Next Article in Journal
Genetic Predictors of Comorbid Course of COVID-19 and MAFLD: A Comprehensive Analysis
Next Article in Special Issue
Clinical Validity of a Machine Learning Decision Support System for Early Detection of Hepatitis B Virus: A Binational External Validation Study
Previous Article in Journal
Characterization of Prunus Necrotic Ringspot Virus and Cherry Virus A Infecting Myrobalan Rootstock
Previous Article in Special Issue
No Hepatitis Delta Virus Seropositivity among Blood Donors with Overt and Occult Hepatitis B Infection in Dalian, Liaoning Province, China
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Hepatitis A Virus and Hepatitis E Virus as Food- and Waterborne Pathogens—Transmission Routes and Methods for Detection in Food

European Union Reference Laboratory for Foodborne Viruses, Swedish Food Agency, Dag Hammarskjölds väg 56 A, 75237 Uppsala, Sweden
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(8), 1725; https://doi.org/10.3390/v15081725
Submission received: 27 June 2023 / Revised: 7 August 2023 / Accepted: 9 August 2023 / Published: 12 August 2023
(This article belongs to the Special Issue Epidemiology and Diagnostics of Hepatitis Viruses)

Abstract

:
Foodborne viruses are an important threat to food safety and public health. Globally, there are approximately 5 million cases of acute viral hepatitis due to hepatitis A virus (HAV) and hepatitis E virus (HEV) every year. HAV is responsible for numerous food-related viral outbreaks worldwide, while HEV is an emerging pathogen with a global health burden. The reported HEV cases in Europe have increased tenfold in the last 20 years due to its zoonotic transmission through the consumption of infected meat or meat products. HEV is considered the most common cause of acute viral hepatitis worldwide currently. This review focuses on the latest findings on the foodborne transmission routes of HAV and HEV and the methods for their detection in different food matrices.

1. Introduction

The estimated number of cases caused by foodborne diseases is 600 million each year, leading to 420,000 premature deaths worldwide [1]. Food can be unsafe for human consumption due to the presence of pathogens such as bacteria, viruses besides parasites and chemical or physical substances [2]. Human pathogenic viruses represent a frequent causative agent of foodborne diseases, having a significant impact on human health and the economy. Although numerous viruses may be foodborne transmitted, noroviruses and hepatitis A virus (HAV) are the major causative agents of foodborne viral outbreaks. Hepatitis E virus (HEV) is a significant cause of acute viral hepatitis worldwide. Additionally, it is an emerging pathogen in economically developed countries, and the foodborne transmission of this zoonotic virus appears to be the major route in Europe (HEV-3) [3,4]. Both HAV and HEV are small RNA viruses but differ in genomic organization and structure.
Hepatitis A virus is a positive-stranded RNA virus belonging to the genus Hepatovirus of the Picornaviridae family [5,6]. HAV is a small virus with a 27–32 nm diameter virion. Two different forms of this infectious virus can be found: the naked, non-enveloped HAV virions which are excreted in the stool, and the quasi-enveloped virions (eHAV), which are released non-lytically from infected cells. eHAV are found in the blood of infected patients or in the supernatant of infected cell cultures [7,8]. HAV is a causative agent of acute infection of the liver; the clinical course of infection ranges from mild to severe while children (<6 years) are often asymptomatic. Symptoms can include fever, malaise, loss of appetite, diarrhoea, nausea, abdominal discomfort, dark-coloured urine and jaundice (a yellowing of the eyes and skin) [2]. Infection by HAV leads to lifelong immunity and does not result in chronic infection or chronic liver disease [9]. The HAV infection is rarely fatal; the WHO estimated that in 2016, 7134 people died from HAV worldwide (accounting for 0.5% of the mortality due to viral hepatitis [1]).
HAV is classified into six genotypes; three genotypes infect humans (I, II and III) and IV, V and VI infect simians. Genotypes I, II and III are divided into seven subtypes (IA, IB, IC, IIA, IIB, IIIA and IIIB). Worldwide, genotype IA is the most reported, which is followed by III, while genotype II has been rarely reported. For the latter, genomic information is sparse [10,11].
Hepatitis E virus (HEV) is a small single-stranded RNA virus that belongs to the Hepeviridae family in the Orthohepevirus genus [12]. Similar to HAV, two different HEV particles exist: the non-enveloped particles that are mainly found in faeces and the quasi-enveloped particles that can be found in serum and cell culture supernatant [13]. Non-enveloped particles showed higher infectivity in cell culture and in uPA-SCID mice model compared to enveloped particles [14,15].
HEV is classified into eight genotypes; HEV-1-4 and HEV-7 have been shown to infect humans; HEV-5 and 6 have been shown to infect only wild boar. HEV-3 infects domestic pig and wild boar, while HEV-7 and HEV-8 has been proposed as a zoonotic camel variant [16]. HEV-3 and HEV-4 are considered zoonotic [17,18]. HEV genotypes are further divided in subtypes [19,20].
HEV-1 strains are endemic to Asia and most countries in sub-Saharan Africa, where it is the main cause of recurrent epidemics. Moreover, HEV-1 is causing large outbreaks in India [21]. HEV-2 is present in Mexico, Nigeria, Chad, Sudan, and the Central African Republic [22]. HEV-3 has a worldwide distribution (Europe, the United States, and other North American countries, Central and Southern Japan, New Zealand, and Australia) while HEV-4 can be found predominantly in Asia (China, northern Japan, and India) [23]. HEV-5 and HEV-6 were detected in Japan [24]. So far, HEV-7 has been identified in the United Arab Emirates, Somalia, Sudan, Egypt, Kenya, Pakistan, Israel, Saudi Arabia and Djibouti [16]. HEV-8 was detected in China [25].
Most of the HEV infections are asymptomatic but HEV can cause symptomatic infection leading to a self-limited, acute hepatitis [26]. HEV can become chronic in immunosuppressed people [27,28]. HEV-1 and HEV-2 can cause severe disease and fulminant hepatitis; pregnant women are affected primarily [29]. HEV infection in pregnant women shows rapid virus replication and has a high incidence of developing fulminant hepatic failure (FHF) or acute liver failure (ALF) [30]. The mortality rate in pregnant women is often 30% or higher particularly in the third trimester [31]. Wang and colleagues identified two mutations in the ORF1 of HEV-1 that enhanced viral replication and may be associated with FHF [32]. Rarely, HEV can lead to ALF not only in pregnant women but also in elderly patients, patients with leukaemia or coinfection with other hepatotropic viruses [33,34,35]. HEV-3 and HEV-4, on the other hand, do not appear to cause fatal infections with fulminant hepatitis in pregnant women, but this has to be confirmed in future studies [36]. However, HEV-4 was shown to be associated with adverse pregnancy outcomes in pregnant women [37,38]. Rarely, acute and chronic and otherwise symptomatic and asymptomatic HEV-3 infections can cause neurological disorders (e.g., neuralgic amyotrophy and Guillain–Barré syndrome) [39].

2. Transmission Routes

The shedding of foodborne viruses originates from the human gastrointestinal tract, and their presence in water and food is a result of contamination by sewage, poor hygiene, or contamination by food handlers [40]. Food can be contaminated at any stage from farm to table; however, often, foodborne viral infections can be traced back to infected food handlers [41]. Foodborne viruses (including HAV and HEV) are typically highly resistant to environmental factors, such as low pH (acidity) and heat [42,43]. They can remain infectious for over a month in food and water [44,45].

2.1. Foodborne Transmission of HAV

The main route of HAV transmission is from person-to-person by the faecal–oral route. HAV outbreaks occur from time to time in high-risk populations, such as men who have sex with men (MSM, sexual transmission) and persons who use injection drugs (bloodborne transmission) [46,47]. Vertical transmission of HAV (from the mother to the foetus) is uncommon, although there are few case reports published [48,49]. In addition, HAV can be transmitted by the ingestion of contaminated food or water. Closed environments and restaurant caterings results in the most common setting of HAV outbreaks. Most European countries are considered as non-endemic or have low endemicity [50], so infection is often related to recent travel to countries with higher rates of HAV. In addition to travelling, HAV infections and outbreaks are frequently associated with imported food products (Table 1).
Berries, green-leaf salads and bivalve molluscan shellfish (BMS)—that are often eaten unprocessed—are the most common food matrices implicated in HAV outbreaks. Ready-to-eat meals contaminated by a food handler or using a contaminated ingredient (e.g., berries on cakes) can cause outbreaks as well. A harmonised investigation in three European countries demonstrated the presence of human enteric viruses in the leafy green vegetable supply chain; however, HAV was only detected in samples from the primary production phase [51]. Due to the complexity of food supply chains and the long incubation time of HAV (with a mean of about 30 days) [52], it is often difficult to identify the actual contaminated source. Table 1 summarises the lately published outbreak studies of HAV containing the investigations in which the implicated food or food handler was confirmed by PCR. In addition to dates and clams, the outbreaks were related to frozen berries (often imported), lettuce and dried tomatoes (Table 1).
Table 1. HAV outbreaks associated to food published in the last 15 years.
Table 1. HAV outbreaks associated to food published in the last 15 years.
YearGenotypeImplicated FoodImported 1No. of CasesOutbreak LocationReference
2021IBmedjool datesY31England and Wales[53]
2021IBmedjool datesY6Australia[54]
2019IAjogaejeot (fermented clams)N South Korea[55]
2019IAblackberriesY20USA[56]
2018–2022IBstrawberriesY65Germany [57]
2018IBstrawberriesY20Sweden and Austria[58]
2018IBfrozen pomegranateY30Australia[59]
2017IBfrozen raspberry/blueberryY14The Netherlands[60]
2016IAraw scallopY292Hawaii[61]
2016IAfrozen strawberries and blackberriesY7New Zealand[62]
2015IAmixed frozen berries packedY67Australia[63]
2014IAkava drinkN4Australia[64]
2013–2014IAmixed frozen berriesY1803Italy, Germany, Ireland, Norway, Austria, Poland, Netherlands, Bulgaria, Denmark[65,66,67]
2013–2014IAfrozen berry mix cakeY33Norway[68]
2012–2013IBfrozen strawberriesY103Denmark, Finland, Norway and Sweden[69]
2012IBpomegranate seeds, a component of frozen fruit blendN9Canada[70]
2012IAmusselsN9The Netherlands[71]
2011IBsemi-dried tomatoes nd8The Netherlands[72]
2011IBsemi-dried tomatoes nd7England[73]
2010IBsemi-dried tomatoes 13The Netherlands[74]
2010IBsemi-dried tomatoes Y59France[75]
2009IBsemi-dried tomatoes Y and N562Australia[76]
2008ndlettucend22California, USA[77]
2008IBRaw clams (Coquina clams)Y100Spain[78]
2008IIIAlettuce or carrotNndSouth Korea[79]
1 Y (yes)/N (no)/nd (not defined).
Bivalves are filter feeders and can therefore bio-accumulate and concentrate microorganisms and viruses from water. Hepatitis A outbreaks are rarely reported in association with the consumption of contaminated bivalve shellfish [80,81], which is possibly due to the low and decreasing incidence of HAV in the European population [82] and due to the long incubation time, making trace-back difficult. The presence of HAV in BMS varies over time and place. Dirks and colleagues studied the presence of HAV (besides norovirus) in bivalve molluscs in the Netherlands between 2013 and 2017. Among the total of 228 oyster and 392 mussel samples, the presence of HAV RNA was detected in only one mussel sample (0.3%, genotype IA) [83]. In the Campania region (Southwest Italy), 289 samples from shellfish production areas and other locations were tested between 2015 and 2017; 8.9% of the samples tested positive for HAV [84]. Shellfish samples (108, fresh and frozen) were collected from three harvesting areas and from restaurants, fish markets, and shellfish markets in Sicily; 13% of the samples tested positive for HAV (14 HAV-positive samples, one HAV 1A and 13 HAV 1B) [85].
In a study assessing the trend of HAV, after a contamination in BMS caused by a community outbreak, a minimum of 5 weeks was required to reduce viral loads to undetectable levels. Moreover, according to their regression analysis, 2–3 months may be required to ensure the removal of residual viral particles present in concentrations below the detection limits of the analytical method [86].
Rarely, drinking water can be a source of HAV infection; several HAV outbreaks have been documented and linked to faecally polluted drinking or recreational water [87].

2.2. Transmission of HEV

Unlike HAV, human-to-human transmission of HEV is uncommon. Secondary attack rates among household contacts of HEV cases was shown to be very low (0.7–2.2%), while it was 50–75% in the case of HAV infections, which is another enterically transmitted virus [88]. The most common sources of HEV infections that occur in humans are contaminated water or food. However, the number of reported transfusion-transmitted HEV (TT-HEV) cases, in blood donation recipients, is also on the rise [89]. In addition, the maternal–foetal transmission of hepatitis E virus has also been reported [90,91].

2.2.1. Waterborne Transmission

HEV-1 and HEV-2 are human specific, and large HEV outbreaks occur due to contaminated water consumption in low-income countries [92,93,94]. An investigation of a tap water-mediated HEV outbreak in China suggests that sporadic cases of waterborne outbreaks of HEV-4 may occur in industrialised countries [95]. Sewage of human and swine origin can be a potential transmission route for HEV, and few infected people can contaminate municipal wastewater treatment plants (WWTP), increasing the potential for further spread of the virus [96]. Coastal waters can become contaminated by human sewage and manure runoff, and therefore, HEV can accumulate in shellfish produced close to land [97,98,99]. The detection of HEV-3 in surface and drinking water indicates the potential risk of HEV-3 transmission through drinking water [100,101,102].

2.2.2. Zoonotic Transmission of HEV

Meat consumption as a source of HEV infection of humans was first described in 2005 [103]. Foodborne transmission in Europe is linked to the consumption of HEV-contaminated pork meat products (liver sausages, salami), undercooked wild boar meat and raw venison. Zoonotic transmission of HEV-3 (predominant in Europe and in America) and HEV-4 occurs frequently, and pigs represent the main reservoir for zoonotic HEV [104].
The numerous contact points between humans and domestic and wild animals from breeding to the food industry redound HEV transmission. The prevalence of HEV in pigs varies between pig farms, farming systems and countries. Generally, both the between-herd and within-herd prevalence of HEV is high (between 20 and 75%), indicating the frequent exposure of pigs to the virus [105,106,107,108,109]. Boxman and colleagues found a high prevalence (16%) of acute HEV infection in pigs in Dutch slaughterhouses [110]. The length of the fattening period has been shown to affect the risk of HEV; a longer fattening period may lower the risk of HEV in shedder animals at slaughter and thus reduce the risk of food contamination [111].
The seroprevalence of immunoglobulin G (IgG) against HEV (anti-HEV IgG) ranges between 10 and 20% in the European human population [112,113,114,115], and exposure to pigs is associated with a significantly higher seroprevalence (in people with contact to pigs) compared with non-exposed humans [116]. A high prevalence of anti-HEV antibodies among swine slaughterhouse workers and farmers has been reported, suggesting an occupational risk of HEV infection [117,118,119,120]. Wu and colleagues studied the risks of HEV infection in workers along the meat supply chain [121]. Their analysis showed that the human HEV infection risk increased along the pork supply chain, with the highest risk at pig slaughterhouses and pork markets; in contrast, no significant higher risk was observed among poultry workers.
Outbreaks of HEV-3 are primarily associated with the consumption of pork products, especially undercooked pork liver or sausages [122,123]. Pig liver is more frequently positive for HEV, while muscles are less often contaminated by HEV-RNA [107]. In accordance with that, the presence of HEV is the highest in products containing liver, which is followed by raw meat sausages (Table 2). Table 2 summarises the recently published studies of the presence of HEV RNA in commercially available pork products and meat cuts. A parametric stochastic model estimating the risk of foodborne exposure showed that products containing liver pose the highest risk at the individual level [124].
Studies on the efficiency of inactivation methods for HEV during meat processing are quite limited [139]. According to a study by Johne and colleagues, high hydrostatic pressure processing (HPP) can be considered as a treatment method for decreasing the risk of foodborne HEV transmission [140].
In addition to swine, wild boar is also an important natural reservoir for the zoonotic transmission of HEV around the world. Since HEV infections are mostly asymptomatic, only two outbreaks associated with the consumption of undercooked venison and wild boar has been published so far [103,141]. Table 3 summarises an updated list of survey studies published of the presence of HEV in wild boar. The prevalence of the virus varies greatly between different geographical regions, as does as the number of samples tested in these surveillance studies.
Although swine and wild boar are the main reservoirs of HEV-3-6, several other animal species act as an HEV host, including domestic and wild ruminants. The first identification of HEV RNA and evidence of zoonotic transmission of HEV in deer has been published in 2003 [142]. Since then, numerous surveillance studies were published on molecular and serologic screening of serum and faecal samples from domestic and wild ruminant species. The virus is present in deer (red deer, Cervus elaphus; roe deer, Capreolus capreolus; fallow deer, Dama dama) and in chamois (Rupicapra rupicapra); however, the prevalence of HEV varies in different geographic regions [143,144,145,146,147]. A high seroprevalence of HEV has been observed in wild reindeer (23.1%) and moose (30% and 19.5%); furthermore, HEV exposure in muskoxen has been reported [148,149]. In a study from Finland, HEV seroprevalence was 9.1% (31/342) in moose and 1.4% (1/70) in white-tailed deer, but HEV RNA could not be detected from samples of seropositive animals [150]. HEV RNA was detected in 7.69% of the moose samples (n = 13) in a surveillance study in Lithuania [147].
HEV-3 and HEV-4 have been detected in bovine liver as well, suggesting that bovine livers may be involved in the zoonotic transmission of HEV to humans [151,152]. Moreover, HEV-3 is also present in sheep and goat populations in Italy [143,153]. Recently, sporadic infections with rabbit HEV (raHEV) have been reported, and immunosuppressed patients (solid organ transplant recipients) have been infected with rabbit HEV (genotype 3ra) [154,155]. In France, 5 of 919 (0.5%) HEV-infected patients during 2015–2016 were infected with a rabbit HEV strain. The source of infection was unclear because none of the patients had direct contact with rabbits. The HEV-3ra infections could be the result of foodborne or waterborne infections [156].
Predominantly, the HEV variants causing human infections belong to HEV species A (Orthohepevirus A, HEV-A) [12]. Orthohepevirus genotype 1 (HEV-C1) circulates in rats and was first isolated in Germany in 2010 [157]. HEV-C1 was earlier considered unable to infect humans, but since then, several rat hepatitis E virus (HEV) cases have been identified in humans [158,159,160,161,162]. Despite the occurrence of HEV-C1 infections in humans, several studies indicating that HEV-C1 infections are very rare among several populations in Europe [163,164,165]. Furthermore, the transmission route of HEV-C1 between rats and humans is elusive. Rat meat consumption is uncommon, suggesting other infection sources and transmission patterns (e.g., rat infestation in domestic premises, water contamination). Further epidemiological investigations would help to identify infection sources and transmission patterns.
HEV-3 and HEV-4 have been found to be excreted in the milk of ruminants [166,167,168]; however, data on the potential milk-borne transmission are still lacking or/and conflicting. The presence of HEV in milk samples from dairy farms was investigated in Germany, but HEV-specific RNA could not be detected in the 400 bulk milk samples [169]. The different results can possibly be due to the differences in the genotype distribution and the differences in the farming systems. Thus, further investigations on the prevalence of HEV in milk could facilitate the understanding of its risk of zoonotic transmission. The zoonotic infection by HEV-7 in a human liver transplant patient who had consumed camel milk demonstrates the ability of this virus to infect humans.
Table 3. HEV RNA detection in wild boar.
Table 3. HEV RNA detection in wild boar.
Year of SamplingSpeciesCountryNo. of SamplesHEV RNA Prevalence; Genotype and SubtypeReference
2018–2022wild boarPortugal144 stool samples 2.80%; HEV-3e, m[170]
2017–2022wild boarChina599, faecal or serum samples2.2%; HEV-4a, d, h[171]
2019–2020wild boarItaly86 liver26.70%; HEV-3c[172]
2019–2020wild boarItaly156 livers5.12%; HEV-3c[173]
2013–2017wild boar and deer Germany1961 wild boar, 559 roe deer, 736 red deer, 316 fallow deerwild boar 13.3% and deer from 4.2%; HEV-3c, f, i[174]
2012–2021wild animals, including 15 speciesJapan3489 serum samples1.2% of wild boar, 0.06% of Sika deer, HEV-3a, b, k HEV-4j[175]
1997–2020wild sika deer (Cervus nippon)Japan395 serum and 199 liver samples of 405 sika deer 0.20%; HEV-3b[176]
2019wild boarGermanyliver, faeces, and muscle samplesmean 8.14%; HEV-3a, c, e, f, k, i[177]
2016–2020wild boarItaly611 livers and 88 paired lungs2.45% of livers, 1.13% of lungs; HEV-3n[178]
2015–2020wild boars, red deer, roe deer and chamois Italy602 wild boar liver, 228 ruminant liver6.97%; HEV-3a, f[179]
2015–2019wild boarPoland470 liver, 433 faeces12.1% of liver, 6.2% of faecal samples[180]
2018–2019wild boarBulgaria3212.50%[181]
2013–2019wild boarJapan1803 serum samples, 1519 liver tissues and 42 gallbladder 3.90%; HEV-5[182]
2015–2016wild boarKorea1859 wild boar bloods1.29%; HEV-3a, HEV-4a, d[183]
2010–2017swine and wild boarsCroatia720 tissue and/or blood samples11.50%; HEV-3a, e[184]
2016–2017wild boarItaly92 livers52.20%; HEV-3c, f[185]
2013–2015wild boarRomania45 liver, 5 spleen 18%; HEV-3[186]
2011–2012wild boarPortugal80 liver samples, 40 stools25% of livers, 10% of stools; HEV-3e[187]
2003–2010wild boar, Iberian pig, deerSpain28710.12% of wild boar, 16.05% of red deer, 0% of Iberian pigs[188]
ndwild boarItaly594 serum and 320 liver4.9% of liver, 0% of serum samples; HEV-3e, f[189]
nd wild boarSweden159 blood samples8.17%; HEV-3f[190]
Genotypes 7 and 8 should be considered as potential human pathogens [191]. A review has been recently published, summarising the current scientific knowledge in HEV detected in milk [192].

3. Methods Used for Detection of HAV and HEV in Different Food Matrices

The low virus concentration and the complexity of food matrices make it difficult to detect viruses in food samples. In general, the virus detection procedure contains three main steps: (1) virus extraction from food matrices, (2) nucleic acid extraction and (3) detection by PCR-based methods.
The first step of the virus detection is virus extraction, which includes the separation and concentration of viruses from food matrices. Methods for the extraction depend on the food composition; three main categories can be distinguished [193]. The first category is composed of carbohydrate- and water-based foods (fruits and vegetables). The second category includes fat- and protein-based foods (ready-to-eat products). Shellfish belongs to the third and separate food category due to the accumulation and concentration of viral particles and other pathogens in the shellfish digestive system. Stals and colleagues have summarised the variety of protocols for virus extraction from food samples [194].
The RNA extraction is mostly performed with commercially available kits that are based on the Boom nucleic acid extraction method using the disruption of viral capsids with guanidine thiocyanate and adsorption of viral RNA to silica magnetic beads or columns with silica [195]. Earlier, RNA was extracted by phenol–chloroform, which ischeaper, but produces hazardous by-products. RT-qPCR is widely used for virus detection because of its sensitivity and specificity. In addition, it delivers quantitative data if standard curves or reference values are used. On the other hand, the PCR is an enzymatic reaction and therefore is sensitive to inhibitors. Food samples are complex and often contain PCR inhibitors. Therefore, proper controls are needed to avoid false negative results (due to inhibition) and false positive results (due to cross-over contamination). Inhibition can be monitored using an external control (EC) RNA, as described, for example, in ISO 15216-1:2017 [196]. The degree of inhibition is measured by comparing the Cq value of a sample well spiked with EC RNA with the Cq value of a control well containing an equal amount of EC RNA in nuclease-free water. Typical PCR inhibitors in food matrices are phenols, polysaccharides (berries), pectin, polyphenol, xylan (green leafy vegetables), algae, and glycogen (bivalves) (summarised by [197]).

3.1. Methods for HAV Detection

A standardised method is available for norovirus and HAV detection in food and has been validated in seven food matrices: bottled water, food surfaces, oysters (Magallana gigas, earlier: Crassostrea gigas), mussels (Mytilus edulis), raspberries, lettuce and green onions [196]. In addition to the validated, high-risk food categories, the standardisation and validation of detecting viruses from other food matrices is needed. For multicomponent foodstuff samples, methods have been described that can be used to detect viruses in composite food products for routine diagnosis [198,199,200].

3.2. Methods for HEV Detection

Unlike HAV and norovirus, no standardised method has been described so far for the detection of HEV in food samples; however, the standardisation process for the detection of HEV has recently been initiated by the International Organisation for Standardisation (ISO; ISO/TC 34/SC 9/WG31 Hepatitis E virus). For pork products, several RT-PCR methods have been described to detect HEV; a recently published review discusses these methods, focusing on the successful use in subsequent studies and surveys [201]. A validated method for HEV RNA detection in meat and meat products has been described by Althof and colleagues [202]. The method was validated in a ring trial with nine independent laboratories using artificially contaminated pork liver sausage samples. The method is currently the basis for the development of a new ISO standard in ISO/TC 34/SC 9 WG 31 (Magnus Simonsson, personal communication). Contaminated milk is a potential risk source for HEV infection, so the assessment of HEV in the milk is of great importance for consumers’ safety [166,203]. A study has been published recently on the detection of HEV in milk-based food products, showing that the HEV molecular assay could be improved by the selection of the extraction procedure that matches with the milk matrix. In addition, the removal of inhibitory substances such as fat and casein from the milk sample increased the performance [204].

3.3. Digital PCR Vs. qPCR

In recent years, digital PCR (dPCR) has been recognised as a useful alternative to qPCR for detecting and quantifying viruses in food samples [205,206,207]. The idea of digital PCR was described already in the 1990s (then referred to as limited dilution PCR [208]), but it was not until the 2010s that the technique became widely commercially available. The sensitivity of the reverse transcription digital PCR (RT-dPCR) assay was similar to that of RT-qPCR, and the methods agreed well in quantification of the presence of HEV in naturally contaminated pig products [209]. Similar to qPCR, dPCR allows the simultaneous quantification of different targets. HAV and norovirus genogroup I and II were quantified simultaneously by triplex droplet reverse transcription digital PCR in food, drinking water, and faecal samples [210]. In general, RT-dPCR can be more expensive and time consuming compared to RT-qPCR, but no external calibration curves are needed for the quantification. In addition, compared to RT-qPCR, RT-dPCR generally offers higher precision in quantification [211] and is less affected by inhibitors [212,213] and primer–template mismatches under certain conditions [214,215,216].

3.4. Molecular Typing

The typing of viral strains allows the identification of the possible source of foodborne outbreaks [217]. Moreover, it helps with studying the epidemiology and transmission routes of the viruses. Online genotyping tools have been developed to identify the HAV and HEV genotypes of a nucleotide sequence (https://www.rivm.nl/mpf/typingtool/hav/ (accessed on 24 April 2023); https://www.rivm.nl/en/Topics/H/HEVNet (accessed on 24 April 2023)).
Next-generation sequencing (NGS) is a sensitive and widely used technique for the molecular typing of pathogens. Yang and colleges identified inter- and intra-host variants of HAV in the clinical specimens and under laboratory culture conditions using NGS [218]. Lately, a multiplex PCR-based NGS was implemented to define HAV genotypes from hepatitis A patients [219]. NGS is becoming frequently applied in food sample testing, primarily for norovirus typing [220,221]. Only few studies have been published so far on the application of NGS for the typing of HAV and HEV in food matrices. The majority of the NGS methods for foodborne virus analysis are amplicon-based and incorporate an RT-PCR amplification step (amplification of subgenomic regions, such as the VP1/P2B junction in case of HAV). These methods can be used for the identification of transmissions among cases with known epidemiological association. As an example, HAV was detected and identified as a source of outbreak from frozen berries by NGS in northern Italy [222]. Yang and colleges developed a sensitive and amplification-independent method for virus characterisation. Celery samples were used as a food matrix, and they were inoculated with high and low copies of viruses (norovirus and HAV) as a single or as a multi-strain mixture [223]. Compared to amplicon-based sequencing, the whole-genome sequencing methods offer a more accurate tracking of virus strains. In a study by Vaughan et al., HAV genetic identity was precisely determined using whole genome (WG) sequences using samples from food-borne outbreaks besides non-outbreak-related samples [224].
Both the ORF 1 and ORF2 regions are used for molecular typing of HEV for Sanger sequencing and NGS, and different subtypes can be determined [130,225,226,227]. HEV-3 subtypes c, e and f have been shown to be the most prevalent subtypes in Europe [4]. Investigations on the HEV strains circulating in pig farms showed that each farm has its unique HEV-3 strain, and the same strain is present in the farms for several years [109,190]. This uniqueness can be helpful in outbreak investigations by facilitating the source tracking.
Sanger sequencing and NGS are still the primarily used methods for viral detection and typing, but third-generation sequencing such as Oxford Nanopore Sequencing, ONT, is becoming a common method for epidemiological studies (e.g., to elucidate viral recombination) [228,229,230,231]. Flint and colleagues used a pre-amplification step to obtain full-length genomic amplicons followed by sequencing on Illumina and ONT platforms in combination to obtain accurate data while reducing the required viral titre [232]. In general, ONT can generate long reads up to 100s of kilobases in a relatively short time and at low cost; however, it is less accurate with a higher error rate compared to Illumina [233,234].

3.5. Emerging Molecular Methods

The rapid and early detection of foodborne pathogens is essential for food safety and for outbreak investigations. The most commonly used methods for nucleic acid detection are PCR-based methods thanks to their accuracy. An important limitation of PCR is the expensive instrumentation and special expertise/trained personnel. Simpler approaches allow more rapid, ease of use methods with less cost (on-site testing). Several techniques have been established over time for nucleic acid-based diagnostics. The main types of these methods are recombinase polymerase amplification (RPA), isothermal-based amplification methods (e.g., loop-mediated isothermal amplification, LAMP), and CRISPR-Cas-based detection methods [235]. Reverse transcription loop-mediated isothermal amplification (RT-LAMP) combines LAMP DNA detection with reverse transcription and can be an alternative to RT-qPCR. In the case of LAMP, the nucleic acid amplification is conducted at a constant temperature (between 60 and 65 °C), so a thermal cycler is not needed. Wu and colleges have developed an RT-LAMP assay for the detection of HAV in different food matrices, including green onion, strawberry, mussel, and milk [236]. Their assay showed comparable sensitivity with RT-qPCR in certain food matrices. The specific amplification of the LAMP method relies on four or six designed primers that bind to six regions specific to the target gene [235]. The mismatches between primers and templates significantly reduce the amplification efficiency of LAMP (especially the two inner LAMP primers FIP and BIP that are typically over 40 nt long, which form mismatches easily with templates of highly variable viruses) [237]. The LAMP assay performance on samples varies according to the level of detectable RNA, and the diagnostic performance may change over time from symptom onset in real-life clinical settings [238]. Generally, this technique has great potential, but further improvement and implementation are needed until more extended application.
Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-associated nuclease (Cas) (CRISPR-Cas)-based methods are a promising alternative for pathogen diagnostics. Approaches such as SHERLOCK (based on CRISPR-Cas13a) and DETECTR (based on CRISPR-Cas12a) are potential methods for the rapid detection and identification of infectious diseases, with simple yet ultra-sensitive tests [239,240,241]. Although these methods are rapidly evolving, there are still many challenges that need to be solved before CRISPR sensing can replace or complement established techniques such as PCR. These challenges mainly are sequence limitations, standardisation, quantification and multiplexing.

4. Concluding Remarks

The globalisation of the food industry favours the foodborne-related HAV outbreaks, as many high-risk products (shellfish, fresh or frozen fruits, and vegetables) are produced in HAV-endemic countries and imported to countries of low endemicity (e.g., Europe). A standardised method helps the accurate quantification and comparison of analysis results between laboratories and between different survey studies. The available ISO method for the detection of HAV in food gives an opportunity for more harmonised and reliable food control; however, even when harmonised standards are used, considerable variation may occur in results [242].
The reported HEV cases in Europe have increased tenfold in the last 20 years partially due to the increasing awareness and the improvement of the detection methods [3]. Yet, the global burden of the disease is considered underestimated and largely unknown. It is difficult to estimate the overall prevalence of HEV, since the methods used in the survey studies are different as well as the samples sizes of these studies. The prevalence varies widely between geographical regions and study populations [243]. A standardised method for HEV detection followed by further research and survey studies of HEV would help achieve a better risk assessment of the food and animal products in the transmission of HEV to humans. Comprehensive and comparable surveillance studies in Europe would help obtain a complete picture regarding the presence and global burden of the virus.
The nucleic acid-based methods (including the most widely used PCR methods) are not able to discriminate between the infectious and the non-infectious viral particles, which may make the interpretation of a positive test result in foods arguable. However, with the usage of proper controls, etc.; the presence of viral RNA suggests that viral contamination of the tested food has occurred somewhere along the supply chain. Viability PCR methods—which amplify nucleic acids only from intact virions—could be an alternative to this problem [244,245], but these methods are not used in routine surveillance currently.
Further method improvements can lead to two different directions that complement each other. One direction could be the improvement of a more accurate but simple and quick detection of viruses, such as CRIPSR-Cas based methods. These methods are extremely useful when a rapid detection is needed (e.g., crisis situations). The other direction—not replacing but complementing the rapid tests—could be the improvement of NGS methods (e.g., more user-friendly bioinformatics software) to help the outbreak investigations and the characterisation of the virus strains circulating in the human population and in the environment.

Author Contributions

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

Funding

This research received no external funding.

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. WHO; GWHO. WHO Estimates of the Global Burden of Foodborne Diseases. Foodborne Disease Burden Epidemiology Reference Group 2007–2015. Available online: https://apps.who.int/iris/bitstream/handle/10665/199350/9789241565165_eng.pdf (accessed on 24 April 2023).
  2. WHO. Hepatitis a Fact Sheet. Available online: https://www.who.int/news-room/fact-sheets/detail/hepatitis-a (accessed on 24 April 2023).
  3. EFSA Panel on Biological Hazards (BIOHAZ); Ricci, A.; Allende, A.; Bolton, D.; Chemaly, M.; Davies, R.; Fernandez Escamez, P.S.; Herman, L.; Koutsoumanis, K.; Lindqvist, R.; et al. Public health risks associated with hepatitis E virus (HEV) as a food-borne pathogen. EFSA J. 2017, 15, e04886. [Google Scholar] [CrossRef] [PubMed]
  4. Aspinall, E.J.; Couturier, E.; Faber, M.; Said, B.; Ijaz, S.; Tavoschi, L.; Takkinen, J.; Adlhoch, C.; On Behalf Of The Country Experts. Hepatitis E virus infection in Europe: Surveillance and descriptive epidemiology of confirmed cases, 2005 to 2015. Eurosurveillance 2017, 22, 30561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Sander, A.L.; Corman, V.M.; Lukashev, A.N.; Drexler, J.F. Evolutionary Origins of Enteric Hepatitis Viruses. Cold Spring Harb. Perspect. Med. 2018, 8, a031690. [Google Scholar] [CrossRef] [PubMed]
  6. Lefkowitz, E.J.; Dempsey, D.M.; Hendrickson, R.C.; Orton, R.J.; Siddell, S.G.; Smith, D.B. Virus taxonomy: The database of the International Committee on Taxonomy of Viruses (ICTV). Nucleic Acids Res. 2018, 46, D708–D717. [Google Scholar] [CrossRef] [Green Version]
  7. Feng, Z.; Hensley, L.; McKnight, K.L.; Hu, F.; Madden, V.; Ping, L.; Jeong, S.H.; Walker, C.; Lanford, R.E.; Lemon, S.M. A pathogenic picornavirus acquires an envelope by hijacking cellular membranes. Nature 2013, 496, 367–371. [Google Scholar] [CrossRef] [Green Version]
  8. McKnight, K.L.; Lemon, S.M. Hepatitis A Virus Genome Organization and Replication Strategy. Cold Spring Harb. Perspect. Med. 2018, 8, a033480. [Google Scholar] [CrossRef]
  9. Lemon, S.M.; Ott, J.J.; Van Damme, P.; Shouval, D. Type A viral hepatitis: A summary and update on the molecular virology, epidemiology, pathogenesis and prevention. J. Hepatol. 2017, 68, 167–184. [Google Scholar] [CrossRef] [Green Version]
  10. Vaughan, G.; Goncalves Rossi, L.M.; Forbi, J.C.; de Paula, V.S.; Purdy, M.A.; Xia, G.; Khudyakov, Y.E. Hepatitis A virus: Host interactions, molecular epidemiology and evolution. Infect. Genet. Evol. 2014, 21, 227–243. [Google Scholar] [CrossRef]
  11. Desbois, D.; Couturier, E.; Mackiewicz, V.; Graube, A.; Letort, M.J.; Dussaix, E.; Roque-Afonso, A.M. Epidemiology and genetic characterization of hepatitis A virus genotype IIA. J. Clin. Microbiol. 2010, 48, 3306–3315. [Google Scholar] [CrossRef] [Green Version]
  12. 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, 001778. [Google Scholar] [CrossRef]
  13. Takahashi, M.; Tanaka, T.; Takahashi, H.; Hoshino, Y.; Nagashima, S.; Jirintai; Mizuo, H.; Yazaki, Y.; Takagi, T.; Azuma, M.; et al. Hepatitis E Virus (HEV) strains in serum samples can replicate efficiently in cultured cells despite the coexistence of HEV antibodies: Characterization of HEV virions in blood circulation. J. Clin. Microbiol. 2010, 48, 1112–1125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Yin, X.; Li, X.; Feng, Z. Role of Envelopment in the HEV Life Cycle. Viruses 2016, 8, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Allweiss, L.; Gass, S.; Giersch, K.; Groth, A.; Kah, J.; Volz, T.; Rapp, G.; Schobel, A.; Lohse, A.W.; Polywka, S.; et al. Human liver chimeric mice as a new model of chronic hepatitis E virus infection and preclinical drug evaluation. J. Hepatol. 2016, 64, 1033–1040. [Google Scholar] [CrossRef]
  16. Santos-Silva, S.; Hemnani, M.; Lopez-Lopez, P.; Goncalves, H.M.R.; Rivero-Juarez, A.; Van der Poel, W.H.M.; Nascimento, M.S.J.; Mesquita, J.R. A Systematic Review of Hepatitis E Virus Detection in Camels. Vet. Sci. 2023, 10, 323. [Google Scholar] [CrossRef]
  17. Pavio, N.; Meng, X.J.; Doceul, V. Zoonotic origin of hepatitis. Curr. Opin. Virol. 2015, 10, 34–41. [Google Scholar] [CrossRef]
  18. Salines, M.; Andraud, M.; Rose, N. From the epidemiology of hepatitis E virus (HEV) within the swine reservoir to public health risk mitigation strategies: A comprehensive review. Vet. Sci. 2017, 48, 31. [Google Scholar]
  19. Smith, D.B.; Simmonds, P.; Izopet, J.; Oliveira-Filho, E.F.; Ulrich, R.G.; Johne, R.; Koenig, M.; Jameel, S.; Harrison, T.J.; Meng, X.J.; et al. Proposed reference sequences for hepatitis E virus subtypes. J. Gen. Virol. 2016, 97, 537–542. [Google Scholar] [CrossRef]
  20. Smith, D.B.; Izopet, J.; Nicot, F.; Simmonds, P.; Jameel, S.; Meng, X.J.; Norder, H.; Okamoto, H.; van der Poel, W.H.M.; Reuter, G.; et al. Update: Proposed reference sequences for subtypes of hepatitis E virus (species Orthohepevirus A). J. Gen. Virol. 2020, 101, 692–698. [Google Scholar] [PubMed]
  21. Pallerla, S.R.; Harms, D.; Johne, R.; Todt, D.; Steinmann, E.; Schemmerer, M.; Wenzel, J.J.; Hofmann, J.; Shih, J.W.K.; Wedemeyer, H.; et al. Hepatitis E Virus Infection: Circulation, Molecular Epidemiology, and Impact on Global Health. Pathogens 2020, 9, 856. [Google Scholar] [CrossRef]
  22. Nelson, K.E.; Labrique, A.B.; Kmush, B.L. Epidemiology of Genotype 1 and 2 Hepatitis E Virus Infections. Cold Spring Harb. Perspect. Med. 2019, 9, a031732. [Google Scholar] [CrossRef]
  23. Wang, J.; Li, N.; Zhang, H.; Li, F.; Fanning, S.; Jiang, T. Detection of Hepatitis E Virus in the Pig Livers and Retail Pork Samples Collected in Selected Cities in China. Foodborne Pathog. Dis. 2021, 18, 97–103. [Google Scholar] [CrossRef]
  24. Sato, Y.; Sato, H.; Naka, K.; Furuya, S.; Tsukiji, H.; Kitagawa, K.; Sonoda, Y.; Usui, T.; Sakamoto, H.; Yoshino, S.; et al. A nationwide survey of hepatitis E virus (HEV) infection in wild boars in Japan: Identification of boar HEV strains of genotypes 3 and 4 and unrecognized genotypes. Arch. Virol. 2011, 156, 1345–1358. [Google Scholar] [CrossRef]
  25. Woo, P.C.; Lau, S.K.; Teng, J.L.; Cao, K.Y.; Wernery, U.; Schountz, T.; Chiu, T.H.; Tsang, A.K.; Wong, P.C.; Wong, E.Y.; et al. New Hepatitis E Virus Genotype in Bactrian Camels, Xinjiang, China, 2013. Emerg. Infect. Dis. 2016, 22, 2219–2221. [Google Scholar] [CrossRef]
  26. Hoofnagle, J.H.; Nelson, K.E.; Purcell, R.H. Hepatitis E. N. Engl. J. Med. 2012, 367, 1237–1244. [Google Scholar] [CrossRef] [PubMed]
  27. Inoue, K.; Yoshiba, M.; Yotsuyanagi, H.; Otsuka, T.; Sekiyama, K.; Fujita, R. Chronic hepatitis A with persistent viral replication. J. Med. Virol. 1996, 50, 322–324. [Google Scholar] [CrossRef]
  28. Dalton, H.R.; Bendall, R.P.; Keane, F.E.; Tedder, R.S.; Ijaz, S. Persistent carriage of hepatitis E virus in patients with HIV infection. N. Engl. J. Med. 2009, 361, 1025–1027. [Google Scholar] [CrossRef]
  29. Jin, H.; Zhao, Y.; Zhang, X.; Wang, B.; Liu, P. Case-fatality risk of pregnant women with acute viral hepatitis type E: A systematic review and meta-analysis. Epidemiol. Infect. 2016, 144, 2098–2106. [Google Scholar] [CrossRef] [Green Version]
  30. Westbrook, R.H.; Dusheiko, G.; Williamson, C. Pregnancy and liver disease. J. Hepatol. 2016, 64, 933–945. [Google Scholar] [CrossRef] [Green Version]
  31. 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] [PubMed] [Green Version]
  32. Wang, B.; Tian, D.; Sooryanarain, H.; Mahsoub, H.M.; Heffron, C.L.; Hassebroek, A.M.; Meng, X.J. Two mutations in the ORF1 of genotype 1 hepatitis E virus enhance virus replication and may associate with fulminant hepatic failure. Proc. Natl. Acad. Sci. USA 2022, 119, e2207503119. [Google Scholar] [CrossRef]
  33. Peron, J.M.; Bureau, C.; Poirson, H.; Mansuy, J.M.; Alric, L.; Selves, J.; Dupuis, E.; Izopet, J.; Vinel, J.P. Fulminant liver failure from acute autochthonous hepatitis E in France: Description of seven patients with acute hepatitis E and encephalopathy. J. Viral Hepat. 2007, 14, 298–303. [Google Scholar] [CrossRef] [PubMed]
  34. Field, Z.; Russin, M.; Murillo Alvarez, R.M.; Madruga, M.; Carlan, S. Acute Hepatitis E: A Rare Cause of Acute Liver Failure in a Patient With Acute Myeloid Leukemia. Cureus 2020, 12, e10628. [Google Scholar] [CrossRef]
  35. Malik, H.; Malik, H.; Uderani, M.; Berhanu, M.; Soto, C.J.; Saleem, F. Fulminant Hepatitis A and E Co-infection Leading to Acute Liver Failure: A Case Report. Cureus 2023, 15, e38101. [Google Scholar] [CrossRef]
  36. Wu, C.; Wu, X.; Xia, J. Hepatitis E virus infection during pregnancy. Virol. J. 2020, 17, 73. [Google Scholar] [CrossRef]
  37. Qian, Z.; Li, T.; Zhang, Y.; Chen, S.; Zhang, H.; Mickael, H.K.; Xiu, D.; Xia, Y.; Cong, C.; Xu, L.; et al. Prevalence of hepatitis E virus and its association with adverse pregnancy outcomes in pregnant women in China. J. Clin. Virol. 2023, 158, 105353. [Google Scholar] [CrossRef] [PubMed]
  38. Li, M.; Bu, Q.; Gong, W.; Li, H.; Wang, L.; Li, S.; Sridhar, S.; Cy Woo, P.; Wang, L. Hepatitis E virus infection and its associated adverse feto-maternal outcomes among pregnant women in Qinhuangdao, China. J. Matern-Fetal Neonatal Med. 2020, 33, 3647–3651. [Google Scholar] [CrossRef]
  39. Kamar, N.; Bendall, R.P.; Peron, J.M.; Cintas, P.; Prudhomme, L.; Mansuy, J.M.; Rostaing, L.; Keane, F.; Ijaz, S.; Izopet, J.; et al. Hepatitis E virus and neurologic disorders. Emerg. Infect. Dis. 2011, 17, 173–179. [Google Scholar] [CrossRef] [PubMed]
  40. Betts, R.; de Blackburn, C.W. Detecting pathogens in food. In Foodborne Pathogens, Hazards, Risk Analysis and Control, 2nd ed.; Woodhead Publishing Series in Food Science, Technology and Nutrition; Woodhead Publishing Limited: Cambridge, UK, 2009; pp. 17–65. [Google Scholar]
  41. Koopmans, M.; Duizer, E. Foodborne viruses: An emerging problem. Int. J. Food Microbiol. 2004, 90, 23–41. [Google Scholar] [CrossRef]
  42. Kingsley, D.H.; Chen, H. Influence of pH, salt, and temperature on pressure inactivation of hepatitis A virus. Int. J. Food Microbiol. 2009, 130, 61–64. [Google Scholar] [CrossRef]
  43. Siegl, G.; Weitz, M.; Kronauer, G. Stability of hepatitis A virus. Intervirology 1984, 22, 218–226. [Google Scholar] [CrossRef] [PubMed]
  44. Rzezutka, A.; Cook, N. Survival of human enteric viruses in the environment and food. FEMS Microbiol. Rev. 2004, 28, 441–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Wolff, A.; Gunther, T.; Johne, R. Stability of Hepatitis E Virus after Drying on Different Surfaces. Food Environ. Virol. 2022, 14, 138–148. [Google Scholar] [CrossRef] [PubMed]
  46. Ndumbi, P.; Freidl, G.S.; Williams, C.J.; Mardh, O.; Varela, C.; Avellon, A.; Friesema, I.; Vennema, H.; Beebeejaun, K.; Ngui, S.L.; et al. Hepatitis A outbreak disproportionately affecting men who have sex with men (MSM) in the European Union and European Economic Area, June 2016 to May 2017. Eurosurveillance 2018, 23, 1700641. [Google Scholar] [CrossRef] [Green Version]
  47. Urbanus, A.T.; van Houdt, R.; van de Laar, T.J.; Coutinho, R.A. Viral hepatitis among men who have sex with men, epidemiology and public health consequences. Eurosurveillance 2009, 14, 19421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Elinav, E.; Ben-Dov, I.Z.; Shapira, Y.; Daudi, N.; Adler, R.; Shouval, D.; Ackerman, Z. Acute hepatitis A infection in pregnancy is associated with high rates of gestational complications and preterm labor. Gastroenterology 2006, 130, 1129–1134. [Google Scholar] [CrossRef]
  49. Leikin, E.; Lysikiewicz, A.; Garry, D.; Tejani, N. Intrauterine transmission of hepatitis A virus. Obstet. Gynecol. 1996, 88 (4 Pt 2), 690–691. [Google Scholar] [CrossRef]
  50. Kroneman, A.; de Sousa, R.; Verhoef, L.; Koopmans, M.P.G.; Vennema, H.; HAVNet Network. Usability of the international HAVNet hepatitis A virus database for geographical annotation, backtracing and outbreak detection. Eurosurveillance 2018, 23, 1700802. [Google Scholar] [CrossRef] [Green Version]
  51. Kokkinos, P.; Kozyra, I.; Lazic, S.; Bouwknegt, M.; Rutjes, S.; Willems, K.; Moloney, R.; de Roda Husman, A.M.; Kaupke, A.; Legaki, E.; et al. Harmonised investigation of the occurrence of human enteric viruses in the leafy green vegetable supply chain in three European countries. Food Environ. Virol. 2012, 4, 179–191. [Google Scholar] [CrossRef]
  52. Koff, R.S. Hepatitis A. Lancet 1998, 351, 1643–1649. [Google Scholar] [CrossRef]
  53. Garcia Vilaplana, T.; Leeman, D.; Balogun, K.; Ngui, S.L.; Phipps, E.; Khan, W.M.; Incident, T.; Balasegaram, S. Hepatitis A outbreak associated with consumption of dates, England and Wales, January 2021 to April 2021. Eurosurveillance 2021, 26, 2100432. [Google Scholar] [CrossRef]
  54. O’Neill, C.; Franklin, N.; Edwards, A.; Martin, T.; O’Keefe, J.; Jackson, K.; Pingault, N.; Glasgow, K. Hepatitis A outbreak in Australia linked to imported Medjool dates, June–September 2021. Commun. Dis. Intell. (2018) 2022, 46, 1–12. [Google Scholar] [CrossRef] [PubMed]
  55. Jeong, H.W.; Kim, M.K.; Yi, H.J.; Kim, D.M.; Jeon, S.J.; Lee, H.K.; Oh, Y.H.; Hwang, Y.O. Hepatitis A virus strains identified in jogaejeot associated with outbreaks in Seoul, South Korea. Lett. Appl. Microbiol. 2021, 73, 107–112. [Google Scholar] [CrossRef] [PubMed]
  56. McClure, M.; Nsubuga, J.; Montgomery, M.P.; Jenkins, E.; Crosby, A.; Schoelen, D.; Basler, C.; Ramachandran, S.; Lin, Y.; Xia, G.L.; et al. A 2019 Outbreak Investigation of Hepatitis A Virus Infections in the United States Linked to Imported Fresh Blackberries. Food Environ. Virol. 2022, 14, 236–245. [Google Scholar] [CrossRef]
  57. Ruscher, C.; Faber, M.; Werber, D.; Stark, K.; Bitzegeio, J.; Michaelis, K.; Sagebiel, D.; Wenzel, J.J.; Enkelmann, J. Resurgence of an international hepatitis A outbreak linked to imported frozen strawberries, Germany, 2018 to 2020. Eurosurveillance 2020, 25, 1900670. [Google Scholar] [CrossRef]
  58. Enkirch, T.; Eriksson, R.; Persson, S.; Schmid, D.; Aberle, S.W.; Lof, E.; Wittesjo, B.; Holmgren, B.; Johnzon, C.; Gustafsson, E.X.; et al. Hepatitis A outbreak linked to imported frozen strawberries by sequencing, Sweden and Austria, June to September 2018. Eurosurveillance 2018, 23, 1800528. [Google Scholar] [CrossRef] [Green Version]
  59. Franklin, N.; Camphor, H.; Wright, R.; Stafford, R.; Glasgow, K.; Sheppeard, V. Outbreak of hepatitis A genotype IB in Australia associated with imported frozen pomegranate arils. Epidemiol. Infect. 2019, 147, e74. [Google Scholar] [CrossRef] [Green Version]
  60. Mollers, M.; Boxman, I.L.A.; Vennema, H.; Slegers-Fitz-James, I.A.; Brandwagt, D.; Friesema, I.H.; Batstra, J.S.; Te Wierik, M.J.M. Successful Use of Advertisement Pictures to Assist Recall in a Food-Borne Hepatitis A Outbreak in The Netherlands, 2017. Food Environ. Virol. 2018, 10, 272–277. [Google Scholar] [CrossRef] [Green Version]
  61. Viray, M.A.; Hofmeister, M.G.; Johnston, D.I.; Krishnasamy, V.P.; Nichols, C.; Foster, M.A.; Balajadia, R.; Wise, M.E.; Manuzak, A.; Lin, Y.; et al. Public health investigation and response to a hepatitis A outbreak from imported scallops consumed raw-Hawaii, 2016. Epidemiol. Infect. 2018, 147, e28. [Google Scholar] [CrossRef] [Green Version]
  62. Hu, X.; Collier, M.G.; Xu, F. Hepatitis A Outbreaks in Developed Countries: Detection, Control, and Prevention. Foodborne Pathog. Dis. 2020, 17, 166–171. [Google Scholar] [CrossRef] [PubMed]
  63. Bozkurt, H.; Phan-Thien, K.Y.; van Ogtrop, F.; Bell, T.; McConchie, R. Outbreaks, occurrence, and control of norovirus and hepatitis a virus contamination in berries: A review. Crit. Rev. Food Sci. Nutr. 2021, 61, 116–138. [Google Scholar] [CrossRef] [PubMed]
  64. Parker, J.A.; Kurien, T.T.; Huppatz, C. Hepatitis A outbreak associated with kava drinking. Commun. Dis. Intell. Q. Rep. 2014, 38, E26–E28. [Google Scholar] [PubMed]
  65. Severi, E.; Verhoef, L.; Thornton, L.; Guzman-Herrador, B.R.; Faber, M.; Sundqvist, L.; Rimhanen-Finne, R.; Roque-Afonso, A.M.; Ngui, S.L.; Allerberger, F.; et al. Large and prolonged food-borne multistate hepatitis A outbreak in Europe associated with consumption of frozen berries, 2013 to 2014. Eurosurveillance 2015, 20, 21192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Scavia, G.; Alfonsi, V.; Taffon, S.; Escher, M.; Bruni, R.; Medici, D.; Pasquale, S.D.; Guizzardi, S.; Cappelletti, B.; Iannazzo, S.; et al. National Italian Task Force On Hepatitis A. A large prolonged outbreak of hepatitis A associated with consumption of frozen berries, Italy, 2013–2014. J. Med. Microbiol. 2017, 66, 342–349. [Google Scholar] [CrossRef]
  67. Gossner, C.M.; Severi, E. Three simultaneous, food-borne, multi-country outbreaks of hepatitis A virus infection reported in EPIS-FWD in 2013: What does it mean for the European Union? Eurosurveillance 2014, 19, 20941. [Google Scholar] [CrossRef] [Green Version]
  68. Guzman-Herrador, B.R.; Panning, M.; Stene-Johansen, K.; Borgen, K.; Einoder-Moreno, M.; Huzly, D.; Jensvoll, L.; Lange, H.; Maassen, S.; Myking, S.; et al. Importance of molecular typing in confirmation of the source of a national hepatitis A virus outbreak in Norway and the detection of a related cluster in Germany. Arch. Virol. 2015, 160, 2823–2826. [Google Scholar] [CrossRef]
  69. C Nordic Outbreak Investigation Team. Joint analysis by the Nordic countries of a hepatitis A outbreak, October 2012 to June 2013: Frozen strawberries suspected. Eurosurveillance 2013, 18, 20520. [Google Scholar]
  70. Swinkels, H.M.; Kuo, M.; Embree, G.; Fraser Health Environmental Health Investigation Team; Andonov, A.; Henry, B.; Buxton, J.A. Hepatitis A outbreak in British Columbia, Canada: The roles of established surveillance, consumer loyalty cards and collaboration, February to May 2012. Eurosurveillance 2014, 19, 20792. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Boxman, I.L.; Verhoef, L.; Vennema, H.; Ngui, S.L.; Friesema, I.H.; Whiteside, C.; Lees, D.; Koopmans, M. International linkage of two food-borne hepatitis A clusters through traceback of mussels, the Netherlands, 2012. Eurosurveillance 2016, 21, 30113. [Google Scholar] [CrossRef] [Green Version]
  72. Fournet, N.; Baas, D.; van Pelt, W.; Swaan, C.; Ober, H.; Isken, L.; Cremer, J.; Friesema, I.; Vennema, H.; Boxman, I.; et al. Another possible food-borne outbreak of hepatitis A in the Netherlands indicated by two closely related molecular sequences, July to October 2011. Eurosurveillance 2012, 17, 20079. [Google Scholar] [CrossRef]
  73. Carvalho, C.; Thomas, H.; Balogun, K.; Tedder, R.; Pebody, R.; Ramsay, M.; Ngui, S. A possible outbreak of hepatitis A associated with semi-dried tomatoes, England, July-November 2011. Eurosurveillance 2012, 17, 20083. [Google Scholar] [CrossRef]
  74. Petrignani, M.; Verhoef, L.; Vennema, H.; van Hunen, R.; Baas, D.; van Steenbergen, J.E.; Koopmans, M.P. Underdiagnosis of foodborne hepatitis A, The Netherlands, 2008–2010(1.). Emerg. Infect. Dis. 2014, 20, 596–602. [Google Scholar] [CrossRef]
  75. Gallot, C.; Grout, L.; Roque-Afonso, A.M.; Couturier, E.; Carrillo-Santisteve, P.; Pouey, J.; Letort, M.J.; Hoppe, S.; Capdepon, P.; Saint-Martin, S.; et al. Hepatitis A associated with semidried tomatoes, France, 2010. Emerg. Infect. Dis. 2011, 17, 566–567. [Google Scholar] [CrossRef]
  76. Donnan, E.J.; Fielding, J.E.; Gregory, J.E.; Lalor, K.; Rowe, S.; Goldsmith, P.; Antoniou, M.; Fullerton, K.E.; Knope, K.; Copland, J.G.; et al. A multistate outbreak of hepatitis A associated with semidried tomatoes in Australia, 2009. Clin. Infect. Dis. 2012, 54, 775–781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Papafragkou, E.; Kulka, M. Review: Approaches to the Viral Extraction, Detection, and Identification of Hepatitis Viruses, HAV and HEV, in Foods. J. AOAC Int. 2016, 99, 130–142. [Google Scholar] [CrossRef]
  78. Pinto, R.M.; Costafreda, M.I.; Bosch, A. Risk assessment in shellfish-borne outbreaks of hepatitis A. Appl. Environ. Microbiol. 2009, 75, 7350–7355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Yoon, Y.K.; Chun, B.C.; Lee, H.K.; Seo, Y.S.; Shin, J.H.; Hong, Y.S.; Sim, H.S.; Kim, J.Y.; Kim, J.Y.; Park, Y.S.; et al. Epidemiological and genetic analysis of a sustained community-wide outbreak of hepatitis A in the Republic of Korea, 2008: A hospital-based case-control study. J. Clin. Virol. 2009, 46, 184–188. [Google Scholar] [CrossRef] [PubMed]
  80. Costantino, A.; Coppola, N.; Spada, E.; Bruni, R.; Taffon, S.; Equestre, M.; Marcantonio, C.; Sagnelli, C.; Dell’Isola, C.; Tosone, G.; et al. Hepatitis A virus strains circulating during 1997–2015 in Campania, a Southern Italy region with periodic outbreaks. J. Med. Virol. 2017, 89, 1931–1936. [Google Scholar] [CrossRef]
  81. Bellou, M.; Kokkinos, P.; Vantarakis, A. Shellfish-borne viral outbreaks: A systematic review. Food Environ. Virol. 2013, 5, 13–23. [Google Scholar] [CrossRef]
  82. European Centre for Disease Prevention and Control. Hepatitis A. In ECDC. Annual Epidemiological Report for 2021; ECDC: Stockholm, Sweden, 2022. [Google Scholar]
  83. Dirks, R.A.M.; Jansen, C.C.C.; Hagele, G.; Zwartkruis-Nahuis, A.J.T.; Tijsma, A.S.L.; Boxman, I.L.A. Quantitative levels of norovirus and hepatitis A virus in bivalve molluscs collected along the food chain in the Netherlands, 2013–2017. Int. J. Food Microbiol. 2021, 344, 109089. [Google Scholar] [CrossRef]
  84. Fusco, G.; Anastasio, A.; Kingsley, D.H.; Amoroso, M.G.; Pepe, T.; Fratamico, P.M.; Cioffi, B.; Rossi, R.; La Rosa, G.; Boccia, F. Detection of Hepatitis A Virus and Other Enteric Viruses in Shellfish Collected in the Gulf of Naples, Italy. Int. J. Environ. Res. Public Health 2019, 16, 2588. [Google Scholar] [CrossRef] [Green Version]
  85. Purpari, G.; Macaluso, G.; Di Bella, S.; Gucciardi, F.; Mira, F.; Di Marco, P.; Lastra, A.; Petersen, E.; La Rosa, G.; Guercio, A. Molecular characterization of human enteric viruses in food, water samples, and surface swabs in Sicily. Int. J. Infect. Dis. 2019, 80, 66–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Suffredini, E.; Proroga, Y.T.R.; Di Pasquale, S.; Di Maro, O.; Losardo, M.; Cozzi, L.; Capuano, F.; De Medici, D. Occurrence and Trend of Hepatitis A Virus in Bivalve Molluscs Production Areas Following a Contamination Event. Food Environ. Virol. 2017, 9, 423–433. [Google Scholar] [CrossRef] [PubMed]
  87. Albert Bosch, R.M.P.; Xavier Abad, F. Survival and transport of enteric viruses in the environment. In Viruses in Foods; Goyal, S.M., Ed.; Springer: Berlin/Heidelberg, Germany, 2006; pp. 151–187. [Google Scholar]
  88. Somani, S.K.; Aggarwal, R.; Naik, S.R.; Srivastava, S.; Naik, S. A serological study of intrafamilial spread from patients with sporadic hepatitis E virus infection. J. Viral Hepat. 2003, 10, 446–449. [Google Scholar] [CrossRef] [PubMed]
  89. Bi, H.; Yang, R.; Wu, C.; Xia, J. Hepatitis E virus and blood transfusion safety. Epidemiol. Infect. 2020, 148, e158. [Google Scholar] [CrossRef] [PubMed]
  90. Khuroo, M.S.; Kamili, S.; Jameel, S. Vertical transmission of hepatitis E virus. Lancet 1995, 345, 1025–1026. [Google Scholar] [CrossRef]
  91. Singh, S.; Mohanty, A.; Joshi, Y.K.; Deka, D.; Mohanty, S.; Panda, S.K. Mother-to-child transmission of hepatitis E virus infection. Indian J. Pediatr. 2003, 70, 37–39. [Google Scholar] [CrossRef]
  92. Tripathy, A.S.; Sharma, M.; Deoshatwar, A.R.; Babar, P.; Bharadwaj, R.; Bharti, O.K. Study of a hepatitis E virus outbreak involving drinking water and sewage contamination in Shimla, India, 2015–2016. Trans. R. Soc. Trop. Med. Hyg. 2019, 113, 789–796. [Google Scholar] [CrossRef]
  93. Malhotra, B.; Deba, F.; Sharma, P.; Trivedi, K.; Tiwari, J.; Chouhan, A.; Singh, R.; Meena, D.; Verma, H.N. Hepatitis E outbreak in Jaipur due to Genotype IA. Indian J. Med. Microbiol. 2020, 38, 46–51. [Google Scholar] [CrossRef]
  94. Baki, A.A.; Haque, W.; Giti, S.; Khan, A.A.; Rahman, M.M.; Jubaida, N.; Rahman, M. Hepatitis E virus genotype 1f outbreak in Bangladesh, 2018. J. Med. Virol. 2021, 93, 5177–5181. [Google Scholar] [CrossRef]
  95. Chen, Y.J.; Cao, N.X.; Xie, R.H.; Ding, C.X.; Chen, E.F.; Zhu, H.P.; Sun, J.M.; Shang, X.P.; Wang, X.X.; Miao, Z.P. Epidemiological investigation of a tap water-mediated hepatitis E virus genotype 4 outbreak in Zhejiang Province, China. Epidemiol. Infect. 2016, 144, 3387–3399. [Google Scholar] [CrossRef] [Green Version]
  96. Miura, T.; Lhomme, S.; Le Saux, J.C.; Le Mehaute, P.; Guillois, Y.; Couturier, E.; Izopet, J.; Abranavel, F.; Le Guyader, F.S. Detection of Hepatitis E Virus in Sewage after an Outbreak on a French Island. Food Environ. Virol. 2016, 8, 194–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Di Profio, F.; Melegari, I.; Palombieri, A.; Sarchese, V.; Arbuatti, A.; Fruci, P.; Marsilio, F.; Martella, V.; Di Martino, B. High prevalence of hepatitis E virus in raw sewage in Southern Italy. Virus Res. 2019, 272, 197710. [Google Scholar] [CrossRef] [PubMed]
  98. Mesquita, J.R.; Oliveira, D.; Rivadulla, E.; Abreu-Silva, J.; Varela, M.F.; Romalde, J.L.; Nascimento, M.S. Hepatitis E virus genotype 3 in mussels (Mytilus galloprovinciallis), Spain. Food Microbiol. 2016, 58, 13–15. [Google Scholar] [CrossRef] [PubMed]
  99. Iaconelli, M.; Purpari, G.; Della Libera, S.; Petricca, S.; Guercio, A.; Ciccaglione, A.R.; Bruni, R.; Taffon, S.; Equestre, M.; Fratini, M.; et al. Hepatitis A and E Viruses in Wastewaters, in River Waters, and in Bivalve Molluscs in Italy. Food Environ. Virol. 2015, 7, 316–324. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, H.; Kjellberg, I.; Sikora, P.; Rydberg, H.; Lindh, M.; Bergstedt, O.; Norder, H. Hepatitis E virus genotype 3 strains and a plethora of other viruses detected in raw and still in tap water. Water Res. 2020, 168, 115141. [Google Scholar] [CrossRef]
  101. Salvador, D.; Neto, C.; Benoliel, M.J.; Caeiro, M.F. Assessment of the Presence of Hepatitis E virus in Surface Water and Drinking Water in Portugal. Microorganisms 2020, 8, 761. [Google Scholar] [CrossRef]
  102. Lhomme, S.; Magne, S.; Perelle, S.; Vaissiere, E.; Abravanel, F.; Trelon, L.; Hennechart-Collette, C.; Fraisse, A.; Martin-Latil, S.; Izopet, J.; et al. Clustered Cases of Waterborne Hepatitis E Virus Infection, France. Viruses 2023, 15, 1149. [Google Scholar] [CrossRef]
  103. Masuda, J.; Yano, K.; Tamada, Y.; Takii, Y.; Ito, M.; Omagari, K.; Kohno, S. Acute hepatitis E of a man who consumed wild boar meat prior to the onset of illness in Nagasaki, Japan. Hepatol. Res. 2005, 31, 178–183. [Google Scholar] [CrossRef]
  104. Meng, X.J. Hepatitis E virus: Animal reservoirs and zoonotic risk. Vet. Microbiol. 2010, 140, 256–265. [Google Scholar] [CrossRef] [Green Version]
  105. Ivanova, A.; Tefanova, V.; Reshetnjak, I.; Kuznetsova, T.; Geller, J.; Lundkvist, A.; Janson, M.; Neare, K.; Velstrom, K.; Jokelainen, P.; et al. Hepatitis E Virus in Domestic Pigs, Wild Boars, Pig Farm Workers, and Hunters in Estonia. Food Environ. Virol. 2015, 7, 403–412. [Google Scholar] [CrossRef]
  106. Monini, M.; Di Bartolo, I.; Ianiro, G.; Angeloni, G.; Magistrali, C.F.; Ostanello, F.; Ruggeri, F.M. Detection and molecular characterization of zoonotic viruses in swine fecal samples in Italian pig herds. Arch. Virol. 2015, 160, 2547–2556. [Google Scholar] [CrossRef]
  107. Chelli, E.; Suffredini, E.; De Santis, P.; De Medici, D.; Di Bella, S.; D’Amato, S.; Gucciardi, F.; Guercio, A.; Ostanello, F.; Perrone, V.; et al. Hepatitis E Virus Occurrence in Pigs Slaughtered in Italy. Animals 2021, 11, 277. [Google Scholar] [CrossRef]
  108. Krog, J.S.; Larsen, L.E.; Breum, S.O. Tracing Hepatitis E Virus in Pigs From Birth to Slaughter. Front. Vet. Sci. 2019, 6, 50. [Google Scholar] [CrossRef] [Green Version]
  109. Wang, H.; Karlsson, M.; Lindberg, M.; Nystrom, K.; Norder, H. Hepatitis E virus strains infecting Swedish domestic pigs are unique for each pig farm and remain in the farm for at least 2 years. Transbound. Emerg. Dis. 2019, 66, 1314–1323. [Google Scholar] [CrossRef] [Green Version]
  110. Boxman, I.L.A.; Verhoef, L.; Dop, P.Y.; Vennema, H.; Dirks, R.A.M.; Opsteegh, M. High prevalence of acute hepatitis E virus infection in pigs in Dutch slaughterhouses. Int. J. Food Microbiol. 2022, 379, 109830. [Google Scholar] [CrossRef]
  111. Ianiro, G.; Monini, M.; De Sabato, L.; Chelli, E.; Cerini, N.; Ostanello, F.; Di Bartolo, I. Dynamic of Hepatitis E Virus (HEV) Shedding in Pigs. Animals 2022, 12, 1063. [Google Scholar] [CrossRef]
  112. Norder, H.; Karlsson, M.; Mellgren, A.; Konar, J.; Sandberg, E.; Lasson, A.; Castedal, M.; Magnius, L.; Lagging, M. Diagnostic Performance of Five Assays for Anti-Hepatitis E Virus IgG and IgM in a Large Cohort Study. J. Clin. Microbiol. 2016, 54, 549–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Wilhelm, B.; Waddell, L.; Greig, J.; Young, I. Systematic review and meta-analysis of the seroprevalence of hepatitis E virus in the general population across non-endemic countries. PLoS ONE 2019, 14, e0216826. [Google Scholar] [CrossRef] [PubMed]
  114. Faber, M.S.; Wenzel, J.J.; Jilg, W.; Thamm, M.; Hohle, M.; Stark, K. Hepatitis E virus seroprevalence among adults, Germany. Emerg. Infect. Dis. 2012, 18, 1654–1657. [Google Scholar] [CrossRef]
  115. Lange, H.; Overbo, J.; Borgen, K.; Dudman, S.; Hoddevik, G.; Urdahl, A.M.; Vold, L.; Sjurseth, S.K. Hepatitis E in Norway: Seroprevalence in humans and swine. Epidemiol. Infect. 2017, 145, 181–186. [Google Scholar] [CrossRef] [PubMed]
  116. Krumbholz, A.; Joel, S.; Dremsek, P.; Neubert, A.; Johne, R.; Durrwald, R.; Walther, M.; Muller, T.H.; Kuhnel, D.; Lange, J.; et al. Seroprevalence of hepatitis E virus (HEV) in humans living in high pig density areas of Germany. Med. Microbiol. Immunol. 2014, 203, 273–282. [Google Scholar] [CrossRef]
  117. Olsen, B.; Axelsson-Olsson, D.; Thelin, A.; Weiland, O. Unexpected high prevalence of IgG-antibodies to hepatitis E virus in Swedish pig farmers and controls. Scand. J. Infect. Dis. 2006, 38, 55–58. [Google Scholar] [CrossRef]
  118. Withers, M.R.; Correa, M.T.; Morrow, M.; Stebbins, M.E.; Seriwatana, J.; Webster, W.D.; Boak, M.B.; Vaughn, D.W. Antibody levels to hepatitis E virus in North Carolina swine workers, non-swine workers, swine, and murids. Am. J. Trop. Med. Hyg. 2002, 66, 384–388. [Google Scholar] [CrossRef] [Green Version]
  119. Galiana, C.; Fernandez-Barredo, S.; Garcia, A.; Gomez, M.T.; Perez-Gracia, M.T. Occupational exposure to hepatitis E virus (HEV) in swine workers. Am. J. Trop. Med. Hyg. 2008, 78, 1012–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Hoan, N.X.; Huy, P.X.; Sy, B.T.; Meyer, C.G.; Son, T.V.; Binh, M.T.; Giang, D.P.; Tu Anh, D.; Bock, C.T.; Wang, B.; et al. High Hepatitis E virus (HEV) Positivity Among Domestic Pigs and Risk of HEV Infection of Individuals Occupationally Exposed to Pigs and Pork Meat in Hanoi, Vietnam. Open Forum Infect. Dis. 2019, 6, ofz306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Wu, J.Y.; Lau, E.H.Y.; Lu, M.L.; Guo, C.; Guo, Z.M.; Yuan, J.; Lu, J.H. An occupational risk of hepatitis E virus infection in the workers along the meat supply chains in Guangzhou, China. One Health 2022, 14, 100376. [Google Scholar] [CrossRef]
  122. Di Cola, G.; Fantilli, A.C.; Pisano, M.B.; Re, V.E. Foodborne transmission of hepatitis A and hepatitis E viruses: A literature review. Int. J. Food Microbiol. 2021, 338, 108986. [Google Scholar] [CrossRef]
  123. Garbuglia, A.R.; Bruni, R.; Villano, U.; Vairo, F.; Lapa, D.; Madonna, E.; Picchi, G.; Binda, B.; Mariani, R.; De Paulis, F.; et al. Hepatitis E Outbreak in the Central Part of Italy Sustained by Multiple HEV Genotype 3 Strains, June-December 2019. Viruses 2021, 13, 1159. [Google Scholar] [CrossRef]
  124. Moro, O.; Suffredini, E.; Isopi, M.; Tosti, M.E.; Schembri, P.; Scavia, G. Quantitative Methods for the Prioritization of Foods Implicated in the Transmission of Hepatititis E to Humans in Italy. Foods 2021, 11, 87. [Google Scholar] [CrossRef]
  125. Thippornchai, N.; Leaungwutiwong, P.; Kosoltanapiwat, N.; Vuong, C.; Nguyen, K.; Okabayashi, T.; Lee, A. Survey of hepatitis E virus in pork products and pig stools in Nakhon Pathom Province, Thailand. Vet. Med. Sci. 2022, 8, 1975–1981. [Google Scholar] [CrossRef] [PubMed]
  126. Soares, V.M.; Dos Santos, E.A.R.; Tadielo, L.E.; Cerqueira-Cezar, C.K.; da Cruz Encide Sampaio, A.N.; Eisen, A.K.A.; de Oliveira, K.G.; Padilha, M.B.; de Moraes Guerra, M.E.; Gasparetto, R.; et al. Detection of adenovirus, rotavirus, and hepatitis E virus in meat cuts marketed in Uruguaiana, Rio Grande do Sul, Brazil. One Health 2022, 14, 100377. [Google Scholar] [CrossRef] [PubMed]
  127. Locus, T.; Lambrecht, E.; Peeters, M.; Suin, V.; Verhaegen, B.; Van Hoorde, K.; Lamoral, S.; Vanwolleghem, T.; Van Gucht, S. Hepatitis E virus in pork meat products and exposure assessment in Belgium. Int. J. Food Microbiol. 2023, 397, 110198. [Google Scholar] [CrossRef]
  128. Harrison, L.; Ramos, T.M.; Wu, X.; DiCaprio, E. Presence of hepatitis E virus in commercially available pork products. Int. J. Food Microbiol. 2021, 339, 109033. [Google Scholar] [CrossRef]
  129. Boxman, I.L.A.; Jansen, C.C.C.; Zwartkruis-Nahuis, A.J.T.; Hagele, G.; Sosef, N.P.; Dirks, R.A.M. Detection and quantification of hepatitis E virus RNA in ready to eat raw pork sausages in the Netherlands. Int. J. Food Microbiol. 2020, 333, 108791. [Google Scholar] [CrossRef]
  130. Pallerla, S.R.; Schembecker, S.; Meyer, C.G.; Linh, L.T.K.; Johne, R.; Wedemeyer, H.; Bock, C.T.; Kremsner, P.G.; Velavan, T.P. Hepatitis E virus genome detection in commercial pork livers and pork meat products in Germany. J. Viral Hepat. 2021, 28, 196–204. [Google Scholar] [CrossRef] [PubMed]
  131. Garcia, N.; Hernandez, M.; Gutierrez-Boada, M.; Valero, A.; Navarro, A.; Munoz-Chimeno, M.; Fernandez-Manzano, A.; Escobar, F.M.; Martinez, I.; Barcena, C.; et al. Occurrence of Hepatitis E Virus in Pigs and Pork Cuts and Organs at the Time of Slaughter, Spain, 2017. Front. Microbiol. 2019, 10, 2990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Boxman, I.L.A.; Jansen, C.C.C.; Hagele, G.; Zwartkruis-Nahuis, A.; Tijsma, A.S.L.; Vennema, H. Monitoring of pork liver and meat products on the Dutch market for the presence of HEV RNA. Int. J. Food Microbiol. 2019, 296, 58–64. [Google Scholar] [CrossRef] [PubMed]
  133. Montone, A.M.I.; De Sabato, L.; Suffredini, E.; Alise, M.; Zaccherini, A.; Volzone, P.; Di Maro, O.; Neola, B.; Capuano, F.; Di Bartolo, I. Occurrence of HEV-RNA in Italian Regional Pork and Wild Boar Food Products. Food Environ. Virol. 2019, 11, 420–426. [Google Scholar] [CrossRef] [PubMed]
  134. Geng, Y.; Zhao, C.; Guo, T.; Xu, Y.; Wang, X.; Huang, W.; Liu, H.; Wang, Y. Detection of Hepatitis E Virus in Raw Pork and Pig Viscera as Food in Hebei Province of China. Foodborne Pathog. Dis. 2019, 16, 325–330. [Google Scholar] [CrossRef] [PubMed]
  135. Hao, X.; Cao, W.; Situ, J.; Zhao, Y.; Huang, F.; Yu, W. Hepatitis E Virus Detected in Pork Products. Food Environ. Virol. 2018, 10, 391–393. [Google Scholar] [CrossRef]
  136. Moor, D.; Liniger, M.; Baumgartner, A.; Felleisen, R. Screening of Ready-to-Eat Meat Products for Hepatitis E Virus in Switzerland. Food Environ. Virol. 2018, 10, 263–271. [Google Scholar] [CrossRef] [Green Version]
  137. Szabo, K.; Trojnar, E.; Anheyer-Behmenburg, H.; Binder, A.; Schotte, U.; Ellerbroek, L.; Klein, G.; Johne, R. Detection of hepatitis E virus RNA in raw sausages and liver sausages from retail in Germany using an optimized method. Int. J. Food Microbiol. 2015, 215, 149–156. [Google Scholar] [CrossRef]
  138. Pavio, N.; Merbah, T.; Thebault, A. Frequent hepatitis E virus contamination in food containing raw pork liver, France. Emerg. Infect. Dis. 2014, 20, 1925–1927. [Google Scholar] [CrossRef]
  139. Cook, N.; D’Agostino, M.; Johne, R. Potential Approaches to Assess the Infectivity of Hepatitis E Virus in Pork Products: A Review. Food Environ. Virol. 2017, 9, 243–255. [Google Scholar] [CrossRef]
  140. Johne, R.; Wolff, A.; Gadicherla, A.K.; Filter, M.; Schluter, O. Stability of hepatitis E virus at high hydrostatic pressure processing. Int. J. Food Microbiol. 2021, 339, 109013. [Google Scholar] [CrossRef]
  141. Rivero-Juarez, A.; Frias, M.; Martinez-Peinado, A.; Risalde, M.A.; Rodriguez-Cano, D.; Camacho, A.; Garcia-Bocanegra, I.; Cuenca-Lopez, F.; Gomez-Villamandos, J.C.; Rivero, A. Familial Hepatitis E Outbreak Linked to Wild Boar Meat Consumption. Zoonoses Public Health 2017, 64, 561–565. [Google Scholar] [CrossRef]
  142. Tei, S.; Kitajima, N.; Takahashi, K.; Mishiro, S. Zoonotic transmission of hepatitis E virus from deer to human beings. Lancet 2003, 362, 371–373. [Google Scholar] [CrossRef] [PubMed]
  143. Palombieri, A.; Robetto, S.; Di Profio, F.; Sarchese, V.; Fruci, P.; Bona, M.C.; Ru, G.; Orusa, R.; Marsilio, F.; Martella, V.; et al. Surveillance Study of Hepatitis E Virus (HEV) in Domestic and Wild Ruminants in Northwestern Italy. Animals 2020, 10, 2351. [Google Scholar] [CrossRef] [PubMed]
  144. Trogu, T.; Ferrari, N.; Formenti, N.; Filipello, V.; Pedrotti, L.; Vigano, R.; Lanfranchi, P.; Luzzago, C. Low Serologic Prevalences Suggest Sporadic Infections of Hepatitis E Virus in Chamois (Rupicapra rupicapra) and Red Deer (Cervus elaphus) in the Italian Alps. J. Wildl. Dis. 2020, 56, 443–446. [Google Scholar] [CrossRef] [PubMed]
  145. Di Bartolo, I.; Ponterio, E.; Angeloni, G.; Morandi, F.; Ostanello, F.; Nicoloso, S.; Ruggeri, F.M. Presence of Hepatitis E Virus in a RED Deer (Cervus elaphus) Population in Central Italy. Transbound. Emerg. Dis. 2017, 64, 137–143. [Google Scholar] [CrossRef] [PubMed]
  146. Fonti, N.; Pacini, M.I.; Forzan, M.; Parisi, F.; Periccioli, M.; Mazzei, M.; Poli, A. Molecular and Pathological Detection of Hepatitis E Virus in Roe Deer (Capreolus capreolus) and Fallow Deer (Dama dama) in Central Italy. Vet. Sci. 2022, 9, 100. [Google Scholar] [CrossRef]
  147. Spancerniene, U.; Grigas, J.; Buitkuviene, J.; Zymantiene, J.; Juozaitiene, V.; Stankeviciute, M.; Razukevicius, D.; Zienius, D.; Stankevicius, A. Prevalence and phylogenetic analysis of hepatitis E virus in pigs, wild boars, roe deer, red deer and moose in Lithuania. Acta Vet. Scand. 2018, 60, 13. [Google Scholar] [CrossRef] [Green Version]
  148. Sacristan, C.; Madslien, K.; Sacristan, I.; Klevar, S.; das Neves, C.G. Seroprevalence of Hepatitis E Virus in Moose (Alces alces), Reindeer (Rangifer tarandus), Red Deer (Cervus elaphus), Roe Deer (Capreolus capreolus), and Muskoxen (Ovibos moschatus) from Norway. Viruses 2021, 13, 224. [Google Scholar] [CrossRef]
  149. Lin, J.; Karlsson, M.; Olofson, A.S.; Belak, S.; Malmsten, J.; Dalin, A.M.; Widen, F.; Norder, H. High prevalence of hepatitis e virus in Swedish moose--a phylogenetic characterization and comparison of the virus from different regions. PLoS ONE 2015, 10, e0122102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Loikkanen, E.; Oristo, S.; Hamalainen, N.; Jokelainen, P.; Kantala, T.; Sukura, A.; Maunula, L. Antibodies against Hepatitis E Virus (HEV) in European Moose and White-Tailed Deer in Finland. Food Environ. Virol. 2020, 12, 333–341. [Google Scholar] [CrossRef]
  151. Bastos, C.; Eisen, A.K.A.; Demoliner, M.; Heldt, F.H.; Filippi, M.; de Abreu Goes Pereira, V.M.; Teixeira, T.A.M.; Roth, L.O.; Gularte, J.S.; Spilki, F.R. Hepatitis E virus genotype 3 in bovine livers slaughtered in the state of Rio Grande do Sul, Brazil. Braz. J. Microbiol. 2022, 53, 1115–1120. [Google Scholar] [CrossRef] [PubMed]
  152. Go, H.J.; Park, B.J.; Ahn, H.S.; Lyoo, E.L.; Kim, D.H.; Lee, J.B.; Park, S.Y.; Song, C.S.; Lee, S.W.; Choi, I.S. Identification of Hepatitis E Virus in Bovine and Porcine Raw Livers. J. Microbiol. Biotechnol. 2019, 29, 2022–2025. [Google Scholar] [CrossRef] [PubMed]
  153. Sarchese, V.; Di Profio, F.; Melegari, I.; Palombieri, A.; Sanchez, S.B.; Arbuatti, A.; Ciuffetelli, M.; Marsilio, F.; Martella, V.; Di Martino, B. Hepatitis E virus in sheep in Italy. Transbound. Emerg. Dis. 2019, 66, 1120–1125. [Google Scholar] [CrossRef]
  154. Sahli, R.; Fraga, M.; Semela, D.; Moradpour, D.; Gouttenoire, J. Rabbit HEV in immunosuppressed patients with hepatitis E acquired in Switzerland. J. Hepatol. 2019, 70, 1023–1025. [Google Scholar] [CrossRef] [Green Version]
  155. Klink, P.; Harms, D.; Altmann, B.; Dorffel, Y.; Morgera, U.; Zander, S.; Bock, C.T.; Hofmann, J. Molecular characterisation of a rabbit Hepatitis E Virus strain detected in a chronically HEV-infected individual from Germany. One Health 2023, 16, 100528. [Google Scholar] [CrossRef]
  156. Abravanel, F.; Lhomme, S.; El Costa, H.; Schvartz, B.; Peron, J.M.; Kamar, N.; Izopet, J. Rabbit Hepatitis E Virus Infections in Humans, France. Emerg. Infect. Dis. 2017, 23, 1191–1193. [Google Scholar] [CrossRef] [Green Version]
  157. Johne, R.; Plenge-Bonig, 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 (Pt 3), 750–758. [Google Scholar] [CrossRef]
  158. Sridhar, S.; Yip, C.C.; Wu, S.; Chew, N.F.; Leung, K.H.; Chan, J.F.; Zhao, P.S.; Chan, W.M.; Poon, R.W.; Tsoi, H.W.; et al. Transmission of Rat Hepatitis E Virus Infection to Humans in Hong Kong: A Clinical and Epidemiological Analysis. Hepatology 2021, 73, 10–22. [Google Scholar] [CrossRef] [PubMed]
  159. 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]
  160. Dremsek, P.; Wenzel, J.J.; Johne, R.; Ziller, M.; Hofmann, J.; Groschup, M.H.; Werdermann, S.; Mohn, U.; Dorn, S.; Motz, M.; et al. Seroprevalence study in forestry workers from eastern Germany using novel genotype 3- and rat hepatitis E virus-specific immunoglobulin G ELISAs. Med. Microbiol. Immunol. 2012, 201, 189–200. [Google Scholar] [CrossRef]
  161. Shimizu, K.; Hamaguchi, S.; Ngo, C.C.; Li, T.C.; Ando, S.; Yoshimatsu, K.; Yasuda, S.P.; Koma, T.; Isozumi, R.; Tsuda, Y.; et al. Serological evidence of infection with rodent-borne hepatitis E virus HEV-C1 or antigenically related virus in humans. J. Vet. Med. Sci. 2016, 78, 1677–1681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Rivero-Juarez, A.; Frias, M.; Perez, A.B.; Pineda, J.A.; Reina, G.; Fuentes-Lopez, A.; Freyre-Carrillo, C.; Ramirez-Arellano, E.; Alados, J.C.; Rivero, A.; et al. Orthohepevirus C infection as an emerging cause of acute hepatitis in Spain: First report in Europe. J. Hepatol. 2022, 77, 326–331. [Google Scholar] [CrossRef]
  163. Faber, M.; Wenzel, J.J.; Erl, M.; Stark, K.; Schemmerer, M. No Evidence for Orthohepevirus C in Archived Human Samples in Germany, 2000–2020. Viruses 2022, 14, 742. [Google Scholar] [CrossRef] [PubMed]
  164. Pankovics, P.; Nemethy, O.; Boros, A.; Par, G.; Szakaly, P.; Reuter, G. Four-year long (2014–2017) clinical and laboratory surveillance of hepatitis E virus infections using combined antibody, molecular, antigen and avidity detection methods: Increasing incidence and chronic HEV case in Hungary. J. Clin. Virol. 2020, 124, 104284. [Google Scholar] [CrossRef]
  165. Parraud, D.; Lhomme, S.; Peron, J.M.; Da Silva, I.; Tavitian, S.; Kamar, N.; Izopet, J.; Abravanel, F. Rat Hepatitis E Virus: Presence in Humans in South-Western France? Front. Med. 2021, 8, 726363. [Google Scholar] [CrossRef]
  166. Huang, F.; Li, Y.; Yu, W.; Jing, S.; Wang, J.; Long, F.; He, Z.; Yang, C.; Bi, Y.; Cao, W.; et al. Excretion of infectious hepatitis E virus into milk in cows imposes high risks of zoonosis. Hepatology 2016, 64, 350–359. [Google Scholar] [CrossRef] [Green Version]
  167. Demirci, M.; Yigin, A.; Unlu, O.; Kilic Altun, S. Detection of HEV RNA amounts and genotypes in raw milks obtained from different animals. Mikrobiyol. Bul. 2019, 53, 43–52. [Google Scholar] [CrossRef]
  168. El-Mokhtar, M.A.; Elkhawaga, A.A.; Sayed, I.M. Assessment of hepatitis E virus (HEV) in the edible goat products pointed out a risk for human infection in Upper Egypt. Int. J. Food Microbiol. 2020, 330, 108784. [Google Scholar] [CrossRef] [PubMed]
  169. Baechlein, C.; Becher, P. No evidence for zoonotic hepatitis E virus infection through dairy milk in Germany. Hepatology 2017, 65, 394–395. [Google Scholar] [CrossRef] [PubMed]
  170. Santos-Silva, S.; Moraes, D.; Lopez-Lopez, P.; Palmeira, J.D.; Torres, R.T.; Sao Jose Nascimento, M.; Dashti, A.; Carmena, D.; Rivero-Juarez, A.; Mesquita, J.R. Survey of Zoonotic Diarrheagenic Protist and Hepatitis E Virus in Wild Boar (Sus scrofa) of Portugal. Animals 2023, 13, 256. [Google Scholar] [CrossRef]
  171. Wu, J.Y.; Meng, X.X.; Wei, Y.R.; Bolati, H.; Lau, E.H.Y.; Yang, X.Y. Prevalence of Hepatitis E Virus (HEV) in Feral and Farmed Wild Boars in Xinjiang, Northwest China. Viruses 2022, 15, 78. [Google Scholar] [CrossRef] [PubMed]
  172. Lorusso, P.; Bonerba, E.; Pandiscia, A.; Mottola, A.; Di Pinto, A.; Piredda, R.; Terio, V. Occurrence of hepatitis E virus (HEV) in Calabrian wild boars. Int. J. Food Microbiol. 2022, 371, 109671. [Google Scholar] [CrossRef]
  173. Ferri, G.; Piccinini, A.; Olivastri, A.; Vergara, A. Hepatitis E virus detection in hunted wild boar (Sus scrofa) livers in Central Italy. Ital. J. Food Saf. 2022, 11, 9979. [Google Scholar] [CrossRef]
  174. Schotte, U.; Martin, A.; Brogden, S.; Schilling-Loeffler, K.; Schemmerer, M.; Anheyer-Behmenburg, H.E.; Szabo, K.; Muller-Graf, C.; Wenzel, J.J.; Kehrenberg, C.; et al. Phylogeny and spatiotemporal dynamics of hepatitis E virus infections in wild boar and deer from six areas of Germany during 2013–2017. Transbound. Emerg. Dis. 2022, 69, e1992–e2005. [Google Scholar] [CrossRef]
  175. Mendoza, M.V.; Yonemitsu, K.; Ishijima, K.; Kuroda, Y.; Tatemoto, K.; Inoue, Y.; Shimoda, H.; Kuwata, R.; Takano, A.; Suzuki, K.; et al. Nationwide survey of hepatitis E virus infection among wildlife in Japan. J. Vet. Med. Sci. 2022, 84, 992–1000. [Google Scholar] [CrossRef]
  176. Takahashi, M.; Nishizono, A.; Kawakami, M.; Fukui, E.; Isogai, E.; Matsuoka, H.; Yamamoto, S.; Mizuo, H.; Nagashima, S.; Murata, K.; et al. Identification of hepatitis E virus in wild sika deer in Japan. Virus Res. 2022, 308, 198645. [Google Scholar] [CrossRef]
  177. Priemer, G.; Cierniak, F.; Wolf, C.; Ulrich, R.G.; Groschup, M.H.; Eiden, M. Co-Circulation of Different Hepatitis E Virus Genotype 3 Subtypes in Pigs and Wild Boar in North-East Germany, 2019. Pathogens 2022, 11, 773. [Google Scholar] [CrossRef] [PubMed]
  178. Pierini, I.; Di Bartolo, I.; Manuali, E.; Pirani, S.; Bazzucchi, M.; Moscati, L.; De Mia, G.M.; Giammarioli, M. Hepatitis E virus (HEV) genotype 3 diversity: Identification of a novel HEV subtype in wild boar in Central Italy. Transbound. Emerg. Dis. 2021, 68, 2121–2129. [Google Scholar] [CrossRef] [PubMed]
  179. Arnaboldi, S.; Righi, F.; Carta, V.; Bonardi, S.; Pavoni, E.; Bianchi, A.; Losio, M.N.; Filipello, V. Hepatitis E Virus (HEV) Spread and Genetic Diversity in Game Animals in Northern Italy. Food Environ. Virol. 2021, 13, 146–153. [Google Scholar] [CrossRef]
  180. Kozyra, I.; Jablonski, A.; Bigoraj, E.; Rzezutka, A. Wild Boar as a Sylvatic Reservoir of Hepatitis E Virus in Poland: A Cross-Sectional Population Study. Viruses 2020, 12, 1113. [Google Scholar] [CrossRef]
  181. Takova, K.; Koynarski, T.; Minkov, I.; Ivanova, Z.; Toneva, V.; Zahmanova, G. Increasing Hepatitis E Virus Seroprevalence in Domestic Pigs and Wild Boar in Bulgaria. Animals 2020, 10, 1521. [Google Scholar] [CrossRef] [PubMed]
  182. Takahashi, M.; Nishizawa, T.; Sato, Y.; Miyazaki, S.; Aikawa, T.; Ashida, K.; Tamaru, T.; Oguro, K.; Hayakawa, F.; Matsuoka, H.; et al. Prevalence and genotype/subtype distribution of hepatitis E virus (HEV) among wild boars in Japan: Identification of a genotype 5 HEV strain. Virus Res. 2020, 287, 198106. [Google Scholar] [CrossRef]
  183. Choe, S.; Song, S.; Park, G.N.; Shin, J.; Kim, K.S.; Cha, R.M.; Hyun, B.H.; Park, B.K.; An, D.J. Detection of subtypes (3a, 4a and 4d) and high prevalence of hepatitis E virus in Korean wild boar. Vet. Microbiol. 2020, 240, 108531. [Google Scholar] [CrossRef]
  184. Jemersic, L.; Prpic, J.; Brnic, D.; Keros, T.; Pandak, N.; Dakovic Rode, O. Genetic diversity of hepatitis E virus (HEV) strains derived from humans, swine and wild boars in Croatia from 2010 to 2017. BMC Infect. Dis. 2019, 19, 269. [Google Scholar] [CrossRef] [Green Version]
  185. De Sabato, L.; Ostanello, F.; De Grossi, L.; Marcario, A.; Franzetti, B.; Monini, M.; Di Bartolo, I. Molecular survey of HEV infection in wild boar population in Italy. Transbound. Emerg. Dis. 2018, 65, 1749–1756. [Google Scholar] [CrossRef]
  186. Porea, D.; Anita, A.; Demange, A.; Raileanu, C.; Oslobanu Ludu, L.; Anita, D.; Savuta, G.; Pavio, N. Molecular detection of hepatitis E virus in wild boar population in eastern Romania. Transbound. Emerg. Dis. 2018, 65, 527–533. [Google Scholar] [CrossRef]
  187. Mesquita, J.R.; Oliveira, R.M.; Coelho, C.; Vieira-Pinto, M.; Nascimento, M.S. Hepatitis E Virus in Sylvatic and Captive Wild Boar from Portugal. Transbound. Emerg. Dis. 2016, 63, 574–578. [Google Scholar] [CrossRef] [PubMed]
  188. Kukielka, D.; Rodriguez-Prieto, V.; Vicente, J.; Sanchez-Vizcaino, J.M. Constant Hepatitis E Virus (HEV) Circulation in Wild Boar and Red Deer in Spain: An Increasing Concern Source of HEV Zoonotic Transmission. Transbound. Emerg. Dis. 2016, 63, e360–e368. [Google Scholar] [CrossRef]
  189. Caruso, C.; Modesto, P.; Bertolini, S.; Peletto, S.; Acutis, P.L.; Dondo, A.; Robetto, S.; Mignone, W.; Orusa, R.; Ru, G.; et al. Serological and virological survey of hepatitis E virus in wild boar populations in northwestern Italy: Detection of HEV subtypes 3e and 3f. Arch. Virol. 2015, 160, 153–160. [Google Scholar] [CrossRef]
  190. Widen, F.; Sundqvist, L.; Matyi-Toth, A.; Metreveli, G.; Belak, S.; Hallgren, G.; Norder, H. Molecular epidemiology of hepatitis E virus in humans, pigs and wild boars in Sweden. Epidemiol. Infect. 2011, 139, 361–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Lee, G.H.; Tan, B.H.; Teo, E.C.; Lim, S.G.; Dan, Y.Y.; Wee, A.; Aw, P.P.; Zhu, Y.; Hibberd, M.L.; Tan, C.K.; et al. Chronic Infection With Camelid Hepatitis E Virus in a Liver Transplant Recipient Who Regularly Consumes Camel Meat and Milk. Gastroenterology 2016, 150, 355–357.e3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  192. Santos-Silva, S.; Goncalves, H.M.R.; Rivero-Juarez, A.; Van der Poel, W.H.M.; Nascimento, M.S.J.; Mesquita, J.R. Detection of hepatitis E virus in milk: Current evidence for viral excretion in a wide range of mammalian hosts. Transbound. Emerg. Dis. 2022, 69, 3173–3180. [Google Scholar] [CrossRef]
  193. Baert, L.; Uyttendaele, M.; Debevere, J. Evaluation of viral extraction methods on a broad range of Ready-to-Eat foods with conventional and real-time RT-PCR for Norovirus GII detection. Int. J. Food Microbiol. 2008, 123, 101–108. [Google Scholar] [CrossRef]
  194. Stals, A.; Baert, L.; Van Coillie, E.; Uyttendaele, M. Extraction of food-borne viruses from food samples: A review. Int. J. Food Microbiol. 2012, 153, 1–9. [Google Scholar] [CrossRef] [PubMed]
  195. Boom, R.; Sol, C.J.; Salimans, M.M.; Jansen, C.L.; Wertheim-van Dillen, P.M.; van der Noordaa, J. Rapid and simple method for purification of nucleic acids. J. Clin. Microbiol. 1990, 28, 495–503. [Google Scholar] [CrossRef]
  196. Lowther, J.A.; Bosch, A.; Butot, S.; Ollivier, J.; Made, D.; Rutjes, S.A.; Hardouin, G.; Lombard, B.; In’t Veld, P.; Leclercq, A. Validation of EN ISO method 15216—Part 1—Quantification of hepatitis A virus and norovirus in food matrices. Int. J. Food Microbiol. 2019, 288, 82–90. [Google Scholar] [CrossRef]
  197. Schrader, C.; Schielke, A.; Ellerbroek, L.; Johne, R. PCR inhibitors—Occurrence, properties and removal. J. Appl. Microbiol. 2012, 113, 1014–1026. [Google Scholar] [CrossRef]
  198. Hennechart-Collette, C.; Niveau, F.; Martin-Latil, S.; Fraisse, A.; Perelle, S. Development of an extraction method to detect enteric viruses in dressed vegetables. Int. J. Food Microbiol. 2019, 311, 108349. [Google Scholar] [CrossRef] [PubMed]
  199. Hennechart-Collette, C.; Dehan, O.; Laurentie, M.; Fraisse, A.; Martin-Latil, S.; Perelle, S. Detection of norovirus, hepatitis A and hepatitis E viruses in multicomponent foodstuffs. Int. J. Food Microbiol. 2021, 337, 108931. [Google Scholar] [CrossRef] [PubMed]
  200. Hennechart-Collette, C.; Dehan, O.; Fraisse, A.; Martin-Latil, S.; Perelle, S. Development of an Extraction Method to Detect Hepatitis A Virus, Hepatitis E Virus, and Noroviruses in Fish Products. Microorganisms 2023, 11, 624. [Google Scholar] [CrossRef]
  201. Cook, N.; D’Agostino, M.; Wood, A.; Scobie, L. Real-Time PCR-Based Methods for Detection of Hepatitis E Virus in Pork Products: A Critical Review. Microorganisms 2022, 10, 428. [Google Scholar] [CrossRef] [PubMed]
  202. Althof, N.; Trojnar, E.; Bohm, T.; Burkhardt, S.; Carl, A.; Contzen, M.; Kilwinski, J.; Mergemeier, S.; Moor, D.; Made, D.; et al. Interlaboratory Validation of a Method for Hepatitis E Virus RNA Detection in Meat and Meat Products. Food Environ. Virol. 2019, 11, 1–8. [Google Scholar] [CrossRef] [PubMed]
  203. Mesquita, J.R.; Santos-Ferreira, N.; Ferreira, A.S.; Albuquerque, C.; Nobrega, C.; Esteves, F.; Cruz, R.; Vala, H.; Nascimento, M.S.J. Increased risk of hepatitis E virus infection in workers occupationally exposed to sheep. Transbound. Emerg. Dis. 2020. online ahead of print. [Google Scholar] [CrossRef]
  204. Sayed, I.M.; Hammam, A.R.A.; Elfaruk, M.S.; Alsaleem, K.A.; Gaber, M.A.; Ezzat, A.A.; Salama, E.H.; Elkhawaga, A.A.; El-Mokhtar, M.A. Enhancement of the Molecular and Serological Assessment of Hepatitis E Virus in Milk Samples. Microorganisms 2020, 8, 1231. [Google Scholar] [CrossRef]
  205. Wang, K.; Liu, L.; Wang, J.; Sun, X.; Han, Q.; Zhou, C.; Xu, X.; Wang, J. Quantification of hepatitis E virus in raw pork livers using droplet digital RT-PCR. Food Microbiol. 2023, 109, 104114. [Google Scholar] [CrossRef]
  206. Persson, S.; Alm, E.; Karlsson, M.; Enkirch, T.; Norder, H.; Eriksson, R.; Simonsson, M.; Ellstrom, P. A new assay for quantitative detection of hepatitis A virus. J. Virol. Methods 2021, 288, 114010. [Google Scholar] [CrossRef]
  207. Persson, S.; Eriksson, R.; Lowther, J.; Ellstrom, P.; Simonsson, M. Comparison between RT droplet digital PCR and RT real-time PCR for quantification of noroviruses in oysters. Int. J. Food Microbiol. 2018, 284, 73–83. [Google Scholar] [CrossRef]
  208. Morley, A.A. Digital PCR: A brief history. Biomol. Detect. Quantif. 2014, 1, 1–2. [Google Scholar] [CrossRef] [Green Version]
  209. Martin-Latil, S.; Hennechart-Collette, C.; Delannoy, S.; Guillier, L.; Fach, P.; Perelle, S. Quantification of Hepatitis E Virus in Naturally-Contaminated Pig Liver Products. Front. Microbiol. 2016, 7, 1183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  210. Han, Y.; Wang, J.; Zhang, S.; Yang, S.; Wang, X.; Han, Y.; Shen, Z.; Xu, X. Simultaneous quantification of hepatitis A virus and norovirus genogroup I and II by triplex droplet digital PCR. Food Microbiol. 2022, 103, 103933. [Google Scholar] [CrossRef] [PubMed]
  211. Hindson, C.M.; Chevillet, J.R.; Briggs, H.A.; Gallichotte, E.N.; Ruf, I.K.; Hindson, B.J.; Vessella, R.L.; Tewari, M. Absolute quantification by droplet digital PCR versus analog real-time PCR. Nat. Methods 2013, 10, 1003–1005. [Google Scholar] [CrossRef] [PubMed]
  212. Racki, N.; Dreo, T.; Gutierrez-Aguirre, I.; Blejec, A.; Ravnikar, M. Reverse transcriptase droplet digital PCR shows high resilience to PCR inhibitors from plant, soil and water samples. Plant Methods 2014, 10, 42. [Google Scholar] [CrossRef]
  213. Coudray-Meunier, C.; Fraisse, A.; Martin-Latil, S.; Guillier, L.; Delannoy, S.; Fach, P.; Perelle, S. A comparative study of digital RT-PCR and RT-qPCR for quantification of Hepatitis A virus and Norovirus in lettuce and water samples. Int. J. Food Microbiol. 2015, 201, 17–26. [Google Scholar] [CrossRef]
  214. Sedlak, R.H.; Nguyen, T.; Palileo, I.; Jerome, K.R.; Kuypers, J. Superiority of Digital Reverse Transcription-PCR (RT-PCR) over Real-Time RT-PCR for Quantitation of Highly Divergent Human Rhinoviruses. J. Clin. Microbiol. 2017, 55, 442–449. [Google Scholar] [CrossRef] [Green Version]
  215. Strain, M.C.; Lada, S.M.; Luong, T.; Rought, S.E.; Gianella, S.; Terry, V.H.; Spina, C.A.; Woelk, C.H.; Richman, D.D. Highly precise measurement of HIV DNA by droplet digital PCR. PLoS ONE 2013, 8, e55943. [Google Scholar] [CrossRef]
  216. Persson, S.; Karlsson, M.; Borsch-Reniers, H.; Ellstrom, P.; Eriksson, R.; Simonsson, M. Missing the Match Might Not Cost You the Game: Primer-Template Mismatches Studied in Different Hepatitis A Virus Variants. Food Environ. Virol. 2019, 11, 297–308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  217. Raymond, P.; Paul, S.; Perron, A.; Bellehumeur, C.; Larocque, E.; Charest, H. Detection and Sequencing of Multiple Human Norovirus Genotypes from Imported Frozen Raspberries Linked to Outbreaks in the Province of Quebec, Canada, in 2017. Food Environ. Virol. 2022, 14, 40–58. [Google Scholar] [CrossRef] [PubMed]
  218. Yang, Z.; Mammel, M.; Whitehouse, C.A.; Ngo, D.; Kulka, M. Inter- and Intra-Host Nucleotide Variations in Hepatitis A Virus in Culture and Clinical Samples Detected by Next-Generation Sequencing. Viruses 2018, 10, 619. [Google Scholar] [CrossRef] [Green Version]
  219. Lee, G.Y.; Kim, W.K.; Cho, S.; Park, K.; Kim, J.; Lee, S.H.; Lee, J.; Lee, Y.S.; Kim, J.H.; Byun, K.S.; et al. Genotyping and Molecular Diagnosis of Hepatitis A Virus in Human Clinical Samples Using Multiplex PCR-Based Next-Generation Sequencing. Microorganisms 2022, 10, 100. [Google Scholar] [CrossRef] [PubMed]
  220. Ollivier, J.L.J.; Desdouits, M.; Schaeffer, J.; Wacrenier, C.; Oude Munnink, B.B.; Besnard, A.M.B.F.; Stapleton, T.; Schultz, A.C.; Aarestrup, F.; Koopmans, M.; et al. Application of Next Generation Sequencing on Norovirus-Contaminated Oyster Samples; EFSA Supporting Publication: Parma, Italy, 2022; p. 144. [Google Scholar]
  221. Bartsch, C.; Hoper, D.; Made, D.; Johne, R. Analysis of frozen strawberries involved in a large norovirus gastroenteritis outbreak using next generation sequencing and digital PCR. Food Microbiol. 2018, 76, 390–395. [Google Scholar] [CrossRef]
  222. Chiapponi, C.; Pavoni, E.; Bertasi, B.; Baioni, L.; Scaltriti, E.; Chiesa, E.; Cianti, L.; Losio, M.N.; Pongolini, S. Isolation and genomic sequence of hepatitis A virus from mixed frozen berries in Italy. Food Environ. Virol. 2014, 6, 202–206. [Google Scholar] [CrossRef] [Green Version]
  223. Yang, Z.; Mammel, M.; Papafragkou, E.; Hida, K.; Elkins, C.A.; Kulka, M. Application of next generation sequencing toward sensitive detection of enteric viruses isolated from celery samples as an example of produce. Int. J. Food Microbiol. 2017, 261, 73–81. [Google Scholar] [CrossRef] [PubMed]
  224. Vaughan, G.; Xia, G.; Forbi, J.C.; Purdy, M.A.; Rossi, L.M.; Spradling, P.R.; Khudyakov, Y.E. Genetic relatedness among hepatitis A virus strains associated with food-borne outbreaks. PLoS ONE 2013, 8, e74546. [Google Scholar] [CrossRef] [Green Version]
  225. Zhai, L.; Dai, X.; Meng, J. Hepatitis E virus genotyping based on full-length genome and partial genomic regions. Virus Res. 2006, 120, 57–69. [Google Scholar] [CrossRef]
  226. Vonlanthen-Specker, I.; Stephan, R.; Sidler, X.; Moor, D.; Fraefel, C.; Bachofen, C. Genetic Diversity of Hepatitis E Virus Type 3 in Switzerland-From Stable to Table. Animals 2021, 11, 3177. [Google Scholar] [CrossRef]
  227. Mikhailov, M.I.; Karlsen, A.A.; Potemkin, I.A.; Isaeva, O.V.; Kichatova, V.S.; Malinnikova, E.Y.; Asadi Mobarkhan, F.A.; Mullin, E.V.; Lopatukhina, M.A.; Manuylov, V.A.; et al. Geographic and Temporal Variability of Hepatitis E Virus Circulation in the Russian Federation. Viruses 2022, 15, 37. [Google Scholar] [CrossRef] [PubMed]
  228. Papp, C.P.; Biedermann, P.; Harms, D.; Wang, B.; Kebelmann, M.; Choi, M.; Helmuth, J.; Corman, V.M.; Thurmer, A.; Altmann, B.; et al. Advanced sequencing approaches detected insertions of viral and human origin in the viral genome of chronic hepatitis E virus patients. Sci. Rep. 2022, 12, 1720. [Google Scholar] [CrossRef] [PubMed]
  229. Asif, K.; O’Rourke, D.; Sabir, A.J.; Shil, P.; Noormohammadi, A.H.; Marenda, M.S. Characterisation of the whole genome sequence of an avian hepatitis E virus directly from clinical specimens reveals possible recombination events between European and USA strains. Infect. Genet. Evol. 2021, 96, 105095. [Google Scholar] [CrossRef]
  230. Liu, Y.; Kearney, J.; Mahmoud, M.; Kille, B.; Sedlazeck, F.J.; Treangen, T.J. Rescuing low frequency variants within intra-host viral populations directly from Oxford Nanopore sequencing data. Nat. Commun. 2022, 13, 1321. [Google Scholar] [CrossRef] [PubMed]
  231. Schuele, L.; Cassidy, H.; Lizarazo, E.; Strutzberg-Minder, K.; Schuetze, S.; Loebert, S.; Lambrecht, C.; Harlizius, J.; Friedrich, A.W.; Peter, S.; et al. Assessment of Viral Targeted Sequence Capture Using Nanopore Sequencing Directly from Clinical Samples. Viruses 2020, 12, 1358. [Google Scholar] [CrossRef]
  232. Flint, A.; Reaume, S.; Harlow, J.; Hoover, E.; Weedmark, K.; Nasheri, N. Genomic analysis of human noroviruses using combined Illumina-Nanopore data. Virus Evol. 2021, 7, veab079. [Google Scholar] [CrossRef] [PubMed]
  233. Linde, J.; Brangsch, H.; Holzer, M.; Thomas, C.; Elschner, M.C.; Melzer, F.; Tomaso, H. Comparison of Illumina and Oxford Nanopore Technology for genome analysis of Francisella tularensis, Bacillus anthracis, and Brucella suis. BMC Genom. 2023, 24, 258. [Google Scholar] [CrossRef]
  234. McNaughton, A.L.; Roberts, H.E.; Bonsall, D.; de Cesare, M.; Mokaya, J.; Lumley, S.F.; Golubchik, T.; Piazza, P.; Martin, J.B.; de Lara, C.; et al. Illumina and Nanopore methods for whole genome sequencing of hepatitis B virus (HBV). Sci. Rep. 2019, 9, 7081. [Google Scholar] [CrossRef] [Green Version]
  235. Notomi, T.; Okayama, H.; Masubuchi, H.; Yonekawa, T.; Watanabe, K.; Amino, N.; Hase, T. Loop-mediated isothermal amplification of DNA. Nucleic Acids Res. 2000, 28, E63. [Google Scholar] [CrossRef] [Green Version]
  236. Wu, R.; Meng, B.; Corredig, M.; Griffiths, M.W. Rapid Detection of Hepatitis A Virus in Foods Using a Bioluminescent Assay in Real-Time (BART) and Reverse Transcription Loop-Mediated Isothermal Amplification (RT-LAMP) Technology. Food Environ. Virol. 2023, 15, 144–157. [Google Scholar] [CrossRef]
  237. Zhou, Y.; Wan, Z.; Yang, S.; Li, Y.; Li, M.; Wang, B.; Hu, Y.; Xia, X.; Jin, X.; Yu, N.; et al. A Mismatch-Tolerant Reverse Transcription Loop-Mediated Isothermal Amplification Method and Its Application on Simultaneous Detection of All Four Serotype of Dengue Viruses. Front. Microbiol. 2019, 10, 1056. [Google Scholar] [CrossRef] [Green Version]
  238. Inaba, M.; Higashimoto, Y.; Toyama, Y.; Horiguchi, T.; Hibino, M.; Iwata, M.; Imaizumi, K.; Doi, Y. Diagnostic accuracy of LAMP versus PCR over the course of SARS-CoV-2 infection. Int. J. Infect. Dis. 2021, 107, 195–200. [Google Scholar] [CrossRef]
  239. Ma, L.; Yin, L.; Li, X.; Chen, S.; Peng, L.; Liu, G.; Ye, S.; Zhang, W.; Man, S. A smartphone-based visual biosensor for CRISPR-Cas powered SARS-CoV-2 diagnostics. Biosens. Bioelectron. 2022, 195, 113646. [Google Scholar] [CrossRef] [PubMed]
  240. Gootenberg, J.S.; Abudayyeh, O.O.; Kellner, M.J.; Joung, J.; Collins, J.J.; Zhang, F. Multiplexed and portable nucleic acid detection platform with Cas13, Cas12a, and Csm6. Science 2018, 360, 439–444. [Google Scholar] [CrossRef] [Green Version]
  241. Chen, J.S.; Ma, E.; Harrington, L.B.; Da Costa, M.; Tian, X.; Palefsky, J.M.; Doudna, J.A. CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity. Science 2018, 360, 436–439. [Google Scholar] [CrossRef] [Green Version]
  242. Livsmendelsverket Swedish Food Agency. Proficiency Testing Scheme EFV05, 2020 Quantification of Norovirus and Hepatitis A Virus in Bivalve Molluscan Shellfish Final Report—Version 1 (2021/06/25); European Union Reference Laboratory for Foodborne Viruses: Uppsala, Sweden, 2020. [Google Scholar]
  243. Li, P.; Liu, J.; Li, Y.; Su, J.; Ma, Z.; Bramer, W.M.; Cao, W.; de Man, R.A.; Peppelenbosch, M.P.; Pan, Q. The global epidemiology of hepatitis E virus infection: A systematic review and meta-analysis. Liver Int. 2020, 40, 1516–1528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  244. Topping, J.R.; Schnerr, H.; Haines, J.; Scott, M.; Carter, M.J.; Willcocks, M.M.; Bellamy, K.; Brown, D.W.; Gray, J.J.; Gallimore, C.I.; et al. Temperature inactivation of Feline calicivirus vaccine strain FCV F-9 in comparison with human noroviruses using an RNA exposure assay and reverse transcribed quantitative real-time polymerase chain reaction-A novel method for predicting virus infectivity. J. Virol. Methods 2009, 156, 89–95. [Google Scholar] [CrossRef]
  245. Sanchez, G.; Elizaquivel, P.; Aznar, R. Discrimination of infectious hepatitis A viruses by propidium monoazide real-time RT-PCR. Food Environ. Virol. 2012, 4, 21–25. [Google Scholar] [CrossRef] [PubMed]
Table 2. HEV RNA detection in meat products.
Table 2. HEV RNA detection in meat products.
Year of SamplingType of SamplesRegion/CountryNo. of SamplesPercentage of HEV RNA DetectionReference
2013–2014pork meat cut, ground pork meat, pig liver, pig intestines, raw pork balls, pork sausages, fermented pork productsNakhon Pathom Province, Thailand2140.5% of pork products, 2.0% of pig liver [125]
2017–2018marketed meat cuts (beef, chicken, pork)Uruguaiana, Rio Grande do Sul, Brazil1310%[126]
2022pork liver pâtés, raw dried hams and raw dried sausagesBelgium5465% of the pork liver pâtés, 15% of raw dried hams and 0% of raw dried sausages[127]
2018ground pork, liverNorthern California, United States20912.6% of ground pork, 45% of pork liver [128]
2017–2019 sausagesthe Netherlands316 [129]
2019–2020liver, liver sausage, liver pateGermany13110%, pork livers (5%), liver sausages (13%) and liver pâté samples (15%)[130]
2017pork cuts and organsSpain450 [131]
2016liver, liver sausage, liver pate, meat cuts, sausages, wild boar meat 52112.7% of livers, 70.7% of liverwurst, 68.9% of liver pate, 0% of the pork chops, 0% of fresh sausages, 0% of wild boar meat [132]
2017–2018raw bacon, meat cuts, pork and wild boar sausages, salami, wild boar salamiSouthern Italy1626.3% of wild boar salamis, 0% all other samples[133]
2017–2018meat cuts, liver, kidney, blood curdHebei Province, China413pig liver 6.1%, kidney 3.1%, and blood curd 1.2%[134]
not detailedpork meat cuts, livers, intestines, spleens, uretersChina10733.3% of meat cuts, 8.3% of pig liver, 18.7% of intestine, 33.3% of spleen, 26.3% of ureter [135]
2016liver sausage, raw meat sausageSwitzerland9018.9% of liver sausages, 5.7% of raw meat sausages[136]
2014–2015liver sausage, salami, wild boar sausagesGermany12020% of raw sausages, 22% of liver sausages[137]
2011sausage, liver sausage, dry salted liver, liver quenellesFrance39430% of sausage, 29% of liver sausages, 25% of liver quenelles, 3% of dried salted liver[138]
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

Nemes, K.; Persson, S.; Simonsson, M. Hepatitis A Virus and Hepatitis E Virus as Food- and Waterborne Pathogens—Transmission Routes and Methods for Detection in Food. Viruses 2023, 15, 1725. https://doi.org/10.3390/v15081725

AMA Style

Nemes K, Persson S, Simonsson M. Hepatitis A Virus and Hepatitis E Virus as Food- and Waterborne Pathogens—Transmission Routes and Methods for Detection in Food. Viruses. 2023; 15(8):1725. https://doi.org/10.3390/v15081725

Chicago/Turabian Style

Nemes, Katalin, Sofia Persson, and Magnus Simonsson. 2023. "Hepatitis A Virus and Hepatitis E Virus as Food- and Waterborne Pathogens—Transmission Routes and Methods for Detection in Food" Viruses 15, no. 8: 1725. https://doi.org/10.3390/v15081725

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

Article Metrics

Back to TopTop