Animal Hepatitis

Several animate being hepatitis viruses, including swine HEV and the avian HEV, are closely related and too members of the Hepeviridae family.

From: Pathobiology of Human being Disease , 2014

Viral Hepatitis

S.A. Weinman , R. Taylor , in Pathobiology of Human Disease, 2014

Virology

The virus is a nonenveloped, unmarried-stranded positive-sense RNA virus that was classified as its own family unit, Hepeviridae. Several animal hepatitis viruses, including swine HEV and the avian HEV, are closely related and also members of the Hepeviridae family. The virus has four genotypes. Genotypes one and 2 cause exclusively man infection. Genotypes 3 and 4 infect animals also and can be transmitted between humans and other animate being species. The viral genome encodes 3 ORFs. The ORF1 polyprotein encodes a methyltransferase, protease, helicase, and polymerase. ORF2 encodes the capsid poly peptide and ORF3 encodes a small protein of unknown function that is involved in virion production.

Read full chapter

URL:

https://www.sciencedirect.com/science/article/pii/B978012386456704209X

Immune Pathogenesis of Viral Hepatitis B and C

Kyong-Mi Chang , in Zakim and Boyer's Hepatology (Sixth Edition), 2012

HBV equally a Stealth Virus Evading Innate Immune Recognition During Acute Infection

Information technology is difficult to study innate immune responses in human being viral hepatitis due to a lack of user-friendly small animal model to sample the relevant compartments (e.thou., site of virus entry, draining lymph nodes, target cells) in a timely style. 41 Still, a remarkable lack of early innate immune induction was demonstrated in acutely HBV-infected chimpanzees. 81 In these animals, HBV DNA and HBcAg became first detectable in the liver at 3 to 5 weeks after inoculation, but without associated liver injury or blazon I IFN response. The onset of astute hepatitis with elevated ALT action occurred several weeks thereafter, coinciding with the detection of T-cell markers (CD3, IFN-γ) in the liver, followed afterward by HBV clearance. These findings suggest that: (ane) HBV is noncytopathic; (two) T cells mediate liver injury; and (3) T cells likewise mediate viral clearance. Further analysis of global gene expression contour showed that no genes (including IFN-inducible genes) correlated with intrahepatic HBV DNA levels during astute hepatitis B, whereas 110 hepatic genes were associated with HBV clearance—the majority of which were associated with T cells. In acutely HBV-infected patients, type I IFN was barely detectable and no higher than those in uninfected controls. 82 These findings in chimpanzees and patients suggested that HBV is a "stealth virus" that spreads without detection by the innate sensing machinery during early infection ( Fig. 8-2, A-C ).

Despite the apparent stealth behavior, HBV infection is resolved in most acutely HBV-infected adults. Thus the ability for early innate allowed abstention cannot explain the long-term virologic event. Nevertheless, it probably does let for the initial establishment of HBV infection because HBV is readily suppressed by innate allowed components including IFN-α, 83 TLR, NK, NKT, and antigen presenting cells. 36,50,84 On the other paw, HBV can activate the cellular components of innate immune response, such as Kupffer cells and NK/NKT cells. 85-87 For example, dynamic changes in NK and NKT prison cell activation and part were observed during acute HBV infection. 88 An early production of immune regulatory cytokine IL-10 was also observed in patients with acute hepatitis B in association with adulterate NK and T-cell response. 82 Thus HBV may be relatively merely not admittedly invisible to the innate immune system, avoiding blazon I IFN merely non other innate immune cells during astute infection.

Read total chapter

URL:

https://www.sciencedirect.com/science/article/pii/B9781437708813000085

Hepatitis E

S.M. Sarin , Manoj Kumar , in Zakim and Boyer'due south Hepatology (6th Edition), 2012

Incidence and Prevalence and Worldwide Disease Patterns

Worldwide, two geographic patterns tin can be differentiated: (1) owned regions or areas of loftier HEV prevalence, in which major outbreaks and a substantial number of sporadic cases occur; and (2) not–endemic regions, in which HEV accounts for a few cases of acute viral hepatitis, mainly amid travelers to endemic regions ( Fig. 33-v ).

HEV in Owned Regions

In such regions epidemics of hepatitis E occur frequently. These epidemics are unremarkably separated by a few years. Such outbreaks have been observed in the Indian subcontinent, China, Southeast and Fundamental Asia, the Eye East, and the northern and western parts of Africa. In North America (United mexican states), two small-scale outbreaks were reported during 1986 to 1987, just no further outbreaks have since been reported. The outbreaks are often large, and affect several hundred to several thou persons. 1 Most reported outbreaks take been related to consumption of fecally contaminated drinking h2o. Their fourth dimension course varies from unimodal outbreaks, which last a few weeks, to prolonged, multipeaked epidemics, with a duration of more than 1 year. The latter represent standing water contamination. The outbreaks frequently follow heavy rainfall and floods, which create conditions that favor mixing of man excreta with sources of drinking water. Some outbreaks have occurred in hot and dry summer months, possibly a event of macerated water flow in rivers leading to an increased concentration of fecal contaminants. In Southeast Asia, recurrent epidemics take been shown to be associated with disposal of homo excreta into rivers and subsequent utilise of water from the same river for drinking, cooking, and personal hygiene. 57 These practices provide conditions that permit continuous fecal contamination of water. Outbreaks of hepatitis E take occurred in underdeveloped urban areas with leaky water pipes passing through soil that is contaminated with sewage. Intermittent water supply in these areas leads to a negative pressure in pipes during periods of no flow, assuasive inward suction of contaminants. 58 Although the dissemination of HEV infection through contagion of food may be possible, few outbreaks related to food-borne manual have been reported from disease-endemic areas. This may exist due to a relatively long incubation period, which makes it difficult to institute a relationship betwixt consumption of a particular food and occurrence of disease. 59

Overall attack rates during hepatitis Eastward outbreaks have ranged from 1% to 15%. The rates are highest among young adults (iii% to xxx%); the reason for this is non clear. Lower attack rates amongst children are related probably to a college proportion of asymptomatic infections than to rarity of infection. Males outnumber females in most outbreaks; this may be due either to their greater take a chance of exposure to HEV infection or to a greater propensity for clinical disease once infected. 59 Hepatitis E outbreaks have characteristically been associated with a loftier attack rate and mortality amongst significant women. The charge per unit of development of acute liver failure among those with symptomatic hepatitis East was besides college among significant women. Once astute liver failure appears, the death charge per unit may be no unlike amidst pregnant women with hepatitis E than in those with other causes of severe liver injury (see Meaning Women subsequently). Because HEV infection during pregnancy is associated with a high attack charge per unit and risk of severe disease, pregnant women are overrepresented among case series of women with liver failure in the endemic areas, and also amid fatal cases during hepatitis E outbreaks. 59

In disease-endemic areas, HEV infection accounts for a large proportion of acute sporadic hepatitis in all age groups. In India HEV infection is the well-nigh common cause of acute sporadic hepatitis and accounts for upwardly to 70% of such cases amidst adults. 59 In these regions, patients with desultory hepatitis E closely resemble those of epidemic hepatitis East in age distribution, severity and elapsing of illness, propensity for worse prognosis among significant women, and absenteeism of chronic sequelae. The road of acquisition of infection in near patients with desultory hepatitis E is unclear. However, given the high potential for fecal contamination of water and food in these areas, these sources are most likely. HEV genomic sequences could be isolated from virtually twoscore% of sewage specimens obtained through all seasons from a large drain in a big Indian city. 60 This indicates ubiquitous circulation of HEV in the population, even in the absenteeism of a disease outbreak.

Unlike several other enterically transmitted infections, person-to-person transmission of HEV from either epidemic or sporadic cases is distinctly uncommon. 61 The exact reason for this is unknown. During outbreaks, secondary attack rates amidst household contacts of patients with hepatitis E take been every bit low as 0.7% to 2.2%. This is much lower than the 50% to 75% infection rate among susceptible household contacts of hepatitis A cases. Even with multiple cases in one family, the fourth dimension interval betwixt cases is curt, indicating a shared main h2o-borne infection rather than person-to-person manual. 59

Maternal–fetal transmission of HEV infection has been reported (run across Modes of Transmission, subsequently). In regions owned for hepatitis E, the presence of HEV viremia among healthy blood donors and manual of this infection to transfusion recipients have been documented. However, the contribution of such transmission to the overall illness brunt remains unclear.

Reservoirs of HEV in Endemic Regions

In affliction-endemic areas, the reservoir of HEV responsible for maintaining the disease in a population has not been clearly adamant. Protracted viremia and prolonged fecal shedding of HEV have been suggested. However, viral shedding in feces lasts for a brusk period, making this possibility unlikely. Another potential reservoir of the virus may exist a continuously circulating pool of individuals with subclinical HEV infection. In an experimental cynomolgus macaque model, HEV-infected animals that lacked biochemical testify of liver injury were institute to excrete large amounts of viable and infectious HEV. 62 Similar fecal shedding of the virus by persons with subclinical HEV infection could lead to continuous maintenance of a source of infection in a disease-owned area, somewhat similar to the situation that existed with poliovirus in areas where information technology was endemic. This pool of infection could, in plow, atomic number 82 to periodic contamination of drinking water supplies.

The issue regarding the beingness of an fauna reservoir in disease-endemic regions remains unresolved. The zoonotic hypothesis for transmission of HEV is based primarily on the following factors: (ane) the high prevalence of anti-HEV antibodies in several animal species; (2) the isolation of HEV genomic sequences from pigs; and (3) the genomic sequence homology between human and animal HEV isolates. Even so, near of the supporting genomic data take been from not–endemic regions. In dissimilarity, data from owned regions are conflicting. Isolates from animals and sporadic human cases take belonged to the same genotype (genotype 4) in Cathay and Vietnam. In India, however, fauna isolates have all been members of genotype four and human being isolates genotype 1. Genotype 1 HEV, which is responsible for the large majority of cases in all endemic countries, has never been isolated from pigs. Also, in experimental studies, genotype 1 virus is unable to infect pigs. Thus zoonotic transmission may not be a major style of distribution in these areas, in item for the widely prevalent genotype i HEV. 59

Thus it appears that in the regions where hepatitis Due east is endemic, the infection is acquired from either an environmental or a human reservoir through poor general sanitation, contaminated drinking water supplies, and lack of attention to personal hygiene. Further data are necessary earlier zoonotic transmission of HEV can exist implicated in these regions.

HEV in Non-Endemic Regions

In non-owned regions, where outbreaks have non been reported, the affliction accounts for only a minority of reported cases of acute viral hepatitis. Until a few years ago, most such cases were found to be related to travel to disease-owned areas. Nonetheless, in recent years isolated cases or small series of cases related to autochthonous transmission of hepatitis E in these regions have been recorded in the United States, Europe (including the United kingdom, France, the Netherlands, Republic of austria, Espana, and Hellenic republic), and developed countries of Asia-Pacific (Japan, Taiwan, Hong Kong, Australia). In a series of xl cases with hepatitis East identified in the United Kingdom, the illness showed seasonal variations with peaks in spring and summer and no cases during Nov and Dec. 63 In the United Kingdom, the disease appeared to exist more than common among residents of littoral and estuarine areas. 64 The mode of transmission in most of these cases could not be identified, although zoonotic distribution has been proposed (see HEV Infection as a Zoonosis, afterwards). Table 33-1 shows epidemiologic differences in hepatitis East in illness-endemic and non-endemic regions.

Read full chapter

URL:

https://www.sciencedirect.com/science/article/pii/B9781437708813000334

INFECTIONS WITH SPECIFIC MICROORGANISMS

Anthony E. Fiore , Beth P. Bong , in Feigin and Cherry's Textbook of Pediatric Infectious Diseases (Sixth Edition), 2009

ROUTES OF TRANSMISSION

Routes of HAV manual are determined by the timing and location of virus replication, circulation, and excretion during infection. HAV replicates in the liver, is excreted in bile, and is found in highest concentration in stool (upward to 10eight infectious particles per milliliter). 311 The highest concentration in stool occurs during the 2-week menses earlier jaundice develops or liver enzymes become elevated, followed by a rapid decline later on jaundice appears (Fig. 180-1). 113, 294, 311 Children and infants may shed HAV for longer periods than adults do. Through the use of polymerase chain reaction (PCR) to dilate viral nucleic acid, HAV RNA has been detected in the stool of infected neonates for as long as six months afterward infection, and some studies have shown excretion in older children and adults 1 to 3 months after the onset of clinical illness. 156, 268, 271, 361 Chronic shedding of HAV does not occur; however, virus may be present in stool during relapsing illness (see the section on relapsing hepatitis A). 293 Viremia occurs during the prodromal phase of infection and extends through the flow of liver enzyme height (see Fig. 180-i), with virus concentrations several orders of magnitude lower than those in stool. 36, 64, 178, 191 In experimentally infected animals, HAV can be detected in saliva during periods of peak excretion in stool, 64 but manual by saliva has not been demonstrated.

Detection of HAV antigen in stool past enzyme immunoassay or detection of HAV RNA in serum or stool past PCR does non demonstrate that an infected person is infectious considering these assays may detect defective as well as infectious viral particles. Nucleic acid amplification past immunocapture PCR requires the presence of intact virus, 39, 164 and HAV RNA may be detected in stool for months with immunocapture PCR. However, the menstruum of infectivity appears to be shorter than the menses when HAV RNA tin can be detected in stool. Information from epidemiologic studies advise that height infectivity occurs during the 2 weeks before the onset of symptoms. For practical purposes, both children and adults with hepatitis A tin exist assumed to exist non-infectious 1 week afterward jaundice appears.

Because of the high concentration of virus in the stool of infected persons, HAV transmission occurs primarily past the fecal-oral route, usually by person-to-person manual in households and extended-family settings and betwixt sexual contacts. 299 Person-to-person transmission results in loftier rates of infection in young children in developing countries and has been the predominant fashion of transmission in the United States, particularly during community-broad outbreaks, as well equally in outbreaks in childcare centers. 22, 32 HAV can remain infectious in the surround, 214 and fecal contagion of food or water can result in mutual-source outbreaks. HAV has been transmitted by transfusion, merely such transmission occurs rarely because the claret donation must occur during the early prodromal phase of the illness or from an asymptomatic person who is viremic. 191 In the United states of america, nucleic acid amplification tests such as PCR now are applied to screening of source plasma used for the industry of plasma-derived products. These assays are sufficiently sensitive to remove nigh units that have HAV, and serosurveillance for HAV infections among clotting factor recipients in the United States indicates that HAV infections are now rare in this hazard grouping. 49

Two published case reports have described intrauterine transmission of HAV during the outset trimester that resulted in fetal meconium peritonitis. 190, 215 Afterward delivery, both infants were found to take a perforated ileum. The gamble of transmission to newborns past pregnant women in whom hepatitis A develops in the third trimester of pregnancy appears to be low. 317 However, newborns who larn HAV infection in this fashion or from a transfusion usually are asymptomatic, and the infection is detected by the development of hepatitis A in hospital staff or other persons having contact with the infant. 271, 342

Read full chapter

URL:

https://www.sciencedirect.com/scientific discipline/article/pii/B9781416040446501850

Infectious Agents and Cancer

Peter M. Howley , in The Molecular Basis of Cancer (4th Edition), 2015

Hepatitis B Virus (Virus-Host Interactions)

HBV is a member of a grouping of the hepadnaviruses (for hepatotropic DNA viruses). HBV is the merely human member of this group of viruses. Other members of this group include the woodchuck hepatitis virus (WHV), the Beechey footing squirrel hepatitis virus (GSHV), the Pekin duck hepatitis B virus (DHBV), and the gray heron hepatitis virus. These viruses share a similar structure, and each is hepatotropic, leading to persistent viral infections of the liver. The animal hepatitis viruses have been very of import contributors to our understanding of the molecular biology of these viruses. Of the hepadnaviruses, just HBV and WHV accept been associated with chronic active hepatitis and HCC.

The reader is referred to the Fields Virology chapter on the molecular biology of the hepadnaviruses for details on the virus and aspects of virus/host cell interactions. 110 Hepatitis B viral particles contain small, circular Deoxyribonucleic acid molecules that are only partially double stranded. The DNA consists of a long strand with a abiding length of 3220 bases and a shorter strand that varies in length from 1700 to 2800 bases in unlike molecules. A map of the HBV Deoxyribonucleic acid genome is shown in Figure 6-four. The virion particles comprise a Deoxyribonucleic acid polymerase activity that is capable of repairing the single-stranded Dna region to make two fully double-stranded molecules, each approximately 3220 bases in length. For this reaction, Dna synthesis initiates at the 3′ end of the short strand that, as noted before, is heterogeneous among different DNA molecules. DNA synthesis terminates at the uniquely located 5′ end of the short strand when information technology is reached. The long strand is not a closed molecule only contains a nick at a unique site approximately 300 base of operations pairs from the 5′ end of the short strand.

The HBV genome has four ORFs and encodes four genes. These ORFs are designated as S and pre-S, C, P, and 10. S and pre-S represent two contiguous reading frames and code for the viral surface glycoproteins. C contains the coding sequences for the core structural protein of the nucleocapsid. The P factor encodes the viral polymerase that contains contrary transcriptase activity. The X ORF encodes a basic polypeptide that has transcriptional transactivation properties that tin upregulate the action of hepadnavirus promoters. The overall construction of the genomes of all of the animal hepadnaviruses is quite similar. The WHV and GSHV genomes are approximately 3300 base of operations pairs in size, and that of DHBV is approximately 3000 base pairs in size. The genomic organization of the diverse hepadnaviruses is similar, and there is all-encompassing nucleotide homology among them. The mammalian hepadnaviruses differ from the avian hepadnaviruses in that the avian hepadnaviruses exercise not contain the X region.

HBV Dna can be establish either gratis or integrated in the host chromosome of the hepatocyte. Free HBV DNA represents intermediate forms of replication for the viral genome and can be detected during acute infections and some chronic stages of HPV infection. Integrated sequences are ordinarily found during chronic virus infection and in HCC. The replication mechanism for the hepadnaviruses, kickoff discovered by Summers and Stonemason for DHBV 111 and later confirmed for HBV, is different from that of other Dna viruses. The replication cycle involves a reverse transcription pace resembling that of the retroviruses in that it goes through an RNA copy of the genome as an intermediate in replication. The hepadnaviruses differ from the retroviruses, however, in that retrovirus virions contain RNA and the intermediate class of replication is integrated DNA. The virions of the hepadnaviruses contain Dna and the intermediate replication course is RNA. Also, integration of the hepadnavirus genome every bit a provirus is not a necessary intermediate step for viral genome replication as it is for a retrovirus. The similarity between the retroviruses and the hepadnaviruses extends to the overall genomic arrangement, in which all of the genes are encoded on only one strand. The society of the genes within the retroviruses (gag, pol, and env) is like to their counterparts for the hepadnaviruses (core, polymerase, and surface antigen). Other subtle differences in the transcriptional programs used to generate the messenger RNAs for these different viruses exist. A farther similarity between these viruses is that some members of each group of these viruses encode transcriptional regulatory factors. For HTLV-1, described later in this chapter, the 10 region encodes the transcriptional activator tax as well as the rex gene product involved in messenger RNA transport to the cytoplasm. The Ten genes encoded by the mammalian hepadnaviruses similarly encode a protein that has been extensively studied and shown to accept a variety of activities, including the ability to role as a transcriptional activator. The role of X in the life wheel of the mammalian hepadnaviruses, however, is all the same non well understood. Although at that place have been studies challenge that the X protein has oncogenic properties, the testify implying a direct role for the X protein in HCC is far from compelling. 110

Primary infection with HBV results either in a subclinical infection or acute hepatitis B, depending on the age of the private, amongst other factors. In adults, 95% of such infections resolve, with clearance of virus from the liver and the blood and with lasting amnesty to reinfection. The remaining 5% of infections do not resolve but develop into a persistent hepatitis with a viremia that usually lasts for the life of the host and can accept a variety of pathologic consequences. Many of these persistent infections have petty associated hepatocellular injury. Approximately twenty% to 25% of persistently infected individuals do develop hepatocellular injury, either chronic persistent hepatitis (in which example the inflammation is limited to periportal areas) or chronic active hepatitis (where there is inflammation and hepatocellular necrosis extending outside of the portal areas). Chronic active hepatitis has significant potential for progression to cirrhosis, hepatic failure, and cancer.

Read full chapter

URL:

https://www.sciencedirect.com/science/commodity/pii/B9781455740666000068

Selected Zoonoses

James K. Play a joke on DVM, MS, DACLAM , ... Lesley A. Colby DVM, DACLAM , in Laboratory Animal Medicine (3rd Edition), 2015

F Viral Hepatitis Infections

Many of the nonhuman primate zoonoses causing systemic infections in humans include hepatitis every bit one component of the disease. All the same, of the viral infections that target the liver every bit the primary site of involvement, only hepatitis A virus (HAV) has proven to be a significant zoonotic pathogen in the laboratory animal facility environment. Nonhuman primates are important experimental hosts in viral hepatitis research and have been used to report hepatitis A, B, C, D, and E infections (Vitral et al., 1998). Other viruses known to induce hepatitis in naturally caused infections of laboratory brute species include the coronavirus, mouse hepatitis virus; the adenovirus, infectious canine hepatitis virus; and the hepadnavirus, woodchuck hepatitis virus. None of these are recognized zoonotic agents.

ane Hepatitis A

Reservoir and Incidence

HAV is a human enterovirus belonging to the family unit Picornaviridae. The principal reservoirs for HAV infection are humans, with nonhuman primate infections resulting from contact with infected humans or other infected nonhuman primates. Still, more than than 100 cases of HAV infection in humans have been associated with newly imported chimpanzees (Dienstag et al., 1976; Hinthorn et al., 1974). At that place are also many other nonhuman primate species naturally susceptible to HAV, including tamarins, owl monkeys (Aotus trivirgatus), African green monkeys, cynomolgus (One thousand. fascicularis) and rhesus macaques, and that could serve as sources for human HAV infection (Burgos-Rodriguez, 2011; Lemon et al., 1990; Shevtsova et al., 1988; Wachtman and Mansfield, 2012).

Mode of Manual

HAV tin can be isolated from the blood and is shed in the feces during the prodromal phase of the disease. It is transmitted by the fecal–oral route, well-nigh commonly via contaminated food or water. Aerosol transmission is not suspected (CDCP-NIH, 2009).

Clinical Signs

The disease in nonhuman primates is much less severe than in humans and is frequently subclinical. Clinical disease develops in the chimpanzee, owl monkey, and several marmoset species, and is characterized past malaise, vomiting, jaundice, and elevated serum levels of hepatic enzymes.

The disease in humans varies from a balmy disease lasting less than ii months to a severely debilitating illness lasting up to six months. Following an incubation flow of approximately 1 month, patients experience an precipitous onset of fever, malaise, anorexia, nausea, joint pain, and intestinal discomfort followed within a few days past jaundice (Fig. 28.3). Children frequently have balmy disease without jaundice, whereas HAV infections in older patients may be fulminant and protracted with prolonged convalescence. Withal, protracted HAV infection is considered an acute infection that is ultimately resolved by the patient; a chronic hepatitis A carrier land has never been shown to exist. Infection confers lifelong immunity (ACIP, 2006).

Figure 28.3. Under a magnification of 100×, this hematoxylin and eosin photomicrograph depicts the cytoarchitectural changes constitute in a liver tissue specimen extracted from a hepatitis A patient. In this item view, note that there are several layers of hepatic parenchyma, which are withal recognizable in the midst of massive necrosis. There is no fat change, but there is an extensive inflammatory infiltrate.

Source: PHIL Library ID# 13020.
Diagnosis and Prevention

Infection is diagnosed past sit-in of elevated IgM-specific anti-HAV or total combined IgM and IgG anti-HAV antibodies in the serum or plasma. Alternatively, detection of HAV RNA in the blood or carrion is considered diagnostic of agile infections. The presence of IgG anti-HAV antibodies is useful in detecting previous infection (ACIP, 2006).

A safe, constructive multidose hepatitis A vaccine is available in the United states of america and is recommended for individuals at high take a chance for exposure to HAV infection, such as persons involved with the intendance of nonhuman primates used in experimental HAV infection studies. Passive protection of such persons can also exist undertaken through the intramuscular assistants of specific immune serum globulin (ISG). Passive protection should be given earlier experimental creature HAV infection studies brainstorm because infected animals start shedding HAV at 7–11 days postinoculation and continue shedding for several weeks. Two different dosages of ISG are recommended, each providing differing durations of passive protection (1–2 months versus 3–5 months) (ACIP, 2006). PEP can be administered post-obit suspected virus exposure. PEP recommendations varies with the historic period and prior vaccination history of the exposed individual and may include administration of a single-antigen hepatitis A vaccine and/or ISG inside 2 weeks of exposure (ACIP, 2007). The use of protective clothing, personal hygiene, and appropriate sanitation practices for equipment and facilities volition as well minimize the potential for zoonotic transmission.

2 Hepatitis Due east Virus

Hepatitis E virus (HEV) is unique among the major hepatitis viruses (A, B, C, and D) in that animals serve as natural reservoirs of the virus (Pavio et al., 2010). HEV is classified in the genus Hepevirus and is currently subdivided into at least four major genotypes, although boosted genotypes are proposed (Maclachlan et al., 2011c). Genotypes one and ii infect but humans; genotypes 3 and four can infect both humans and a range of other species involved in zoonotic transmission of the virus. HEV infects people worldwide, causing a sometimes fatal viral hepatitis (Lhomme et al., 2013). Its detection in fauna populations has steadily risen in the recent past, thereby prompting increased concern of its zoonotic potential.

Reservoir and Incidence

In humans, a large proportion of the enterically transmitted astute viral hepatitis cases in developing countries in Asia, Africa, and Mexico are due to HEV infections. In industrialized countries, serologic testify of HEV exposure is high and widespread, although clinical HEV infection is only sporadically diagnosed. Hepatitis E may be disproportionally fatal in pregnant women, with over a 25% case fatality rate as compared to 1% in the general population (Meng, 2005). All the same, it is unclear what function other underlying health factors may take played in this increased fatality charge per unit (Meng, 2010a). In industrialized countries, identified cases unremarkably occur sporadically, presumably due to zoonotic transmission from ane or more beast reservoirs or post-obit consumption of contaminated meat or shellfish (Lhomme et al., 2013; Meng, 2010a). In contrast, outbreaks most commonly occur in developing countries secondary to fecal contamination of food or water (Maclachlan et al., 2011c; Meng, 2005). Straight human-to-human being manual is rare (Pavio et al., 2010).

Domestic pigs can be naturally infected with HEV and shed large quantities of virus in their carrion. Under experimental conditions, swine HEV strains are infectious for nonhuman primates and the human strains of HEV are infectious to pigs. In improver, hog handlers (due east.g., swine veterinarians, swine farmers) have a significantly higher rate of seroconversion to HEV than does the general population (Huang et al., 2002). Swine infection is widespread and worldwide. In the United States, the sero­prevalence charge per unit of swine HEV infection ranges between 50 and 100% in some herds (Lhomme et al., 2013). Infection occurs in farmed pigs betwixt 2 and 4 months of historic period, presumably by fecal–oral transmission, with a transient viremia followed past fecal shedding (Huang et al., 2002; Meng, 2011). Given this, domestic pigs are recognized as likely reservoirs of HEV for the human population.

An avian strain of HEV exists with a high incidence of seroconversion in farmed chickens. The virus is at present believed to exist the etiologic agent of big liver and spleen disease (hepatitis-splenomegaly syndrome) of chickens. Still, disease has not resulted following experimental manual of the avian virus to other species including nonhuman primates, suggesting that the avian strain does non nowadays a significant zoonotic threat. Other animals including farmed rabbits, wild rats, wild boar, bats, mongeese, and deer are susceptible to HEV infection and are being evaluated as potential sources of human being affliction (Cossaboom et al., 2011; Lhomme et al., 2013; Meng, 2011). Of these, rabbit HEV is of greatest concern as antibody titers, suggestive of infection, have been detected in rabbit farms in the United States and China, and experimental infection of pigs has been demonstrated (Cossaboom et al., 2012). Some species (e.1000., tamarins, owls monkeys, and cynomolgus monkeys) are susceptible to experimental infection and antibody response to HEV has been detected in wild-defenseless macaques. Nonhuman primates are not considered likely disease reservoirs although disease manual from nonhuman primates to humans should non be discounted (Wachtman and Mansfield, 2012).

Style of Transmission

In human populations, hepatitis E was originally believed to result most commonly from human fecal contamination of drinking water in areas owned for the illness. Recent research has focused on the function that many animal species may play in zoonotic disease transmission from the feces of infected animals and the consumption of undercooked, contaminated creature tissues (meat and liver) (Lhomme et al., 2013). Aerosol transmission is non suspected. Given the suspected routes of virus transmission to humans, the risk within an animal facility should be minimal. All the same, personnel are brash to observe proper PPE practices to preclude possible HEV exposure.

Clinical Signs

Affliction is frequently self-limiting and asymptomatic, although an acute hepatitis can develop with classical signs of liver interest including jaundice, anorexia, and hepatomegaly. A fulminant, fatal hepatitis of pregnant women has been attributed to HEV infection, although this disease course has non been observed in naturally or experimentally infected pigs or experimentally infected rhesus macaques (Lhomme et al., 2013; Meng, 2010b). Chronic disease may develop in the immunocompromised including transplant recipients (Zhou et al., 2013). Infected pigs most frequently appear clinically normal despite microscopic lesions of hepatitis (Meng et al., 1997).

Diagnosis and Prevention

Propagation of HEV in cell civilization has been problematic, hindering its evaluation and development of diagnostic tests. Disease diagnosis in both humans and pigs is based on the detection of anti-HEV antibodies or the presence of HEV RNA in serum or feces (Pavio et al., 2010). Treatment is express to provision of supportive intendance.

A hepatitis East vaccine is non currently bachelor.

3 Other Viral Hepatitis Agents

Humans are considered natural hosts for viral hepatitis types B, C, and D, all of which are transmitted parenterally by exposure to blood or other actual fluids. Hepatitis B virus (HBV), caused by a homo hepadnavirus, has been widely studied experimentally in the chimpanzee although the gibbon, orangutan, and wooly monkey are known to be susceptible (MacDonald et al., 2000). Natural HBV infection was first noted past demonstration of HBV surface antigen in cynomologus in 1985 (Kornegay et al., 1985). Recently, wild-living cynomolgus macaques from Mauritius Island were diagnosed with natural, chronic infection of a genotype of HBV distinct from the human genotype. The disease appears relatively benign in the infected animals. The zoonotic adventure of the cynomolgus genotype is even so unknown (Dupinay et al., 2013). It is postulated that infected cynomolgus monkeys may serve as a useful beast model of the illness. Other natural hepadnavirus infections of animals (woodchuck, ground squirrel, and duck) are used as animal models of HBV infection, but none are transmissible to humans (Blanchard and Russell-Lodrigue, 2012). The chimpanzee has been used equally an experimental model for the study of hepatitis C and D viruses. Thus, the concern for hepatitis B, C, and D every bit zoonoses is minimal in the laboratory fauna facility surround except where these agents are used in experimental animate being studies. In these cases, personnel should adhere to appropriate precautions when handling nonhuman primates.

Read full chapter

URL:

https://www.sciencedirect.com/science/commodity/pii/B9780124095274000286

Preventive and therapeutic vaccines * Viral Immunology

Johannes Scholz , ... Reimar Johne , in Current Opinion in Virology, 2020

Last remarks

In club to clarify HEV pathogenicity and replication in more detail, substantial development can be recorded in the field of reverse genetics for HEV and related viruses. RGSs are available for most human being-pathogenic HEV types as well as for the most of import animal HEV-related viruses. However, for some viruses, for case, genotype two, rabbit HEV and fish HEV, RGSs are however missing. As a weakness of current RGSs, most of the cDNA clones showed only slow replication kinetics in cell culture while simply some genotype iii-based systems propagate to higher virus titers. Recent improvements in cell civilisation conditions [16,17 ] and the introduction of beneficial mutations into the HEV genome [17 ] may contribute to overcome these constrains. The RGSs for HEV and related viruses represent essential tools for basic and applied research, investigating virus replication and pathogenicity, the fashion of action for antivirals and consecration of resistance, likewise as for targeted development of vaccines in future.

Read full article

URL:

https://world wide web.sciencedirect.com/science/commodity/pii/S187962572030047X

Past, present and futurity of hepatitis Due east virus infection: Zoonotic perspectives

Shahid Faraz Syed , ... Muhammad Asif Arain , in Microbial Pathogenesis, 2018

half-dozen Determination and future perspectives

The panorama of HEV research is changing drastically with new and frequent developments in the field. Current inquiry is focused on understanding virus biological science, characterizing the host immune response during infection and developing strategies for prevention of HEV as a zoonotic illness. HEV 239, a condom vaccine being used successfully in people in China, has also been shown to provide protective immunity in rabbits, which could be very helpful in preventing animal HEV infection and zoonotic manual. The development of an effective vaccine to immunize wild animals and minimize the risk of HEV transmission to people or domestic animals is needed, but this task is challenging and has received very little attention in the by. Because cross-species transmission and host tropisms of zoonotic HEV genotypes are not yet understood, surveillance studies of swine and wildlife reservoirs similar deer and wild boar should exist conducted to identify all possible human exposure pathways. The expanding beast host range of HEV compels application of deep sequencing by using metagenomics approaches. In this style, animal species tin be tested to demonstrate whether they harbor virus that is closely related to human strains or genetically divergent, such equally avian HEV. Ultimately, the design of a vaccine based on common human and animal HEV epitopes may prove equally effective against zoonotic genotypes and is crucially of import to prevent zoonotic HEV transmission. This may not be easily attainable, as recent reports of antigenic variation betwixt HEV genotypes and quasi-enveloped viral particles that are shielded from neutralizing antibodies are emerging challenges to vaccine efficacy. Nevertheless, overcoming these challenges and others that ascend should be worth the effort.

Read full article

URL:

https://www.sciencedirect.com/science/article/pii/S0882401018304698

Identifying and defusing weapons of mass inflammation in carcinogenesis

Lorne J. Hofseth , Lei Ying , in Biochimica et Biophysica Acta (BBA) - Reviews on Cancer, 2006

Metals, especially iron and copper, can also play a key role in free radical generation and propagation. Therefore, metallic chelators are a potential source of high cancer risk, reactive species overload affliction prevention. The copper chelating amanuensis, tetrathiomolybdate, has been shown to protect against animate being hepatitis associated with copper overload [50]. Because the metal binding protein, metallothionein, is upregulated in high cancer hazard, reactive species overload diseases (possibly as an adaptive response), this could prove to be a benign molecule for treatment and chemoprevention. Mice deficient in metallothionein are more susceptible to gastritis [51] and hepatitis [52]. Copper chelation therapy is a useful strategy for the treatment of the high liver cancer risk, copper overload disease, Wilson's disease. Fe chelators have also been useful in disease treatment, including the iron overload disease, hemochromatosis (Table i; [53]).

Read total commodity

URL:

https://www.sciencedirect.com/science/commodity/pii/S0304419X05000466

Emerging viruses: intraspecies transmission • Viral Immunology

Harini Sooryanarain , Xiang-Jin Meng , in Current Opinion in Virology, 2019

Viral factors contributing to the host range and cantankerous-species infection of HEV

Ii major events, point mutations and recombination, attribute to the genetic variations and evolution of HEV genome. Accumulating bear witness suggest that the ORF1 appears to exist responsible for determining the host range and cross-species infection. It has been documented that point mutations affect HEV heterogeneity, and contribute to viral pathogenesis and susceptibility [100 ]. Recombination events tin occur within unlike HEV genotypes, and between homo and other animal HEV strains. An extensive heterogeneity in the hypervariable region (HVR) and macro domain of HEV genome is seen amidst the immunocompromised patients developing a chronic disease as compared to patients with resolving HEV infections [101]. Insertion and/or deletions in HVR of HEV ORF1 contributes to viral replication efficiency and host adaptations [102,103]. By utilizing intergenotypic chimeric viruses betwixt homo-exclusive genotype 1 HEV, and zoonotic genotypes 3 and four HEVs, it was plant that the HEV ORF2 capsid cistron does not determine host range [104,105]. Instead, the ORF1 is idea to be involved in host tropism of HEV. It has been shown that insertion of host RPS17 gene within the HEV HVR led to increased adjustability and infection of the virus in cells from different fauna origins [106 ]. A clinical isolate of HEV from a chronically-infected patient with a 258 bp duplicated HVR fragment and a 24 bp RdRp-derived fragment was associated with increased viral replication [107 ]. HVR appears to be exchangeable between different HEV genotypes, yet the chimeric viruses displayed a reduced virus replication kinetics [103]. Thus, it appears that HVR may contain genotype-specific sequences that are crucial for viral RNA replication and host adaptability, which may farther influence the disease outcome.

Read full article

URL:

https://www.sciencedirect.com/scientific discipline/article/pii/S187962571830110X