Sei sulla pagina 1di 13

Review

J Vet Intern Med 2013;27:1334–1346

Review of Equine Piroplasmosis


L.N. Wise, L.S. Kappmeyer, R.H. Mealey, and D.P. Knowles
Equine piroplasmosis is caused by one of 2 erythrocytic parasites Babesia caballi or Theileria equi. Although the genus
of the latter remains controversial, the most recent designation, Theileria, is utilized in this review. Shared pathogenesis
includes tick-borne transmission and erythrolysis leading to anemia as the primary clinical outcome. Although both para-
sites are able to persist indefinitely in their equid hosts, thus far, only B. caballi transmits across tick generations. Patho-
genesis further diverges after transmission to equids in that B. caballi immediately infects erythrocytes, whereas T. equi
infects peripheral blood mononuclear cells. The recent re-emergence of T. equi in the United States has increased
awareness of these tick-borne pathogens, especially in terms of diagnosis and control. This review focuses in part on fac-
tors leading to the re-emergence of infection and disease of these globally important pathogens.
Key words: Anemia; Babesia caballi; Erythrocytic parasite; Theileria equi; Tick-transmitted.

quine piroplasmosis is an infectious, tick-borne that 2 separate and distinct parasites could infect
E disease caused by the hemoprotozoan parasites
Theileria equi and Babesia caballi. Piroplasmosis,
equid erythrocytes, one significantly larger than the
other.13 The larger of the parasites was termed
which is also known in the literature as equine babesi- Piroplasma caballi, only to be later reclassified as
osis, theileriosis (concerning T. equi), and biliary fever, B. caballi.14 Whereas B. caballi is considered a classic
affects all equid species, including horses, donkeys, “babesia” species, the taxonomy of B. equi remains
mules, and zebras.1,2 Infection with either or both of controversial. Based in part on finding an extra-eryth-
these obligate, intraerythrocytic organisms can cause rocytic stage within equine PBMC, B. equi was reclas-
varying degrees of hemolytic anemia and associated sified as T. equi in 1998.11 Molecular phylogenetic
systemic illness. As T. equi infection “silently” re- investigations indicated that the organism possesses
emerged in the United States, questions are being characteristics of both babesia and theileria lineages,
raised concerning the tick-vector-parasite-host require- possibly placing it between the two.15–18 Recent geno-
ments necessary for the development of clinical dis- mic analysis supports the concept of a new genus for
ease. The parasites and their natural tick vectors are T. (B.) equi.10 Additional data are needed to deter-
endemic to most countries with tropical and subtropical mine the final placement of this parasite and there-
climates.3–5 The goals of control and disease eradication fore this review will use the most recent designation,
vary tremendously between endemic and non-endemic T. equi.
nations. Recent outbreaks of infection with limited dis-
ease expression in the United States and the Netherlands Epidemiology/Ecology
coupled with the identification of novel vectors within
the United States have prompted a renewed interest in Piroplasmosis occurs in most countries worldwide
this historically important disease.6–9 and infection is maintained within equine populations
Taxonomy of the causative agents of piroplasmosis as long as competent vectors are present.19 For
has been in question since their discovery and T. equi, the reservoir is the persistently infected equid;
remains controversial for T. equi.10,11 Currently, the however, for B. caballi, both infected horses and the
parasites are classified within the phylum Apicom- primary tick vector are reservoirs.20–24 Although the
plexa, which contains other hemoprotozoan such as precise tick-vector-parasite-host requirements for infec-
Plasmodium and Theileria. The parasite, termed Piro- tion or clinical disease are not known, the outcome of
plasma equi (reclassified later as B. equi), was first increasing densities of infected horses and ticks in an
recognized in South Africa as the causative agent of area is an increase in infection and potentially disease.25
disease in 1901.12 A few years later, it was discovered Clinically silent transmission appears common.7 The
risk of life-threatening clinical disease increases with
the presence of factors such as immunological naivety
From the Department of Veterinary Microbiology and Pathology, and increased density of infected ticks and horses.26
Washington State University (Wise, Mealey, Knowles); the Animal Although numerous studies have been published
Disease Research Unit, U.S. Department of Agriculture,
Agricultural Research Service (Wise, Kappmeyer, Knowles), and
regarding the epidemiology and distribution of infec-
the OIE Reference Laboratory for Equine Piroplasmosis (Knowles), tion within specific countries and regions, these
Pullman, WA. publications should be interpreted with caution given
Corresponding author: L.N. Wise, DVM, DACVIM, Depart- the profound variation in experimental design, sample
ment of Veterinary Microbiology and Pathology, Washington State population, and diagnostic testing. Particularly impor-
University, PO Box 646630, Pullman, WA 99164; e-mail: tant is the previous use of the complement fixation test
nwise@vetmed.wsu.edu.
(CFT) for detection of antibody against B. caballi
Submitted February 5, 2013; Revised April 15, 2013;
Accepted July 16, 2013.
or T. equi. Abundant data have shown that the CFT
Published 2013. This article is a U.S. Government work and is in lacks sensitivity in detecting persistent infections.27
the public domain in the U.S.A. Ixodid tick vectors occur in tropical, subtropical,
10.1111/jvim.12168 and some temperate climates.28 T. equi is more preva-
Equine Piroplasmosis 1335

lent than B. caballi, yet B. caballi has been identified had been at some time associated with the ranch. An
in more northern regions of the Northern Hemisphere investigation of this property allowed identification of
than T. equi.1 Given the currently available informa- 2 previously unrecognized competent vectors of T. equi
tion on geographic distribution of infected horses within the United States (Amblyomma cajennense
according to the Office International des Epizooties/ and Dermacentor variabilis). Ticks collected from the
The World Organisation for Animal Health (OIE), infected horses on the premises were capable of biolog-
Central and South America, Cuba, Africa, Asia, the ically transmitting disease to na€ıve horses.9
Middle East and Southern Europe are considered Competent ticks and iatrogenic blood transfers are
endemic regions.8 Within South America, the disease is efficient modes of transmission.19–21,25,40 With over 850
readily identified in all regions with the exception of tick species worldwide and approximately 85 within
the southernmost areas of Chile and Argentina. The the United States, the potential for transmission is
prevalence and distribution of the infection within the high.28 However, the presence of a competent tick vec-
Caribbean nations are questionable, but the disease tor and infected horses within the same area does not
has been reported on most islands, including Trinidad always lead to further infection or disease. Many fac-
and Cuba.8,29,30 Disease is widespread in Africa and tors must be considered including season, climate,
Asia with the highest prevalence reported in South host-specificity, and the particulars of a competent
Africa.31,32 Not all countries report identified cases to tick’s life cycle.25
the OIE, making an accurate understanding of the cur- The life cycle of a tick involves 4 life stages: egg,
rent parasite distribution difficult. Countries such as larva, nymph, and adult. After hatching from an egg,
Mexico and China are not considered endemic because the larva feeds on its host and molts into a nymph.
the OIE routinely receives no information regarding The nymph then feeds and molts into an adult.
distribution of piroplasmosis cases in those countries. Females and males proceed through these life stages,
Yet, articles from both Mexico and China have been but the female dies after laying her eggs. Adult male
published citing cases within those countries.33–35 ticks seeking females have the potential to feed on
Compiling a list of currently non-endemic regions is multiple hosts. Ticks are classified as hard ticks (Ixodi-
equally challenging given the difference in surveillance, dae) or soft ticks (Argasidae).28 Both are vectors for
import/export restrictions, and disease reporting that pathogen transmission, yet only hard ticks are natural
occurs. Current disease status for those countries that vectors for B. caballi and T. equi.
report to the OIE can be found on their website.36 Tick transmission can occur via 3 forms: intrasta-
Detection of infection within the United States in dial, transtadial, or transovarial. Intrastadial transmis-
recent years has placed the “piroplasmosis free” status sion occurs when acquisition and transmission of the
of this country under scrutiny.7,37 The 1st case of B. cab- parasite occurs within 1 life stage (no stage transition
alli was identified in a horse in 1961 in southern Florida before transmission). Transtadial describes acquisition
and while the source of infection was never definitively of infection in 1 stage and the ability for the same tick
determined, it was speculated that it was attributable to to transmit the infection during subsequent life stages.
importation of horses from Cuba.38,39 During the fol- The parasite is maintained within the tick as it devel-
lowing decade during extensive surveillance programs, ops. Transovarial transmission occurs when the female
several hundred total cases of both B. caballi and acquires parasites, which enter ovaries and are trans-
T. equi were diagnosed in 7 states. Dermacentor (Ano- mitted to offspring, allowing maintenance of the para-
center) nitens, a tick capable of transmitting B. caballi, sites across tick generations.20
was identified in affected Florida counties, yet this docu- Multiple ixodid tick species have been identified as
mented vector’s role in disease transmission was never either natural or experimental vectors of piroplasmosis.
confirmed. All infected horses were deported, quaran- B caballi is transmitted by 15 separate species (7 Der-
tined, or treated with various drugs until a negative macentor [Anocenter] sp., 6 Hyalomma sp., and 2
serologic result was obtained. Intense surveillance of Rhiphicephalus sp.) and T. equi by 14 species (4
horses and ticks continued in Florida until 1988 when Dermacentor sp., 4 Hyalomma sp., 5 Rhiphicephalus
the United States was deemed free of disease. To (Boophilus) sp., and A. cajennense). B. caballi is trans-
maintain this status, USDA APHIS improved restric- mitted transtadially and transovarially by its vectors.41
tions on importation of horses from endemic areas. T. equi is generally transmitted through transstadial
In 2008, 20 T. equi-infected horses were identified and intrastadial transmission.20 Transovarial transmis-
on 7 separate premises in Florida.37 All affected horses sion of T. equi occurs, but the precise role in epidemi-
were associated with horses that had been imported ology has not been detailed.42,43
from Mexico and all were engaged in illegal horse rac- Discovery of the vector D. reticulatus in the Nether-
ing. Given the history and distribution of infected lands in 2010 combined with recognition of a subclini-
horses, inappropriate management practices including cally B. caballi-infected horse led to a surveillance of
needle sharing and “blood doping” were assumed to that area that resulted in identification of several of
be the mode of transmission. No tick vectors were T. equi- and B. caballi-infected horses.6 Before 2009,
identified despite aggressive surveillance. More recently only 2 tick species known to transmit T. equi naturally
in 2009, an outbreak of T. equi was identified on a had been identified within the southernmost parts of
ranch in southern Texas involving approximately 400 the United States: D. nitens and R. microplus. Within
horses.7 All infected horses resided on the premises or the Texas outbreak, infected horses were found to be
1336 Wise et al

infested with 4 different tick species: A. cajennense, and invade other erythrocytes. T. equi’s initial invasion
A. maculatum, Dermacentor (Anocenter) nitens, and is different in that it first enters PBMCs.11 This life
D. variabilis. A. cajennense was the most abundant, cycle event is part of the justification for the most
being identified on approximately 79% of the infected recent taxonomic classification as Theileria.10 Inside
horses, followed by A. maculatum (19%), D. variabilis PBMCs, T. equi sporozoites develop into large schizo-
(16%), and D. nitens (3%). Before this outbreak, nts and after approximately 9 days, merozoites are
A. cajennense had not been identified as a vector for released and invade erythrocytes. For both parasites,
T. equi. The geographic distribution of these 3 host asexual replication results in an expanding population
ticks within the United States appears to be limited to of merozoites and parasitized erythrocytes. Some mer-
Texas and Florida.44 The role of A. maculatum in the ozoites develop into gametocyte forms within equine
outbreak currently remains unclear and although peripheral blood. Upon ingestion of merozoites (and/
D. nitens is a known vector of B. caballi, evidence or gametocytes) by a competent tick, the parasites
supporting transmission of T. equi is lacking. Adult undergo sexual reproduction, with gametocytes devel-
D. variabilis ticks collected from horses on the ranch oping into gametes, which combine to form zygotes
were able to experimentally transmit disease to na€ıve within the tick midgut. The zygotes develop differently
horses, but their role as a natural vector in this depending on the tick species and the parasite. After a
outbreak remains questionable.9 period of 6–24 days, continued development results in
the presence of sporozoites within the salivary gland of
Pathogenesis (Transmission/Life Cycle of the tick.43,47,48
Parasites) Transmission can occur iatrogenically through inap-
propriate mixing of the infected and uninfected
The life cycle of both B. caballi and T. equi involves blood.37 This occurs most frequently during the prac-
distinct stages that occur in the host and tick45,46 tice of needle sharing between positive and na€ıve
(Fig 1, 2). Both parasites progress through 3 life horses, but use of any blood-contaminated equipment
stages: the sporozoite (asexual transmission stage), the could result in transmission.49 Infection can also be
merozoite (asexual blood stage), and the gametocyte caused when chronically infected horses serve as blood
(sexual blood stage). The development of these para- donors to na€ıve horses. The illegal practice of “blood
sites within the tick can vary depending on species of doping” (prerace blood transfusions) was implicated in
tick involved. Regardless of species variation, for both the 2008 Florida outbreak.37 Experimental infection
B. caballi and T. equi, infectious sporozoites are trans- can be achieved through intravenous and subcutaneous
mitted through the tick-saliva to the equid host. The routes as well as tick transmission.20,50–52
duration of the extrinsic incubation period (acquisition Although some details of pathogenesis remain
feed; parasite replication time in the tick vector and unknown, infection with either T. equi or B. caballi
transmission feed) is not defined for B. caballi or causes erythrocyte lysis resulting in varying degrees of
T. equi. Once within the host, B. caballi sporozoites hemolytic anemia. Physical rupture of erythrocytes
directly invade erythrocytes where they multiply and during release of merozoite stages causes intravascular
develop first into trophozoites and then into merozo- hemolytic anemia. Infected red blood cells are removed
ites. After erythrocyte rupture, merozoites are released from the circulation by splenic macrophages further

Fig 1. Life cycle of Babesia caballi. Illustration by Massaro Ueti.


Equine Piroplasmosis 1337

Fig 2. Life cycle of Theileria equi. Illustration by Massaro Ueti.

contributing to hemolytic anemia. Nonparasitized likely that individual horse genetics or geographic iso-
erythrocytes are also removed from circulation, but late/strain differences could influence the prevalence of
the reason for this phenomenon is unknown. Data placental transmission. Exposure to semen from an
from experimentally infected splenectomized donkeys infected stallion is not considered to be a means of
indicate that the biochemical structure of erythrocyte transmission, yet blood contamination during breeding
membranes changes dramatically during infection with practices could present a transmission risk.61
T. equi.53 It was suggested that this conformational In most cases, the horses become persistently
change causes decreased deformability of the red cells, infected and become inapparent carriers. The inappar-
which could lead to reduced microvascular blood flow. ent carrier state is life-long with T. equi and possibly
Plasma levels of malondialdehyde (marker of lipid per- for B. caballi. A number of accounts indicate that
oxidation) are significantly increased, suggesting that horses infected with B. caballi can undergo self-clear-
an accumulation of oxidative ions also contributes to ance of the parasite without treatment.1,3,5 It is
erythrocyte lysis.53 These parasites also alter coagula- assumed that persistent subclinical infection is due in
tion in infected horses through unknown mechanisms. part to sequestration of the organism and immune
B. caballi-infected erythrocytes cause microthrombi by evasion strategies. Various theories for location of par-
clumping within small vessels, leading to venous stasis asite sequestration in asymptomatic horses have been
and vasculitis.52,54 Varying degrees of thrombocytope- reported, including capillaries, central nervous system
nia and prolonged clotting times have been reported vasculature, and bone marrow.4,62,63 The mechanism(s)
during infection with T. equi and B. caballi.55 Hypoth- of persistent infection remain unknown.
eses regarding the pathogenesis of decreased platelet After transmission, depending on factors including
counts include immune-mediated destruction, splenic parasite dose and immunity, clinical signs develop
sequestration and/or excess consumption as is within 10–30 days for B. caballi and 12–19 days for
observed in disseminated intravascular coagulation. T. equi.3 The fatality rate of na€ıve horses in endemic
Severe piroplasmosis can result in hypercoagulability, nations has been estimated at 5–10% dependent on
systemic inflammatory response syndrome, and subse- parasite, transmission dose, overall health of the horse,
quent multiorgan system dysfunction.56 and administration of treatment.4 Uniformly, infection
Placental transmission from infected carrier mares to with T. equi typically results in more severe clinical
their fetuses has been documented.15,57–59 This trans- disease than B. caballi.4 Disease signs and severity can
mission can result in abortion (most commonly in late vary significantly from 1 region to another.7,31
gestation), stillbirth, or neonatal infection and can
occur across placentas that are histologically normal. Clinical Disease
The prevalence of this type of transmission is
unknown. The natural outbreak documented in Texas Clinical disease can manifest in different forms. With
of 2009 resulted in infection of pregnant mares, none acute T. equi infection, clinical signs are usually related
of which transmitted infection to their foals based on to marked hemolysis and resulting anemia. Although
serial negative polymerase chain reaction (PCR) B. caballi-infected horses do become anemic as well,
results. Conversely, T. equi has been reported to be the rare cases of acute death from B. caballi have
responsible for 11% of all abortions in South Africa.60 occurred reportedly as a result of multiple organ
Based on the variation in reported occurrence, it is dysfunction related to systemic formation of micro-
1338 Wise et al

thrombi and development of disseminated intravascu- mission via ticks, placentally or iatrogenically, these
lar coagulation.56 In these cases, the exact clinical signs horses represent the largest challenge to non-endemic
vary depending on the organ system affected. nations attempting to prevent apparently healthy carri-
Horses with acute infection initially develop nonspe- ers from crossing their borders.20,73
cific signs such as high fevers, sometimes in excess of An appropriate list of differential diagnoses should
104°F, lethargy, anorexia, weight loss, and peripheral be determined based on whether the horse resides in or
edema.4 Petechiations caused by thrombocytopenia are has visited an endemic region. In general, acute onset
often observed on mucous membranes, including the of the aforementioned clinical signs could also be
nictitating membrane.3 Signs of hemolytic anemia fol- caused by equine infectious anemia virus, African
low and include icteric or pale mucous membranes, horse sickness virus, equine viral arteritis virus, equine
tachycardia, tachypnea, weakness, and pigmenturia ehrlichiosis, purpura hemorrhagica, immune-mediated
(because of either hemoglobinuria or bilirubinuria).53,64 hemolytic anemia, and red maple leaf toxicity.8,74
Some horses show signs of gastrointestinal complica- Results of laboratory analyses may aid in diagno-
tions including colic or impactions followed by diar- sis. Most horses regardless of clinical syndrome exhi-
rhea. Other less common clinical presentations include bit some degree of anemia characterized by decreased
secondary development of pneumonia, pulmonary packed cell volume, hemoglobin, and erythrocyte
edema, cardiac arrhythmias, catarrhal enteritis, lamini- count. Although acutely infected horses can have
tis, and central nervous system disease characterized profound anemia with packed cell volumes (PCV) as
by ataxia, myalgia, and seizures.32,39,64,65 Temporary low as 10%, the PCV rarely falls below 20%.52,54,64
or permanent infertility has been reported in stal- Red blood cell indices, MCV, MCH, and MCHC are
lions.56 Acute renal failure occurs as a result of hemo- variable.53 Thrombocytopenia is commonly identi-
globin-induced pigment nephropathy and systemic fied.3,52,54,55,64 One report noted a decrease in platelet
responses to severe inflammation (hypotension) can count in 39% of T. equi infections, 80% of B. caballi
worsen the kidney disease.3 Severe infections can also infections, and 100% of dual infection.54,64,75 Clotting
culminate in liver failure or disseminated intravascular times can be prolonged or normal. The leukogram
coagulation.56 can vary depending on infection stage and severity.76
Fulminant, abrupt onset of signs of disease, termed Fibrinogen concentration can be elevated and albu-
peracute disease, has been documented. Collapse and min concentration can vary depending on hydration
sudden death from overwhelming T. equi can occur and status, chronicity of the disease, and associated con-
introduction of na€ıve horses into an endemic region can ditions that result in protein loss.55 Hyperbilirubin-
lead to rapid onset of severe disease. In the 1930s, relo- emia is often observed and the liver enzyme
cation of a group of na€ıve horses into an endemic area activities, ALP, AST, and GGT can be elevated.64
of southern France resulted in a 69% fatality rate.26 A These elevations are attributed to reduced blood flow
report from Jordan documented 5 presumed inapparent to the liver, which can in severe cases result in centri-
carriers undergoing strenuous exercise that immediately lobular necrosis. Hypophosphatemia and hypoferremia
after completion of the exercise, developed profound are common, attributed to altered erythrocytic metabo-
weakness with two dying suddenly.66 Recrudescence of lism.77 Infected erythrocytes can be identified in sternal
marked T. equi parasitemia was assumed. Neonatal bone marrow aspirates of asymptomatic horses, but util-
foals infected in utero with T. equi can present with ity of this test as a diagnostic tool is limited.62
acute, severe signs.57,58,67–71 These foals can exhibit clin- Gross and histopathologic findings at necropsy vary
ical signs at birth or can become ill at 2–3 days of age. depending on the severity of disease and associated com-
Clinical signs are often nonspecific, such as weakness plications. Gross examination might demonstrate evi-
and decreased suckling, but progress to resemble those dence of anemia as well as varying degrees of icterus,
of an infected adult, including icterus, fever, and anemia edema, and splenomegaly. Other findings can include
(with or without petechiations and hemoglobuinuria). pulmonary edema and congestion, cardiac hemor-
Cases of B. caballi fetal and neonatal infection have rhages, hydropericardium, hydrothorax, hepatomegaly,
been reported but are rare.70,72 ascites, enlarged discolored kidneys, and lymphadenop-
Chronic T. equi or B. caballi infection can result athy.3 Histopathologic findings can include centrilobu-
only in nonspecific signs, including lethargy, partial lar necrosis of the liver, renal tubular necrosis with
anorexia, weight loss, and poor performance. Mild hemoglobin casts, and microthrombi within the liver
anemia might be present and the spleen might be and lungs.32 Pulmonary tissue examination can demon-
enlarged upon rectal palpation. It has been suggested strate congestion, edema, and hemosiderin-laden mac-
that splenic enlargement is caused by the increased rate rophages within the pulmonary alveolar walls.
of extravascular hemolysis that occurs within the Parasites can be observed within red blood cells within
spleen in less severely affected horses.3,5,52 blood vessels and within macrophages in the lymph
Importantly, horses infected with either T. equi or nodes.3,39,78
B. caballi in both endemic and non-endemic regions The apparent global variation in clinical disease
are most commonly inapparent carriers with no appre- might be caused by a variety of factors contributing to
ciable signs of disease. Pregnancy in carrier mares can emergence of infection and disease. Infection with
result in abortion or neonatal infection.15,32 Because T. equi in South Africa often results in severe disease,
inapparent carriers can serve as reservoirs for trans- requiring treatment.31,32 In contrast, in the outbreak
Equine Piroplasmosis 1339

identified in the United States, in which 475 horses


were affected, only 1 horse was reported to exhibit
mild clinical signs.7 It is difficult to compare these
instances because the differences between these T. equi
strains are unknown, but it at minimum provides a
single comparison between non-endemic and endemic
countries.

Immunity
The response of the equine immune system to infec-
tion with T. equi or B. caballi is not completely
defined, but is undoubtedly complex and multifaceted.
It is well accepted that infection with either parasite
results in carrier status, which confers protection
against disease. There is no documented cross-protec-
tion between T. equi and B. caballi, as horses can be
infected with both parasites simultaneously.4
The spleen plays a necessary role in control of most Fig 3. Electron micrograph of a splenic macrophage from a
hemoprotozoan parasites. A horse with a spleen is typ- severe combined immunodeficiency foal showing phagocytized
ically able to overcome acute T. equi-induced disease, Theileria equi-infected erythrocytes. Arrow denotes the organism
inside an erythrocyte and the asterisk illustrates the nucleus of
whereas splenectomized horses invariably succumb to
the splenic macrophage. Image courtesy of Lowell Kappmeyer.
disease with parasitemias that can reach 80%.51,53,79
Inapparent carriers of T. equi will also develop termi-
nal disease upon splenectomy.80 Splenectomized horses
inoculated experimentally with B. caballi may or may genome.93 In the acute stages of T. equi infection, high
not develop obvious clinical disease, but death caused levels of IgGa (now IgG1) and IgGb (now IgG4&7)
by infection has been documented.81 This discrepancy correlate with control, whereas IgG(T) levels (now pre-
may be attributable to differences in parasite strain, dominately IgG5 and to a lesser extent IgG3) increase
infective dose, the overall health of the horse, or a after resolution of parasitemia during the chronic
combination of factors. phase of infection.92 Antibodies are first detected
Although essential in other hemoprotozoan infec- within 7–11 days after natural infection and peak at
tions like Babesia bovis, the function of cell-mediated 30–45 days. All examined subclasses remain detectable
immunity in piroplasmosis has yet to be fully deter- into the chronic/inapparent stage of disease.
mined.82,83 Production of nitric oxide by macrophages The correlates of adaptive immune responses to
might be an essential effector mechanism of immune T. equi infection are currently unknown. Donkeys vac-
control against experimental B. caballi infection.84 cinated with T. equi immunogen were able to mount a
Importantly, innate immune responses and the pres- protective response characterized by high antimerozo-
ence of a spleen are not sufficient to control T. equi ite antibody titers and merozoite-specific lymphocyte
infection, because spleen-intact foals with severe com- proliferation.83 Recently, it was documented that
bined immunodeficiency (SCID) are unable to control SCID foals that received repeated infusions of T. equi
T. equi parasitemia85 (Fig 3). Although innate immu- hyperimmune plasma before inoculation with the para-
nity is unaffected, SCID foals lack functional T and B site were able to delay the time to peak parasitemia.94
lymphocytes and are incapable of mounting antigen- All foals developed disease, but partial protection was
specific antibody and cellular immune responses.85–91 associated predominantly with transfused IgG3-specific
Inoculation of these foals with T. equi resulted in ful- antimerozoite antibodies. Overall, additional research
minant, severe infection within 7 days, and subsequent is needed to define the protective roles of antibody and
death. Terminal parasitemias ranged between 29 and cellular adaptive immune responses against T. equi.
41% and PCV decreased by 50% of the normal value. Even less is known about protective immune
Thus, adaptive immune responses are required to responses against B. caballi infection. Infected horses
control T. equi parasitemia, but are not required for produce antibodies to rhoptry associated protein-1
lysis of erythrocytes. (RAP-1), which is utilized on serologic detection of
Antibody responses correlate with control of parasit- infection. This conserved apical merozoite protein
emia.92 T. equi-infected horses produce antibodies remains partially uncharacterized in B. caballi, but in
against immunodominant merozoite proteins termed B. bovis plays a pivotal role in induction of humoral
equi merozoite antigens (EMAs), which are surface immunity.95
expressed on merozoites.85 The exact role of this anti- In most endemic areas, foals that ingest colostrum
body response in immunity and persistence remains from a carrier mare are protected from infection and
unclear. The nomenclature of equine immunoglobulin clinical disease for the first 1–5 months and can be
G has recently been adjusted to accommodate the 7 protected up to 9 months of age.3,56 As maternal anti-
unique IgG heavy chain genes discovered in the equine bodies decline, the foal becomes susceptible and most
1340 Wise et al

young horses in endemic nations are infected by age 2. ites are smaller and typically measure 2–3 lm in
It has also been suggested that foals can be born as length.3 The percent of infected erythrocytes during
healthy inapparent carriers of T. equi, which would clinical disease caused by T. equi is usually between 1
also infer some level of protection.56 and 5%, but in severe cases can exceed 20%.56 In
Rarely, inapparent T. equi carriers can exhibit cases of chronic or inapparent infection, parasite num-
relapses of clinical disease associated with stress, stren- bers remain too low for reliable detection on blood
uous exercise, immunosuppression, and steroid smear.99
administration.66,96,97 Experimental treatment with Several serologic tests were developed to increase
beclamethasone at a dose of 0.1 mg/kg once daily for diagnostic sensitivity, especially in those carrier horses
5 days before and 5 days after inoculation with T. equi exhibiting no clinical signs. These tests include the
resulted in a 50% increase in parasitemia as compared CFT, indirect immunofluorescence assay, western blot,
with controls.96 These relapses have not been reported and competitive enzyme-linked immunosorbent assay.
for B. caballi. The CFT relies on activation of complement upon
specific interaction of antibody and antigen.99 A posi-
Diagnosis tive result is defined as a positive reaction at a dilution
of 1 : 5. Infected horses seroconvert on CFT approxi-
Various diagnostic modalities can be used alone or mately 8–11 days after infection with titers beginning
in combination to diagnose infection. During manage- to decline at 2–3 months.99 CFT is a very specific test,
ment of an outbreak within a non-endemic nation, yet lacks sensitivity, especially in chronic infection or
involvement of the state and national regulatory agen- after treatment.27 Horses can transiently become nega-
cies is required and often, multiple diagnostic methods tive 3–15 months after treatment for B. caballi and
will be utilized in an effort to obtain the most accurate 24 months for T. equi.99,100 IgG(T), now classified as
information. Only a few laboratories in the world are IgG5 and to a lesser extent IgG3, remains elevated in
authorized to perform certain tests, so proper handling chronic T. equi infections. IgG(T) does not fix comple-
of samples is crucial. ment and recently, it was demonstrated that IgG3 fixes
Light microscopy can be used to identify the organ- complement, whereas IgG5 does not.93,101 Thus, it is
isms within the erythrocytes. A thin blood smear, not surprising that the CFT lacks sensitivity for diag-
stained with Giemsa, Wright’s, or Diff-Quik®, may nosis of chronic or inapparent T. equi infection. Cross-
reveal organisms during the acute stage of infection. reactivity between antibodies against T. equi and
The smears must be thoroughly examined since even B. caballi when using the CFT has been reported.99,102
during severe infection, the percent parasitemia Regardless of the fact that the CFT was previously the
remains so low that false-negative results are not official regulatory test for establishing piroplasmosis
uncommon.5,98 The piroplasms of T. equi and B. caballi status before travel to a non-endemic country, the
can be easily distinguished from one another. Within the CFT is not considered the diagnostic test of choice for
erythrocyte, B. caballi typically appears as 2 large pyri- chronic infection.98
form (pear-shaped) merozoites that measure approxi- Indirect immunofluorescent antibody tests (IFAT)
mately 2–5 lm in length (Fig 4). During clinical infection demonstrates high specificity, but lacks sensitivity. It
with B. caballi, the percentage of erythrocytes parasitized is, however, considered more sensitive than the CFT.99
is typically less than 1% and may be less than 0.1%. In this test, fluorescently labeled antibodies react with
T. equi merozoites occur within erythrocytes as poly- antigen bound to a glass slide. A sample is considered
morphic, small piroplasms occasionally in a distinct positive if strong fluorescence is noted at a dilution of
Maltese cross-formation (Fig 5). The T. equi merozo- 1 : 80 or higher. Experimental intravenous infection

Fig 4. Equine erythrocyte containing Babesia caballi merozoites. Fig 5. Equine erythrocyte containing Theileria equi merozoites
Diff-Quik®, 9100 oil magnification. Image courtesy of Peter in characteristic Maltese cross-formation. Diff-Quik®, 9100 oil
Awinda. magnification.
Equine Piroplasmosis 1341

with T. equi or B. caballi resulted in positive IFAT Polymerase chain reaction tests for the presence of
results at days 3–20 post infection.100 Titers were more the organism of interest by amplifying and detecting
consistently detected and remained elevated longer specific fractions of the DNA. This test is exquisitely
with the IFAT versus the CFT.103 Typically, the IFAT sensitive and thus far has only been utilized in research
is used as an adjunct test to aid in analysis of CFT settings for the detection of T. equi. Three variations
results, but it remains one of the prescribed tests for of PCR include real-time PCR, nested PCR, and
equine piroplasmosis recommended by the OIE.98 nested PCR with hybridization.113–117 Comparison of
Until recently, Western blot (or immunoblot) has these tests and the results from these tests is difficult,
been utilized primarily in a research setting for diagno- given that no standardization exists between laborato-
sis of T. equi and B. caballi. The National Veterinary ries. Nested PCR for T. equi using the EMA-1 gene
Services Laboratory in Ames, Iowa, is now offering an sequence has been shown to detect a positive result
immunoblot as an adjunct diagnostic tool for detection equivalent to a percent parasitemia of 0.000006%.115
of B. caballi infection.104 Research is under way to val- One report also indicated that nested PCR detects
idate these tests for use in routine diagnosis. 3.69 more infections that microscopy and 2.29 more
Since 2004, the competitive inhibition enzyme-linked than traditional PCR.30 The validity of nested PCR
immunosorbent assay (cELISA) has been one of the routinely in the United States has been questioned as a
regulatory tests prescribed by the OIE for international diagnostic tool for horses in South Africa.110,118 Upon
horse transport.98 The test is considered to be the most examination of the genetic composition of EMA-1, it
sensitive means of detection of chronic T. equi infec- was recognized that the strains from around the world
tion.27 The cELISA for T. equi utilizes recombinant were not 100% homologous. These discoveries further
EMA-1 and specific monoclonal antibodies.105 EMA-1 emphasize the issues involved in standardization of
is an immunodominant, highly conserved surface anti- PCR as a diagnostic test. Thus far, the use of nested
gen specific to T. equi. Horses infected with T. equi are PCR for detection of B. caballi DNA in chronically
detectable with the cELISA as early as 21 days after infected horses has proven unreliable. As it is currently
experimental infection and approximately 5 weeks performed, it is unlikely that nested PCR will ever be
after tick transmission.105 The EMA-1 cELISA is vali- standardized and commercially marketed for detection
dated for use against multiple different strains of of T. equi or B. caballi infection.119
T. equi found around the world.27 Generation of a With increasing use of PCR in laboratory settings,
recombinant form of this epitope and associated the validity of a positive cELISA result has come into
monoclonal antibodies allowed standardization of this question. Despite treatment and apparent clearance of
test and markedly increased sensitivity as compared T. equi, as demonstrated by negative PCR and trans-
with the other serologic tests.27,106,107 mission studies, cELISA results remain positive, some-
A recombinant form of RAP-1 was also developed times for up to 24 months after clearance.7,120 The
for the B. caballi cELISA.108 This test, upon compari- transmission risk that these PCR-negative, seropositive
son with CFT using 300 equine serum samples from horses pose must be determined.
around the world, was able to diagnose infection in
25% more cases than with the CFT. However, the Treatment
currently available RAP-1 cELISA relies on the recog-
nition of epitopes that are not conserved across all In endemic regions, treatment of piroplasmosis is
B. caballi strains. Because of sequence heterogeneity used only as a means of decreasing clinical signs and
between the recombinant RAP-1 used in the test and reducing fatalities. Clearance of the organism serves
South African isolates of B. caballi, the test is unable no purpose in these countries as life-long immunity
to detect infected horses in South Africa.109 Although (premunity) is assumed to be conferred with chronic,
similar differences exist between the recombinant inapparent infection. In non-endemic regions attempt-
EMA-1 used in the T. equi cELISA and South African ing to remain free of piroplasmosis, treatment of
T. equi isolates, the cELISA continues to detect infected horses with the intent of clearance (chemoster-
infected horses in South Africa.110 Both cELISAs are ilization) is desired. T. equi infections are more typi-
marketed by VMRD (Pullman, WA) and are not cally difficult to treat than B. caballi infections.
available to general practitioners. Lastly, a field vali- Numerous drugs have been reported to have variable
dated ELISA using whole T. equi merozoite antigen efficacy in inhibiting T. equi and B. caballi both in cell
has been described as an easy, economical, and reliable culture and in vivo, making the literature difficult to
test.111 interpret.117,121–128 Historically, it was reported that
Recently, the previously reported data on the speci- B. caballi infection was self-limiting with clearance
ficity and sensitivity of the CFT and cELISA were noted after several years, yet this is not always the
statistically analyzed.25,106,108,112 Overall, the sensitivity case.63 Chemotherapeutic clearance of T. equi in the
of the CFT to detect T. equi is 47% and the cELISA horse has been previously reported, yet the research
is 96%. The specificities of the 2 tests are 94% and was conducted before the development of tests with
95%, respectively. For testing of B. caballi, the sensi- increased sensitivity for parasite persistence.122,123
tivity of the CFT is 88% and the cELISA is 91%. The Until recently, it was widely accepted that chemosteril-
specificities of the 2 tests for B. caballi are 98% and ization of a T. equi-infected horse was unachievable.
70%, respectively. Data collected during the outbreak in Texas indicate
1342 Wise et al

that T. equi can be eliminated from an infected horse recommended.122 Information regarding treatment of
with appropriate dosing of imidocarb diproprionate.7 T. equi-infected neonatal foals with ID is limited with
For alleviation of clinical signs, several drugs have only one case report.71 When a nursing mare is given a
been used with success, yet imidocarb, in its diproprio- single dose of at 2.4 mg/kg IM, ID is detectable in the
nate salt form (ID), is considered to be the most effective. milk 2 hours after administration.132 It remains
The alternate form of this drug, composed of dihydro- unclear if this could lead to toxicity in the suckling
chloride salt, will cause more severe muscle damage at foal. One report of administration of ID to pregnant
the site of injection.122,129 ID, a carbanilide derivative, is mares followed by induced abortion resulted in circu-
typically administered to horses intramuscularly. lating levels of ID in each fetus comparable to the
Although its mechanism of action remains unclear, pro- dam’s serum concentrations.60
posed mechanisms include inhibition of inositol entry Diminazene aceturate and diaminazene diaceturate
into infected erythrocytes or alteration in metabolism of have been used with success against T. equi and
polyamines.130,131 After intramuscular (IM) injection, B. caballi at a dose of 3.5 mg/kg IM every 48 hours
ID is rapidly eliminated from the plasma, yet remains for 2 treatments.134 Diminazene aceturate is more
sequestered in certain tissues of the body.132 Reported effective than diaminazene diaceturate, but both drugs
dosages for alleviation of clinical signs vary, yet most have been reported to cause significant injection site
sources indicate that 2.2–4.4 mg/kg given IM once is muscle damage. Efficacy of both drugs increases with
effective. If necessary, lower dosages can be repeated the 2nd dosage, yet no chemosterilization has been
at 24–72 hour intervals for 2–3 treatments. In non- reported. Signs of toxicity include respiratory distress
endemic nations where chemotherapeutic clearance of and lethargy.
the organism is desired, animals infected with B. cabal- The antibiotic oxytetracycline when administered IV
li can be cleared with a dose of 4.4 mg/kg IM every at a dose of 5–6 mg/kg once daily for 7 days is effective
72 hours for 4 treatments.117 For clearance of T. equi, against T. equi, but not against B. caballi.64 Other drugs
data from both naturally and experimentally infected reported to have efficacy in treatment of babesiosis
horses indicate that the same dose is effective.7,120 Of include amicarbilade isothionate, euflavine, artesunate
the 25 naturally infected treated horses, one remained and arteether (arteminsin derivatives), buparvaquone,
positive for T. equi after the initial treatment and had and atovaquone, yet these drugs are no longer com-
to undergo a 2nd treatment regimen to obtain chemo- monly utilized in practice.56,124,125 Ponazuril inhibits
sterilization.7 In the study using ID to attempt clear- T. equi organisms in vitro.135 No in vivo research has
ance in experimentally T. equi-infected horses, 1 horse been conducted to date, yet it is possible that this class
remained resistant to parasite elimination.120 Cur- of drugs could offer additional treatment options in
rently, clearance is determined by negative PCR results the horse.
and inability to transmit the parasite to a na€ıve sple- Aside from antiprotozoal drugs, acutely infected
nectomized horse via blood transfusion, but studies horses often require supportive care including but not
are under way to more clearly define parasite elimina- limited to intravenous fluids, nonsteroidal anti-inflam-
tion.7 In the United States, if a horse is diagnosed with matory drugs, pain management, and blood transfu-
T. equi, the owner and veterinarian can enroll the sions. Adequate hydration is essential upon initiation
horse in the USDA controlled treatment program as of and during treatment with imidocarb.
to ensure appropriate quarantine, treatment, and sub-
sequent release of cleared horses.120 Prevention/Vaccination
ID has anticholinesterase activity, so reactions to the
drug often present as sweating, signs of agitation, colic, Prevention of infection in endemic nations is virtu-
and diarrhea.129,133 Typically, these signs are transient ally impossible, and it is assumed that the premunity
and rarely life-threatening. Effects can be prevented with conferred with initial infection acts to protect the horse
an intravenous dose of glycopyrolate at 0.0025 mg/kg from recurrent disease upon subsequent exposures. In
once or reversed with a single intravenous dose of atro- non-endemic nations, the cornerstone of protection is
pine at 0.2 mg/kg. Both of these anticholinergic drugs regulation of equine movement between endemic
cause adverse effects as well. Administration of the nations. Depending on the non-endemic country in
anticholinergic n-butylscopolaminea can lessen clinical question, horses must test negative for T. equi and
signs without addition of adverse effects.7 As ID B. caballi on the serologic test designated specifically
undergoes hepatic and renal clearance, periportal hepa- by the country of import, typically the cELISA or the
tic necrosis and renal tubular necrosis can occur with IFAT. If positive, horses are generally denied entrance
toxicity.129,133 Horses undergoing treatment with this unless for tightly regulated athletic events. All
drug should be monitored carefully for development of imported horses from endemic nations undergo strict
complications. Transient azotemia, elevations in uri- quarantine and are examined thoroughly for ticks.
nary GGT/creatinine ratios, or both as well as tran- Application of acaricides before removal from the
sient elevations in liver enzyme activities (AST, ALT, endemic nation is used to ensure that ticks are not
ALP, and SDH) can be observed during treatment, introduced with the horses. The regulatory system put
but typically resolve with discontinuation of the in place by the OIE has been successful, yet isolated
drug. Donkeys and mules are exquisitely sensitive cases continue to occur in non-endemic nations. These
to ID, therefore its use in these species is not isolated cases are rarely caused by tick transmission
Equine Piroplasmosis 1343

and are most often linked to the use of blood contami- treatment of these parasites is paramount to control
nated equipment and practices involving needle sharing these insidious diseases, while promoting growth in the
or blood transfusions from untested donors.37,39,78 In movement of horses internationally.
the United States, a horse that is identified as positive
on cELISA must be immediately quarantined and the
state and federal authorities must be notified. Until a
reliable means of sterilization is identified, these horses Footnote
must remain quarantined, be exported, donated to a
research facilities, or euthanized. The most appropriate Buscopan; Boehringer Ingelheim, Rheinland-Pfalz, Germany
action is determined by the state and federal regula-
tory officials on a case-by-case basis.25,120
Non-endemic nations that border endemic nations
cannot completely prevent introduction of ticks, so dil- Acknowledgments
igent measures must be taken to reduce horses’ contact Conflict of Interest Declaration: The authors disclose
with ticks. This includes routine application of acari- no conflict of interest.
cides, surveillance for the presence of ticks, and
reduction in vegetation.73 A variety of chemical are
available to reduce tick exposure, but not all effective References
acaricides conform to EPA regulations, so careful 1. Friedhoff KT, Tenter AM, Muller I. Haemoparasites of
selection and use of these products is necessary.25 equines: Impact on international trade of horses. Rev Sci Tech
Although no direct scientific evidence exists in horses, 1990;9:1187–1194.
systemic administration of the dewormer ivermectin 2. Schein E. Equine babesiosis. In: Ristic M, ed. Babesiosis
likely aids in control of blood-feeding ticks. Being of Domestic Animals and Man. Boca Raton, FL: CRC;
aware of the habitat and the seasonality of ticks in 1988:197–208.
3. de Waal DT. Equine piroplasmosis: A review. Br Vet J
your area is important. Guidelines for examining
1992;148:6–14.
horses for ticks and appropriate removal of ticks have
4. Maurer FD. Equine piroplasmosis—Another emerging dis-
been published by the USDA.73 ease. J Am Vet Med Assoc 1962;141:699–702.
Several studies have assessed the potential use of 5. Friedhoff KT, Soule C. An account on equine babesioses.
vaccination to induce immunity to B. caballi and Rev Sci Tech 1996;15:1191–1201.
T. equi infection, yet no vaccine is commercially 6. Butler CM, Sloet van Oldruitenborgh-Oosterbaan MM,
available. Immunization with whole merozoite induced Stout TAE, et al. Prevalence of the causative agents of equine
protection in 4 donkeys.83 The correlates of protective piroplasmosis in the South West of The Netherlands and the
immunity in horses are unknown, and until these are identification of two autochthonous clinical Theileria equi infec-
elucidated, developing an effective vaccine will be tions. Vet J 2012;193:381–385.
7. Ueti MW, Mealey RH, Kappmeyer LS, et al. Re-emergence
difficult.
of the apicomplexan Theileria equi in the United States: Elimina-
Vaccination and treatment strategies are dependent
tion of persistent infection and transmission risk. PLoS One
on the infection status (endemic versus non-endemic) of 2012;7:e44713.
the region. A transmission blocking vaccine in the 8. World Animal Health Information Database (WAHID).
absence of full compliance in at-risk horses would not 2012. Available at: http://www.oie.int/fileadmin/Home/eng/Ani-
be appropriate in an endemic region. A vaccine prevent- mal_Health_in_the_World/docs/pdf/EQUINE_PRIOPLASMO-
ing transmission or clinical disease and death is needed SIS_FINAL.pdf. Accessed April 1, 2013.
for na€ıve horses moved into endemic regions. Chemo- 9. Scoles GA, Hutcheson HJ, Schlater JL, et al. Equine piro-
therapeutics, which aid in control of acute parasitemia plasmosis associated with Amblyomma cajennense ticks, Texas,
and associated clinical signs, but do not eliminate USA. Emerg Infect Dis 2011;17:1903–1905.
10. Kappmeyer LS, Thiagarajan M, Herndon DR, et al. Com-
infection, are important in endemic regions. In non-
parative genomic analysis and phylogenetic position of Theileria
endemic regions, the goal is remaining infection free;
equi. BMC Genomics 2012;13:603.
therefore, when infected horses are detected, a safe 11. Mehlhorn H, Schein E. Redescription of Babesia equi
chemotherapeutic with efficacy for eliminating persis- Laveran, 1901 as Theileria equi Mehlhorn, Schein 1998. Parasitol
tent infection is needed. To remain infection free, nec- Res 1998;84:467–475.
essary tools include accurate screening diagnostic tests 12. Laveran A. Contribution a l’etude du Piroplasma equi. CR
and thorough knowledge of tick populations and their Seances Soc Biol Paris 1901;53:2.
ability to transmit B. caballi, T. equi, both. 13. Koch R. BeitraE  ge zur entwicklungsgeschichte der
Increasing globalization of the equine industry cou- piroplasmen. Z Hyg Infektionskrankh 1906;54:9.
pled with constantly changing climates provides major 14. Nuttal GH, Strickland C. Die parasiten der pferdepiro-
plasmose response der biliary fever. Zentralbl Bakteriol 1910;1:2.
challenges for controlling persistent infections as those
15. Allsopp MT, Lewis BD, Penzhorn BL. Molecular evidence
caused by T. equi and B. caballi. As with all vector-
for transplacental transmission of Theileria equi from carrier
borne infections, measures must be taken to enhance mares to their apparently healthy foals. Vet Parasitol
control through surveillance of equine populations and 2007;148:130–136.
detailed knowledge of vector competence and habitat. 16. Allsopp MT, Allsopp BA. Molecular sequence evidence
Detailed information regarding life cycles, transmis- for the reclassification of some Babesia species. Ann N Y Acad
sion, immune responses, accurate diagnostics, and Sci 2006;1081:509–517.
1344 Wise et al

17. Allsopp MT, Cavalier-Smith T, De Waal DT, et al. Phy- 37. Short MA, Clark CK, Harvey JW, et al. Outbreak of
logeny and evolution of the piroplasms. Parasitology 1994;108(Pt equine piroplasmosis in Florida. J Am Vet Med Assoc
2):147–152. 2012;240:588–595.
18. Uilenberg G. Babesia—A historical overview. Vet Parasi- 38. Bryant JE, Anderson JB, Willers KH. Control of equine
tol 2006;138:3–10. piroplasmosis in Florida. J Am Vet Med Assoc 1969;154:1034–
19. Thompson PH. Ticks as vectors of equine piroplasmosis. J 1036.
Am Vet Med Assoc 1969;155:454–457. 39. Taylor WM, Bryant JE, Anderson JB, et al. Equine piro-
20. Ueti MW, Palmer GH, Scoles GA, et al. Persistently plasmosis in the United States—A review. J Am Vet Med Assoc
infected horses are reservoirs for intrastadial tick-borne transmis- 1969;155:915–919.
sion of the apicomplexan parasite Babesia equi. Infect Immun 40. Ueti MW, Palmer GH, Kappmeyer LS, et al. Ability of
2008;76:3525–3529. the vector tick Boophilus microplus to acquire and transmit Babe-
21. Schwint ON, Knowles DP, Ueti MW, et al. Transmission sia equi following feeding on chronically infected horses with
of Babesia caballi by Dermacentor nitens (Acari: Ixodidae) is low-level parasitemia. J Clin Microbiol 2005;43:3755–3759.
restricted to one generation in the absence of alimentary reinfec- 41. De Waal DT. The transovarial transmission of Babesia
tion on a susceptible equine host. J Med Entomol 2008;45:1152– caballi by Hyalomma truncatum. Onderstepoort J Vet Res
1155. 1990;57:99–100.
22. Stiller D, Goff WL, Johnson LW, et al. Dermacentor varia- 42. Ikadai H, Sasaki M, Ishida H, et al. Molecular evidence
bilis and Boophilus microplus (Acari: Ixodidae): Experimental vec- of Babesia equi transmission in Haemaphysalis longicornis. Am J
tors of Babesia equi to equids. J Med Entomol 2002;39:667–670. Trop Med Hyg 2007;76:694–697.
23. Holbrook AA, Anthony DW, Johnson AJ. Observations 43. Moltmann UG, Mehlhorn H, Schein E, et al. Ultrastruc-
on the development of Babesia caballi (Nuttall) in the tropical tural study on the development of Babesia equi (Coccidia: Piro-
horse tick Dermacentor nitens Neumann. J Protozool plasmia) in the salivary glands of its vector ticks. J Protozool
1968;15:391–396. 1983;30:218–225.
24. Roby TO, Anthony DW. Transmission of equine piroplas- 44. Estrada-Pena A, Guglielmone AA, Mangold AJ. The
mosis by Dermacentor nitens Neumann. J Am Vet Med Assoc distribution and ecological ‘preferences’ of the tick Amblyomma
1963;142:768–769. cajennense (Acari: Ixodidae), an ectoparasite of humans and
25. United States Department of Agriculture APHIS National other mammals in the Americas. Ann Trop Med Parasitol
Center for Risk Analysis. Equine piroplasmosis domestic path- 2004;98:283–292.
ways assessment 2011: Pathways assessment for the spread of the 45. Schein E, Rehbein G, Voigt WP, et al. Babesia equi (Lave-
causative agents of equine piroplasmosis from the movement of a ran 1901) 1. Development in horses and in lymphocyte culture.
horse from a quarantined premises within the contiguous United Tropenmed Parasitol 1981;32:223–227.
States Updated December 2011. Available at: http://www.aphis. 46. Moltmann UG, Mehlhorn H, Schein E, et al. Fine struc-
usda.gov/animal_health/emergingissues/downloads/epdomestic- ture of Babesia equi Laveran, 1901 within lymphocytes and
pathwaysfinalapril11.pdf. Accessed February 12, 2013. erythrocytes of horses: An in vivo and in vitro study. J Parasitol
26. Basset J, Auger L. Piroplasmose vraie du cheval (P. cabal- 1983;69:111–120.
li) dans le sudest. Comp Rend Soc Biol 1931;107:629. 47. Zapf F, Schein E. The development of Babesia (Theileria)
27. Knowles DP Jr, Kappmeyer LS, Stiller D, et al. Antibody equi (Laveran, 1901) in the gut and the haemolymph of the
to a recombinant merozoite protein epitope identifies horses vector ticks. Hyalomma species. Parasitol Res 1994;80:297–302.
infected with Babesia equi. J Clin Microbiol 1992;30:3122–3126. 48. Guimaraes AM, Lima JD, Ribeiro MF, et al. Ultrastruc-
28. Sonenshine DE. Biology of Ticks. New York, NY: Oxford ture of sporogony in Babesia equi in salivary glands of adult
University Press; 1991. female Boophilus microplus ticks. Parasitol Res 1998;84:69–74.
29. Asgarali Z, Coombs DK, Mohammed F, et al. A serologi- 49. Gerstenberg C, Allen WR, Phipps LP. The mechanical
cal study of Babesia caballi and Theileria equi in Thoroughbreds transmission of Babesia equi infection in a British herd of horses.
in Trinidad. Vet Parasitol 2007;144:167–171. In: Proceedings of the Eighth International Conference on Equine
30. Rampersad J, Cesar E, Campbell MD, et al. A field evalu- Infectious Diseases. Dubai, United Arab Emirates, 1998:100.
ation of PCR for the routine detection of Babesia equi in horses. 50. Simpson CF, Neal FC. Ultrastructure of Babesia equi in
Vet Parasitol 2003;114:81–87. ponies treated with imidocarb. Am J Vet Res 1980;41:267–271.
31. Viljoen A, Saulez MN, Donnellan CM, et al. After-hours 51. Kuttler KL, Gipson CA, Goff WL, et al. Experimental
equine emergency admissions at a university referral hospital Babesia equi infection in mature horses. Am J Vet Res
(1998–2007): Causes and interventions. J S Afr Vet Assoc 1986;47:1668–1670.
2009;80:169–173. 52. Allen PC, Frerichs WM, Holbrook AA. Experimental
32. de Waal DT, van Heerden J. Equine piroplasmosis. In: acute Babesia caballi infections. I. Red blood cell dynamics. Exp
Coetzer JAW, Tustin RC, eds. Infectious Diseases of Livestock, Parasitol 1975;37:67–77.
2nd ed. Cape Town, South Africa: Oxford University Press; 53. Ambawat HK, Malhotra DV, Kumar S, et al. Erythrocyte
2004:425–433. associated haemato-biochemical changes in Babesia equi infection
33. Xu Y, Zhang S, Huang X, et al. Seroepidemiologic studies experimentally produced in donkeys. Vet Parasitol 1999;85:319–
on Babesia equi and Babesia caballi infections in horses in Jilin 324.
province of China. J Vet Med Sci 2003;65:1015–1017. 54. de Waal DT, van Heerden J, Potgieter FT. An investiga-
34. Xuan X, Chahan B, Huang X, et al. Diagnosis of equine tion into the clinical pathological changes and serological
piroplasmosis in Xinjiang province of China by the enzyme- response in horses experimentally infected with Babesia equi and
linked immunosorbent assays using recombinant antigens. Vet Babesia caballi. Onderstepoort J Vet Res 1987;54:561–568.
Parasitol 2002;108:179–182. 55. Allen PC, Frerichs WM, Holbrook AA. Experimental
35. Cantu-Martinez MA, Segura-Correa JC, Silva-Paez ML, acute Babesia caballi infections. II. Response of platelets and
et al. Prevalence of antibodies to Theileria equi and Babesia caballi fibrinogen. Exp Parasitol 1975;37:373–379.
in horses from northeastern Mexico. J Parasitol 2012;98:869–870. 56. Donnellan CM, Marais HJ. Equine piroplasmosis. In:
36. World Organization of Animal Health (OIE) Website. Mair TS, Hutchinson RE, eds. Infectious Diseases of the Horse.
Available at: http://www.oie.int. Updated: 2013. April 1, 2013. Cambridgeshire, England, UK: EVJ Ltd; 2009:333–340.
Equine Piroplasmosis 1345

57. Georges KC, Ezeokoli CD, Sparagano O, et al. A case of 81. De Waal DT, Van Heerden J, Van den Berg SS, et al. Iso-
transplacental transmission of Theileria equi in a foal in Trinidad. lation of pure Babesia equi and Babesia caballi organisms in sple-
Vet Parasitol 2011;175:363–366. nectomized horses from endemic areas in South Africa.
58. Chhabra S, Ranjan R, Uppal S, et al. Transplacental Onderstepoort J Vet Res 1988;55:33–35.
transmission of Babesia equi (Theileria equi) from carrier mares 82. Banerjee DP, Singh B, Gautam OP, et al. Cell-mediated
to foals. J Parasit Dis 2012;36:31–33. immune response in equine babesiosis. Trop Anim Health Prod
59. Phipps LP, Otter A. Transplacental transmission of The- 1977;9:153–158.
ileria equi in two foals born and reared in the United Kingdom. 83. Kumar S, Malhotra DV, Dhar S, et al. Vaccination of
Vet Rec 2004;154:406–408. donkeys against Babesia equi using killed merozoite immunogen.
60. Lewis BD, Penzhorn BL, Volkmann DH. Could treatment Vet Parasitol 2002;106:19–33.
of pregnant mares prevent abortions due to equine piroplasmo- 84. Hanafusa Y, Cho KO, Kanemaru T, et al. Pathogenesis
sis? J S Afr Vet Assoc 1999;70:90–91. of Babesia caballi infection in experimental horses. J Vet Med Sci
61. Metcalf ES. The role of international transport of equine 1998;60:1127–1132.
semen on disease transmission. Anim Reprod Sci 2001;68:229– 85. Knowles DP Jr, Kappmeyer LS, Perryman LE. Specific
237. immune responses are required to control parasitemia in Babesia
62. Pitel P-H, Pronost S, Scrive T, et al. Molecular detection equi infection. Infect Immun 1994;62:1909–1913.
of Theileria equi and Babesia caballi in the bone marrow of 86. Magnuson NS, Perryman LE, Wyatt CR, et al. Continu-
asymptomatic horses. Vet Parasitol 2010;170:182–184. ous cultivation of equine lymphocytes: Evidence for occasional T
63. Holbrook AA. Biology of equine piroplasmosis. J Am Vet cell-like maturation events in horses with hereditary severe com-
Med Assoc 1969;155:453–454. bined immunodeficiency. J Immunol 1984;133:2518–2524.
64. Zobba R, Ardu M, Niccolini S, et al. Clinical and labora- 87. McGuire TC, Banks KL, Poppie MJ. Combined immuno-
tory findings in equine piroplasmosis. J Eq Vet Sci 2008;28:301– deficiency in horses: Characterization of the lymphocyte defect.
308. Clin Immunol Immunopathol 1975;3:555–566.
65. Diana A, Guglielmini C, Candini D, et al. Cardiac 88. Perryman LE, McGuire TC, Crawford TB. Maintenance
arrhythmias associated with piroplasmosis in the horse: A case of foals with combined immunodeficiency: Causes and control of
report. Vet J 2007;174:193–195. secondary infections. Am J Vet Res 1978;39:1043–1047.
66. Hailat NQ, Lafi SQ, al-Darraji AM, et al. Equine babesio- 89. Perryman LE, O’Rourke KI, McGuire TC. Immune
sis associated with strenuous exercise: Clinical and pathological responses are required to terminate viremia in equine infectious
studies in Jordan. Vet Parasitol 1997;69:1–8. anemia lentivirus infection. J Virol 1988;62:3073–3076.
67. Al-Saad K. Acute babesiosis in foals. J Anim Vet Adv 90. Shin EK, Perryman LE, Meek K. A kinase-negative muta-
2009;8:5. tion of DNA-PK(CS) in equine SCID results in defective coding
68. Butler CM, van Gils JA, van der Kolk JH. A literature and signal joint formation. J Immunol 1997;158:3565–3569.
review of equine piroplasmosis after an episode of acute babesio- 91. Wiler R, Leber R, Moore BB, et al. Equine severe com-
sis in a Dutch Standardbred foal after a stay in Normandy. bined immunodeficiency: A defect in V(D)J recombination and
Tijdschr Diergeneeskd 2005;130:726–731. DNA-dependent protein kinase activity. Proc Natl Acad Sci U S
69. Erbsloh JK. Babesiosis in the newborn foal. J Reprod Fer- A 1995;92:11485–11489.
til Suppl 1975;23:725–726. 92. Cunha CW, McGuire TC, Kappmeyer LS, et al. Develop-
70. Purchase HS. Piroplasmosis in foal at birth. Vet Rec ment of specific immunoglobulin Ga (IgGa) and IgGb antibodies
1947;59:449. correlates with control of parasitemia in Babesia equi infection.
71. Silvey RE. Babesiosis in a foal. Vet Rec 1996;139:428. Clin Vaccine Immunol 2006;13:297–300.
72. Neitz WO. Classification, transmission, and biology of 93. Lewis MJ, Wagner B, Woof JM. The different effector func-
piroplasms of domestic animals. Ann N Y Acad Sci 1956;64:56– tion capabilities of the seven equine IgG subclasses have implica-
111. tions for vaccine strategies. Mol Immunol 2008;45:818–827.
73. United States Department of Agriculture Animal and 94. Mealey RH, Kappmeyer LS, Ueti MW, et al.Protective
Plant Inspection Service. Equine piroplasmosis. Updated effects of passively transferred merozoite-specific antibodies
November 2012. Available at: http://www.aphis.usda.gov/ani- against Theileria equi in horses with severe combined immunode-
mal_health/animal_diseases/piroplasmosis. Accessed February ficiency. Clin Vaccine Immunol 2012;19:100–104.
12, 2013. 95. Yokoyama N, Okamura M, Igarashi I. Erythrocyte inva-
74. Alward A, Corriher CA, Barton MH, et al. Red maple sion by Babesia parasites: Current advances in the elucidation of
(Acer rubrum) leaf toxicosis in horses: A retrospective study of 32 the molecular interactions between the protozoan ligands and
cases. J Vet Intern Med 2006;20:1197–1201. host receptors in the invasion stage. Vet Parasitol 2006;138:22–32.
75. Camacho AT, Guitian FJ, Pallas E, et al. Theileria (Babe- 96. Oladosu LA. Effects of intravenous corticosteroid on the
sia) equi and Babesia caballi infections in horses in Galicia, pathogenicity of Babesia equi infection of donkeys (Equus asinus).
Spain. Trop Anim Health Prod 2005;37:293–302. Zentralbl Veterinarmed B 1988;35:509–514.
76. Rudolph W, Correa J, Zurita L, et al. Equine piroplasmo- 97. Oladosu LA, Olufemi BE. Haematology of experimental
sis: Leukocytic response to Babesia equi (Laveran, 1901) infection babesiosis and ehrlichiosis in steroid immunosuppressed horses.
in Chile. Br Vet J 1975;131:601–609. Zentralbl Veterinarmed B 1992;39:345–352.
77. Frerichs WM, Holbrook AA. Feeding mechanisms of 98. Equine piroplasmosis terrestrial manual 2008. Updated:
Babesia equi. J Protozool 1974;21:707–709. 2012. http://www.oie.int/fileadmin/Home/eng/Health_standards/
78. Mahoney DF, Wright IG, Frerichs WM, et al. The identi- tahm/2.05.08_EQUINE_PIROPLASMOSIS.pdf. Accessed April
fication of Babesia equi in Australia. Aust Vet J 1977;53:461–464. 1, 2013.
79. Frerichs WM, Johnson AJ, Holbrook AA. Equine piro- 99. Br€uning A. Equine piroplasmosis an update on diagnosis,
plasmosis: Attempts to infect laboratory animals with Babesia treatment and prevention. Br Vet J 1996;152:139–151.
equi. Am J Vet Res 1969;30:1333–1336. 100. Weiland G. Species-specific serodiagnosis of equine piro-
80. Guimar~ aes AM, Lima JD, Tafuri WL, et al. Clinical and plasma infections by means of complement fixation test (CFT),
histopathological aspects of splenectomized foals infected by immunofluorescence (IIF), and enzyme-linked immunosorbent
Babesia equi. J Eq Vet Sci 1997;17:211–216. assay (ELISA). Vet Parasitol 1986;20:43–48.
1346 Wise et al

101. McGuire TC, Van Hoosier GL Jr, Henson JB. The 117. Schwint ON, Ueti MW, Palmer GH, et al. Imidocarb
complement-fixation reaction in equine infectious anemia: dipropionate clears persistent Babesia caballii infection with elim-
Demonstration of inhibition by IgG (T). J Immunol ination of transmission potential. Antimicrob Agents Chemother
1971;107:1738–1744. 2009;53:4327–4332.
102. Donnelly J, Phipps LP, Watkins KL. Evidence of mater- 118. Ueti MW, Palmer GH, Kappmeyer LS, et al. Expression
nal antibodies to Babesia equi and B caballi in foals of seroposi- of equi merozoite antigen 2 during development of Babesia equi
tive mares. Equine Vet J 1982;14:126–128. in the midgut and salivary gland of the vector tick Boophilus
103. Tenter AM, Friedhoff KT. Serodiagnosis of experimental microplus. J Clin Microbiol 2003;41:5803–5809.
and natural Babesia equi and B. caballi infections. Vet Parasitol 119. Yang S, Rothman RE. PCR-based diagnostics for infec-
1986;20:49–61. tious diseases: Uses, limitations, and future applications in acute-
104. Animal and Plant Health Inspection Service Website. care settings. Lancet Infect Dis 2004;4:337–348.
Animal Health Laboratory Information and Services. Updated 120. Grause JF, Ueti MW, Nelson JT, et al. Efficacy of imi-
November 2012. Available at: http://www.aphis.usda.gov/ docarb dipropionate in eliminating Theileria equi from experi-
animal_health/lab_info_services. Accessed February 12, 2013. mentally infected horses. Vet J 2013;196:541–546.
105. Knowles DP Jr, Perryman LE, Kappmeyer LS, et al. 121. Nagai A, Yokoyama N, Matsuo T, et al. Growth-inhibi-
Detection of equine antibody to Babesia equi merozoite proteins tory effects of artesunate, pyrimethamine, and pamaquine against
by a monoclonal antibody-based competitive inhibition enzyme- Babesia equi and Babesia caballi in in vitro cultures. Antimicrob
linked immunosorbent assay. J Clin Microbiol 1991;29:2056– Agents Chemother 2003;47:800–803.
2058. 122. Frerichs WM, Allen PC, Holbrook AA. Equine piroplas-
106. Knowles DP Jr, Perryman LE, Goff WL, et al. A mono- mosis (Babesia equi): Therapeutic trials of imidocarb dihydro-
clonal antibody defines a geographically conserved surface chloride in horses and donkeys. Vet Rec 1973;93:73–75.
protein epitope of Babesia equi merozoites. Infect Immun 123. Frerichs WM, Holbrook AA. Treatment of equine piro-
1991;59:2412–2417. plasmosis (B caballi) with imidocarb dipropionate. Vet Rec
107. Cunha CW, Kappmeyer LS, McGuire TC, et al. Confor- 1974;95:188–189.
mational dependence and conservation of an immunodominant 124. Kumar S, Gupta AK, Pal Y, et al. In-vivo therapeutic
epitope within the Babesia equi erythrocyte-stage surface protein efficacy trial with artemisinin derivative, buparvaquone and imi-
equi merozoite antigen 1. Clin Diagn Lab Immunol 2002;9: docarb dipropionate against Babesia equi infection in donkeys. J
1301–1306. Vet Med Sci 2003;65:1171–1177.
108. Kappmeyer LS, Perryman LE, Hines SA, et al. Detection 125. Zaugg JL, Lane VM. Evaluations of buparvaquone as a
of equine antibodies to Babesia caballi by recombinant B. caballi treatment for equine babesiosis (Babesia equi). Am J Vet Res
rhoptry-associated protein 1 in a competitive-inhibition enzyme- 1989;50:782–785.
linked immunosorbent assay. J Clin Microbiol 1999;37:2285– 126. Bork S, Yokoyama N, Ikehara Y, et al. Growth-inhibi-
2290. tory effect of heparin on Babesia parasites. Antimicrob Agents
109. Bhoora R, Quan M, Zweygarth E, et al. Sequence heter- Chemother 2004;48:236–241.
ogeneity in the gene encoding the rhoptry-associated protein-1 127. Bork S, Yokoyama N, Matsuo T, et al. Growth inhibi-
(RAP-1) of Babesia caballi isolates from South Africa. Vet Paras- tory effect of triclosan on equine and bovine Babesia parasites.
itol 2010;169:279–288. Am J Trop Med Hyg 2003;68:334–340.
110. Bhoora R, Quan M, Matjila PT, et al. Sequence heteroge- 128. Bork S, Yokoyama N, Matsuo T, et al. Clotrimazole,
neity in the equi merozoite antigen gene (ema-1) of Theileria equi ketoconazole, and clodinafop-propargyl as potent growth inhibi-
and development of an ema-1-specific TaqMan MGB assay for the tors of equine Babesia parasites during in vitro culture. J Parasi-
detection of T. equi. Vet Parasitol 2010;172:33–45. tol 2003;89:604–606.
111. Kumar S, Kumar Y, Malhotra DV, et al. Standardisa- 129. Meyer C, Guthrie AJ, Stevens KB. Clinical and clinico-
tion and comparison of serial dilution and single dilution enzyme pathological changes in 6 healthy ponies following intramuscular
linked immunosorbent assay (ELISA) using different antigenic administration of multiple doses of imidocarb dipropionate. J S
preparations of the Babesia (Theileria) equi parasite. Vet Res Afr Vet Assoc 2005;76:26–32.
2003;34:71–83. 130. Bacchi CJ, Nathan HC, Hutner SH, et al. Prevention by
112. Katz J, Dewald R, Nicholson J. Procedurally similar polyamines of the curative effect of amicarbalide and imidocarb
competitive immunoassay systems for the serodiagnosis of Babe- for Trypanosoma brucei infections in mice. Biochem Pharmacol
sia equi, Babesia caballi, Trypanosoma equiperdum, and Burk- 1981;30:883–886.
holderia mallei infection in horses. J Vet Diagn Invest 131. McHardy N, Woollon RM, Clampitt RB, et al. Efficacy,
2000;12:46–50. toxicity and metabolism of imidocarb dipropionate in the treat-
113. Battsetseg B, Xuan X, Ikadai H, et al. Detection of ment of Babesia ovis infection in sheep. Res Vet Sci 1986;41:14–
Babesia caballi and Babesia equi in Dermacentor nuttalli adult 20.
ticks. Int J Parasitol 2001;31:384–386. 132. Belloli C, Crescenzo G, Lai O, et al. Pharmacokinetics of
114. Bashiruddin JB, Camma C, Rebelo E. Molecular detec- imidocarb dipropionate in horses after intramuscular administra-
tion of Babesia equi and Babesia caballi in horse blood by PCR tion. Equine Vet J 2002;34:625–629.
amplification of part of the 16S rRNA gene. Vet Parasitol 133. Adams LG. Clinicopathological aspects of imidocarb
1999;84:75–83. dipropionate toxicity in horses. Res Vet Sci 1981;31:54–61.
115. Nicolaiewsky TB, Richter MF, Lunge VR, et al. Detec- 134. Rashid H, Chaudhry M, Rashid H, et al. Comparative
tion of Babesia equi (Laveran, 1901) by nested polymerase chain efficacy of diminazene diaceturate and diminazene aceturate for
reaction. Vet Parasitol 2001;101:9–21. the treatment of babesiosis in horses. Trop Anim Health Prod
116. Bhoora R, Quan M, Franssen L, et al. Development and 2008;40:463–467.
evaluation of real-time PCR assays for the quantitative detection 135. Wise LN, Ueti MW, Kappmeyer LS, et al. In vitro activ-
of Babesia caballi and Theileria equi infections in horses from ity of ponazuril against Theileria equi. Vet Parasitol
South Africa. Vet Parasitol 2010;168:201–211. 2012;185:282–285.

Potrebbero piacerti anche