice probability of ac- man monoclonal antibody against HeV glycoprotein quiring rabies from equines in veterinarian is low but (G) protein is considered to be most promising pas- possibility cannot completely be ignored due to sever- sive immunotherapy (Bossart et al., 2011). ity of infection and variation in expression of symp- toms. In equines, among furious and dumb form, par- Equine Rabies alytic or dumb form is more reported with the signs of Rabies is a well-documented zoonotic disease that rubbing of site of wound, gradual lameness and colic causes huge fatalities throughout the world. Rabies (Krebs et al., 2000, 2001; Weese, 2002). Animal han- is a fatal neurological disease of mammals which is dlers, veterinarians and horse owners are at highest not an exception in equines, affects horses, mules and risk for the transmission of rabies virus to them. Hu- donkeys (Pawaiya et al., 2010b). It is caused by Lyssa- mans experience various symptoms like ataxia, loss of virus of Rhabdoviridae family. The major animal in- awareness, paralysis of muscles, etc. Vaccines are avail- volved is the dog and affected dogs can transmit the able for animals and humans that can prevent rabies,

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 36 Advances in Animal and Veterinary Sciences however history of vaccination does not necessarily able for R. equi infection of equines as well as humans exclude the risk of rabies, as one report described that till date (Khurana, 2015). even among 21 vaccinated horse, 5 animals could ac- quire the rabies (Green et al., 1999). Initial diagno- Human beings acquire infection mainly through in- sis can be made by differentially diagnosing all un- halation of dust harboring , from domestic differentiated neurological diseases or encephalitis animals including equines and wound, however man from Rabies. Animals may die due to cardiac arrest to man transmission is thought to be rare (Weinstock within 2-7 days or 14 days after appearance of clini- and Brown, 2002). cal symptoms as nerves are affected in this disease. In infected animals, central nervous system components, Agar gel diffusion test developed byNakazawa et saliva and salivary glands all are rich source of rabies al. (1987) and ELISA by Giguere et al. (2003) have virus hence any contact between infected saliva and not been found to be of much promise for diagnosis breached skin or mucous membrane of horse or han- of R. equi serologically. In India, diagnosis through dler, jockey, owner must be avoided by taking biosecu- post-mortem examination (Garg et al., 1985; Saxena rity precautions. If any case is observed or suspected, and Narwal, 2009) and isolation of R. equi from clin- higher authorities must be notified. ical samples (Khurana et al., 2009). Various PCR as- says have been developed (Sellon et al., 2001; Arriaga An overview on equine zoonosis is presented in Fig- et al., 2002; Ladron et al., 2003; Oldfield et al., 2004; ure 1. Ocampo-Sosa et al., 2007; Pusterla et al., 2007; Letek et al., 2008; Monego et al., 2009). The most valua- BACTERIAL PATHOGENS ble diagnostic procedures are combination of cultural methods along with PCR assay (Khurana, 2015). Rhodococcus equi Infection Rhodococcus equi is a gram positive, aerobic soil actin- Rifampicin along with macrolides is the drug of omycete responsible primarily for severe respiratory choice for treatment of R. equi infection. Rifampic- disease of young foals with high mortality rate (Pres- in resistance have been reported which is posing a cott, 1991; Yager et al., 1991; Khurana et al., 2009; challenge in therapy (Asoh et al., 2013; Burton et al., Giguere et al., 2011a, b; Khurana, 2014; Khurana 2013; Goldstein, 2014; Liu et al., 2014). Proper man- et al., 2014; Khurana, 2015). R. equi also causes ex- agement and sanitation at farms is very important for tra-pulmonary complications in equines including control of disease at equine farms. Hygiene is impor- enteritis, arthritis and in abdomen (Giguere tant in immunocompromised human beings. and Prescott, 1997). R. equi was first recovered from lung of a foal as equi (Magnusson, Anthrax 1923) and was reclassified as R. equi (Goodfellow and Anthrax is caused by Bacillus anthracis, an extremely Alderson, 1977). This organism is emerging as an im- resistant spore forming bacteria. Horses generally get portant pathogen in AIDS patients (Weinstock and infection by grazing in areas contaminated by anthrax. Brown, 2002), drug therapy (Mizuno et al., 2005) and The typical incubation period is 3 to 7 days. Bacte- some other immunosuppressive conditions (Napoleao ria multiply and disseminate throughout the body et al., 2005). The most common manifestation of hu- through blood and lymphatic system, and release le- man R. equi infections is , others include thal toxin causing cell death and breakdown of tissues. fever, diarrhoea, abscesses in various internal organs and arthritis. Symptoms appear very rapidly which include high fe- ver, agitation, chills, colic, anorexia, dullness, laboured Virulence associated protein A (Vap A), a cell surface breathing and seizures. Bloody diarrhoea, swelling lipoprotein is essentially required for virulence in foals around the neck may also be observed. Chest, abdo- whereas virulence associated protein B (Vap B) is of- men and genitals may also get swollen. Death may ten associated with disease in human beings and pigs. occur in 2 to 3 days after occurrence of first symp- Intracellular localization of R. equi is responsible for toms. This is controlled by vaccination in endemic ar- its prolonged and difficult therapeutic management. eas, early diagnosis and treatment, burning and burial No suitable serodiagnostic test or vaccination is avail- of dead animals.

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 37 Advances in Animal and Veterinary Sciences Horses are usually less susceptible than ruminants, but reported. reports of outbreaks of anthrax are available in horses from Minnesota and North Dakota. Horses may have This organism is very important from biological war- prolonged course of disease. Affected horses exhibit fare angle due to high rate of mortality and ability of symptoms of marked pyrexia, colic, dyspnea, and sub- small number of organisms to establish the infection. cutaneous edema and death may occur abruptly. The diagnosis of glanders is done by isolation and al- lergic test (Mallein test) which is prescribed test for Human beings contract infection from contaminated international trade. The test is not very specific, so it horses and their accessories and are often associated should be used along with complement fixation test with three forms cutaneous, gastrointestinal and in- which is also prescribed for international trade. The halation (Hicks et al., 2012). One more form of injec- test is not very specific, so it should be used along tional anthrax has also been reported (Lalitha et al., with complement fixation test (CFT), which is also 1988; Beaumont, 2010; Booth et al., 2011; Jallali et prescribed for international trade. Various serological al., 2011). Spores of anthrax bacilli are so sturdy that tests for diagnosis of glanders include complement they can survive for years in the environment and that fixation test (CFT), indirect haemagglutination assay can cause major problem for human population. (IHA) and ELISA. Singha et al. (2014) have devel- oped an indirect ELISA using truncated TssB protein Control of anthrax is done through vaccination, early for serodiagnosis of glanders. detection and reporting, quarantine, antibiotic thera- py of exposed animals, burning or burial of dead an- Various PCR assays have been developed (Grishki- imals that had suspected or confirmed anthrax infec- na and Samygi, 2010; Zhang et al., 2012) which have tion. Vaccination of horses is done only in endemic been found to be rapid, sensitive and specific. Janse et farms and areas. al. (2013) developed a multiplex qPCR for detection and differentiation of B. mallei and B. pseudomallei. Glanders Diagnosis in human beings is generally done by CFT Glanders is a contagious and fatal disease of horse, and imaging studies. mules and donkey with zoonotic potential (Malik et al., 2010; Varga et al., 2012; Verma et al., 2014a). This No vaccine is available for prevention of glanders for is a disease known since ancient times and was iden- animals or humans (Burtnick et al., 2012). In case of tified in th4 century BC by Hippocrates (Colahan et death suspected due to glanders, the carcass should al., 1999). The disease is caused by a non-spore form- not be opened and must be buried deep or incinerated. ing gram negative bacillus called mallei. Most common mode of transmission of this organism Brucellosis is inhalation and ingestion of contaminated feed and Brucellosis is one of the most important disease prob- water. The disease occurs in chronic form in horses, lems of animals and human beings. This is caused by where bacteria are found in nasal discharges and skin in equines, and is manifested by fistu- lesions (OIE 2004, 2008), whereas in mules and don- lous withers, poll evil, lameness due to joint infection keys the disease occurs in acute form (Hunting, 1913; and rarely late abortions in mares. Horizontal transfer Gulati and Gautam, 1962). The acute form of glanders of Brucella spp. to horses from cattle and pigs has been involves pulmonary, cutaneous and nasal sites ( Jubb documented (Forbes, 1990). Brucellosis is considered et al., 1993), and is characterized by pyrexia, cough, to be an occupational disease from public health point discharges from nostrils, ulcers on nasal mucosa and of view that mainly affects slaughter house workers, nodules on the skin finally leading to death. butchers and veterinarians. This causes undulant fever in human beings. Chronic form in horse also shows all three clinical manifestations which include pulmonary, nasal and Ehizibolo et al. (2011) have reported a sero-preva- cutaneous forms. The human glanders is not very lence of 14.7% of equine brucellosis in Nigeria. Ta- common, but it is having very high mortality rate of hamtan et al. (2010) have reported a sero-prevalence 90-95% in untreated septicaemic infection and 50% of 2.5% for brucellosis in horses in Iran. A sero-prev- in treated humans. Human outbreaks have not been alence of 5.88%, 12.89% and 5.78% has been reported

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 38 Advances in Animal and Veterinary Sciences from Egypt (Montasser et al., 1999), India (Sharma into acute toxic enterocolitis or septicaemia in various et al., 1979) and Pakistan (Ahmed and Munir, 1995a, vertebrate hosts including horses and human beings. b), respectively. Etiological agent is Gram negative bacteria of Salmo- nella genus with multiple serotypes. Horses of all age The incubation period in humans varies from 1-3 groups are susceptible. Multidrug resistant S. Typhi- weeks. The symptoms may include irregular fever, murium DT104 ( subspecies enter- headache, weakness, malaise, profuse sweating espe- ica Serotype Typhimurium Definitive Type 104) has cially during night. Coughing, chest pain, irritation, been recovered from many horses in Ontario and has insomnia, depression are occasionally encountered. In significant zoonotic potential due to its high lethality many patients, the symptoms last for 2 to 4 weeks and rate in human beings as well. Initially S. Typhimuri- are followed by spontaneous recovery. Others devel- um DT104 was isolated from cattle in 1988 in Eng- op recurrent bouts at 2-14 day intervals. Most people land and Wales but consequently was reported from with this undulant form recover completely in 3 to 12 sheep, pigs, poultry and horse. Fecal-oral route with months. A few patients become chronically ill, with high inoculums size containing more number of bac- symptoms of chronic fatigue, depressive episodes and teria is the most common way for zoonotic spread of arthritis. Relapses can be seen months after the initial salmonellosis in human population, though in immu- symptoms, even in successfully treated cases. Occa- nocompromised persons low inoculums may also pro- sional complications include arthritis, endocarditis, duce the disease. Higher mortality rate and resistance granulomatous hepatitis, meningitis, uveitis, orchitis, to commonly preferred antibiotics further aids to the cholecystitis, osteomyelitis, and rare cases of enceph- potential of this pathogen for zoonotic transmission. alitis. Asymptomatic infections are also common in Suspected cases should be monitored and treated sep- humans. arately. Strict follow up of personal hygienic meas- ures and proper disinfection of stables, contaminat- Diagnosis relies on the detection of circulating anti- ed equipments and utensils will help in reducing the bodies followed by the bacteriological isolation and menace of zoonotic transmission (Fone and Barker, serum agglutination tests [Rose Bengal plate agglu- 1994; Weese et al., 2001a; Weese, 2002). tination test (RBPT) and standard tube agglutina- tion test (STAT)]. Molecular diagnostic assays can be equi Subspecies used for detection instead of serological tests because zooepidemicus Infection serological assays like RBPT, STAT have the disad- Streptococcus equi subspecies zooepidemicus is consid- vantage of false positive reaction against other gram ered opportunistic pathogen in horse, but causes in- negative bacteria (Karthik et al., 2014a). Various mo- fection in cattle, sheep, goat, pig and dog also. S. equi lecular assays like polymerase chain reaction (PCR), subspecies zooepidemicus has more than 98% DNA se- Real time-PCR, etc., can be employed for detection quence homology with S. equi subspecies equi. Pelko- of the pathogen. Recent field assays like loop medi- nen et al. (2013) have shown that S. equi subspecies ated isothermal amplification assay and lateral flow zooepidemicus is transmitted from horse to human be- assay can be employed (Karthik et al., 2014a, 2014b). ings and they found that human and equine isolates were identical or closely related. Lindahl et al. (2013) Brucellosis being intracellular requires the need for reported an outbreak of repiratory disease due to S. antibiotics that can target intracellular pathogen. equi subspecies equi. Downar et al. (2001) have doc- Treatment in humans can be done with combination umented infection of Streptococcal meningitis from of various antibiotics like doxycyline, streptomycin close contact with an infected horse (Downar et al. and rifampicin. Combinational therapy is followed to (2001)). reduce the infection quickly and also to reduce the toxicity (MacMillan et al., 1982; Yousefi-Nooraie et S. equi subspecies zooepidemicus is β-haemolytic strep- al., 2012). tococci in Lancefield group C (Lancefield, 1933). The equine Lancefield group C streptococci are differenti- Salmonellosis ated biochemically by their ability to ferment sorbitol, Salmonellosis is an enteric disease with clinical man- lactose and trehalose (Quinn et al., 1994). In addition, ifestation of chronic diarrhea, fever which may turn PCR can be used to genetically identify different spe-

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 39 Advances in Animal and Veterinary Sciences cies and subspecies (Alber et al., 2004; Baverud et al., quent international movement and trading of horses 2007; Preziuso et al., 2010). (Weese et al., 2005, 2006). Most of the methicillin re- sistant Staphylococcus organisms affect soft tissues and difficile Infection joints in horses but these are resistant to treatment Clostridium difficile (C. difficile) is an anaerobic caus- with antibiotics. Some instances pneumonia, metritis ative agent for colitis in horses and human ( Jones et and sinusitis are also documented in horses (Smiet et al., 1987; George et al., 1978; Weese et al., 2001b). al., 2012). There is always danger of transfer of these C. difficileassociated diarrhoea can be mild, self-lim- drug resistance bacteria from animals to humans, and iting or peracute leading to fatal outcomes in horses. these methicillin resistant organisms can jump both It affects all ages of horses ranging from neonatal to ways from human to animal and also from animal to adults. In acute cases of diarrhea rapid diagnosis is human (Weese and Lefebvre, 2007). Veterinarians based on detection of bacterial toxins in fecal samples. top the list of humans suffering from this methicillin C. difficile is a well-recognized pathogen of human resistant Staphylococcus spp. transmission form horses also. The infection in man varies from mild to severe (van Duijkeren et al., 2011). Bacteriophage therapy pseudomembranous colitis leading to intestinal perfo- has yielded good response against MRSA (Karthik et ration and death. Transmission between animals and al., 2014c). Similarly, broad spectrum beta lactamase humans has been poorly studied. Equines suffering producing Enterobacteriacea can also be transmitted with C. difficile diarrhoea should be considered infec- from horses to human (Boyen et al., 2013). tious, particularly to people undergoing antimicrobial or chemotherapeutic treatment. Proper precautions Tuberculosis (gloves, gowns and boots) and close attention to per- Tuberculosis (TB) affects different mammals and sonal hygiene may prevent the chance of zoonotic many variants including extremely drug resistant TB transmission. Sporicidal disinfectant (5–10% bleach has emerged which poses serious threat to human be- solution) should be used to clean the contaminated ings (Karthik, 2012; Karthik et al., 2013). Incidences equipment and the areas under use. The veterinarians of TB are sparse in horses mainly due to established who develop acute diarrhoea following contact with control programmes, however few workers have re- suspected or confirmed case of infection in a horse ported the disease as confirmed by the presence of should take medical help to confirm the infection. causative agent (Pavlik et al., 2004). TB is a zoonotic disease caused by Mycobacterium tuberculosis, M. bo- Methicillin Resistant Staphylococcus spp. vis, and members of M. tuberculosis complex in several In the era of antibiotic resistance, methicillin resist- mammalian hosts including horses though they are ant Staphylococcus aureus (MRSA) is one of the prime considered comparatively resistant. Horses residing concerns. Different Staphylococcus spp. can harbour in close proximity with infected cattle acquire the in- horses of which S. aureus and its variant MRSA are fection as evidenced from the earlier reports showing the important species as they can be transmitted to presence of M. tuberculosis and M. bovis as well and humans and can cause major infections. In the early advocate a possibility of interspecies transmission and phase of MRSA, concern was restricted to humans zoonotic prospective of M. tuberculosis. Horses infect- and community associated MRSA which were in- ed with pulmonary TB illustrate lesions of multiple creasing throughout the world. Livestock associated tuberculoid granulomas in lung with manifestation MRSA has spread recently among animals (Hart- of granulomatous lymphadenitis in mediastinal and mann et al., 1997; McCarthy et al., 2012). Though the tracheobronchial lymph nodes, which can be further report of MRSA in horses was late but there are arrays confirmed by laboratory culture techniques and quan- of documentation after its first report in later part of titative real-time PCR (Keck et al., 2010; Blahutkova nineteenth century (Stull et al., 2012). Literature re- et al., 2011; Konstantin et al., 2012). veal that approximately 10% of healthy vigorous hors- es cart MRSA in their nasal passages, intestinal tracts PARASITIC PATHOGENS and on their skin, thereafter these colonized horses act as reservoirs of MRSA in the community which Cryptosporidiosis are efficiently capable of transmitting MRSA in the Equine cryptosporidiosis is caused by a protozoal human population in and across the globe due to fre- pathogen, Cryptosporidium parvum, causing enter-

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 40 Advances in Animal and Veterinary Sciences ic disease in several species including humans and is sis of global concern caused by spirochetes of genus most commonly associated with foals and immuno- Leptospira (Ebani et al., 2012). Leptospira spp. is en- deficient animals. Infected horses shed oocysts with demic in various parts of the world. Various serovars the shedding rate of 0–21% which have infective po- like Pomana, Icterohaemorrhagiae and Bratislava are tential, horses develop asymptomatic cryptosporidi- recorded in horses but risk of zoonotic transmission osis. High prevalence of disease with 71% infection of from horses is not immense. Hall rate is documented in foals and zoonotic transmission and Bryan (1952) and other workers have reported of Cryptosporidium from foal to handling veterinari- leptospirosis in horse, which harbor the infection as ans is also reported (Coleman et al., 1989; Cole et al., accidental host (Hall and Bryan, 1952; Hogg, 1974; 1998). Humans express symptoms of copious watery Barwick et al., 1998). Symptoms in horse include diarrhea which may lead to critical grave condition. anorexia, fever, lethargyness, renal dysfunction, jaun- In immunocompromised persons, the disease exists dice; abortion and still birth can also occur in preg- in a self-limiting form. Zoonotic implications occur nant mares (Divers et al., 1992; Timoney et al., 2011). due to high shedding rates, hence precautions must Equine recurrent uveitis or moon blindness occurs be taken while dealing with diarrhoic animals to min- after weeks or months after the onset of systemic imize the risk of zoonosis (Snyder et al., 1978; Kon- leptospirosis in horses. Symptoms in human include kle et al., 1997; Majewska et al., 1999; McKenzie and jaundice, fever, muscular pain, vomiting, uveitis etc. Diffay, 2000). (Hartskeerl et al., 2004; Verma et al., 2013). Micro- scopic agglutination test is the gold standard test for Giardiasis diagnosis of leptospirosis but several disadvantages Fecal shedding of Giardia oocysts from horses is an makes it difficult to perform. Various recombinant indication of zoonotic risk. Giardiasis caused by Gi- proteins are used in ELISA and latex agglutination ardia intestinalis is the most common intestinal para- assay formats for diagnosis of leptospirosis which are sitic disease characterized by mild or severe diarrhea. effective in diagnosis (Deneke et al., 2014). Dermato- Zoonotic transmission of Giardia is supported by phytosis (ringworm) is a fungal dermatologic disease feco-oral route. Asymptomatic shedding of Giardia of zoonotic concern affecting variety of animals in- in 25% of adult horses and 71% cumulative infection cluding horses, caused by species of Microsporum or rate in foals further suggest possibility of zoonotic po- Trichophyton. Horses are mainly affected by T. equi- tential of this pathogen in horses besides other species num with the clinical presentation of mild or subclini- (Xiao and Herd, 1994). Fecal cyst detection and ELI- cal form of disease to severe lesions imitating to pem- SA are available for diagnosis for giardiasis (Rishniw phigus foliaceus. Dermatophytosis can be transmitted et al., 2010). from horses to persons in contact through direct and indirect routes (Pascoe, 1976; Pier and Zancanella, Other Zoonotic Diseases of Equines 1993; Huovinen et al., 1998). Several important zoonoses shared between man and equines have been described above, however all the Preparedness and strategic planning possible zoonotic threats related to equines are not to counter equine zoonosis detailed and still there are many others like Borna To protect the public and animal health of equine virus, Nipah virus infection as geographically limited sector from devastating effects of zoonoses, equine zoonotic, dourine, crytosporidisis (Sellon, 2007; Xiao, emergency preparedness plans are required to be im- 2008), giardiasis, leptospirosis, dermatophytosis, Hal- plemented by collaboration of local, regional, state icephalobus gingivalis (Micronema deletrix) under officials, academic institutions, tribal and other allied certain specific conditions, are also among the sever- government agencies to safeguard the equine industry al other infections capable of being transmitted from and personnel involved by providing public education, equines to human beings thus adding to the disease following integrated surveillance plans, epidemiolog- burden. Besides viral and bacterial pathogens, para- ical disease investigations and prevention strategies sitic, protozoan and fungal agents are also involved in on zoonotic threats of already mentioned emerging equine zoonoses. Leptospirosis is another important equine zoonotic diseases. As swiftly as possible any disease of public health concern that can be transmit- suspected incidence should be detected and con- ted from various animals. Leptospirosis is a zoono- trolled, and containment of the incidence should be

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 41 Advances in Animal and Veterinary Sciences arranged to protect the health and environment for 2008, 2013e; Koff et al., 2013). Adaptation of regular stabilizing the economy. Local veterinary surgeon and and judicious vaccination strategies, DIVA strategy, state personnel should assure educating equine vet- prime boost regimens and giving booster vaccina- erinarians, owners, trainers and farm managers about tions must be implemented appropriately. Emphasis equine industry organizations and effective disease should be given in utilizing potential of novel and management practices. alternative/complementary immunomodulatory and treatment regimens comprising of immunotherapy, In the current era of increasing global population, glo- cytokine therapy, si-RNAs, avian egg antibodies, toll balization trends, tourism expansion, ecosystem and like receptors, phages, enzybiotics, probiotics, nutri- biodiversity changes like global warming, emerging tional immunomodulation, herbs and nanomedicines drug resistance, need for effective therapeutics and for devising appropriate prevention and control pro- vaccines, immune stresses, we need to strengthen grammes to counter infectious pathogens including research and development programmes, implement zoonosis (Mahima et al., 2012; Dhama et al., 2013f, strategic and planned veterinary and medical ap- 2014a; Malik et al., 2013; Tiwari et al., 2014). RNA proaches, the one world health one medicine concept, interference has been used successfully in various par- multi-disciplinary and international level surveillance asitic infections which can be adopted for other dis- /networking employing geographical information eases (Sudhakar et al., 2013). Implementation of good system (GIS), early warning systems and to tack- management practices, strict biosecurity rules, proper le emerging/re-emerging infectious diseases of ani- hygiene and sanitation procedures, follow up of isola- mals and their increasing zoonotic and pandemic risk tion and quarantine, and trade restrictions need to be (Slingenbergh et al., 2004; Kahn et al., 2007; Jones taken care of very timely for checking and controlling et al., 2008; Bergquist, 2011; Dhama et al., 2013a, the transmission and spread of zoonotic pathogens. 2013b, 2013c, 2014a; Tiwari et al., 2013; Verma et al., This holistic approach would pave road to lower the 2014b). Important vectors and reservoirs of infectious disease incidences / outbreaks as well as in controlling zoonotic pathogens need to be brought under control the zoonotic pathogens of equines and their public to prevent the spread of infectious agents and disease health concerns. risks and threats to equines and related public health concerns (Daszak et al., 2000; Bengis et al., 2004; CHALLENGES AHEAD AND Zinsstag et al., 2007; Dhama et al., 2013d). Recent FUTURE OUTLOOK advances in diagnostics and molecular detection tools for delivering rapid and confirmatory diagnosis of zo- Lack of awareness, inadequate communication be- onotic infectious pathogens of animals and affecting tween veterinarians and public health organizations, humans need to be explored to their full potential, in- and weak surveillance systems for zoonoses are the cluding PCR, real-time PCR, multiplex PCR, LAMP, major problem areas. Infections that affect animals recombinant protein based diagnostics, biosensors, and humans fall in ‘no man’s land’. In most hu- biochips, microarrays, gene sequencing, phylogenet- man-animal episodes, neither the human nor the vet- ic analysis and nanodiagnostics (Schmitt and Hen- erinary health systems have the capacity to deal with derson, 2005; Belak, 2007; Belak et al., 2009; Bollo, the outbreaks. Strengthening of risk assessments and 2007; Ratcliff et al., 2007; Balamurugan et al., 2010; early warning systems, laboratory capacity for diag- Bergquist, 2011; Deb and Chakraborty, 2012; Dha- nosis, monitoring and treatment are the much needed ma et al., 2012, 2014b; Ayyar and Arora, 2013). Apart priorities. Monitoring and testing of all antivenom from conventional killed and live vaccines, due prior- or antitoxin produced from horse serum against vari- ity need to be given for developing effective and saf- ous pathogens like Brucella, Streptococcus, Burkholderia er new generation prophylactics comprising of DNA mallei and other viral pathogens using different spe- vaccines, plant based (edible) vaccines, reverse genet- cific diagnostic assays can prevent spread of zoonot- ics vaccines, vector vaccines, protein/peptide vaccines, ic diseases. Zoonotic illnesses can infect humans by gene deleted mutant vaccines, reassortant vaccines, entering the body in a variety of ways: animal bites, chimeric vaccines, virus like particles (VLP), vaccine insect bites, by ingestion, by inhalation, through cuts/ cocktails, and vaccine delivery systems (oral, spray scratches and through the eyes or contact with other administration) (Meeusen et al., 2007; Dhama et al., mucous membranes. A combination of precautions

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 42 Advances in Animal and Veterinary Sciences including breaking the transmission cycle especial- dx.doi.org/10.1111/j.1439-0450.2004.00799.x ly in arboviral infections is effective in preventing • Aljofan M (2013). Hendra and nipah infection: zoonotic infections. Appropriate follow up of early emerging paramyxoviruses. Virus Res. diagnosis; maintain high personal hygiene and pre- 177(2): 119-126. http://dx.doi.org/10.1016/j. cautions while handling animals, fomites, tissues and virusres.2013.08.002 • Arriaga JM, Cohen ND, Derr JN, Chaffin MK, various specimens effectively reduces the probability Martens RJ (2002). Detection of Rhodococcus of disease transmission to a great extent. Physicians, equi by polymerase chain reaction using veterinarians and public health professionals must species-specific nonproprietary primers. J. Vet. work together to recognize and control zoonotic dis- Diagn. Invest. 14(4): 347-353. http://dx.doi. eases. Approaches to the control of zoonoses differ org/10.1177/104063870201400416 according to the type of zoonoses, because majority of • Asoh N, Watanabe H, Fines-Guyon M, Watanabe direct and cyclozoonoses and some saprozoonoses are K, Oishi K, Kositsakulchai W, Sanchai T, most effectively controlled by techniques involving Kunsuikmengrai K, Kahintapong S, Khanawa the animal host, and methods used to combat these B, Tharavichitkul P, Sirisanthana T, Nagatake T diseases are almost entirely the responsibility of veter- (2013). Emergence of rifampin-resistant Rhodococcus inary medicine. The control of metazoonoses may be equi with several types of mutations in rpoB gene among AIDS patients in northern Thailand. J. directed at the infected vertebrate host, at the infected Clin. Microbiol. 41(6): 2337-2340. http://dx.doi. invertebrate vector or both. National and internation- org/10.1128/JCM.41.6.2337-2340.2003 al agencies with mandates to control zoonotic diseases • Ayyar BV, Arora S (2013). Antibody-based biosensors should coordinate their activities and share resources for detection of Veterinary viral pathogens. Adv. to accomplish prevention and control of zoonotic dis- Anim. Vet. Sci. 1(4S): 37-44. eases. The key success to prevent and control equine • Balamurugan V, Venkatesan G, Sen A, Annamalai L, zoonotic pathogens lies in optimum utilization of re- Bhanuprakash V, Singh RK (2010). Recombinant cent developments in advances in diagnostics, surveil- protein-based viral disease diagnostics in veterinary lance /networking, vaccines, therapeutics and good medicine. Expert Rev. Mol. Diagn. 10(6): 731-753. management practices. Proper hygienic and sanitary http://dx.doi.org/10.1586/erm.10.61 practice while handling horses or any other animals • Barwick RS, Mohammed HO, McDonough PL, White ME (1998). Epidemiological features can prevent spread of infectious agents not only from of equine Leptospira interrogans of human animals but also vice versa. Veterinarians who are at significance. Prev. Vet. Med. 36: 153–165. http:// most risk should wear protective clothings as gloves, dx.doi.org/10.1016/S0167-5877(98)00069-5 mask, boots etc., while examining animals to prevent • Baverud V, Johansson SK, Aspan A (2007). Real- spread of zoonotic infection. time PCR for detection and differentiation of Streptococcus equi subsp. equi and Streptococcus REFERENCES equi subsp. zooepidemicus. Vet. Microbiol. 124(3-4): 219-229. http://dx.doi.org/10.1016/j. • Aguilar PV, Robich RM, Turell MJ, O’Guinn ML, vetmic.2007.04.020 Klein TA, Huaman A, Guevara C, Rios Z, Tesh RB, • Beamont G (2010). Anthrax in a Scottish intravenous Watts DM, Olson J, Weaver SC (2007). Endemic drug user. J. Forensic Leg. Med. 17(8): 443-445. eastern equine encephalitis in the amazon region of http://dx.doi.org/10.1016/j.jflm.2010.09.008 Peru. Am. J. Trop. Med. Hyg. 76(2): 293–298. • Beck C, Jimenez-Clavero MA, Leblond A, Durand B, • Ahmed R, Munir M (1995a). Epidemiological Nowotny N, Laparc-Goffart I, Zientara S, Jourdain investigations of brucellosis in horses, dogs, cats and E, Lecollinet S (2013). Flaviviruses in Europe: poultry. Pak. Vet. J. 15(2): 85-88. Complex circulation patterns and their consequences • Ahmed R, Munir M (1995b). Epidemiological for diagnosis and control of West Nile disease. Int. J. investigations of brucellosis in Pakistan. Pak. Vet. J. Environ. Res. Public health. 10: 6049-6083. http:// 15(4): 169-172. dx.doi.org/10.3390/ijerph10116049 • Alber J, El Sayed A, Lammler C, Hassan AA, Weiss • Belak S (2007). Molecular diagnosis of viral diseases, R, Zschock M (2004). Multiplex polymerase chain present trends and future aspects: A view from the reaction for identification and differentiation of OIE collaborating centre for the application of Streptococcus equi subsp. zooepidemicus and polymerase chain reaction methods for diagnosis Streptococcus equi subsp. Equi. J. Vet. Med. B Infect. of viral diseases. Veterinary Medocone. Vaccine. Dis. Vet. Public Health. 51(10): 455-458. http:// 25(30): 5444-5452. http://dx.doi.org/10.1016/j.

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 43 Advances in Animal and Veterinary Sciences vaccine.2006.11.068 plasma. Biologicals. 32(3): 115-128. http://dx.doi. • Belak S, Thoren P, Le Blanc N, Viljoen G (2009). org/10.1016/j.biologicals.2004.07.001 Advances in viral disease diagnostic and molecular • Burtnick MN, Heiss C, Roberts RA, Schweizer epidemiological technologies. Expert Rev. Mol. HP, Azadi P, Brett PJ (2012). Development of Diagn. 9(4): 367-381. http://dx.doi.org/10.1586/ capsular polysaccharide-based glycoconjugates for erm.09.19 immunization against and glanders. • Bengis RG, Leighton FA, Fischer JR, Artois M, Front Cell. Infect. Microbiol. 2: 108. http://dx.doi. Mörner T, Tate CM (2004). The role of wildlife org/10.3389/fcimb.2012.00108 http://dx.doi. in emerging and re-emerging zoonoses. Revue org/10.3389/fcimb.2012.00148 Scientifique et Technique de l Office International • Burton AJ, Giguère S, Sturgill TL , Berghaus LJ, des Epizooties. 23(2): 497-511. Slovis NM, Whitman JL, Levering C, Kuskie • Bergquist R (2011). New tools for epidemiology: KR, Cohen ND (2013). Macrolide-and rifampin- a space odyssey. Memórias do Instituto Oswaldo resistant Rhodococcus equi on a horse breeding farm, Cruz. 106(7): 892-900. http://dx.doi.org/10.1590/ Kentucky, USA. Emerg. Infect. Dis. 19(2): 282-285. S0074-02762011000700016 http://dx.doi.org/10.3201/eid1902.121210 • Blahutkova M, Fictum P, Skoric M (2011). • Carossino M, Thiry E, Grandiere A, Barrandeguy ME Mycobacteruim avium subsp. Hominissuis infection (2014). Novel vaccination approaches against equine in two sibling Fjord horses diagnosed using alphavirus encephalitides. Vaccine. 32(3): 311-319. quantitative real time PCR: a case report. Veterinarni http://dx.doi.org/10.1016/j.vaccine.2013.11.071 Medicina. 56: 294-301. • Chen W, Tesh RB, Rico-Hesse R (1990). Genetic • Blitvich BJ (2008). Transmission dynamics and variation of Japanese encephalitis virus in nature. changing epidemiology of West Nile virus. Anim. J. Gen. Virol. 71(Pt 12): 2915-2922. http://dx.doi. Health Res. Rev. 9(1): 71-86. http://dx.doi. org/10.1099/0022-1317-71-12-2915 org/10.1017/S1466252307001430 • Chen W, Rico-Hesse R, Tesh RB (1992). A new • Bollo E (2007). Nanotechnologies applied to genotype of Japanese encephalitis virus from veterinary diagnostics. Vet. Res. Commun. 1: 145- Indonesia. Am. J. Trop. Med. Hyg. 47(1): 61-69. 147. http://dx.doi.org/10.1007/s11259-007-0080-x • Colahan PT, Mayhew IG, Merritt AM, Moore JN • Booth MG, Hood J, Brooks TJ, Hart A (2011). (1999). Equine Medicine and Surgery. Vol. 1-2, 5th Health protection Scotland anthrax clinical Edn. Mosby Inc, St. Louis. Pp. 536-537. network. Anthrax infection in drug users. Lancet. • Cole DJ, Cohen ND, Snowden K, Smith R (1998). 375: 1345-1346. http://dx.doi.org/10.1016/S0140- Prevalence of and risk factors for fecal shedding of 6736(10)60573-9 Cryptosporidium parvum oocysts in horses. J. Am. • Bossart KN, Geisbert TW, Feldmann H, Zhu ZY, Vet. Med. Assoc. 213(9): 1296-1302. Feldmann F, Geisbert JB, Yang LY, Feng YR, Brining • Coleman SU, Klei TR, French DD, Chapman MR, D, Scott D, Wang YP, Dimitrov AS, Callison J, Chan Corstvet RE (1989). Prevalence of Cryptosporidium YP, Hickey AC, Dimitrov DS, Broder CC, Rockx B sp. in equids in Louisiana. Am. J. Vet. Res. 50(4): (2011). A neutralizing human monoclonal antibody 575-577. protects African green monkeys from hendra virus • Croser EL, Marsh GA (2013). The changing face of challenge. Sci. Transl. Med. 3(105): 105ra103. henipaviruses. Vet. Microbiol. 167(1-2): 151-158. • Bowen GS, Calisher CH (1976). Virological http://dx.doi.org/10.1016/j.vetmic.2013.08.002 and serological studies of Venezuelan equine • Daszak P, Cunningham AA, Hyatt AD (2000). encephalomyelitis in humans. J. Clin. Microbiol. Emerging infectious diseases of wildlife: threats 4(1): 22-27. to biodiversity and human health. Science. • Boyen F, Smet A, Hermans K, Butaye P, Martens 287(5452): 443-449. http://dx.doi.org/10.1126/ A, Martel A, Haesebrouck F (2013). Methicillin science.287.5452.443 resistant staphylococci and broad-spectrum • De Filette M, Ulbert S, Diamond M, Sanders NN b-lactamase producing in (2012). Recent progress in West Nile virus diagnosis horses. Vet. Microbiol. 167(1-2): 67–77. http:// and vaccination. Vet. Res. 43: 16. http://dx.doi. dx.doi.org/10.1016/j.vetmic.2013.05.001 org/10.1186/1297-9716-43-16 • Brandler S, Tangy F (2013). Vaccines in development • Deb R, Chakraborty S (2012). Trends in Veterinary against West Nile Virus. Viruses. 5(10): 2384–2409. diagnostics. J. Vet. Sci. Technol. 3: 1. http://dx.doi.org/10.3390/v5102384 • Deneke Y, Sabarinath T, Gogia N, Lalsiamthara • Burnouf T, Griffiths E, Padilla A, Seddik S, Stephanoe J, Viswas KN, Chaudhuri P (2014). Evaluation of MA, Gutie´rrez JM (2004). Assessment of the viral recombinant LigB antigen-based indirect ELISA safety of antivenoms fractionated from equine and latex agglutination test for the serodiagnosis NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 44 Advances in Animal and Veterinary Sciences of bovine leptospirosis in India. Mol. Cell Probes. Biol. Agri. Sci. 1(1): 1-12. 28(4): 141-146. http://dx.doi.org/10.1016/j. • Dhama K, Chakraborty S, Mahima Wani MY, Verma mcp.2014.01.001 AK, Deb R, Tiwari R, Kapoor S (2013f ). Novel and • Dhama K, Mahendran M, Gupta PK, Rai A (2008). emerging therapies safeguarding health of humans DNA vaccines and their applications in veterinary and their companion animals: A review. Pak. J. Biol. practice: current perspectives. Vet. Res. Commun. Sci. 16(3): 101-111. http://dx.doi.org/10.3923/ 32(5): 341-356. http://dx.doi.org/10.1007/s11259- pjbs.2013.101.111 008-9040-3 • Dhama K, Chakraborty S, Tiwari R, Verma AK, • Dhama K, Pawaiya RVS, Kapoor S, Mathew T Saminathan M, Amarpal, Malik YS, Nikousefat (2010a). West Nile virus infection in horses. In: Z, Javdani M, Khan, RU (2014a). A concept paper Advances in Medical and Veterinary Virology, on novel technologies boosting production and Immunology, and Epidemiology - Vol. 7 : Tropical safeguarding health of humans and animals. Res. Viral Diseases of Large Domestic Animals- Part 1, Opin. Anim. Vet. Sci. 4(7): 353-370. Editor : Thankam Mathew, Thajema Publishers, 31 • Dhama K, Karthik K, Chakraborty S, Tiwari R, Glenview Dr. West Orange NJ 07052-1010, USA / Kapoor S, Kumar A, Thomas P (2014b). Loop- Xlibris Corporation, United Kingdom, ISBN 978- mediated isothermal amplification of DNA 1-4415-8160-0. Pp. 372-392. (LAMP) – a new diagnostic tool lights the world • Dhama K, Pawaiya RVS, Kapoor S (2010b). Hendra of diagnosis of Animal and Human Pathogens: virus infection in horses. In: Advances in Medical A review. Pak. J. Biol. Sci. 17(2): 151-166. http:// and Veterinary Virology, Immunology, and dx.doi.org/10.3923/pjbs.2014.151.166 Epidemiology - Vol. 7 : Tropical Viral Diseases of • Divers TJ, Byars TD, Shin SJ (1992). Renal dysfunction Large Domestic Animals- Part 1, Editor : Thankam associated with infection of Leptospira interrogans Mathew, Thajema Publishers, 31 Glenview Dr., in a horse. J. Am. Vet. Med. Assoc. 201(9): 1391– West Orange NJ 07052-1010, USA / Xlibris 1392. Corporation, United Kingdom, ISBN 978-1-4415- • Downar J, Willey BM, Sutherland JW, Mathew 8160-0. Pp. 292-308. K, Low DE (2001). Streptococcal meningitis • Dhama K, Wani MY, Tiwari R, Kumar D (2012). resulting from contact with an infected horse. J. Molecular diagnosis of animal diseases: the current Clin. Microbiol. 39(6): 2358–2359. http://dx.doi. trends and perspectives. Livest. Sphere. 1(5): 6-10. org/10.1128/JCM.39.6.2358-2359.2001 • Dhama K, Chakraborty S, Kapoor S, Tiwari R, Kumar • Dupuy LC, Richards MJ, Ellefsen B, Chau L, A, Deb R, Rajagunalan S, Singh R, Vora K, Natesan Luxembourg A, Hannaman D, Livingstone BD, S (2013a). One world, one health - veterinary Schmaljohn CS (2011). A DNA vaccine for perspectives. Adv. Anim. Vet. Sci. 1(1): 5-13. Venezuelan equine encephalitis virus delivered by • Dhama K, Tiwari R, Chakraborty S, Kumar A, intramuscular electroporation elicits high levels of Karikalan M, Singh R, Rai RB (2013b). Global neutralizing antibodies in multiple animal models warming and emerging infectious diseases of and provides protective immunity to mice and animals and humans: Current scenario, challenges, nonhuman primates. Clin. Vaccine Immunol. 18(5): solutions and future perspectives – A review. Int. J. 707-716. http://dx.doi.org/10.1128/CVI.00030-11 Curr. Res. 5(7): 1942-1958. • Ebani VV, Bertelloni F, Pinzauti P, Cerri D (2012). • Dhama K, Verma AK, Tiwari R, Chakraborty S, Vora Seroprevalence of Leptospira spp. and Borrelia K, Kapoor S, Deb R, Karthik K, Singh R, Munir burgdorferi sensu lato in Italian horses. Ann. Agric. M, Natesan S (2013c). A perspective on application Environ. Med. 19(2): 237–240. of Geographical Information System (GIS); and • Ehizibolo DO, Gusi AM, Ehizibolo PO, Mbuk EU, advanced tracking tool for disease surveillance and Ocholi RA (2011). Serologic prevance of brucellosis monitoring in Veterinary epidemiology. Adv. Anim. in horse stables in two nortern states of Nigeria. J. Vet. Sci. 1(1): 14-24. Equine Sci. 22(1): 17-19. http://dx.doi.org/10.1294/ • Dhama K, Karthik K, Chakraborty S, Tiwari R, jes.22.17 Kapoor S. (2013d). Wildlife: a hidden warehouse of • Engler RJ, Mangiafico JA, Jahrling P, Ksiazek TG, zoonosis – A review. Int. J. Curr. Res. 5(7): 1866- Pedrotti-krueger M, Peters CJ (1992). Venezuelan 1879. equine encephalitis-specific immunoglobulin • Dhama K, Wani MY, Deb R, Karthik K, Tiwari responses: live attenuated TC-83 versus inactivated R, Barathidasan R, Kumar A, Mahima, Verma C-84 vaccine. J. Med. Microbiol. 38(4): 305-310. AK, Singh SD (2013e). Plant based oral vaccines • Fone D, Barker R (1994). Associations between for human and animal pathogens-a new era of human and farm animal infections with Salmonella prophylaxis: current and future prospective. J. Exp. typhimurium DT104 in Herefordshire. Comun. Dis.

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 45 Advances in Animal and Veterinary Sciences Rep. CDR Rev. 4: R136–R140. Adv. Anim. Vet. Sci. 2 (4S): 17 – 23. http://dx.doi. • Forbes LB (1990). Brucella abortus infection in 14 org/10.14737/journal.aavs/2014/2.4s.17.23 farm dogs. J. Am. Vet. Med. Assoc. 196: 911–916. • Guy B, Guirakhoo F, Barban V, Higgs S, Monath TP, • Garg DN, Manchanda VP, Chandramani NK (1985). Lang J (2010). Preclinical and clinical development Etiology of post-natal foal mortality. Ind. J. Comp. of YFV 17D-based chimeric vaccines against Microbiol. Immunol. Infect. Dis. 6(1): 29-35. dengue, west Nile and Japanese encephalitis • George RH, Symonds JM, Dimock F, Brown JD, viruses. Vaccine. 28(3): 632-649. http://dx.doi. Arabi Y, Shinagawa N (1978). Identification of org/10.1016/j.vaccine.2009.09.098 Clostridium difficile as a cause of pseudomembranous • Hall CE, Bryan JT (1952). A case of leptospirosis in a colitis. Br. Med. J. 1: 695. horse. Cornell Vet. 44: 345–348. • Giguere S, Hernandez J, Baskin J, Prescott JF, Takai • Halstead SB, Thomas SJ (2011). New Japanese S, Miller C (2003). Performance of five serological encephalitis vaccines: alternatives to production in assays for diagnosis of Rhodococcus equi pneumonia mouse brain. Expert Rev. Vaccines. 10(3): 355-364. in foals. Clin. Diagn. Lab. Immunol. 10(2): 241–245. http://dx.doi.org/10.1586/erv.11.7 • Gigue`re S, Prescott JF (1997). Clinical manifestations, • Hartmann FA, Trostle SS, Klohnen AAO (1997). diagnosis, treatment, and prevention of Rhodococcus Isolation of methicillin- resistant Staphyloccus aureus equi infections in foals. Vet. Microbiol. 56: 313-334. from a postoperative wound infection in a horse. J. • Giguère S, Cohen ND, Keith Chaffin M, Hines Am. Vet. Med. Assoc. 211(5): 590–592. SA, Hondalus MK, Prescott JF, Slovis NM • Hartskeerl RA, Goris MG, Brem S, Meyer P, Kopp (2011a). Rhodococcus equi: Clinical manifestations, H, Gerhards H, Wollanke B (2004). Classification virulence, and immunity. J. Vet. Intern. Med. 25(6): of leptospira from the eyes of horses suffering 1221-1230. http://dx.doi.org/10.1111/j.1939- from recurrent uveitis. J. Vet. Med. B Infect. Dis. 1676.2011.00804.x Vet. Public Health. 51(3): 110-115. http://dx.doi. • Giguère S, Cohen ND, Keith Chaffin M, Slovis NM, org/10.1111/j.1439-0450.2004.00740.x Hondalus MK, Hines SA, Prescott JF (2011b). • Hayes EB, Komar N, Nasci RS, Montogomery SP, Diagnosis, treatment, control, and prevention of O’Leary DR, Campbell GL (2005). Epidemiology infections caused by Rhodococcus equi in foals. J. and transmission dynamics of West Nile virus Vet. Intern. Med. 25(6): 1209-1220. http://dx.doi. disease. Emerg. Infect. Dis. 11(8): 1167-1173. org/10.1111/j.1939-1676.2011.00835.x http://dx.doi.org/10.3201/eid1108.050289a • Giltner LT, Shahan MS (1933). The 1933 outbreak • Hayes EB, Gubler DJ (2006). West Nile virus: of infectious equine encephalomyelitis in the eastern epidemiology and clinical features of an emerging states. North Am. Vet. 14(11): 25-27. epidemic in the United States. Annu. Rev. Med. • Go YY, Balasuriya UBR, Lee C (2014). Zoonotic 57: 181-197. http://dx.doi.org/10.1146/annurev. encephalitides caused by arboviruses: transmission med.57.121304.131418 and epidemiology of alphaviruses and flaviviruses. • Hicks CW, Sweeney DA, Cui X, Li Y, Eichacker PQ Clin. Exp. Vaccine Res. 3(1): 58-77. http://dx.doi. (2012). An overview of anthrax infection including org/10.7774/cevr.2014.3.1.58 recently identified form of disease in injection drug • Goldstein BP (2014). Resistance to rifampicin: a users. Intensive Care Med. 38(7): 1092-1104. http:// review. J. Antibiot. 67(9): 625-630. http://dx.doi. dx.doi.org/10.1007/s00134-012-2541-0 org/10.1038/ja.2014.107 • Hogg GG (1974). The isolation of Leptospira pomona • Goodfellow M, Alderson G (1977). The actinomycete- from a sick foal. Aust. Vet. J. 50: 326. http://dx.doi. genus Rhodococcus: A home for “rhodochrous org/10.1111/j.1751-0813.1974.tb05336.x complex”. J. Gen. Microbiol. 100(1): 99-122. http:// • Honnold SP, Baken RR, Fisher D, Lind CM, Cohen dx.doi.org/10.1099/00221287-100-1-99 JW, Eccleston LT, Spurgers KB, Maheshwari RK, • Green SL, Smith L, Vernau W, Beacock SM (1999). Glass PJ (2014) Second generation inactivated Rabies in horses: 21 cases (1970–1990). J. Am. Vet. eatern equine encephalitis virus vaccine candidates Med. Assoc. 200: 1133– 1137. protect mice against lethal aerosol challenge. PLoS • Grishkina TA, Samygin VM (2010). Molecular One. 9(8): e104708. http://dx.doi.org/10.1371/ methods of detection and identification of journal.pone.0104708 pathogenic Burkholderia. Zh Mikrobiol. Epidemiol. • Howitt B (1938). Recovery of virus of equine Immunobiol. 5: 98-105. encephalomyelitis from the brain of a child. Science. • Gulati, RL, Gautam OP (1962). Glanders in mules. 88(2289): 455-456. http://dx.doi.org/10.1126/ Indian Vet. J. 39: 588-593. science.88.2289.455 • Gulati BR, Gupta AK, Kadian SK (2014). West Nile • Hudson LC, Weinstock D, Jordan T, Bold-Fletcher virus infection among animals and humans in India. NO (1996). Clinical presentation of experimentally NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 46 Advances in Animal and Veterinary Sciences induced rabies in horses. Zentralbl Veterinarmed B. (2014a). Loop-mediated isothermal amplification 43(5):277-85. (LAMP) test for specific and rapid detection of • Hunting W (1913). Glanders. In: A System of Brucella abortus in cattle. Vet. Q. http://dx.doi.org/ Veterinary Medicine, Hoarse, E.W. (Ed.). Balliere 10.1080/01652176.2014.966172 Tindall and Cox, London. • Karthik K, Rathore R, Thomas P, Arun TR, Viswas • Huovinen S, Tunnela E, Kuijpers AF, Suhonen KN, Dhama K, Agarwal RK (2014b). New closed R, Huovinen P (1998). Human onychomycosis tube loop mediated isothermal amplification assay caused by Trichophyton equinum transmitted from a for prevention of product cross contamination. racehorse. Br. J. Dermatol. 138(6): 1082–1084. http:// MethodsX. 1: e137-e143. http://dx.doi. dx.doi.org/10.1046/j.1365-2133.1998.02286.x org/10.1016/j.mex.2014.08.009 • Jallali N, Hettianatchy S, Gordon AC, Jain A (2011). • Karthik K, Muneeswaran NS, Manjunathachar HV, The surgical management of injectional anthrax. J. Gopi M, Elamurugan A, Kalaiyarasu S. (2014c). Plast. Recontr. Aesthet. Surg. 64(2): 276-277. http:// Bacteriophages: Effective Alternative to Antibiotics. dx.doi.org/10.1016/j.bjps.2010.06.003 Adv. Anim. Vet. Sci. 2(3S): 1 – 7. http://dx.doi. • Janse I, Hamidjaja RA, Hendriks AC, von Rotterdam org/10.14737/journal.aavs/2014/2.3s.1.7 BJ (2013). Multiplex qPCR for reliable detection • Keck N, Dutruel H, Smyej F, Nodet M, Boschiroli and differentiation ofBurkholderia mallei and ML (2010). Tuberculosis due to Mycobacterium bovis Burkholderia pseudomallei. BMC Infect. Dis. 13: 86. in a Camargue horse. Vet. Rec. 166(16): 499-500. http://dx.doi.org/10.1186/1471-2334-13-86 http://dx.doi.org/10.1136/vr.b4785 • Jones KE, Patel NG, Levy MA, Storeygard A, Balk • Khurana SK, Malik P, Virmani N, Singh BR (2009). D (2008). Global trends in emerging infectious Prevalence of Rhodococcus equi infection in foals. Ind. diseases. Nature. 451: 990-993. http://dx.doi. J. Vet. Res. 18(1): 20-22. org/10.3201/eid1409.080585 • Khurana SK (2014). Rhodococcus equi infection. In: • Jones RL, Adney WS, Shideler RK (1987). Isolation SR Garg, ed. Zoonoses: bacterial Diseases. Daya of Clostridium difficile and detection of cytotoxin Publishing House. New Delhi. India. Pp. 390-401. in the feces of diarrheic foals in the absence of • Khurana SK, Kanu Priya, Singh N, Singha H, Punia S antimicrobial treatment. J. Clin. Microbiol. 25(7): (2014). Comparative analysis of whole cell proteins 1225–1227. of Rhodococcus equi isolates using SDS-PAGE. Int. • Jubb KVF, Kennedy PC, Palmer N (1993). Pathology J. Bioassays. 3(3): 1803-1805. of Domestic Animals. Vol. 2, 4th Edn. Academic • Khurana SK (2015). Current understanding Press, San Diego. Pp. 253-255. http://dx.doi. of Rhodococcus equi infection and its zoonotic org/10.1016/B978-0-08-057133-1.50008-4 implications. Adv. Anim. Vet. Sci. 3(1): 1-10. http:// • Kahn LH, Kaplan B, Steele JH (2007). Confronting dx.doi.org/10.14737/journal.aavs/2015/3.1.1.10 zoonoses through closer collaboration between • Koff WC, Burton DR, Johnson PR, Walker BD, medicine and veterinary medicine (as ‘one King CR, Nabel GJ, Ahmed R, Bhan MK, Plotkin medicine’). Veterinaria Italiana. 43(1): 5-19. SA (2013). Accelerating next generation vaccine • Kapoor S, Dhama K, Pawaiya, RVS, Mahendran M, development for global disease prevention. Science. Mathew T (2010). Eastern, Western and Venezuelan 340 (6136): 1232910. http://dx.doi.org/10.1126/ equine encephalomyelitis. In: Advances in Medical science.1232910 and Veterinary Virology, Immunology, and • Konkle DM, Nelson KM, Lunn DP (1997). Epidemiology - Vol. 7 : Tropical Viral Diseases of Nosocomial transmission of Cryptosporidium in Large Domestic Animals- Part 1, Editor : Thankam a veterinary hospital. J. Vet. Int. Med. 11(6): 340– Mathew, Thajema Publishers, 31 Glenview Dr., 343. http://dx.doi.org/10.1111/j.1939-1676.1997. West Orange NJ 07052-1010, USA / Xlibris tb00477.x Corporation, United Kingdom, ISBN 978-1-4415- • Konstantin PL, Rena G, Javan E, Alexis L, Ray 8160-0. Pp. 90-109. Waters W, Horst P, Bodmer T, Janssens JP, Aloisio • Karthik, K (2012). Tuberculosis Goes Wild: Emphasis F, Graubner C, Grosclaude E, Piersigilli A, Schiller on Elephants. J. Vet. Adv. 2(11): 534-538. I (2012). Pulmonary Disease due to Mycobacterium • Karthik K, Kesavan M, Tamilmahan P, Saravanan tuberculosis in a Horse: Zoonotic Concerns and M and Dashprakash M (2013). Neutrophils in Limitations of Antemortem Testing. Vet. Med. Int. tuberculosis: will code be unlocked? Vet. World. http://dx.doi.org/10.1155/2012/642145 6(2): 118-121. http://dx.doi.org/10.5455/ • Krebs JW, Rupprecht CE, Childs JE (2000). Rabies vetworld.2013.118-121 surveillance in the United States during 1999. J. • Karthik K, Rathore R, Thomas P, Arun TR, Viswas Am. Vet. Med. Assoc. 217(12): 1799-1811. http:// KN, Agarwal RK, Manjunathachar HV, Dhama K dx.doi.org/10.2460/javma.2000.217.1799

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 47 Advances in Animal and Veterinary Sciences • Krebs JW, Mondul AM, Rupprecht CE, Childs, JE Appearance of multidrug-resistant virulent (2001). Rabies surveillance in the United States Rhodococcus equi clinical isolates obtained in China. during 2000. J. Am. Vet. Med. Assoc. 219: 1687–1699. J. Clin. Microbiol. 52(2): 703. http://dx.doi. http://dx.doi.org/10.2460/javma.2001.219.1687 org/10.1128/JCM.02925-13 • Kumaragurubaran K, Kaliaperumal K (2013). • Lundstrom K (2014). Alphavirus-based vaccines. DNA Vaccine: the miniature miracle. Vet. Viruses. 6(6): 2392-2415. http://dx.doi. World. 6(4): 228-232. http://dx.doi.org/10.5455/ org/10.3390/v6062392 vetworld.2013.228-232 • Mackenzie RM, De SJ, Parra D (1976). Venezuelan • Ladro´n N, Ferna´ndez M, Agu¨ero J, Zo¨rn BG, equine encephalitis virus: comparison of infectivity Va´zquez-Boland JA, Navas J (2003). Rapid and virulence of strains V-38 and P676 in donkeys. identification of Rhodococcus equi by a PCR Am. J. Trop. Med. Hyg. 25(3): 494-499. assay targeting the choE gene. J. Clin. Microbiol. • MacMillan AP, Baskerville A, Hambleton P, Corbel 41(7): 3241–3245. http://dx.doi.org/10.1128/ MJ (1982). Experimental Brucella abortus infection JCM.41.7.3241-3245.2003 in the horse: observations during the three months • Lalitha MK, Anandi V, Walter N, DevaduttaJO, following inoculation. Res. Vet. Sci. 33(3): 351–359. Pulimood BM (1988). Primary anthrax presenting • Madsen C, Hooper I, Lundberg L, Shafagati N, as an injection . Ind. J. Pathol. Microbiol. 31: Johnson A, Senina S, de la Feunte C, Hoover LI, 254-256. Fredricksen BL, Dinman J, Jacobs JL, Kuhn-Hall • Lalloo DG, Theakston RD (2003). Snake antivenoms. K (2014). Small molecule inhibitors of Ago2 J. Toxicol. Clin. Toxicol. 41: 277-90. http://dx.doi. decrease Venezuelan equine encephalitis virus org/10.1081/CLT-120021113 replication. Antiviral Res. 112: 26-37. http://dx.doi. • Lambert AJ, Martin DA, Lanciotti RS (2003). org/10.1016/j.antiviral.2014.10.002 Detection of North American and western equine • Magnusson H (1923). Spezifische infektiose encephalitis viruses by nucleic acid amplification Pneumonie beim Fohlen. Ein neuer Eiterreger beim assays. J. Clin. Microbiol. 41(1): 379-385. http:// Pferd. Arch. Wiss. Prakt. Tierhelkd. 50: 22-38. dx.doi.org/10.1128/JCM.41.1.379-385.2003 • Mahalingam S, Herrero LJ, Playford EG, Spann • Lancefield RC (1993). A serological differentiation of K, Herring B, Rolph MS, Middleton D, McCall human and other groups of hemolytic streptococci. B, Field H, Wang LF (2012). Hendra virus: an J. Exp. Med. 57(4): 571-595. emerging paramyxovirus in Australia. Lancet. Infect. • Letek M, Ocampo-Sosa AA, Sanders M, Fogarty Dis. 12(10): 799-807. http://dx.doi.org/10.1016/ U, Buckley T, Leadon DP, Gonza´lez P, Scortti M, S1473-3099(12)70158-5 Meijer WG, Parkhill J, Bentley S, Va´zquez-Boland • Mahima Rahal A, Deb R, Latheef SK, Samad HA, JA (2008). Evolution of the Rhodococcus equi vap Tiwari R, Verma AK, Kumar A, Dhama K (2012). pathogenicity island seen through comparison of Immunomodulatory and therapeutic potentials of host-associated vapA and vapB virulence plasmids. herbal, traditional / indigenous and ethnoveterinary J. Bacteriol. 190: 5797-5805. http://dx.doi. medicines. Pak. J. Biol. Sci. 15(16): 754-774. http:// org/10.1128/JB.00468-08 dx.doi.org/10.3923/pjbs.2012.754.774 • Lewis L, Taylor HG, Sorem MB, Norcross • Majewska AC, Werner A, Sulima P, Luty T (1999). JW, Kindsvatter VH (1947). Japanese B Survey on equine cryptosporidiosis in Poland and encephalitis: Clinical observation in an outbreak the possibility of zoonotic transmission. Ann. Agri. on Okinawa Shima. Arch. Neurol. Psychiatry. Environ. Med. 6: 161–165. 57(4): 430-463. http://dx.doi.org/10.1001/ • Malik P, Khurana SK, Dwivedi SK (2010). Re- archneurpsyc.1947.02300270048004 emergence of glanders in India- Report of • Lindahl S, Aspán A, Båverud V, Paillot R, Pringle Maharashtra state. Ind. J. Microbiol. 50(3): 345-348. J, Rash NL, Söderlund R, Waller AS (2013). http://dx.doi.org/10.1007/s12088-010-0027-8 Outbreak of respiratory disease in horses caused by • Malik YS, Sharma K, Jeena LM, Kumar N, Sircar S, Streptococcus equi subsp. Zooepidemicus ST-24. Rajak KK, Dhama K (2013). Toll-like receptors: the Vet. Microbiol. 166(1-2), 281-285. http://dx.doi. innate immune receptors with ingenious anti-viral org/10.1016/j.vetmic.2013.05.006 paradigm. South Asian J. Exp. Biol. 3(5): 207-213. • Linssen B, Kinney RM, Aguilar P, Russel KL, Watts • Martin DA, Muth DA, Brown T, Johnson AJ, DM, Kaaden OR, Pfeffer M (2000). Development Karabatsos N, Roehrig JT (2000). Standardization of reverse transcription-PCR assays specific for of immunoglobulin M capture enzyme-linked detection of equine encephalitis viruses. J. Clin. immunosorbent assays for routine diagnosis of Microbiol. 38(4): 1527-1535. arboviral infections. J. Clinical Microbiol. 38(5): • Liu H, Wang Y, Yan J, Wang C, He H (2014). 1823–1826. NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 48 Advances in Animal and Veterinary Sciences • McKenzie DM, Diffay BC (2000). Diarrhoea results of a household-based seroepidemiological associated with cryptosporidal oocyst shedding in a survey. Lancet. 358(9278): 261-264. http://dx.doi. quarterhorse stallion. Aust. Vet. J. 78: 27–28. http:// org/10.1016/S0140-6736(01)05480-0 dx.doi.org/10.1111/j.1751-0813.2000.tb10351.x • Mukarim A, Dechassa T, Mahendra P (2015). Equine • McCarthy AJ, Breathnach AS, Lindsay JA (2012). Bacterial and Viral Zoonosis: A Systematic Review. Detection of mobile-genetic element variation Austin. J. Trop. Med. Hyg. 1(1): 1001-1006. between colonizing and infecting hospital-associated • Murray PK (1996). The evolving story of the equine methicillin-resistant Staphylococcus aureus isolates. morbillivirus. Aust. Vet. J. 74(3): 214. http://dx.doi. J. Clin. Microbiol. 50(3): 1073–1075. org/10.1111/j.1751-0813.1996.tb15406.x • Meeusen EN, Walker J, Peter A, Pastorate PP, • Nakazawa M, Isayama Y, Kashiwazaki M, Yasui T Jungersen G (2007). Current status of veterinary (1987). Diagnosis of Rhodococcus equi in foals by vaccines. Clin. Microbiol. Rev. 20(3): 489-510. agar gel diffusion test with protein antigen. Vet. http://dx.doi.org/10.1128/CMR.00005-07 Microbiol. 41(7): 3241-3245. • Meyer KF, Haring CM, Howitt B (1931). The etiology • Napoleão F, Damasco PV, Camello TC, do Vale of epizootic encephalomyelitis in horses in San MD, de Andrade AF, Hirata RJ, de Mattos- Joachin Valley, 1930. Science. 74(1913): 227-228. Guaraldi AL (2005). Pyogenic liver abscess due • Mishra N, Kalaiyarasu S, Nagarajan S, Rao to Rhodococcus equi in an immunocompetent host. MV, George A, Sridevi R, Behera SP, Dubey J. Clin. Microbiol. 43(2):1002-1004. http://dx.doi. SC, McCracken T, Newman SH (2012). Serological org/10.1128/JCM.43.2.1002-1004.2005 evidence of West Nile virus infection in wild • Ocampo-Sosa AA, Lewis DA, Navas J, Quigley migratory and resident water birds in Eastern and F, Callejo R, Scortti M, Leadon DP, Fogarty Northern India. Comp. Immunol. Microbiol. Infect. U, Va´zquez-Boland JA (2007). Molecular Dis. 35(6): 591-8. http://dx.doi.org/10.1016/j. epidemiology of Rhodococcus equi based on traA, vapA, cimid.2012.08.002 and vapB virulence plasmid markers. J. Infect. Dis. • Mizuno Y, Sato F, Sakamoto M, Yoshikawa K, 196(5): 763-769. http://dx.doi.org/10.1086/519688 Yoshida M, Shiba K, Onodera S, Matsuura R, • OIE (2004). Glanders. In: Manual of Diagnostic Takai SJ (2005). VapB-positive Rhodococcus equi Tests and Vaccines for Terrestrial Animals, OIE infection in an HIV-infected patient in Japan. J. (Ed.). 5th Edn. OIE, Paris. Infect. Chemother. 11(1): 37-40. http://dx.doi. • OIE (2008). Glanders. In: OIE Terrestrial Manual, org/10.1007/s10156-004-0355-X OIE (Ed.). Organisation for Animal Health, New • Molaei G, Cummings RF, Su T, Armstrong PM, York. Williams GA, Cheng M, Webb JP, Andreadis • Oldfield C, Bonella H, Renwick L, Dodson HI, TG (2010). Vector-host interactions governing Alderson G, Goodfellow M (2004). Rapid epidemiology of West Nile Virus in Southern determination of vapA/vapB genotype in California. Am. J. Trop. Med. Hyg. 83(6): 1269-1282. Rhodococcus equi using a differential polymerase http://dx.doi.org/10.4269/ajtmh.2010.10-0392 chain reaction method. Antonie Van Leeuwenhoek. • Monego F, Maboni F, Krewer C, Vargas A, Costa 85(4):317-326. http://dx.doi.org/10.1023/ M, Loreto E (2009). Molecular characterization of B:ANTO.0000020383.66622.4d Rhodococcus equi from horse-breeding farms by • Paessler S, Fayzulin RZ, Anishchenko M, Greene IP, means of multiplex PCR for the vap gene family. Weaver SC, Frolov I (2003). Recombinant sindbis/ Curr. Microbiol. 58(4): 399-403. http://dx.doi. Venezuelan equine encephalitis virus is highly org/10.1007/s00284-009-9370-6 attenuated and immunogenic. J. Virol. 77(17): 9278- • Montasser AM, Saleh S, Ibrahim SI, Gilaby SE 9286. http://dx.doi.org/10.1128/JVI.77.17.9278- (1999). Recent studies on brucellosis in domestic 9286.2003 animals in Egypt. In: 5th Science Congres Egyptian • Paessler S, Weaver SC (2009). Vaccines for Society for Cattle Diseases. Assiut, Egypt. Venezuelan equine encephalitis. Vaccine. 27 • Morrison AC, Forshey BM, Notyce D, Astete H, (Suppl. 4): D80 - D85. http://dx.doi.org/10.1016/j. Lopez V, Rocha C, Carrion R, Carey C, Eza D, vaccine.2009.07.095 Mongomery JM, Kochel TJ (2008). Venezuelan • Pascoe RR (1976). Studies on the prevalence of equine encephalitis virus in Iquitos, Peru: urban ringworm among horses in racing and breeding transmission of sylvatic strain. PLoS. Negl. Trop. stables. Aust. Vet. J. 52(9): 419–421. http://dx.doi. Dis. 2: e349. http://dx.doi.org/10.1371/journal. org/10.1111/j.1751-0813.1976.tb09515.x pntd.0000349 • Paterson BJ, Mackenzie JS, Durrheim DN, Smith D • Mostashari F, Bunning ML, Kitsutani PT (2001). (2011). A review of epidemiology and surveillance Epidemic West Nile encephalitis, New York, 1999: of viral zoonotic encephalitis and the impact on

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 49 Advances in Animal and Veterinary Sciences human health in Australia. NSW Public Health 58: 197-205. http://dx.doi.org/10.1111/j.1865- Bull. 22(5-6): 99-104. http://dx.doi.org/10.1071/ 1682.2010.01196.x NB10076 • Prescott JF (1991). Rhodococcus equi an animal and • Pavlik I, Jahn P, Dvorska L, M Bartos, L Novotny, human pathogen. Clin. Microbiol. Rev. 4(1): 20-34. R Halouzka (2004). Mycobacterial infections • Preziuso, S., Laus, F., Tejeda, A.R., Valente, C. & Cuteri, in horses: a review of the literature. Veterinarni V (2010). Detection of Streptococcus dysgalactiae Medicina. 49: 427-440. subsp. equisimilis in equine nasopharyngeal swabs • Pawaiya RVS, Dhama K, Kapoor S, Mahendran M, by PCR. J. Vet. Sci. 11(1). 67-72. http://dx.doi. Mathew T (2010a). Japanese encephalitis in horses. org/10.4142/jvs.2010.11.1.67 In: Advances in Medical and Veterinary Virology, • Pusterla N, Wilson WD, Mapes S, Leutenegger CM Immunology, and Epidemiology - Vol. 7 : Tropical (2007). Diagnostic evaluation of real-time PCR Viral Diseases of Large Domestic Animals- Part 1, in the detection of Rhodococcus equi in faeces and Editor : Thankam Mathew, Thajema Publishers, 31 nasopharyngeal swabs from foals with pneumonia. Glenview Dr., West Orange NJ 07052-1010, USA Vet. Rec. 161(8): 272-275. http://dx.doi. / Xlibris Corporation, United Kingdom, ISBN 978- org/10.1136/vr.161.8.272 1-4415-8160-0. 322-344. • Quinn PJ, Carter ME, Markey B, Carter GR (1994). • Pawaiya, RVS, Dhama K, Kapoor S, Mahendran M The streptococci and related cocci. In: Clinical (2010b). Equine rabies. In: Advances in Medical Veterinary Microbiology. pp. 127-136. London: and Veterinary Virology, Immunology, and Wolfe Publishing. Mosby-Year Book Europe Epidemiology - Vol. 7 : Tropical Viral Diseases of Limited. Large Domestic Animals- Part 1, Editor : Thankam • Ratcliff RM, Chang G, Kok T, Sloots TP (2007). Mathew, Thajema Publishers, 31 Glenview Dr., Molecular diagnosis of medical viruses. Curr. West Orange NJ 07052-1010, USA / Xlibris Issues Mol. Biol. 9(2): 87-102. Corporation, United Kingdom, ISBN 978-1-4415- • Rishniw M, Liotta J, Bellosa M, Bowman D, Simpson 8160-0. Pp. 200-223 KW (2010). Comparison of 4 Giardia diagnostic • Pelkonen S, Lindahl SB, Suomala P, Karhukorpi J, tests in diagnosis of naturally acquired canine Vuorinen S, Koivula I, Vaisanen T, Pentikainen chronic subclinical giardiasis. J. Vet. Intern. Med. J, Autio T, Tuuminen T. (2013). Transmission 24(2):293-297. http://dx.doi.org/10.1111/j.1939- of Streptococcus equi subspecies zooepidemicus 1676.2010.0475.x infection from horses to humans. Emerg. Infect. • Rosen L. (1986). The natural history of Japanese Dis. 19(7): 1041-1048. http://dx.doi.org/10.3201/ encephalitis virus. Annu. Rev. Microbiol. 40: eid1907.121365 395-414. http://dx.doi.org/10.1146/annurev. • Pfeffer M, Dobler G (2010). Emergence of zoonotic mi.40.100186.002143 arboviruses by animal trade and migration. Parasit. • Saxena V, Narwal PS (2009). Rhodococcus equi infection Vectors. 3(1): 35. http://dx.doi.org/10.1186/1756- in foals. J. Remount Vet. Corps. 48: 27-31. 3305-3-35 • Schmitt B, Henderson L (2005). Diagnostic tools for • Pier AC, Zancanella PJ (1993). Immunization animal diseases. Revue scientifique et technique de of horses against dermaytophytosis caused by l office international des epizooties. 24(1): 243-250. Trichophyton equinum. Equine Pract. 15: 23–27. • Sellon DC, Besser TE, Vivrette SL, McConnico RS • Plowright RK, Eby P, Hudson PJ, Smith IL, (2001). Comparison of nucleic acid amplification, Westcott D, Bryden WL, Middleton D, Reid PA, serology, and microbiologic culture for diagnosis McFarlane RA, Martin G, Tabor GM, Skerratt of Rhodococcus equi pneumonia in foals. J. Clin. LF, Anderson DL, Crameri G, Quammen D, Microbiol. 39(4): 1289-1293. http://dx.doi. Jordan D, Freeman P, Wang LF, Epstein JH, Marsh org/10.1128/JCM.39.4.1289-1293.2001 GA, Kung NY, McCallum H (2015). Ecological • Sellon DC (2007). Miscellaneous parasitic diseases. dynamics of emerging bat virus spillover. Proc. R. Ed: DC Sellon, MT Long, WB Saunders, St Louis. Soc. B 282: 20142124. http://dx.doi.org/10.1098/ Pp. 473-480. rspb.2014.2124 • Selvey LA, Taylor R, Arklay A, Gerrard J (1996). • Porter RS, Leblond A, Lecolilinet S, Tritz P, Screening of bat carers for antibodies to morbillivirus. Cantile C, Kutasi O, Zientara S, Pradier S, van Commun. Dis. Intell. 20(22): 477-478. Galen G, Speybroek N, Saegerman C (2011). • Selvey LA, Wells RM, McCormack JG, Ansford Clinical diagnosis of West Nile fever in equids by AJ, Murray K, Rogers RJ Lavercombe PS, Selleck classification and regression tree (CART) analysis P, Sheridan JW (1995). Infection of humans and and comparative study of clinical appearance in horses by a newly described morbillivirus. Med. J. three European countries. Transbound. Emerg. Dis. Aust. 162(12): 642-645. NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 50 Advances in Animal and Veterinary Sciences • Sharma VD, Sethi MS, Yadav MP, Dube DC (1979). veterinary teaching hospital. J. Equine Vet. Sero-epidemiologic investigations on brucellosis in Sci. 32: S5–S6. http://dx.doi.org/10.1016/j. the states of Uttar Pradesh (U.P.) and Delhi (India). jevs.2012.08.015 Int. J. Zoonoses. 6(2): 75-81. • Sudhakar NR, Manjunathachar HV, Karthik K, Sahu • Singh A, Mitra M, Sampath G, Venugopal P, Rao JV, S, Gopi M, Shantaveer SB, Madhu DN, Maurya Krishnamurthy B, Gupta MK, Krishna SS, Sudhakar PS, Nagaraja KH, Shinde S, Tamilmahan P (2013). B, Rao NB, Kaushik Y, Gopinathan K, Hegde NR, RNA interference in parasites; prospects and pitfalls. Gore MM, Mohan YK, Ella KM (2015). A japanese Adv. Anim. Vet. Sci. 1(2S): 1 – 6. encephalitis vaccine from India induces durable and • Tahamtan Y, Namavari MM, Mohammadi G, Jula cross-protective immunity against temporally and GM (2010). Prevalence of brucellosis in horse spatially wide-ranging global field strains. J. Infect. north-east of Iran. J. Equine Vet. Sci. 30(7): 376- Dis. pii: jiv023. http://dx.doi.org/10.1093/infdis/ 378. http://dx.doi.org/10.1016/j.jevs.2010.05.007 jiv023 • Tauber E, Kolluritsch H, Korinek M, Rendi-Wagner • Singha H, Malik P, Goyal SK, Khurana SK, P, Jilma B, Firbas C, Schranz S, Jong E, Klingler Mukhopadyaya C, Eshwara VK, Singh RK (2014). A, Dewasthaly S, Klade CS (2007). Safety and Optimization and validation of indirect ELISA immunogenicity of a vero-cell-derived inactivated using truncated TssB protein for serodiagnosis of japanese encephalitis vaccine: a non-inferiority, glanders amongst equines. Scientific World J. 2014: phase III, randomized controlled trial. Lancet. 469407. http://dx.doi.org/10.1155/2014/469407 370(9602): 1847-1853. http://dx.doi.org/10.1016/ • Slingenbergh J, Gilbert M, de Balog K, Wint W (2004). S0140-6736(07)61780-2 Ecological sources of zoonotic diseases. Revue • Taylor KG, Paessler (2013). Pathogenesis of scientifique et technique de l office international des Venezuelan equine encephalitis. Vet. Microbiol. epizooties. 23(2): 467-484. 167(1-2): 145-150. http://dx.doi.org/10.1016/j. • Smiet E, Grinwis GCM, van den Top JGB, Sloet vetmic.2013.07.012 van Oldruitenborgh-Oosterbaan MM (2012). • TenBroeck C, Merrill MH (1933). A serological Equine mammary gland disease with a focus on difference between eastern and western equine : a review and case study. Equine encephalomyelitis virus. Proc. Soc. Exp. Biol. Med. Vet. Edu. 24: 357–366. http://dx.doi.org/10.1111/ 31: 217-220. http://dx.doi.org/10.3181/00379727- j.2042-3292.2011.00352.x 31-7066C • Smith JF, Davis K, Hart MK, Luwig GV, McClain DJ, • Theakston RD, Warrell DA, Griffiths E (2003). Parker MD, Pratt WD (1997). Medical aspects of Report of a WHO workshop on the standardization chemical and Washington, D.C.: and control of antivenoms. Toxicon. 41(5): 541-557. Office of The Surgeon General, Borden Institute, http://dx.doi.org/10.1016/S0041-0101(02)00393- Walter Reed Army Medical Centre, Office of The 8 Surgeon General United States Army. Chapter 28: • Tigrett WD, Downs, WG (1962). Studies on the virus Viral Encephalitides: Pp. 561-569. of Venezuelan equine encephalomyelitis in Trinidad, • Smithburn KC, Hughes TP, Burke AW, Paul JH W.I.I. The 1943-1944 epizootic. Am. J. Trop. Med. (1940). A neurotropic virus isolated from the blood Hyg. 11: 822-834. of a native of Uganda. Am. J. Trop. Med. 20(4): 471- • Timoney JF, Kalimuthusamy N, Velineni S, Donahue 492. JM, Artiushin SC, Fettinger M (2011). A unique • Snyder SP, England JJ, McChesney AE (1978). genotype of Leptospira interrogans serovar Pomona Cryptosporidiosis in immunodeficient Arabian type kennewicki is associated with equine abortion. foals. Vet. Pathol. 15: 12–17. http://dx.doi. Vet. Microbiol. 150(3-4): 349–353. http://dx.doi. org/10.1177/030098587801500102 org/10.1016/j.vetmic.2011.02.049 • Strauss JH, Calisher CH, Dalgarno L, Dalrymple, • Tiwari R, Chakraborty S, Dhama K, Rajagunalan S, JM, Frey TK, Petterson RF, Rice CM, Spaan WJM Singh SV (2013). Antibiotic resistance- an emerging (1995). In: Murphy FA, Fauquet CM, Bishop health problem: causes, worries, challenges and DHL, Ghabrial SA, Jarwis, AW, Martelli GP, Mayo solutions- a review. Int. J. Curr. Res. 5(7): 1880-1892 MA, Summers MD, editors. Virus Taxonomy. • Tiwari R, Chakraborty S, Dhama K, Wani MY, Kumar Sixth Report of the International Committee on A, Kapoor S (2014). Wonder world of phages: Taxonomy of Viruses. New York: Springer-Verlag. potential biocontrol agents safeguarding biosphere p. 428-433. and health of animals and humans - current scenario • Stull JW, Slavic´ D, Rousseau J, Weese JS (2012). and perspectives. Pak. J. Biol. Sci. 17(3): 316-328. Staphylococcus delphini and methicillin-resistant http://dx.doi.org/10.3923/pjbs.2014.316.328 Staphylococcus pseudintermedius in horses at a • Tretyakova I, Lukashevich IS, Glass P, Wang E, Weaver

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 51 Advances in Animal and Veterinary Sciences S, Pushko P (2013). Novel vaccine Venezuelan http://dx.doi.org/10.1093/infdis/128.3.271 equine encephalitis combines advantages of DNA • Watts DM, Callahan J, Rossi C, Oberste MS, Roehrig immunization and a live attenuated vaccine. Vaccine. JT, Wooster MT, Smith JF, Cropp CB, Gentrau 31(7): 1019-1025. http://dx.doi.org/10.1016/j. EM, Karabatsos N, Gubler DJ, Hayes CG (1998). vaccine.2012.12.050 Venezuelan equine encephalitis febrile cases among • Trobaugh DW, Ryman KD, Klimstra WB (2014). humans in the Peruvian Amazon river region. Am. Can understanding the virulence mechanisms of J. Trop. Med. Hyg. 58(1): 35-40. RNA viruses lead us to a vaccine against eastern • Watts DM, Lavera V, Callahan J, Rossi C, Oberste equine encephalitis virus and other alphaviruses? MS, Roehrig JT, Cropp CB, Karabatsos N, Smith Expert Rev. Vaccines. 13(12): 1423-1425. http:// JF, Gubler DJ, Wooster MT, Nelson WM, Hayes dx.doi.org/10.1586/14760584.2014.944168 CG (1997). Oropouche virus infections among • Tsarev SA, Sanders ML, Vaughn DW, Innis BL Peruvian army troopsin the Amazon region of Peru. (2000). Phylogenetic analysis suggests only one Am. J. Trop. Med. Hyg. 56(6): 661-667. serotype of Japanese encephalitis virus. Vaccine. • Weaver SC, Barrett AD (2004). Transmission cycles, 18(Suppl 2): 36-43. http://dx.doi.org/10.1016/ host range, evolution and emergence of arboviral S0264-410X(00)00039-6 disease. Nat. Rev. Microbiol. 2(10): 789-801. http:// • Van Duijkeren E, Ten Horn L, Wagenaar JA, de dx.doi.org/10.1038/nrmicro1006 Bruijn M, Laarhoven L, Verstappen K, de Weerd • Weese JS, Baird JD, Poppe C, Archambault M W, Meessen N, Duim B (2011). Suspected horse- (2001a). Emergence of Salmonella typhimurium to-human transmission of MRSA ST398. Emerg. definitive type 104 (DT104) as an important cause Infect. Dis. 17(6): 1137–1139. http://dx.doi. of salmonellosis in horses in Ontario. Can. Vet. J. org/10.3201/eid1706.101330 42(10): 788–792. • Van-den Hurk AF, Ritchie SA, Johansen CA, • Weese JS, Staempfli HR, Prescott JF (2001b). A Mackenzie JS, Smith GA (2008). Domestic pigs prospective study of the roles of Clostridium difficile and Japanese encephalitis virus infection, Australia. and enterotoxigenic Clostridium perfringens in Emerg. Infect. Dis. 14(11): 1736-1738. http:// equine diarrhoea. Equine Vet. J. 33(4): 403–409. dx.doi.org/10.3201/eid1411.071368 http://dx.doi.org/10.2746/042516401776249534 • Van-den Hurk AF, Ritchie SA, Mackenzie JS • Weese JS (2002). A review of equine zoonotic diseases: (2009). Ecology and geographical expansion of risks in veterinary medicine. AEEP Proceedings. 48: Japanese encephalitis virus. Annu. Rev. Entomol. 362-369. 54: 17-35. http://dx.doi.org/10.1146/annurev. • Weese JS, Rousseau J, Traub-Dargatz JL, Willey ento.54.110807.090510 BM, McGeer AL, Low DE (2005). Community- • Varga JJ, Vigil A, DeShazer D, Waag DM, Felgner Associated methicillin-resistant Staphylococcus P, Goldberg JB (2012). Distinct human antibody aureus in horses and humans who work with horses. response to the biological warfare agent Burkholderia J. Am. Vet. Med. Assoc. 226: 580-583. http://dx.doi. mallei. Virulence. 3(6): 510-514. http://dx.doi. org/10.2460/javma.2005.226.580 org/10.4161/viru.22056 • Weese JS, Caldwell F, Willey BM, Kreiswirth BN, • Verma A, Stevenson B, Adler B (2013). Leptospirosis McGeer A, Rousseau J, Low DE (2006). An in horses. Vet. Microbiol. 167: 61–66. http://dx.doi. outbreak of methicillin-resistant Staphylococcus org/10.1016/j.vetmic.2013.04.012 aureus skin infections resulting from horse to human • Verma AK, Saminathan M, Neha, Tiwari R, Dhama transmission in a veterinary hospital. Vet. Microbiol. K, Singh SV (2014a). Glanders-A re-emerging 114(1-2): 160-164. http://dx.doi.org/10.1016/j. zoonotic disease: a review. J. Biol. Sci. 14(1): 38-51. vetmic.2005.11.054 http://dx.doi.org/10.3923/jbs.2014.38.51 • Weese JS, Lefebvre SL (2007). Risk factors • Verma AK, Dhama K, Chakraborty S, Kumar A, for methicillin-resistant Staphylococcus aureus Tiwari R, Rahal A, Mahima, Singh SV (2014b). colonization in horses admitted to a veterinary Strategies for combating and eradicating important teaching hospital. Can. Vet. J. 48(9): 921–926. infectious diseases of animals with particular • Weinstock DM, Brown AE (2002). Rhodococcus equi: reference to India: Present and future perspectives. An emerging pathogen. Clin. Infect. Dis. 34(10): Asian J. Anim. Vet. Adv. 9(2): 77-106. http://dx.doi. 1379-1385. http://dx.doi.org/10.1086/340259 org/10.3923/ajava.2014.77.106 • Williamson MM, Hooper PT, Selleck PW, Gleeson • Walton TE, Alvarez JO, Buckwalter RM, Johnson LJ, Daniels PW, Westbury HA, Murray PK KM (1973). Experimental infection of horses with (1998). Transmission studies of Hendra virus enzootic and epizootic strains of Venezuelan equine (equine morbillivirus) in fruit bats, horses and encephalomyelitis virus. J. Infect. Dis. 128: 271-282. cats. Aust. Vet. J. 76(12): 813–818. http://dx.doi.

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 52 Advances in Animal and Veterinary Sciences org/10.1111/j.1751-0813.1998.tb12335.x Dis. 12(5): 372-379. http://dx.doi.org/10.1089/ • Xiao L, Feng Y (2008). Zoonotic crytosporidiosis. vbz.2011.0709 FEMS Immunol. Med. Microbiol. 52(3): • Young NA (1972). Origin of epidemics of Venezuelan 309-323. http://dx.doi.org/10.1111/j.1574- equine encephalitis. J. Infect. Dis. 125(5): 565-567. 695X.2008.00377.x http://dx.doi.org/10.1093/infdis/125.5.565 • Xiao L, Herd RP (1994). Epidemiology of equine • Yousefi-Nooraie R, Mortaz-Hejri S, Mehrani M, Cryptosporidium and Giardia infections. Sadeghipour P (2012). Antibiotics for treating Equine Vet. J. 26(1): 14–17. http://dx.doi. human brucellosis. Cochrane Database of Systematic org/10.1111/j.2042-3306.1994.tb04322.x Reviews. 10. http://dx.doi.org/10.1002/14651858. • Yager JA, Prescott CA, Kramar DP, Hannah H, Yager CD007179.pub2 JA, Prescott CA, Kramar DP, Hannah H, Balson • Zacks MA, Paessler S (2010). Encephalitic GA, Croy B.A (1991). The effect of experimental alphaviruses. Vet. Microbiol. 140(3-4): 281-286. infection with Rhodococcus equi on immunodeficient http://dx.doi.org/10.1016/j.vetmic.2009.08.023 mice. Vet. Microbiol. 28(4): 363-376. http://dx.doi. • Zhang B, Wear DJ, Kim HS, Weina P, Stojadinovic org/10.1016/0378-1135(91)90071-M A,Izadjoo M (2012). Development of hydrolysis • Yeh J, Lee J, Park J, Seo H, Park J, Moon J, Cho I, Lee probe based real-time PCR for identification of J, Park S, Song C, Choi I (2010). Fast duplex one- virulent gene targets of Burkholderia pseudomallei step reverse transcriptase PCR for rapid differential and B. mallei-a retrospective study on archival cases detection of west Nile and Japanese encephalitis. J. of service members with melioidosis and glanders. Clin. Microbiol. 48(11): 4010-4014. http://dx.doi. Military Med. 177: 216-221. http://dx.doi. org/10.1128/JCM.00582-10 org/10.7205/MILMED-D-11-00232 • Yeh J, Lee J, Park J, Seo H, Moon J, Cho I, Kim H, • Zinsstag J, Schelling E, Roth F, Bonfoh B, De Yang Y, Ahn K, Kyung S, Choi I, Lee J (2012). A Savigny D, Tanner M (2007). Human benefits of diagnostic algorithm to serologically differentiate animal interventions for zoonosis control. Emerg. west Nile virus from Japanese encephalitis virus Infect. Dis. 13: 527-531. http://dx.doi.org/10.3201/ infections and its validation in field surveillance eid1304.060381 of poultry and horse. Vector-Borne Zoonotic

NE US Academic Publishers March 2015 | Volume 3 | Special issue 2 | Page 53