Next Article in Journal / Special Issue
Stocking Density and Homogeneity, Considerations on Pandemic Potential
Previous Article in Journal / Special Issue
Livestock Reservoir Hosts: An Obscured Threat to Control of Human Schistosomiasis in Nigeria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Extrahepatic Replication Sites of Hepatitis E Virus (HEV)

by
Kush Kumar Yadav
1,2,* and
Scott P. Kenney
1,2
1
Center for Food Animal Health, Department of Veterinary Preventive Medicine, The Ohio State University, Wooster, OH 43210, USA
2
Center for Food Animal Health, Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA
*
Author to whom correspondence should be addressed.
Zoonotic Dis. 2023, 3(1), 68-84; https://doi.org/10.3390/zoonoticdis3010007
Submission received: 29 December 2022 / Revised: 6 March 2023 / Accepted: 7 March 2023 / Published: 14 March 2023
(This article belongs to the Special Issue Feature Papers of Zoonotic Diseases 2021–2022)

Abstract

:

Simple Summary

Hepatitis E virus is one of the emerging pathogens causing lethal effects to pregnant and immunosuppressed individuals. Recent progress in HEV demonstrated the ability of the virus to cross the natural body barriers such as blood–brain and blood–testis barriers. Extrahepatic existence of HEV was related to clinical manifestations via different case reports, case–control studies, and prospective studies. Knowledge about HEV-related extrahepatic diseases is very important for clinicians, as this would give them a clearer picture of organs involved in hepatitis E virus pathogenesis and spread. This summarization of the extrahepatic replication sites will help in designing treatment regimens and selection of samples for screening of hepatitis E viruses in cases of sporadic outbreaks.

Abstract

Hepatitis E virus (HEV) is an emerging viral disease known to cause acute viral hepatitis globally. Various genotypes of HEV have been identified that produce genotype specific lesions depending on the HEV targeted population. Pregnant or immunosuppressed individuals develop significantly more severe hepatitis E in comparison to the general population. In the last 40 years, we discovered that the tropism of HEV is not restricted to the liver, and virus replication was demonstrated in multiple organs. Out of the 10 body systems described in humans, HEV produces lesions causing a broad range of extrahepatic clinical manifestations in each of them. Affected body systems include nervous and musculoskeletal, cardiovascular, digestive, endocrine, integumentary, renal, respiratory, immune, and reproductive systems producing systemic lesions. All extrahepatic signs are caused by either direct HEV replication in these tissues, or indirectly by various immune mediated mechanisms. Extrahepatic replication features of HEV allowed it to cross the placental barrier, blood–brain barrier (BBB), and blood–testis barrier (BTB) that do not typically grant entry to viruses in general. Thus, in this review, we summarized the extrahepatic replication sites of HEV, listed the body systems where HEV invaded, and described multiple animal models including immunocompetent and immunosuppressed that were used to study the extrahepatic replication sites of HEV.

1. Introduction

Hepatitis E virus (HEV) is the primary cause of acute viral hepatitis in humans [1]. Clinical manifestations include asymptomatic infection, generally seen in immunocompetent individuals, acute self-limiting hepatitis, persistent hepatitis in immunosuppressed and pregnant populations, and extrahepatic manifestations [2]. HEV was reported to cause about 70,000 deaths and 3000 stillbirths annually [3].
The recent reclassification of the family Hepeviridae comprises two subfamilies: Orthohepevirinae and Parahepevirinae. A collection of terrestrial and arboreal animals are included on the infection list of the four genera within the Orthohepevirinae. Most cases of hepatitis E in humans are caused by the species Paslahepevirus balayani, which consist of eight genotypes (gt1–gt8), five of which are infectious to humans (gt1–gt4, gt7) [4,5]. HEV (gt1 and gt2) are obligated to humans, while HEV gt3 and HEV gt4 have zoonotic importance, as they travel via the food chain (pig and undercooked pork products) to develop a disease in humans [6]. Avihepevirus, Rocahepevirus, and Chirohepevirus are the other three genera predominantly circulating in birds, rodents, and bats, respectively. Notably, Rocahepevirus species ratti (rat HEV) was initially isolated from rats but was believed to have negligible zoonotic ability because of extreme genetic and antigenic divergence from HEV gt1 [7,8]. Recently, it was revealed that rat HEV can cause disease in humans via reports from Hong Kong [9,10,11].
HEV is a single stranded, positive-sense RNA virus with a 7.2 kb genome size. It is comprised of three open reading frames (ORFs), while some strains (Paslahepevirus gt1) also contain a fourth ORF, ORF4 [12,13,14,15]. The largest ORF in size is ORF1 (1693 amino acids) that encodes for nonstructural proteins (Burma strain). The translation of ORF2 (660 amino acids) and ORF3 (112–114 amino acids) is from the subgenomic RNA that encodes for structural proteins and a phosphoprotein/viroporin, respectively. Distinct functional domains: (a) methyl transferase (MT), (b) Y domain, (c) papain-like cysteine protease (PCP), (d) proline-rich hinge domain, (e) X domain, (f) RNA helicase, and (g) RNA-dependent RNA polymerase (RdRp) were reported in ORF1 based on computer-aided alignment and similarity prediction of the nucleotide sequence [2,16].
The shortage of appropriate in vitro models and in vivo models led to difficulties in understanding the pathogenesis of HEV failing to mimic the complete pathology demonstrated in humans [17,18]. Even though the fecal-oral route is considered as the main mode of HEV spread, the journey of virus particles from gastrointestinal tract to the liver and then to different organs has not been elaborated well. A very recent meta-assessment in 2018 revealed HEV prevalence up to 9% in the USA, 4.2% in Brazil, and up to 1% in the Caribbean [19]. Extrahepatic replication related to HEV acute and chronic infections have been reported by several publications [20]. Temporal associations between the infection and the extra-hepatic manifestations were made after eliminating other potential etiologies that could imitate these types of manifestations.

2. Extrahepatic Replication of HEV

2.1. Insights of Extrahepatic Replication

Even though HEV is known as a primary cause for acute hepatitis cases, chronic, and extrahepatic clinical diseases pertaining to different body systems cannot be neglected. Successful HEV replication in an organ can only be defined either by the presence of a negative-strand-specific reverse transcriptase PCR/in situ hybridization for replication complex RNA or via immunohistochemistry (IHC)/immunofluorescence assays (IFA) targeting the subgenomic RNA encoded proteins. Some HEV reports lacked the information on the negative sense RNA and IHC, limiting our complete understanding on the replication sites of HEV.
Multiple studies evaluating the vertical transmission of gt1 HEV in the fetus highlighted the successful in vitro HEV replication in the stromal cells [21], placental cells [22], both proven by IFA, and ex vivo replication in the maternal fetal interface recognized by in situ hybridization [23]. Additionally, HEV antigen was reported via IHC in the maternal and fetal side of the placental tissues collected from the HEV positive pregnant individuals after delivery [24].
The blood–testis barrier and blood–brain barriers limit immune cell trafficking into the immune privileged sites such as the testis and central nervous system, respectively [25]. There was very little knowledge about the replication of HEV in the immune privileged sites. Recently, the existence of gt3 HEV was demonstrated in the cerebrospinal fluid (CSF) [26] via the presence of a negative strand RNA in pigs and gt4 HEV via IHC in the macaque’s testis [27].
The female reproductive organs’ role in the HEV pathogenesis was one of the prime research interest areas for several years. Extensive research was conducted to understand the factors enhancing HEV virulence in pregnant women [2]. HEV tissue tropism in the ovary, ovum, and uterus was demonstrated in various species such as rabbits via IHC [28] and in BALB/c mice [29] via the detection of negative strand RNA. Even after 40 years of HEV discovery, the mechanisms behind HEV pregnancy mortality were not identified. Furthermore, pathology of HEV infection in the non-pregnant female reproductive system is completely unknown.
Of the various organs, the pancreas was extensively reported to harbor HEV replication. HEV was associated with 2.1% of acute pancreatitis cases, particularly in young males [30]. Experimental inoculation of miniature pigs with HEV gt3 demonstrated higher titers of HEV in the pancreas than the liver, highlighting necroptosis [31]. Interestingly, HEV antigens were described earlier in lymphoid tissues even before the noted classical organ of infection, “liver” via IHC [31].
It is very interesting to note that extrahepatic replication related to HEV is not limited to a genotype. From the listed studies, extrahepatic replication in males and non-pregnant females are related to gt3/gt4 HEV. One of the studies reported no evidence of gt1 HEV in the male reproductive system of humans [32]. However, an exception was seen when gt1 HEV acute infection was related to the digestive disorder, acalculous cholesystitis [33]. HEV gt1 was related with the female reproductive organs only during the pregnancy. Genotype specific lesions illustrate the need to understand the mechanisms behind the extrahepatic replication of HEV.

2.2. Pathogenesis of Extrahepatic Replication

There are many unknowns in the pathogenesis of extrahepatic manifestations due to HEV infection. Direct or indirect mechanisms were postulated with HEV induced pathogenesis. Direct mechanisms include HEV replication in the infected tissues, developing cellular damage. However, indirect mechanisms relate to cross-reactive immune triggers, development of immune complexes, or by indicated secondary infection [34]. Direct mechanisms were reported in vitro supporting the complete replication of HEV viral RNA and translation of viral capsid protein in some tissue types such as neuronal cells and human neuronal derived cells [35,36]. On the other side, humoral and cellular immune responses are responsible for the indirect mechanisms and are expected to be relevant in the pathogenesis of extrahepatic manifestations.
Remarkably, extrahepatic clinical manifestations are easily seen in immunocompetent patients rather than immunocompromised patients [37,38,39]. One report demonstrated that neurological manifestations were significantly more common in immunocompetent patients [n = 137] than immunocompromised patients [n = 63] (22.6% as compared to 3.2%) [37]. Similarly, a second report demonstrated neuralgic amyotrophy only in immunocompetent patients [38]. Likewise, pleiotropic neurologic disorders were reported in HEV-infected immunocompetent patients [39]. Such findings imply that immune-mediated mechanisms could be responsible for these extra hepatic disease manifestations.
Four decades of research advancements in HEV clearly demonstrated that the liver is not the only organ where HEV replication occurred. Neuronal cells, intestines, and human placenta are also important replication sites for HEV in humans [22,23,36,40,41]. Out of the 10 organ systems described in the human body, HEV was shown to affect all of these systems (nervous and musculoskeletal, cardiovascular, digestive, endocrine, integumentary, renal, respiratory, immune, reproductive systems) causing lesions. Thus, these extrahepatic manifestations need to be studied and characterized for the correct diagnosis of HEV infection. This review aims to summarize data pertaining to the extrahepatic manifestation of HEV reported in humans and recapitulated in animal models.

3. List of Body Systems Affected by HEV in Humans

Homeostasis is the ability of the body system to maintain a balance or equilibrium internally against external forces [42]. Invading organisms, such as HEV, trigger systemic physiological responses that disrupt normal homeostasis in the body systems when trying to establish itself in the host. Disturbance in homeostasis can be attributed to different clinical diseases which depend on the organ tropism of the pathogen and the subsequent pathogen–host–organ interactions.
Here, we list the system related clinical diseases that were demonstrated during HEV infection in humans.

3.1. Nervous and Musculoskeletal System

Neurologic manifestations of HEV are the most common extrahepatic manifestations and are increasingly being recognized as a complication of HEV infection. Europe (74%) and South East Asia Region (SEAR) nations (15%), mainly France, India, and Bangladesh reported multiple neurological disorders. Of the various disorders, the most common HEV neurological disorders are neuralgic amyotrophy (39%) and Guillain–Barre syndrome (37%) [43]. Numbers of patients reported with neurological disorders while having an HEV infection are presented below (Table 1).
Of the various reported neurological disorders associated with HEV, myasthenia gravis, encephalopathy, encephalitic parkinsonism, vestibular neuritis were reported only in individual cases (Figure 1). Myasthenia gravis was reported in a 33-year-old immunocompetent female with acute HEV infection [66]. Encephalopathy was reported in a 66-year-old, immunosuppressed renal transplant woman [72]. Acute encephalitic parkinsonism was reported in a 17-year-old sportsman with acute HEV infection [73]. Vestibular neuritis was observed in a 92-year-old female with acute HEV infection.
Of the various musculoskeletal disorders related to HEV infection, transverse myelitis and myositis were observed only in single patients. Transverse myelitis was reported in a 62-year-old Caucasian woman with acute HEV infection [74]. Myositis was observed in a 57-year-old man with a history of alcoholic chronic liver disease with acute HEV infection [76].

3.2. Cardiovascular System

Cryoglobulinemia is the most described cardiovascular disorder associated with HEV infection followed by monoclonal gammopathy, lymphocytosis, and thrombocytopenia (Table 2).
Of the various cardiovascular system disorders, cardiac arrhythmia, anemia, leukocytosis, massive hemolysis, long QT syndrome, Torsades de Pointes, and metabolic acidosis were reported only in one patient (Figure 2). Cardiac arrhythmia was reported in a 73-year-old male with acute HEV infection [57]. Anemia was reported in a 5-year-old male with acute HEV infection. The child was coinfected with hepatitis B, and parvovirus [79]. Leukocytosis was reported in a 32-year-old male with acute HEV infection [30]. Massive hemolysis was reported in a 48-year-old male that resulted in renal failure with acute HEV infection [80]. Long QT syndrome and Torsades de Pointes was reported in a 62-year-old female with acute HEV infection [82]. Metabolic acidosis was reported in a 32-year-old male with acute HEV infection [30].

3.3. Digestive System

Pancreatitis is the most attributed digestive system disorder associated with HEV infection followed by pancreatic pseudocyst and acalculous cholecystitis (Table 3).
Of the three digestive system disorders, pancreatic pseudocyst and acalculous cholecystitis associated with HEV were observed only in single patients (Figure 3). Pancreatic pseudocyst was observed in a 35-year-old man with acute HEV infection resulted in fatal outcome [87]. Acalculous cholecystitis was observed in a 24-year-old female with acute HEV infection [33].

3.4. Other Body Systems

An endocrine system disorder, autoimmune thyroiditis, was reported in a middle-aged woman with acute hepatitis E virus infection [88] (Table 4). An integumentary system disorder, cutaneous T cell lymphoproliferative, was reported in a 62-year-old Caucasian male with acute HEV infection [89]. A renal system disorder such as acute kidney injury was reported in a 32-year old Indian male with acute hepatitis infection [30]. Similarly, massive hemolysis causing renal failure was reported during acute hepatitis E infection in 48 year old man [80]. Cryoglobulinemic membranoproliferative glomerulonephritis, another renal system disorder, was reported in a 46-year-old man infected with gt3 HEV infection after the third kidney transplantation [90]. In contrast, another report of HEV induced cryoglobulinemic glomerulonephritis was from non-immunocompromised 48-year-old native French man [91].
Interestingly, a respiratory system disorder, pleural effusion was reported during coinfection of hepatitis E and hepatitis A viruses [92]. Immune system disorders, acute allograft dysfunction was reported in a 56-year-old male patient after 17 years of liver transplant with acute HEV infection [93]. Histologic reports of his liver demonstrated portal infiltration by lymphocytes during the acute HEV infection [93]. Reproductive system disorder, infertility was reported in 185 males of which 28.11% (52/185) were HEV positive [27]. Also, it was found that in some patients the blood–testis barrier was destroyed by HEV infection [27].

4. List of the Animal Models Studied with Involved Body Systems and Extrahepatic Replication Sites for HEV

To dissect the in vivo characteristics of HEV and determine its replicative sites other than the liver, it is important to understand the viral pathogenesis utilizing various animal models. Multiple animal models were used to understand the molecular pathobiology of HEV and to gain an understanding of the extrahepatic replication sites’ role in invading the immune system of the host. Although immunocompetent animal models depict the susceptibility to distinct HEV strains and mimic human disease [94], recapitulating the immunosuppressed individual’s scenario can only be done utilizing the immunosuppressed animal models. Thus, immunosuppressed animal models gained popularity mimicking the immunosuppressed solid organ transplant recipients. Such immunosuppressed animal models can be used to understand the disease severity and the role of extrahepatic sites in the maintenance of HEV. Below, we list the HEV genotypes and/or strains, body systems, and extrahepatic sites involved during the infection in immunocompetent and immunosuppressed animal models (Table 5).

5. Notable Abilities of HEV

In general, reorganization of intracellular membranes (endoplasmic reticulum, golgi apparatus, mitochondria, endosomes and/or lysosomes) to establish replication sites is the unique attribute of positive-stranded RNA viruses. These replication complexes function as platforms to enhance viral and cellular cofactors at local level and offer a secure setting which decreases the innate immune system detection of viral proteins and nucleic acids [116]. Genetic plasticity is one of the advantageous attributes of RNA viruses, thus, they can promptly produce drug-resistant viral populations. This helps them to escape the host immune system under the pressure of homeostasis. Lack of proofreading attributed to RdRp is responsible for such variability [117]. A report suggested that during viral replication, hundreds of progenies (quasispecies) are produced with a mutation rate comprising 10−6 to 10−4 substitutions per nucleotide resulting in the alteration of genomic sequences attributed to one or few nucleotide substitutions [118]. In addition, the propagation of a well-adapted viral population depends upon the quasispecies fitness ability indicating Darwinian evolution and natural selection [118].
ORF1 encodes the replicative machinery of HEV which contains methyltransferase, RNA helicase, X and Y domains, protease, and RdRp [119]. Polyprotein processing into distinct domains during the HEV life cycle remains debatable but is generally seen in other viruses [120,121,122,123,124,125,126]. Moreover, the hypervariable region (HVR) of the HEV ORF1 exhibits sequence differences even among isolates of identical virus genotypes [127]. Acute hepatitis patients were reported to consist of HEV with naïve genomic rearrangements associated with HEV persistence and long term use of ribavirin. It resulted in ribavirin insensitivity to HEV with an observed increase in the HEV quasispecies [128,129,130,131,132]. A recombination was demonstrated between HEV genotypes and fragments of human genes and HEV strains. For example, human RPS17 (ribosomal protein S17) inclusion in the hypervariable region boosted replication in hepatoma cells [133].
In addition, HEV is found in two forms: nonenveloped and quasi-enveloped. Both were demonstrated to take different replicative cycles with differences in infectivity [134]. For example, effective replication of KM01 (genotype 4 HEV) strain isolated from pig fecal samples was demonstrated in A549 cells and Huh7.5.1 cells following an extensive period (more than 11 years) of serial passaging. Feces derived or bile-derived HEV (nonenveloped) and cell culture-derived (enveloped) HEV was reported in A549 cells when inoculated with the KM01 strain. Fairly distinct infectivity was noted between two forms (5.34% of enveloped HEV versus 10.80% of non-enveloped HEV in A549 cells) suggesting non-enveloped HEV is more infectious when compared to enveloped HEV [134]. Unlike non-enveloped HEV, the entry of enveloped HEV needs Rab5 and Rab7, small GTPases participating in endosomal trafficking [135]. Interestingly, the infectivity of enveloped HEV was enhanced when the virus was treated with 1% NP-40 to eliminate the quasi-enveloped membranes [134].
The in vitro blood-brain barrier (BBB) model and in vivo HEV infection study in pigs revealed HEV virions, both quasi-enveloped and nonenveloped, possessed the ability to compromise the BBB and enter the central nervous system (CNS) [26]. Furthermore, HEV can cross the placental barrier and was demonstrated to transmit via vertical transmission [23,24]. In addition, HEV is shown to be persistent in the ejaculate of chronically infected men [27,136]. No noted difference could be observed between enveloped HEV, when compared morphologically to serum derived HEV particles, suggesting its ability to cross the blood–testis barrier (BTB) [26,27,136]. Interestingly, HEV was also reported in the muscle of wild boars [137,138,139]. Thus, with the increase in host organ tropism, HEV is slowly becoming an even more important emerging pathogen. HEV currently ranks 6th on a list of 50 priority zoonotic viral pathogens, only behind Lassa, SARS-CoV-2, Ebola, Seoul, and Nipah viruses, and ahead of viral pathogens such as Monkeypox, Marburg, Rabies, Simian immunodeficiency virus, and other coronaviruses currently of global relevance [140]. HEV being entrenched as a food borne zoonosis possesses a higher spillover risk estimate than some known zoonotic pathogens which necessitates a better understanding of HEV pathogenesis and its virulence mechanisms in relation to body system preference.

6. Conclusions

The increasing host range of HEV guided numerous innovative experimental and naturally existing animal models capable of recapitulating extrahepatic manifestations. These models offer prospects for potential HEV study with strains from humans, macaques, pigs, wild boar, rabbits, gerbils, and mice that could be used to understand the extrahepatic replication role in HEV pathogenesis. In addition, there could be multiple reservoir cells for HEV in the human body that still need to be investigated for the application of proper treatment and prevention strategies against HEV. Summarizing the body systems involved in the HEV replication gives us a somewhat clearer picture of organs involved in HEV pathogenesis and spread. This summary will help in designing treatment regimens and selection of samples for screening of HEV in cases of a sporadic outbreak.

Author Contributions

Conceptualization, K.K.Y. and S.P.K.; writing-original draft preparation, K.K.Y.; writing—review and editing, K.K.Y. and S.P.K. All authors have read and agreed to the published version of the manuscript.

Funding

Additional salaries and research support were provided by state and federal funds appropriated to the Ohio Agricultural Research and Development Center, The Ohio State University, and from the research funds of National Institute of Allergy and Infectious Diseases (#R21AI151736).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Lhomme, S.; Marion, O.; Abravanel, F.; Izopet, J.; Kamar, N. Clinical Manifestations, Pathogenesis and Treatment of Hepatitis E Virus Infections. J. Clin. Med. 2020, 9, 331. [Google Scholar] [CrossRef] [Green Version]
  2. Yadav, K.K.; Kenney, S.P. Hepatitis E Virus Immunopathogenesis. Pathogens 2021, 10, 1180. [Google Scholar] [CrossRef]
  3. Rein, D.B.; Stevens, G.A.; Theaker, J.; Wittenborn, J.S.; Wiersma, S.T. The global burden of hepatitis E virus genotypes 1 and 2 in 2005. Hepatol. Baltim. Md. 2012, 55, 988–997. [Google Scholar] [CrossRef]
  4. Wang, B.; Yang, X.L. Chirohepevirus from Bats: Insights into Hepatitis E Virus Diversity and Evolution. Viruses 2022, 14, 905. [Google Scholar] [CrossRef] [PubMed]
  5. Purdy, M.A.; Drexler, J.F.; Meng, X.J.; Norder, H.; Okamoto, H.; Van der Poel, W.H.M.; Reuter, G.; de Souza, W.M.; Ulrich, R.G.; Smith, D.B. ICTV Virus Taxonomy Profile: Hepeviridae 2022. J. Gen. Virol. 2022, 103, 1778. [Google Scholar] [CrossRef]
  6. Legrand-Abravanel, F.; Kamar, N.; Sandres-Saune, K.; Garrouste, C.; Dubois, M.; Mansuy, J.M.; Muscari, F.; Sallusto, F.; Rostaing, L.; Izopet, J. Characteristics of autochthonous hepatitis E virus infection in solid-organ transplant recipients in France. J. Infect. Dis. 2010, 202, 835–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Johne, R.; Plenge-Bönig, A.; Hess, M.; Ulrich, R.G.; Reetz, J.; Schielke, A. Detection of a novel hepatitis E-like virus in faeces of wild rats using a nested broad-spectrum RT-PCR. J. Gen. Virol. 2010, 91, 750–758. [Google Scholar] [CrossRef] [PubMed]
  8. Sridhar, S.; Situ, J.; Cai, J.-P.; Yip, C.C.-Y.; Wu, S.; Zhang, A.J.-X.; Wen, L.; Chew, N.F.-S.; Chan, W.-M.; Poon, R.W.-S. Multimodal investigation of rat hepatitis E virus antigenicity: Implications for infection, diagnostics, and vaccine efficacy. J. Hepatol. 2021, 74, 1315–1324. [Google Scholar] [CrossRef] [PubMed]
  9. Rivero-Juarez, A.; Frias, M.; Lopez-Lopez, P.; de Los Angeles Risalde, M.; Brieva, T.; Machuca, I.; Camacho, A.; Martinez-Peinado, A.; Gomez-Villamandos, J.C.; Rivero, A. Hepatitis E virus (HEV) infection in anti-HEV immunoglobulin G-carrying patients after successful hepatitis C virus treatment: Reactivation or reinfection? Clin. Infect. Dis. 2017, 64, 964–966. [Google Scholar]
  10. Andonov, A.; Robbins, M.; Borlang, J.; Cao, J.; Hatchette, T.; Stueck, A.; Deschambault, Y.; Murnaghan, K.; Varga, J.; Johnston, L. Rat hepatitis E virus linked to severe acute hepatitis in an immunocompetent patient. J. Infect. Dis. 2019, 220, 951–955. [Google Scholar] [CrossRef]
  11. Sridhar, S.; Yip, C.C.; Wu, S.; Cai, J.; Zhang, A.J.-X.; Leung, K.-H.; Chung, T.W.; Chan, J.F.; Chan, W.-M.; Teng, J.L. Rat hepatitis E virus as cause of persistent hepatitis after liver transplant. Emerg. Infect. Dis. 2018, 24, 2241. [Google Scholar] [CrossRef] [Green Version]
  12. Kenney, S.P.; Meng, X.J. Hepatitis E Virus Genome Structure and Replication Strategy. Cold Spring Harb. Perspect. Med. 2019, 9, a031724. [Google Scholar] [CrossRef]
  13. Chandra, V.; Taneja, S.; Kalia, M.; Jameel, S. Molecular biology and pathogenesis of hepatitis E virus. J. Biosci. 2008, 33, 451–464. [Google Scholar] [CrossRef]
  14. Nair, V.P.; Anang, S.; Subramani, C.; Madhvi, A.; Bakshi, K.; Srivastava, A.; Shalimar; Nayak, B.; Ranjith Kumar, C.T.; Surjit, M. Endoplasmic Reticulum Stress Induced Synthesis of a Novel Viral Factor Mediates Efficient Replication of Genotype-1 Hepatitis E Virus. PLoS Pathog. 2016, 12, e1005521. [Google Scholar] [CrossRef]
  15. Yadav, K.K.; Boley, P.A.; Fritts, Z.; Kenney, S.P. Ectopic Expression of Genotype 1 Hepatitis E Virus ORF4 Increases Genotype 3 HEV Viral Replication in Cell Culture. Viruses 2021, 13, 10075. [Google Scholar] [CrossRef] [PubMed]
  16. Koonin, E.V.; Gorbalenya, A.E.; Purdy, M.A.; Rozanov, M.N.; Reyes, G.R.; Bradley, D.W. Computer-assisted assignment of functional domains in the nonstructural polyprotein of hepatitis E virus: Delineation of an additional group of positive-strand RNA plant and animal viruses. Proc. Natl. Acad. Sci. USA 1992, 89, 8259–8263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Kenney, S.P.; Meng, X.J. Hepatitis E Virus: Animal Models and Zoonosis. Annu. Rev. Anim. Biosci. 2019, 7, 427–448. [Google Scholar] [CrossRef]
  18. Okamoto, H. Culture systems for hepatitis E virus. J. Gastroenterol. 2013, 48, 147–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Horvatits, T.; Ozga, A.K.; Westhölter, D.; Hartl, J.; Manthey, C.F.; Lütgehetmann, M.; Rauch, G.; Kriston, L.; Lohse, A.W.; Bendall, R.; et al. Hepatitis E seroprevalence in the Americas: A systematic review and meta-analysis. Liver Int. Off. J. Int. Assoc. Study Liver 2018, 38, 1951–1964. [Google Scholar] [CrossRef]
  20. Wu, J.; Xiang, Z.; Zhu, C.; Yao, Y.; Bortolanza, M.; Cao, H.; Li, L. Extrahepatic manifestations related to hepatitis E virus infection and their triggering mechanisms. J. Infect. 2021, 83, 298–305. [Google Scholar] [CrossRef] [PubMed]
  21. El-Mokhtar, M.A.; Othman, E.R.; Khashbah, M.Y.; Ismael, A.; Ghaliony, M.A.; Seddik, M.I.; Sayed, I.M. Evidence of the extrahepatic replication of Hepatitis E virus in human endometrial stromal cells. Pathogens 2020, 9, 295. [Google Scholar] [CrossRef] [Green Version]
  22. Knegendorf, L.; Drave, S.A.; Dao Thi, V.L.; Debing, Y.; Brown, R.J.P.; Vondran, F.W.R.; Resner, K.; Friesland, M.; Khera, T.; Engelmann, M.; et al. Hepatitis E virus replication and interferon responses in human placental cells. Hepatol. Commun. 2018, 2, 173–187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gouilly, J.; Chen, Q.; Siewiera, J.; Cartron, G.; Levy, C.; Dubois, M.; Al-Daccak, R.; Izopet, J.; Jabrane-Ferrat, N.; El Costa, H. Genotype specific pathogenicity of hepatitis E virus at the human maternal-fetal interface. Nat. Commun. 2018, 9, 4748. [Google Scholar] [CrossRef] [Green Version]
  24. Ratho, R.K.; Thakur, V.; Arya, S.; Singh, M.P.; Suri, V.; Das, A. Placenta as a site of HEV replication and inflammatory cytokines modulating the immunopathogenesis of HEV in pregnant women. J. Med. Virol. 2022, 94, 3457–3463. [Google Scholar] [CrossRef] [PubMed]
  25. Muldoon, L.L.; Alvarez, J.I.; Begley, D.J.; Boado, R.J.; Del Zoppo, G.J.; Doolittle, N.D.; Engelhardt, B.; Hallenbeck, J.M.; Lonser, R.R.; Ohlfest, J.R.; et al. Immunologic privilege in the central nervous system and the blood-brain barrier. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2013, 33, 13–21. [Google Scholar] [CrossRef] [Green Version]
  26. Tian, D.; Li, W.; Heffron, C.L.; Wang, B.; Mahsoub, H.M.; Sooryanarain, H.; Hassebroek, A.M.; Clark-Deener, S.; LeRoith, T.; Meng, X.-J. Hepatitis E virus infects brain microvascular endothelial cells, crosses the blood–brain barrier, and invades the central nervous system. Proc. Natl. Acad. Sci. USA 2022, 119, e2201862119. [Google Scholar] [CrossRef] [PubMed]
  27. Huang, F.; Long, F.; Yu, W.; Situ, J.; Fu, L.; He, Z.; Dong, H.; Yang, C.; Li, Y.; Yang, F. High prevalence of hepatitis E virus in semen of infertile male and causes testis damage. Gut 2018, 67, 1199–1201. [Google Scholar] [CrossRef]
  28. Ahn, H.-S.; Han, S.-H.; Kim, Y.-H.; Park, B.-J.; Kim, D.-H.; Lee, J.-B.; Park, S.-Y.; Song, C.-S.; Lee, S.-W.; Choi, C.; et al. Adverse fetal outcomes in pregnant rabbits experimentally infected with rabbit hepatitis E virus. Virology 2017, 512, 187–193. [Google Scholar] [CrossRef]
  29. Yang, W.; Chen, S.; Mickael, H.K.; Xu, L.; Xia, Y.; Cong, C.; Zhang, Y.; Qian, Z.; Li, T.; Wei, D.; et al. Uterine Injury Caused by Genotype 4 Hepatitis E Virus Infection Based on a BALB/c Mice Model. Viruses 2021, 13, 1950. [Google Scholar] [CrossRef]
  30. Vallabhajosyula, S.; Prabhu, M.; Stanley, W.; Vallabhajosyula, S. 510: Multiorgan Dysfunction from Hepatitis E infection. Crit. Care Med. 2018, 46, 240. [Google Scholar] [CrossRef]
  31. Jung, S.; Seo, D.J.; Yeo, D.; Wang, Z.; Min, A.; Zhao, Z.; Song, M.; Choi, I.S.; Myoung, J.; Choi, C. Experimental infection of hepatitis E virus induces pancreatic necroptosis in miniature pigs. Sci. Rep. 2020, 10, 12022. [Google Scholar] [CrossRef]
  32. El-Mokhtar, M.A.; Seddik, M.I.; Osman, A.O.B.; Mahmoud, A.A.; Mandour, S.A.; Radwan, E.; Ali, M.; Ismael, A.E.; Twisy, H.O.; Karam-Allah Ramadan, H.; et al. No evidence of HEV genotype 1 infections harming the male reproductive system. Virology 2021, 554, 37–41. [Google Scholar] [CrossRef]
  33. Fujioka, K.; Nishimura, T.; Seki, M.; Kinoshita, M.; Mishima, N.; Irimajiri, S.; Yamato, M. Genotype 1 hepatitis E virus infection with acute acalculous cholecystitis as an extrahepatic symptom: A case report. Trop. Med. Health 2016, 44, 18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Feng, Z. Causation by HEV of extrahepatic manifestations remains unproven. Liver Int. Off. J. Int. Assoc. Study Liver 2016, 36, 477–479. [Google Scholar] [CrossRef] [Green Version]
  35. Zhou, X.; Huang, F.; Xu, L.; Lin, Z.; de Vrij, F.M.S.; Ayo-Martin, A.C.; van der Kroeg, M.; Zhao, M.; Yin, Y.; Wang, W.; et al. Hepatitis E Virus Infects Neurons and Brains. J. Infect. Dis. 2017, 215, 1197–1206. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Drave, S.A.; Debing, Y.; Walter, S.; Todt, D.; Engelmann, M.; Friesland, M.; Wedemeyer, H.; Neyts, J.; Behrendt, P.; Steinmann, E. Extra-hepatic replication and infection of hepatitis E virus in neuronal-derived cells. J. Viral Hepat. 2016, 23, 512–521. [Google Scholar] [CrossRef] [PubMed]
  37. Abravanel, F.; Pique, J.; Couturier, E.; Nicot, F.; Dimeglio, C.; Lhomme, S.; Chiabrando, J.; Saune, K.; Péron, J.M.; Kamar, N.; et al. Acute hepatitis E in French patients and neurological manifestations. J. Infect. 2018, 77, 220–226. [Google Scholar] [CrossRef]
  38. Ripellino, P.; Pasi, E.; Melli, G.; Staedler, C.; Fraga, M.; Moradpour, D.; Sahli, R.; Aubert, V.; Martinetti, G.; Bihl, F.; et al. Neurologic complications of acute hepatitis E virus infection. Neurol. R Neuroimmunol. Neuroinflamm. 2020, 7, 643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Perrin, H.B.; Cintas, P.; Abravanel, F.; Gerolami, R.; d’Alteroche, L.; Raynal, J.-N.; Alric, L.; Dupuis, E.; Prudhomme, L.; Vaucher, E. Neurologic disorders in immunocompetent patients with autochthonous acute hepatitis E. Emerg. Infect. Dis. 2015, 21, 1928. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Marion, O.; Lhomme, S.; Nayrac, M.; Dubois, M.; Pucelle, M.; Requena, M.; Migueres, M.; Abravanel, F.; Peron, J.M.; Carrere, N.; et al. Hepatitis E virus replication in human intestinal cells. Gut 2020, 69, 901–910. [Google Scholar] [CrossRef]
  41. Sayed, I.M.; Seddik, M.I.; Gaber, M.A.; Saber, S.H.; Mandour, S.A.; El-Mokhtar, M.A. Replication of Hepatitis E Virus (HEV) in Primary Human-Derived Monocytes and Macrophages In Vitro. Vaccines 2020, 8, 239. [Google Scholar] [CrossRef]
  42. Billman, G.E. Homeostasis: The Underappreciated and Far Too Often Ignored Central Organizing Principle of Physiology. Front. Physiol. 2020, 11, 200. [Google Scholar] [CrossRef] [Green Version]
  43. Jha, A.K.; Kumar, G.; Dayal, V.M.; Ranjan, A.; Suchismita, A. Neurological manifestations of hepatitis E virus infection: An overview. World J. Gastroenterol. 2021, 27, 2090–2104. [Google Scholar] [CrossRef]
  44. Avila, J.D.; Lacomis, D.; Lam, E.M. Neuralgic Amyotrophy Associated With Hepatitis E Virus Infection: First Case in the United States. J. Clin. Neuromuscul. Dis. 2016, 18, 96–100. [Google Scholar] [CrossRef]
  45. Garofoli, R.; Zauderer, J.; Seror, P.; Roren, A.; Guerini, H.; Rannou, F.; Drapé, J.L.; Nguyen, C.; Lefèvre-Colau, M.M. Neuralgic amyotrophy and hepatitis E infection: 6 prospective case reports. RMD Open 2020, 6, e001401. [Google Scholar] [CrossRef]
  46. Dalton, H.R.; van Eijk, J.J.J.; Cintas, P.; Madden, R.G.; Jones, C.; Webb, G.W.; Norton, B.; Pique, J.; Lutgens, S.; Devooght-Johnson, N.; et al. Hepatitis E virus infection and acute non-traumatic neurological injury: A prospective multicentre study. J. Hepatol. 2017, 67, 925–932. [Google Scholar] [CrossRef]
  47. Dartevel, A.; Colombe, B.; Bosseray, A.; Larrat, S.; Sarrot-Reynauld, F.; Belbezier, A.; Lagrange, E.; Bouillet, L. Hepatitis E and neuralgic amyotrophy: Five cases and review of literature. J. Clin. Virol. Off. Publ. Pan Am. Soc. Clin. Virol. 2015, 69, 156–164. [Google Scholar] [CrossRef]
  48. Deroux, A.; Brion, J.P.; Hyerle, L.; Belbezier, A.; Vaillant, M.; Mosnier, E.; Larrat, S.; Morand, P.; Pavese, P. Association between hepatitis E and neurological disorders: Two case studies and literature review. J. Clin. Virol. Off. Publ. Pan Am. Soc. Clin. Virol. 2014, 60, 60–62. [Google Scholar] [CrossRef]
  49. Fong, F.; Illahi, M. Neuralgic amyotrophy associated with hepatitis E virus. Clin. Neurol. Neurosurg. 2009, 111, 193–195. [Google Scholar] [CrossRef]
  50. Fritz, M.; Berger, B.; Schemmerer, M.; Endres, D.; Wenzel, J.J.; Stich, O.; Panning, M. Pathological Cerebrospinal Fluid Findings in Patients With Neuralgic Amyotrophy and Acute Hepatitis E Virus Infection. J. Infect. Dis. 2018, 217, 1897–1901. [Google Scholar] [CrossRef]
  51. Jones, K.; Van Eijk, J.; Ripellino, P.; Madden, R.; Herrod, J.; Lissmann, R.; Webb, G.; Abdelrahim, M.; Ashraf, H.; Almasri, O. Clinical phenotype and outcome of hepatitis E virus associated neuralgic amyotrophy; an international retrospective comparative cohort study. J. Hepatol. 2017, 1, S59. [Google Scholar] [CrossRef]
  52. Sánchez-Azofra, M.; Romero-Portales, M.; Tortajada-Laureiro, L.; García-Samaniego, J.; Mora-Sanz, P. Hepatitis E virus in neurological disorders: A case of Parsonage-Turner syndrome. Rev. Esp. Enfermadades Dig. REED 2018, 110, 402–404. [Google Scholar] [CrossRef]
  53. Scanvion, Q.; Perez, T.; Cassim, F.; Outteryck, O.; Lanteri, A.; Hatron, P.-Y.; Lambert, M.; Morell-Dubois, S. Neuralgic amyotrophy triggered by hepatitis E virus: A particular phenotype. J. Neurol. 2017, 264, 770–780. [Google Scholar] [CrossRef]
  54. Silva, M.; Wicki, B.; Tsouni, P.; Cunningham, S.; Doerig, C.; Zanetti, G.; Aubert, V.; Sahli, R.; Moradpour, D.; Kuntzer, T. Hepatitis E virus infection as a direct cause of neuralgic amyotrophy. Muscle Nerve 2016, 54, 325–327. [Google Scholar] [CrossRef]
  55. Swinnen, B.; Boeynaems, S.; Schrooten, M.; Saegeman, V.; Claeys, K.G.; Van Damme, P. Anterior interosseous mononeuropathy associated with HEV infection. Neurol. R Neuroimmunol. Neuroinflamm. 2018, 5, e429. [Google Scholar] [CrossRef] [Green Version]
  56. Velay, A.; Kack-Kack, W.; Abravanel, F.; Lhomme, S.; Leyendecker, P.; Kremer, L.; Chamouard, P.; Izopet, J.; Fafi-Kremer, S.; Barth, H. Parsonage-Turner syndrome due to autochthonous acute genotype 3f hepatitis E virus infection in a nonimmunocompromised 55-year-old patient. J. Neurovirol. 2017, 23, 615–620. [Google Scholar] [CrossRef]
  57. Woolson, K.L.; Forbes, A.; Vine, L.; Beynon, L.; McElhinney, L.; Panayi, V.; Hunter, J.G.; Madden, R.G.; Glasgow, T.; Kotecha, A.; et al. Extra-hepatic manifestations of autochthonous hepatitis E infection. Aliment. Pharmacol. Ther. 2014, 40, 1282–1291. [Google Scholar] [CrossRef]
  58. Al-Saffar, A.; Al-Fatly, B. Acute Motor Axonal Neuropathy in Association with Hepatitis E. Front. Neurol. 2018, 9, 62. [Google Scholar] [CrossRef] [Green Version]
  59. Choudhary, M.C.; Bajpai, V.; Anand, L.; Gupta, E. Guillain-Barré syndrome in a patient of acute Hepatitis E virus infection associated with genotype 1: Case report and literature review. Intractable Rare Dis. Res. 2019, 8, 43–47. [Google Scholar] [CrossRef] [Green Version]
  60. Fukae, J.; Tsugawa, J.; Ouma, S.; Umezu, T.; Kusunoki, S.; Tsuboi, Y. Guillain-Barré and Miller Fisher syndromes in patients with anti-hepatitis E virus antibody: A hospital-based survey in Japan. Neurol. Sci Off. J. Ital. Neurol. Soc. Ital. Soc. Clin. Neurophysiol. 2016, 37, 1849–1851. [Google Scholar] [CrossRef]
  61. Lei, J.H.; Tian, Y.; Luo, H.Y.; Chen, Z.; Peng, F. Guillain-Barré syndrome following acute co-super-infection of hepatitis E virus and cytomegalovirus in a chronic hepatitis B virus carrier. J. Med. Virol. 2017, 89, 368–372. [Google Scholar] [CrossRef]
  62. Salim, O.J.; Davidson, A.; Li, K.; Leach, J.P.; Heath, C. Brainstem encephalitis and acute polyneuropathy associated with hepatitis E infection. BMJ Case Rep. 2017, 2017, 220799. [Google Scholar] [CrossRef]
  63. Stevens, O.; Claeys, K.G.; Poesen, K.; Saegeman, V.; Van Damme, P. Diagnostic Challenges and Clinical Characteristics of Hepatitis E Virus-Associated Guillain-Barré Syndrome. JAMA Neurol. 2017, 74, 26–33. [Google Scholar] [CrossRef]
  64. van den Berg, B.; van der Eijk, A.A.; Pas, S.D.; Hunter, J.G.; Madden, R.G.; Tio-Gillen, A.P.; Dalton, H.R.; Jacobs, B.C. Guillain-Barré syndrome associated with preceding hepatitis E virus infection. Neurology 2014, 82, 491–497. [Google Scholar] [CrossRef]
  65. Wang, Y.; Wang, S.; Wu, J.; Jiang, Y.; Zhang, H.; Li, S.; Liu, H.; Yang, C.; Tang, H.; Guo, N.; et al. Hepatitis E virus infection in acute non-traumatic neuropathy: A large prospective case-control study in China. EBioMedicine 2018, 36, 122–130. [Google Scholar] [CrossRef] [Green Version]
  66. Belbezier, A.; Deroux, A.; Sarrot-Reynauld, F.; Larrat, S.; Bouillet, L. Myasthenia gravis associated with acute hepatitis E infection in immunocompetent woman. Emerg. Infect. Dis. 2014, 20, 908–910. [Google Scholar] [CrossRef]
  67. Belliere, J.; Abravanel, F.; Nogier, M.B.; Martinez, S.; Cintas, P.; Lhomme, S.; Lavayssière, L.; Cointault, O.; Faguer, S.; Izopet, J.; et al. Transfusion-acquired hepatitis E infection misdiagnosed as severe critical illness polyneuromyopathy in a heart transplant patient. Transpl. Infect. Dis. Off. J. Transplant. Soc. 2017, 19, 12784. [Google Scholar] [CrossRef]
  68. Despierres, L.A.; Kaphan, E.; Attarian, S.; Cohen-Bacrie, S.; Pelletier, J.; Pouget, J.; Motte, A.; Charrel, R.; Gerolami, R.; Colson, P. Neurologic disorders and hepatitis E, France, 2010. Emerg. Infect. Dis. 2011, 17, 1510–1512. [Google Scholar] [CrossRef]
  69. Murkey, J.A.; Chew, K.W.; Carlson, M.; Shannon, C.L.; Sirohi, D.; Sample, H.A.; Wilson, M.R.; Vespa, P.; Humphries, R.M.; Miller, S.; et al. Hepatitis E Virus-Associated Meningoencephalitis in a Lung Transplant Recipient Diagnosed by Clinical Metagenomic Sequencing. Open Forum Infect. Dis. 2017, 4, ofx121. [Google Scholar] [CrossRef]
  70. Jha, A.K.; Nijhawan, S.; Nepalia, S.; Suchismita, A. Association of Bell’s Palsy with Hepatitis E Virus Infection: A Rare Entity. J. Clin. Exp. Hepatol. 2012, 2, 88–90. [Google Scholar] [CrossRef] [Green Version]
  71. Yazaki, Y.; Sugawara, K.; Honda, M.; Ohnishi, H.; Nagashima, S.; Takahashi, M.; Okamoto, H. Characteristics of 20 Patients with Autochthonous Acute Hepatitis E in Hokkaido, Japan: First Report of Bilateral Facial Palsy Following the Infection with Genotype 4 Hepatitis E Virus. Tohoku J. Exp. Med. 2015, 236, 263–271. [Google Scholar] [CrossRef] [Green Version]
  72. de Vries, M.A.; Samijn, J.P.; de Man, R.; Boots, J.M. Hepatitis E-associated encephalopathy in a renal transplant recipient. BMJ Case Rep. 2014, 2014, 204244. [Google Scholar] [CrossRef] [Green Version]
  73. Pasha, S.A.; Pasha, S.A.; Suhasini, T.; Rao, D.A. Hepatitis E Virus-Associated Acute Encephalitic Parkinsonism. J. Assoc. Physicians India 2018, 66, 92–93. [Google Scholar]
  74. Sarkar, P.; Morgan, C.; Ijaz, S. Transverse myelitis caused by hepatitis E: Previously undescribed in adults. BMJ Case Rep. 2015, 2015, 209031. [Google Scholar] [CrossRef] [Green Version]
  75. Bennett, S.; Li, K.; Gunson, R.N. Hepatitis E virus infection presenting with paraesthesia. Scott. Med. J. 2015, 60, e27–e29. [Google Scholar] [CrossRef]
  76. Mengel, A.M.; Stenzel, W.; Meisel, A.; Büning, C. Hepatitis E-induced severe myositis. Muscle Nerve 2016, 53, 317–320. [Google Scholar] [CrossRef]
  77. Premkumar, M.; Rangegowda, D.; Vashishtha, C.; Bhatia, V.; Khumuckham, J.S.; Kumar, B. Acute viral hepatitis e is associated with the development of myocarditis. Case Rep. Hepatol. 2015, 2015, 458056. [Google Scholar] [CrossRef]
  78. Dougherty, T.; Showkat, B.; Adam, M.; Borum, M. Acute myopericarditis due to Hepatitis E virus infection: The first reported case in the western hemisphere. J. Gastrointest. Dig. Syst. 2016, 6, 2. [Google Scholar] [CrossRef]
  79. Kishore, J.; Sen, M. Parvovirus B19-induced thrombocytopenia and anemia in a child with fatal fulminant hepatic failure coinfected with hepatitis A and E viruses. J. Trop. Pediatr. 2009, 55, 335–337. [Google Scholar] [CrossRef] [Green Version]
  80. Karki, P.; Malik, S.; Mallick, B.; Sharma, V.; Rana, S.S. Massive Hemolysis Causing Renal Failure in Acute Hepatitis E Infection. J. Clin. Transl. Hepatol. 2016, 4, 345–347. [Google Scholar] [CrossRef] [Green Version]
  81. Marion, O.; Abravanel, F.; Del Bello, A.; Esposito, L.; Lhomme, S.; Puissant-Lubrano, B.; Alric, L.; Faguer, S.; Izopet, J.; Kamar, N. Hepatitis E virus-associated cryoglobulinemia in solid-organ-transplant recipients. Liver Int. Off. J. Int. Assoc. Study Liver 2018, 38, 2178–2189. [Google Scholar] [CrossRef] [PubMed]
  82. Aiqin, Z.; Dongming, X.; Kejun, T.; Yun, Z. Long Qt Syndrome and Torsades De Points in a patient with acute hepatitis E virus infection: An unusual case. Heart 2012, 98, E222–E223. [Google Scholar] [CrossRef] [Green Version]
  83. Deniel, C.; Coton, T.; Brardjanian, S.; Guisset, M.; Nicand, E.; Simon, F. Acute pancreatitis: A rare complication of acute hepatitis E. J. Clin. Virol. Off. Publ. Pan Am. Soc. Clin. Virol. 2011, 51, 202–204. [Google Scholar] [CrossRef] [PubMed]
  84. Raj, M.; Kumar, K.; Ghoshal, U.C.; Saraswat, V.A.; Aggarwal, R.; Mohindra, S. Acute Hepatitis E-Associated Acute Pancreatitis: A Single Center Experience and Literature Review. Pancreas 2015, 44, 1320–1322. [Google Scholar] [CrossRef]
  85. Sinha, S.; Jha, R.; Lakhtakia, S.; Narayan, G. Acute pancreatitis following kidney transplantation—Role of viral infections. Clin. Transplant. 2003, 17, 32–36. [Google Scholar] [CrossRef]
  86. Thakur, A.; Basu, P.P. Acute Non-Fulminant Viral Hepatitis E Presenting with Acute Pancreatitis-An Unusual Presentation. Malays. J. Med. Sci. MJMS 2017, 24, 102–105. [Google Scholar] [CrossRef]
  87. Somani, S.K.; Ghosh, A.; Awasthi, G. Severe acute pancreatitis with pseudocyst bleeding due to hepatitis E virus infection. Clin. J. Gastroenterol. 2009, 2, 39–42. [Google Scholar] [CrossRef]
  88. Dumoulin, F.L.; Liese, H. Acute hepatitis E virus infection and autoimmune thyroiditis: Yet another trigger? BMJ Case Rep. 2012, 2012, 5441. [Google Scholar] [CrossRef] [Green Version]
  89. Mallet, V.; Bruneau, J.; Zuber, J.; Alanio, C.; Leclerc-Mercier, S.; Roque-Afonso, A.M.; Kraft, A.R.M.; Couronné, L.; Roulot, D.; Wedemeyer, H.; et al. Hepatitis E virus-induced primary cutaneous CD30(+) T cell lymphoproliferative disorder. J. Hepatol. 2017, 67, 1334–1339. [Google Scholar] [CrossRef]
  90. Del Bello, A.; Guilbeau-Frugier, C.; Josse, A.G.; Rostaing, L.; Izopet, J.; Kamar, N. Successful treatment of hepatitis E virus-associated cryoglobulinemic membranoproliferative glomerulonephritis with ribavirin. Transpl. Infect. Dis. Off. J. Transplant. Soc. 2015, 17, 279–283. [Google Scholar] [CrossRef]
  91. Guinault, D.; Ribes, D.; Delas, A.; Milongo, D.; Abravanel, F.; Puissant-Lubrano, B.; Izopet, J.; Kamar, N. Hepatitis E Virus-Induced Cryoglobulinemic Glomerulonephritis in a Nonimmunocompromised Person. Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 2016, 67, 660–663. [Google Scholar] [CrossRef]
  92. Kumar, K.; Kumar, H.K.; Manjunath, V.; Umesh, L. Pleural effusion: A rare complication of co-infection of hepatitis A and hepatitis E. Ann. Trop. Med. Public Health 2012, 5, 532. [Google Scholar] [CrossRef]
  93. Hillebrandt, K.H.; Arsenic, R.; Hofmann, J.; Eurich, D.; Gül, S.; Strücker, B.; Sauer, I.M.; Pratschke, J.; Stockmann, M.; Raschzok, N. Acute Graft Dysfunction 17 Years After Liver Transplant: A Challenging Clinical and Histologic Manifestation of Hepatitis E. Exp. Clin. Transplant. Off. J. Middle East Soc. Organ Transplant. 2018, 16, 348–351. [Google Scholar] [CrossRef]
  94. Corneillie, L.; Banda, D.H.; Meuleman, P. Animal Models for Hepatitis E virus. Viruses 2019, 11, 564. [Google Scholar] [CrossRef] [Green Version]
  95. Li, W.; Sun, Q.; She, R.; Wang, D.; Duan, X.; Yin, J.; Ding, Y. Experimental infection of Mongolian gerbils by a genotype 4 strain of swine hepatitis E virus. J. Med. Virol. 2009, 81, 1591–1596. [Google Scholar] [CrossRef] [PubMed]
  96. Hong, Y.; He, Z.J.; Tao, W.; Fu, T.; Wang, Y.K.; Chen, Y. Experimental infection of Z:ZCLA Mongolian gerbils with human hepatitis E virus. World J. Gastroenterol. 2015, 21, 862–867. [Google Scholar] [CrossRef]
  97. Xu, L.D.; Zhang, F.; Chen, C.; Peng, L.; Luo, W.T.; Chen, R.; Xu, P.; Huang, Y.W. Revisiting the Mongolian Gerbil Model for Hepatitis E Virus by Reverse Genetics. Microbiol. Spectr. 2022, 10, e0219321. [Google Scholar] [CrossRef]
  98. Soomro, M.H.; Shi, R.; She, R.; Yang, Y.; Wang, T.; Wu, Q.; Li, H.; Hao, W. Molecular and structural changes related to hepatitis E virus antigen and its expression in testis inducing apoptosis in Mongolian gerbil model. J. Viral Hepat. 2017, 24, 696–707. [Google Scholar] [CrossRef] [PubMed]
  99. Yu, W.; Hao, X.; Li, Y.; Yang, C.; Li, Y.; He, Z.; Huang, F. Vertical transmission of hepatitis E virus in pregnant rhesus macaques. Sci. Rep. 2020, 10, 17517. [Google Scholar] [CrossRef] [PubMed]
  100. Longer, C.F.; Denny, S.L.; Caudill, J.D.; Miele, T.A.; Asher, L.V.S.; Myint, K.S.A.; Huang, C.-C.; Engler, W.F.; LeDuc, J.W.; Binn, L.N.; et al. Experimental Hepatitis E: Pathogenesis in Cynomolgus Macaques (Macaca fascicularis). J. Infect. Dis. 1993, 168, 602–609. [Google Scholar] [CrossRef]
  101. Bottino, F.O.; Gardinali, N.R.; Salvador, S.B.S.; Figueiredo, A.S.; Cysne, L.B.; Francisco, J.S.; de Oliveira, J.M.; Machado, M.P.; Pinto, M.A. Cynomolgus monkeys (Macaca fascicularis) experimentally and naturally infected with hepatitis E virus: The bone marrow as a possible new viral target. PLoS ONE 2018, 13, e0205039. [Google Scholar] [CrossRef] [Green Version]
  102. Gardinali, N.R.; Guimarães, J.R.; Melgaço, J.G.; Kevorkian, Y.B.; Bottino, F.O.; Vieira, Y.R.; da Silva, A.C.; Pinto, D.P.; da Fonseca, L.B.; Vilhena, L.S.; et al. Cynomolgus monkeys are successfully and persistently infected with hepatitis E virus genotype 3 (HEV-3) after long-term immunosuppressive therapy. PLoS ONE 2017, 12, e0174070. [Google Scholar] [CrossRef]
  103. Balayan, M.S.; Usmanov, R.K.; Zamyatina, N.A.; Djumalieva, D.I.; Karas, F.R. Experimental hepatitis E infection in domestic pigs. J. Med. Virol. 1990, 32, 58–59. [Google Scholar] [CrossRef]
  104. Meng, X.J.; Halbur, P.G.; Haynes, J.S.; Tsareva, T.S.; Bruna, J.D.; Royer, R.L.; Purcell, R.H.; Emerson, S.U. Experimental infection of pigs with the newly identified swine hepatitis E virus (swine HEV), but not with human strains of HEV. Arch. Virol. 1998, 143, 1405–1415. [Google Scholar] [CrossRef]
  105. Meng, X.-J.; Halbur, P.G.; Shapiro, M.S.; Govindarajan, S.; Bruna, J.D.; Mushahwar, I.K.; Purcell, R.H.; Emerson, S.U. Genetic and Experimental Evidence for Cross-Species Infection by Swine Hepatitis E Virus. J. Virol. 1998, 72, 9714–9721. [Google Scholar] [CrossRef] [Green Version]
  106. Cao, D.; Cao, Q.M.; Subramaniam, S.; Yugo, D.M.; Heffron, C.L.; Rogers, A.J.; Kenney, S.P.; Tian, D.; Matzinger, S.R.; Overend, C.; et al. Pig model mimicking chronic hepatitis E virus infection in immunocompromised patients to assess immune correlates during chronicity. Proc. Natl. Acad. Sci. USA 2017, 114, 6914–6923. [Google Scholar] [CrossRef] [Green Version]
  107. Liu, B.; Chen, Y.; Zhang, M.; Chen, T.; Zhang, Y.; DanBaZhaXi; Xu, S.; Zhao, Q.; Zhou, E.M. Identification and pathogenicity of hepatitis E Virus from laboratory Bama miniature pigs. BMC Vet. Res. 2022, 18, 99. [Google Scholar] [CrossRef]
  108. Liu, B.; Chen, Y.; Sun, Y.; Nan, Y.; Li, H.; Du, T.; Hiscox, J.A.; Zhao, Q.; Zhou, E.-M. Experimental infection of rabbit with swine-derived hepatitis E virus genotype 4. Vet. Microbiol. 2019, 229, 168–175. [Google Scholar] [CrossRef]
  109. Han, J.; Lei, Y.; Liu, L.; Liu, P.; Xia, J.; Zhang, Y.; Zeng, H.; Wang, L.; Wang, L.; Zhuang, H. SPF rabbits infected with rabbit hepatitis E virus isolate experimentally showing the chronicity of hepatitis. PLoS ONE 2014, 9, e99861. [Google Scholar] [CrossRef] [Green Version]
  110. Xia, J.; Liu, L.; Wang, L.; Zhang, Y.; Zeng, H.; Liu, P.; Zou, Q.; Wang, L.; Zhuang, H. Experimental infection of pregnant rabbits with hepatitis E virus demonstrating high mortality and vertical transmission. J. Viral Hepat. 2015, 22, 850–857. [Google Scholar] [CrossRef]
  111. He, Q.; Zhang, F.; Shu, J.; Li, S.; Liang, Z.; Du, M.; Liu, X.; Liu, T.; Li, M.; Yin, X.; et al. Immunocompromised rabbit model of chronic HEV reveals liver fibrosis and distinct efficacy of different vaccination strategies. Hepatol. Baltim. Md. 2022, 76, 788–802. [Google Scholar] [CrossRef] [PubMed]
  112. Huang, F.; Zhang, W.; Gong, G.; Yuan, C.; Yan, Y.; Yang, S.; Cui, L.; Zhu, J.; Yang, Z.; Hua, X. Experimental infection of Balb/c nude mice with Hepatitis E virus. BMC Infect. Dis. 2009, 9, 93. [Google Scholar] [CrossRef] [Green Version]
  113. Yang, C.; Hao, X.; Li, Y.; Long, F.; He, Q.; Huang, F.; Yu, W. Successful Establishment of Hepatitis E Virus Infection in Pregnant BALB/c Mice. Viruses 2019, 11, 451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Maneerat, Y.; Clayson, E.T.; Myint, K.S.; Young, G.D.; Innis, B.L. Experimental infection of the laboratory rat with the hepatitis E virus. J. Med. Virol. 1996, 48, 121–128. [Google Scholar] [CrossRef]
  115. Sridhar, S.; Wu, S.; Situ, J.; Shun, E.H.-K.; Li, Z.; Zhang, A.J.-X.; Hui, K.; Fong, C.H.-Y.; Poon, V.K.-M.; Chew, N.F.-S.; et al. A small animal model of chronic hepatitis E infection using immunocompromised rats. JHEP Rep. 2022, 4, 100546. [Google Scholar] [CrossRef] [PubMed]
  116. Heaton, N.S.; Randall, G. Multifaceted roles for lipids in viral infection. Trends Microbiol. 2011, 19, 368–375. [Google Scholar] [CrossRef]
  117. Figlerowicz, M.; Alejska, M.; Kurzyńska-Kokorniak, A.; Figlerowicz, M. Genetic variability: The key problem in the prevention and therapy of RNA-based virus infections. Med. Res. Rev. 2003, 23, 488–518. [Google Scholar] [CrossRef]
  118. Lauring, A.S.; Andino, R. Quasispecies theory and the behavior of RNA viruses. PLoS Pathog. 2010, 6, e1001005. [Google Scholar] [CrossRef] [Green Version]
  119. Cao, D.; Meng, X.J. Molecular biology and replication of hepatitis E virus. Emerg. Microbes Infect. 2012, 1, e17. [Google Scholar] [CrossRef]
  120. Ropp, S.; Tam, A.; Beames, B.; Purdy, M.; Frey, T. Expression of the hepatitis E virus ORF1. Arch. Virol. 2000, 145, 1321–1337. [Google Scholar] [CrossRef]
  121. Sehgal, D.; Thomas, S.; Chakraborty, M.; Jameel, S. Expression and processing of the Hepatitis E virus ORF1 nonstructural polyprotein. Virol. J. 2006, 3, 38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Suppiah, S.; Zhou, Y.; Frey, T.K. Lack of processing of the expressed ORF1 gene product of hepatitis E virus. Virol. J. 2011, 8, 245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Parvez, M.K. Molecular characterization of hepatitis E virus ORF1 gene supports a papain-like cysteine protease (PCP)-domain activity. Virus Res. 2013, 178, 553–556. [Google Scholar] [CrossRef] [PubMed]
  124. Perttilä, J.; Spuul, P.; Ahola, T. Early secretory pathway localization and lack of processing for hepatitis E virus replication protein pORF1. J. Gen. Virol. 2013, 94, 807–816. [Google Scholar] [CrossRef]
  125. Paliwal, D.; Panda, S.K.; Kapur, N.; Varma, S.P.K.; Durgapal, H. Hepatitis E virus (HEV) protease: A chymotrypsin-like enzyme that processes both non-structural (pORF1) and capsid (pORF2) protein. J. Gen. Virol. 2014, 95, 1689–1700. [Google Scholar] [CrossRef]
  126. Spall, V.E.; Shanks, M.; Lomonossoff, G. Polyprotein processing as a strategy for gene expression in RNA viruses. In Seminars in Virology; Academic Press: Cambridge, MA, USA, 1997; pp. 15–23. [Google Scholar]
  127. Smith, D.B.; Vanek, J.; Ramalingam, S.; Johannessen, I.; Templeton, K.; Simmonds, P. Evolution of the hepatitis E virus hypervariable region. J. Gen. Virol. 2012, 93, 2408. [Google Scholar] [CrossRef] [Green Version]
  128. Todt, D.; Gisa, A.; Radonic, A.; Nitsche, A.; Behrendt, P.; Suneetha, P.V.; Pischke, S.; Bremer, B.; Brown, R.J.; Manns, M.P. In vivo evidence for ribavirin-induced mutagenesis of the hepatitis E virus genome. Gut 2016, 65, 1733–1743. [Google Scholar] [CrossRef] [Green Version]
  129. Debing, Y.; Gisa, A.; Dallmeier, K.; Pischke, S.; Bremer, B.; Manns, M.; Wedemeyer, H.; Suneetha, P.V.; Neyts, J. A mutation in the hepatitis E virus RNA polymerase promotes its replication and associates with ribavirin treatment failure in organ transplant recipients. Gastroenterology 2014, 147, 1008–1011.e1007. [Google Scholar] [CrossRef]
  130. Debing, Y.; Ramière, C.; Dallmeier, K.; Piorkowski, G.; Trabaud, M.-A.; Lebossé, F.; Scholtès, C.; Roche, M.; Legras-Lachuer, C.; de Lamballerie, X. Hepatitis E virus mutations associated with ribavirin treatment failure result in altered viral fitness and ribavirin sensitivity. J. Hepatol. 2016, 65, 499–508. [Google Scholar] [CrossRef]
  131. Todt, D.; Walter, S.; Brown, R.J.; Steinmann, E. Mutagenic effects of ribavirin on hepatitis E virus—Viral extinction versus selection of fitness-enhancing mutations. Viruses 2016, 8, 283. [Google Scholar] [CrossRef] [Green Version]
  132. Lhomme, S.; Nicot, F.; Jeanne, N.; Dimeglio, C.; Roulet, A.; Lefebvre, C.; Carcenac, R.; Manno, M.; Dubois, M.; Peron, J.-M. insertions and duplications in the polyproline region of the hepatitis E virus. Front. Microbiol. 2020, 11, 331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. van Tong, H.; Hoan, N.X.; Wang, B.; Wedemeyer, H.; Bock, C.T.; Velavan, T.P. Hepatitis E Virus Mutations: Functional and Clinical Relevance. EBioMedicine 2016, 11, 31–42. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Ji, H.; Chen, S.; He, Q.; Wang, W.; Gong, S.; Qian, Z.; Zhang, Y.; Wei, D.; Yu, W.; Huang, F. The different replication between nonenveloped and quasi-enveloped hepatitis E virus. J. Med. Virol. 2021, 93, 6267–6277. [Google Scholar] [CrossRef] [PubMed]
  135. Yin, X.; Ambardekar, C.; Lu, Y.; Feng, Z. Distinct Entry Mechanisms for Nonenveloped and Quasi-Enveloped Hepatitis E Viruses. J. Virol. 2016, 90, 4232–4242. [Google Scholar] [CrossRef] [Green Version]
  136. Horvatits, T.; Wißmann, J.E.; Johne, R.; Groschup, M.H.; Gadicherla, A.K.; Schulze Zur Wiesch, J.; Eiden, M.; Todt, D.; Reimer, R.; Dähnert, L.; et al. Hepatitis E virus persists in the ejaculate of chronically infected men. J. Hepatol. 2021, 75, 55–63. [Google Scholar] [CrossRef]
  137. Anheyer-Behmenburg, H.E.; Szabo, K.; Schotte, U.; Binder, A.; Klein, G.; Johne, R. Hepatitis E Virus in Wild Boars and Spillover Infection in Red and Roe Deer, Germany, 2013–2015. Emerg. Infect. Dis. 2017, 23, 130–133. [Google Scholar] [CrossRef] [Green Version]
  138. De Sabato, L.; Amoroso, M.G.; Ianiro, G.; Esposito, C.; De Grossi, L.; Fusco, G.; Barone, A.; Martini, E.; Ostanello, F.; Di Bartolo, I. Detection of Hepatitis E Virus in Livers and Muscle Tissues of Wild Boars in Italy. Food Env. Virol 2020, 12, 1–8. [Google Scholar] [CrossRef]
  139. Ferri, G.; Lauteri, C.; Festino, A.R.; Piccinini, A.; Olivastri, A.; Vergara, A. Hepatitis E Virus Detection in Hunted Wild Boar Liver and Muscle Tissues in Central Italy. Microorganisms 2022, 10, 1628. [Google Scholar] [CrossRef]
  140. Grange, Z.L.; Goldstein, T.; Johnson, C.K.; Anthony, S.; Gilardi, K.; Daszak, P.; Olival, K.J.; O’Rourke, T.; Murray, S.; Olson, S.H.; et al. Ranking the risk of animal-to-human spillover for newly discovered viruses. Proc. Natl. Acad. Sci. USA 2021, 118, e2002324118. [Google Scholar] [CrossRef]
Figure 1. Summary of neurological and musculoskeletal clinical disorders presented in relation to HEV infection. The numbers are extracted from case reports, retrospective studies, case–control studies, case series, and cross-sectional studies. Every disorder and their references are listed in Table 1.
Figure 1. Summary of neurological and musculoskeletal clinical disorders presented in relation to HEV infection. The numbers are extracted from case reports, retrospective studies, case–control studies, case series, and cross-sectional studies. Every disorder and their references are listed in Table 1.
Zoonoticdis 03 00007 g001
Figure 2. Summary of the cardiovascular clinical disorders presented in relation to HEV infection. The numbers are isolated from case reports, retrospective studies, case-control studies, case series, and cross-sectional studies. Every reported disorder and their references are listed in Table 2.
Figure 2. Summary of the cardiovascular clinical disorders presented in relation to HEV infection. The numbers are isolated from case reports, retrospective studies, case-control studies, case series, and cross-sectional studies. Every reported disorder and their references are listed in Table 2.
Zoonoticdis 03 00007 g002
Figure 3. Summary of the digestive clinical disorders presented in relation to HEV infection. The numbers are isolated from case reports, retrospective studies, case-control studies, case series, and cross-sectional studies. Every disorder and their references were listed in Table 3.
Figure 3. Summary of the digestive clinical disorders presented in relation to HEV infection. The numbers are isolated from case reports, retrospective studies, case-control studies, case series, and cross-sectional studies. Every disorder and their references were listed in Table 3.
Zoonoticdis 03 00007 g003
Table 1. Nervous and musculoskeletal system disorders related to HEV infection.
Table 1. Nervous and musculoskeletal system disorders related to HEV infection.
SNClinical DiseaseNumber of PatientsReferences
1Neuralgic amyotrophy102[44,45,46,47,48,49,50,51,52,53,54,55,56]
2Guillain–Barre syndrome36[39,57,58,59,60,61,62,63,64,65]
3Myasthenia gravis1[66]
4Polyneuromyopathy2[57,67]
5Mononeuritis multiplex6[39]
6Meningo-radiculitis5[39,68]
7Cerebral ischemia5[46,65]
8Epilepsy2[46]
9Encephalitis5[46,48,65,69]
10Facial Nerve Palsy3[46,70,71]
11Encephalopathy1[72]
12Encephalitic Parkinsonism1[73]
13Transverse myelitis1[74]
14Peripheral neuropathy3[57,65,75]
15Vestibular neuritis1[57]
16Myositis1[76]
Table 2. Cardiovascular system disorders related to HEV infection.
Table 2. Cardiovascular system disorders related to HEV infection.
SNClinical DiseaseNumber of PatientsReferences
1Cardiac arrhythmia1[57]
2Myocarditis2[77,78]
3Anemia1[79]
4Thrombocytopenia13[57,79]
5Lymphocytosis14[57]
6Lymphopenia8[57]
7Leukocytosis1[30]
8Massive hemolysis1[80]
9Monoclonal gammopathy17[57]
10Cryoglobulinemia51[81]
11Long QT syndrome and Torsades de pointes1[82]
12Metabolic acidosis1[30]
13Acute myeloid leukemia2[57]
Table 3. Digestive system disorders related to HEV infection.
Table 3. Digestive system disorders related to HEV infection.
SNClinical DiseaseNumber of PatientsReferences
1Pancreatitis22[30,83,84,85,86]
2Pancreatic Pseudocyst1[87]
3Acalculous cholecystitis1[33]
Table 4. Other body systems disorders related to HEV infection.
Table 4. Other body systems disorders related to HEV infection.
SNClinical DiseaseBody SystemNumber of PatientsReferences
1Autoimmune ThyroiditisEndocrine1[88]
2Cutaneous T-Cell lymphoproliferative disorderIntegumentary1[89]
3Acute Kidney InjuryRenal2[30,80]
4Cryoglobulinemic membranoproliferative glomerulonephritisRenal2[90,91]
5Pleural EffusionRespiratory1[92]
6Acute Graft DysfunctionImmune1[93]
7InfertilityReproductive52[27]
Table 5. List of animal models used to study HEV extrahepatic replication.
Table 5. List of animal models used to study HEV extrahepatic replication.
SNAnimal ModelHEV Genotype/StrainBody System InvolvedExtrahepatic SitesDemonstration of Viral Replication ViaReferences
1Mongolian GerbilHuman Gt4Digestive, Renal, LymphaticKidneys,
Spleen,
Small Intestine
IHC and positive strand RT-qPCR (not confirmatory)[95]
Gt1 (clinical human sample)Digestive, Renal, LymphaticSpleen,
Kidney
Positive strand RT-qPCR (not confirmatory)[96]
Human Gt3Digestive, LymphaticSpleenIHC and positive strand RT-qPCR (not confirmatory)[97]
Swine HEV Gt4 (CHN-HB-HD-L2)Digestive,
Renal,
Lymphatic,
Respiratory,
Reproductive
Brain,
Spinal cord,
Spleen,
Peripheral blood monocytes,
Pancreas,
Lung,
Lymph node,
Kidney,
Duodenum,
Jejunum,
Ileum,
Colon, Placenta,
Urine
IHC and negative strand RT-qPCR[98]
2Pregnant Rhesus MacaquesGt4 (KM01)Digestive, Fetal (Digestive, Renal)Spleen,
Kidneys,
Intestine
IHC and positive strand RT-qPCR (not confirmatory)[99]
Cynomolgus MacaquesHuman derived HEVDigestiveNot listedPositive strand RT-qPCR (not confirmatory)[100]
Human Gt3Digestive, Nervous, LymphaticsBone marrowIHC and negative strand RT-qPCR[101]
Immunocompromised Cynomolgus MonkeyGt3Digestive, LymphaticSpleen,
Duodenum,
Colon,
Lymph node,
Pancreas
IHC and negative strand RT-qPCR[102]
3PigunknownDigestiveNot listedVisualization of virus-like particles[103]
Swine HEVDigestive, RespiratoryNot listedPositive strand RT-qPCR (not confirmatory)[104]
US2DigestiveNot listedPositive strand RT-qPCR (not confirmatory)[105]
Immunosuppressed pigsHuman HEV (US2 strain), Gt3Digestive, ImmuneNot listedPositive strand RT-qPCR (not confirmatory)[106]
4Miniature pigsGt3Digestive,
Endocrine, Respiratory, Renal,
Nervous
Pancreas,
Kidney,
Brain,
Peyer’s patches,
Lungs (bronchioles)
IHC and positive strand RT-qPCR (not confirmatory)[31,107]
5RabbitSwine derived HEV Gt4Digestive, Lymphatic,SpleenNegative strand RT-qPCR[108]
Rabbit derived HEV Gt3Digestive, lymphaticSpleenNegative strand RT-qPCR[108]
SPF rabbitsRabbit HEVDigestive,
Renal,
Lymphatic,
Respiratory,
Nervous
Stomach,
Duodenum,
Kidney,
Bile,
Lung,
Bladder,
Brain
IHC and negative strand RT-qPCR[109]
Swine Gt4Mild digestiveNot listedIHC and negative strand RT-qPCR[109]
Pregnant rabbitsRabbit HEV (CHN-BJ-R14)Digestive, ReproductivePlacentaIHC and negative strand RT-qPCR[110]
Rabbit gt3 (KOR-Rb-1)Digestive, ReproductiveUterusPositive strand RT-qPCR (not confirmatory)[28]
Immunosuppressed RabbitRabbit derived HEV-3raDigestive, Renal, NervousKidney, Duodenum,
Jejunum,
Cecum,
Colon,
Urine,
Cerebrospinal fluid
Positive strand RT-qPCR (not confirmatory)[111]
6BALB/c nude miceSwine HEV Gt4Digestive, Lymphatic, RenalSpleen,
Kidney,
Jejunum,
Ileum,
Colon
IHC and positive strand RT-qPCR (not confirmatory)[112]
Pregnant BALB/c miceSwine derived Gt4 (KM01)Digestive, Reproductive, Renal, Placenta and neonatal liverSpleen, Kidney,
Colon, Uterus,
Placenta
IHC and negative strand RT-qPCR[113]
7RatHuman feces derived (TK-037/92)Digestive, LymphaticSpleen,
Mesenteric lymph nodes,
Small intestine
IHC and positive strand RT-qPCR (not confirmatory)[114]
Immunocompromised ratsHuman derived rat strain (CCY)DigestiveNot listedIHC and positive strand RT-qPCR (not confirmatory)[115]
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

Yadav, K.K.; Kenney, S.P. Extrahepatic Replication Sites of Hepatitis E Virus (HEV). Zoonotic Dis. 2023, 3, 68-84. https://doi.org/10.3390/zoonoticdis3010007

AMA Style

Yadav KK, Kenney SP. Extrahepatic Replication Sites of Hepatitis E Virus (HEV). Zoonotic Diseases. 2023; 3(1):68-84. https://doi.org/10.3390/zoonoticdis3010007

Chicago/Turabian Style

Yadav, Kush Kumar, and Scott P. Kenney. 2023. "Extrahepatic Replication Sites of Hepatitis E Virus (HEV)" Zoonotic Diseases 3, no. 1: 68-84. https://doi.org/10.3390/zoonoticdis3010007

Article Metrics

Back to TopTop