Neorickettsia risticii

Last updated

Neorickettsia risticii
Neorickettsia risticii phylogenetic tree.jpg
Neorickettsia risticii phylogenetic tree based on 16S rRNA gene sequences
Scientific classification
Domain:
Phylum:
Class:
Order:
Family:
Genus:
Species:
N. risticii
Binomial name
Neorickettsia risticii (Dumler et al., 2001)
Synonyms

Ehrlichia risticii

Neorickettsia risticii, formerly Ehrlichia risticii, is an obligate intracellular gram negative bacteria that typically lives as an endosymbiont in parasitic flatworms, specifically flukes. [1] N. risticii is the known causative agent of equine neorickettsiosis (also known as Potomac horse fever (PHF)), [2] which gets its name from its discovery near the Potomac River in Maryland and Virginia. [3] N. risticii was first recovered from horses in this region in 1984 but was not recognized as the causative agent of PHF until 1979. [3] Potomac horse fever is currently endemic in the United States but has also been reported with lower frequency in other regions, including Canada, Brazil, Uruguay, and Europe. [4] PHF is a condition that is clinically important for horses since it can cause serious signs such as fever, diarrhea, colic, and laminitis. [5] PHF has a fatality rate of approximately 30%, making this condition one of the concerns for horse owners in endemic regions [5] N. risticii is typically acquired in the middle to late summer near freshwater streams or rivers, as well as on irrigated pastures. [6] This is a seasonal infection because it relies on the ingestion of an arthropod vector, which is more commonly found on pasture in the summer months. [7] Although N. risticii is a well known causative agent for PHF in horses, it may act as a potential pathogen in cats and dogs as well. [8] Not only has N. risticii been successfully cultured from monocytes of dogs and cats, [6] but cats have become clinically ill after experimental infection with the bacteria. [9] In addition, N. risticii has been isolated and cultured from human histiocytic lymphoma cells. [6]

Contents

Taxonomy, morphology, and identification

Neorickettsia risticii is a member of the Order Rickettsiales and Family Anaplasmataceae within the Class Alphaproteobacteria. [10] N. risticii are obligate intracellular organisms [10] of equine monocytes/macrophages and glandular intestinal epithelium. [6] Individual organisms are small, pleomorphic (coccoid to ellipsoidal) gram negative aerobes that lack LPS and peptidoglycan in their cell walls. [11] [12] They have two cell forms: a reticular body type form that is large and light, and a smaller elementary body type form that is electron-dense. [6] [12] [10] They can exist as individual cells or as morulae (colonies similar in appearance to mulberries) and replicate in parasitophorous vacuoles via binary fission within the host cell. [12] [10] Identification can be done based on cell culture isolation, serology, or PCR assay. [1] [6] [13] However, N. risticii is rarely identified in monocytes from peripheral blood smears. [6] PCR assays can be performed on blood and feces (antemortem identification) or on fresh or formalin fixed tissues (postmortem identification). [6] N. risticii can be viewed using light microscopy with a variety of stains: Giemsa and Romanowsky stains will mark the bacteria dark blue to purple, [12] Machiavello will stain red, and hematoxylin eosin (H&E) will stain pale blue. [6]

Transmission

Proposed life cycle and mode of transmission of Neorickettsia risticii. Proposed life cycle and mode transmission of Neorickettsia risticii.png
Proposed life cycle and mode of transmission of Neorickettsia risticii.

The exact mode of transmission of N. risticii has been undetermined for many years due to its complex life cycle. N. risticii is transmitted vertically inside a trematode (fluke), Acanthatrium oregonense, and the transmission of N. risticii is closely related to the life cycle of the fluke. [14] [15] As the fluke develops and gets into the next host, N. risticii is passed on to the next host along with it. [15] The proposed mode of transmission involves two intermediate hosts (an aquatic snail and an aquatic insect such as caddisflies and mayflies). [14] The egg of the fluke is ingested by an aquatic snail. Then, the cercaria form of the fluke develops and emerges from the snail into the environment. [14] The cercaria then invades the second intermediate host, the larval or nymph stage of an aquatic insect. The cercaria develops into a metacercaria as the insect develops into an adult. [14] The adult insect is then ingested by an insectivore such as a bat or a swallow. [14] [15] It is thought that bats and swallows serve as the definitive hosts for the fluke and natural reservoirs of N. risticii. [14]

It is proposed that horses can be exposed to N. risticii through two routes. Horses can be exposed through skin penetration by infected cercaria form of the fluke. This is commonly seen when horses are grazing near contaminated water sources. [16] Another route of exposure is through directly ingesting the second intermediate host, aquatic insects, containing the infected metacercariae. [14]

Pathogenesis

It is proposed that N. risticii is an obligate intracellular bacterium which prefer replicating in monocytes of the host. [17] Once phagocytized by the monocyte, the pathogen prevents lysosomal fusion with the phagosome thus escaping the host defense mechanism. [14] Once N. risticii establishes infection inside the host, the primary targets are the epithelial cells, mast cells, and macrophages located in the host's large intestine. [14] The infected intestinal epithelial cells suffer from impaired electrolyte transport and loss of microvilli, which contribute to decreased electrolyte absorption and increased water loss in the large intestine. [18] Therefore, diarrhea is one of the common clinical signs of equine neorickettsiosis.

Clinical significance

Neorickettsia risticii is the infectious cause of Equine neorickettsiosis, or the colloquially termed Potomac Horse Fever (PHF). [5] This disease is acquired when horses ingest a trematode host that is infected with the bacteria. [5] Because of the presence of the arthropod vector, it seems that this disease is of concern in the summer, when warmer weather permits these vectors to be present on pasture. [7] N. risticii is able to stay inside these trematodes through their development stages, and can also be transmitted to future generations through a transovarial transmission route. [11] Once infected, the horse is not contagious to other horses, as the infection must be spread by the intermediate host ingestion. [5]

This disease can cause horses to become feverish, experience liquid diarrhea, show a quiet demeanor and go off their food, which can lead to colic and laminitis. [5] Intestinal lesions previously seen with PHF include pronounced enterocolitis with ulcerative erosions and evident reduction in villus projections. [19] The lesions will result in the horse having severe gastrointestinal disease, and reduced ability to absorb nutrients. Lesions that were linked to the presence of the bacteria in the blood include hepatitis, kidney tubule inflammation, loss of renal arterial blood supply, and inflammation of the adrenal glands. [19] There are also changes to the blood in response to infection, and early PHF can result in reduced white blood cells, including low neutrophils and low lymphocytes, while progression of PHF can result in an inflammatory response and increased white blood cells. [19] Replication of N. risticii happens within the epithelial lining of the intestines, and within myeloid cells such as macrophages, monocytes, and mast cells. [11] The condition can be fatal in 3 out of 10 horses that are infected, but up to 1/3 of horses infected have been shown to be asymptomatic, so there is variability in the disease presentation. [5] Due to the severe symptoms associated with the disease, and fatality rate, this condition is of concern for horse owners.

If a pregnant mare is infected with N. risticii and has symptoms of PHF, there is a chance that it may cause abortion and lesions in the fetus. Studies have been done on mares that were infected either naturally or artificially with N. risticii and experienced abortion. The fetuses had lesions of liver inflammation, heart inflammation, enterocolitis, and inflammation of the mesenteric lymph nodes. [20] Abortion seems to be a notable complication of PHF, as was showcased in a study that infected pregnant mares with N. risticii resulting in abortion in just over half of the subjects. [20] This infection does not cause inflammation of the placenta, and lesions are generally absent from the placenta or very minor, yet it does cause lesions in the fetus itself, which is quite unique. [20] PHF is not a well documented cause for equine abortion, and is often not chosen as a differential diagnosis. [20] However, it could be argued that N. risticii requires more research in this area, and could be a more common cause of equine abortion than was previously thought.

Diagnostics

Diagnosis of Potomac Horse Fever (PHF) is most commonly based on serological techniques and therefore requires collecting a blood sample from an infected horse. Neorickettsia risticii is detected in the blood using an indirect immunofluorescent antibody (IFA) test or by PCR identification. [21] The IFA serological test detects the presence of IgG and IgM antibodies against N. risticii in the blood, however does not differentiate whether an animal is actively infected or has had previous exposure. [22] The use of IFA at a single point in time does not provide an accurate diagnosis and therefore requires IFA testing at multiple time points to detect increasing antibody titres. Increasing antibody titres in conjunction with clinical signs is suggestive of an active PHF infection. [22] PCR assays can be performed on both the blood and stool of an infected animal and allows the detection of N. risticii in these samples. [4] PCR is the test of choice in vaccinated animals because these animals will have antibodies against N. risticii , which interferes with the IFA test. [22]

Cell culture isolation can also be used for the detection and isolation of Neorickettsia risticii, although it is a more time-consuming technique that requires specialized equipment and is not routinely used as a diagnostic method. [1] N. risticii is an obligate intracellular microorganism and as such is more technically difficult to culture, isolate, and ship to a diagnostic laboratory. [4] Cell culture isolation detects the presence of live bacteria and can be used to isolate bacteria from an infected animal, the environment, or a parasitic host. [1] Strain variation among isolates can be determined by identifying surface antigens or by performing whole-genome sequencing. [4] Molecular analysis surface antigens has found significant genetic and phenotypic variation in strains of N. risticii, these variations may contribute to vaccination failures. [23]

Treatment and prevention

The main clinical presentation of Neorickettsia risticii is Potomac Horse Fever (PHF). Timely diagnosis and treatment of PHF is important for preventing the disease from progressing and causing clinical signs such as laminitis, endotoxemia and colic. Successfully preventing this progression can greatly increase an infected horse's chance of survival. [24] Treatment of PHF utilizes a combination of antimicrobial and supportive therapy. [25] Oxytetracycline has been found to be the most successful antimicrobial treatment for PHF and has been correlated to increased survival time. [24] When treatment is delayed until after the onset of clinical signs, additional therapy with doxycycline, demeclocycline or rifampin has been associated with higher antibody titer levels than those of untreated animals. [26] In addition to antimicrobial treatment, supportive therapy for pain management, dehydration and gastrointestinal function should also be utilized to treat laminitis, endotoxemia and colic, respectively. Laminitis can be treated with a combination of cryotherapy and an opioid, such as butorphanol or morphine. [27] Intravenous lactated Ringer's solution can be given for fluid and electrolyte replenishment and Flunixin Meglumine can be used to treat signs of colic, both of which can help offset signs of endotoxemia and prevent endotoxic shock. [4]

There are currently two types of vaccines for Potomac Horse Fever available. One is an inactive monovalent vaccine that protects against PHF only, and the other is an inactive multivalent vaccine that is combined with a rabies vaccination. There is some evidence to show that the multivalent PHF/rabies vaccine provides lower immunogenicity in contrast to the monovalent PHF vaccine when it is given with a separate rabies vaccine. [28] The efficacy of these vaccines has been reported to be quite poor due to the fact that they contain only one of several strains of N. risticii, but studies have also shown that vaccinated horses that do become infected tend to show less severe clinical signs when compared to unvaccinated horses. [29] The vaccines are recommended to be given annually, but horses in endemic regions can be given a second dose 3–4 weeks following the initial vaccine to help increase the effectiveness. Pregnant mares should also be given an additional vaccine 4–6 weeks prior to foaling. [30]

See also

Related Research Articles

Ehrlichiosis is a tick-borne disease of dogs usually caused by the rickettsial agent Ehrlichia canis. Ehrlichia canis is the pathogen of animals. Humans can become infected by E. canis and other species after tick exposure. German Shepherd Dogs are thought to be susceptible to a particularly severe form of the disease; other breeds generally have milder clinical signs. Cats can also be infected.

<span class="mw-page-title-main">Babesiosis</span> Malaria-like parasitic disease caused by infection with the alveoate Babesia or Theileria

Babesiosis or piroplasmosis is a malaria-like parasitic disease caused by infection with a eukaryotic parasite in the order Piroplasmida, typically a Babesia or Theileria, in the phylum Apicomplexa. Human babesiosis transmission via tick bite is most common in the Northeastern and Midwestern United States and parts of Europe, and sporadic throughout the rest of the world. It occurs in warm weather. People can get infected with Babesia parasites by the bite of an infected tick, by getting a blood transfusion from an infected donor of blood products, or by congenital transmission . Ticks transmit the human strain of babesiosis, so it often presents with other tick-borne illnesses such as Lyme disease. After trypanosomes, Babesia is thought to be the second-most common blood parasite of mammals. They can have major adverse effects on the health of domestic animals in areas without severe winters. In cattle, the disease is known as Texas cattle fever or redwater.

H3N8 is a subtype of the species Influenza A virus that is endemic in birds, horses and dogs. It is the main cause of equine influenza and is also known as equine influenza virus. In 2011, it was reported to have been found in seals. Cats have been experimentally infected with the virus, leading to clinical signs, shedding of the virus and infection of other cats. In 2022 and 2023, three people in China were infected with H3N8, with one fatality, marking the first time a human has died from this strain of flu.

<span class="mw-page-title-main">Bovine malignant catarrhal fever</span> Species of virus

Bovine malignant catarrhal fever (BMCF) is a fatal lymphoproliferative disease caused by a group of ruminant gamma herpes viruses including Alcelaphine gammaherpesvirus 1 (AlHV-1) and Ovine gammaherpesvirus 2 (OvHV-2) These viruses cause unapparent infection in their reservoir hosts, but are usually fatal in cattle and other ungulates such as deer, antelope, and buffalo. In Southern Africa the disease is known as snotsiekte, from the Afrikaans.

<span class="mw-page-title-main">Equine protozoal myeloencephalitis</span> Central nervous system disease of horses

Equine protozoal myeloencephalitis (EPM) is a disease that affects the central nervous system of horses. It is caused by a protozoal infection that is brought about by the apicomplexan parasites Sarcocystis neurona or Neospora hughesi. Most cases are caused by S. neurona. The lifecycle and transmission of N. hughesi is not well understood. The parasites create lesions in both the brain and spinal cord of the affected horses leading to neurological issues. Most horses infected with S. neurona do not exhibit neurological symptoms consistent with EPM.

<i>Anaplasma phagocytophilum</i> Species of bacterium

Anaplasma phagocytophilum is a Gram-negative bacterium that is unusual in its tropism to neutrophils. It causes anaplasmosis in sheep and cattle, also known as tick-borne fever and pasture fever, and also causes the zoonotic disease human granulocytic anaplasmosis.

<span class="mw-page-title-main">Anaplasmosis</span> Medical condition

Anaplasmosis is a tick-borne disease affecting ruminants, dogs, and horses, and is caused by Anaplasma bacteria. Anaplasmosis is an infectious but not contagious disease. Anaplasmosis can be transmitted through mechanical and biological vector processes. Anaplasmosis can also be referred to as "yellow bag" or "yellow fever" because the infected animal can develop a jaundiced look. Other signs of infection include weight loss, diarrhea, paleness of the skin, aggressive behavior, and high fever.

<span class="mw-page-title-main">Ehrlichiosis</span> Medical condition

Ehrlichiosis is a tick-borne bacterial infection, caused by bacteria of the family Anaplasmataceae, genera Ehrlichia and Anaplasma. These obligate intracellular bacteria infect and kill white blood cells.

Potomac Horse Fever (PHF) is a potentially-fatal febrile illness affecting horses caused by the intracellular bacterium Neorickettsia risticii. PHF is also known as Shasta River Crud and Equine Monocytic Ehrlichiosis. It was first described in areas surrounding the Potomac River northwest of Washington, D.C., in the 1980s, but cases have been described in many other parts of the United States, such as Minnesota, California, and Pennsylvania. Currently, it is found in more than 40 U.S. states and Canada.

Equine viral arteritis (EVA) is a disease of horses caused by a virus of the species Alphaarterivirus equid, an RNA virus. It is the only species in the genus Alphaarterivirus, and that is the only genus in the Equarterivirinae subfamily. The virus which causes EVA was first isolated in 1953, but the disease has afflicted equine animals worldwide for centuries. It has been more common in some breeds of horses in the United States, but there is no breed "immunity". In the UK, it is a notifiable disease. There is no known human hazard.

<span class="mw-page-title-main">Antibody-dependent enhancement</span> Antibodies rarely making an infection worse instead of better

Antibody-dependent enhancement (ADE), sometimes less precisely called immune enhancement or disease enhancement, is a phenomenon in which binding of a virus to suboptimal antibodies enhances its entry into host cells, followed by its replication. The suboptimal antibodies can result from natural infection or from vaccination. ADE may cause enhanced respiratory disease, but is not limited to respiratory disease. It has been observed in HIV, RSV virus and Dengue virus and is monitored for in vaccine development.

Neorickettsia is a genus of bacteria. Species or strains in this genus are coccoid or pleomorphic cells that reside in cytoplasmic vacuoles within monocytes and macrophages of dogs, horses, bats, and humans.

Ehrlichia chaffeensis is an obligate intracellular, Gram-negative species of Rickettsiales bacteria. It is a zoonotic pathogen transmitted to humans by the lone star tick. It is the causative agent of human monocytic ehrlichiosis.

<i>Ehrlichia ewingii</i> Species of bacterium

Ehrlichia ewingii is a species of rickettsiales bacteria. It has recently been associated with human infection, and can be detected via PCR serological testing. The name Ehrlichia ewingii was proposed in 1992.

Ehrlichiosis ewingii infection is an infectious disease caused by an intracellular bacteria, Ehrlichia ewingii. The infection is transmitted to humans by the tick, Amblyomma americanum. This tick can also transmit Ehrlichia chaffeensis, the bacteria that causes human monocytic ehrlichiosis (HME).

<span class="mw-page-title-main">Mosquito-borne disease</span> Diseases caused by bacteria, viruses or parasites transmitted by mosquitoes

Mosquito-borne diseases or mosquito-borne illnesses are diseases caused by bacteria, viruses or parasites transmitted by mosquitoes. Nearly 700 million people get a mosquito-borne illness each year, resulting in over tens million deaths. The devastation is almost equivalent to the entire 3 year COVID-19 global pandemic.

<i>Juga</i> Genus of gastropods

Juga is a genus of freshwater snails with a gill and an operculum, aquatic gastropod mollusks in the family Semisulcospiridae.

Ehrlichia canis is an obligate intracellular bacterium that acts as the causative agent of ehrlichiosis, a disease most commonly affecting canine species. This pathogen is present throughout the United States, South America, Asia, Africa and recently in the Kimberley region of Australia. First defined in 1935, E. canis emerged in the United States in 1963 and its presence has since been found in all 48 contiguous United States. Reported primarily in dogs, E. canis has also been documented in felines and humans, where it is transferred most commonly via Rhipicephalus sanguineus, the brown dog tick.

Purpura haemorrhagica is a rare complication of equine strangles and is caused by bleeding from capillaries which results in red spots on the skin and mucous membranes together with oedema (swelling) of the limbs and the head. Purpura hemorrhagica is more common in younger animals.

Anaplasma bovis is gram negative, obligate intracellular organism, which can be found in wild and domestic ruminants, and potentially a wide variety of other species. It is one of the last species of the Family Anaplasmaceae to be formally described. It preferentially infects host monocytes, and is often diagnosed via blood smears, PCR, and ELISA. A. bovis is not currently considered zoonotic, and does not frequently cause serious clinical disease in its host. This organism is transmitted by tick vectors, so tick bite prevention is the mainstay of A. bovis control, although clinical infections can be treated with tetracyclines. This organism has a global distribution, with infections noted in many areas, including Korea, Japan, Europe, Brazil, Africa, and North America.

References

  1. 1 2 3 4 Teymournejad O, Lin M, Bekebrede H, Kamr A, Toribio RE, Arroyo LG, et al. (February 2020). "Neorickettsia Species That Causes Potomac Horse Fever". mBio. 11 (1) (published 25 February 2020). doi:10.1128/mBio.03429-19. ISSN   2150-7511. PMC   7042704 . PMID   32098825. Wikidata   Q89881646. (erratum)
  2. Gibson KE, Rikihisa Y, Zhang C, Martin C (February 2005). "Neorickettsia risticii is vertically transmitted in the trematode Acanthatrium oregonense and horizontally transmitted to bats". Environmental Microbiology. 7 (2): 203–12. doi:10.1111/j.1462-2920.2004.00683.x. PMID   15658987.
  3. 1 2 Chaichanasiriwithaya W, Rikihisa Y, Yamamoto S, Reed S, Crawford TB, Perryman LE, Palmer GH (December 1994). "Antigenic, morphologic, and molecular characterization of new Ehrlichia risticii isolates". Journal of Clinical Microbiology. 32 (12): 3026–33. doi:10.1128/JCM.32.12.3026-3033.1994. PMC   264219 . PMID   7533780.
  4. 1 2 3 4 5 Xiong Q, Bekebrede H, Sharma P, Arroyo LG, Baird JD, Rikihisa Y (October 2016). Dozois CM (ed.). "An Ecotype of Neorickettsia risticii Causing Potomac Horse Fever in Canada". Applied and Environmental Microbiology. 82 (19): 6030–6. doi:10.1128/AEM.01366-16. PMC   5038023 . PMID   27474720.
  5. 1 2 3 4 5 6 7 Roier, Erica C.R.; Costa, Renata L.; Pires, Marcus S.; Vilela, Joice A.R.; Santos, Tiago M. dos; Santos, Huarrisson A.; Baldani, Cristiane D.; Massard, Carlos L. (October 2016). "Epidemiological survey of Neorickettsia risticii in equids from the State of Rio de Janeiro, Brazil". Pesquisa Veterinária Brasileira. 36 (10): 939–946. doi: 10.1590/S0100-736X2016001000004 .
  6. 1 2 3 4 5 6 7 8 9 Pusterla N, Madigan JE (2014). "Neorickettsia risticii". Equine Infectious Diseases. Elsevier. pp. 347–351.e2. doi:10.1016/b978-1-4557-0891-8.00040-3. ISBN   978-1-4557-0891-8.
  7. 1 2 McLaughlin B, Gough J (June 1996). "Potomac horse fever in southwestern Ontario". The Canadian Veterinary Journal. 37 (6): 367–8. PMC   1576412 . PMID   8689598.
  8. Ayllón T, Diniz PP, Breitschwerdt EB, Villaescusa A, Rodríguez-Franco F, Sainz A (February 2012). "Vector-borne diseases in client-owned and stray cats from Madrid, Spain". Vector Borne and Zoonotic Diseases. 12 (2): 143–50. doi:10.1089/vbz.2011.0729. PMID   22022820.
  9. Lappin MR, Griffin B, Brunt J, Riley A, Burney D, Hawley J, et al. (April 2006). "Prevalence of Bartonella species, haemoplasma species, Ehrlichia species, Anaplasma phagocytophilum, and Neorickettsia risticii DNA in the blood of cats and their fleas in the United States". Journal of Feline Medicine and Surgery. 8 (2): 85–90. doi:10.1016/j.jfms.2005.08.003. PMID   16290092. S2CID   19187668.
  10. 1 2 3 4 Yu XJ, Walker DH (2006). "The Order Rickettsiales". In Dworkin M, Falkow S, Rosenberg E, Schleifer KH (eds.). The Prokaryotes. New York, NY: Springer New York. pp. 493–528. doi:10.1007/0-387-30745-1_20. ISBN   978-0-387-25495-1.
  11. 1 2 3 Lin M, Zhang C, Gibson K, Rikihisa Y (October 2009). "Analysis of complete genome sequence of Neorickettsia risticii: causative agent of Potomac horse fever". Nucleic Acids Research. 37 (18): 6076–91. doi:10.1093/nar/gkp642. PMC   2764437 . PMID   19661282.
  12. 1 2 3 4 Rikihisa Y (October 2006). "New findings on members of the family Anaplasmataceae of veterinary importance". Annals of the New York Academy of Sciences. 1078 (1): 438–45. Bibcode:2006NYASA1078..438R. doi:10.1196/annals.1374.083. PMID   17114752. S2CID   21179916.
  13. Lappin MR, Griffin B, Brunt J, Riley A, Burney D, Hawley J, et al. (April 2006). "Prevalence of Bartonella species, haemoplasma species, Ehrlichia species, Anaplasma phagocytophilum, and Neorickettsia risticii DNA in the blood of cats and their fleas in the United States". Journal of Feline Medicine and Surgery. 8 (2): 85–90. doi:10.1016/j.jfms.2005.08.003. PMID   16290092. S2CID   19187668.
  14. 1 2 3 4 5 6 7 8 9 Sellon DC, Long MT (2014). "Acknowledgments". Equine Infectious Diseases. Elsevier. pp. xi. doi:10.1016/b978-1-4557-0891-8.00078-6. ISBN   978-1-4557-0891-8.
  15. 1 2 3 Madigan JE, Pusterla N, Johnson E, Chae JS, Pusterla JB, Derock E, Lawler SP (July 2000). "Transmission of Ehrlichia risticii, the agent of Potomac horse fever, using naturally infected aquatic insects and helminth vectors: preliminary report". Equine Veterinary Journal. 32 (4): 275–9. doi:10.2746/042516400777032219. PMID   10952374.
  16. "Potomac Horse Fever: A Water-borne Equestrian Worry". Horse Canada. Retrieved 2020-11-15.
  17. "Potomac Horse Fever - Digestive System". Merck Veterinary Manual. Retrieved 2020-11-15.
  18. Rikihisa Y, Johnson GC, Wang YZ, Reed SM, Fertel R, Cooke HJ (May 1992). "Loss of absorptive capacity for sodium and chloride in the colon causes diarrhoea in Potomac horse fever". Research in Veterinary Science. 52 (3): 353–362. doi:10.1016/0034-5288(92)90037-3. PMID   1352409.
  19. 1 2 3 Heller MC, McClure J, Pusterla N, Pusterla JB, Stahel S (May 2004). "Two cases of Neorickettsia (Ehrlichia) risticii infection in horses from Nova Scotia". The Canadian Veterinary Journal. 45 (5): 421–3. PMC   548627 . PMID   15206592.
  20. 1 2 3 4 Coffman EA, Abd-Eldaim M, Craig LE (November 2008). "Abortion in a horse following Neorickettsia risticii infection". Journal of Veterinary Diagnostic Investigation. 20 (6): 827–30. doi: 10.1177/104063870802000622 . PMID   18987240. S2CID   3208314.
  21. Gibson KE, Pastenkos G, Moesta S, Rikihisa Y (June 2011). "Neorickettsia risticii surface-exposed proteins: proteomics identification, recognition by naturally-infected horses, and strain variations". Veterinary Research. 42 (1): 71. doi: 10.1186/1297-9716-42-71 . PMC   3127766 . PMID   21635728.
  22. 1 2 3 Mott J, Rikihisa Y, Zhang Y, Reed SM, Yu CY (September 1997). "Comparison of PCR and culture to the indirect fluorescent-antibody test for diagnosis of Potomac horse fever". Journal of Clinical Microbiology. 35 (9): 2215–9. doi:10.1128/JCM.35.9.2215-2219.1997. PMC   229942 . PMID   9276390.
  23. Biswas B, Vemulapalli R, Dutta SK (August 1998). "Molecular basis for antigenic variation of a protective strain-specific antigen of Ehrlichia risticii". Infection and Immunity. 66 (8): 3682–8. doi:10.1128/IAI.66.8.3682-3688.1998. PMC   108402 . PMID   9673249.
  24. 1 2 Bertin FR, Reising A, Slovis NM, Constable PD, Taylor SD (2013). "Clinical and clinicopathological factors associated with survival in 44 horses with equine neorickettsiosis (Potomac horse Fever)". Journal of Veterinary Internal Medicine. 27 (6): 1528–34. doi: 10.1111/jvim.12209 . PMID   24118378.
  25. "Potomac Horse Fever - Digestive System". Merck Veterinary Manual. Retrieved 2020-11-10.
  26. Rikihisa Y, Jiang BM (March 1989). "Effect of antibiotics on clinical, pathologic and immunologic responses in murine Potomac horse fever: protective effects of doxycycline". Veterinary Microbiology. 19 (3): 253–62. doi:10.1016/0378-1135(89)90071-0. PMID   2497577.
  27. Durán MC, Marqués FJ (March 2016). "Detection of Neorickettsia risticii, the agent of Potomac horse fever, in a Gypsy Vanner stallion from Manitoba". The Canadian Veterinary Journal. 57 (3): 293–5. PMC   4751770 . PMID   26933267.
  28. McKenzie HC, Funk RA, Trager L, Werre SR, Crisman M (November 2019). "Immunogenicity of Potomac horse fever vaccine when simultaneously co-administered with rabies vaccine in a multivalent vaccine or as two monovalent vaccines at separate sites". Equine Veterinary Journal. 51 (6): 774–778. doi:10.1111/evj.13096. PMC   6850380 . PMID   30859618.
  29. Dutta SK, Vemulapalli R, Biswas B (February 1998). "Association of deficiency in antibody response to vaccine and heterogeneity of Ehrlichia risticii strains with Potomac horse fever vaccine failure in horses". Journal of Clinical Microbiology. 36 (2): 506–12. doi:10.1128/JCM.36.2.506-512.1998. PMC   104568 . PMID   9466767.
  30. "Potomac Horse Fever | AAEP". aaep.org. Retrieved 2020-11-10.