Common search

Tuesday, November 20, 2012

Book on hepatitis from page 106 to 117

Book on hepatitis from page 106 to 117

106  Hepatology 2012
Tanji Y, Hijikata M, Satoh S, et al. Hepatitis C virus-encoded nonstructural protein NS4A has
versatile functions in viral protein processing. J Virol 1995;69:1575-81. (Abstract)
Thiel HJ, Collett MS, Gould EA et al. Family Flaviviridae, In: Fauquet CM, Mayo MA, Maniloff J
et al., eds. Virus Taxonomy:VIIIth Report of the International Committee on
Taxonomy of Viruses. San Diego: Academic Press; 2005:979-96.
Thomssen R, Bonk S, Propfe C, et al. Association of hepatitis C virus in human sera with
lipoprotein. Med Microbiol Immunol 1992;181:293-300. (Abstract)
Thomssen R, Bonk S, Thiele A. Density heterogeneities of hepatitis C virus in human sera due
to the binding of beta-lipoproteins and immunoglobulins. Med Microbiol Immunol
1993;182:329-34. (Abstract)
Timpe JM, Stamataki Z, Jennings A, et al. Hepatitis C virus cell-cell transmission in hepatoma
cells in the presence of neutralizing antibodies. Hepatology 2007;47:17-24. (Abstract)
Tomei L, Failla C, Santolini E, et al. NS3 is a Serine protease required for processing of
hepatitis C virus polyprotein. J Virol 1993;67:4017-26. (Abstract)
Tomei L, Failla C, Vitale RL, et al. A central hydrophobic domain of the hepatitis C virus NS4A
protein is necessary and sufficient for the activation of the NS3 protease. J Gen Virol
1996;77:1065-70. (Abstract)
Tong MJ, el-Farra NS, Reikes AR et al. Clinical outcomes after transfusion-associated hepatitis
C. N Engl J Med 1995;332:1463-6. (Abstract)
Tsukiyama-Kohara K, Iizuka N, Kohara M, et al. Internal ribosome entry site within hepatitis C
virus RNA. J Virol 1992;66:1476-83. (Abstract)
Valli MB, Serafino A, Crema A, et al. Transmission in vitro of hepatitis C virus from persistently
infected human B-cells to hepatoma cells by cell-to-cell contact. J Med Virol
2006;78:192-201. (Abstract)
Valli MB, Crema A, Lanzilli G, et al. Molecular and cellular determinants of cell-to-cell
transmission of HCV in vitro. J Med Virol 2007;79:1491-9. (Abstract)
Varaklioti A, Vassilaki N, Georgopoulou U, et al. Alternate translation occurs within the core
coding region of the hepatitis C viral genome. J Biol Chem 2002;17713-21. (Abstract)
Wakita T, Pietschmann T, Kato T, et al. Production of infectious hepatitis C virus in tissue
culture from a cloned viral genome. Nat Med 2005;11:791-6. (Abstract)
Walewski JL, Keller TR, Stump DD, et al. Evidence for a new hepatitis C virus antigen encoded
in an overlapping reading frame. RNA 2001;7:710-21. (Abstract)
Wang C, Sarnow P, Siddiqui A. Translation of human hepatitis C virus RNA in cultured cells is
mediated by an internal ribosome-binding mechanism. J Virol 1993;67:3338-44.
(Abstract)
Wang H, Shen XT, Ye R, et al. Roles of the polypyrimidine tract and 3' noncoding region of
hepatitis C virus RNA in the internal ribosome entry site-mediated translation. Arch
Virol 2005;150:1085-99. (Abstract)
Washburn ML, Bility MT, Zhang L, et al. A humanized mouse model to study hepatitis C virus
infection, immune response, and liver disease. Gastroenterology 2011;140:1334-44.
(Abstract)
Watashi K, Ishii N, Hijikata M, et al. Cyclophilin B is a functional regulator of hepatitis C virus
RNA polymerase. Mol Cell 2005;19:111-22.
Webb NR, Connell PM, Graf GA, et al. SR-BII, an isoform of the scavenger receptor BI
containing an alternate cytoplasmic tail, mediates lipid transfer between high-density
lipoprotein and cells. J Biol Chem 1998;273:15241-8. (Abstract)
Weihofen A, Binns K, Lemberg MK, et al. Identification of signal peptide peptidase, a
presenelin-type aspartic protease. Science 2002;296:2215-8. (Abstract)
Weiner AJ, Brauer MJ, Rosenblatt J, et al. Variable and hypervariable domains are found in the
regions of HCV corresponding to the flavivirus envelope and NS1 proteins and the
pestivirus envelope glycoproteins. Virology 1991;180:842-8. (Abstract)
Witteveldt J, Evans MJ, Bitzegeio J, et al. CD81 is dispensable for hepatitis C virus cell-to-cell
transmission in hepatoma cells. J Gen Virol 2009;90:48-58. (Abstract)
Wölk B, Sansonno D, Kräusslich HG, et al. Subcellular localization, stability, and transcleavage
competence of the hepatitis C virus NS3-NS4A complex expressed in tetracycline-regulated cell lines. J Virol 2000;74:2293-304. (Abstract)
Wünschmann S, Medh JD, Klinzmann D, et al. Characterization of hepatitis C virus (HCV) and
HCV E2 interactions with CD81 and the low-density lipoprotein receptor. J Virol
2000;74:10055-62. (Abstract)
Xu Z, Choi J, Yen TS, et al. Synthesis of a novel hepatitis C virus protein by ribosomal
frameshift. EMBO J 2001;20:3840-8. (Abstract)
HCV Virology  107
Xu Z, Choi J, Lu W, et al. Hepatitis C virus F protein is a short-lived protein associated with the
endoplasmic reticulum. J Virol 2003;77:1578-83. (Abstract)
Yanagi M, St Claire M, Emerson SU. In vivo analysis of the 3' untranslated region of the
hepatitis C virus after in vitro mutagenesis of an infectious cDNA clone. Proc Natl
Acad Sci USA 1999;96:2291-5. (Abstract)
Yang F, Robotham JM, Nelson HB, et al. Cyclophilin A is an essential cofactor for hepatitis C
virus infection and the principal mediator of cyclosporine resistance in vitro. J Virol
2008;82:5269-78. (Abstract)
Yi M and Lemon SM. 3' Nontranslated RNA signals required for replication of hepatitis C virus
RNA. J Virol 2003;77:3557-68. (Abstract)
Yi M, Villanueva RA, Thomas DL, et al. Production of infectious genotype 1a hepatitis C virus
(Hutchinson strain) in cultured hepatoma cells. Proc Natl Acad Sci USA
2006;103:2310-5. (Abstract)
You S and Rice CM. 3´ RNA elements in hepatitis C virus replication: kissing partners and long
poly(U). J Virol 2008;82:184-95. (Abstract)
Yu X, Qiao M, Atanasov I, et al. Cryo-electron microscopy and three-dimensional
reconstructions of hepatitis C virus particles. Virology 2007;367:126-34. (Abstract)
Zeisel MB, Koutsoudakis G, Schnober EK, et al. Scavenger receptor class B type I is a key host
factor for hepatitis C virus infection required for an entry step closely linked to CD81.
Hepatology 2007;46:1722-31. (Abstract)
Zhang J, Yamada O, Yoshida H, et al. Autogenous translational inhibition of core protein:
implication for switch from translation to RNA replication in hepatitis C virus. Virology
2002;293:141-50. (Abstract)
Zhang LQ, Randall G, Higginbottom A, et al. CD81 is required for hepatitis C virus glycoprotein-mediated viral infection. J Virol 2004;78:1448-55. (Abstract)
Zheng A, Yuan F, Li Y, et al. Claudin-6 and claudin-9 function as additional coreceptors for
hepatitis C virus. J Virol 2007;81:12465-71. (Abstract)
Zhong J, Gastaminza P, Cheng G, et al. Robust hepatitis C virus infection in vitro. Proc Natl
Acad Sci USA 2005;102:9294-9. (Abstract)
108  Hepatology 2012
7.  Prophylaxis and Vaccination
Heiner Wedemeyer
Introduction
Understanding the biology and modes of transmission of hepatitis viruses has
significantly improved over the last decades. Still, prophylactic vaccines are only
available against HAV and HBV. Although an enormous amount of basic and
clinical research has been performed to develop a vaccine against hepatitis C, it is
very unlikely that a prophylactic or therapeutic HCV vaccine will be licensed in the
next few years. A first Phase III vaccine trial against hepatitis E has been successful
in China; nevertheless, it is currently unknown if or when this vaccine will become
available in other countries. Prophylaxis of HCV, HDV (for HBV-infected patients)
and HEV infection therefore must still occur by avoiding all routes of exposure to
the respective hepatitis viruses discussed in detail in Chapters 1-4.
Prophylaxis of hepatitis viruses
Hepatitis A and E
The hepatitis A and E viruses are usually transmitted by oral ingestion of
contaminated food or water. Thus, particular caution is warranted when individuals
from low endemic areas such as western Europe and the US travel to countries with
a high prevalence of HAV and HEV infections. In addition, hepatitis E can also be a
zoonosis. A German case-control study identified 32% of all reported HEV
infections as being autochthonous infections, meaning not associated with travelling
to endemic countries (Wichmann 2008). In these patients consumption of offal and
wild boar meat was independently associated with HEV infection. This may have
significant implications for immunosuppressed patients as cases of chronic hepatitis
E with the development of advanced fibrosis have been described in patients after
organ transplantation (Kamar 2008, Pischke 2010). HEV has frequently been
detected in the meat of pigs; Danish farmers show a higher prevalence of HEV
antibodies. Importantly, zoonotic HEV infection is usually caused by HEV
genotype 3 while HEV genotype 1 can be found in travelling-associated hepatitis E.
Prophylaxis and Vaccination  109
HAV and HEV can also be transmitted by blood transfusion although cases are
extremely rare.
Hepatitis B and D
HBV and HDV were transmitted frequently by blood transfusion before HBsAg
testing of all blood products was introduced in the 1970s. Since then, vertical
transmission and sexual exposure have become the most frequent routes of HBV
infection. Medical procedures still represent a potential source for HBV
transmissions and thus strict and careful application of standard hygienic
precautions for all medical interventions are absolutely mandatory, and not only in
endemic areas. This holds true in particular for immunocompromised individuals
who are highly susceptible to HBV infection as HBV is characterized by a very high
infectivity (Wedemeyer 1998). Moreover, immunosuppressed patients are at risk for
reactivation of occult HBV infection after serological recovery from hepatitis B.
Treatments with high doses of steroids and rituximab have especially been
identified as major risk factors for HBV reactivation (Lalazar 2007, Loomba 2008).
After a new diagnosis of HBV infection, all family members of the patient need to
be tested for their immune status against HBV. Immediate active vaccination is
recommended for all anti-HBc-negative contact persons. HBsAg-positive
individuals should use condoms during sexual intercourse if it is not known if the
partner has been vaccinated. Non-immune individuals who have experienced an
injury and were exposed to HBsAg-positive fluids should undergo passive
immunization with anti-HBs as soon as possible, preferentially within 2-12 hours
(Cornberg 2011).
Hepatitis C
Less than 1% of individuals who are exposed to HCV by an injury with
contaminated needles develop acute HCV infection. At Hannover Medical School,
not a single HCV seroconversion occurred after 166 occupational exposures with
anti-HCV positive blood in a period of 6 years (2000-2005). A systematic review of
the literature identified 22 studies including a total of 6,956 injuries with HCV
contaminated needles. Only 52 individuals (0.75%) became infected. The risk of
acute HCV infection was lower in Europe at 0.42% compared to eastern Asia at
1.5% (Kubitschke 2007). Thus, the risk of acquiring HCV infection after a needle-stick injury is lower than frequently reported. Worldwide differences in HCV se-roconversion rates may suggest that genetic factors provide some level of natural
resistance against HCV. Factors associated with a higher risk of HCV transmission
are likely to be the level of HCV viremia in the index patient, the amount of
transmitted fluid and the duration between contamination of the respective needle
and injury. Suggested follow-up procedures after needlestick injury are shown in
Figure 1.
Sexual intercourse with HCV-infected persons has clearly been identified as a risk
for HCV infection, as about 10-20% of patients with acute hepatitis C report this as
a potential risk factor (Deterding 2009; Table 1). However, there is also evidence
that the risk of acquiring HCV sexually is extremely low in individuals in stable
partnerships who avoid injuries. Cohort studies including more than 500 HCV-infected patients followed over periods of more than 4 years could not identify any
cases of confirmed HCV transmission. Thus, guidelines generally do not
110  Hepatology 2012
recommend the use of condoms in monogamous relationships (EASL 2011).
However, this does not hold true for HIV-positive homosexual men. Several
outbreaks of acute hepatitis C have been described in this scenario (Fox 2008, Low
2008, van de Laar 2009). Transmitted cases had more sexual partners, increased
levels of high-risk sexual behaviour (in particular, fisting) and were more likely to
have shared drugs via a nasal or anal route than controls (Turner 2006).
Due to the low HCV prevalence in most European countries and due to a
relatively low vertical transmission rate of 1-6%, general screening of pregnant
women for anti-HCV is not recommended. Interestingly, transmission may be
higher for girls than for boys (European Pediatric Hepatitis C Virus Network 2005).
Transmission rates may be higher in HIV-infected women so pregnant women
should be tested for hepatitis C. Other factors possibly associated with high
transmission rates are the level of HCV viremia, maternal intravenous drug use, and
specific HLA types of the children. Cesarean sections are not recommended for
HCV RNA positive mothers as there is no clear evidence that these reduce
transmission rates. Children of HCV-infected mothers should be tested for HCV
RNA after 1 month as maternal anti-HCV antibodies can be detected for several
months after birth. Mothers with chronic hepatitis C can breast-feed their children as
long as they are HIV-negative and do not use intravenous drugs (European Pediatric
Hepatitis C Virus Network 2001, EASL 2011).
The Spanish Acute HCV Study Group has identified hospital admission as a
significant risk factor for acquiring HCV infection in Spain (Martinez-Bauer 2008).
The data are in line with reports from Italy (Santantonio 2006) and the USA (Corey
2006). We have reported data from the German Hep-Net Acute HCV Studies and
found 38 cases (15% of the entire cohort) of acute HCV patients who reported a
medical procedure as the most likely risk factor for having acquired HCV
(Deterding 2008). The majority of those were hospital admissions with surgery in
30 cases; other invasive procedures, including dental treatment, were present in only
4 cases. Medical procedures were significantly more often the probable cause of
infection in patients older than 30 years of age (p=0.002) but not associated with
disease severity or time from exposure to onset of symptoms. Thus, medical
treatment per se still represents a significant risk factor for HCV infection – even in
developed countries. Strict adherence to universal precaution guidelines is urgently
warranted.
Vaccination against hepatitis A
The first active vaccine against HAV was licensed in 1995. The currently available
inactive vaccines are manufactured from cell culture-adapted HAV, grown either in
human fibroblasts or diploid cells (Nothdurft 2008). Two doses of the vaccine are
recommended. The second dose should be given between 6 and 18 months after the
first dose. All vaccines are highly immunogenic and basically all vaccinated healthy
persons develop protective anti-HAV antibodies. Similar vaccine responses are
obtained in both children and adults and no relevant regional differences in response
to HAV vaccination have been observed. The weakest vaccine responses have been
described for young children receiving a 0, 1, 2 months schedule (Hammitt 2008).
Patients with chronic liver disease do respond to vaccination but may display lower
anti-HAV titers (Keeffe 1998). A combined vaccine against HAV and HBV is
Prophylaxis and Vaccination  111
available that needs to be administered three times, on a 0, 1, and 6 months
schedule. More than 80% of healthy individuals have detectable HAV antibodies by
day 21 applying an accelerated vaccine schedule of 0, 7 and 21 days using the
combined HAV/HBV vaccine, and all study subjects were immune against HAV by
2 months (Kallinowski 2003).
HAV vaccines are very well tolerated and no serious adverse events have been
linked with the administration of HAV vaccines (Nothdurft 2008). The vaccine can
safely be given together with other vaccines or immunoglobulins without
compromising the development of protective antibodies.
Vaccination is recommended for non-immune individuals who plan to travel to
endemic countries, medical health professionals, homosexual men, persons in
contact with hepatitis A patients, and individuals with chronic liver diseases. Some
studies have suggested that patients with chronic hepatitis C have a higher risk of
developing fulminant hepatitis A (Vento 1998) although this finding has not been
confirmed by other investigators (Deterding 2006). The implementation of
childhood vaccination programs has led to significant and impressive declines of
HAV infections in several countries, justifying further efforts aiming at controlling
the spread of HAV in endemic countries (Hendrickx 2008). It is important to
highlight that most studies have confirmed that HAV vaccination is cost-effective
(Rein 2008, Hollinger 2007).
Long-term follow-up studies after complete HAV vaccination have been
published. Interestingly, anti-HAV titers sharply decline during the first year after
vaccination but remain detectable in almost all individuals for at least 10-15 years
after vaccination (Van Herck 2011). Based on these studies it was estimated that
protective anti-HAV antibodies should persist for at least 27-30 years after
successful vaccination (Hammitt 2008, Bovier 2010).
Vaccination against hepatitis B
The hepatitis B vaccine is the first vaccine able to reduce the incidence of cancer. In
Taiwan, a significant decline in cases of childhood hepatocellular carcinoma has
been observed since the implementation of programs to vaccinate all infants against
HBV (Chang 1997). This landmark study impressively highlighted the usefulness of
universal vaccination against HBV in endemic countries. Controversial discussions
are ongoing regarding to what extent universal vaccination against HBV may be
cost-effective in low-endemic places such as the UK, the Netherlands or
Scandinavia (Zuckerman 2007). In 1992 the World Health Organization
recommended general vaccination against hepatitis B. It  should be possible to
eradicate hepatitis B by worldwide implementation of this recommendation,
because  humans are the only epidemiologically relevant host for HBV. 179
countries have introduced a hepatitis B vaccine in their national infant immunization
schedules by the end of 2010, including parts of India and the Sudan (WHO 2011).
The first plasma-derived hepatitis B vaccine was approved by FDA in 1981.
Recombinant vaccines consisting of HBsAg produced in yeast became available in
1986. In the US, two recombinant vaccines are licensed (Recombivax® and
Engerix-B®) while additional vaccines are used in other countries. The vaccines are
administered three times, on a 0, 1, and 6 months schedule.
112  Hepatology 2012
Who should be vaccinated? (The German Guidelines (Cornberg 2011))
−  Hepatitis B high-risk persons working in health care settings including trainees,
students, cleaning personnel;
−  Personnel in psychiatric facilities or comparable welfare institutions for
cerebrally damaged or disturbed patients; other persons who are at risk because
of blood contact with possibly infected persons dependent on the risk
evaluation, e.g., persons giving first aid professionally or voluntarily,
employees of ambulance services, police officers, social workers, and prison
staff who have contact with drug addicts;
−  Patients with chronic kidney disease, dialysis patients, patients with frequent
blood or blood component transfusions (e.g., hemophiliacs), patients prior to
extensive surgery (e.g., before operations using heart-lung machine. The
urgency of the operation and the patient’s wish for vaccination protection are
of primary importance);
−  Persons with chronic liver disease including chronic diseases with liver
involvement as well as HIV-positive persons without HBV markers;
−  Persons at risk of contact with HBsAg carriers in the family or shared housing,
sexual partners of HBsAg carriers;
−  Patients in psychiatric facilities or residents of comparable welfare institutions
for cerebrally damaged or disturbed persons as well as persons in sheltered
workshops;
−  Special high-risk groups, e.g., homosexually active men, regular drug users,
sex workers, prisoners serving extended sentences;
−  Persons at risk of contacting HBsAg carriers in facilities (kindergarten,
children’s homes, nursing homes, school classes, day care groups);
−  Persons travelling to regions with high hepatitis B prevalence for an extended
period of time or with expected close contact with the local population;
−  Persons who have been injured by possibly contaminated items, e.g., needle
puncture (see post-exposition prophylaxis);
−  Infants of HBsAg-positive mothers or of mothers with unknown HBsAg status
(independent of weight at birth) (see post-exposition prophylaxis).
Routine testing for previous contact with hepatitis B is not necessary before
vaccination unless the person belongs to a risk group and may have acquired
hepatitis B before. Pre-vaccine testing is usually not cost-effective in populations
with anti-HBc prevalence below 20%. Vaccination of an HBsAg-positive individual
can be performed without any danger – however, it is ineffective.
Efficacy of vaccination against hepatitis B
A response to HBV vaccination is determined by the development of anti-HBs
antibodies, detectable in 90-95% of individuals one month after a complete
vaccination schedule (Wedemeyer 2007, Coates 2001). Responses are lower in
elderly people and much weaker in immunocompromised persons such as organ
transplant recipients, patients receiving hemodialysis and HIV-infected individuals.
In case of vaccine non-response, another three courses of vaccine should be
administered and the dose of the vaccine should be increased. Other possibilities to
Prophylaxis and Vaccination  113
increase the immunogenicity of HBV vaccines include intradermal application and
coadministration of adjuvants and cytokines (Cornberg 2011). The response to
vaccination should be controlled in high-risk individuals such as medical health
professionals and immunocompromised persons. Some guidelines also recommend
testing elderly persons after vaccinations as vaccine response does decline more
rapidly in the elderly (Wolters 2003).
Post-exposure prophylaxis
Non-immune persons who have been in contact with HBV-contaminated materials
(e.g., needles) or who have had sexual intercourse with an HBV-infected person
should undergo active-passive immunization (active immunization plus hepatitis B
immunoglobulin) as soon as possible – preferentially within the first 48 hours of
exposure to HBV. Individuals previously vaccinated but who have an anti-HBs titer
of <10 IU/L should also be vaccinated both actively and passively. No action is
required if an anti-HBs titer of >100 IU/l is documented; active vaccination alone is
sufficient for persons with intermediate anti-HBs titers between 10 and 100 IU/L
(Cornberg 2011).
Safety of HBV vaccines
Several hundred million individuals have been vaccinated against hepatitis B. The
vaccine is very well tolerated. Injection site reactions in the first 1-3 days and mild
general reactions are common, although they are usually not long lasting. Whether
there is a causal relationship between the vaccination and the seldomly observed
neurological disorders occurring around the time of vaccination is not clear. In the
majority of these case reports the concomitant events most likely occurred
coincidentally and are independent and not causally related. That hepatitis B
vaccination causes and induces acute episodes of multiple sclerosis or other
demyelating diseases are repeatedly discussed (Geier 2001, Hernan 2004, Girard
2005). However, there are no scientific facts proving such a relationship. Numerous
studies have not been able to find a causal relationship between the postulated
disease and the vaccination (Sadovnick 2000, Monteyne 2000, Ascherio 2001,
Confavreux 2001, Schattner 2005).
Long-term immunogenicity of hepatitis B vaccination
Several studies have been published in recent years investigating the long-term
efficacy of HBV vaccination. After 10-15 years, between one third and two thirds of
vaccinated individuals have completely lost anti-HBs antibodies and only a minority
maintain titers of >100 IU/L. However, in low/intermediate endemic countries such
as Italy, this loss in protective humoral immunity did not lead to many cases of
acute or even chronic HBV infection (Zanetti 2005). To what extent memory B and
T cell responses contribute to a relative protection against HBV in the absence of
anti-HBs remains to be determined. Nevertheless, in high-endemic countries such as
Gambia a significant proportion of infants develop anti-HBc indicating active HBV
infection (18%) and some children develop chronic hepatitis B (van der Sande
2007). Thus, persons at risk should receive booster immunization if HBs antibodies
have been lost.
114  Hepatology 2012
Prevention of vertical HBV transmission
Infants of HBsAg-positive mothers should be immunized actively and passively
within 12 hours of birth. This is very important as the vertical HBV transmission
rate can be reduced from 95% to <5% (Ranger-Rogez 2004). Mothers with high
HBV viremia, of >1 million IU/ml, should receive in addition antiviral therapy with
a potent HBV polymerase inhibitor (European Association For The Study Of The
Liver 2009, Peterson 2011, Han 2011). Tenofovir and telbivudine have been
classified as Category B drugs by the FDA and can therefore be given during
pregnancy as no increased rates of birth defects have been reported. If active/passive
immunization has been performed, there is no need to recommend cesarean section.
Mothers of vaccinated infants can breastfeed unless antiviral medications are being
taken by the mother, which can pass through breast milk.
Vaccination against hepatitis C
No prophylactic or therapeutic vaccine against hepatitis C is available. As re-infections after spontaneous or treatment-induced recovery from hepatitis C virus
infection have frequently been reported, the aim of a prophylactic vaccine will very
likely be not to prevent completely an infection with HCV but rather to modulate
immune responses in such a way that the frequency of evolution to a chronic state
can be reduced (Torresi 2011).
HCV specific T cell responses play an important role in the natural course of
HCV infection. The adaptive T cell response is mediated both by CD4+ helper T
cells and CD8+ killer T cells. Several groups have consistently found an association
between a strong, multispecific and maintained HCV-specific CD4+ and CD8+ T
cell response and the resolution of acute HCV infection. While CD4+ T cells seem
to be present for several years after recovery, there are conflicting data whether
HCV-specific CD8+ T cells responses persist or decline over time (Wiegand 2007).
However, several studies have observed durable HCV-specific T cells in HCV-seronegative individuals who were exposed to HCV by occupational exposure or as
household members of HCV-positive partners, but who never became HCV RNA
positive. These observations suggest that HCV-specific T cells may be induced
upon subclinical exposure and may contribute to protection against clinically
apparent HCV infection. T cell responses are usually much weaker in chronic
hepatitis C. The frequency of specific cells is low but also effector function of
HCV-specific T cells is impaired. Different mechanisms are discussed as being
responsible for this impaired T cell function, including higher frequencies of
regulatory T cells (Tregs), altered dendritic cell activity, upregulation of inhibitory
molecules such as PD-1, CTL-A4 or 2B4 on T cells and escape mutations. HCV
proteins can directly or indirectly contribute to altered functions of different
immune cells (Rehermann 2009).
To what extent humoral immune responses against HCV contribute to
spontaneous clearance of acute hepatitis C is less clear. Higher levels of neutralizing
antibodies early during the infection are associated with viral clearance (Pestka
2007). Antibodies with neutralizing properties occur at high levels during chronic
infection, although HCV constantly escapes these neutralizing antibodies (von Hahn
2007). Yet, no completely sterilizing humoural anti-HCV immunity exists in the
long-term after recovery (Rehermann 2009). Attempts to use neutralizing antibodies
Prophylaxis and Vaccination  115
to prevent HCV re-infection after liver transplant have not been successful (Gordon
2011).
Few Phase I vaccine studies based either on vaccination with HCV peptides, HCV
proteins alone or in combination with distinct adjuvants or recombinant viral vectors
expressing HCV proteins have been completed (Torresi 2011). HCV-specific T cells
or antibodies against HCV were induced by these vaccines in healthy individuals.
Studies in chimpanzees have shown that it is very unlikely that a vaccine will be
completely protective against heterologous HCV infections. However, a reasonable
approach might be the development of a vaccine that does not confer 100%
protection against acute infection but prevents progression of acute hepatitis C to
chronic infection. In any case, there are no vaccine programs that have reached
Phase III yet (Halliday 2011). Therapeutic vaccination against hepatitis C has also
been explored (Klade 2008, Wedemeyer 2009, Torresi 2011). These studies show
that induction of HCV-specific humoural or cellular immune responses is possible
even in chronically infected individuals. The first studies showed a modest antiviral
efficacy of HCV vaccination in some patients (Sallberg 2009, Habersetzer 2011,
Wedemeyer 2011). Therapeutic vaccination was also able to enhance responses to
interferon  α  and ribavirin treatment (Pockros 2010, Wedemeyer 2011). Future
studies will need to explore the potential role of HCV vaccines in combination with
direct acting antivirals against hepatitis C.
Vaccination against hepatitis E
A Phase II vaccine trial performed in Nepal with 200 soldiers showed a vaccine
efficacy of 95% for an HEV recombinant protein (Shrestha 2007). However, the
development of this vaccine has been stopped. Since then, in September 2010, data
from a very large Phase III trial were reported involving about 110,000 individuals
in China (Zhu 2010). The vaccine efficacy of HEV 239 was 100% after three doses
to prevent cases of symptomatic acute hepatitis E. However, it is currently unknown
if this HEV genotype 1 vaccine also prevents against zoonotic HEV genotype 3
infections. Moreover, vaccine efficacy in special risk groups such patients with end-stage liver disease, immunocompromised individuals or elderly persons is unknown.
Finally, the duration of protection needs to be determined (Wedemeyer 2011). It is
currently unknown if and when the Chinese vaccine HEV-239 will become
available in other countries. Until then, preventive hygienic measures remain the
only option to avoid HEV infection.
References
Ascherio A, Zhang SM, Hernan MA, Olek MJ, Coplan PM, Brodovicz K, Walker AM. Hepatitis B
vaccination and the risk of multiple sclerosis. N Engl J Med 2001;344:327-32.
(Abstract)
Bovier PA, Bock J, Ebengo TF, et al. Predicted 30-Year Protection After Vaccination With an
Aluminum-Free Virosomal Hepatitis A Vaccine. Journal of Medical Virology
2010;82:1629-34. (Abstract)
Chang MH, Chen CJ, Lai MS, et al. Universal hepatitis B vaccination in Taiwan and the
incidence of hepatocellular carcinoma in children. Taiwan Childhood Hepatoma
Study Group. N Engl J Med 1997;336:1855-9. (Abstract)
Coates T, Wilson R, Patrick G, Andre F, Watson V. Hepatitis B vaccines: assessment of the
seroprotective efficacy of two recombinant DNA vaccines. Clin Ther 2001;23: 392-403. (Abstract)
116  Hepatology 2012
Confavreux C, Suissa S, Saddier P, Bourdes V, Vukusic S. Vaccinations and the risk of relapse
in multiple sclerosis. Vaccines in Multiple Sclerosis Study Group. N Engl J Med
2001;344:319-26. (Abstract)
Corey K.E, Ross A.S, Wurcel A, Schulze Zur W.J, Kim A.Y, Lauer G.M, Chung R.T. Outcomes
and treatment of acute hepatitis C virus infection in a United States population. Clin
Gastroenterol Hepatol 2006;4:1278-82. (Abstract)
Cornberg M Protzer U, Petersen J, et al. Prophylaxis, Diagnosis and Therapy of Hepatitis B
Virus Infection - The German Guideline. Zeitschrift Fur Gastroenterologie
2011;49:871-930.
Deterding K, Tegtmeyer B, Cornberg M, et al. Hepatitis A virus infection suppresses hepatitis C
virus replication and may lead to clearance of HCV. J Hepatol 2006;45:770-8.
(Abstract)
Deterding K, Wiegand J, Gruner N, Wedemeyer H. Medical procedures as a risk factor for HCV
infection in developed countries: Do we neglect a significant problem in medical
care? J Hepatol 2008;48:1019-20. (Abstract)
Deterding K, Wiegand J, Gruner N, et al. The German Hep-Net acute hepatitis C cohort: impact
of viral and host factors on the initial presentation of acute hepatitis C virus infection.
Z Gastroenterol 2009;47:531-40. (Abstract)
EASL Clinical Practice Guidelines: Management of hepatitis C virus infection. J Hepatol
2011;55:245-64.
European Association For The Study Of The Liver (2008). EASL Clinical Practice Guidelines:
Management of chronic hepatitis B. J Hepatol 2009. (Abstract)
European Paediatric Hepatitis C Virus Network. Effects of mode of delivery and infant feeding
on the risk of mother-to-child transmission of hepatitis C virus. BJOG 2001;108:371-7. (Abstract)
European Paediatric Hepatitis C Virus Network. A significant sex--but not elective cesarean
section--effect on mother-to-child transmission of hepatitis C virus infection. J Infect
Dis 2005;192:1872-9. (Abstract)
Fox J, Nastouli E, Thomson E, Muir D, McClure M, Weber J, Fidler S. Increasing incidence of
acute hepatitis C in individuals diagnosed with primary HIV in the United Kingdom.
AIDS 2008;22:666-8. (Abstract)
Geier MR, Geier DA. Immunologic reactions and hepatitis B vaccine. Ann Intern Med
2001;134:1155. (Abstract)
Girard M. Autoimmune hazards of hepatitis B vaccine. Autoimmun Rev 2005;4:96-100.
(Abstract)
Gordon FT, Chung RT, Curry MP, et al. Molrine Monoclonal Antibody MBL-HCV1 Suppresses
Return of HCV Following Liver Transplantation. Hepatology 2011.
Habersetzer F, Honnet G, Bain C, et al. A Poxvirus Vaccine Is Safe, Induces T-Cell Responses,
and Decreases Viral Load in Patients With Chronic Hepatitis C. Gastroenterology
2011;141:890-9. (Abstract)
Halliday J, Klenerman P, Barnes E. Vaccination for hepatitis C virus: closing in on an evasive
target. Expert Review of Vaccines 2011;10:659-72. (Abstract)
Hammitt LL, Bulkow L, Hennessy TW, Zanis C, Snowball M, Williams JL, et al. Persistence of
antibody to hepatitis A virus 10 years after vaccination among children and adults. J
Infect Dis 2008;198:1776-82. (Abstract)
Han GR, Cao MK, Zhao W, et al. A prospective and open-label study for the efficacy and safety
of telbivudine in pregnancy for the prevention of perinatal transmission of hepatitis B
virus infection. J Hepatol 2011;55:1215-21. (Abstract)
Hendrickx G, Van Herck K, Vorsters A, et al. Has the time come to control hepatitis A globally?
Matching prevention to the changing epidemiology. J Viral Hepat 2008;15 Suppl 2:1-15. (Abstract)
Hernan MA, Jick SS, Olek MJ, Jick H. Recombinant hepatitis B vaccine and the risk of multiple
sclerosis: a prospective study. Neurology 2004;63:838-42. (Abstract)
Hollinger FB, Bell B, Levy-Bruhl D, Shouval D, Wiersma S, Van Damme P. Hepatitis A and B
vaccination and public health. J Viral Hepat 2007;14 Suppl 1:1-5. (Abstract)
Kallinowski B, Jilg W, Buchholz L, Stremmel W, Engler S. Immunogenicity of an accelerated
vaccination regime with a combined hepatitis a/b vaccine in patients with chronic
hepatitis C. Z Gastroenterol 2003;41:983-90. (Abstract)
Kamar N, Selves J, Mansuy JM, et al. Hepatitis E virus and chronic hepatitis in organ-transplant
recipients. N Engl J Med 2008;358:811-17. (Abstract)

Book on hepatitis from Page 96 to 105

Book on hepatitis from Page 96 to 105

96  Hepatology 2012
needs NS4A as a cofactor (Failla 1994, Tanji 1995, Bartenschlager 1995, Lin 1995,
Tomei 1996).
HCV RNA replication
The complex process of HCV RNA replication is poorly understood. The key
enzyme for viral RNA replication is NS5B, an RNA-dependent RNA polymerase
(RdRp) of HCV (Behrens 1996). In addition, several cellular as well as viral factors
have been reported to be part of the HCV RNA replication complex. One important
viral factor for the formation of the replication complex appears to be NS4B, which
is able to induce an ER-derived membranous web containing most of the non-structural HCV proteins including NS5B (Egger 2002). This web could serve as the
platform for the next steps of viral RNA replication. The RdRp uses the previously
released genomic positive-stranded HCV RNA as a template for the synthesis of an
intermediate minus-stranded RNA. Recently it has been reported that the cellular
peptidyl-prolyl isomerases cyclophilin A, B and C (Cyp A, Cyp B, and Cyp C)
could stimulate binding of the RdRp to the viral RNA resulting in increased HCV
RNA synthesis (Watashi 2005, Nakagawa 2005, Yang 2008, Heck 2009). However,
these reports are in part inconsistent and further studies are needed in order to
investigate the involvement of cyclophilins in HCV RNA replication.
After the viral polymerase has bound to its template, the NS3 helicase is assumed
to unwind putative secondary structures of the template RNA in order to facilitate
the synthesis of minus-strand RNA (Jin 1995, Kim 1995). In turn, again with the
assistance of the NS3 helicase, the newly synthesized antisense RNA molecule
serves as the template for the synthesis of numerous plus-stranded RNA. The
resulting sense RNA may be used subsequently as genomic RNA for HCV progeny
as well as for polyprotein translation.
Assembly and release
After the viral proteins, glycoproteins, and the genomic HCV RNA have been
synthesized these single components have to be arranged in order to produce
infectious virions. As is the case for all other steps in the HCV lifecycle viral
assembly is a multi-step procedure involving most viral components along with
many cellular factors. Investigation of viral assembly and particle release is still in
its infancy since the development of in vitro models for the production and release
of infectious HCV occurred only recently. Previously, it was reported that core
protein molecules were able to self-assemble in vitro, yielding nucleocapsid-like
particles. More recent findings suggest that viral assembly takes place within the
endoplasmic reticulum (Gastaminza 2008) and that lipid droplets (LD) are involved
in particle formation (Moradpour 1996, Barba 1997, Miyanari 2007, Shavinskaya
2007, Appel 2008). It appears that LD-associated core protein targets viral non-structural proteins and the HCV RNA replication complex including positive- and
negative-stranded RNA from the endoplasmic reticulum to the LD (Miyanari 2007).
Beside the core protein, LD-associated NS5A seems to play a key role in the
formation of infectious viral particles (Appel 2008). Moreover, E2 molecules are
detected in close proximity to LD-associated membranes. Finally, spherical virus-like particles associated with membranes can be seen very close to the LD. Using
HCV Virology  97
specific antibodies the virus-like particles were shown to contain core protein as
well as E2 glycoprotein molecules indicating that these structures may represent
infectious HCV (Miyanari 2007). However, the precise mechanisms for the
formation and release of infectious HCV particles are still unknown.
Model systems for HCV research
For a long time HCV research was limited due to a lack of small animal models and
efficient cell culture systems. The development of the first HCV replicon system
(HCV RNA molecule, or region of HCV RNA, that replicates autonomously from a
single origin of replication) 10 years after the identification of the hepatitis C virus
offered the opportunity to investigate the molecular biology of HCV infection in a
standardized manner (Lohmann 1999).
HCV replicon systems. Using total RNA derived from the explanted liver of an
individual chronically infected with HCV genotype 1b, the entire HCV ORF
sequence was amplified and cloned in two overlapping fragments. The flanking
NTRs were amplified and cloned separately and all fragments were assembled into
a modified full-length sequence. Transfection experiments with in vitro transcripts
derived from the full-length clones failed to yield viral replication. For this reason,
two different subgenomic replicons consisting of the 5’IRES, the neomycin
phosphotransferase gene causing resistance to the antibiotic neomycin, the IRES
derived from the encephalomyocarditis virus (EMCV) and the NS2-3’NTR or NS3-3’NTR sequence, respectively, were generated (Figure 3).
Figure 3. Structure of subgenomic HCV replicons (Lohmann 1999). This figure illustrates
the genetic information of in vitro transcripts used for Huh7 transfection. A) Full-length transcript
derived from the explanted liver of a chronically infected subject. B) Subgenomic replicon
lacking the structural genes and the sequence encoding p7. C) Subgenomic replicon lacking C,
E1, E2, p7, and NS2 genes. neo, neomycin phosphotransferase gene; E-I, IRES of the
encephalomyocarditis virus (EMCV).
98  Hepatology 2012
In vitro transcripts derived from these constructs lacking the genome region
coding for the structural HCV proteins were used to transfect the hepatoma cell line
Huh7 (Lohmann 1999). The transcripts are bicistronic, i.e., the first cistron
containing the HCV IRES enables the translation of the neomycin
phosphotransferase as a tool for efficient selection of successfully transfected cells
and the second cistron containing the EMCV IRES directs translation of the HCV-specific proteins. Only some Huh7 clones can replicate replicon-specific RNA in
titres of approximately 10
8
positive-stranded RNA copies per microgram total RNA.
Moreover, all encoded HCV proteins are detected predominantly in the cytoplasm
of the transfected Huh7 cells. The development of this replicon is a milestone in
HCV research with regard to the investigation of HCV RNA replication and HCV
protein analyses.
More recently, the methodology has been improved in order to achieve
significantly higher replication efficiency. Enhancement of HCV RNA replication
was achieved by the use of replicons harbouring cell culture-adapted point
mutations or deletions within the NS genes (Blight 2000, Lohmann 2001, Krieger
2001). Further development has led to the generation of selectable full-length HCV
replicons, i.e., genomic replicons that also contain genetic information for the
structural proteins Core, E1, and E2 (Pietschmann 2002,  Blight 2002). This
improvement offered the opportunity to investigate the influence of the structural
proteins on HCV replication. Thus it has been possible to analyse the intracellular
localisation of these proteins althoughviral assembly and release has not been
achieved.
Another important milestone was reached when a subgenomic replicon based on
the HCV genotype 2a strain JFH-1 was generated (Kato 2003). This viral strain
derived from a Japanese subject with fulminant hepatitis C (Kato 2001). The
corresponding replicons showed higher RNA replication efficiency than previous
replicons. Moreover, cell lines distinct from Huh7, such as HepG2 or HeLa were
transfected efficiently with transcripts derived from the JFH-1 replicon (Date 2004,
Kato 2005).
HCV pseudotype virus particles (HCVpp).  The generation of retroviral
pseudotypes bearing HCV E1 and E2 glycoproteins (HCVpp) offers the opportunity
to investigate E1-E2-dependent HCVpp entry into Huh7 cells and primary human
hepatocytes (Bartosch 2003a, Hsu 2003, Zhang 2004). In contrast to the HCV
replicons where cells were transfected with HCV-specific synthetic RNA
molecules, this method allows a detailed analysis of the early steps in the HCV life
cycle, e.g., adsorption and viral entry.
Infectious HCV particles in cell culture (HCVcc). Transfection of Huh7 and
‘cured’ Huh7.5 cells with full-length JFH-1 replicons led for the first time to the
production of infectious HCV virions (Zhong 2005, Wakita 2005). The construction
of a chimera with the core NS2 region derived from HCV strain J6 (genotype 2a)
and the remaining sequence derived from JFH-1 improved infectivity. Importantly,
the secreted viral particles are infectious in cell culture (HCVcc) (Wakita 2005,
Zhong 2005, Lindenbach 2005) as well as in chimeric mice with human liver grafts
as well as in chimpanzees (Lindenbach 2006).
An alternative strategy for the production of infectious HCV particles was
developed (Heller 2005): a full-length HCV construct (genotype 1b) was placed
between two ribozymes in a plasmid containing a tetracycline-responsive promoter.
HCV Virology  99
Huh7 cells were transfected with those plasmids, resulting in efficient viral
replication with HCV RNA titres of up to 10
7
copies/ml cell culture supernatant.
The development of cell culture systems that allow the production of infectious
HCV represents a breakthrough for HCV research and it is now possible to
investigate the whole viral life cycle from viral adsorption to virion release. These
studies will help to better understand the mechanisms of HCV pathogenesis and
they should significantly accelerate the development of HCV-specific antiviral
compounds.
Small animal models.  Very recently, substantial progress was achieved in
establishing two mouse models for HCV infection viagenetically humanized mice
(Dorner 2011). In this experiment immunocompetent mice were transduced using
viral vectors containing the genetic information of four human proteins involved in
adsorption and entry of HCV into hepatocytes (CD81, SR-BI, CLDN1, OCLN).
This humanisation procedure enabled the authors to infect the transduced mice with
HCV. Although this mouse model does not enable complete HCV replication in
murine hepatocytes it will be useful to investigate the early steps of HCV infection
in vivo. Moreover, the approach should be suitable for the evaluation of HCV entry
inhibitors and vaccine candidates.
A second group of investigators have chosen another promising strategy for
HCV-specific humanisation of mice. After depleting murine hepatocytes human
CD34(+) hematopoietic stem cells and hepatocyte progenitors were cotransplantated
into transgenic mice leading to efficient engraftment of human leukocytes and
hepatocytes, respectively (Washburn 2011). A portion of the humanised mice
became infectable with primary HCV isolates resulting in low-level HCV RNA in
the murine liver. As a consequence HCV infection induced liver inflammation,
hepatitis, and fibrosis. Furthermore, due to the cotransplantation of CD34(+) human
hematopoietic stem cells, an HCV-specific T cell immune response could be
detected.
Both strategies are promising and have already delivered new insights into viral
replication and the pathogenesis of HCV. However, the methods lack some
important aspects and need to be improved. As soon as genetically humanised mice
that are able to replicate HCV completely are created they can be used for the
investigation of HCV pathogenesis and HCV-specific immune responses. The
Washburn method should be improved in order to achieve higher HCV replication
rates. Moreover, reconstitution of functional human B cells would make this mouse
model suitable to study the important HCV-specific antibody response.
References
Acton SL, Scherer PE, Lodish HF, et al. Expression cloning of SR-BI, a CD36-related class B
scavenger receptor. J Biol Chem 1994;269:21003-9. (Abstract)
Acton S, Rigotti A, Landschulz KT, et al. Identification of scavenger receptor BI as a high-density lipoprotein receptor. Science 1996;271:518-20. (Abstract)
Agnello V, Abel G, Elfahal M, et al. Hepatitis C virus and other Flaviviridae viruses enter cells
via low-density lipoprotein receptor. Proc Natl Acad Sci U S A 1999;96:12766-71.
(Abstract)
Ago H, Adashi T, Yoshida A, et al. Crystal structure of the RNA-dependent RNA polymerase of
hepatitis C virus. Structure 1999;7:1417-26. (Abstract)
Ait-Goughoulte M, Hourioux C, Patient R, et al. Core protein cleavage by signal peptide
peptidase is required for hepatitis C virus-like particle assembly. J Gen Virol
2006;87:855-60. (Abstract)
100  Hepatology 2012
Akazawa D, Date T, Morikawa K, et al. CD81 expression is important for the permissiveness of
Huh7 cell clones for heterogeneous hepatitis C virus infection. J Virol 81:5036-5045.
(Abstract)
Alter HJ, Holland PV, Purcell RH, et al. Transmissible agent in non-A, non-B hepatitis. Lancet
1978;1:459-63. (Abstract)
Andre P, Komurian-Pradel F, Deforges S, et al. Characterization of low- and very-low-density
hepatitis C virus RNA-containing particles. J Virol 2002;76:6919-28. (Abstract)
Appel N, Zayas M, Miller S, et al. Essential role of domain III of nonstructural protein 5A for
hepatitis C virus infectious particle assembly. PLoS Pathog 2008;4:e1000035.
(Abstract)
Barba G, Harper F, Harada T, et al. Hepatitis C virus core protein shows a cytoplasmic
localization and associates to cellular lipid storage droplets. Proc Natl Acad Sci U S A
1997;94:1200-5. (Abstract)
Bartenschlager R, Ahlborn-Laake L, Mous J, et al. Nonstructural protein 3 of hepatitis C virus
encodes a serine-type proteinase required for cleavage at the NS3/4 and NS4/5
junctions. J Virol 1993;67:3835-44. (Abstract)
Bartenschlager R, Lohmann V, Wilkinson T, et al. Complex formation between the NS3
serinetype proteinase of the hepatitis C virus and NS4A and its importance for
polyprotein maturation. J Virol 1995;69:7519-28. (Abstract)
Barth H, Schäfer C, Adah MI, et al. Cellular binding of hepatitis C virus envelope glycoprotein
E2 requires cell surface heparan sulphate. J Biol Chem 2003;278:41003-12.
(Abstract)
Bartosch B, Dubuisson J, Cosset FL. Infectious hepatitis C virus pseudoparticles containing
functional E1-E2 envelope protein complexes. J Exp Med 2003a;197:633-42.
(Abstract)
Bartosch B, Vitelli A, Granier C, et al. Cell entry of hepatitis C virus requires a set of
coreceptors that include CD81 and SR-B1 scavenger receptor. J Biol Chem
2003b;278:41624-30. (Abstract)
Behrens SE, Tomei L, DeFrancesco R. Identification and properties of the RNA-dependent
RNA polymerase of hepatitis C virus. EMBO J 1996;15:12-22. (Abstract)
Bertaux C, Dragic T. Different domains of CD81 mediate distinct stages of hepatitis C virus
pseudoparticle entry. J Virol 2006;80:4940-8. (Abstract)
Blanchard E, Belouzard S, Goueslain L, et al. Hepatitis C virus entry depends on clathrin-mediated endocytosis. J Virol 2006;80:6964-72. (Abstract)
Blight KJ and Rice CM. Secondary structure determination of the conserved 98-base sequence
at the 3´terminus of hepatitis C virus genome RNA. J Virol 1997;71:7345-52.
(Abstract)
Blight KJ, Kolykhalov AA, Rice CM. Efficient initiation of HCV RNA replication in cell culture.
Science 2000;290:1972-4. (Abstract)
Blight KJ, McKeating JA, Rice CM. Highly permissive cell lines for subgenomic and genomic
hepatitis C virus RNA replication. J Virol 2002;76:13001-14. (Abstract)
Bradley DW, McCaustland KA, Cook EH, et al. Post-transfusion non-A, non-B hepatitis in
chimpanzees: physiochemical evidence that the tubule-forming agent is a small,
enveloped virus. Gastro 1985;88:773-9. (Abstract)
Bradley D, McCaustland K, Krawczynski K, et al. Hepatitis C virus: buoyant density of the factor
VIII-derived isolate in sucrose. J Med Virol 1991;34:206-8. (Abstract)
Bradrick SS, Walters RW, Gromeier M. The hepatitis C virus 3'-untranslated region or a poly(A)
tract promote efficient translation subsequent to the initiation phase. Nucleic Acids
Res 2006;34:1293-303. (Abstract)
Branch AD, Stumpp DD, Gutierrez JA et al. The hepatitis virus alternate reading frame (ARF)
and its family of novel products: the alternate reading frame protein/F-protein, the
double-frameshift protein, and others. Semin Liv Dis 2005;25:105-17.
Bressanelli S, Tomei L, Roussel A, et al. Crystal structure of the RNA-dependent RNA
polymerase of hepatitis C virus. Proc Natl Acad Sci USA 1999;96:13034-9. (Abstract)
Bukh J, Miller RH, Purcell RH. Biology and genetic heterogeneity of hepatitis C virus. Clin Exp
Rheumatol 1995;Suppl 13:S3-7. (Abstract)
Choo QL, Kuo G, Weiner AJ, et al. Isolation of a cDNA clone derived from a blood-borne non-A,
non-B viral hepatitis genome. Science 1989;244:359-62. (Abstract)
Choo QL, Richman KH, Han JH, et al. Genetic organization and diversity of the hepatitis C
virus. Proc Natl Acad Sci U S A 1991;88:2451-5. (Abstract)
HCV Virology  101
Cocquerel L, Meunier JC, Pillez A, et al. A retention signal necessary and sufficient for
endoplasmic reticulum localization maps to the transmembrane domain of hepatitis C
virus glycoprotein E2. J Virol 1998;72:2183-91. (Abstract)
Cocquerel L, Duvet S, Meunier JC, et al. The transmembrane domain of hepatitis C virus
glycoprotein E1 is a signal for static retention in the endoplasmic reticulum. J Virol
1999;73:2641-9. (Abstract)
Cocquerel L, Meunier JC, Op de Beeck A, et al. Coexpression of hepatitis C virus envelope
proteins E1 and E2 in cix improves the stability of membrane insertion of E2. J Gen
Virol 2001;82:1629-35. (Abstract)
Codran A, Royer C, Jaeck D, et al. Entry of hepatitis C virus pseudotypes into primary human
hepatocytes by clathrin-dependent endocytosis. J Gen Virol 2006;87:2583-93.
(Abstract)
Cormier EG, Tsamis F, Kajumo F, et al. CD81 is an entry coreceptor for hepatitis C virus. Proc
Natl Acad Sci USA 2004;101:7270-4. (Abstract)
Date T, Kato T, Miyamoto M, et al. Genotype 2a hepatitis C virus subgenomic replicon can
replicate in HepG2 and IMY-N9 cells. J Biol Chem 2004;279:22371-6. (Abstract)
Dorner M, Horwitz JA, Robbins JB, et al. A genetically humanized mouse model for hepatitis C
virus infection. Nature 2011;474:208-11. (Abstract)
Duvet S, Cocquerel L, Pillez A, et al. Hepatitis C virus glycoprotein complex localization in the
endoplasmic reticulum involves a determinant for retention and not retrieval. J Biol
Chem 1998;273:32088-95. (Abstract)
Duvet S, Op de Beeck A, Cocquerel L, et al. Glycosylation of the hepatitis C virus envelope
protein E1 occurs posttranslationally in a mannosylphosphoryldolichol-deficient CHO
mutant cell line. Glycobiology 2002;12:95-101. (Abstract)
Eckart MR, Selby M, Maisiarz F, et al. The hepatitis C virus encodes a serine protease involved
in processing of the putative non-structural proteins from the viral polyprotein
precursor. Biochem Biophys Res Commun 1993;192:399-406. (Abstract)
Egger D, Wölk B, Gosert R, et al. Expression of hepatitis C virus proteins induce distinct
membrane alterations including candidate viral replication complex. J Virol
2002;76:5974-84. (Abstract)
Einav S, Elazar M, Danieli T, et al. A nucleotide binding motif in hepatitis C virus (HCV) NS4B
mediates HCV RNA replication. J Virol 2004;78:11288-95. (Abstract)
Elazar M, Liu P, Rice CM, et al. An N-terminal amphipathic helix in hepatitis C virus (HCV)
NS4B mediates membrane association, correct localization of replication complex
proteins, and HCV RNA replication. J Virol 2004;78:11393-400. (Abstract)
Enomoto N, Sakuma I, Asahina Y, et al. Comparison of full-length sequences of interferon
sensitive and resistant hepatitis C virus 1b. Sensitivity to interferon is conferred by
amino acid substitutions in the NS5A region. J Clin Investig 1995;96:224-30.
(Abstract)
Enomoto N, Sakuma I, Asahina Y, et al. Mutations in the nonstructural protein 5A gene and
response to interferon in patients with chronic hepatitis C virus 1b infection. N Engl J
Med 1996;334:77-81. (Abstract)
Evans MJ, von Hahn T, Tscherne DM, et al. Claudin-1 is a hepatitis C virus co-receptor
required for a late step in entry. Nature 2007;446:801-5. (Abstract)
Failla C, Tomei L, DeFrancesco R. Both NS3 and NS4A are required for proteolytic processing
of hepatitis C virus nonstructural proteins. J Virol 1994;68:3753-60. (Abstract)
Farci P, Shimoda A, Wong D, et al. Prevention of hepatitis C virus infection in chimpanzees by
hyperimmune serum against the hypervariable region 1 of the envelope 2 protein.
Proc Nat. Acad Sci USA 1996;93:15394-9. (Abstract)
Feinstone SM, Kapikian AZ, Purcell RH, et al. Transfusion-associated hepatitis not due to viral
hepatitis type A or B. N Engl J Med 1975;292:767-70.
Feinstone SM, Mihalik KB, Purcell AH, et al. Inactivation of hepatitis B virus and non-A, non-B
hepatitis by chloroform. Infect Immun 1983;41:816-21. (Abstract)
Flint M, Maidens C, Loomis-Price LD, et al. Characterization of hepatitis C virus E2 glycoprotein
interaction with a putative cellular receptor, CD81. J Virol 1999;73:6235-44.
(Abstract)
Friebe P, Lohmann V, Krieger N, et al. Sequences in the 5' nontranslated region of hepatitis C
virus required for RNA replication. J Virol 2001;75:12047-57. (Abstract)
Friebe P and Bartenschlager R. Genetic analysis of sequences in the 3' nontranslated region of
hepatitis C virus that are important for RNA replication. J Virol 2002;76:5326-38.
(Abstract)
102  Hepatology 2012
Friebe P, Boudet J, Simorre JP, et al. Kissing-loop interaction in the 3' end of the hepatitis C
virus genome essential of RNA replication. J Virol 2005;79:380-92. (Abstract)
Fried MW, Shiffman ML, Reddy KR, et al. Peginterferon Alfa-2a plus Ribavirin for Chronic
Hepatitis C Virus Infection. N Engl J Med 2002;347:975-82. (Abstract)
Fukushi S, Katayama K, Kurihara C, et al. Complete 5' noncoding region is necessary for the
efficient internal initiation of hepatitis C virus RNA. Biochem Biophys Res Commun
1994;199:425-32. (Abstract)
Furuse M, Fujita K, Hiiragi T, et al. Claudin-1 and -2: novel integral membrane proteins
localizing at tight junctions with no sequence similarity to occludin. J Cell Biol
1998;141:1539-50. (Abstract)
Gardner JP, Durso RJ, Arrigale RR, et al. L-SIGN (CD209L) is a liver-specific capture receptor
for hepatitis C virus. Proc Nat. Acad Sci USA 2003;100:4498-503.
Gastaminza P, Cheng G, Wieland S, et al. Cellular determinants of hepatitis C virus assembly,
maturation, degradation, and secretion. J Virol 2008;82:2120-9. (Abstract)
Germi R, Crance JM, Garin D, et al. Cellular glycosaminglycans and low density lipoprotein
receptor are involved in hepatitis C virus adsorption. J Med Virol 2002;68:206-15.
(Abstract)
Grakoui A, McCourt DW, Wychowski C, et al. Characterization of hepatitis C virus-encoded
serine proteinase: determination of proteinase-dependent polyprotein cleavage sites.
J Virol 1993a;67:2832-43. (Abstract)
Grakoui A, McCourt DW, Wychowski C, et al. A second hepatitis C virus-encoded proteinase.
Proc Natl Acad Sci USA 1993b;90:10583-7. (Abstract)
Gretton SN, Taylor AI, McLauchlan J. Mobility of the hepatitis C virus NS4B protein on the
endoplasmic reticulum membrane and membrane-associated foci. J Gen Virol
2005;86:1415-21. (Abstract)
Griffin SD, Beales LP, Clarke DS, et al. The p7 protein of hepatitis C virus forms an ion channel
that is blocked by the antiviral drug, Amantadine. FEBS Lett 2003;535:34-8.
(Abstract)
Grove J, Huby T, Stamataki Z, et al. Scavenger receptor BI and BII expression levels modulate
hepatitis C virus infectivity. J Virol 2007;81: 3162-9. (Abstract)
Haqshenas G, Mackenzie JM, Dong X, et al. Hepatitis C virus p7 protein is localized in the
endoplasmic reticulum when it is encoded by a replication-competent genome. J Gen
Virol 2007;88:134-42. (Abstract)
Heck JA, Meng X, Frick DN. Cyclophilin B stimulates RNA synthesis by the HCV RNA
dependent RNA polymerase. Biochem Pharmacol 2009;1173-80. (Abstract)
Helle F, Dubuisson J. Hepatitis C virus entry into host cells. Cell Mol Life Sci 2008, 65:100-12.
Hellen CU, Pestova TV. Translation of hepatitis C virus RNA. J Viral Hepat
1999;6:79-87.
Heller TS, Saito J, Auerbach T, et al. An in vitro model of hepatitis C virion production. Proc
Nat. Acad Sci USA 2005;102:2579-83. (Abstract)
Heo TH, Chang JH, Lee JW, et al. Incomplete humoral immunity against hepatitis C virus is
linked with distinct recognition of putative multiple receptors by E2 envelope
glycoprotein. J Immunol 2004;173:446-55. (Abstract)
Hollinger FB, Gitnick GL, Aach RD, et al. Non-A, non-B hepatitis transmission in chimpanzees:
a project of the Transfusion-transmitted Viruses Study Group. Intervirology
1978;10:60-8. (Abstract)
Honda M, Beard MR, Ping LH, et al. A phylogenetically conserved stem-loop structure at the 5´
border of the internal ribosome entry site of hepatitis C virus is required for cap-independent viral translation. J Virol 1999;73:1165-74.
Hsu M, Zhang J, Flint M, et al. Hepatitis C virus glycoproteins mediate pH-dependent cell entry
of pseudotyped retroviral particles. Proc Natl Acad Sci USA 2003;100:7271-6.
(Abstract)
Hüssy P, Langen H, Mous J, et al. Hepatitis C virus core protein: carboxy-terminal boundaries
of two processed species suggest cleavage by a signal peptide peptidase. Virology
1996;224-93-104. (Abstract)
Ito T and Lai MMC. Determination of the secondary structure of and cellular protein binding to
the 3'-untranslated region of the hepatitis C virus RNA genome. J Virol 1997;71:8698-706. (Abstract)
Jennings TA, Chen Y, Sikora D, et al. RNA unwinding activity of the hepatitis C virus NS3
helicase is modulated by the NS5B polymerase. Biochemistry 2008;47:1126-35.
(Abstract)
HCV Virology  103
Jeong SH, Qiao M, Nascimbeni M, et al. Immunization with hepatitis C virus-like particles
induces humoral and cellular immune responses in honhuman primates. J Virol
2004;78:6995-7003. (Abstract)
Jin L and Peterson DL. Expression, isolation, and characterization of the hepatitis C virus
ATPase/RNA helicase. Arch Biochem Biophys 1995;323:47-53. (Abstract)
Jones CT, Murray CL, Eastman DK, et al. Hepatitis C virus p7 and NS2 proteins are essential
for production of infectious virus. J Virol 2007;81:8374-83. (Abstract)
Kaito M, Watanabe S, Tsukiyama-Kohara K, et al. Hepatitis C virus particle detected by
immunoelectron microscopy study. J Gen Virol 1994;75:1755-60. (Abstract)
Kapadia SB, Barth H, Baumert T, et al. Initiation of hepatitis C virus infection is dependent on
cholesterol and cooperativity between CD81 and scavenger receptor B type I. J Virol
2007;81:374-83. (Abstract)
Kato J, Kato N, Yoshida H, et al. Hepatitis C virus NS4A and NS4B proteins suppress
translation in vivo. J Med Virol 2002;66:187-99. (Abstract)
Kato N. Molecular virology of hepatitis C virus. Acta Med Okayama 2001;55:133-159. (Abstract)
Kato T, Furusaka A, Miyamoto M, et al. Sequence analysis of hepatitis C virus isolated from a
fulminant hepatitis patient. J Med Virol 2001;64:334-9. (Abstract)
Kato T, Date T, Miyamoto M, et al. Efficient replication of the genotype 2a hepatitis C virus
subgenomic replicon. Gastroenterology 2003;125:1808-17. (Abstract)
Kato T, Date T, Miyamoto M, et al. Nonhepatic cell lines HeLa and 293 support efficient
replication of hepatitis C virus genotype 2a subgenomic replicon. J Virol 2005;79:592-6. (Abstract)
Kim DW, Gwack Y, Han JH, et al. C-terminal domain of the hepatitis C virus NS3 protein
contains an RNA helicase activity. Biochem Biophys Res Commun 1995;215:160-6.
(Abstract)
Kim YK, Kim CS, Lee SH, et al. Domains I and II in the 5' nontranslated region of the HCV
genome are required for RNA replication. Biochem Biophys Res Commun
2002;290:105-12. (Abstract)
Kolykhalov AA, Feinstone SM, Rice SM. Identification of a highly conserved sequence element
at the 3´terminus of hepatitis C virus genome RNA. J Virol 1996;70:3363-71.
(Abstract)
Kolykhalov AA, Mihalik K, Feinstone SM, et al. Hepatitis C virus-encoded enzymatic activities
and conserved RNA elements in the 3' nontranslated region are essential for virus
replication in vivo. J Virol 2000;74:2046-51.. (Abstract)
Komurian-Pradel F, Rajoharison A, Berland JL, et al. Antigenic relevance of F protein in chronic
hepatitis C virus infection. Hepatology 2004;40:900-9. (Abstract)
Kou YH, Chou SM, Wang YM, et al. Hepatitis C virus NS4A inhibits cap-dependent and the
viral IRES-mediated translation through interacting with eukaryotic elongation factor
1A. J Biomed Sci 2006;13:861-74. (Abstract)
Koutsoudakis G, Kaul A, Steinmann E, et al. Characterization of the early steps of hepatitis C
virus infection by using luciferase reporter viruses. J Virol 2006;80:5308-20.
(Abstract)
Krieger N, Lohmann V, Bartenschlager R. Enhancement of hepatitis C virus RNA replication by
cell culture-adaptive mutations. J Virol 2001;75:4614-24. (Abstract)
Kuo G, Choo QL, Alter HJ, et al. An assay for circulating antibodies to a major etiologic virus of
human non-A, non-B hepatitis. Science 1989;244:362-4. (Abstract)
Lavillette D, Tarr AW, Voisset C, et al. Characterization of host-range and cell entry properties
of the major genotypes and subtypes of hepatitis C virus. Hepatology 2005;41:265-74. (Abstract)
Lavillette D, Pécheur EI, Donot P, et al. Characterization of fusion determinants points to the
involvement of three discrete regions of both E1 and E2 glycoproteins in the
membrane fusion process of hepatitis C virus. J Virol 2007;81:8752-65. (Abstract)
Lesburg CA, Cable MB, Ferrari E, et al. Crystal structure of the RNA-dependent RNA
polymerase from hepatitis C virus reveals a fully encircled active site. Nat Struct Biol
1999;6:937-43. (Abstract)
Lin C, Thomson JA, Rice CM. A central region in the hepatitis C virus NS4A protein allows
formation of an active NS3-NS4A serine proteinase complex in vivo and in vitro. J
Virol 1995;69:4373-80. (Abstract)
Lindenbach BD, Evans MJ, Syder AJ, et al. Complete replication of hepatitis C virus in cell
culture. Science 2005;309:623-6. (Abstract)
104  Hepatology 2012
Lindenbach BD, Meuleman P, Ploss A, et al. Cell culture-grown hepatitis C virus is infectious in
vivo and can be recultured in vitro. Proc Natl Acad Sci USA 2006;103:3805-9.
(Abstract)
Liu S, Yang W, Shen L, et al. Tight junction proteins claudin-1 and occludin control hepatitis C
virus entry and are downregulated during infection to prevent superinfection. J Virol
2009;83:2011-4. (Abstract)
Lohmann V, Körner F, Koch JO, et al. Replication of subgenomic hepatitis C virus RNAs in a
hepatoma cell line. Science 1999;285:110-3. (Abstract)
Lohmann V, Körner F, Dobierzewska A, et al. Mutations in hepatitis C virus RNAs conferring
cell culture adaptation. J Virol 2001;75:1437-49. (Abstract)
Lozach PY, Lortat-Jacob H, de Lacroix de Lavalette A, et al. DC-SIGN and L-SIGN are high
affinity binding receptors for hepatitis C virus glycoprotein E2. J Biol Chem
2003;278:20358-66. (Abstract)
Manns MP, McHutchinson JG, Gordon SC, et al. Peginterferon alfa-2b plus ribavirin compared
with interferon alfa-2b plus ribavirin for initial treatment of chronic hepatitis C: a
randomised trial. Lancet 2001;358:958-65. (Abstract)
Matsumoto M, Hwang SB, Jeng KS, et al. Homotypic interaction and multimerization of
hepatitis C virus core protein. Virology 1996;218:43-51. (Abstract)
McKeating JA, Zhang LQ, Logvinoff C, et al. Diverse hepatitis C virus glycoproteins mediate
viral infection in a CD81-dependent manner. J Virol 2004;78:8496-505. (Abstract)
McLauchlan J, Lemberg MK, Hope G, et al. Intramembrane proteolysis promotes trafficking of
hepatitis C virus core protein to lipid droplets. EMBO J 2002;21:3980-8. (Abstract)
Meertens L, Bertaux C, Dragic T. Hepatitis C virus entry requires a critical postinternalization
step and delivery to early endosomes via clathrin-coated vesicles. J Virol.
2006;80:11571-8. (Abstract)
Meertens L, Bertaux C, Cukierman L, et al. The tight junction proteins claudin-1, -6, and -9 are
entry cofactors for hepatitis C virus. J Virol 2008;82:3555-60. (Abstract)
Miyanari Y, Atsuzawa K, Usuda N, et al. The lipid droplet is an important organelle for hepatitis
C virus production. Nat Cell Biol 2007;9:961-9. (Abstract)
Molina S, Castet V, Fournier-Wirth C, et al. The low-density lipoprotein receptor plays a role in
the infection of primary human hepatocytes by hepatitis C virus. J Hepatol
2007;46:411-9. (Abstract)
Monazahian M, Böhme I, Bonk S, et al. Low-density lipoprotein receptor as a candidate
receptor for hepatitis C virus. J Med Virol 1999;57:223-9. (Abstract)
Moradpour D, Englert C, Wakita T, et al. Characterization of cell lines allowing tightly regulated
expression of hepatitis C virus core protein. Virology 1996;222:52-63. (Abstract)
NahmiasY, Casali M, Barbe L, et al. Liver endothelial cells promote LDL-R expression and the
uptake of HCV-like particles in primary rat and human hepatocytes. Hepatology
2006;43:257-65. (Abstract)
Nakagawa M, Sakamoto N, Tanabe Y, et al. Suppression of hepatitis C virus replication by
cyclosporin A is mediated by blockade of cyclophilins. Gastroenterology 2005;129-1031-41. (Abstract)
Ndjomou J, Pybus OG, Matz B. Phylogenetic analysis of hepatitis C virus isolates indicates a
unique pattern of endemic infection in Cameroon. J Gen Virol 2003;84:2333-41.
(Abstract)
Ohba K, Mizokami M, Lau JYN, et al. Evolutionary relationship of hepatitis C, pesti-, flavi-,
plantviruses, and newly discovered GB hepatitis agents. FEBS Lett 1996;378:232-4.
(Abstract)
Op de Beeck A, Montserret R, Duvet S, et al. The transmembrane domains of hepatitis C virus
envelope glycoprotein E1 and E2 play a major role in heterodimerization. J Biol
Chem 2000;275:31428-37. (Abstract)
Otto GA, Lukavsky PJ, Lancaster AM, et al. Ribosomal proteins mediate the hepatitis C virus
IRES-HeLa 40S interaction. RNA 2002;8:913-23. (Abstract)
Pavlovic D, Neville DC, Argaud O, et al. The hepatitis C virus p7 protein forms an ion channel
that is inhibited by long-alkyl-chain iminosugar derivatives. Proc Nat. Acad Sci USA
2003;100:6104-8. (Abstract)
Pawlotsky JM, Chevalier S, McHutchinson JG. The hepatitis C virus life cycle as a target for
new antiviral therapies. Gastroenterology 2007;132:1979-98. (Abstract)
Pietschmann T, Lohmann V, Kaul A, et al. Persistent and transient replication of full-length
hepatitis C virus genomes in cell culture. J Virol 2002;76:4008-21. (Abstract)
HCV Virology  105
Pileri P, Uematsu Y, Campagnoli S, et al. Binding of hepatitis C virus to CD81. Science 1998,
282:938-941. Ploss A, Evans MJ, Gaysinskaya VA, et al. Human occluding is a
hepatitis C virus entry factor required for infection of mouse cells. Nature
2009;457:882-6. (Abstract)
Ploss A, Evans MJ, Gaysinskaya VA, et al. Human occluding is a hepatitis C virus entry factor
required for infection of mouse cells. Nature 2009;457:882-6. (Abstract)
Pöhlmann S, Zhang J , Baribaud F, et al. Hepatitis C virus glycoproteins interact with DC-SIGN
and DC-SIGNR. J Virol 2003;77:4070-80. (Abstract)
Poynard T, Bedossa P, Opolon P et al. Natural history of liver fibrosis progression in patients
with chronic hepatitis C. Lancet 1997;349:825-33. (Abstract)
Prince AM, Huima-Byron T, Parker TS, et al. Visualization of hepatitis C virions and putative
defective interfering particles isolated from low-density lipoproteins. J Viral Hepat
1996;3:11-7. (Abstract)
Qiao M, Ashok M, Bernard KA, et al. Induction of sterilizing immunity against West Nile virus
(WNV) by immunization with WNV-like particles produced in insect cells. J Infect Dis
2004;190:2104-8. (Abstract)
Roussel J, Pillez A, Montpellier C, et al. Characterization of the expression of the hepatitis C
virus F protein. J Gen Virol 2003;84:1751-9. (Abstract)
Sakai A, Claire MS, Faulk K, et al. The p7 polypeptide of hepatitis C virus is critical for infectivity
and contains functionally important genotype-specific sequences. Proc Nat. Acad Sci
USA 2003;100:11646-51. (Abstract)
Santolini E, Migliaccio G, La Monica N. Biosynthesis and biochemical properties of the hepatitis
C virus core protein. J Virol 1994;68:3631-41. (Abstract)
Santolini E, Pacini L, Fipaldini C, et al. The NS2 protein of hepatitis C virus is a transmembrane
polypeptide. J Virol 1995;69:7461-71. (Abstract)
Scarselli E, Ansuini H, Cerino R, et al. The human scavenger receptor class B type I is a novel
candidate receptor for the hepatitis C virus. EMBO J 2002;21:5017-25. (Abstract)
Schmidt-Mende J, Bieck E, Hugle T, et al. Determinants for membrane association of the
hepatitis C virus RNA-dependent RNA polymerase. J Biol Chem 2001;276:44052-63.
(Abstract)
Shavinskaya A, Boulant S, Penin F, et al. The lipid droplet binding domain of hepatitis C virus
core protein is a major determinant for efficient virus assembly. J Biol Chem
2007;282:37158-69. (Abstract)
Shimizu YK, Feinstone SM, Kohara M, et al. Hepatitis C virus: detection of intracellular virus
particles by electron microscopy. Hepatology 1996a;23:205-9. (Abstract)
Shimizu YK, Igarashi H, Kiyohara T, et al. A hyperimmune serum against a synthetic peptide
corresponding to the hypervariable region 1 of hepatitis C virus can prevent viral
infection in cell cultures. Virology 1996b;223:409-12. (Abstract)
Simmonds P. The origin and evolution of hepatitis viruses in humans. J Gen Virol 2001, 82:693-712. Simmonds P. Genetic diversity and evolution of hepatitis C virus - 15 years on. J
Gen Virol 2004;85:3173-88. (Abstract)
Simmonds P, Bukh J, Combet C, et al. Consensus proposals for a unified system of
nomenclature of hepatitis C virus genotypes. Hepatology 2005;42:962-73. (Abstract)
Simons JN, Leary TP, Dawson GJ, et al. Isolation of novel virus-like sequences associated with
human hepatitis. Nat Med 1995;1:564-9. (Abstract)
Sizova DV, Kolupaeva VG, Pestova TV, et al. Specific interaction of eukaryotic translation
initiation factor 3 with the 5' nontranslated regions of hepatitis C virus and classical
swine fever virus RNAs. J Virol 1998;72:4775-82. (Abstract)
Song Y, Friebe P, Tzima E, et al. The hepatitis C virus RNA 3'-untranslated region strongly
enhances translation directed by the internal ribosome entry site. J Virol
2006;80:11579-88. (Abstract)
Spahn CM, Kieft JS, Grassucci RA, et al. Hepatitis C virus IRES RNA-induced changes in the
conformation of the 40s ribosomal subunit. Science 2001;291:1959-62. (Abstract)
Takahashi K, Kishimoto S, Yoshizawa H, et al. p26 protein and 33-nm particle associated with
nucleocapsid of hepatitis C virus recovered from the circulation of infected hosts.
Virology 1992;191:431-4. (Abstract)
Tanaka T, Kato N, Cho MJ, et al. A novel sequence found at the 3' terminus of hepatitis C virus
genome. Biochem Biophys Res 1995;215:744-9. (Abstract)
Tanaka T, Kato N, Cho MJ, et al. Structure of the 3' terminus of the hepatitis C virus genome. J
Virol 1996;70:3307-12. (Abstract)

Book on hepatitis from page 87 to 95

Book on hepatitis from page 86 to 95

86  Hepatology 2012
Taxonomy and genotypes
HCV is a small-enveloped virus with one single-stranded positive-sense RNA
molecule of approximately 9.6 kb. It is a member of the Flaviviridae family. This
viral family contains three genera, flavivirus, pestivirus, and hepacivirus. To date,
only two members of the hepacivirus genus have been identified, HCV and GB
virus B (GBV-B), a virus that had been initially detected together with the then-unclassified virus GB virus A (GBV-A) in a surgeon with active hepatitis (Thiel
2005, Ohba 1996, Simons 1995). However, the natural hosts for GBV-B and GBV-C  seem to be monkeys of the Saguinus  species (tamarins). Analyses of viral
sequences and phylogenetic comparisons support HCV’s membership in a distinct
genus from flavivirus or pestivirus (Choo 1991). The error-prone RNA polymerase
of HCV together with the high replication rate of the virus is responsible for the
large interpatient genetic diversity of HCV strains. Moreover, the extent of viral
diversification of HCV strains within a single HCV-positive individual increases
significantly over time, resulting in the development of quasispecies (Bukh 1995).
Comparisons of HCV nucleotide sequences derived from individuals from
different geographical regions revealed the presence of six major HCV genotypes
with a large number of subtypes within each genotype (Simmonds 2004, Simmonds
2005). Sequence divergence of genotypes and subtypes is 20% and 30%,
respectively. HCV strains belonging to the major genotypes 1, 2, 4, and 5 are found
in sub-Saharan Africa whereas genotypes 3 and 6 are detected with extremely high
diversity in South East Asia. This suggests that these geographical areas could be
the origin of the different HCV genotypes. The emergence of different HCV
genotypes in North America and Europe and other non-tropical countries appears to
represent more recent epidemics introduced from the countries of the original HCV
endemics (Simmonds 2001, Ndjomou 2003). Besides epidemiological aspects,
determination of the HCV genotype plays an important role for the initiation of anti-HCV treatment since the response of different genotypes varies significantly with
regard to specific antiviral drug regimens, e.g., genotype 1 is most resistant to the
current therapy of the combination of pegylated interferon α and ribavirin (Manns
2001, Fried 2002).
Viral structure
Structural analyses of HCV virions are very limited since the virus is difficult to
cultivate in cell culture systems, a prerequisite for yielding sufficient virions for
electron microscopy. Moreover, serum-derived virus particles are associated with
serum low-density lipoproteins (Thomssen 1992), which makes it difficult to isolate
virions from serum/plasma of infected subjects by centrifugation. Visualization of
HCV virus-like particles via electron microscopy succeeded only rarely (Kaito
1994, Shimizu 1996a, Prince 1996) and it was a point of controversy if the detected
structures really were HCV virions. Nevertheless, these studies suggest that HCV
has a diameter of 55-65 nm confirming size prediction of the NANBH agent by
ultra-filtration (Bradley 1985). Various forms of HCV virions appear to exist in the
blood of infected individuals: virions bound to very low density lipoproteins
(VLDL), virions bound to low density lipoproteins (LDL), virions complexed with
immunoglobulins, and free circulating virions (Bradley 1991, Thomssen 1992,
HCV Virology  87
Thomssen 1993, Agnello 1999, Andre 2002). The reasons for the close association
of a major portion of circulating virions with LDL and VLDL remain unexplained.
One possible explanation is that HCV theoretically enters hepatocytes via the LDL
receptor (Agnello 1999, Nahmias 2006). Moreover, it is speculated that the
association with LDL and/or VLDL protects the virus against neutralization by
HCV-specific antibodies.
The design and optimization of subgenomic and genomic HCV replicons in the
human hepatoma cell line Huh7 offered for the first time the possibility to
investigate HCV RNA replication in a standardized manner (Lohmann 1999, Ikeda
2002, Blight 2002). However, despite the high level of HCV gene expression, no
infectious viral particles are actually produced. Therefore, it cannot be used for
structural analysis of free virions.
Infectious HCV particles have been achieved in cell culture by using recombinant
systems (Heller 2005, Lindenbach 2005, Wakita 2005, Zhong 2005, Yu 2007).
However, even in these in vitro systems the limited production of viral particles
prevents 3D structural analysis (Yu 2007). It was also shown by cryoelectron
microscopy (cryoEM) and negative-stain transmission electron microscopy that
HCV virions isolated from cell culture have a spherical shape with a diameter of
approximately 50 to 55 nm (Heller 2005, Wakita 2005, Yu 2007) confirming earlier
results that measured the size of putative native HCV particles from the serum of
infected individuals (Prince 1996). The outer surface of the viral envelope seems to
be smooth. Size and morphology are therefore very similar to other members of the
Flaviviridae family such as the dengue virus and the West Nile virus (Yu 2007).
Modifying a baculovirus system (Jeong 2004, Qiao 2004) the same authors were
able to produce large quantities of HCV-like particles (HCV-LP) in insect cells (Yu
2007). Analysing the HCV-LPs by cryoEM it was demonstrated that the HCV E1
protein is present in spikes located on the outer surface of the LPs.
Using 3D modeling of the HCV-LPs together with genomic comparison of HCV
and well-characterized flaviviruses it is assumed that 90 copies of a block of two
heterodimers of HCV proteins E1 and E2 form the outer layer of the virions with a
diameter of approximately 50 nm (Yu 2007). This outer layer surrounds the lipid
bilayer that contains the viral nucleocapsid consisting of several molecules of the
HCV core (C) protein. An inner spherical structure with a diameter of
approximately 30-35 nm has been observed (Wakita 2005) suggesting the
nucleocapsid that harbours the viral genome (Takahashi 1992).
Genome organization
The genome of the hepatitis C virus consists of one 9.6 kb single-stranded RNA
molecule with positive polarity. Similar to other positive-strand RNA viruses, the
genomic RNA of hepatitis C virus serves as messenger RNA (mRNA) for the
translation of viral proteins. The linear molecule contains a single open reading
frame (ORF) coding for a precursor polyprotein of approximately 3000 amino acid
residues (Figure 1). During viral replication the polyprotein is cleaved by viral as
well as host enzymes into three structural proteins (core, E1, E2) and seven non-structural proteins (p7, NS2, NS3, NS4A, NS4B, NS5A, NS5B). An additional
protein (termed F [frameshift] or ARF [alternate reading frame]) is predicted as a
result of ribosomal frameshifting during translation within the core region of the
88  Hepatology 2012
genomic RNA (Xu 2001, Walewski 2001, Varaklioti 2002, Branch 2005). Detection
of anti-F protein antibodies in the serum of HCV-positive subjects indicates that the
protein is expressed during infection in vivo (Walewski 2001, Komurian-Pradel
2004).
The structural genes encoding the viral  core protein and the viral envelope
proteins E1 and E2 are located at the 5’ terminus of the open reading frame
followed downstream by the coding regions for the non-structural proteins p7, NS2,
NS3, NS4A, NS4B, NS5A, and NS5B (Figure 1). The structural proteins are
essential components of the HCV virions, whereas the non-structural proteins are
not associated with virions but are involved in RNA replication and virion
morphogenesis.
The ORF is flanked by 5’ and 3’ nontranslated regions (NTR; also called
untranslated regions, UTR or noncoding regions, NCR) containing nucleotide
sequences relevant for the regulation of viral replication. Both NTRs harbour highly
conserved regions compared to the protein encoding regions of the HCV genome.
The high grade of conservation of the NTRs makes them candidates i) for improved
molecular diagnostics, ii) as targets for antiviral therapeutics, and iii) as targets for
an anti-HCV vaccine.
Figure 1. Genome organization and polyprotein processing. A) Nucleotide positions
correspond to the HCV strain H77 genotype 1a, accession number NC_004102.
nt, nucleotide; NTR, nontranslated region. B) Cleavage sites within the HCV precursor
polyprotein for the cellular signal peptidase the signal peptide peptidase (SPP) and the viral
proteases NS2-NS3 and NS3-NS4A, respectively.
The 5’NTR is approximately 341 nucleotides long with a complex secondary
structure of four distinct domains (I-IV) (Fukushi 1994, Honda 1999). The first 125
nucleotides of the 5’NTR spanning domains I and  II have been shown to be
essential for viral RNA replication (Friebe 2001, Kim 2002). Domains II-IV build
an internal ribosome entry side (IRES) involved in ribosome binding and
subsequent cap-independent initiation of translation (Tsukiyama-Kohara 1992,
Wang 1993).
The 3’NTR consists of three functionally distinct regions: a variable region, a
poly U/UC tract of variable length, and the highly conserved X tail at the 3’
terminus of the HCV genome (Tanaka 1995, Kolykhalov 1996, Blight 1997). The
HCV Virology  89
variable region of approximately 40 nucleotides is not essential for RNA
replication. However, deletion of this sequence led to significantly decreased
replication efficiency (Yanagi 1999, Friebe 2002). The length of the poly U/UC
region varies in different HCV strains ranging from 30 to 80 nucleotides
(Kolykhalov 1996). The minimal length of that region for active RNA replication
has been reported to be 26 homouridine nucleotides in cell culture (Friebe 2002).
The highly conserved 98-nucleotide X tail consists of three stem-loops (SL1-SL3)
(Tanaka 1996, Ito 1997, Blight 1997) and deletions or nucleotide substitutions
within that region are most often lethal (Yanagi 1999, Kolykhalov 2000, Friebe
2002, Yi 2003). Another so-called “kissing-loop” interaction of the 3’X tail SL2 and
a complementary portion of the NS5B encoding region has been described (Friebe
2005). This interaction induces a tertiary RNA structure of the HCV genome that is
essential for HCV replication in cell culture systems (Friebe 2005, You 2008).
Finally, both NTRs appear to work together in a long-range RNA-RNA interaction
possibly resulting in temporary genome circularization (Song 2006).
Genes and proteins
As described above, translation of the HCV polyprotein is initiated through
involvement of some domains in NTRs of the genomic HCV RNA. The resulting
polyprotein consists of ten proteins that are co-translationally or post-translationally
cleaved from the polyprotein. The N-terminal proteins C, E1, E2, and p7 are
processed by a cellular signal peptidase (SP) (Hijikata 1991). The resulting
immature core protein still contains the E1 signal sequence at its C terminus.
Subsequent cleavage of this sequence by a signal peptide peptidase (SPP) leads to
the mature core protein (McLauchlan 2002). The non-structural proteins NS2 to
NS5B of the HCV polyprotein are processed by two virus-encoded proteases (NS2-NS3 and NS3) with the NS2-NS3 cysteine protease cleaving at the junction of NS2-NS3 (Santolini 1995) and the NS3 serine protease cleaving the remaining functional
proteins (Bartenschlager 1993, Eckart 1993, Grakoui 1993a, Tomei 1993).
The positions of viral nucleotide and amino acid residues correspond to the HCV
strain H77 genotype 1a, accession number NC_004102. Some parameters
characterizing HCV proteins are summarised in Table 1.
Core. The core-encoding sequence starts at codon AUG at nt position 342 of the
H77 genome, the start codon for translation of the entire HCV polyprotein. During
translation the polyprotein is transferred to the endoplasmic reticulum (ER) where
the core protein (aa 191) is excised by a cellular signal peptidase (SP). The C
terminus of the resulting core precursor still contains the signal sequence for ER
membrane translocation of the E1 ectodomain (aa 174-191). This protein region is
further processed by the cellular intramembrane signal peptide peptidase (SPP)
leading to removal of the E1 signal peptide sequence (Hüssy 1996, McLauchlan
2002, Weihofen 2002).
The multifunctional core protein has a molecular weight of 21 kilodalton (kd). In
vivo, the mature core molecules are believed to form homo-multimers located
mainly at the ER membrane (Matsumoto 1996). They have a structural function
since they form the viral capsid that contains the HCV genome. In addition, the core
protein has regulatory functions including particle assembly, viral RNA binding,
and regulation of RNA translation (Ait-Goughoulte 2006, Santolini 1994).
90  Hepatology 2012
Moreover, protein expression analyses indicate that the core protein may be
involved in many other cellular reactions such as cell signalling, apoptosis, lipid
metabolism, and carcinogenesis (Tellinghuisen 2002). However, these preliminary
findings need to be analysed further.
Table 1. Overview of the size of HCV proteins.*
Protein  No. of aa  aa position
in ref. seq.
MW of protein
Core immature  191  1-191  23 kd
Core mature  174  1-174  21 kd
F-protein or ARF-protein  126-161    ~ 16-17 kd
E1  192  192-383  35 kd
E2  363  384-746  70 kd
p7  63  747-809  7 kd
NS2  217  810-1026  21 kd
NS3  631  1027-1657  70 kd
NS4A  54  1658-1711  4 kd
NS4B  261  1712-1972  27 kd
NS5A  448  1973-2420  56 kd
NS5B  591  2421-3011  66 kd
* aa, amino acid; MW, molecular weight; kd, kilodalton; ref. seq., reference sequence (HCV
strain H77; accession number NC_004102).
E1 and E2. Downstream of the core coding region of the HCV RNA genome two
envelope glycoproteins are encoded, E1 (gp35, aa 192) and E2 (gp70, aa 363).
During translation at the ER both proteins are cleaved from the precursor
polyprotein by a cellular SP. Inside the lumen of the ER both polypeptides
experience N-linked glycosylation post-translationally (Duvet 2002). Both
glycoproteins E1 and E2 harbour 5 and 11 putative N-glycosylation sites,
respectively.
E1 and E2 are type I transmembrane proteins with a large hydrophilic ectodomain
of approximately 160 and 334 aa and a short transmembrane domain (TMD) of 30
aa. The TMD are responsible for the anchoring of the envelope proteins in the
membrane of the ER and ER retention (Cocquerel 1998, Duvet 1998, Cocquerel
1999, Cocquerel 2001). Moreover, the same domains have been reported to
contribute to the formation of E1-E2 heterodimers (Op de Beeck 2000). The E1-E2
complex is involved in adsorption of the virus to its putative receptors tetraspanin
CD81 and low-density lipoprotein receptor inducing fusion of the viral envelope
with the host cell plasma membrane (Agnello 1999, Flint 1999, Wunschmann
2000). However, the precise mechanism of host cell entry is still not understood
completely. Several other host factors have been identified to be involved in viral
entry. These candidates include the scavenger receptor B type I (Scarselli 2002,
Kapadia 2007), the tight junction proteins claudin-1 (Evans 2007) and occludin
(Ploss 2009), the C-type lectins L-SIGN and DC-SIGN (Gardner 2003, Lozach
2003, Pöhlmann 2003) and heparan sulfate (Barth 2003).
Two hypervariable regions have been identified within the coding region of E2.
These regions termed hypervariable region 1 (HVR1) and 2 (HVR2) differ by up to
80% in their amino acid sequence (Weiner 1991, Kato 2001). The first 27 aa of the
E2 ectodomain represent HVR1, while the HVR2 is formed by a stretch of seven
HCV Virology  91
amino acids (position 91-97). The high variability of the HVRs reflects exposure of
these domains to HCV-specific antibodies. In fact, E2-HVR1 has been shown to be
the most important target for neutralizing antibodies (Farci 1996, Shimizu 1996b).
However, the combination of the mutation of the viral genome with the selective
pressure of the humoral immune response leads to viral escape via epitope
alterations. This makes the development of vaccines that induce neutralizing
antibodies challenging.
The p7 protein. The small p7 protein (63 aa) is located between the E2 and NS2
regions of the polyprotein precursor. During translation the cellular SP cleaves the
E2-p7 as well as the p7-NS2 junction. The functional p7 is a membrane protein
localised in the endoplasmic reticulum where it forms an ion channel (Haqshenas
2007, Pavlovic 2003, Griffin 2003). The p7 protein is not essential for RNA
replication since replicons lacking the p7 gene replicate efficiently (Lohmann 1999,
Blight 2000), however, it has been suggested that p7 plays an essential role for the
formation of infectious virions (Sakai 2003, Haqshenas 2007).
NS2. The non-structural protein 2 (p21, 217 aa) together with the N-terminal
portion of the NS3 protein form the NS2-3 cysteine protease which catalyses
cleavage of the polyprotein precursor between NS2 and NS3 (Grakoui 1993b,
Santolini 1995). The N-terminus of the functional NS2 arises from the cleavage of
the p7-NS2 junction by the cellular SP. Moreover, after cleavage from the NS3 the
protease domain of NS2 seems to play an essential role in the early stage of virion
morphogenesis (Jones 2007).
NS3. The non-structural protein 3 (p70, 631 aa) is cleaved at its N terminus by the
NS2-NS3 protease. The N terminus (189 aa) of the NS3 protein has a serine
protease activity. However, in order to develop full activity of the protease the NS3
protease domain requires a portion of NS4A (Faila 1994, Bartenschlager 1995, Lin
1995, Tanji 1995, Tomei 1996). NS3 together with the NS4A cofactor are
responsible for cleavage of the remaining downstream cleavages of the HCV
polyprotein precursor. Since the NS3 protease function is essential for viral
infectivity it is a promising target in the design of antiviral treatments.
The C-terminal portion of NS3 (442 aa) has ATPase/helicase activity, i.e., it
catalyses the binding and unwinding of the viral RNA genome during viral
replication (Jin 1995, Kim 1995). However, recent findings indicate that other non-structural HCV proteins such as the viral polymerase NS5B may interact
functionally with the NS3 helicase (Jennings 2008). These interactions need to be
investigated further in order to better understand the mechanisms of HCV
replication.
NS4A. The HCV non-structural protein 4A (p4) is a 54 amino acid polypeptide
that acts as a cofactor of the NS3 serine protease (Faila 1994, Bartenschlager 1995,
Lin 1995, Tanji 1995, Tomei 1996). Moreover, this small protein is involved in the
targeting of NS3 to the endoplasmic reticulum resulting in a significant increase of
NS3 stability (Wölk 2000).
NS4B. The NS4B (p27) consists of 217 amino acids. It is an integral membrane
protein localized in the endoplasmic reticulum. The N-terminal domain of the NS4B
has an amphipathic character that targets the protein to the ER. This domain is
crucial in HCV replication (Elazar 2004, Gretton 2005) and therefore an interesting
target for the development of anti-HCV therapeutics or vaccines. In addition, a
nucleotide-binding motif (aa 129-134) has been identified (Einav 2004). Although
92  Hepatology 2012
the function of NS4B is still unknown, it has been demonstrated that the protein
induces a membranous web that may serve as a platform for HCV RNA replication
(Egger 2002).
NS5A. The NS5A protein (p56; 458 aa) is a membrane-associated phosphoprotein
that appears to have multiple functions in viral replication. It is phosphorylated by
different cellular protein kinases indicating an essential but still not understood role
of NS5A in the HCV replication cycle. In addition, NS5A has been found to be
associated with several other cellular proteins (MacDonald 2004) making it difficult
to determine the exact functions of the protein. One important property of NS5A is
that it contains a domain of 40 amino acids, the so-called IFN-a sensitivity-determining region (ISDR) that plays a significant role in the response to IFN-a-based therapy (Enomoto 1995, Enomoto 1996). An increasing number of mutations
within the ISDR showed positive correlation with sustained virological response to
IFN-a-based treatment.
NS5B.  The non-structural protein 5B (p66; 591 aa) represents the RNA-dependent RNA polymerase of HCV (Behrens 1996). The hydrophobic domain (21
aa) at the C terminus of NS5B inserts into the membrane of the endoplasmic
reticulum, while the active sites of the polymerase are located in the cytoplasm
(Schmidt-Mende 2001).
The cytosolic domains of the viral enzyme form the typical polymerase right-handed structure with “palm”, “fingers”, and “thumb” subdomains (Ago 1999,
Bressanelli 1999, Lesburg 1999). In contrast to mammalian DNA and RNA
polymerases the fingers and thumb subdomains are connected resulting in a fully
enclosed active site for nucleotide triphosphate binding. This unique structure
makes the HCV NS5B polymerase an attractive target for the development of
antiviral drugs.
Using the genomic HCV RNA as a template, the NS5B promotes the synthesis of
minus-stranded RNA that then serves as a template for the synthesis of genomic
positive-stranded RNA by the polymerase.
Similar to other RNA-dependent polymerases, NS5B is an error-prone enzyme
that incorporates wrong ribonucleotides at a  rate of approximately 10
-3
per
nucleotide per generation. Unlike cellular polymerases, the viral NS5B lacks a
proof-reading mechanism leading to the conservation of misincorporated
ribonucleotides. These enzyme properties together with the high rate of viral
replication promote a pronounced intra-patient as well as inter-patient HCV
evolution.
F protein, ARFP. In addition to the ten proteins derived from the long HCV
ORF, the F (frameshift) or ARF (alternate reading frame) or core+1 protein has
been reported (Walewski 2001, Xu 2001, Varaklioti 2002). As the designations
indicate the ARFP is the result of a -2/+1 ribosomal frameshift between codons 8
and 11 of the core protein-encoding region. The ARFP length varies from 126 to
161 amino acids depending on the corresponding genotype. In vitro studies have
shown that ARFP is a short-lived protein located in the cytoplasm (Roussel 2003)
primarily associated with the endoplasmic reticulum (Xu 2003). Detection of anti-F
protein antibodies in the serum of HCV-positive subjects indicates that the protein is
expressed during infection in vivo  (Walewski 2001, Komurian-Pradel 2004).
However, the functions of ARFP in the viral life cycle are still unknown and remain
to be elucidated.
HCV Virology  93
Viral lifecycle
Due to the absence of a small animal model system and efficient in vitro HCV
replication systems it has been difficult to investigate the viral life cycle of HCV.
The recent development of such systems has offered the opportunity to analyse in
detail the different steps of viral replication.
Figure 2. Current model of the HCV lifecycle. Designations of cellular components are in red.
For a detailed illustration of viral translation and RNA replication, see Pawlotsky 2007.
Abbreviations: HCV +ssRNA, single stranded genomic HCV RNA with positive polarity; rough
ER, rough endoplasmic reticulum; PM, plasma membrane. For other abbreviations see text.
Adsorption and viral entry
The most likely candidate as receptor for HCV is the tetraspanin CD81 (Pileri
1998). CD81 is an ubiquitous 25 kd molecule expressed on the surface of a large
variety of cells including hepatocytes and PBMCs. Experimental binding of anti-CD81 antibodies to CD81 were reported to inhibit HCV entry into Huh7 cells and
primary human hepatocytes (Hsu 2003, Bartosch 2003a, Cormier 2004, McKeating
2004, Zhang 2004, Lindenbach 2005, Wakita 2005). Moreover, gene silencing of
CD81 using specific siRNA molecules confirmed the relevance of CD81 in viral
entry (Bartosch 2003b,  Cormier 2004,  Zhang 2004,  Akazawa 2007). Finally,
expression of CD81 in cell lines lacking CD81 made them permissive for HCV
entry (Zhang 2004, Lavillette 2005, Akazawa 2007). However, more recent studies
have shown that CD81 alone is not sufficient for HCV viral entry and that co-factors such as scavenger receptor B type I (SR-BI) are needed (Bartosch 2003b,
Hsu 2003,  Scarselli 2002, Kapadia 2007). Moreover, it appears that CD81 is
involved in a post-HCV binding step (Cormier 2004, Koutsoudakis 2006, Bertaud
2006). These findings together with the identification of other host factors involved
94  Hepatology 2012
in HCV cell entry generate the current model for the early steps of HCV infection
(Helle 2008).
Adsorption of HCV to its target cell is the first step of viral entry. Binding is
possibly initiated by the interaction of the HCV E2 envelope glycoprotein and the
glycosaminglycan heparan sulfate on the surface of host cells (Germi 2002, Barth
2003, Basu 2004, Heo 2004). Moreover, it is assumed that HCV initiates hepatocyte
infection via LDL receptor binding (Agnello 1999, Monazahian 1999, Wünschmann
2000, Nahmias 2006, Molina 2007). This process may be mediated by VLDL or
LDL and is reported to be associated with HCV virions in human sera (Bradley
1991,  Thomssen 1992,  Thomssen  1993). After initial binding the HCV E2
glycoprotein interacts with the SR-BI in cell culture (Scarselli 2002). SR-BI is a
protein expressed on the surface of the majority of mammalian cells. It acts as a
receptor for LDL as well as HDL (Acton 1994, Acton 1996) emphasizing the role of
these compounds for HCV infectivity. Alternative splicing of the SR-BI transcript
leads to the expression of a second isoform of the receptor SR-BII (Webb 1998),
which also may be involved in HCV entry into target cells (Grove 2007). As is the
case for all steps of viral entry the exact mechanism of the HCVE2/SR-BI
interaction remains unknown. In some studies it has been reported that HCV
binding to SR-BI is a prerequisite for the concomitant or subsequent interaction of
the virus with CD81 (Kapadia 2007, Zeisel 2007). The multi-step procedure of
HCV cell entry was shown to be even more complex since a cellular factor termed
claudin-1 (CLDN1) has been newly identified as involved in this process (Evans
2007). CLDN1 is an integral membrane protein that forms a backbone of tight
junctions and is highly expressed in the liver (Furuse 1998). Inhibition assays reveal
that CLDN1 involvement occurs downstream of the HCV-CD81 interaction (Evans
2007). Recent findings suggest that CLDN1 could also act as a compound enabling
cell-to-cell transfer of hepatitis C virus independently of CD81 (Timpe 2007).
Furthermore, it was reported that two other members of the claudin family claudin-6
and claudin-9 may play a role in HCV infection (Zheng 2007, Meertens 2008). The
fact that some human cell lines were not susceptible to HCV infection despite
expressing SR-BI, CD81, and CLDN1 indicates that other cellular factors are
involved in viral entry (Evans 2007). Very recently, a cellular four-transmembrane
domain protein named occludin (OCLN) was identified to represent an additional
cellular factor essential for the susceptibility of cells to HCV infection (Liu 2009,
Ploss 2009). Similar to claudin-1, OCLN is a component of the tight junctions in
hepatocytes. All tested cells expressing SR-BI, CD81, CLDN1, and OCLN were
susceptible to HCV. Although the precise mechanism of HCV uptake in hepatocytes
is still not clarified, these four proteins may represent the complete set of host cell
factors necessary for cell-free HCV entry.
After the complex procedure of binding to the different host membrane factors
HCV enters the cell in a pH-dependent manner indicating that the virus is
internalized via clathrin-mediated endocytosis (Bartosch 2003b,  Hsu 2003,
Blanchard 2006, Codran 2006). The acidic environment within the endosomes is
assumed to trigger HCV E1-E2 glycoprotein-mediated fusion of the viral envelope
with the endosome membrane (Blanchard 2006, Meertens 2006, Lavillette 2007).
In summary, HCV adsorption and viral entry into the target cell is a very complex
procedure that is not yet fully understood. Despite having identified several host
HCV Virology  95
factors that probably interact with the viral glycoproteins, the precise mechanisms
of interaction need to continue to be investigated.
Besides the infection of cells through cell-free HCV it has been documented that
HCV can also spread via cell-to-cell transmission (Valli 2006, Valli 2007). This
transmission path may differ significantly with regard to the cellular factors needed
for HCV entry into cells. CD81 is dispensable for cell-to-cell transmission in
cultivated hepatoma cells (Witteveldt 2009). These findings require further
investigation in order to analyze the process of cell-to-cell transmission of HCV
both in vitro and in vivo. Antiviral treatment strategies must account for the cellular
pathways of both cell-free virus and HCV transmitted via cell-to-cell contact.
Translation and posttranslational processes
As a result of the fusion of the viral envelope and the endosomic membrane, the
genomic HCV RNA is released into the cytoplasm of the cell. As described above,
the viral genomic RNA possesses a nontranslated region (NTR) at each terminus.
The 5’NTR consists of four distinct domains, I-IV. Domains II-IV form an internal
ribosome entry side (IRES) involved in ribosome-binding and subsequent cap-independent initiation of translation (Fukushi 1994,  Honda 1999,  Tsukiyama-Kohara 1992, Wang 1993). The HCV-IRES binds to the 40S ribosomal subunit
complexed with eukaryotic initiation factors 2 and 3 (eIF2 and eIF3), GTP, and the
initiator tRNA resulting in the 48S preinitiation complex (Spahn 2001, Otto 2002,
Sizova 1998, reviewed in Hellen 1999). Subsequently, the 60S ribosomal subunit
associates with that complex leading to the formation of the translational active
complex for HCV polyprotein synthesis at the endoplasmic reticulum. HCV RNA
contains a large ORF encoding a polyprotein precursor. Posttranslational cleavages
lead to 10 functional viral proteins Core, E1, E2, p7, NS2-NS5B. The viral F protein
(or ARF protein) originates from a ribosomal frameshift within the first codons of
the core-encoding genome region (Walewski 2001, Xu 2001, Varaklioti 2002).
Besides several other cellular factors that have been reported to be involved in HCV
RNA translation, various viral proteins and genome regions have been shown to
enhance or inhibit viral protein synthesis (Zhang 2002, Kato 2002, Wang 2005, Kou
2006, Bradrick 2006, Song 2006).
The precursor polyprotein is processed by at least four distinct peptidases. The
cellular signal peptidase (SP) cleaves the N-terminal viral proteins immature core
protein, E1, E2, and p7 (Hijikata 1991), while the cellular signal peptide peptidase
(SPP) is responsible for the cleavage of the E1 signal sequence from the C-terminus
of the immature core protein, resulting in the mature form of the core (McLauchlan
2002). The E1 and E2 proteins remain within the lumen of the ER where they are
subsequently N-glycosylated with E1 having 5 and E2 harbouring 11 putative N-glycosylation sites (Duvet 2002).
In addition to the two cellular peptidases HCV encodes two viral enzymes
responsible for cleavage of the non-structural proteins NS2 to NS5B within the
HCV polyprotein precursor. The zinc-dependent NS2-NS3 cysteine protease
consisting of the NS2 protein and the N-terminal portion of NS3 autocatalytically
cleaves the junction between NS2 and NS3 (Santolini 1995), whereas the NS3
serine protease cleaves the remaining functional proteins (Bartenschlager 1993,
Eckart 1993, Grakoui 1993a, Tomei 1993). However, for its peptidase activity NS3

.

Powered By Blogger

Search This Blog