Next Article in Journal
A Baculovirus Expression Vector Derived Entirely from Non-Templated, Chemically Synthesized DNA Parts
Next Article in Special Issue
A Survey of Henipavirus Tropism—Our Current Understanding from a Species/Organ and Cellular Level
Previous Article in Journal
Correction: Kaewchim et al. Neutralizing and Enhancing Epitopes of the SARS-CoV-2 Receptor-Binding Domain (RBD) Identified by Nanobodies. Viruses 2023, 15, 1252
Previous Article in Special Issue
Henipavirus Matrix Protein Employs a Non-Classical Nuclear Localization Signal Binding Mechanism
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Animal Models for Henipavirus Research

by
Declan D. Pigeaud
1,2,
Thomas W. Geisbert
1,2,3 and
Courtney Woolsey
1,3,*
1
Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
2
Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
3
Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(10), 1980; https://doi.org/10.3390/v15101980
Submission received: 1 September 2023 / Revised: 19 September 2023 / Accepted: 21 September 2023 / Published: 22 September 2023
(This article belongs to the Special Issue Henipaviruses: An Ongoing Threat to Livestock and Humans)

Abstract

:
Hendra virus (HeV) and Nipah virus (NiV) are zoonotic paramyxoviruses in the genus Henipavirus (HNV) that emerged nearly thirty years ago. Outbreaks of HeV and NiV have led to severe respiratory disease and encephalitis in humans and animals characterized by a high mortality rate. Despite the grave threat HNVs pose to public health and global biosecurity, no approved medical countermeasures for human use currently exist against HeV or NiV. To develop candidate vaccines and therapeutics and advance the field’s understanding of HNV pathogenesis, animal models of HeV and NiV have been instrumental and remain indispensable. Various species, including rodents, ferrets, and nonhuman primates (NHPs), have been employed for HNV investigations. Among these, NHPs have demonstrated the closest resemblance to human HNV disease, although other animal models replicate some key disease features. Here, we provide a comprehensive review of the currently available animal models (mice, hamsters, guinea pigs, ferrets, cats, dogs, nonhuman primates, horses, and swine) to support HNV research. We also discuss the strengths and limitations of each model for conducting pathogenesis and transmission studies on HeV and NiV and for the evaluation of medical countermeasures.

1. Introduction

Hendra virus (HeV) and Nipah virus (NiV) are zoonotic pathogens found within the Henipavirus (HNV) genus (family Paramyxoviridae) that emerged in the 1990s [1,2,3]. Following the identification of HeV and NiV, other HNV members were later discovered, including Cedar virus [4], Mojiang virus [5], Ghana virus [6,7], and Langya virus [8]. Both HeV and NiV cause severe respiratory and neurological diseases in humans and certain livestock [1,2]. Fruit bats of the Pteropus genus are the natural reservoirs of NiV and HeV and replicating virus has been isolated in several bat species local to endemic regions of outbreaks [9,10,11]. The suspected natural hosts of Mojiang and Langya viruses are rats and shrews, respectively [5,8].
HeV first emerged in September of 1994, after several horses at a stable in Brisbane, Australia, developed a severe respiratory illness that was lethal in 65% of animals infected [12]. Two humans who came into direct contact with the fatally infected horses developed a severe influenza-like illness, which was lethal in one individual [1,12,13,14]. In 2004, 2008, and 2009, subsequent human cases occurred in veterinary or husbandry staff who came into contact with infected horses [15,16,17]. Additional equine outbreaks have occurred since then, but no other human cases have been reported. Since HeV’s emergence, there have been at least 63 spillover events in horses with an overall fatality rate of ~75% [18]. Only seven human cases of HeV have been recorded, 4 of which were fatal, and all occurred after direct contact with infected, symptomatic horses [19]. A second genotype of HeV (HeV-g2) was discovered in flying foxes [20] and horses [21]. HeV-g2 shares ~83% sequence homology to the prototype HeV. This genotype is lethal in horses, yet its pathogenicity in humans and nonhuman primates is presently unknown.
NiV was identified following an outbreak among pig farmers, first in Malaysia and then in Singapore during 1998 and 1999, resulting in 265 total cases and 105 deaths, corresponding to a mortality rate of 39% [2,3,22,23]. Epidemiological investigations early in these outbreaks found that many NiV-infected patients had direct contact with blood, tissues, or secretions of pigs suspected of NiV infection [2,3]. In Malaysia, as many as 93% of patients reported direct contact with swine in the two weeks prior to developing symptoms of NiV encephalitis [2]. A case-control study identified that close contact with pigs was the primary source of the NiV outbreak in Malaysia [24]. Furthermore, this initial outbreak of NiV in Malaysia and Singapore was quelled by the culling of an estimated 1 million pigs in Malaysia and the cessation of pig exports from Malaysia to Singapore [22]. Overall, the total number of NiV cases recorded exceeds 650, and the continual emergence of the virus in Bangladesh and India increases these numbers almost annually [25].
The ominous potential of NiV was swiftly realized as it manifested as febrile encephalitis and severe respiratory disease in humans, with case fatality rates (CFR) of up to 100% in certain outbreaks [26,27,28]. Genetic analysis has identified at least two strains of NiV responsible for outbreaks in different geographical areas: NiV-Malaysia (NiV-M) and NiV-Bangladesh (NiV-B) [29,30]. NiV-M and NiV-B share ~92% nucleotide sequence homology [30] and may exhibit divergent pathologies in humans [23,31]. The Malaysia strain caused the initial outbreak of NiV from 1998–1999 in Malaysia and Singapore and resulted in a high percentage of patients with neurological disease, with most encephalitic cases correlating with lethality in these outbreaks [26,27,28]. An additional outbreak of the Malaysia strain in the Philippines in 2014 was associated with a ~82% CFR in patients presenting with acute encephalitis syndrome [32]. The Bangladesh strain of NiV has caused repeated and near-annual outbreaks in Bangladesh and India between 2001 and 2023 [27,28,33,34]. The outbreaks caused by NiV-B have had higher CFRs, averaging about 75% [31], with human-to-human transmission also observed [35,36]. A 2018 outbreak of NiV-B in India resulted in 21 deaths among 23 total cases (91% CFR) [33], and a current outbreak of NiV-B in Bangladesh has resulted in 11 cases with eight deaths [34]. Subsequent outbreaks of NiV-B that have occurred in Bangladesh and/or India have reported a higher proportion of respiratory disease observed than in Malaysia [31,37]. Although imprecise characterizations have distinguished the Malaysia strain for causing neurological disease and the Bangladesh strain for causing pulmonary disease, it is important to note that both strains inflict pulmonary and neurological disease in both humans and animal models.
Viral entry of HNVs is initiated following binding of the attachment (G) glycoprotein to host-cell Ephrin-B2 or Ephrin-B3 ligands, after which the fusion (F) glycoprotein mediates membrane fusion between the viral and host-cell membranes, enabling viral entry to the cell [38,39]. Ephrin B2 is present in various cell types, including neurons, endothelial cells, smooth muscle surrounding arteries, placental tissue, spleen, and the lining of lymph nodes’ sinuses. In contrast, ephrin B3 is predominantly expressed in the central nervous system (CNS), particularly the spinal cord midline, and functions to prevent corticospinal tract axons from recrossing the midline [40]. Ephrin B3 is also expressed on lymphoid cells, which could potentially explain the occurrence of acute lymphoid necrosis induced by NiV infection; while lymphocytes are not directly permissive to NiV, the virus can bind these cells as a vehicle for spread into tissues [38,41]. The extensive species tropism of HNVs is primarily attributed to the conserved protein sequence of ephrin B2 and B3 across numerous species [42]. Despite this high conservation, HNVs do not cause disease in immunocompetent mice after intraperitoneal challenge [43]. Hamsters and ferrets have been the most valuable small in vivo models, particularly for triaging candidate vaccines and treatments, but these fail to reflect some major aspects of NiV disease in humans [44]. The animal model that appears to reproduce human NiV infection most faithfully is African green monkeys (AGMs) [44,45], which we will discuss in detail below.
This review comprehensively evaluates the suitability of current animal models for conducting studies on the pathogenesis and transmission of HeV and NiV and assessment of experimental countermeasures. A detailed examination of the strengths and limitations of each model is offered to facilitate an informed selection of the optimal model for the intended research purpose. Figure 1 describes the general suitability of several animal models for specific research objectives.

2. Henipavirus Infection in Humans

2.1. Hendra Virus

HeV infection in humans produces an acute respiratory and encephalitic disease. At present, seven human patients are confirmed to have been infected with HeV, five of whom died. The incubation period ranges from as few as four days to two weeks following exposure. All cases initially exhibit non-specific, flu-like symptoms of fever, cough, myalgia, nausea, and lethargy, which subsequently progress to acute respiratory distress and encephalitis in fatal cases [13,15,16]. Findings in human cases of HeV infection included pneumonitis, respiratory failure, renal failure, and arterial thrombosis [15]. Neurological signs and symptoms are presented as headache and confusion, and, in fatal cases, tonic-clonic seizure and ataxia [13]. Notably, a single case of fatal relapsing encephalitis occurred approximately 13 months after acute signs of HeV disease resolved [46]. Importantly, no human-to-human transmission of HeV has been observed.

2.2. Nipah Virus

Clinically, the incubation period for NiV infection ranges from a few days to two weeks [23,31]. NiV can cause atypical pneumonia [23] or necrotizing alveolitis with hemorrhage, pulmonary edema, and aspiration pneumonia [47], leading to acute respiratory distress syndrome. The pulmonary syndrome is presented as an influenza-like illness, with hypoxemia and diffuse alveolar shadowing in chest X-rays. NiV also causes severe encephalitis with cognitive, sensory, and motor neurological signs and symptoms [22,23,31,48]. Clinical signs reported in NiV encephalitic syndrome include areflexia, hypotonia, abnormal pupillary and doll’s eye reflex, tachycardia, hypertension, myoclonus, meningism, convulsions, and seizures. Brain MRI scans in acute NiV encephalitis show disseminated, small, discrete hyperintense lesions in both grey and white matter. Targeting endothelial cells by NiV leads to systemic vasculitis and infection of multiple organ systems, including the kidneys, liver, heart, and brain [49,50]. Pulmonary or encephalitic disease can be lethal, and many survivors of encephalitic infection face long-term neurological sequelae [51]. Late-onset and relapsing encephalitis due to prior NiV infection has been reported months to years after initial infection, with the longest time to relapse described so far being 11 years [52,53,54].

3. Animal Models of HNV Infection—Initial Studies

The first animal challenge studies with HeV (previously equine morbillivirus) were performed in healthy adult horses to reproduce the severe respiratory disease observed during the initial outbreak [1,12,13]. The experiments successfully recapitulated clinical signs of disease seen in natural infection, including anorexia, fever, and respiratory distress, with the primary accompanying pathology being pulmonary edema and histological evidence of syncytial cell formation of vascular endothelium across lung, brain, spleen, heart, and kidneys [1]. Shortly after the initial equine experiments, a pilot study was performed to identify the susceptibility of several common laboratory species, including mice, guinea pigs, rats, chickens, rabbits, cats, and dogs with 5 × 104 TCID50 of HeV administered intraperitoneally (i.p.) [55]. Only in guinea pigs and cats were clinical signs of disease observed. A wide range of species were then investigated for suitability as models of HNV disease.

4. Mice

The mouse model for HNV disease has not been widely used in the field for studies of pathogenesis or medical countermeasures due to initial challenge experiments failing to produce clinical disease in immunocompetent mice (Table 1). Recent studies employing transgenic mice with modifications aimed to dampen the host antiviral immune response have yielded positive findings, suggesting that the model may be of utility if further refined (Table 2). A mouse model for HNV disease would be highly useful due to plentiful reagent availability, ease of handling, space requirements in Biosafety Level (BSL)-4 containment, and low cost.

4.1. Immunocompetent Mice

Initial challenge studies employing immunocompetent, juvenile mice (Table 1) did not result in clinical disease or seroconversion when inoculated i.p. with 5 × 103 TCID50 of HeV-prototype or 1 × 107 PFU of NiV-M [43,55]. Mice surviving infection with NiV were re-challenged and developed an antibody response but still did not show clinical signs of disease [43]. A later study demonstrated that intracerebral (i.c.) NiV-M infections with 1 × 105 PFU were 100% lethal by day 5 post-infection in C57BL/6 mice, whereas i.p. inoculation with a 1 × 106 PFU dose in this model did not result in disease [58]. A subsequent study examining the susceptibility of juvenile (8-week-old) and aged (12-month-old) BALB/c and C57BL/6 mice to HeV found that a 5 × 104 TCID50 dose administered intranasally (i.n.) was lethal in aged mice of both genetic backgrounds [56]. This study also examined subcutaneous (s.c.) HeV challenge (5 × 104 TCID50), but mice did not exhibit clinical signs, and only a small minority developed a neutralizing antibody response. In contrast, juvenile BALB/c and C57BL/6 mice did not develop clinical signs of disease; however, histological assessment of asymptomatic mice did uncover encephalitic lesions and viral antigen deposition. Clinical signs of disease observed in aged mice challenged with HeV in this study were largely neurological, with depression, ataxia, hypersensitivity, and tremors reported, with mice succumbing to disease or meeting euthanasia criteria between 11- and 21-days post-infection. Encephalitis characterized by neuronal degradation, perivascular cuffing, and non-suppurative meningitis was identified in all C57BL/6 mice showing clinical signs of disease [56]. Antigen deposition was detected in the lungs of animals, and live virus was re-isolated in BALB/c mice; however, clinical signs of respiratory distress were not observed, and no histological lesions were present in major target organs of HeV infection [56].

4.2. Immunodeficient Mice

The susceptibility of immunocompromised mice has also been evaluated, with type I interferon receptor knockout (IFNAR-KO) mice demonstrating uniform lethality for both NiV and HeV (Table 2) [58]. I.p. infection with 1 × 106 PFU in 3– (HeV) and 9–10-week-old (NiV) IFNAR-KO mice achieved uniform lethality [58]. Groups of 3- and 12-week-old IFNAR-KO mice were next i.n. challenged with 1 × 106 PFU of HeV, but neither group developed clinical disease. Nevertheless, HeV genomic RNA was detected in the brain at levels lower than groups challenged i.p. [58]. Interestingly, immunocompetent mice supported higher levels of HeV RNA in the brain following i.n. infection as compared to those inoculated via the s.c. route at an identical challenge dose [56]. Among many potential reasons, this disparity may be attributed to the i.n. route delivering more infectious particles in close anatomical proximity to the olfactory bulb. Not only was i.n. challenge lethal in aged immunocompetent mice, but the challenge dose was also lower than that used in the IFNAR-KO mice [56,58]. Taken together, these findings indicate that lethality is both dose- and route-dependent. It is unclear how lethality was higher in immunocompetent mice inoculated with a lower dose than that used in the IFNAR-KO mouse model. These discrepancies warrant further investigation.
IFNAR-KO mice infected i.p. with NiV-M showed clinical signs consistent with neurological disease, including head tilt and locomotor disabilities that progressed to paralysis in the late stages of infection. Breathing difficulty was seen only in some i.n. challenged animal. Other clinical signs included hunched posture, decreased grooming, and weight loss. The LD50 for NiV-M in IFNAR-KO mice inoculated via the i.p. route was determined to be 8 × 103 PFU. Transgenic mice deficient in multiple innate pattern recognition signaling adaptor proteins including myeloid differentiation primary response 88 (MyD88), TIR-domain-containing adapter-inducing interferon-b (TRIF), mitochondrial antiviral-signaling protein (MAVS), and stimulator of interferon genes (STING) (MyD88/TRIF/MAVS/STING-KO) were also assessed for susceptibility to NiV infection. These adaptor molecules are implicated in the antiviral sensing activities of toll-like receptors (TLRs) and RIG-I-like receptors (RLRs), which detect DNA and RNA typically associated with viruses and their replication cycle. MyD88/TRIF/MAVS/STING-KO mice developed lethal disease following i.p. challenge with 1 × 106 PFU of NiV-M, and clinical signs were consistent with those observed in IFNAR-KO studies [61]. Principal clinical signs aligned with a neurological-skewed disease course as hunched posture, prostration, and paralysis were observed, with all (MyD88/TRIF/MAVS/STING-KO) subjects succumbing to disease by 11 days post-infection [62]. Gross or histopathologic lesions were not described by the authors of this study; however, antigen was detected in the brain.
Table 2. Experimental findings in immunodeficient mouse models of HNV Disease.
Table 2. Experimental findings in immunodeficient mouse models of HNV Disease.
Species/Genetic Background (Age)Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
IFNAR-KO
(3-week-old)
HeV
(prototype)
1 × 106
PFU
i.p.100%Weight loss, aggressiveness, prostration, paralysis[58]
IFNAR-KO
(6-week-old)
~80%
IFNAR-KO
(18-week-old)
50%
IFNAR-KO1 × 106
PFU
i.n.NoneNone
NOD/SCID/γcnullNiV-M1 × 105 TCID50intra-graftNoneNone[63]
1 × 105 TCID50i.d.
IFNAR-KONiV-M (UMMC1)1 × 106
PFU
i.p.100%
(5/5)
Weight loss, aggressiveness, prostration, paralysis[58]
1 × 106
PFU
i.n60%
(3/5)
1 × 105
PFU
i.c.100%
(5/5)
a rNiV-FlucNP8 × 104
PFU
i.n.50%
(2/4)
Hunched posture[64]
8 × 105
PFU
i.p.100%
(4/4)
Hunched posture, lethargy, ataxia, seizure
C57BL/6 MyD88/MAVS-KONiV-M (UMMC1)1 × 106
PFU
i.p.66%
(4/6)
Lordosis, prostration, paralysis[61]
C57BL/6 IFNAR-KO1 × 106
PFU
83%
(5/6)
C57BL/6 MyD88/TRIF/MAVS/STING-KONiV-M (UMMC1)1 × 106
PFU
i.p.100%
(5/5)
Yes, not described[62]
C57BL/6 MyD88/TRIF/MAVS-KO1 × 106
PFU
40%
(2/5)
C57BL/6 IFNAR-KO1 × 106
PFU
80%
(4/5)
C57BL/6 MyD88 KO1 × 106
PFU
NoneNone
C57BL/6 MyD88/TRIF-KO1 × 106
PFU
None
IFNAR-KO
(37–44-day-old)
NiV-M1 × 104 TCID50i.p.50%
(4/8)
Clinical signs including weight loss b[59]
1 × 106 TCID5063%
(5/8)
1 × 107 TCID5075%
(6/8)
a Recombinant NiV containing a luciferase reporter gene. b Comprehensive numerical clinical scores were described in this study, with individual signs not specified for subjects.
Currently, no published studies employ mice as a model to study antiviral countermeasures to HNV infection. The use of immunocompromised mouse strains clouds interpretations of the immune response to HNV infection or medical countermeasures. Additional challenge studies should be performed to refine the challenge route and dose across multiple genotypes. Since intranasal inoculation of HeV is uniformly lethal in immunocompetent mice, determination of LD50 should be conducted. Because of the relevancy to NiV pathogenesis and transmission, aerosol challenge studies should also be carried out. Lastly, to detect pathogenicity differences, challenge studies should be performed with multiple strains, including NiV-B, the strain responsible for the most frequent, recent, and lethal outbreaks in India and Bangladesh [33,34].

5. Syrian Golden Hamster

The Syrian golden hamster (SGH; Mesocricetus auratus) model is the most common rodent model employed in HNV pathogenesis, prophylaxis, and treatment studies. Both HeV and NiV infections produce disease in SGHs that is highly similar to that observed in human cases. The SGH model was first developed for NiV in 2003 [43] and HeV in 2009 [65]. Following these initial studies, continued investigation of challenge route, dosing, and HNV strains has enabled refinement of the model (Table 3). The SGH model has been used to evaluate the in vivo efficacy of antivirals, including favipiravir [66], ribavirin [67], and chloroquine [68] against lethal NiV challenge. Experimental vaccine countermeasures to HNVs spanning a variety of platforms, including ChAdOx1 [69], virus-like particle [70], mRNA [71], and recombinant vesicular stomatitis virus [72] based vaccines, have been described. Monoclonal antibody [73] and passive immune sera treatment studies [74] have also been studied in this model.
SGHs are highly susceptible to lethal HeV infection, with an LD50 calculated to be 12 PFU in seven-week-old animals challenged via the i.p. route [65]. Inoculation with doses as low as 1 × 103 PFU i.p. resulted in death within six days of challenge [65]. Unlike humans and AGMs, the Bangladesh strain is less pathogenic than the Malaysia strain in hamsters [79]. HeV infection of SGHs resulted in acute respiratory disease marked by labored breathing and diffuse pulmonary infiltrates on thoracic radiography alongside neurological signs, including paralysis, limb trembling, and seizure [65,78]. Antigen was detected in the brain, lungs, heart, kidney, liver, and spleen as well as within blood vessel walls within 2–3 days of i.p. infection with 1 × 103 PFU of HeV [65]. A higher challenge dose (1 × 105 PFU) of HeV produced more severe lesions in the same organs. The challenge dose and route of HeV in the SGH model results in varying clinical signs and disease courses, with high-dose i.n. challenge producing a more severe respiratory disease state and rapid death, and low-dose producing a more neurological skewed disease course [78]. The lethality of HeV infection in SGH does decrease in response to the age of the hamster, with a dose of 100 LD50 being completely lethal in seven-week-old hamsters but only ~84% lethal in 11-week-old hamsters [65]. Furthermore, while seven-week-old hamsters all died by day 5 post-infection, fatally infected SGHs in the 11-week-old group died by day 12 post-infection. The potentially decreased sensitivity to lethal HeV infection with increased age must be accounted for in vaccine studies, as prime-boost dose regiments may require higher challenge doses to achieve uniform lethality in control animals.
NiV infection of SGHs results in respiratory and neurological signs of disease consistent with human infection, including acute interstitial pneumonia, systemic vasculitis, encephalitis, and death. Uniform lethality is achieved through multiple challenge routes, including i.n., i.p., and small particle aerosol. A number of experiments have been performed to elucidate the LD50 of NiV-M and NiV-B delivered i.p. and i.n., and there is some variation in these values [79,83]. As was seen with the HeV challenge, the disease severity of NiV-infected SGH is route and dose-dependent, and there are differences in pathogenicity observed between NiV-M and NiV-B. Initial studies determined the LD50 of NiV-M in SGH to be 2.7 × 102 PFU and 4.7 × 104 PFU via the i.p. and i.n. routes, respectively [43]. Doses of 1 × 104 PFU or 1 × 103 TCID50 are uniformly lethal when delivered via the i.p. route. Lethality is more reliably achieved with lower doses via the i.p. route. Time-to-death also varied depending on the challenge route, with i.p. challenged animals succumbing to the disease within 5–9 days post-infection and 9–15 days for i.n. [43]. A recent study sought to robustly determine whether differences in lethality, disease course, and time to death varied between i.n. and i.p. challenge routes. This report found that the i.p. challenge disseminated the virus more rapidly to tissues and that there was no difference in survival when the same dose was delivered [83]. Initial dose susceptibility employing serial dilutions of NiV-M ranging from 1 × 104 to 1 × 106 TCID50 i.n. and 1 × 103 to 1 × 105 TCID50 i.p. were uniformly lethal; however, a dose of 1 × 102 TCID50 i.p. resulted in 1 of 4 animals surviving, which is consistent with other studies [79,83]. The challenge of SGHs with 1 × 102 TCID50 of NiV-B delivered i.p. resulted in 100% survival in a separate study by a different group [79]. Challenge of SGH via the i.n. route produces a respiratory-skewed disease, and i.p. challenge results in more neurological signs. Lesion severity in the lungs diverges at the early stages of the disease. Severe broncho-interstitial pneumonia and thickening of alveolar walls occurred at days 2 and 4 post-infection, whereas i.p. challenged animals showed only mild interstitial pneumonia [83]. Viral RNA was detected in high abundance starting at day 2 post-infection in i.n. challenged subjects; however, no RNA was observed at this time point in i.p. challenged animals. This is likely because the i.n. delivery ensures large amounts of virus travel deep into the lungs, producing acute and severe respiratory disease. Meningitis was observed only in i.p. challenged animals, and no viral RNA was found in the brain of i.n. challenged animals [83].
The challenge of SGHs with whole-body aerosols containing NiV-M at doses of 1 × 102 to 1 × 105 PFU resulted in uniform lethality at the highest dose, with an estimated LD50 of less than 1 × 102 PFU [80]. Animals exhibited clinical signs of disease starting five days post-infection, including weight loss and decreased grooming, which progressed to severe respiratory and neurological signs of disease. The mean time-to-death occurred around seven days post-infection for animals receiving a 1 × 105 PFU dose [80]. Prominent pathological findings included vasculitis of medium and large blood vessels, edema, broncho-interstitial pneumonia, and mild meningitis. As seen with other routes of challenge, lower challenge doses resulted in incomplete lethality and extended disease courses. Notably, there was no correlation between dose and whether the infection produced a neurological or respiratory-focused disease, as animals in all dose groups demonstrated a mix of respiratory and neurological disease. These studies suggest that the aerosol challenge of NiV in the SGH model is useful for evaluating medical countermeasures against potential natural routes of exposure.
There is no evidence of HeV transmission between SGHs through direct contact or aerosols. Naïve animals co-housed with HeV-infected subjects did not show clinical signs of disease or seroconvert by study endpoint [65]. Similar studies evaluated viral shedding and transmission of NiV via direct contact, aerosol, and fomites. Virus shedding detected by titration was identified in oral swabs with doses as low as 1 × 103 TCID50 i.n.; however, the virus was detected in oral, nasal, rectal, and urogenital swabs from animals inoculated with a higher dose of 1 × 107 TCID50 of NiV-M [77]. Naïve SGHs exposed to aerosols from NiV-M infected subjects did not seroconvert nor exhibit clinical signs of disease. However, viral shedding was detected by qRT-PCR in naïve SGHs co-housed with NiV-M infected subjects or those exposed to bedding, food, and water from infected animals [77]. None of the naïve animals in the fomite or aerosol transmission group seroconverted by study endpoint; however, several animals from the direct contract transmission group did, which is consistent with other direct-contact NiV transmission experiments in the SGH model [43]. Foodborne transmission of NiV has also been investigated; ~63% of SGHs drinking simulated date palm sap contaminated with 5 × 108 TCID50 of NiV-B developed neurological disease [85].

6. Guinea Pig

The guinea pig (Cavia porcellus) was among the first rodents to be experimentally infected with Hendra virus (Table 4) [55]. Four out of five guinea pigs challenged via the s.c. route with 5 × 104 TCID50 of HeV succumbed to disease 8–13 days post-infection after clinical signs of acute respiratory distress were observed [55,86]. Only one of twelve guinea pigs challenged i.n. with 3 × 104 TCID50 of HeV showed clinical signs of dyspnea and was euthanized; however, all asymptomatic animals seroconverted and remained clinically well through the study endpoint [87]. Intradermal inoculation also failed to produce clinical disease, and animals did not seroconvert [87]. The guinea pig model recapitulated the systemic vasculitis seen in human HeV infection, with principal histological findings including syncytial cell formation in vascular endothelium, fibrinoid degeneration of arteries and veins, and thrombi [86,87]. Vasculitis in the lungs, heart, kidney, urinary bladder, and brain was marked by infiltration of mononuclear cells, eosinophils and fibroblasts into vessel walls and surrounding structures. In contrast to the disease seen in humans and horses, signs of severe pulmonary edema or acute encephalitis were absent. Pregnant guinea pigs challenged i.p. with 5 × 104 TCID50 of HeV showed clinical signs consistent with previous studies, yet infection was abortive in one subject, and HeV was re-isolated from fetal tissues [88]. Placental tissue also showed multi-focal necrosis and vascular degeneration.
Limited studies have been performed to establish guinea pigs as a model for NiV disease. An initial study showed guinea pigs i.n. infected with 6 × 105 PFU of NiV-M did not result in clinical disease or seroconversion by the study endpoint, and challenge with 1 × 107 PFU i.p. resulted in only low fever and weight loss [43]. The lack of clinical disease in guinea pigs challenged i.n. with NiV or HeV indicates that this challenge route is not conducive to productive infection. Guinea pigs challenged with a lower i.p. dose of 5 × 104 TCID50 produced similarly mild clinical signs, including ruffled fur and slight ataxia 7–9 days post-infection [90]. A subsequent study in guinea pigs exposed to 6 × 104 PFU of NiV-M resulted in severe clinical signs of disease and 92% lethality within 4–8 days post-infection [91]. Histopathological lesions were present in guinea pig lung, spleen, brain, bladder, uterus, kidney and lymph nodes, and consisted of epithelial syncytial cell formation and fibrinoid necrosis. It is unclear why 1 × 107 PFU i.p. did not result in clinical signs, but 6 × 104 PFU caused nearly uniformly lethal disease. Except for the ataxia reported in one guinea pig challenged with 5 × 104 TCID50, clinical neurological signs are not reported for NiV challenge in guinea pigs. However, histopathological lesions can include viral and eosinophilic inclusions, meningeal vasculitis, and lymphocytic meningitis [91]. The most severe lesions are found in the urogenital tract. Further studies are needed in this model to determine appropriate infective doses and whether NiV-B produces similar disease.

7. Ferret

The domesticated ferret (Mustela putorius furo) is a well-characterized model that effectively recapitulates human NiV and HeV disease (Table 5). In ferrets, infection with HNVs through the combined intranasal/intratracheal (i.t./i.n.) or oronasal (o.n.) route leads to the onset of fever between days 4 and 7 post-challenge [92,93]. This fever progresses rapidly, resulting in severe respiratory distress and neurological signs appearing between days 6 and 10, depending on the challenge dose. Clinical manifestations in ferrets include coughing, nasal discharge, dyspnea, cerebral edema, tremors, and partial or complete limb paralysis. Unlike hamsters, there is no observable connection between the challenge dose and the clinical outcome in ferrets.
Gross pathological changes mainly manifest within the respiratory tract and include scattered pinpoint hemorrhagic lesions throughout the lung surface (Figure 2). These lesions stand out as they are notably different from the extensive hemorrhagic lesions found in hamsters and AGMs [78,103,104]. Histopathological alterations are primarily concentrated in the lungs, spleen, and kidneys [92] and include acute focal necrotizing alveolitis and pulmonary vasculitis, as well as necrosis in the spleen and kidneys. While vasculitis and encephalitis are typically absent, occasional instances of nonsuppurative meningitis can be observed. The presence of viral antigen is detected in syncytial cells of small blood vessels and alveolar walls in the lungs, along with necrotic glomerular and tubular epithelium in the kidneys. Despite minimal histopathological changes in the brain, HNV antigen is identified in meningeal blood vessels, choroid plexus endothelium, and neurons. Antigen detection is noted in various tissues, including the respiratory tract, brain, liver, spleen, and kidneys [92,98].
The ferret model has been utilized to compare potential distinctions in transmission and pathogenesis between the Malaysia and Bangladesh strains of NiV [98]. Throughout the infection course in ferrets, notably higher virus levels are retrieved from oral secretions in animals infected with NiV-B compared to NiV-M. Nevertheless, no endeavors have been undertaken to study inter-animal transmission in this model. Beyond transmission and pathogenesis investigations, ferret models have also proven effective in evaluating vaccines and therapeutics [92,93,94]. The main disadvantage of the ferret model is that limited reagents exist to conduct advanced immunological analyses.

8. Nonhuman Primates

Experimental challenge studies of NiV and HeV have been performed in a diverse array of nonhuman primates species, AGMs (Chlorocebus aethiops) [103,104], cynomolgus macaques (Macaca fascicularis) [105], and the common marmoset (Callithrix jacchus) [106]. Lethal infection is reliably achieved in AGMs across multiple routes that simulate natural routes of exposure, including inhaled small particle aerosol and i.t./i.n. inoculation. Conversely, infection of cynomolgus macaques results in mild to no disease or inconsistent lethality [105]. AGMs are ideal for evaluating promising medical countermeasures to NiV and HeV as they faithfully recapitulate the respiratory and neurological disease observed in human cases.

8.1. African Green Monkey

As mentioned, the AGM model is the gold standard for both NiV and HeV infection, as it accurately recapitulates human pathogenesis and is amenable to multiple challenge routes, including aerosol. Moreover, a wide range of immunological reagents are available for detailed characterization of the host response [105,107].
The challenge of AGMs i.t. with 4 × 105 TCID50 of HeV resulted in a uniformly lethal disease course highly similar to that observed in humans with animals succumbing to disease or meeting euthanasia criteria 7–9 days following the challenge (Table 6) [104]. Clinical signs of disease became apparent 5–6 days after infection and included frothy nasal discharge, labored breathing, mild depression, seizures, muscle fasciculations, and radiology depicting pulmonary congestion and consolidation by days 7 and 8, indicative of interstitial pneumonia [104].
Lungs of HeV-infected AGMs were discolored, wet and heavy upon necropsy with areas of multifocal congestion and hemorrhage. Pathological findings were similar in NiV-infected AGMs (Figure 3). Infection of AGMs i.t. with NiV-M with doses ranging from 2.5 × 103–1.3 × 106 PFU resulted in mostly lethal disease with death occurring between 9- and 12-days following challenge [103]. Subjects demonstrated clinical signs of disease consistent with human NiV infection (acute respiratory distress, inappetence, lethargy, depression, fever, neurological signs) and NiV RNA was detectable in plasma samples as early as 7 dpi and in nasal swabs 4 dpi [103]. Notably, AGM models for both Malaysia and Bangladesh strains of NiV have been established [103] that are consistent with human disease [116,120]. In line with the high mortality rates in humans reported in outbreaks caused by the latter strain, NiV-B appears more pathogenic than NiV-M in the AGM model under identical experimental conditions, resulting in uniform lethality (NiV-B) as opposed to partial lethality (NiV-M) [116].
Severe neurological signs of disease, including seizure, headache, dizziness, myoclonus, and dysphasia, were reported in human NiV-M infections, with over 50% of patients presenting with reduced levels of consciousness and brain-stem dysfunction [23]. Notably, 78% of patients in the outbreak received either oral or intravenous ribavirin. Interestingly, treatment of HeV infection AGMs with ribavirin increased the propensity of animals to exhibit neurological signs (muscle fasciculations, seizure) and a delayed disease course (by 1–2 days) but ultimately did not improve survival [104]. Human patients infected with HeV have also received ribavirin therapy, and consistent with what was observed in AGMs, this treatment does not appear to benefit outcomes, although the small number of HeV patients prohibits definitive conclusions from being reached. One of the more promising treatments tested in NiV and HeV-infected AGMs is monoclonal antibodies targeting the surface glycoproteins of HNVs, including m102.4 [108,116]. Remdesivir (GS-5734) was also found to be protective [121]. A variety of vaccine countermeasures have been evaluated in the AGM model against NiV and HeV, including rVSV displaying NiV-B (G) glycoprotein [107,122], soluble HeV (G) glycoprotein [109,117], and a chimpanzee adenovirus platform (ChAdOx1) displaying the NiV-M (G) glycoprotein [123]. Thus, the AGM model is versatile and can be used to explore HNV pathogenesis (including neuropathologies) and to develop medical countermeasures.

8.2. Cynomolgus Macaque

Infection of cynomolgus macaques with 5 × 105 PFU of HeV via the i.t./i.n. route caused very mild clinical disease in some animals with signs of agitation, increased aggressiveness, and abdominal breathing, accompanied by decreased white blood cell counts and increased levels of liver enzymes ALT and AST (Table 7) [105]. HeV RNA is detectable throughout the course of infection in cynomolgus macaques starting three days post-challenge in blood samples and oral, nasal, and rectal swabs, with levels similar to that of HeV-infected AGMs. No evidence of pathological lesions was observed at necropsy, nor was viral antigen detected. HeV-infected macaques seroconverted and achieved neutralizing antibody titers approximately equal to those seen to protect AGMs from lethal HNV challenge successfully. Assessment of global transcriptional changes related to immune signaling revealed that surviving HeV-infected macaques showed upregulated levels of transcripts mapped to adaptive immunity signaling pathways, including B-cell antigen presentation and differentiation into plasma cells. Instead, downregulated transcripts were implicated in complement activation, apoptosis, and cytotoxic granule release [105]. Additionally, macaques displayed lower levels of MCP-1, a chemoattractant involved in the trans-endothelial migration of monocytes, likely reducing the systemic spread of the virus following infection. Infection with 5 × 105 PFU i.t./i.n. of NiV-M or NiV-B produced limited signs of clinical illness similar to what was observed following the HeV challenge. The lack of lethal infection, clinical disease, or obvious pathological lesions following challenge with HNVs indicates that the model is of little utility for the study of medical countermeasure efficacy against HNV infection; however, further study of the immune response could inform the development of immunomodulatory drugs.

8.3. Common Marmoset

The marmoset model is the most recent nonhuman primate model of HNV disease to be developed and recapitulates the respiratory features of human NiV infection (Table 7). Seven-year-old marmosets challenged via the i.t./i.n. route with 6.33 × 104 PFU of NiV-B succumbed to disease 8–11 days after infection and showed clinical signs of anorexia, hyperventilation, hunched posture, and hind-limb tremors within eight days following challenge [106]. X-ray of the chest revealed opacities starting at day seven post exposure. Principal gross pathological findings included pulmonary edema and multifocal hemorrhage, with diffuse, macroscopic necrosis of the liver. Histopathological lesions included syncytial cells in the heart, kidney, and pulmonary vessels. Viral antigen was identified in the heart, pulmonary vessels, liver, kidney, and spleen; however, no lesions or antigens were found in the brain [106]. The marmoset model appears to replicate the respiratory pathology and systemic vasculitis caused by NiV infection in humans and AGMs, but the model lacks lesions in the brain typical of NiV encephalitis. One key advantage of the marmoset species as a model for HNV disease is their small size, which permits greater numbers of animals to be housed and included in studies, thereby increasing statistical power. However, the availability of immunological reagents may prohibit some studies from being performed, and the pathogenesis of HeV in this model has not been studied.

8.4. Squirrel Monkey

In a single study, squirrel monkeys were exposed to a NiV-M isolate using either intravenous (i.v.) or i.n. routes (Table 7) [124]. Both infection routes used viral doses of either 1 × 103 or 1 × 107 PFU. While clinical signs were observed in monkeys infected through both i.n. and i.v. routes, the duration of the disease was longer in i.n. infected animals (lasting seven days) compared to monkeys infected i.v. (lasting 2–3 days). Among i.v. infected monkeys, 3 out of 4 animals succumbed to the disease. Monkeys infected i.n. exhibited milder states of disease, with only 2 out of 4 animals showing signs before recovering after 3–7 days of illness [124]. The observed clinical indicators in these monkeys closely resembled those documented in human infections, with notable involvement of neurological and respiratory systems. NiV-specific RNA was detectable in various organs exclusively in monkeys infected i.v., accompanied by mild histologic lesions, primarily characterized by lung parenchymal inflammation. Unlike human infections, significant vasculitis and brain abnormalities were less pronounced. Nonetheless, immunohistochemistry demonstrated viral antigen localization in the brain, lungs, spleen, and kidney extravascular parenchyma, thus confirming viral presence in these organs [124].

9. Other Models of Note

HNVs emerged via zoonosis in livestock amplifying reservoirs horses (HeV) and pigs (NiV). Initial studies of HeV in horses were necessary to determine the etiology and characterize the causative agent of the outbreaks. Since horses and pigs can transmit HNVs to humans, the development of vaccine countermeasures to break transmission has been evaluated in these models. Other species, including cats and dogs, have been evaluated for HNV permissiveness [55]. The cat model was briefly pursued and used to evaluate an experimental vaccine for NiV infection [125], which proved effective. No further countermeasure testing was reported in this model. Similarly, only a single experimental challenge study in dogs has been performed to investigate the pathogenesis and transmission of HeV in this model. Dogs and cats are not consistently used as animal models for HNV disease for a variety of reasons, including their status as companion animals, their inability to fully recapitulate the HNV disease observed in humans, and the existence of better-suited species for biomedical research. Epidemiologic surveillance and observational studies of peridomestic animals, including cats and dogs, may be of use in outbreaks due to the potential for livestock to transmit HNVs to these species, which may then be capable of subsequent transmission to humans.

9.1. Horses

Natural HeV infection of horses leads to a severe and often fatal disease characterized by a sudden onset of influenza-like signs, including fever, inappetence, lethargy, respiratory signs of frothy nasal discharge, coughing, tachypnea, and neurological signs of aggression, muscle tremors, unsteadiness, head-tilt, and altered behavior [1]. Disease progression can be rapid, with horses deteriorating within a matter of days. Naturally, infected animals die or are typically humanely euthanized 5–15 days after showing clinical signs. Due to the severity of the disease and the potential for transmission to humans, HeV infection of horses is of significant concern for both equine and public health. Experimental infection of horses (Table 8) via i.v., i.n., or o.n. routes with HeV-containing tissue homogenate from fatal equine cases or cell culture isolates of HeV resulted in rapid and fatal disease characterized by severe pulmonary edema, congestion, focal hemorrhagic lesions and cyanosis of the lungs, and enlarged spleen [126]. Histopathological findings included the presence of syncytial cells in pulmonary vessels and the kidney, alveolar edema, and systemic vasculitis found in the brain, lungs, lymph nodes, kidney, spleen, uterus, and intestine [126,127]. Natural infection of horses with NiV has been reported [32]. A veterinary vaccine formulated with the soluble HeV-G glycoprotein is currently approved for use in Australia to protect horses from HeV infection and has demonstrated efficacy in preventing outbreaks [128].

9.2. Pigs

Experimental infection of pigs with NiV or HeV produces clinical disease with both respiratory and neurological features but typically does not result in fatal outcomes (Table 9). NiV infection of humans in Malaysia and Singapore in 1999 was preceded by widespread infection of pigs on farms, with animals typically displaying a characteristic barking cough and occasionally muscle spasms, myoclonus, trembling, and fever [129]. Although experimental infection often produces a mild, self-limiting disease with clinical signs evident by day 7 post-infection, NiV is shed in nasal secretions in pigs [130]. Gross macroscopic lesions are present in the lungs and include reddish-purple consolidation and hemorrhage. Histopathological lesions include syncytial cell formation, moderate to severe meningitis, and fibrinoid necrosis of pulmonary vessels [130,131]. Landrace pigs challenged with 6.6 × 107 PFU of HeV via the o.n. route resulted in the development of clinical signs, including depression, inappetence, and weight loss as soon as four days post-infection [89]. Gottingen minipigs challenged with 2 × 107 PFU i.n. developed clinical signs of the disease 3–7 days post-infection, and at necropsy, had evident petechial hemorrhage of the lungs and lymph nodes, with histological lesions including pulmonary edema and syncytial cells. Landrace pigs developed more severe interstitial pneumonia and extensive syncytial cell formation in the alveoli and bronchiolar epithelium; infiltration of inflammatory cells was observed in the bronchial lymph nodes [89]. A variety of veterinary vaccines have been experimentally evaluated in this model [131,132,133].

9.3. Dogs

Dogs were among the first species to be experimentally infected with HeV [55]. Several dogs have been observed to be directly infected with HeV or NiV, and serosurveillance suggests infections of dogs during NiV outbreaks are frequent. Livestock-to-dog and dog-to-human transmission events pose a potential transmission risk during outbreaks. Dogs are not a natural host of HeV or NiV but potentially serve as amplifying reservoirs during epidemics. Infection of dogs with Hendra virus has been described in two experimental challenge studies (Table 10) and a case report of natural HeV infection [138], which generally describe subclinical illness with limited evidence of pathologies in the respiratory or central nervous system.
HeV is not highly pathogenic in canines, but viremia and oral shedding suggest dogs are capable of transmitting the virus to humans. The challenge of dogs with 5 × 103 TCID50 of the prototype HeV strain via the s.c. route produced no clinical signs of disease, animals did not seroconvert, and gross or histopathological lesions were absent upon necropsy [55]. In 2013, a dog was found to be naturally infected with HeV following contact with an HeV-infected horse during an outbreak and was subsequently euthanized following confirmation of infection [138]. The dog did not show clinical signs of illness, but gross- and histopathological lesions were discovered at necropsy. Primary gross lesions included reddening of the lungs, frothy edematous fluid in the trachea and bronchi, and hyperplasia of mandibular, tonsillar, bronchial, and mediastinal lymph nodes. Significant vasculitis and fibrinoid necrosis were identified in the lymph nodes, kidneys, and brain, in addition to the spleen, lungs, intestines, and liver [138]. Further histopathological lesions were identified throughout the brain, including the brain stem and meninges. Although the virus was not isolated from tissues, the dog was seropositive and viral antigen was present in the kidney [138].
Experimental challenge of beagles with HeV dosed at 2 × 106 TCID50 via the o.n. route was not lethal and produced only mild clinical signs of disease, including inappetence and low fever at days 6 and 10 post-infection [139]. The virus was re-isolated from oral swabs at days 2 and 4 post-infection and from respiratory tissues at days 2, 4, 6, and 8; however, it was not recovered from the blood at any point [139]. Gross pathological lesions at necropsy were generally consistent with those observed in natural infection and included mild consolidation of the lungs, hyperplasia of the tonsils and tracheobronchial lymph nodes, and areas of focal hemorrhage and congestion of the mediastinal and retropharyngeal lymph nodes [138,139]. Histopathological lesions of the lungs included bronchointerstitial pneumonia, vasculitis, and antigen deposition in the bronchiolar epithelium and vascular endothelium. Limited syncytial cell formation was observed in the bronchiole lymph nodes. Notably, no evidence of neuroinfection from clinical signs or histopathological lesions was observed in experimentally infected dogs [138,139]. These findings suggest that experimental and natural HNV infections produce mild disease in dogs. Oral swabs collected from HeV-infected dogs at days 2 and 4 following the challenge were used to inoculate naïve ferrets. Ferrets developed fever, depression, and paralysis and were euthanized 7–9 days after infection, demonstrating that virus shedding from HeV-infected dogs was infective to naïve, susceptible animals [139]. This finding suggests that dogs may potentially pose a risk to humans, horses, and other peridomestic animals during HeV outbreaks.
During the 1998–1999 outbreak of NiV in Malaysia, it was reported that domestic dogs were also infected [140]. Serosurveillance in April of 1999 identified that 57% of 63 dogs sampled were positive for NiV-reactive antibodies, and additional measurements from the same area in May of 1999 saw a non-significant decrease in percent positivity to 26%, suggesting that dog-to-dog transmission was not taking place [141]. Limited data is available for dogs confirmed to have been actively infected with NiV, and no studies involving experimental infection of dogs with NiV have been published to date. During the 1999 outbreak, one moribund and one deceased dog were necropsied, with clinical signs observed in the moribund dog being respiratory distress, fever, nasal discharge, and conjunctivitis [142]. Gross findings at necropsy were consistent with human respiratory pathologies: heavy lungs with visible congestion, mottling, and consolidation of all lobes in addition to reddening of the renal capsules and cortices [142]. Histological lesions were reported in the lungs (severe pulmonary edema, interstitial pneumonia), kidneys (glomerular atrophy and syncytia), and brain (nonsuppurative meningitis). Virus isolation or antigen deposition in tissues was not described [142]. This report and evidence of seroprevalence during active NiV outbreaks indicate that close attention should be paid to dogs during outbreaks. Furthermore, disease control practices should be enforced to reduce and prevent contact between dogs and known amplifying reservoirs of NiV. Further epidemiological surveillance and investigation of apparent NiV- or HeV-infected animals during outbreaks should be a priority to understand modes of HNV transmission further.

9.4. Cat

Experimental challenge of cats revealed high susceptibility to both HeV and NiV by a variety of challenge routes and doses (Table 11). The challenge of cats with ~1 × 103–5 × 104 TCID50 of HeV s.c., o.n., or oral (o.r.) routes produces influenza-like clinical signs characterized by fever and respiratory distress [55,143]. Gross pathology in HeV-infected felines was characteristic of acute HeV disease with focal hemorrhagic lesions of the lungs, pulmonary edema and hydrothorax, and the enlargement of the spleen and mesenteric lymph nodes [86]. Histopathologic lesions were consistent with those observed in horses, including systemic vasculitis with syncytial cells, fibrinoid necrosis, and mononuclear inflammatory cell infiltration in the lungs. The virus was isolated from the respiratory tract, liver, spleen, kidney, lymph node, bladder, pleural fluid, urine, and brain. Neurological signs were not reported [86,143].
Infection of domestic shorthair cats with 5 × 102–5 × 103 TCID50 of NiV-M via the s.c. or o.n. route resulted in clinical disease characterized by fever, depression, and respiratory distress. NiV infection produced lesions highly similar to HeV-infected cats, with focal hemorrhagic necrosis of the pleura and wet and heavy lungs with edema and hydrothorax. Histopathological lesions of the lungs included bronchitis and alveolar hemorrhage. Vasculitis with syncytial cell formation and inflammatory cell infiltrates was observed in large arterial vessels of the lungs and endothelial cells of the lungs, spleen, lymph nodes, and brain [125,130]. Meningitis was also found. NiV antigen was detected in the lungs, spleen, bladder, and lymph nodes, where the infectious virus was recoverable, in addition to the tonsil [125,130]. Transmission of NiV to or from cats has not been documented during outbreaks; however, vertical transmission of NiV was observed in a cat discovered to be pregnant at necropsy [145], with infectious virus found in the placenta, fetus and uterine fluid.

10. Future Directions

Well-characterized animal models for both NiV and HeV are currently required for investigating the efficacy of medical countermeasures against these pathogens. Here, we describe several highly reliable models for recapitulating human infection by HNVs. AGMs closely mimic the pathogenesis and clinical signs of NiV and HeV disease in humans and are the gold standard for evaluating medical countermeasures, including vaccines, antivirals, and monoclonal antibodies. Hamsters and ferrets are the most well-characterized small- and mid-sized animal models for HNV disease, and continued optimization and refinement of these models has allowed for pathogenesis and countermeasure efficacy studies for HeV and NiV. Continued development of these models should be pursued to allow for better harmonization of challenge dosing and clinical scoring. The field should also prioritize the development and validation of species-specific reagents for hamsters and ferrets, as the lack of commercial availability fetters opportunities for more precise characterization of HNV pathogenesis. Models consistently recapitulating the CNS pathologies of HNV infection would also prove useful, especially for studying the recrudescence and chronic encephalitis seen in human NiV infection. This would allow for the evaluation of CNS-based therapeutics for HNV disease.
The seasonal nature of NiV and HeV virus outbreaks in their respective endemic areas continues to pose a risk to public health. The frequency and severity of NiV outbreaks in Bangladesh and India appear to be increasing, with a recent outbreak resulting in 11 cases and eight deaths earlier this year. The wide geographic distribution of natural reservoirs of HNVs and the emergence of novel HNVs, including Cedar, Mojiang and Langya, suggest continued efforts are needed to understand these viruses. Animal models have been pivotal in our study of HeV and NiV disease pathogenesis and for evaluating medical countermeasures to potentially protect against future outbreaks of these deadly viruses.

Author Contributions

Conceptualization, D.D.P. and C.W.; writing—original draft preparation, D.D.P. and C.W.; writing—review and editing, T.W.G.; resources, T.W.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Murray, K.; Selleck, P.; Hooper, P.; Hyatt, A.; Gould, A.; Gleeson, L.; Westbury, H.; Hiley, L.; Selvey, L.; Rodwell, B.; et al. A Morbillivirus That Caused Fatal Disease in Horses and Humans. Science 1995, 268, 94–97. [Google Scholar] [CrossRef] [PubMed]
  2. Chua, K.B.; Goh, K.J.; Wong, K.T.; Kamarulzaman, A.; Tan, P.S.; Ksiazek, T.G.; Zaki, S.R.; Paul, G.; Lam, S.K.; Tan, C.T. Fatal encephalitis due to Nipah virus among pig-farmers in Malaysia. Lancet 1999, 354, 1257–1259. [Google Scholar] [CrossRef] [PubMed]
  3. Paton, N.I.; Leo, Y.S.; Zaki, S.R.; Auchus, A.P.; Lee, K.E.; Ling, A.E.; Chew, S.K.; Ang, B.; Rollin, P.E.; Umapathi, T.; et al. Outbreak of Nipah-virus infection among abattoir workers in Singapore. Lancet 1999, 354, 1253–1256. [Google Scholar] [CrossRef] [PubMed]
  4. Marsh, G.A.; De Jong, C.; Barr, J.A.; Tachedjian, M.; Smith, C.; Middleton, D.; Yu, M.; Todd, S.; Foord, A.J.; Haring, V.; et al. Cedar Virus: A Novel Henipavirus Isolated from Australian Bats. PLoS Pathog. 2012, 8, e1002836. [Google Scholar] [CrossRef] [PubMed]
  5. Wu, Z.; Yang, L.; Yang, F.; Ren, X.; Jiang, J.; Dong, J.; Sun, L.; Zhu, Y.; Zhou, H.; Jin, Q. Novel Henipa-like Virus, Mojiang Paramyxovirus, in Rats, China, 2012. Emerg. Infect. Dis. 2014, 20, 1064–1066. [Google Scholar] [CrossRef]
  6. Drexler, J.F.; Corman, V.M.; Müller, M.A.; Maganga, G.D.; Vallo, P.; Binger, T.; Gloza-Rausch, F.; Cottontail, V.M.; Rasche, A.; Yordanov, S.; et al. Bats host major mammalian paramyxoviruses. Nat. Commun. 2012, 3, 796. [Google Scholar] [CrossRef]
  7. Hayman, D.T.S.; Suu-Ire, R.; Breed, A.C.; McEachern, J.A.; Wang, L.; Wood, J.L.N.; Cunningham, A.A. Evidence of Henipavirus Infection in West African Fruit Bats. PLoS ONE 2008, 3, e2739. [Google Scholar] [CrossRef]
  8. Zhang, X.-A.; Li, H.; Jiang, F.-C.; Zhu, F.; Zhang, Y.-F.; Chen, J.-J.; Tan, C.-W.; Anderson, D.E.; Fan, H.; Dong, L.-Y.; et al. A Zoonotic Henipavirus in Febrile Patients in China. N. Engl. J. Med. 2022, 387, 470–472. [Google Scholar] [CrossRef]
  9. Halpin, K.; Young, P.L.; Field, H.E.; Mackenzie, J.S. Isolation of Hendra virus from pteropid bats: A natural reservoir of Hendra virus. J. Gen. Virol. 2000, 81, 1927–1932. [Google Scholar] [CrossRef]
  10. Anderson, D.E.; Islam, A.; Crameri, G.; Todd, S.; Islam, A.; Khan, S.U.; Foord, A.; Rahman, M.Z.; Mendenhall, I.H.; Luby, S.P.; et al. Isolation and Full-Genome Characterization of Nipah Viruses from Bats, Bangladesh. Emerg. Infect. Dis. 2019, 25, 166–170. [Google Scholar] [CrossRef]
  11. Chua, K.B.; Koh, C.L.; Hooi, P.S.; Wee, K.F.; Khong, J.H.; Chua, B.H.; Chan, Y.P.; Lim, M.E.; Lam, S.K. Isolation of Nipah virus from Malaysian Island flying-foxes. Microbes Infect. 2002, 4, 145–151. [Google Scholar] [CrossRef] [PubMed]
  12. Murray, K.; Rogers, R.; Selvey, L.; Selleck, P.; Hyatt, A.; Gould, A.; Gleeson, L.; Hooper, P.; Westbury, H. A novel morbillivirus pneumonia of horses and its transmission to humans. Emerg. Infect. Dis. 1995, 1, 31–33. [Google Scholar] [CrossRef] [PubMed]
  13. Selvey, L.A.; Wells, R.M.; McCormack, J.G.; Ansford, A.J.; Murray, K.; Rogers, R.J.; Lavercombe, P.S.; Selleck, P.; Sheridan, J.W. Infection of humans and horses by a newly described morbillivirus. Med. J. Aust. 1995, 162, 642–645. [Google Scholar] [CrossRef] [PubMed]
  14. Rogers, R.J.; Douglas, I.C.; Baldock, F.C.; Glanville, R.J.; Seppanen, K.T.; Gleeson, L.J.; Selleck, P.N.; Dunn, K.J. Investigation of a second focus of equine morbillivirus infection in coastal Queensland. Aust. Vet. J. 1996, 74, 243–244. [Google Scholar] [CrossRef] [PubMed]
  15. Playford, E.G.; McCall, B.; Smith, G.; Slinko, V.; Allen, G.; Smith, I.; Moore, F.; Taylor, C.; Kung, Y.H.; Field, H. Human Hendra virus encephalitis associated with equine outbreak, Australia, 2008. Emerg. Infect. Dis. 2010, 16, 219–223. [Google Scholar] [CrossRef]
  16. Hanna, J.N.; McBride, W.J.; Brookes, D.L.; Shield, J.; Taylor, C.T.; Smith, I.L.; Craig, S.B.; Smith, G.A. Hendra virus infection in a veterinarian. Med. J. Aust. 2006, 185, 562–564. [Google Scholar] [CrossRef]
  17. Field, H.; Schaaf, K.; Kung, N.; Simon, C.; Waltisbuhl, D.; Hobert, H.; Moore, F.; Middleton, D.; Crook, A.; Smith, G.; et al. Hendra virus outbreak with novel clinical features, Australia. Emerg. Infect. Dis. 2010, 16, 338–340. [Google Scholar] [CrossRef]
  18. Eby, P.; Peel, A.J.; Hoegh, A.; Madden, W.; Giles, J.R.; Hudson, P.J.; Plowright, R.K. Pathogen spillover driven by rapid changes in bat ecology. Nature 2023, 613, 340–344. [Google Scholar] [CrossRef]
  19. Halpin, K.; Rota, P. A Review of Hendra Virus and Nipah Virus Infections in Man and Other Animals; Springer: Dordrecht, The Netherlands, 2015; pp. 997–1012. [Google Scholar]
  20. Wang, J.; Anderson, D.E.; Halpin, K.; Hong, X.; Chen, H.; Walker, S.; Valdeter, S.; Van Der Heide, B.; Neave, M.J.; Bingham, J.; et al. A new Hendra virus genotype found in Australian flying foxes. Virol. J. 2021, 18, 197. [Google Scholar] [CrossRef]
  21. Annand, E.J.; Horsburgh, B.A.; Xu, K.; Reid, P.A.; Poole, B.; De Kantzow, M.C.; Brown, N.; Tweedie, A.; Michie, M.; Grewar, J.D.; et al. Novel Hendra Virus Variant Detected by Sentinel Surveillance of Horses in Australia. Emerg. Infect. Dis. 2022, 28, 693–704. [Google Scholar] [CrossRef]
  22. Chua, K.B.; Bellini, W.J.; Rota, P.A.; Harcourt, B.H.; Tamin, A.; Lam, S.K.; Ksiazek, T.G.; Rollin, P.E.; Zaki, S.R.; Shieh, W.; et al. Nipah virus: A recently emergent deadly paramyxovirus. Science 2000, 288, 1432–1435. [Google Scholar] [CrossRef] [PubMed]
  23. Goh, K.J.; Tan, C.T.; Chew, N.K.; Tan, P.S.; Kamarulzaman, A.; Sarji, S.A.; Wong, K.T.; Abdullah, B.J.; Chua, K.B.; Lam, S.K. Clinical features of Nipah virus encephalitis among pig farmers in Malaysia. N. Engl. J. Med. 2000, 342, 1229–1235. [Google Scholar] [CrossRef] [PubMed]
  24. Parashar, U.D.; Sunn, L.M.; Ong, F.; Mounts, A.W.; Arif, M.T.; Ksiazek, T.G.; Kamaluddin, M.A.; Mustafa, A.N.; Kaur, H.; Ding, L.M.; et al. Case-Control Study of Risk Factors for Human Infection with a New Zoonotic Paramyxovirus, Nipah Virus, during a 1998–1999 Outbreak of Severe Encephalitis in Malaysia. J. Infect. Dis. 2000, 181, 1755–1759. [Google Scholar] [CrossRef]
  25. Sharma, V.; Kaushik, S.; Kumar, R.; Yadav, J.P.; Kaushik, S. Emerging trends of Nipah virus: A review. Rev. Med. Virol. 2019, 29, e2010. [Google Scholar] [CrossRef] [PubMed]
  26. Ang, B.S.P.; Lim, T.C.C.; Wang, L. Nipah Virus Infection. J. Clin. Microbiol. 2018, 56. [Google Scholar] [CrossRef] [PubMed]
  27. Gómez Román, R.; Wang, L.F.; Lee, B.; Halpin, K.; de Wit, E.; Broder, C.C.; Rahman, M.; Kristiansen, P.; Saville, M. Nipah@20: Lessons Learned from Another Virus with Pandemic Potential. mSphere 2020, 5. [Google Scholar] [CrossRef]
  28. Soman Pillai, V.; Krishna, G.; Valiya Veettil, M. Nipah Virus: Past Outbreaks and Future Containment. Viruses 2020, 12, 465. [Google Scholar] [CrossRef]
  29. Harcourt, B.H.; Tamin, A.; Ksiazek, T.G.; Rollin, P.E.; Anderson, L.J.; Bellini, W.J.; Rota, P.A. Molecular characterization of Nipah virus, a newly emergent paramyxovirus. Virology 2000, 271, 334–349. [Google Scholar] [CrossRef]
  30. Harcourt, B.H.; Lowe, L.; Tamin, A.; Liu, X.; Bankamp, B.; Bowden, N.; Rollin, P.E.; Comer, J.A.; Ksiazek, T.G.; Hossain, M.J.; et al. Genetic characterization of Nipah virus, Bangladesh, 2004. Emerg. Infect. Dis. 2005, 11, 1594–1597. [Google Scholar] [CrossRef]
  31. Hossain, M.J.; Gurley, E.S.; Montgomery, J.M.; Bell, M.; Carroll, D.S.; Hsu, V.P.; Formenty, P.; Croisier, A.; Bertherat, E.; Faiz, M.A.; et al. Clinical presentation of nipah virus infection in Bangladesh. Clin. Infect. Dis. 2008, 46, 977–984. [Google Scholar] [CrossRef]
  32. Ching, P.K.; de los Reyes, V.C.; Sucaldito, M.N.; Tayag, E.; Columna-Vingno, A.B.; Malbas, F.F., Jr.; Bolo, G.C., Jr.; Sejvar, J.J.; Eagles, D.; Playford, G.; et al. Outbreak of henipavirus infection, Philippines, 2014. Emerg. Infect. Dis. 2015, 21, 328–331. [Google Scholar] [CrossRef] [PubMed]
  33. Arunkumar, G.; Chandni, R.; Mourya, D.T.; Singh, S.K.; Sadanandan, R.; Sudan, P.; Bhargava, B. Outbreak Investigation of Nipah Virus Disease in Kerala, India, 2018. J. Infect. Dis. 2019, 219, 1867–1878. [Google Scholar] [CrossRef] [PubMed]
  34. World Health Organization. Nipah Virus Infection—Bangladesh. Available online: https://www.who.int/emergencies/disease-outbreak-news/item/2023-DON442 (accessed on 3 August 2023).
  35. Gurley, E.S.; Montgomery, J.M.; Hossain, M.J.; Bell, M.; Azad, A.K.; Islam, M.R.; Molla, M.A.; Carroll, D.S.; Ksiazek, T.G.; Rota, P.A.; et al. Person-to-person transmission of Nipah virus in a Bangladeshi community. Emerg. Infect. Dis. 2007, 13, 1031–1037. [Google Scholar] [CrossRef]
  36. Homaira, N.; Rahman, M.; Hossain, M.J.; Epstein, J.H.; Sultana, R.; Khan, M.S.; Podder, G.; Nahar, K.; Ahmed, B.; Gurley, E.S.; et al. Nipah virus outbreak with person-to-person transmission in a district of Bangladesh, 2007. Epidemiol. Infect. 2010, 138, 1630–1636. [Google Scholar] [CrossRef]
  37. Chong, V.; Hossain, M.; Chong, H. Differences in epidemiologic and clinical features of Nipah virus encephalitis between the Malaysian and Bangladesh outbreaks. Neurol. Asia 2008, 13, 23–26. [Google Scholar]
  38. Bonaparte, M.I.; Dimitrov, A.S.; Bossart, K.N.; Crameri, G.; Mungall, B.A.; Bishop, K.A.; Choudhry, V.; Dimitrov, D.S.; Wang, L.-F.; Eaton, B.T.; et al. Ephrin-B2 ligand is a functional receptor for Hendra virus and Nipah virus. Proc. Natl. Acad. Sci. USA 2005, 102, 10652–10657. [Google Scholar] [CrossRef] [PubMed]
  39. Xu, K.; Broder, C.C.; Nikolov, D.B. Ephrin-B2 and ephrin-B3 as functional henipavirus receptors. Semin. Cell Dev. Biol. 2012, 23, 116–123. [Google Scholar] [CrossRef]
  40. Negrete, O.A.; Wolf, M.C.; Aguilar, H.C.; Enterlein, S.; Wang, W.; Mühlberger, E.; Su, S.V.; Bertolotti-Ciarlet, A.; Flick, R.; Lee, B. Two Key Residues in EphrinB3 Are Critical for Its Use as an Alternative Receptor for Nipah Virus. PLoS Pathog. 2006, 2, e7. [Google Scholar] [CrossRef]
  41. Lee, B. Envelope–Receptor Interactions in Nipah Virus Pathobiology. Ann. N. Y. Acad. Sci. 2007, 1102, 51–65. [Google Scholar] [CrossRef]
  42. Eaton, T.B.; Broder, C.C.; Wang, L.-F. Hendra and Nipah Viruses: Pathogenesis and Therapeutics. Curr. Mol. Med. 2005, 5, 805–816. [Google Scholar] [CrossRef]
  43. Wong, K.T.; Grosjean, I.; Brisson, C.; Blanquier, B.; Fevre-Montange, M.; Bernard, A.; Loth, P.; Georges-Courbot, M.-C.; Chevallier, M.; Akaoka, H.; et al. A Golden Hamster Model for Human Acute Nipah Virus Infection. Am. J. Pathol. 2003, 163, 2127–2137. [Google Scholar] [CrossRef] [PubMed]
  44. Geisbert, T.W.; Feldmann, H.; Broder, C.C. Animal challenge models of henipavirus infection and pathogenesis. Curr. Top. Microbiol. Immunol. 2012, 359, 153–177. [Google Scholar] [CrossRef] [PubMed]
  45. de Wit, E.; Munster, V.J. Animal models of disease shed light on Nipah virus pathogenesis and transmission. J. Pathol. 2015, 235, 196–205. [Google Scholar] [CrossRef] [PubMed]
  46. Wong, K.T.; Robertson, T.; Ong, B.B.; Chong, J.W.; Yaiw, K.C.; Wang, L.F.; Ansford, A.J.; Tannenberg, A. Human Hendra virus infection causes acute and relapsing encephalitis. Neuropathol. Appl. Neurobiol. 2009, 35, 296–305. [Google Scholar] [CrossRef]
  47. Wong, K.T.; Shieh, W.J.; Kumar, S.; Norain, K.; Abdullah, W.; Guarner, J.; Goldsmith, C.S.; Chua, K.B.; Lam, S.K.; Tan, C.T.; et al. Nipah virus infection: Pathology and pathogenesis of an emerging paramyxoviral zoonosis. Am. J. Pathol. 2002, 161, 2153–2167. [Google Scholar] [CrossRef]
  48. Ong, K.C.; Wong, K.T. Henipavirus Encephalitis: Recent Developments and Advances. Brain Pathol. 2015, 25, 605–613. [Google Scholar] [CrossRef]
  49. Maisner, A.; Neufeld, J.; Weingartl, H. Organ- and endotheliotropism of Nipah virus infections in vivo and in vitro. Thromb. Haemost. 2009, 102, 1014–1023. [Google Scholar] [CrossRef]
  50. Wong, K.T.; Ong, K.C. Pathology of acute henipavirus infection in humans and animals. Pathol. Res. Int. 2011, 2011, 567248. [Google Scholar] [CrossRef]
  51. Sejvar, J.J.; Hossain, J.; Saha, S.K.; Gurley, E.S.; Banu, S.; Hamadani, J.D.; Faiz, M.A.; Siddiqui, F.M.; Mohammad, Q.D.; Mollah, A.H.; et al. Long-term neurological and functional outcome in Nipah virus infection. Ann. Neurol. 2007, 62, 235–242. [Google Scholar] [CrossRef]
  52. Tan, C.T.; Goh, K.J.; Wong, K.T.; Sarji, S.A.; Chua, K.B.; Chew, N.K.; Murugasu, P.; Loh, Y.L.; Chong, H.T.; Tan, K.S.; et al. Relapsed and late-onset Nipah encephalitis. Ann. Neurol. 2002, 51, 703–708. [Google Scholar] [CrossRef]
  53. Chong, H.; Tan, C. Relapsed and late-onset Nipah encephalitis, a report of three cases. Neurol. J. Southeast. Asia 2003, 8, 109–112. [Google Scholar]
  54. Abdullah, S.; Chang, L.; Rahmat, K.; Goh, K.; Tan, C. Late-onset Nipah virus encephalitis 11 years after the initial outbreak: A case report. Neurol. Asia 2012, 17, 71–74. [Google Scholar]
  55. Westbury, H.; Hooper, P.; Selleck, P.; Murray, P. Equine morbillivirus pneumonia: Susceptibility of laboratory animals to the virus. Aust. Vet. J. 1995, 72, 278–279. [Google Scholar] [CrossRef] [PubMed]
  56. Dups, J.; Middleton, D.; Yamada, M.; Monaghan, P.; Long, F.; Robinson, R.; Marsh, G.A.; Wang, L.F. A new model for Hendra virus encephalitis in the mouse. PLoS ONE 2012, 7, e40308. [Google Scholar] [CrossRef] [PubMed]
  57. Dups, J.; Middleton, D.; Long, F.; Arkinstall, R.; Marsh, G.A.; Wang, L.-F. Subclinical infection without encephalitis in mice following intranasal exposure to Nipah virus-Malaysia and Nipah virus-Bangladesh. Virol. J. 2014, 11, 102. [Google Scholar] [CrossRef]
  58. Dhondt, K.P.; Mathieu, C.; Chalons, M.; Reynaud, J.M.; Vallve, A.; Raoul, H.; Horvat, B. Type I interferon signaling protects mice from lethal henipavirus infection. J. Infect. Dis. 2013, 207, 142–151. [Google Scholar] [CrossRef]
  59. Welch, S.R.; Spengler, J.R.; Genzer, S.C.; Coleman-McCray, J.D.; Harmon, J.R.; Sorvillo, T.E.; Scholte, F.E.M.; Rodriguez, S.E.; O’Neal, T.J.; Ritter, J.M.; et al. Single-dose mucosal replicon-particle vaccine protects against lethal Nipah virus infection up to 3 days after vaccination. Sci. Adv. 2023, 9, eadh4057. [Google Scholar] [CrossRef]
  60. Edwards, S.J.; Rowe, B.; Reid, T.; Tachedjian, M.; Caruso, S.; Blasdell, K.; Watanabe, S.; Bergfeld, J.; Marsh, G.A. Henipavirus-induced neuropathogenesis in mice. Virology 2023, 587, 109856. [Google Scholar] [CrossRef]
  61. Iampietro, M.; Aurine, N.; Dhondt, K.P.; Dumont, C.; Pelissier, R.; Spanier, J.; Vallve, A.; Raoul, H.; Kalinke, U.; Horvat, B. Control of Nipah Virus Infection in Mice by the Host Adaptors Mitochondrial Antiviral Signaling Protein (MAVS) and Myeloid Differentiation Primary Response 88 (MyD88). J. Infect. Dis. 2020, 221, S401–S406. [Google Scholar] [CrossRef]
  62. Iampietro, M.; Dumont, C.; Mathieu, C.; Spanier, J.; Robert, J.; Charpenay, A.; Dupichaud, S.; Dhondt, K.P.; Aurine, N.; Pelissier, R.; et al. Activation of cGAS/STING pathway upon paramyxovirus infection. iScience 2021, 24, 102519. [Google Scholar] [CrossRef]
  63. Valbuena, G.; Halliday, H.; Borisevich, V.; Goez, Y.; Rockx, B. A Human Lung Xenograft Mouse Model of Nipah Virus Infection. PLoS Pathog. 2014, 10, e1004063. [Google Scholar] [CrossRef] [PubMed]
  64. Yun, T.; Park, A.; Hill, T.E.; Pernet, O.; Beaty, S.M.; Juelich, T.L.; Smith, J.K.; Zhang, L.; Wang, Y.E.; Vigant, F.; et al. Efficient Reverse Genetics Reveals Genetic Determinants of Budding and Fusogenic Differences between Nipah and Hendra Viruses and Enables Real-Time Monitoring of Viral Spread in Small Animal Models of Henipavirus Infection. J. Virol. 2015, 89, 1242–1253. [Google Scholar] [CrossRef] [PubMed]
  65. Guillaume, V.; Wong, K.T.; Looi, R.Y.; Georges-Courbot, M.C.; Barrot, L.; Buckland, R.; Wild, T.F.; Horvat, B. Acute Hendra virus infection: Analysis of the pathogenesis and passive antibody protection in the hamster model. Virology 2009, 387, 459–465. [Google Scholar] [CrossRef] [PubMed]
  66. Dawes, B.E.; Kalveram, B.; Ikegami, T.; Juelich, T.; Smith, J.K.; Zhang, L.; Park, A.; Lee, B.; Komeno, T.; Furuta, Y.; et al. Favipiravir (T-705) protects against Nipah virus infection in the hamster model. Sci. Rep. 2018, 8, 7604. [Google Scholar] [CrossRef]
  67. Georges-Courbot, M.C.; Contamin, H.; Faure, C.; Loth, P.; Baize, S.; Leyssen, P.; Neyts, J.; Deubel, V. Poly(I)-Poly(C12U) but Not Ribavirin Prevents Death in a Hamster Model of Nipah Virus Infection. Antimicrob. Agents Chemother. 2006, 50, 1768–1772. [Google Scholar] [CrossRef]
  68. Freiberg, A.N.; Worthy, M.N.; Lee, B.; Holbrook, M.R. Combined chloroquine and ribavirin treatment does not prevent death in a hamster model of Nipah and Hendra virus infection. J. Gen. Virol. 2010, 91, 765–772. [Google Scholar] [CrossRef]
  69. Van Doremalen, N.; Lambe, T.; Sebastian, S.; Bushmaker, T.; Fischer, R.; Feldmann, F.; Haddock, E.; Letko, M.; Avanzato, V.A.; Rissanen, I.; et al. A single-dose ChAdOx1-vectored vaccine provides complete protection against Nipah Bangladesh and Malaysia in Syrian golden hamsters. PLoS Neglected Trop. Dis. 2019, 13, e0007462. [Google Scholar] [CrossRef]
  70. Walpita, P.; Cong, Y.; Jahrling, P.B.; Rojas, O.; Postnikova, E.; Yu, S.; Johns, L.; Holbrook, M.R. A VLP-based vaccine provides complete protection against Nipah virus challenge following multiple-dose or single-dose vaccination schedules in a hamster model. NPJ Vaccines 2017, 2, 21. [Google Scholar] [CrossRef]
  71. Lo, M.K.; Spengler, J.R.; Welch, S.R.; Harmon, J.R.; Coleman-McCray, J.D.; Scholte, F.E.M.; Shrivastava-Ranjan, P.; Montgomery, J.M.; Nichol, S.T.; Weissman, D.; et al. Evaluation of a Single-Dose Nucleoside-Modified Messenger RNA Vaccine Encoding Hendra Virus-Soluble Glycoprotein Against Lethal Nipah virus Challenge in Syrian Hamsters. J. Infect. Dis. 2019, 221, S493–S498. [Google Scholar] [CrossRef]
  72. Monath, T.P.; Nichols, R.; Tussey, L.; Scappaticci, K.; Pullano, T.G.; Whiteman, M.D.; Vasilakis, N.; Rossi, S.L.; Campos, R.K.; Azar, S.R.; et al. Recombinant vesicular stomatitis vaccine against Nipah virus has a favorable safety profile: Model for assessment of live vaccines with neurotropic potential. PLoS Pathog. 2022, 18, e1010658. [Google Scholar] [CrossRef]
  73. Guillaume, V.; Contamin, H.; Loth, P.; Grosjean, I.; Courbot, M.C.G.; Deubel, V.; Buckland, R.; Wild, T.F. Antibody Prophylaxis and Therapy against Nipah Virus Infection in Hamsters. J. Virol. 2006, 80, 1972–1978. [Google Scholar] [CrossRef] [PubMed]
  74. Guillaume, V.; Contamin, H.; Loth, P.; Georges-Courbot, M.C.; Lefeuvre, A.; Marianneau, P.; Chua, K.B.; Lam, S.K.; Buckland, R.; Deubel, V.; et al. Nipah virus: Vaccination and passive protection studies in a hamster model. J. Virol. 2004, 78, 834–840. [Google Scholar] [CrossRef]
  75. Lo, M.K.; Bird, B.H.; Chattopadhyay, A.; Drew, C.P.; Martin, B.E.; Coleman, J.D.; Rose, J.K.; Nichol, S.T.; Spiropoulou, C.F. Single-dose replication-defective VSV-based Nipah virus vaccines provide protection from lethal challenge in Syrian hamsters. Antivir. Res. 2014, 101, 26–29. [Google Scholar] [CrossRef] [PubMed]
  76. Yoneda, M.; Georges-Courbot, M.C.; Ikeda, F.; Ishii, M.; Nagata, N.; Jacquot, F.; Raoul, H.; Sato, H.; Kai, C. Recombinant measles virus vaccine expressing the Nipah virus glycoprotein protects against lethal Nipah virus challenge. PLoS ONE 2013, 8, e58414. [Google Scholar] [CrossRef] [PubMed]
  77. De Wit, E.; Bushmaker, T.; Scott, D.; Feldmann, H.; Munster, V.J. Nipah Virus Transmission in a Hamster Model. PLoS Neglected Trop. Dis. 2011, 5, e1432. [Google Scholar] [CrossRef]
  78. Rockx, B.; Brining, D.; Kramer, J.; Callison, J.; Ebihara, H.; Mansfield, K.; Feldmann, H. Clinical outcome of henipavirus infection in hamsters is determined by the route and dose of infection. J. Virol. 2011, 85, 7658–7671. [Google Scholar] [CrossRef]
  79. DeBuysscher, B.L.; de Wit, E.; Munster, V.J.; Scott, D.; Feldmann, H.; Prescott, J. Comparison of the Pathogenicity of Nipah Virus Isolates from Bangladesh and Malaysia in the Syrian Hamster. PLoS Neglected Trop. Dis. 2013, 7, e2024. [Google Scholar] [CrossRef]
  80. Escaffre, O.; Hill, T.; Ikegami, T.; Juelich, T.L.; Smith, J.K.; Zhang, L.; Perez, D.E.; Atkins, C.; Park, A.; Lawrence, W.S.; et al. Experimental Infection of Syrian Hamsters With Aerosolized Nipah Virus. J. Infect. Dis. 2018, 218, 1602–1610. [Google Scholar] [CrossRef]
  81. Ploquin, A.; Szécsi, J.; Mathieu, C.; Guillaume, V.; Barateau, V.; Ong, K.C.; Wong, K.T.; Cosset, F.-L.; Horvat, B.; Salvetti, A. Protection Against Henipavirus Infection by Use of Recombinant Adeno-Associated Virus–Vector Vaccines. J. Infect. Dis. 2012, 207, 469–478. [Google Scholar] [CrossRef]
  82. Yoneda, M.; Guillaume, V.; Sato, H.; Fujita, K.; Georges-Courbot, M.-C.; Ikeda, F.; Omi, M.; Muto-Terao, Y.; Wild, T.F.; Kai, C. The Nonstructural Proteins of Nipah Virus Play a Key Role in Pathogenicity in Experimentally Infected Animals. PLoS ONE 2010, 5, e12709. [Google Scholar] [CrossRef]
  83. Findlay-Wilson, S.; Flett, L.; Salguero, F.J.; Ruedas-Torres, I.; Fotheringham, S.; Easterbrook, L.; Graham, V.; Dowall, S. Establishment of a Nipah Virus Disease Model in Hamsters, including a Comparison of Intranasal and Intraperitoneal Routes of Challenge. Pathogens 2023, 12, 976. [Google Scholar] [CrossRef]
  84. Mathieu, C.; Guillaume, V.; Volchkova, V.A.; Pohl, C.; Jacquot, F.; Looi, R.Y.; Wong, K.T.; Legras-Lachuer, C.; Volchkov, V.E.; Lachuer, J.; et al. Nonstructural Nipah Virus C Protein Regulates both the Early Host Proinflammatory Response and Viral Virulence. J. Virol. 2012, 86, 10766–10775. [Google Scholar] [CrossRef] [PubMed]
  85. De Wit, E.; Prescott, J.; Falzarano, D.; Bushmaker, T.; Scott, D.; Feldmann, H.; Munster, V.J. Foodborne Transmission of Nipah Virus in Syrian Hamsters. PLoS Pathog. 2014, 10, e1004001. [Google Scholar] [CrossRef] [PubMed]
  86. Hooper, P.T.; Westbury, H.A.; Russell, G.M. The Lesions of Experimental Equine Morbillivirus Disease in Cats and Guinea Pigs. Vet. Pathol. 1997, 34, 323–329. [Google Scholar] [CrossRef] [PubMed]
  87. Williamson, M.M.; Hooper, P.T.; Selleck, P.W.; Westbury, H.A.; Slocombe, R.F. A guinea-pig model of Hendra virus encephalitis. J. Comp. Pathol. 2001, 124, 273–279. [Google Scholar] [CrossRef]
  88. Williamson, M.M.; Hooper, P.T.; Selleck, P.W.; Westbury, H.A.; Slocombe, R.F. Experimental hendra virus infection in pregnant guinea-pigs and fruit Bats (Pteropus poliocephalus). J. Comp. Pathol. 2000, 122, 201–207. [Google Scholar] [CrossRef] [PubMed]
  89. Li, M.; Embury-Hyatt, C.; Weingartl, H.M. Experimental inoculation study indicates swine as a potential host for Hendra virus. Vet. Res. 2010, 41, 33. [Google Scholar] [CrossRef] [PubMed]
  90. Middleton, D.J.; Morrissy, C.J.; van der Heide, B.M.; Russell, G.M.; Braun, M.A.; Westbury, H.A.; Halpin, K.; Daniels, P.W. Experimental Nipah virus infection in pteropid bats (Pteropus poliocephalus). J. Comp. Pathol. 2007, 136, 266–272. [Google Scholar] [CrossRef] [PubMed]
  91. Torres-Velez, F.J.; Shieh, W.-J.; Rollin, P.E.; Morken, T.; Brown, C.; Ksiazek, T.G.; Zaki, S.R. Histopathologic and Immunohistochemical Characterization of Nipah Virus Infection in the Guinea Pig. Vet. Pathol. 2008, 45, 576–585. [Google Scholar] [CrossRef]
  92. Bossart, K.N.; Zhu, Z.; Middleton, D.; Klippel, J.; Crameri, G.; Bingham, J.; McEachern, J.A.; Green, D.; Hancock, T.J.; Chan, Y.-P. A neutralizing human monoclonal antibody protects against lethal disease in a new ferret model of acute nipah virus infection. PLoS Pathog. 2009, 5, e1000642. [Google Scholar] [CrossRef]
  93. Pallister, J.; Middleton, D.; Wang, L.-F.; Klein, R.; Haining, J.; Robinson, R.; Yamada, M.; White, J.; Payne, J.; Feng, Y.-R. A recombinant Hendra virus G glycoprotein-based subunit vaccine protects ferrets from lethal Hendra virus challenge. Vaccine 2011, 29, 5623–5630. [Google Scholar] [CrossRef] [PubMed]
  94. Pallister, J.A.; Klein, R.; Arkinstall, R.; Haining, J.; Long, F.; White, J.R.; Payne, J.; Feng, Y.-R.; Wang, L.-F.; Broder, C.C. Vaccination of ferrets with a recombinant G glycoprotein subunit vaccine provides protection against Nipah virus disease for over 12 months. Virol. J. 2013, 10, 237. [Google Scholar] [CrossRef] [PubMed]
  95. Leon, A.J.; Borisevich, V.; Boroumand, N.; Seymour, R.; Nusbaum, R.; Escaffre, O.; Xu, L.; Kelvin, D.J.; Rockx, B. Host gene expression profiles in ferrets infected with genetically distinct henipavirus strains. PLoS Neglected Trop. Dis. 2018, 12, e0006343. [Google Scholar] [CrossRef] [PubMed]
  96. Mire, C.E.; Chan, Y.-P.; Borisevich, V.; Cross, R.W.; Yan, L.; Agans, K.N.; Dang, H.V.; Veesler, D.; Fenton, K.A.; Geisbert, T.W.; et al. A Cross-Reactive Humanized Monoclonal Antibody Targeting Fusion Glycoprotein Function Protects Ferrets Against Lethal Nipah Virus and Hendra Virus Infection. J. Infect. Dis. 2020, 221, S471–S479. [Google Scholar] [CrossRef] [PubMed]
  97. Clayton, B.A.; Middleton, D.; Arkinstall, R.; Frazer, L.; Wang, L.-F.; Marsh, G.A. The Nature of Exposure Drives Transmission of Nipah Viruses from Malaysia and Bangladesh in Ferrets. PLoS Neglected Trop. Dis. 2016, 10, e0004775. [Google Scholar] [CrossRef] [PubMed]
  98. Clayton, B.A.; Middleton, D.; Bergfeld, J.; Haining, J.; Arkinstall, R.; Wang, L.; Marsh, G.A. Transmission Routes for Nipah Virus from Malaysia and Bangladesh. Emerg. Infect. Dis. 2012, 18, 1983–1993. [Google Scholar] [CrossRef]
  99. Pallister, J.; Middleton, D.; Crameri, G.; Yamada, M.; Klein, R.; Hancock, T.J.; Foord, A.; Shiell, B.; Michalski, W.; Broder, C.C.; et al. Chloroquine Administration Does Not Prevent Nipah Virus Infection and Disease in Ferrets. J. Virol. 2009, 83, 11979–11982. [Google Scholar] [CrossRef]
  100. Satterfield, B.A.; Borisevich, V.; Foster, S.L.; Rodriguez, S.E.; Cross, R.W.; Fenton, K.A.; Agans, K.N.; Basler, C.F.; Geisbert, T.W.; Mire, C.E. Antagonism of STAT1 by Nipah virus P gene products modulates disease course but not lethal outcome in the ferret model. Sci. Rep. 2019, 9, 16710. [Google Scholar] [CrossRef]
  101. Satterfield, B.A.; Cross, R.W.; Fenton, K.A.; Agans, K.N.; Basler, C.F.; Geisbert, T.W.; Mire, C.E. The immunomodulating V and W proteins of Nipah virus determine disease course. Nat. Commun. 2015, 6, 7483. [Google Scholar] [CrossRef]
  102. Satterfield, B.A.; Cross, R.W.; Fenton, K.A.; Borisevich, V.; Agans, K.N.; Deer, D.J.; Graber, J.; Basler, C.F.; Geisbert, T.W.; Mire, C.E. Nipah Virus C and W Proteins Contribute to Respiratory Disease in Ferrets. J. Virol. 2016, 90, 6326–6343. [Google Scholar] [CrossRef]
  103. Geisbert, T.W.; Daddario-DiCaprio, K.M.; Hickey, A.C.; Smith, M.A.; Chan, Y.P.; Wang, L.F.; Mattapallil, J.J.; Geisbert, J.B.; Bossart, K.N.; Broder, C.C. Development of an acute and highly pathogenic nonhuman primate model of Nipah virus infection. PLoS ONE 2010, 5, e10690. [Google Scholar] [CrossRef] [PubMed]
  104. Rockx, B.; Bossart, K.N.; Feldmann, F.; Geisbert, J.B.; Hickey, A.C.; Brining, D.; Callison, J.; Safronetz, D.; Marzi, A.; Kercher, L.; et al. A novel model of lethal Hendra virus infection in African green monkeys and the effectiveness of ribavirin treatment. J. Virol. 2010, 84, 9831–9839. [Google Scholar] [CrossRef] [PubMed]
  105. Prasad, A.N.; Woolsey, C.; Geisbert, J.B.; Agans, K.N.; Borisevich, V.; Deer, D.J.; Mire, C.E.; Cross, R.W.; Fenton, K.A.; Broder, C.C.; et al. Resistance of Cynomolgus Monkeys to Nipah and Hendra Virus Disease Is Associated with Cell-Mediated and Humoral Immunity. J. Infect. Dis. 2020, 221, S436–S447. [Google Scholar] [CrossRef] [PubMed]
  106. Stevens, C.S.; Lowry, J.; Juelich, T.; Atkins, C.; Johnson, K.; Smith, J.K.; Panis, M.; Ikegami, T.; tenOever, B.; Freiberg, A.N.; et al. Nipah Virus Bangladesh Infection Elicits Organ-Specific Innate and Inflammatory Responses in the Marmoset Model. J. Infect. Dis. 2023, 228, 604–614. [Google Scholar] [CrossRef] [PubMed]
  107. Woolsey, C.; Borisevich, V.; Fears, A.C.; Agans, K.N.; Deer, D.J.; Prasad, A.N.; O’Toole, R.; Foster, S.L.; Dobias, N.S.; Geisbert, J.B.; et al. Recombinant vesicular stomatitis virus-vectored vaccine induces long-lasting immunity against Nipah virus disease. J. Clin. Investig. 2023, 133, e164946. [Google Scholar] [CrossRef]
  108. Bossart, K.N.; Geisbert, T.W.; Feldmann, H.; Zhu, Z.; Feldmann, F.; Geisbert, J.B.; Yan, L.; Feng, Y.R.; Brining, D.; Scott, D.; et al. A neutralizing human monoclonal antibody protects african green monkeys from hendra virus challenge. Sci. Transl. Med. 2011, 3, 105ra103. [Google Scholar] [CrossRef]
  109. Mire, C.E.; Geisbert, J.B.; Agans, K.N.; Feng, Y.R.; Fenton, K.A.; Bossart, K.N.; Yan, L.; Chan, Y.P.; Broder, C.C.; Geisbert, T.W. A recombinant Hendra virus G glycoprotein subunit vaccine protects nonhuman primates against Hendra virus challenge. J. Virol. 2014, 88, 4624–4631. [Google Scholar] [CrossRef]
  110. Prasad, A.N.; Agans, K.N.; Sivasubramani, S.K.; Geisbert, J.B.; Borisevich, V.; Mire, C.E.; Lawrence, W.S.; Fenton, K.A.; Geisbert, T.W. A Lethal Aerosol Exposure Model of Nipah Virus Strain Bangladesh in African Green Monkeys. J. Infect. Dis. 2020, 221, S431–S435. [Google Scholar] [CrossRef]
  111. Lara, A.; Cong, Y.; Jahrling, P.B.; Mednikov, M.; Postnikova, E.; Yu, S.; Munster, V.; Holbrook, M.R. Peripheral immune response in the African green monkey model following Nipah-Malaysia virus exposure by intermediate-size particle aerosol. PLoS Negl. Trop. Dis. 2019, 13, e0007454. [Google Scholar] [CrossRef]
  112. Hammoud, D.A.; Lentz, M.R.; Lara, A.; Bohannon, J.K.; Feuerstein, I.; Huzella, L.; Jahrling, P.B.; Lackemeyer, M.; Laux, J.; Rojas, O.; et al. Aerosol exposure to intermediate size Nipah virus particles induces neurological disease in African green monkeys. PLoS Neglected Trop. Dis. 2018, 12, e0006978. [Google Scholar] [CrossRef]
  113. Cline, C.; Bell, T.M.; Facemire, P.; Zeng, X.; Briese, T.; Lipkin, W.I.; Shamblin, J.D.; Esham, H.L.; Donnelly, G.C.; Johnson, J.C.; et al. Detailed analysis of the pathologic hallmarks of Nipah virus (Malaysia) disease in the African green monkey infected by the intratracheal route. PLoS ONE 2022, 17, e0263834. [Google Scholar] [CrossRef] [PubMed]
  114. Johnston, S.C.; Briese, T.; Bell, T.M.; Pratt, W.D.; Shamblin, J.D.; Esham, H.L.; Donnelly, G.C.; Johnson, J.C.; Hensley, L.E.; Lipkin, W.I.; et al. Detailed Analysis of the African Green Monkey Model of Nipah Virus Disease. PLoS ONE 2015, 10, e0117817. [Google Scholar] [CrossRef] [PubMed]
  115. Cong, Y.; Lentz, M.R.; Lara, A.; Alexander, I.; Bartos, C.; Bohannon, J.K.; Hammoud, D.; Huzella, L.; Jahrling, P.B.; Janosko, K.; et al. Loss in lung volume and changes in the immune response demonstrate disease progression in African green monkeys infected by small-particle aerosol and intratracheal exposure to Nipah virus. PLoS Neglected Trop. Dis. 2017, 11, e0005532. [Google Scholar] [CrossRef]
  116. Mire, C.E.; Satterfield, B.A.; Geisbert, J.B.; Agans, K.N.; Borisevich, V.; Yan, L.; Chan, Y.P.; Cross, R.W.; Fenton, K.A.; Broder, C.C.; et al. Pathogenic Differences between Nipah Virus Bangladesh and Malaysia Strains in Primates: Implications for Antibody Therapy. Sci. Rep. 2016, 6, 30916. [Google Scholar] [CrossRef]
  117. Bossart, K.N.; Rockx, B.; Feldmann, F.; Brining, D.; Scott, D.; LaCasse, R.; Geisbert, J.B.; Feng, Y.-R.; Chan, Y.-P.; Hickey, A.C.; et al. A Hendra Virus G Glycoprotein Subunit Vaccine Protects African Green Monkeys from Nipah Virus Challenge. Sci. Transl. Med. 2012, 4, 146ra107. [Google Scholar] [CrossRef] [PubMed]
  118. Foster, S.L.; Woolsey, C.; Borisevich, V.; Agans, K.N.; Prasad, A.N.; Deer, D.J.; Geisbert, J.B.; Dobias, N.S.; Fenton, K.A.; Cross, R.W.; et al. A recombinant VSV-vectored vaccine rapidly protects nonhuman primates against lethal Nipah virus disease. Proc. Natl. Acad. Sci. USA 2022, 119, e2200065119. [Google Scholar] [CrossRef] [PubMed]
  119. de Wit, E.; Feldmann, F.; Cronin, J.; Goldin, K.; Mercado-Hernandez, R.; Williamson, B.N.; Meade-White, K.; Okumura, A.; Callison, J.; Weatherman, S.; et al. Distinct VSV-based Nipah virus vaccines expressing either glycoprotein G or fusion protein F provide homologous and heterologous protection in a nonhuman primate model. eBioMedicine 2023, 87, 104405. [Google Scholar] [CrossRef] [PubMed]
  120. Geisbert, J.B.; Borisevich, V.; Prasad, A.N.; Agans, K.N.; Foster, S.L.; Deer, D.J.; Cross, R.W.; Mire, C.E.; Geisbert, T.W.; Fenton, K.A. An Intranasal Exposure Model of Lethal Nipah Virus Infection in African Green Monkeys. J. Infect. Dis. 2020, 221, S414–S418. [Google Scholar] [CrossRef]
  121. Lo, M.K.; Feldmann, F.; Gary, J.M.; Jordan, R.; Bannister, R.; Cronin, J.; Patel, N.R.; Klena, J.D.; Nichol, S.T.; Cihlar, T.; et al. Remdesivir (GS-5734) protects African green monkeys from Nipah virus challenge. Sci. Transl. Med. 2019, 11, eaau9242. [Google Scholar] [CrossRef]
  122. Mire, C.E.; Geisbert, J.B.; Agans, K.N.; Versteeg, K.M.; Deer, D.J.; Satterfield, B.A.; Fenton, K.A.; Geisbert, T.W. Use of Single-Injection Recombinant Vesicular Stomatitis Virus Vaccine to Protect Nonhuman Primates Against Lethal Nipah Virus Disease. Emerg. Infect. Dis. 2019, 25, 1144–1152. [Google Scholar] [CrossRef]
  123. Van Doremalen, N.; Avanzato, V.A.; Goldin, K.; Feldmann, F.; Schulz, J.E.; Haddock, E.; Okumura, A.; Lovaglio, J.; Hanley, P.W.; Cordova, K.; et al. ChAdOx1 NiV vaccination protects against lethal Nipah Bangladesh virus infection in African green monkeys. NPJ Vaccines 2022, 7, 171. [Google Scholar] [CrossRef] [PubMed]
  124. Marianneau, P.; Guillaume, V.; Wong, K.T.; Badmanathan, M.; Looi, R.Y.; Murri, S.; Loth, P.; Tordo, N.; Wild, T.F.; Horvat, B.; et al. Experimental Infection of Squirrel Monkeys with Nipah Virus. Emerg. Infect. Dis. J. 2010, 16, 507. [Google Scholar] [CrossRef] [PubMed]
  125. Mungall, B.A.; Middleton, D.; Crameri, G.; Bingham, J.; Halpin, K.; Russell, G.; Green, D.; McEachern, J.; Pritchard, L.I.; Eaton, B.T.; et al. Feline model of acute nipah virus infection and protection with a soluble glycoprotein-based subunit vaccine. J. Virol. 2006, 80, 12293–12302. [Google Scholar] [CrossRef] [PubMed]
  126. Hooper, P.T.; Ketterer, P.J.; Hyatt, A.D.; Russell, G.M. Lesions of Experimental Equine Morbillivirus Pneumonia in Horses. Vet. Pathol. 1997, 34, 312–322. [Google Scholar] [CrossRef] [PubMed]
  127. Marsh, G.; Haining, J.; Hancock, T.; Robinson, R.; Foord, A.; Barr, J.; Riddell, S.; Heine, H.; White, J.; Crameri, G.; et al. Experimental Infection of Horses with Hendra Virus/Australia/Horse/2008/Redlands. Emerg. Infect. Dis. 2011, 17, 2232–2238. [Google Scholar] [CrossRef]
  128. Halpin, K.; Graham, K.; Durr, P.A. Sero-Monitoring of Horses Demonstrates the Equivac® HeV Hendra Virus Vaccine to Be Highly Effective in Inducing Neutralising Antibody Titres. Vaccines 2021, 9, 731. [Google Scholar] [CrossRef]
  129. Chua, K.B. Nipah virus outbreak in Malaysia. J. Clin. Virol. 2003, 26, 265–275. [Google Scholar] [CrossRef]
  130. Middleton, D.J.; Westbury, H.A.; Morrissy, C.J.; van der Heide, B.M.; Russell, G.M.; Braun, M.A.; Hyatt, A.D. Experimental Nipah Virus Infection in Pigs and Cats. J. Comp. Pathol. 2002, 126, 124–136. [Google Scholar] [CrossRef]
  131. Weingartl, H.M.; Berhane, Y.; Caswell, J.L.; Loosmore, S.; Audonnet, J.-C.; Roth, J.A.; Czub, M. Recombinant Nipah Virus Vaccines Protect Pigs against Challenge. J. Virol. 2006, 80, 7929–7938. [Google Scholar] [CrossRef]
  132. Pickering, B.S.; Hardham, J.M.; Smith, G.; Weingartl, E.T.; Dominowski, P.J.; Foss, D.L.; Mwangi, D.; Broder, C.C.; Roth, J.A.; Weingartl, H.M. Protection against henipaviruses in swine requires both, cell-mediated and humoral immune response. Vaccine 2016, 34, 4777–4786. [Google Scholar] [CrossRef]
  133. Pedrera, M.; Macchi, F.; McLean, R.K.; Franceschi, V.; Thakur, N.; Russo, L.; Medfai, L.; Todd, S.; Tchilian, E.Z.; Audonnet, J.-C.; et al. Bovine Herpesvirus-4-Vectored Delivery of Nipah Virus Glycoproteins Enhances T Cell Immunogenicity in Pigs. Vaccines 2020, 8, 115. [Google Scholar] [CrossRef] [PubMed]
  134. Berhane, Y.; Weingartl, H.M.; Lopez, J.; Neufeld, J.; Czub, S.; Embury-Hyatt, C.; Goolia, M.; Copps, J.; Czub, M. Bacterial Infections in Pigs Experimentally Infected with Nipah Virus. Transbound. Emerg. Dis. 2008, 55, 165–174. [Google Scholar] [CrossRef] [PubMed]
  135. Stachowiak, B.; Weingartl, H.M. Nipah Virus Infects Specific Subsets of Porcine Peripheral Blood Mononuclear Cells. PLoS ONE 2012, 7, e30855. [Google Scholar] [CrossRef] [PubMed]
  136. Weingartl, H.; Czub, S.; Copps, J.; Berhane, Y.; Middleton, D.; Marszal, P.; Gren, J.; Smith, G.; Ganske, S.; Manning, L.; et al. Invasion of the Central Nervous System in a Porcine Host by Nipah Virus. J. Virol. 2005, 79, 7528–7534. [Google Scholar] [CrossRef] [PubMed]
  137. Kasloff, S.B.; Leung, A.; Pickering, B.S.; Smith, G.; Moffat, E.; Collignon, B.; Embury-Hyatt, C.; Kobasa, D.; Weingartl, H.M. Pathogenicity of Nipah henipavirus Bangladesh in a swine host. Sci. Rep. 2019, 9, 5230. [Google Scholar] [CrossRef] [PubMed]
  138. Kirkland, P.D.; Gabor, M.; Poe, I.; Neale, K.; Chaffey, K.; Finlaison, D.S.; Gu, X.; Hick, P.M.; Read, A.J.; Wright, T.; et al. Hendra Virus Infection in Dog, Australia, 2013. Emerg. Infect. Dis. 2015, 21, 2182–2185. [Google Scholar] [CrossRef]
  139. Middleton, D.; Riddell, S.; Klein, R.; Arkinstall, R.; Haining, J.; Frazer, L.; Mottley, C.; Evans, R.; Johnson, D.; Pallister, J. Experimental Hendra virus infection of dogs: Virus replication, shedding and potential for transmission. Aust. Vet. J. 2017, 95, 10–18. [Google Scholar] [CrossRef]
  140. Field, H.; Young, P.; Yob, J.M.; Mills, J.; Hall, L.; Mackenzie, J. The natural history of Hendra and Nipah viruses. Microbes Infect. 2001, 3, 307–314. [Google Scholar] [CrossRef]
  141. Mills, J.N.; Alim, A.N.M.; Bunning, M.L.; Lee, O.B.; Wagoner, K.D.; Amman, B.R.; Stockton, P.C.; Ksiazek, T.G. Nipah Virus Infection in Dogs, Malaysia, 1999. Emerg. Infect. Dis. 2009, 15, 950–952. [Google Scholar] [CrossRef]
  142. Hooper, P.; Zaki, S.; Daniels, P.; Middleton, D. Comparative pathology of the diseases caused by Hendra and Nipah viruses. Microbes Infect. 2001, 3, 315–322. [Google Scholar] [CrossRef]
  143. Westbury, H.; Hooper, P.; Brouwer, S.; Selleck, P. Susceptibility of cats to equine morbillivirus. Aust. Vet. J. 1996, 74, 132–134. [Google Scholar] [CrossRef] [PubMed]
  144. Williamson, M.; Hooper, P.; Selleck, P.; Gleeson, L.; Daniels, P.; Westbury, H.; Murray, P. Transmission studies of Hendra virus (equine morbilli-virus) in fruit bats, horses and cats. Aust. Vet. J. 1998, 76, 813–818. [Google Scholar] [CrossRef] [PubMed]
  145. Mungall, B.A.; Middleton, D.; Crameri, G.; Halpin, K.; Bingham, J.; Eaton, B.T.; Broder, C.C. Vertical Transmission and Fetal Replication of Nipah Virus in an Experimentally Infected Cat. J. Infect. Dis. 2007, 196, 812–816. [Google Scholar] [CrossRef] [PubMed]
Figure 1. General suitability of AGMs, Ferrets, SGH, and Mouse models by study type.
Figure 1. General suitability of AGMs, Ferrets, SGH, and Mouse models by study type.
Viruses 15 01980 g001
Figure 2. Representative gross pathology of lung, spleen, and urinary bladder from ferrets challenged with rNiV-M. Adapted from [101]. Representative gross pathology of the lung (a), spleen (b) and urinary bladders (c) taken from ferrets infected with NiV-M (ac). Inserts show magnified regions of each specimen. Findings included multifocal to coalescing hemorrhage and necrosis of all lung lobes (a), splenomegaly and multifocal necrosis in the spleen (b), and large hemorrhagic lesions in the urinary bladder (c).
Figure 2. Representative gross pathology of lung, spleen, and urinary bladder from ferrets challenged with rNiV-M. Adapted from [101]. Representative gross pathology of the lung (a), spleen (b) and urinary bladders (c) taken from ferrets infected with NiV-M (ac). Inserts show magnified regions of each specimen. Findings included multifocal to coalescing hemorrhage and necrosis of all lung lobes (a), splenomegaly and multifocal necrosis in the spleen (b), and large hemorrhagic lesions in the urinary bladder (c).
Viruses 15 01980 g002
Figure 3. Representative pathogenesis of experimental NiV infection in the African Green Monkey, adapted from [103]. Panels (A,B,D,E); AGM euthanized nine days post-challenge with 1.3 × 106 PFU of NiV i.t./o.n. Panel (C); AGM succumbed to infection 11 days post challenge with 8.1 × 104 PFU of NiV i.t./o.n. (A) Serosanguineous, frothy oronasal exudate. (B) Wet, heavy, congested, and hemorrhagic lungs. (C) Lung, right diaphragmatic lobe by immunohistochemical (IHC) stain depicting fibrin deposition in and around alveolar spaces (arrows); 40× magnification. (D) Lung, right diaphragmatic lobe by PTAH connective tissue stain showing an abundance of polymerized fibrin in and around alveolar spaces (arrows); 100× magnification. (E) Localization of NiV antigen by IHC stain within a lung blood vessel with endothelial syncytia (arrow) and immunopositive cells (brown) abundant in alveolar septae; 400× magnification.
Figure 3. Representative pathogenesis of experimental NiV infection in the African Green Monkey, adapted from [103]. Panels (A,B,D,E); AGM euthanized nine days post-challenge with 1.3 × 106 PFU of NiV i.t./o.n. Panel (C); AGM succumbed to infection 11 days post challenge with 8.1 × 104 PFU of NiV i.t./o.n. (A) Serosanguineous, frothy oronasal exudate. (B) Wet, heavy, congested, and hemorrhagic lungs. (C) Lung, right diaphragmatic lobe by immunohistochemical (IHC) stain depicting fibrin deposition in and around alveolar spaces (arrows); 40× magnification. (D) Lung, right diaphragmatic lobe by PTAH connective tissue stain showing an abundance of polymerized fibrin in and around alveolar spaces (arrows); 100× magnification. (E) Localization of NiV antigen by IHC stain within a lung blood vessel with endothelial syncytia (arrow) and immunopositive cells (brown) abundant in alveolar septae; 400× magnification.
Viruses 15 01980 g003
Table 1. Experimental findings in immunocompetent mouse models of HNV Disease.
Table 1. Experimental findings in immunocompetent mouse models of HNV Disease.
Species (Age)Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
C57BL/6
(aged)
HeV (Redlands/Horse/2008)5 × 104
TCID50
i.n.100%
(5/5)
Depression, ataxia, hypersensitivity, tremors[56]
BALB/c
(aged)
60%
(3/5)
C57BL/6
(juvenile)
s.c.NoneNone
BALB/c
(juvenile)
C57BL/6
(aged)
NiV-B (human/2004/Rajbari, R1)5 × 104
TCID50
i.n.NoneNone[57]
C57BL/6
(juvenile)
BALB/c
(aged)
BALB/c
(juvenile)
C57BL/6
(aged)
NiV-M (Malaysia/human/99)5 × 104
TCID50
i.n.NoneNone
C57BL/6
(juvenile)
BALB/c
(aged)
BALB/c
(juvenile)
UnknownHeV
(prototype)
5 × 103
TCID50
s.c.NoneNone[55]
Swiss
(juvenile)
HeV
(prototype)
6 × 105
PFU
i.n.NoneNone[43]
1 × 107
PFU
i.p.
C57BL/6NiV-M
(UMMC1)
1 × 106
PFU
i.p.NoneNone[58]
1 × 105
PFU
i.c.100%
(5/5)
Weight loss, aggressiveness, prostration, paralysis[58]
Suckling Mice
(2–3-day-old)
NiV-M1 × 100
TCID50
i.c.15%
(9–13) a
Clinical signs including weight loss b[59]
1 × 101
TCID50
66%
(9–13) a
1 × 102
TCID50
100%
(9–13) a
1 × 103
TCID50
100%
(9–13) a
1 × 104
TCID50
100%
(9–13) a
1 × 105
TCID50
100%
(9–13) a
1 × 106
TCID50
100%
(9–13) a
BALB/c
(13-week-old)
NiV-B (human/2004/Rajbari, R1)5 × 104
TCID50
i.n.NoneNone[60]
rNiV-WTNone
rNiV-FHeVNone
rNiV-GHeVNone
rNiV-MHeVNone
HeV (Redlands/Horse/2008)5 × 104
TCID50
i.n.NoneNone
rHeV-WT40%
(2/5)
Depression (2/5), increased respiration (2/5), hypersensitivity to stimuli (2/5), piloerection (1/5), hunched posture (1/5)
rHeV-FNiVNoneNone
rHeV-GNiVNone
rHeV-MNiVNone
a A range of 9–13 animals per group. b Comprehensive numerical clinical scores were described in this study, with individual signs not specified for subjects.
Table 3. Experimental findings in Syrian golden hamster models of HNV Disease.
Table 3. Experimental findings in Syrian golden hamster models of HNV Disease.
AgeVirus
(Isolate)
DoseRoute% LethalityClinical DiseaseReference
8–10-week-oldNiV-M1 × 104
TCID50
i.p.80% (4/5)Weight loss[68]
HeV
(prototype)
1 × 104
TCID50
i.p.100% (5/5)Head tilt, weight loss, hypothermia,
7-week-oldHeV
(prototype)
1 × 105
PFU
i.p.100% (6/6)Paralysis, trembling limbs, breathing difficulties, serosanguinous nasal discharge[65]
1 × 104
PFU
100% (6/6)
1 × 103
PFU
100%
(6/6)
1 × 102
PFU
83%
(5/6)
1 × 101
PFU
67%
(4/6)
1 × 100
PFU
NoneNone
11-week-old1.2 × 103
PFU
80%
(4/5)
Paralysis, trembling limbs, breathing difficulties, serosanguinous nasal discharge
7-week-old1.2 × 103
PFU
100%
(5/5)
11-week-old1.2 × 104
PFU
100%
(5/5)
7-week-old1.2 × 104
PFU
100%
(5/5)
2-month-oldNiV-M6 × 105
PFU
i.n.100%
(2/2)
Ataxia, imbalance[43]
1 × 107
PFU
i.p.100%
(2/2)
7–14-week-old1 × 104
PFU
i.p.100%
(6/6)
Tremors, limb paralysis
1 × 103
PFU
83%
(5/6)
1 × 102
PFU
67%
(4/6)
1 × 101
PFU
None (0/6)
1 × 100
PFU
None
(0/6)
7–14-week-old1 × 106
PFU
i.n.83%
(5/6)
Imbalance, limb paralysis, lethargy, muscle twitching, breathing difficulties
1 × 105
PFU
83%
(5/6)
1 × 104
PFU
50%
(3/6)
1 × 103
PFU
50% (3/6)
1 × 102
PFU
None (0/6)
1 × 101
PFU
None
(0/6)
~10-week-old aNiV-M6.84 × 104
TCID50
i.p.100%
(10/10)
Severe clinical signs of disease, not described[71]
~10-week-oldNiV-M1 × 105
TCID50
i.p.100%
(3/3)
Neurological signs, respiratory distress[75]
8-week-oldNiV
(unknown)
2 × 103
TCID50
i.p.90%
(9/10)
Not described[76]
UnknownNiV-M b1 × 103
PFU
i.p.100%
(unknown)
Not described[74]
6–8-week-oldNiV-M1 × 107
TCID50
i.n.100%
(14/14) c
Severe signs of disease, respiratory distress[77]
1 × 105
TCID50
67%
(4/6)
Weight loss
1 × 103
TCID50
33%
(2/6)
Weight loss, neurological signs
6-week-oldNiV-M1 × 105
TCID50
i.n.100%
(5/5)
Weight loss, severe acute respiratory distress[78]
1 × 102
TCID50
100%
(5/5)
Weight loss, neurological signs (complete paralysis, seizure, muscle fasciculations)
HeV
(prototype)
1 × 105
TCID50
i.n.100%
(5/5)
Weight loss, severe acute respiratory distress (labored breathing, serosanguineous oronasal exudates)
1 × 102
TCID50
100%
(5/5)
Weight loss, respiratory disease, neurological signs (complete paralysis, seizure, muscle fasciculations)
5–6-week-oldNiV-M1 × 105
TCID50
i.p.100%
(5/5)
Respiratory distress (labored breathing, hunched posture), neurological signs (one animal)[79]
1 × 104
TCID50
100%
(5/5)
Respiratory distress (labored breathing, hunched posture)
1 × 103
TCID50
100%
(5/5)
Neurological signs (imbalance, partial paralysis, seizure)
1 × 102
TCID50
60%
(3/5)
Neurological signs (imbalance, partial paralysis, seizure)
1 × 101
TCID50
None
(0/5)
Neurological signs
1 × 100
TCID50
None
(0/5)
5–6-week-oldNiV-B1 × 105
TCID50
i.p.80%
(4/5)
Respiratory or neurological, both respiratory and neurological (one animal)
1 × 104
TCID50
80%
(4/5)
Respiratory or neurological, both respiratory and neurological (one animal)
1 × 103
TCID50
60%
(5/5)
Neurological signs
1 × 102
TCID50
None
(0/5)
1 × 101
TCID50
40%
(2/5)
1 × 100
TCID50
None
(0/5)
5–6-week-oldNiV-M1 × 105
TCID50
i.n.100%
(5/5)
Not described
5–6-week-oldNiV-B1 × 105
TCID50
100%
(5/5)
4–5-week-oldNiV-M1 × 105
PFU
aerosol100%
(5/5)
Weight loss, scruffy coat (3/5), Respiratory and neurological signs (2/5)[80]
2 × 104
TCID50
60%
(3/5)
Neurological signs (3/5), Respiratory and neurological signs (2/5)
8 × 102
TCID50
80%
(4/5)
Neurological signs (3/5), Respiratory and neurological signs (2/5)
2.5 × 102
TCID50
80%
(4/5)
Neurological signs (3/5), Respiratory and neurological signs (2/5)
4–5-week-oldrNiV-FlucNP2 × 105
PFU
aerosol100%
(5/5)
Weight loss, scruffy coat, labored breathing (4/5), Respiratory and neurological symptoms (1/5)
8.5 × 104
TCID50
100%
(5/5)
Respiratory and neurological signs (4/5), weight loss
9.5 × 103
TCID50
100%
(5/5)
Respiratory and neurological signs (4/5), weight loss, weight loss (1/5), respiratory only (1/5)
2.5 × 102
TCID50
60%
(3/5)
Respiratory and neurological signs (2/5), weight loss, respiratory only (1/5)
6.6 × 101
TCID50
60%
(3/5)
Respiratory and neurological signs (2/5), weight loss, respiratory only (1/5)
UnknownNiV-M (UMMC1) d1 × 104
PFU
i.p.100%
(6/6)
Weight loss, dyspnea, tremor, paralysis[81]
UnknownHeV
(prototype) d
1 × 104
PFU
100%
(6/6)
Weight loss, dyspnea, tremor, paralysis
8-week-oldrNiV(W-) e1 × 105
PFU
i.p.83%
(5/6)
Neurological signs[82]
1 × 104
PFU
83%
(5/6)
1 × 103
PFU
66%
(4/6)
1 × 101
PFU
50%
(3/6)
1 × 100
PFU
16%
(1/6)
6–11-week-oldNiV-M1 × 105
TCID50
i.n.100%
(10/10)
Respiratory signs (dyspnea, wasp waist), ataxia (2/10)[83]
1 × 104
TCID50
100%
(4/4)
Dyspnea (1/4), ataxia (2/4)
NiV-M1 × 103
TCID50
i.p.100%
(8/8)
Neurological signs, ataxia (2/8), respiratory signs (2/8).
1 × 102
TCID50
75%
(3/4)
Neurological signs
8-week-oldNiV-M (UMMC1)1 × 102
PFU
i.p.100%
Unknown f
Not described[84]
1 × 103
PFU
100%
Unknown f
NiVΔC1 g1 × 102
PFU
i.p.~30–40%
Unknown f
Not described
1 × 103
PFU
~80–90%
Unknown f
NiVΔC2 g1 × 102
PFU
i.p.~60%
Unknown f
Not described
1 × 103
PFU
~70%
Unknown f
5–7-
week-old
NiV-M1 × 106
TCID50
i.n.76%
(~35/46) h
Clinical signs including weight loss i[59]
1 × 105
TCID50
50%
Unknown
1 × 103
TCID50
12%
Unknown
1 × 104
TCID50
i.p.100%
Unknown
a Age at the time of challenge. b Received a nonspecific vaccinia vaccine. c Animals from two control groups combined. d Received a nonspecific AAV-GFP vaccine. e Recombinant NiV with W accessory protein knockout. f Study specifies 5 or 6 animals per group. g Recombinant NiV mutants lacking C protein expression. h Includes historical data. i Comprehensive numerical clinical scores were described in this study, with individual signs not specified for subjects.
Table 4. Experimental findings in guinea pig models of HNV Disease.
Table 4. Experimental findings in guinea pig models of HNV Disease.
Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
HeV
(prototype)
5 × 103
TCID50
s.c.80%
(4/5)
Respiratory distress, inappetence[55]
5 × 104
TCID50
s.c.75%
(9/12)
Weakness, lethargy, head tilt, depression, abortion (1/12) [88]
3.4 × 106
PFU
i.n.33%
(2/6)
Depression, inappetence, inactivity, weight loss, nasal hemorrhage (1/6) [89]
NiV-M6 × 105
PFU
i.n.NoneNone[43]
1 × 107
PFU
i.p.NoneTransient fever and weight loss
5 × 104
TCID50
i.p.38%
(3/8)
Ruffled fur, ataxia (1/8) [90]
6 × 104
PFU
i.p.92%
(24/26)
Severe clinical disease (features not described)[91]
Table 5. Experimental findings in ferret models of HNV Disease.
Table 5. Experimental findings in ferret models of HNV Disease.
Virus (Isolate)DoseRouteTreatment% LethalityClinical DiseaseReference
NiV-M
(EUKK 19817)
5 × 104
TCID50
o.n.N/A100%
(2/2)
Severe depression, orthopnea, expiratory dyspnea, serous nasal discharge, cough, subcutaneous edema of the head [92]
5 × 103
TCID50
100% (2/2)Severe depression, orthopnea, cutaneous ecchymoses, vomiting, hypothermia
5 × 102
TCID50
50%
(1/2)
Obtundation, tremor, and hind limb paralysis
5 × 101
TCID50
None
(0/2)
None
5 × 103
TCID50
PBS
(IV)
100%
(2/2)
Weight loss, fever, depression, reduced play activity, subcutaneous edema of the head, cutaneous hemorrhages, inappetence (1/2), diarrhea (1/2), blood in the mouth (1/2)
mAb a (IV)66%
(2/3)
Fever, depression, weight loss, reduced play activity, inappetence, diarrhea, crusting nasal discharge, hind limb paralysis (1/2), generalized tremor (1/3)
HeV
(Redlands 2008)
5 × 104
TCID50
o.n.N/A100%
(2/2)
Fever, depression, decreased grooming, generalized tremors[93]
5 × 103
TCID50
100%
(2/2)
5 × 102
TCID50
100%
(2/2)
5 × 101
TCID50
100%
(2/2)
5 × 103
TCID50
HeVsG
100 μg b
None
(0/2)
None
HeVsG
20 μg b
None
(0/2)
HeVsG
4 μg b
50%
(1/2)
Reduced play activity, weakness, tremor
CpG
Adjuvant c
100%
(2/2)
Reduced play activity, fever, hind limb paralysis
NiV-B (human/2004/Rajbari, R1)5 × 103
TCID50
unknownHeVsG
100 μg b,d
None
(1/1)
None[94]
HeVsG
20 μg b,d
None
(2/2)
HeVsG
4 μg b,d
None
(1/1)
HeVsG
100 μg b,e
None
(1/1)
HeVsG
20 μg b,e
None
(1/1)
HeVsG
4 μg b,e
None
(2/2)
CpG
Adjuvant c
100%
(2/2)
Fever
CpG
Adjuvant c
100%
(2/2)
Fever, reduced playfulness
NiV-M (patient isolate 1998 Malaysia outbreak)1 × 105
TCID50
i.n.N/A100%
(4/4)
Labored breathing, fever, mild paralysis, generalized tremors, subcutaneous edema of head and neck, lack of grooming, hunched posture, ataxia, continuous licking, imbalance, myoclonus, head tilt, hind-limb paralysis, seizures[95]
1 × 103
TCID50
100%
(4/4)
1 × 102
TCID50
100%
(4/4)
1 × 101
TCID50
25%
(1/4)
NiV-B (2004 patient isolate)1 × 105
TCID50
i.n.N/A100%
(4/4)
1 × 103
TCID50
100%
(4/4)
1 × 102
TCID50
75%
(3/4)
1 × 101
TCID50
25%
(1/4)
HeV
(prototype)
1 × 105
TCID50
i.n.N/A100%
(4/4)
1 × 103
TCID50
100%
(4/4)
1 × 102
TCID50
100%
(4/4)
1 × 101
TCID50
75%
(3/4)
NiV-M (isolate 1999011924)5 × 103
PFU
i.n.N/A100%
(1/1)
Fever, facial edema, nasal and ocular discharge, sneezing, depression, loss of appetite, labored breathing, head and neck myoclonus[96]
mAb fNone
(0/3)
Fever (1/3)
mAb gNone
(0/3)
Fever (1/3), minor facial and ear twitching (1/3)
HeV
(prototype)
5 × 103
PFU
i.n.N/A100%
(1/1)
Fever, nasal and ocular discharge, sneezing, loss of appetite, depression, facial edema, labored breathing
mAb gNone
(0/3)
Mild fever
NiV-B (human/2004/Rajbari, R1)5 × 103
PFU
o.n.N/A100%
(4/4)
Fever, ataxia, agitation, facial edema, disorientation, tachypnea/dyspnea[97]
contact h100%
(4/4)
NiV-M (Malaysia/Human/99)5 × 103
PFU
o.n.N/A100%
(4/4)
contact h100%
(4/4)
NiV-B (human/2004/Rajbari, R1)5 × 103
TCID50
o.n.N/A100%
(8/8)
Hunched posture, agitation, sneezing, weight loss, licking, dehydration, vomiting, myoclonus of forelimbs, hindlimbs, flank, or tail, nasal discharge, facial edema, oral mucosa hemorrhage, ataxia, ventral neck edema, paralysis[98]
NiV-M (Malaysia/Human/99)5 × 103
TCID50
o.n.N/A100%
(6/6) i
Severe ataxia, facial and hind limb tremors, head tilt, torticollis, sneezing, nasal discharge, facial edema, hemorrhage of rectal mucosa, dyspnea, hemorrhage from nose and mouth, spastic paralysis of right forelimb, myoclonus of right trunk, extensive cutaneous petechial hemorrhage, hind limb paralysis, hunched posture, muscular fasciculation over flank, weight loss, recumbency
NiV-M
(EUKK 19817)
5 × 103
TCID50
o.n.Vehicle
control j
100%
(2/2)
Fever, depression, hind-limb paralysis, myoclonus, urinary incontinence, subcutaneous edema of neck and throat, cutaneous petechial hemorrhage, serosanguinous oral secretions[99]
Antiviral k100%
(3/3)
Antiviral l100%
(3/3)
rNiVM-wt5 × 103
PFU
i.n.N/A100%
(5/5)
Fever, respiratory distress, lethargy, inappetence, depression, ocular, oral, and nasal discharge, ataxia, severe hypothermia, myoclonus, weight loss, hindlimb paresis, rales, hyperglycemia[100]
rNiVM-PY166E5 × 103
PFU
i.n.N/A100%
(5/5)
Fever, depression, lethargy, inappetence, oral and nasal discharge, seizure, weight loss, aggressiveness, hyperglycemia, ataxia, hindlimb paresis
rNiVM-PD116–1355 × 103
PFU
i.n.N/A100%
(5/5)
Fever, depression, lethargy, inappetence, sneezing, myoclonus, facial tremor, nasal discharge, weight loss, hindlimb paresis, quadriparesis, hypothermia, seizure
rNiVM-wt5 × 103
PFU
i.n.N/A100%
(5/5)
Depression, lethargy, inappetence, ocular and nasal discharge, myoclonus, ataxia, weight loss, sneezing, obtundation, nasal and oral frothing, facial edema, tremor[101]
rNiVM-WKO5 × 103
PFU
i.n.N/A100%
(5/5)
Depression, lethargy, dehydration, sneezing, rales, nasal and oral frothing, severe ataxia, seizure, hypothermia, inappetence, hypersalivation, tremors, ocular and nasal discharge, obtundation
rNiVM-VKO5 × 103
PFU
i.n.N/ANone
(0/5)
Lethargy, nasal discharge, depression, tremors, mild ataxia
rNiVM-wt5 × 103
PFU
i.n.N/A100%
(5/5)
Depression, lethargy, inappetence, ocular and nasal discharge, ataxia, hindlimb myoclonus, tremors, nasal and oral frothing, facial edema, rales, hypothermia, sneezing, weight loss, obtundation[102]
rNiVM-CKO5 × 103
PFU
i.n.N/A100%
(5/5)
Depression, lethargy, sneezing, ataxia, hypothermia, inappetence, myoclonus, rales, ocular and nasal discharge, weight loss
rNiVM- CKOWKO5 × 103
PFU
i.n.N/A60%
(3/5)
Depression, lethargy, sneezing, nasal discharge, rales, hindlimb myoclonus and paralysis, severe tremors, seizures, weight loss, facial myoclonus, dilated pupils, facial edema, obtundation, severe hypothermia, visual deficit, aggressiveness
a mAb m102.4, 50 mg per animal, given 24 h prior to infection with NiV-M. b HeVsG is an adjuvanted, soluble HeV (G) glycoprotein vaccine containing a dose indicated in micrograms with a CpG adjuvant. c Ferrets received 150 μg CPG ODN 2007 dinucleotide adjuvant. d Vaccinated 20 days prior to challenge. e Vaccinated 434 days prior to challenge. f Received mAb h5B3.1, 20 mg/kg intraperitoneally on days 1 and 3 post-challenge. g Received mAb h5B3.1, 20 mg/kg intraperitoneally on days 3 and 5 post-challenge. h Ferrets were infected through cohabitation with directly exposed ferrets and inoculation with oronasal secretions of directly exposed ferrets at two time points. i Excluded one animal from the group due to euthanasia for reasons unrelated to NiV-M infection. j 20% sucrose. k Treated with chloroquine 24 h before challenge. l Treated with chloroquine 10 h after challenge.
Table 6. Experimental findings in AGM models of HNV Disease.
Table 6. Experimental findings in AGM models of HNV Disease.
Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
HeV
(prototype)
4 × 105
TCID50
i.t.100%
(4/4)
Fever, severe respiratory distress (increased breathing rate, open mouth breathing), interstitial pneumonia (radiology)[108]
5 × 105
PFU
100%
(4/4)
Fever, depression, lethargy, inappetence, labored breathing, hind limb paralysis (1/4), dehydration[109]
4 × 105
TCID50
100%
(3/3)
Nasal discharge, labored breathing[104]
HeV
(prototype)
4 × 105
TCID50
i.t.100%
(9/9) a
Nasal discharge, labored breathing, seizure (1/9), muscle fasciculations (5/9)
NiV-M1.3 × 106
PFU
i.t., oral100%
(1/1)
Depression, lethargy, fever, inappetence, severe dyspnea, labored breathing[103]
7.0 × 103
PFU
None
(0/1)
Depression, lethargy, fever, inappetence, severe dyspnea, labored breathing, nausea, lymphadenopathy, ecchymotic rash at the venipuncture site, muscle twitches, behavioral changes
8.1 × 104
PFU
100%
(1/1)
Depression, lethargy, fever, inappetence, severe dyspnea, labored breathing, nausea, pleural effusions (X-ray)
6.5 × 104
PFU
i.t.100%
(1/1)
Depression, lethargy, fever, inappetence, labored breathing, pleural effusions (X-ray)
5.9 × 104
PFU
100%
(1/1)
Depression, lethargy, inappetence, labored breathing, X-ray showed pleural effusions
2.3 × 104
PFU
100%
(1/1)
Depression, lethargy, fever, inappetence, labored breathing, loss of balance, pleural effusions (X-ray)
7.0 × 103
PFU
100%
(1/1)
Depression, lethargy, inappetence, labored breathing, pleural effusions (X-ray)
2.5 × 103
PFU
100%
(1/1)
Depression, lethargy, inappetence, labored breathing, pleural effusions (X-ray)
NiV-B (isolate 200401066)8.81 × 102
PFU
aerosol100%
(1/1)
Anorexia[110]
1.33 × 103
PFU
100%
(1/1)
Depression, lethargy, recumbency, anorexia, severe dyspnea, hypothermia
9.95 × 103
PFU
100%
(1/1)
Depression, lethargy, anorexia, fever, severe dyspnea, pulmonary consolidation (X-ray)
1.31 × 104
PFU
100%
(1/1)
Anorexia, mild dyspnea
NiV-M~64 × 101
PFU b
aerosol100%
(3/3)
Weight loss, cough, lethargy, inappetence, pulmonary consolidation (CT scan), neutrophilia (2/6), anemia (2/6), monocytosis (2/6), lymphopenia (3/6), hypoalbuminemia, increased globulin, increased respiratory rate only on day of euthanasia[111,112]
~7 × 102
PFU b
66%
(2/3)
NiV-M2.41 × 105
PFU
aerosol100%
(4/4)
Decreased activity, labored breathing, increased respiratory rates, unresponsiveness, fever, continuous head twitch (1/4)[113]
NiV-M2.5 × 104
PFU
i.t.50%
(2/4)
Decreased responsiveness, tachypnea, fever, tachycardia, tachypnea (3/4), hypotension (2/4), hypothermia (2/4), tremors (1/4), bloody oronasal exudate (1/4), lymphadenopathy, weight loss (1/4)[114]
NiV-M1.3 × 104
PFU
i.t.100%
(3/3)
Lethargy, cough, breathing difficulty, inappetence[115]
4.03 × 104
PFU
aerosol100%
(3/3)
NiV-M (isolate 199902916)5 × 105
PFU
i.t./i.n.50%
(2/4)
Depression, lethargy (3/4), fever, inappetence, dyspnea, lymphopenia, thrombocytopenia, hypoalbuminemia, nasal exudate (3/4), tremors (2/4)[116]
NiV-B (isolate 200401066)5 × 105
PFU
i.t./i.n.100%
(4/4)
Fever (2/4), depression, inappetence, severe dyspnea, lymphopenia, thrombocytopenia, hypoalbuminemia, labored breathing (3/4), nasal exudate (2/4), with epistaxis (1/4), tremors (1/4)
100%
(2/2) c
Fever, depression, lethargy, inappetence, severe dyspnea, labored breathing, nasal exudate with epistaxis (1/2)
NiV-B (isolate 200401066)5 × 105
PFU
i.t./i.n.100%
(6/6)
Aggressiveness, pulmonary edema (3/6),
abdominal (3/6) or open mouth breathing (2/6), lethargy (4/6), depression (1/6), dyspnea (1/6), anorexia (3/6), recumbency (3/6), bradypnea (2/6), nasal exudate (1/6), hypothermia (1/6)
[105]
NiV-M (isolate 199902916)5 × 105
PFU
i.t./i.n.50%
(1/2)
Depression, abdominal breathing, anorexia (1/2), lethargy (1/2), recumbency (1/2), nasal exudates, pulmonary consolidation (x-ray, 1/2), tremors (1/2)
HeV-prototype (isolate 9409-30-1800)5 × 105
PFU
i.t./i.n.80%
(4/5)
Anorexia (2/5), abdominal breathing (2/5), severe bradypnea (1/5), lethargy (1/5), dyspnea (1/5), mild tachypnea (1/5), pulmonary edema (1/5), depression (1/5)
NiV-M1 × 105
TCID50
i.t.100%
(1/1) d
Anorexia, depression, decreased activity, hunched posture, increase in respiratory rate, acute respiratory distress, decrease in platelet count[117]
NiV-B (isolate 200401066)5 × 105
PFU
i.t./i.n.100%
(6/6) e
Increased respiration rates, tachypnea, dyspnea, depression[118]
NiV (unknown isolate/strain)1 × 108
TCID50
i.n./oralNone
(0/1)
Weight loss, severe illness[76]
1 × 106
TCID50
None
(0/1)
1 × 108
TCID50
i.p.100%
(1/1)
Weight loss, severe depression, anorexia, decreased activity
1 × 106
TCID50
100%
(1/1)
NiV-M (isolate 199901924)1 × 105
TCID50
i.t./i.n.100%
(2/2) e
Clinical signs not described[119]
NiV-B (isolate 200401066)1 × 105
TCID50
i.t./i.n.100%
(2/2) e
Clinical signs not described
a All animals treated with ribavirin pre- or post-infection. b Approximate average exposure dose across animals in each group. c Infused with IV saline as the control for mAb treatment. d Received adjuvant as the control for vaccine study. e Received nonspecific VSV vaccine.
Table 7. Experimental findings in other nonhuman primate models of henipavirus disease.
Table 7. Experimental findings in other nonhuman primate models of henipavirus disease.
SpeciesVirus (Isolate)DoseRoute% LethalityClinical DiseaseReference
Cynomolgus MacaqueNiV-B
(isolate 200401066)
5 × 105
PFU
i.t./i.n.None
(0/3)
Pulmonary edema, abdominal breathing, fever (2/3), lymphadenitis (1/3)[105]
NiV-M
(isolate 199902916)
None
(0/3)
None
HeV
(prototype)
None
(0/2)
Agitation (1/2), increased aggressiveness (1/2), abdominal breathing (1/2)
Common
Marmoset
NiV-B
(isolate 200401066)
6.33 × 104
PFU
i.t./i.n.100%
(4/4)
Hyperventilation (4/4), anorexia (3/4), lethargy (2/4), open mouth breathing (1/4), hunched posture (1/4), hindlimb tremors (1/4)[106]
Squirrel
Monkey
NiV-M (UMMC1)1 × 103
PFU
i.v.100%
(2/2)
Uncoordinated motor movements, prostration, coma[124]
1 × 107
PFU
50%
(1/2)
Uncoordinated motor movements, prostration, coma, anorexia, depression
1 × 103
PFU
i.n.None
(0/0)
Anorexia, seizure
1 × 107
PFU
None
(0/0)
Anorexia, seizure, ocular edema
Table 8. Experimental findings in equine models of henipavirus disease.
Table 8. Experimental findings in equine models of henipavirus disease.
Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
HeV
(prototype)
N/A ai.v./i.n.100%
(2/2)
Fever, weakness, labored breathing, agitation[126]
2 × 107
TCID50
i.v./i.n. aerosol100%
(2/2)
Mild temperature increase, labored breathing, depression
HeV (Australia/Horse/2008/Redlands)2 × 106
TCID50
o.n.100%
(3/3)
Increased heart rate, depression, anorexia, serous nasal discharge, dyspnea, agitation, irritability, panting respiration[127]
a Animals challenged with a homogenate of blood, spleen, and lung from horses infected naturally in an HeV outbreak.
Table 9. Experimental findings in swine models of henipavirus disease.
Table 9. Experimental findings in swine models of henipavirus disease.
Species (Age)Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
LandraceHeV
(prototype)
6.6 × 107
PFU
o.n.100%
(2/2)
Fever, inappetence, severe depression, respiratory distress, recumbency[89]
Gottingen MinipigsHeV
(prototype)
2.0 × 107
PFU
i.n.N/A a
(0/5)
Fever (1/5), mild depression, cough, respiratory distress, transient neurological signs
Landrace
(7–8-week-old)
NiV-M b2.5 × 105
PFU
i.n.N/A a
(0/4)
Fever, lethargy, labored breathing (1/4), cough (1/4), unwilling to stand (1/4)[131]
Landrace
(9-week-old) c
NiV-M b5 × 105
PFU
i.n.None
(0/2)
Mild temperature increase[132]
Landrace
(9-week-old) c
HeV
(prototype)
5 × 105
PFU
i.n.None
(0/1)
Mild temperature increase
Landrace
(4-week-old)
NiV-M
(human isolate)
2.5 × 105
PFU
o.n.16%
(1/6)
Transient temperature increase, exudative epidermitis (1/6), mild to severe depression (3/6), shivering, unwilling to stand (3/6), inappetence, shiver, hunched posture (2/4), cough (2/6), increased respiratory rate [134]
Landrace
(4–6-week-old)
NiV-M b 2.5 × 105
PFU
i.n.31%
(5/16)
Not described[135]
Conventional
(6-week-old)
NiV-M5 × 103
TCID50
s.c.66%
(2/3)
Loss of consciousness, lateral recumbency, ataxia, unwillingness to stand, muscle fasciculations, nasal discharge, shivering, persistent cough upon stimulation, mild increase in temperature[130]
oralNone
(0/3)
No signs attributed to NiV infection
Landrace
(4-week-old)
NiV-M
(human isolate)
2.5 × 105
PFU
o.n. + ocular18%
(2/11) d
Wide stance, difficulty standing, unsteady balance, restless, lethargy, unwilling to walk, sawhorse stance, depression, severe shivering, seizure[136]
Landrace
(5–6-week-old)
rNiV-B e2.5 × 105
PFU
o.n.None
(0/10)
None[137]
a Animals were sacrificed at specific time points but did show clinical signs prior to euthanasia. b Human isolate passaged through experimentally infected pigs. c Approximate age at the time of challenge. d Animals euthanized due to the severity of the disease; however, animals were sacrificed at scheduled time points per study design. e Recombinant NiV-B recovered from reverse genetics system.
Table 10. Experimental findings in canine models of henipavirus disease.
Table 10. Experimental findings in canine models of henipavirus disease.
Species (Age)Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
UnknownHeV
(prototype)
5 × 103
TCID50
s.c.None
(0/2)
None[55]
Beagle
(5–8-month-old)
HeV (Australia/Horse/2008/Redlands)2 × 106
TCID50
o.n.None
(0/6)
Mild inappetence, mild conjunctivitis, tonsillar hyperplasia, transient mild temperature increase[139]
Table 11. Experimental findings in feline models of henipavirus disease.
Table 11. Experimental findings in feline models of henipavirus disease.
Species (Age)Virus (Isolate)DoseRoute% LethalityClinical DiseaseReference
Domestic shorthair cat (12–24-month-old)NiV-M5 × 103
TCID50
s.c.100%
(2/2)
Fever, increased respiratory rate, inappetence, depression[125]
5 × 102
TCID50
100%
(4/4)
Domestic shorthair catNiV-M5 × 103
TCID50
o.n.50%
(1/2)
Fever, depression, increased respiratory rates, vomiting, decreased grooming, dyspnea, open mouth breathing[130]
UnknownHeV
(prototype)
5 × 103
TCID50
s.c.100%
(2/2)
Inappetence, increased respiratory rate[55]
UnknownHeV
(prototype)
5 × 104
TCID50
o.r.100%
(2/2)
Open mouth breathing, dyspnea[144]
Unknown
(8-month-old)
HeV
(prototype)
1 × 103.6
TCID50
o.r.100%
(2/2)
Depression, fever, increased respiratory rate[143]
i.n.100%
(2/2)
s.c.100%
(2/2)
N/AContact a50%
(1/2)
UnknownHeV
(prototype)
5 × 103
TCID50
s.c.100%
(2/2)
Not described[86]
a Naïve cat housed with an animal infected via the s.c. route.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pigeaud, D.D.; Geisbert, T.W.; Woolsey, C. Animal Models for Henipavirus Research. Viruses 2023, 15, 1980. https://doi.org/10.3390/v15101980

AMA Style

Pigeaud DD, Geisbert TW, Woolsey C. Animal Models for Henipavirus Research. Viruses. 2023; 15(10):1980. https://doi.org/10.3390/v15101980

Chicago/Turabian Style

Pigeaud, Declan D., Thomas W. Geisbert, and Courtney Woolsey. 2023. "Animal Models for Henipavirus Research" Viruses 15, no. 10: 1980. https://doi.org/10.3390/v15101980

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop