Open access peer-reviewed chapter

Immune Disorders in HIV-Infected Patients Coinfected with Hepatitis C Virus

Written By

Konstantin Shmagel and Evgeniya Saidakova

Submitted: 31 January 2018 Reviewed: 27 March 2018 Published: 05 November 2018

DOI: 10.5772/intechopen.76810

From the Edited Volume

Advances in HIV and AIDS Control

Edited by Samuel Okware

Chapter metrics overview

1,326 Chapter Downloads

View Full Metrics

Abstract

In Russia, more than half of HIV-infected people are coinfected with hepatitis C. Both viruses interact with the immune system compounding the disease course. HIV infection accelerates the onset of hepatitis-mediated liver fibrosis and cirrhosis. Hepatitis C slows down the recovery of CD4+ T-lymphocytes during antiretroviral treatment and fuels the already intense chronic inflammation. In the present review, we discuss coinfection prevalence and reasons for its abundance, provide extensive coverage of the known mechanisms that give rise to the detrimental health effects in HIV/hepatitis C-coinfected patients, and report our own data on the double infection consequences in people with discordant immunologic response to treatment.

Keywords

  • HIV infection
  • hepatitis C
  • HIV/HCV coinfection
  • innate immunity
  • adaptive immunity
  • discordant immunologic response
  • highly active antiretroviral therapy

1. Introduction

More than any other infectious disease, HIV infection claims to be called “the coinfection illness” [1]. Coinfections can significantly change the illness pattern and the immune activation profile [2, 3, 4] and typically lead to the rise in morbidity and mortality [5, 6, 7]. The coinfection most often associated with HIV is hepatitis C virus (HCV) infection. This is due to the worldwide prevalence of both illnesses (there are approximately 40 million HIV-infected and about 120 million HCV-infected subjects worldwide) and the overlap in infection transmission routes [8, 9]. In Western Europe and the United States, the proportion of hepatitis C chronically infected patients among HIV-positive people is 25–30% [10], and in Eastern Europe, it is more than 50% [11]. In Russia, the increase in injection drug use has led to a significant rise in the prevalence of HIV/HCV coinfection. Its level among drug users reaches 93% [12]. The problem is complicated by the rise in non-AIDS-defining morbidity and mortality in HIV/HCV-coinfected subjects [13, 14].

There is considerable evidence that HIV infection adversely affects the course of a hepatitis C infection. When HIV/HCV coinfection is compared with HCV monoinfection, a more rapid fibrosis [15, 16] and liver cirrhosis [16, 17] are observed. Coinfected subjects also have an increased risk of hepatocellular carcinoma, which occurs at an earlier age and in a shorter time interval after HCV infection [18, 19, 20]. It was found that HIV/HCV-coinfected patients compared to HCV-monoinfected patients were more resistant to interferon therapy. In HIV-seronegative subjects infected with HCV genotype 1, 50–80% can achieve a complete recovery. However, in HIV-seropositive individuals coinfected with the same HCV type, interferon therapy is successful only in 20–35% of patients [21]. This accounts for the increased mortality rate among HIV-/HCV-coinfected patients when compared with HIV-monoinfected patients [22, 23].

Less is known about the effect of hepatitis C on the natural course of HIV infection. Among the negative influences, one can point to direct viral effects, hepatocyte destruction by immunocompetent cells, hepatic cell apoptosis, immune activation, and specific antiviral immune response alterations [24, 25, 26]. The complexity of the problem is largely due to the lack of knowledge about the biology of both HIV and HCV. It remains unknown whether the viruses interact with each other and in what ways that interaction might be expressed.

Advertisement

2. Liver fibrosis in HIV/HCV coinfection

Evidence indicates that the HCV viral load is lower in hepatitis C-monoinfected patients when compared to HIV/HCV-coinfected patients [27, 28]. Similar results were obtained when estimating the viral load in hepatic tissue [29]. In addition, multiyear cohort studies state that in patients with hepatitis C the HCV RNA blood level significantly increases after exposure to HIV [30, 31]. HCV replication enhancement in coinfection is attributed to both the development of immunodeficiency and the direct impact of HIV. While attempting to determine the mechanism(s) of these effects, it was shown that inactivated HIV or its component (gp120) can intensify viral replication in HCV-infected hepatoma cells in vitro [32]. This effect of HIV was shown to be due to transforming growth factor-beta 1 (TGF-β1) synthesis (antibodies against the cytokine blocked the HCV replication enhancement). Researchers also noted that HIV engages CCR5 or CXCR4 co-receptors for the related intracellular signal induction. Those data are significant not only for demonstrating the ability of HIV to increase HCV replication (with a monoinfection of hepatitis C, viral load is usually not associated with the disease severity) but also for illuminating the possible pathogenetic mechanism of fibrosis in HIV/HCV coinfection.

In many studies, HIV/HCV-coinfected patients demonstrated an inverse correlation between the CD4+ T-cell count and the HCV viral load [33, 34, 35, 36, 37]. Moreover, in those patients, low CD4+ T-lymphocyte quantity was used as a liver fibrosis predictor [34, 38, 39]. This suggests a negative impact of HIV infection on the course of hepatitis C through the development of CD4+ T-cell deficiency. It should be noted that a decrease in the CD4+ T-lymphocyte count is also found in those monoinfected with HCV. Indeed, the majority of HIV-seronegative subjects with liver cirrhosis have a reduced CD4+ T-cell count [40, 41]. Most researchers state that HIV infection, accompanied by a profound depletion in the CD4+ T-lymphocyte pool, is a prominent mediator of the accelerated liver fibrosis development in HIV-/HCV-coinfected people [42, 43, 44]. Based on those results and the opinions of leading specialists, the European AIDS Clinical Society recommends the early administration of highly active antiretroviral therapy (ART) to HCV-coinfected patients not only to optimize their hepatitis C management but also to slow down the development of fibrosis [45].

The main cellular element involved in the process of hepatic tissue fibrosis is the liver stellate cell (LSC) [46, 47, 48, 49] located around the sinuses and usually not showing high activity until the organ is damaged [50, 51]. However, various destructive processes in the liver are accompanied by the reaction of hepatocytes, endotheliocytes, and Kupffer cells to produce various humoral factors [52]. Of those, TGF-β1 [53, 54] and PDGF (platelet-derived growth factor) have the most pronounced effect on LSC [52, 55]. Both cytokines induce LSC activation and differentiation into myofibroblast-like cells, which actively synthesize extracellular matrix proteins [56]. However, it should be noted that TGF-β1 and PDGF blood concentrations (as opposed to analyzing the hyaluronic acid or hepatocytes’ growth factor content) have no high diagnostic value for the detection of fibrosis [57, 58, 59, 60]. Moreover, it has recently been established that HIV influences the liver by infecting hepatocytes and liver stellate cells [61].

Advertisement

3. Anti-HCV immunity in HIV/HCV coinfection

Protection against HCV is implemented by various factors with an important role for interferons, natural killer (NK) cells, neutralizing antibodies, and T-lymphocytes. Type I interferons (IFN-α and IFN-β) and type III interferon (IFN-λ) are synthesized in response to the virus and induce interferon-stimulated gene (ISG) expression [62, 63]. In the cytosol, the pathogen’s RNA is detected by the RIG-I (retinoic acid-inducible gene I) sensors, protein kinase R, and MDA5 (melanoma differentiation-associated protein 5). The first two mediate the interferon response at the early stages of the disease, and the third one mediates at the later infection phase [64, 65]. In endosomes, the virus is primarily detected by Toll-like receptor 3 (TLR3) that also triggers the IFN production and the ISG expression [66]. In hepatocytes of HCV-infected patients, the viral RNA and ISGs’ mRNA are detected simultaneously, which confirms the connection between the cell genetic response and the presence of the pathogen [67]. The result of the activated ISG status in HCV infection leads to viral replication inhibition [68, 69]. However, prolonged ISG expression has a negative effect on the process of HCV spontaneous elimination [70, 71] and on the results of interferon and ribavirin combination therapy [72, 73].

NK cells play an important role in the pathogenesis of an HCV infection. It was found that in the healthy liver they represent the majority of the innate immune cells [74]. In the acute phase of the disease, NK cells affected by the virus are activated, produce IFN-γ, and perform cytotoxic functions [75]. In the chronic infection phase, IFN-γ and tumor necrosis factor (TNF)-α synthesis are reduced [76, 77, 78] even though the NK cell cytotoxic potential remains high [79, 80]. Since the protective effect of IFN-γ was demonstrated in HCV-infected hepatoma cells [81] and in experiments with chimpanzees given primary and repeated infections [82], it is believed that reduced IFN-γ production weakens the NK cell’s antiviral activity. Thus, in the chronic stage of HCV infection, in spite of being activated and ready to perform cytotoxic functions, the NK lymphocytes are unable to effectively resist HCV due to the failure of IFN-γ production. However, the saved killing function can produce a positive result. NK cells from HCV-infected subjects are capable of killing activated LSC by NKG2D- and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-dependent apoptosis, which allows them to be considered as active participants in liver fibrosis suppression [83].

The role of neutralizing antibodies (nAB) in protection against an HCV infection is not yet sufficiently understood. Based on known cases of spontaneous recovery before nAB emerge [84] and based on the ability of some patients with hypogammaglobulinemia to control the infection [85, 86], it could be concluded that the humoral immune response does not determine resistance to the disease. At the same time, there is evidence of a protective function for antibodies directed against HCV surface proteins. HCV envelope glycoprotein E1 and glycoprotein E2 seroconversion is usually observed a few weeks after an infection [87]. The ability of viral envelope-specific antibodies to block the infectious process was demonstrated in chimpanzees [88, 89] and in mice with genetically humanized liver [90, 91]. The emergence of nAB in the acute phase of HCV infection is accompanied by an alteration in the virus and its escape from immune control [92]. The authors also showed that despite the increased virus flexibility, high antibody titers significantly increase the chance for clearance of the infection. The acute-phase nAB titers are usually low in patients subsequently entering the chronic stage of the disease.

As was established in monkeys with induced CD4+ or CD8+ T-cell deficiency [93, 94], T-lymphocytes play an important role in the development of hepatitis C. It should be noted that the HCV-specific T-cell response usually develops 2–3 months after the infection [95, 96], although according to some authors such a “slow” reaction has little effect on the disease outcome [84]. It seems that the quality of the immune response achieved by CD4+ and CD8+ T-cells is a more important factor [97, 98]. It was found that in the acute phase of the infection, patients spontaneously clearing HCV compared to subjects in whom the disease became chronic had a more robust CD4+ T-lymphocyte response, which manifested in more active proliferation and cytokine (IFN-γ, TNF-α, and IL-2) production [99, 100, 101, 102]. Later, it was found that the expression of PD-1 (programmed cell death protein 1) and CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) inhibitory molecules was increased on the surface of CD4+ T-cells in patients chronically infected with HCV [103]. Blocking of PD-1 ligand (PD-L1/PD-L2), IL-10, and TGF-β1 in cultured lymphocytes isolated from the blood of these patients increased the virus-specific expansion of CD4+ T-lymphocytes. Neutralization of IL-10 and TGF-β1 enhanced the synthesis of IFN-γ, IL-2, and TNF-α. Further studies revealed that IL-21-producing CD4+ T-lymphocytes are lost in individuals with a chronic hepatitis C infection [104]. It has also been demonstrated that the deficiency of Th17 cells synthesizing IL-21 limits the HCV-specific CD8+ T-lymphocyte function and survival. The inability of CD4+ and CD8+ T-cells to control viral replication leads to their exhaustion. According to the authors, in chronic HCV infection, the increase in regulatory T-cell number and activity is aimed at suppressing an ineffective immune response and reducing inflammation. The other side of that process is fibrosis intensification. Thus, based on the above data, one can conclude that CD4+ T-lymphocytes are the key cells in protection against HCV. Hence, it becomes clear why a low CD4+ T-cell count is a negative predictor for liver fibrosis development in HIV-/HCV-coinfected patients.

Advertisement

4. Detrimental effects of hepatitis C on the course of HIV infection

It is more difficult to assess the effect of hepatitis C on the natural course of HIV infection. One of the parameters characterizing that effect is CD4+ T-cell count reconstitution after the administration of ART. To date, the accumulated data indicate slowing of the CD4+ T-lymphocyte restoration process in HIV-positive subjects coinfected with hepatitis C [35, 105, 106, 107, 108]. The rate of CD4+ T-cell counts increases after receiving ART was reduced sevenfold in coinfected individuals compared with HIV-monoinfected patients [35]. The authors also established an association between impaired immunity regeneration and the level of HCV replication. In another study, it was demonstrated that in hepatitis C-positive patients, ineffective ART-mediated restoration affected not only the total CD4+ T-lymphocyte numbers but also their naive subset [106]. However, it should be noted that not all researchers support the idea of the negative effect of HCV coinfection on the treatment-induced CD4+ T-cell response [109, 110]. Still, an extensive multicenter study involving 22,533 patients showed that immune regeneration during ART is slower in coinfected patients, and the lower the nadir CD4+ T-lymphocyte level, the more pronounced the effect [111]. However, as noted in the paper, the differences in the CD4+ T-cell recovery between HIV + HCV+ and HIV + HCV− subjects are canceled by early treatment administration and uninterrupted long-term ART.

HIV infection causes severe devastation in the lymphoid structures of the digestive tract. It is accompanied by intestinal epithelial barrier destruction [112, 113] and the entry of microbes and their products into the bloodstream [114]. The increase in intestinal permeability is due to the direct destructive effect of HIV on the intestinal epithelium [115] followed by the development of inflammation and tissue remodeling [116]. Another cause for the pathological changes to the epithelial barrier is the deficiency of lymphocytes producing IL-17 and IL-22 which are necessary to maintain the epithelial lining integrity [117, 118]. To date, the role of microbial translocation in the immune system activation has been well established [119, 120, 121]. In addition, it has been shown that the blood levels of lipopolysaccharide (LPS) and soluble macrophage receptor CD14 (sCD14, capable of binding LPS) in HIV-infected patients can be used to predict disease progression and mortality [122, 123, 124]. Recently, in a large cohort of non-treated HIV-infected patients, an association between LPS-dependent immune activation and intestinal damage markers in serum was demonstrated [125]. However, a relationship between the immune system activation and viral load in blood was not found. Other data obtained in a study of bacterial-induced immune activation in patients with suppressed viral load also confirm its independence from the HIV load in blood [121, 126, 127]. It is assumed that the immune activation is mediated through TLRs [128, 129, 130].

It is widely accepted that liver cirrhosis compounds microbial translocation from the intestine into the bloodstream. A comparison of sCD14 blood levels in HIV/HCV-coinfected subjects, with varying degrees of liver fibrosis, showed that in patients with higher cirrhosis the soluble receptor concentration was more than in subjects with minimal organ destruction [131, 132, 133]. In HIV infection, the role of cirrhosis in the enhancement of the systemic inflammatory process was demonstrated while comparing two variants: compensated and uncompensated inflammations [134]. When uncompensated, a significantly higher level of LPS-binding protein (LBP) was detected in the patients’ blood. The increase in the LBP content was accompanied by the rise in sCD14, sTNFR-I, and IL-6 concentrations.

Entry of large amounts of microbial products into the liver can have a negative impact on its function in HIV-infected individuals [135]. Liver cells actively express TLRs [136, 137, 138, 139] that can interact with bacterial molecules and cause a marked inflammatory response in liver tissue [140, 141]. However, in experiments, it has been shown that prolonged exposure to LPS leads to a gradual decrease in the hepatocyte’s sensitivity and a weakening of its ability to capture the bacterial lipopolysaccharide [142]. This causes a decrease in the detoxification function of the liver in patients monoinfected with HIV. The presence of hepatitis and cirrhosis additionally contributes to the decrease in LPS clearance by hepatocytes [143]. Consequently, in coinfection, microbial products entering the blood should cause strong immune activation. Indeed, the level of immune activation in HIV/HCV coinfection is higher than that in HIV [144, 145] or HCV [146, 147] monoinfection alone.

Activation of CD4+ and CD8+ T-cells is often followed by apoptosis: a phenomenon called “activation-induced cell death” (AICD) [148, 149]. AICD can be achieved through several mechanisms involving T-cell receptor stimulation, CD95, and various cytokines [150]. The role of innate immunity receptors in this phenomenon has also been established. Addition of various TLR ligands to cultured T-lymphocytes from healthy donors induced CD38 expression on CD4+ and CD8+ T-cells in short-term (less than 24 h) cultures [129]. Long-term culture (7 days) with TLR ligands led to a pronounced CD69 expression on CD8+ T-lymphocytes and Ki-67 expression in CD4+ T-cells. In such a case, CD8+ elements retained viability, and cycling CD4+ T-lymphocytes died. The data presented show how CD4+ T-cells, activated by microbial products, can die in HIV/HCV-coinfected patients. It is known that in untreated HIV-infected patients, HCV coinfection increases the apoptosis indices, but that effect is canceled by ART [151]. Later, the same authors found that in HIV/HCV coinfection, CD4+ T-lymphocytes are sensitized to Fas-induced apoptosis [152].

Advertisement

5. HIV/HCV-coinfected immunological nonresponders

A current concern is that as many as 30% of treated HIV-infected patients experience poor CD4+ T-cell restoration despite prolonged suppression of viral replication during ART (discordant immune response). These “immune nonresponders” (INRs) contrast with “immune responders” who recover their CD4+ T-cells to the normal range (>500/μL) while being treated. The discordant immune response is linked to an additional risk of mortality and non-AIDS-defining morbidities [153]. Still, the mechanisms responsible for insufficient CD4+ T-cell recovery during ART have not been fully discerned.

Our own studies have provided some additional information on the negative impact of hepatitis C on the natural course of an HIV infection. In our Russian cohort, we showed that HCV coinfection is a sufficient risk factor for the formation of a discordant CD4+ T-lymphocyte response to ART [154]. At the same time, we found that in coinfected patients when compared with patients monoinfected with HIV the increase in systemic inflammation was associated with liver damage and destruction of the hepatic barrier that protects against microbial products coming from the intestine [155]. INRs have a higher degree of hepatic tissue destruction and express more sclerosing processes [156]. We also found that HIV/HCV-coinfected INR subjects are characterized not only by a deep CD4+ T-lymphocyte deficiency but also by decreased IL-2 production. Earlier, it was demonstrated [157] that through the secretion of this cytokine, T-cells can stimulate the natural killer cell’s antifibrotic activity (realized through the killing of liver stellate cells). Thus, in HIV/HCV-coinfected INR patients, the IL-2 deficiency may be regarded as a liver fibrosis-accelerating factor.

In summary, it can be concluded that HIV/HCV-coinfected INRs are characterized by both a CD4+ T-lymphocyte deficiency and reduced IL-2 production which leads to liver destruction and cirrhosis formation. Under such conditions increased intestinal permeability (due to HIV infection) is supplemented by destruction of the hepatic barrier, accompanied by the entry of microbial products into the bloodstream. As a result, despite the ART-mediated HIV replication suppression, a pronounced systemic inflammation develops, leading to non-AIDS-defining diseases. Therefore, in coinfection, not only HIV infection therapy but also hepatitis С treatment should be actively carried out.

Advertisement

6. Conclusion

In HIV/HCV coinfection, the level of immune activation is higher than in HIV and HCV monoinfections. The transition of immunocompetent cells to the activated state is accompanied not only by their loss through the AICD mechanism but also leads to the development of non-AIDS-defining diseases, especially against the background of ART administration.

It can also be concluded that HIV infection exerts a greater influence on the natural course of a hepatitis C infection than the opposite. The key link in this influence is the depletion of CD4+ T-cells. However, not only CD4+ T-lymphocyte deficiency determines the HIV-negative impact on the development of hepatitis C. Even on the background of ART, HIV infection has a pronounced suppressive effect on NK cell activity against HCV. In HIV/HCV coinfection, a decrease in natural killer cell numbers and their ability to respond to IL-2 is observed. More importantly, the production of IFN-γ by those cells is also significantly impaired [158]. The outcome is the rapid development of liver fibrosis and cirrhosis.

As to changing the course of HIV infection against the background of hepatitis C, many questions remain. It is necessary to identify the main CD4+ and CD8+ T-cell subsets which are affected by HCV infection. In addition, studies of functional changes in T-lymphocytes like exhaustion [159, 160, 161], senescence [162, 163, 164], and loss of cytokine receptors [165, 166] must occur. It is also important to understand the nature of the immune system activation in HIV/HCV coinfection which is important in predicting the development of non-AIDS-defining diseases. Solving these issues requires further experimental and clinical research.

Advertisement

Acknowledgments

This work was supported by the Russian Foundation for Basic Research (17-54-30006).

Advertisement

Conflict of interest

The authors have no conflicts of interest to disclose.

References

  1. 1. Boulougoura A, Sereti I. HIV infection and immune activation: The role of coinfections. Current Opinion in HIV and AIDS. 2016;11(2):191-200
  2. 2. Appay V, Kelleher AD. Immune activation and immune aging in HIV infection. Current Opinion in HIV and AIDS. 2016;11(2):242-249
  3. 3. Taiwo B, Barcena L, Tressler R. Understanding and controlling chronic immune activation in the HIV-infected patients suppressed on combination antiretroviral therapy. Current HIV/AIDS Reports. 2013;10(1):21-32
  4. 4. Masia M, Robledano C, de la Tabla VO, Antequera P, Lumbreras B, Hernandez I, et al. Coinfection with human herpesvirus 8 is associated with persistent inflammation and immune activation in virologically suppressed HIV-infected patients. PLoS One [Internet]. 2014;9(8):e105442
  5. 5. Matthews PC, Geretti AM, Goulder PJ, Klenerman P. Epidemiology and impact of HIV coinfection with hepatitis B and hepatitis C viruses in Sub-Saharan Africa. Journal of Clinical Virology. 2014;61(1):20-33
  6. 6. Deffur A, Mulder NJ, Wilkinson RJ. Co-infection with Mycobacterium tuberculosis and human immunodeficiency virus: An overview and motivation for systems approaches. Pathogens and Disease. 2013;69(2):101-113
  7. 7. Effros RB. The silent war of CMV in aging and HIV infection. Mechanisms of Ageing and Development [Internet]. Sep 2016;158:46-52. DOI: 10.1016/j.mad.2015.09.003
  8. 8. Webster DP, Klenerman P, Dusheiko GM. Hepatitis C. Lancet. 2015;385(9973):1124-1135
  9. 9. Platt L, Easterbrook P, Gower E, McDonald B, Sabin K, McGowan C, et al. Prevalence and burden of HCV co-infection in people living with HIV: A global systematic review and meta-analysis. The Lancet Infectious Diseases. 2016;16(7):797-808
  10. 10. Alter MJ. Epidemiology of viral hepatitis and HIV co-infection. Journal of Hepatology. 2006;44(1 Suppl):S6-S9
  11. 11. Peters L, Mocroft A, Lundgren J, Grint D, Kirk O, Rockstroh J. HIV and hepatitis C co-infection in Europe, Israel and Argentina: A EuroSIDA perspective. BMC Infectious Diseases [Internet]. 2014;14(Suppl. 6):S13
  12. 12. Rhodes T, Platt L, Judd A, Mikhailova LA, Sarang A, Wallis N, et al. Hepatitis C virus infection, HIV co-infection, and associated risk among injecting drug users in Togliatti, Russia. International Journal of STD & AIDS. 2005;16(11):749-754
  13. 13. Chen TY, Ding EL, Seage Iii GR, Kim AY. Meta-analysis: Increased mortality associated with hepatitis C in HIV-infected persons is unrelated to HIV disease progression. Clinical Infectious Diseases. 2009;49(10):1605-1615
  14. 14. Puoti M, Moioli MC, Travi G, Rossotti R. The burden of liver disease in human immunodeficiency virus-infected patients. Seminars in Liver Disease. 2012;32(2):103-113
  15. 15. Macias J, Berenguer J, Japon MA, Giron JA, Rivero A, Lopez-Cortes LF, et al. Fast fibrosis progression between repeated liver biopsies in patients coinfected with human immunodeficiency virus/hepatitis C virus. Hepatology. 2009;50(4):1056-1063
  16. 16. Deng LP, Gui XE, Zhang YX, Gao SC, Yang RR. Impact of human immunodeficiency virus infection on the course of hepatitis C virus infection: A meta-analysis. World Journal of Gastroenterology. 2009;15(8):996-1003
  17. 17. Hernandez MD, Sherman KE. HIV/hepatitis C coinfection natural history and disease progression. Current Opinion in HIV and AIDS. 2011;6(6):478-482
  18. 18. Garcia-Samaniego J, Rodriguez M, Berenguer J, Rodriguez-Rosado R, Carbo J, Asensi V, et al. Hepatocellular carcinoma in HIV-infected patients with chronic hepatitis C. The American Journal of Gastroenterology. 2001;96(1):179-183
  19. 19. Puoti M, Bruno R, Soriano V, Donato F, Gaeta GB, Quinzan GP, et al. Hepatocellular carcinoma in HIV-infected patients: Epidemiological features, clinical presentation and outcome. AIDS. 2004;18(17):2285-2293
  20. 20. Brau N, Fox RK, Xiao P, Marks K, Naqvi Z, Taylor LE, et al. Presentation and outcome of hepatocellular carcinoma in HIV-infected patients: A U.S.-Canadian multicenter study. Journal of Hepatology. 2007;47(4):527-537
  21. 21. Operskalski EA, Kovacs A. HIV/HCV co-infection: Pathogenesis, clinical complications, treatment, and new therapeutic technologies. Current HIV/AIDS Reports. 2011;8(1):12-22
  22. 22. Sabin CA, Walker AS, Dunn D. HIV/HCV coinfection, HAART, and liver-related mortality. Lancet. 2004;364(9436):757-758 author reply 8
  23. 23. Hernando V, Alejos B, Monge S, Berenguer J, Anta L, Vinuesa D, et al. All-cause mortality in the cohorts of the Spanish AIDS Research Network (RIS) compared with the general population: 1997-2010. BMC Infectious Diseases. 2013;13:382
  24. 24. Rotman Y, Liang TJ. Coinfection with hepatitis C virus and human immunodeficiency virus: Virological, immunological, and clinical outcomes. Journal of Virology. 2009;83(15):7366-7374
  25. 25. Roe B, Hall WW. Cellular and molecular interactions in coinfection with hepatitis C virus and human immunodeficiency virus. Expert Reviews in Molecular Medicine. 2008;10:e30
  26. 26. Kim AY, Chung RT. Coinfection with HIV-1 and HCV—A one-two punch. Gastroenterology. 2009;137(3):795-814
  27. 27. Sherman KE, O’Brien J, Gutierrez AG, Harrison S, Urdea M, Neuwald P, et al. Quantitative evaluation of hepatitis C virus RNA in patients with concurrent human immunodeficiency virus infections. Journal of Clinical Microbiology. 1993;31(10):2679-2682
  28. 28. Thomas DL, Astemborski J, Vlahov D, Strathdee SA, Ray SC, Nelson KE, et al. Determinants of the quantity of hepatitis C virus RNA. The Journal of Infectious Diseases. 2000;181(3):844-851
  29. 29. Bonacini M, Govindarajan S, Blatt LM, Schmid P, Conrad A, Lindsay KL. Patients co-infected with human immunodeficiency virus and hepatitis C virus demonstrate higher levels of hepatic HCV RNA. Journal of Viral Hepatitis. 1999;6(3):203-208
  30. 30. Beld M, Penning M, Lukashov V, McMorrow M, Roos M, Pakker N, et al. Evidence that both HIV and HIV-induced immunodeficiency enhance HCV replication among HCV seroconverters. Virology. 1998;244(2):504-512
  31. 31. Eyster ME, Fried MW, Di Bisceglie AM, Goedert JJ. Increasing hepatitis C virus RNA levels in hemophiliacs: Relationship to human immunodeficiency virus infection and liver disease. Multicenter Hemophilia Cohort Study. Blood. 1994;84(4):1020-1023
  32. 32. Lin W, Weinberg EM, Tai AW, Peng LF, Brockman MA, Kim KA, et al. HIV increases HCV replication in a TGF-beta1-dependent manner. Gastroenterology. 2008;134(3):803-811
  33. 33. Martinez-Sierra C, Arizcorreta A, Diaz F, Roldan R, Martin-Herrera L, Perez-Guzman E, et al. Progression of chronic hepatitis C to liver fibrosis and cirrhosis in patients coinfected with hepatitis C virus and human immunodeficiency virus. Clinical Infectious Diseases. 2003;36(4):491-498
  34. 34. Benhamou Y, Bochet M, Di Martino V, Charlotte F, Azria F, Coutellier A, et al. Liver fibrosis progression in human immunodeficiency virus and hepatitis C virus coinfected patients. The Multivirc Group. Hepatology. 1999;30(4):1054-1058
  35. 35. Potter M, Odueyungbo A, Yang H, Saeed S, Klein MB. Canadian Co-infection Cohort Study I. Impact of hepatitis C viral replication on CD4+ T-lymphocyte progression in HIV-HCV coinfection before and after antiretroviral therapy. AIDS. 2010;24(12):1857-1865
  36. 36. Falconer K, Gonzalez VD, Reichard O, Sandberg JK, Alaeus A. Spontaneous HCV clearance in HCV/HIV-1 coinfection associated with normalized CD4 counts, low level of chronic immune activation and high level of T cell function. Journal of Clinical Virology. 2008;41(2):160-163
  37. 37. Rohrbach J, Robinson N, Harcourt G, Hammond E, Gaudieri S, Gorgievski M, et al. Cellular immune responses to HCV core increase and HCV RNA levels decrease during successful antiretroviral therapy. Gut. 2010;59(9):1252-1258
  38. 38. Reiberger T, Ferlitsch A, Sieghart W, Kreil A, Breitenecker F, Rieger A, et al. HIV-HCV co-infected patients with low CD4+ cell nadirs are at risk for faster fibrosis progression and portal hypertension. Journal of Viral Hepatitis. 2010;17(6):400-409
  39. 39. Mohsen AH, Easterbrook PJ, Taylor C, Portmann B, Kulasegaram R, Murad S, et al. Impact of human immunodeficiency virus (HIV) infection on the progression of liver fibrosis in hepatitis C virus infected patients. Gut. 2003;52(7):1035-1040
  40. 40. McGovern BH, Golan Y, Lopez M, Pratt D, Lawton A, Moore G, et al. The impact of cirrhosis on CD4+ T cell counts in HIV-seronegative patients. Clinical Infectious Diseases. 2007;44(3):431-437
  41. 41. Rashkin S, Rouster S, Goodman ZD, Sherman KE. T-helper cells and liver fibrosis in hepatitis C virus-monoinfected patients. Journal of Viral Hepatitis. 2010;17(3):222-226
  42. 42. Mandorfer M, Payer BA, Schwabl P, Steiner S, Ferlitsch A, Aichelburg MC, et al. Revisiting liver disease progression in HIV/HCV-coinfected patients: The influence of vitamin D, insulin resistance, immune status, IL28B and PNPLA3. Liver International. 2015;35(3):876-885
  43. 43. Kooij KW, Wit FW, van Zoest RA, Schouten J, Kootstra NA, van Vugt M, et al. Liver fibrosis in HIV-infected individuals on long-term antiretroviral therapy: Associated with immune activation, immunodeficiency and prior use of didanosine. AIDS. 2016;30(11):1771-1780
  44. 44. Swanson S, Ma Y, Scherzer R, Huhn G, French AL, Plankey MW, et al. Association of HIV, hepatitis C virus, and liver fibrosis severity with the enhanced liver fibrosis score. The Journal of Infectious Diseases. 2016;213(7):1079-1086
  45. 45. Mandorfer M, Schwabl P, Steiner S, Reiberger T, Peck-Radosavljevic M. Advances in the management of HIV/HCV coinfection. Hepatology International. 2016;10(3):424-435
  46. 46. Hui AY, Friedman SL. Molecular basis of hepatic fibrosis. Expert Reviews in Molecular Medicine. 2003;5(5):1-23
  47. 47. Zhao Q, Qin CY, Zhao ZH, Fan YC, Wang K. Epigenetic modifications in hepatic stellate cells contribute to liver fibrosis. The Tohoku Journal of Experimental Medicine. 2013;229(1):35-43
  48. 48. Friedman SL. Hepatic stellate cells: Protean, multifunctional, and enigmatic cells of the liver. Physiological Reviews. 2008;88(1):125-172
  49. 49. Lee UE, Friedman SL. Mechanisms of hepatic fibrogenesis. Best Practice & Research. Clinical Gastroenterology. 2011;25(2):195-206
  50. 50. Marrone G, Shah VH, Gracia-Sancho J. Sinusoidal communication in liver fibrosis and regeneration. Journal of Hepatology. 2016
  51. 51. Yin C, Evason KJ, Asahina K, Stainier DY. Hepatic stellate cells in liver development, regeneration, and cancer. The Journal of Clinical Investigation. 2013;123(5):1902-1910
  52. 52. Greuter T, Shah VH. Hepatic sinusoids in liver injury, inflammation, and fibrosis: New pathophysiological insights. Journal of Gastroenterology. 2016;51(6):511-519
  53. 53. Rossmanith W, Schulte-Hermann R. Biology of transforming growth factor beta in hepatocarcinogenesis. Microscopy Research and Technique. 2001;52(4):430-436
  54. 54. Matsuzaki K. Modulation of TGF-beta signaling during progression of chronic liver diseases. Frontiers in Bioscience (Landmark Ed). 2009 Jan 1;14:2923-2934
  55. 55. Hernandez-Gea V, Friedman SL. Pathogenesis of liver fibrosis. Annual Review of Pathology. 2011;6:425-456
  56. 56. Tsukamoto H, Zhu NL, Asahina K, Mann DA, Mann J. Epigenetic cell fate regulation of hepatic stellate cells. Hepatology Research. 2011;41(7):675-682
  57. 57. Anatol P, Robert F, Danuta P. Effect of interferon alpha2b plus ribavirin treatment on selected growth factors in respect to inflammation and fibrosis in chronic hepatitis C. World Journal of Gastroenterology. 2005;11(12):1854-1858
  58. 58. Jain MK, Adams-Huet B, Terekhova D, Kushner LE, Bedimo R, Li X, et al. Acute and chronic immune biomarker changes during interferon/ribavirin treatment in HIV/HCV co-infected patients. Journal of Viral Hepatitis. 2015;22(1):25-36
  59. 59. El-Bassiouni NE, Nosseir MM, Madkour ME, Zoheiry MM, Bekheit IW, Ibrahim RA, et al. Role of fibrogenic markers in chronic hepatitis C and associated hepatocellular carcinoma. Molecular Biology Reports. 2012;39(6):6843-6850
  60. 60. Nath NC, Rahman MA, Khan MR, Hasan MS, Bhuiyan TM, Hoque MN, et al. Serum hyaluronic acid as a predictor of fibrosis in chronic hepatitis B and C virus infection. Mymensingh Medical Journal. 2011;20(4):614-619
  61. 61. Kong L, Cardona Maya W, Moreno-Fernandez ME, Ma G, Shata MT, Sherman KE, et al. Low-level HIV infection of hepatocytes. Virology Journal. 2012;9:157
  62. 62. Ciccaglione AR, Marcantonio C, Tritarelli E, Tataseo P, Ferraris A, Bruni R, et al. Microarray analysis identifies a common set of cellular genes modulated by different HCV replicon clones. BMC Genomics. 2008;9:309
  63. 63. Su AI, Pezacki JP, Wodicka L, Brideau AD, Supekova L, Thimme R, et al. Genomic analysis of the host response to hepatitis C virus infection. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(24):15669-15674
  64. 64. Hiet MS, Bauhofer O, Zayas M, Roth H, Tanaka Y, Schirmacher P, et al. Control of temporal activation of hepatitis C virus-induced interferon response by domain 2 of nonstructural protein 5A. Journal of Hepatology. 2015;63(4):829-837
  65. 65. Arnaud N, Dabo S, Akazawa D, Fukasawa M, Shinkai-Ouchi F, Hugon J, et al. Hepatitis C virus reveals a novel early control in acute immune response. PLOS Pathogens [Internet]. 2011;7(10):e1002289
  66. 66. Li K, Li NL, Wei D, Pfeffer SR, Fan M, Pfeffer LM. Activation of chemokine and inflammatory cytokine response in hepatitis C virus-infected hepatocytes depends on Toll-like receptor 3 sensing of hepatitis C virus double-stranded RNA intermediates. Hepatology. 2012;55(3):666-675
  67. 67. Wieland S, Makowska Z, Campana B, Calabrese D, Dill MT, Chung J, et al. Simultaneous detection of hepatitis C virus and interferon stimulated gene expression in infected human liver. Hepatology. 2014;59(6):2121-2130
  68. 68. Schoggins JW, Wilson SJ, Panis M, Murphy MY, Jones CT, Bieniasz P, et al. A diverse range of gene products are effectors of the type I interferon antiviral response. Nature. 2011;472(7344):481-485
  69. 69. Metz P, Dazert E, Ruggieri A, Mazur J, Kaderali L, Kaul A, et al. Identification of type I and type II interferon-induced effectors controlling hepatitis C virus replication. Hepatology. 2012;56(6):2082-2093
  70. 70. Terczynska-Dyla E, Bibert S, Duong FH, Krol I, Jorgensen S, Collinet E, et al. Reduced IFNλ4 activity is associated with improved HCV clearance and reduced expression of interferon-stimulated genes. Nature Communications. 2014;5:5699
  71. 71. Scagnolari C, Monteleone K, Cacciotti G, Antonelli G. Role of interferons in chronic hepatitis C infection. Current Drug Targets. 2017;18(7):844-850
  72. 72. Dill MT, Duong FH, Vogt JE, Bibert S, Bochud PY, Terracciano L, et al. Interferon-induced gene expression is a stronger predictor of treatment response than IL28B genotype in patients with hepatitis C. Gastroenterology. 2011;140(3):1021-1031
  73. 73. Lanford RE, Guerra B, Bigger CB, Lee H, Chavez D, Brasky KM. Lack of response to exogenous interferon-alpha in the liver of chimpanzees chronically infected with hepatitis C virus. Hepatology. 2007;46(4):999-1008
  74. 74. Norris S, Collins C, Doherty DG, Smith F, McEntee G, Traynor O, et al. Resident human hepatic lymphocytes are phenotypically different from circulating lymphocytes. Journal of Hepatology. 1998;28(1):84-90
  75. 75. Amadei B, Urbani S, Cazaly A, Fisicaro P, Zerbini A, Ahmed P, et al. Activation of natural killer cells during acute infection with hepatitis C virus. Gastroenterology. 2010;138(4):1536-1545
  76. 76. Holder KA, Stapleton SN, Gallant ME, Russell RS, Grant MD. Hepatitis C virus-infected cells downregulate NKp30 and inhibit ex vivo NK cell functions. Journal of Immunology. 2013;191(6):3308-3318
  77. 77. Yoon JC, Lim JB, Park JH, Lee JM. Cell-to-cell contact with hepatitis C virus-infected cells reduces functional capacity of natural killer cells. Journal of Virology. 2011;85(23):12557-12569
  78. 78. Mondelli MU, Oliviero B, Mele D, Mantovani S, Gazzabin C, Varchetta S. Natural killer cell functional dichotomy: A feature of chronic viral hepatitis? Frontiers in Immunology [Internet]. 2012;3:351
  79. 79. Oliviero B, Varchetta S, Paudice E, Michelone G, Zaramella M, Mavilio D, et al. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology, 60. 2009;137(3):1151, 60 e1-7
  80. 80. Ahlenstiel G, Titerence RH, Koh C, Edlich B, Feld JJ, Rotman Y, et al. Natural killer cells are polarized toward cytotoxicity in chronic hepatitis C in an interferon-alfa-dependent manner. Gastroenterology. 2010;138(1):325-335 e1-2
  81. 81. Frese M, Schwarzle V, Barth K, Krieger N, Lohmann V, Mihm S, et al. Interferon-gamma inhibits replication of subgenomic and genomic hepatitis C virus RNAs. Hepatology. 2002;35(3):694-703
  82. 82. Major ME, Mihalik K, Puig M, Rehermann B, Nascimbeni M, Rice CM, et al. Previously infected and recovered chimpanzees exhibit rapid responses that control hepatitis C virus replication upon rechallenge. Journal of Virology. 2002;76(13):6586-6595
  83. 83. Glässner A, Eisenhardt M, Krämer B, Körner C, Coenen M, Sauerbruch T, et al. NK cells from HCV-infected patients effectively induce apoptosis of activated primary human hepatic stellate cells in a TRAIL-, FasL- and NKG2D-dependent manner. Laboratory Investigation. 2012;92(7):967-977
  84. 84. Terilli RR, Cox AL. Immunity and hepatitis C: A review. Current HIV/AIDS Reports. 2013;10(1):51-58
  85. 85. Christie JM, Healey CJ, Watson J, Wong VS, Duddridge M, Snowden N, et al. Clinical outcome of hypogammaglobulinaemic patients following outbreak of acute hepatitis C: 2 year follow up. Clinical and Experimental Immunology. 1997;110(1):4-8
  86. 86. Semmo N, Lucas M, Krashias G, Lauer G, Chapel H, Klenerman P. Maintenance of HCV-specific T-cell responses in antibody-deficient patients a decade after early therapy. Blood. 2006;107(11):4570-4571
  87. 87. Lauer GM, Walker BD. Hepatitis C virus infection. The New England Journal of Medicine. 2001;345(1):41-52
  88. 88. Farci P, Shimoda A, Wong D, Cabezon T, De Gioannis D, Strazzera A, et al. Prevention of hepatitis C virus infection in chimpanzees by hyperimmune serum against the hypervariable region 1 of the envelope 2 protein. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(26):15394-15399
  89. 89. Morin TJ, Broering TJ, Leav BA, Blair BM, Rowley KJ, Boucher EN, et al. Human monoclonal antibody HCV1 effectively prevents and treats HCV infection in chimpanzees. PLOS Pathogens [Internet]. 2012;8(8):e1002895
  90. 90. Law M, Maruyama T, Lewis J, Giang E, Tarr AW, Stamataki Z, et al. Broadly neutralizing antibodies protect against hepatitis C virus quasispecies challenge. Nature Medicine. 2008;14(1):25-27
  91. 91. Vanwolleghem T, Bukh J, Meuleman P, Desombere I, Meunier JC, Alter H, et al. Polyclonal immunoglobulins from a chronic hepatitis C virus patient protect human liver-chimeric mice from infection with a homologous hepatitis C virus strain. Hepatology. 2008;47(6):1846-1855
  92. 92. Dowd KA, Netski DM, Wang XH, Cox AL, Ray SC. Selection pressure from neutralizing antibodies drives sequence evolution during acute infection with hepatitis C virus. Gastroenterology. 2009;136(7):2377-2386
  93. 93. Grakoui A, Shoukry NH, Woollard DJ, Han JH, Hanson HL, Ghrayeb J, et al. HCV persistence and immune evasion in the absence of memory T cell help. Science. 2003;302(5645):659-662
  94. 94. Shoukry NH, Grakoui A, Houghton M, Chien DY, Ghrayeb J, Reimann KA, et al. Memory CD8+ T cells are required for protection from persistent hepatitis C virus infection. The Journal of Experimental Medicine. 2003;197(12):1645-1655
  95. 95. Thimme R, Bukh J, Spangenberg HC, Wieland S, Pemberton J, Steiger C, et al. Viral and immunological determinants of hepatitis C virus clearance, persistence, and disease. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(24):15661-15668
  96. 96. Shin EC, Park SH, Demino M, Nascimbeni M, Mihalik K, Major M, et al. Delayed induction, not impaired recruitment, of specific CD8(+) T cells causes the late onset of acute hepatitis C. Gastroenterology. 2011;141(2):686-695, 95 e1
  97. 97. Neumann-Haefelin C, Thimme R. Success and failure of virus-specific T cell responses in hepatitis C virus infection. Digestive Diseases. 2011;29(4):416-422
  98. 98. Klenerman P, Thimme R. T cell responses in hepatitis C: The good, the bad and the unconventional. Gut. 2012;61(8):1226-1234
  99. 99. Diepolder HM, Zachoval R, Hoffmann RM, Wierenga EA, Santantonio T, Jung MC, et al. Possible mechanism involving T-lymphocyte response to non-structural protein 3 in viral clearance in acute hepatitis C virus infection. Lancet. 1995;346(8981):1006-1007
  100. 100. Missale G, Bertoni R, Lamonaca V, Valli A, Massari M, Mori C, et al. Different clinical behaviors of acute hepatitis C virus infection are associated with different vigor of the anti-viral cell-mediated immune response. The Journal of Clinical Investigation. 1996;98(3):706-714
  101. 101. Smyk-Pearson S, Tester IA, Klarquist J, Palmer BE, Pawlotsky JM, Golden-Mason L, et al. Spontaneous recovery in acute human hepatitis C virus infection: Functional T-cell thresholds and relative importance of CD4 help. Journal of Virology. 2008;82(4):1827-1837
  102. 102. Urbani S, Amadei B, Fisicaro P, Tola D, Orlandini A, Sacchelli L, et al. Outcome of acute hepatitis C is related to virus-specific CD4 function and maturation of antiviral memory CD8 responses. Hepatology. 2006;44(1):126-139
  103. 103. Raziorrouh B, Ulsenheimer A, Schraut W, Heeg M, Kurktschiev P, Zachoval R, et al. Inhibitory molecules that regulate expansion and restoration of HCV-specific CD4+ T cells in patients with chronic infection. Gastroenterology. 2011;141(4):1422-1431, 31 e1-6
  104. 104. Kared H, Fabre T, Bedard N, Bruneau J, Shoukry NH. Galectin-9 and IL-21 mediate cross-regulation between Th17 and Treg cells during acute hepatitis C. PLOS Pathogens [Internet]. 2013;9(6):e1003422
  105. 105. Greub G, Ledergerber B, Battegay M, Grob P, Perrin L, Furrer H, et al. Clinical progression, survival, and immune recovery during antiretroviral therapy in patients with HIV-1 and hepatitis C virus coinfection: The Swiss HIV Cohort Study. Lancet. 2000;356(9244):1800-1805
  106. 106. Santin M, Mestre M, Shaw E, Barbera MJ, Casanova A, Niubo J, et al. Impact of hepatitis C virus coinfection on immune restoration during successful antiretroviral therapy in chronic human immunodeficiency virus type 1 disease. European Journal of Clinical Microbiology & Infectious Diseases. 2008;27(1):65-73
  107. 107. Taye S, Lakew M. Impact of hepatitis C virus co-infection on HIV patients before and after highly active antiretroviral therapy: An immunological and clinical chemistry observation, Addis Ababa, Ethiopia. BMC Immunology [Internet]. 2013;14:23
  108. 108. Weis N, Lindhardt BO, Kronborg G, Hansen AB, Laursen AL, Christensen PB, et al. Impact of hepatitis C virus coinfection on response to highly active antiretroviral therapy and outcome in HIV-infected individuals: A nationwide cohort study. Clinical Infectious Diseases. 2006;42(10):1481-1487
  109. 109. Yacisin K, Maida I, Rios MJ, Soriano V, Nunez M. Hepatitis C virus coinfection does not affect CD4 restoration in HIV-infected patients after initiation of antiretroviral therapy. AIDS Research and Human Retroviruses. 2008;24(7):935-940
  110. 110. Rockstroh JK, Mocroft A, Soriano V, Tural C, Losso MH, Horban A, et al. Influence of hepatitis C virus infection on HIV-1 disease progression and response to highly active antiretroviral therapy. The Journal of Infectious Diseases. 2005;192(6):992-1002
  111. 111. Tsiara CG, Nikolopoulos GK, Dimou NL, Bagos PG, Saroglou G, Velonakis E, et al. Effect of hepatitis C virus on immunological and virological responses in HIV-infected patients initiating highly active antiretroviral therapy: A meta-analysis. Journal of Viral Hepatitis. 2013;20(10):715-724
  112. 112. Mohan M, Kaushal D, Aye PP, Alvarez X, Veazey RS, Lackner AA. Focused examination of the intestinal epithelium reveals transcriptional signatures consistent with disturbances in enterocyte maturation and differentiation during the course of SIV infection. PLoS One [Internet]. 2013;8(4):e60122
  113. 113. Sharpstone D, Neild P, Crane R, Taylor C, Hodgson C, Sherwood R, et al. Small intestinal transit, absorption, and permeability in patients with AIDS with and without diarrhoea. Gut. 1999;45(1):70-76
  114. 114. Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends in Microbiology. 2013;21(1):6-13
  115. 115. Nazli A, Chan O, Dobson-Belaire WN, Ouellet M, Tremblay MJ, Gray-Owen SD, et al. Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLOS Pathogens [Internet]. 2010;6(4):e1000852
  116. 116. Smith AJ, Schacker TW, Reilly CS, Haase AT. A role for syndecan-1 and claudin-2 in microbial translocation during HIV-1 infection. JAIDS Journal of Acquired Immune Deficiency Syndromes. 2010;55(3):306-315
  117. 117. Gordon SN, Cervasi B, Odorizzi P, Silverman R, Aberra F, Ginsberg G, et al. Disruption of intestinal CD4+ T cell homeostasis is a key marker of systemic CD4+ T cell activation in HIV-infected individuals. Journal of Immunology. 2010;185(9):5169-5179
  118. 118. Klatt NR, Estes JD, Sun X, Ortiz AM, Barber JS, Harris LD, et al. Loss of mucosal CD103+ DCs and IL-17+ and IL-22+ lymphocytes is associated with mucosal damage in SIV infection. Mucosal Immunology. 2012;5(6):646-657
  119. 119. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nature Medicine. 2006;12(12):1365-1371
  120. 120. Estes JD, Harris LD, Klatt NR, Tabb B, Pittaluga S, Paiardini M, et al. Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections. PLOS Pathogens [Internet]. 2010;6(8):e1001052
  121. 121. Marchetti G, Bellistri GM, Borghi E, Tincati C, Ferramosca S, La Francesca M, et al. Microbial translocation is associated with sustained failure in CD4+ T-cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy. AIDS. 2008;22(15):2035-2038
  122. 122. Leon A, Leal L, Torres B, Lucero C, Inciarte A, Arnedo M, et al. Association of microbial translocation biomarkers with clinical outcome in controllers HIV-infected patients. AIDS. 2015;29(6):675-681
  123. 123. Marchetti G, Cozzi-Lepri A, Merlini E, Bellistri GM, Castagna A, Galli M, et al. Microbial translocation predicts disease progression of HIV-infected antiretroviral-naive patients with high CD4(+) cell count. AIDS. 2011;25(11):1385-1394
  124. 124. Sandler NG, Wand H, Roque A, Law M, Nason MC, Nixon DE, et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. The Journal of Infectious Diseases. 2011;203(6):780-790
  125. 125. Perkins MR, Bartha I, Timmer JK, Liebner JC, Wolinsky D, Gunthard HF, et al. The interplay between host genetic variation, viral replication, and microbial translocation in untreated HIV-infected individuals. The Journal of Infectious Diseases. 2015;212(4):578-584
  126. 126. Jiang W, Lederman MM, Hunt P, Sieg SF, Haley K, Rodriguez B, et al. Plasma levels of bacterial DNA correlate with immune activation and the magnitude of immune restoration in persons with antiretroviral-treated HIV infection. The Journal of Infectious Diseases. 2009;199(8):1177-1185
  127. 127. Marchetti G, Gori A, Casabianca A, Magnani M, Franzetti F, Clerici M, et al. Comparative analysis of T-cell turnover and homeostatic parameters in HIV-infected patients with discordant immune-virological responses to HAART. AIDS. 2006;20(13):1727-1736
  128. 128. Bukh AR, Melchjorsen J, Offersen R, Jensen JMB, Toft L, Stovring H, et al. Endotoxemia is associated with altered innate and adaptive immune responses in untreated HIV-1 infected individuals. PLoS One [Internet]. 2011;6(6):e21275
  129. 129. Funderburg N, Luciano AA, Jiang W, Rodriguez B, Sieg SF, Lederman MM. Toll-like receptor ligands induce human T cell activation and death, a model for HIV pathogenesis. PLoS One [Internet]. 2008;3(4):e1915
  130. 130. Novati S, Sacchi P, Cima S, Zuccaro V, Columpsi P, Pagani L, et al. General issues on microbial translocation in HIV-infected patients. European Review for Medical and Pharmacological Sciences. 2015;19(5):866-878
  131. 131. Sandler NG, Koh C, Roque A, Eccleston JL, Siegel RB, Demino M, et al. Host response to translocated microbial products predicts outcomes of patients with HBV or HCV infection. Gastroenterology. 2011;141(4):1220-30, 30 e1-3
  132. 132. Balagopal A, Philp FH, Astemborski J, Block TM, Mehta A, Long R, et al. Human immunodeficiency virus-related microbial translocation and progression of hepatitis C. Gastroenterology. 2008;135(1):226-233
  133. 133. Marchetti G, Nasta P, Bai F, Gatti F, Bellistri GM, Tincati C, et al. Circulating sCD14 is associated with virological response to pegylated-interferon-alpha/ribavirin treatment in HIV/HCV co-infected patients. PLoS One [Internet]. 2012;7(2):e32028
  134. 134. de Oca Arjona MM, Marquez M, Soto MJ, Rodriguez-Ramos C, Terron A, Vergara A, et al. Bacterial translocation in HIV-infected patients with HCV cirrhosis: Implication in hemodynamic alterations and mortality. Journal of Acquired Immune Deficiency Syndromes. 2011;56(5):420-427
  135. 135. Scarpellini E, Valenza V, Gabrielli M, Lauritano EC, Perotti G, Merra G, et al. Intestinal permeability in cirrhotic patients with and without spontaneous bacterial peritonitis: Is the ring closed? The American Journal of Gastroenterology. 2010;105(2):323-327
  136. 136. Su GL, Klein RD, Aminlari A, Zhang HY, Steinstraesser L, Alarcon WH, et al. Kupffer cell activation by lipopolysaccharide in rats: Role for lipopolysaccharide binding protein and toll-like receptor 4. Hepatology. 2000;31(4):932-936
  137. 137. Jiang W, Sun R, Wei H, Tian Z. Toll-like receptor 3 ligand attenuates LPS-induced liver injury by down-regulation of toll-like receptor 4 expression on macrophages. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(47):17077-17082
  138. 138. Guo J, Loke J, Zheng F, Hong F, Yea S, Fukata M, et al. Functional linkage of cirrhosis-predictive single nucleotide polymorphisms of toll-like receptor 4 to hepatic stellate cell responses. Hepatology. 2009;49(3):960-968
  139. 139. Nakamoto N, Kanai T. Role of toll-like receptors in immune activation and tolerance in the liver. Frontiers in Immunology [Internet]. 2014;5:e221
  140. 140. Paik YH, Schwabe RF, Bataller R, Russo MP, Jobin C, Brenner DA. Toll-like receptor 4 mediates inflammatory signaling by bacterial lipopolysaccharide in human hepatic stellate cells. Hepatology. 2003;37(5):1043-1055
  141. 141. Kopydlowski KM, Salkowski CA, Cody MJ, van Rooijen N, Major J, Hamilton TA, et al. Regulation of macrophage chemokine expression by lipopolysaccharide in vitro and in vivo. Journal of Immunology. 1999;163(3):1537-1544
  142. 142. Scott MJ, Liu S, Shapiro RA, Vodovotz Y, Billiar TR. Endotoxin uptake in mouse liver is blocked by endotoxin pretreatment through a suppressor of cytokine signaling-1-dependent mechanism. Hepatology. 2009;49(5):1695-1708
  143. 143. Lumsden AB, Henderson JM, Kutner MH. Endotoxin levels measured by a chromogenic assay in portal, hepatic and peripheral venous blood in patients with cirrhosis. Hepatology. 1988;8(2):232-236
  144. 144. Kovacs A, Al-Harthi L, Christensen S, Mack W, Cohen M, Landay A. CD8(+) T cell activation in women coinfected with human immunodeficiency virus type 1 and hepatitis C virus. The Journal of Infectious Diseases. 2008;197(10):1402-1407
  145. 145. Gonzalez VD, Falconer K, Blom KG, Reichard O, Morn B, Laursen AL, et al. High levels of chronic immune activation in the T-cell compartments of patients coinfected with hepatitis C virus and human immunodeficiency virus type 1 and on highly active antiretroviral therapy are reverted by alpha interferon and ribavirin treatment. Journal of Virology. 2009;83(21):11407-11411
  146. 146. Feuth T, Arends JE, Fransen JH, Nanlohy NM, van Erpecum KJ, Siersema PD, et al. Complementary role of HCV and HIV in T-cell activation and exhaustion in HIV/HCV coinfection. PLoS One [Internet]. 2013;8(3):e59302
  147. 147. Hodowanec AC, Brady KE, Gao W, Kincaid SL, Plants J, Bahk M, et al. Characterization of CD4(+) T-cell immune activation and interleukin 10 levels among HIV, hepatitis C virus, and HIV/HCV-coinfected patients. Journal of Acquired Immune Deficiency Syndromes. 2013;64(3):232-240
  148. 148. Budd RC. Activation-induced cell death. Current Opinion in Immunology. 2001;13(3):356-362
  149. 149. Green DR, Droin N, Pinkoski M. Activation-induced cell death in T cells. Immunological Reviews. 2003;193:70-81
  150. 150. Brenner D, Krammer PH, Arnold R. Concepts of activated T cell death. Critical Reviews in Oncology/Hematology. 2008;66(1):52-64
  151. 151. Körner C, Krämer B, Schulte D, Coenen M, Mauss S, Fatkenheuer G, et al. Effects of HCV co-infection on apoptosis of CD4+ T-cells in HIV-positive patients. Clinical Science (London, England). 2009;116(12):861-870
  152. 152. Körner C, Tolksdorf F, Riesner K, Krämer B, Schulte D, Nattermann J, et al. Hepatitis C coinfection enhances sensitization of CD4(+) T-cells towards Fas-induced apoptosis in viraemic and HAART-controlled HIV-1-positive patients. Antiviral Therapy. 2011;16(7):1047-1055
  153. 153. Lederman MM, Calabrese L, Funderburg NT, Clagett B, Medvik K, Bonilla H, et al. Immunologic failure despite suppressive antiretroviral therapy is related to activation and turnover of memory CD4 cells. The Journal of Infectious Diseases. 2011;204(8):1217-1226
  154. 154. Shmagel N, Shmagel K, Chereshnev V. Clinical aspects of inefficiency of highly active antiretroviral therapy. Infectious Diseases. 2011;9(11):5-10
  155. 155. Shmagel KV, Saidakova EV, Shmagel NG, Korolevskaya LB, Chereshnev VA, Robinson J, et al. Systemic inflammation and liver damage in HIV/hepatitis C virus coinfection. HIV Medicine. 2016 Sep;17(8):581-589
  156. 156. Shmagel NG, Shmagel KV, Saidakova EV, Korolevskaya LB, Chereshnev VA. Discordant response of CD4(+) T cells to antiretroviral therapy in HIV-infected patients coinfected with hepatitis C virus is accompanied by increased liver damage. Doklady. Biochemistry and Biophysics. 2015;465:358-360
  157. 157. Glassner A, Eisenhardt M, Kokordelis P, Kramer B, Wolter F, Nischalke HD, et al. Impaired CD4(+) T cell stimulation of NK cell anti-fibrotic activity may contribute to accelerated liver fibrosis progression in HIV/HCV patients. Journal of Hepatology. 2013;59(3):427-433
  158. 158. Goeser F, Glassner A, Kokordelis P, Wolter F, Lutz P, Kaczmarek DJ, et al. HIV mono-infection is associated with an impaired anti-hepatitis C virus activity of natural killer cells. AIDS. 2016;30(3):355-363
  159. 159. Kahan SM, Wherry EJ, Zajac AJ. T cell exhaustion during persistent viral infections. Virology. 2015;479-480:180-193
  160. 160. Fuertes Marraco SA, Neubert NJ, Verdeil G, Speiser DE. Inhibitory receptors beyond T cell exhaustion. Frontiers in Immunology. 2015;6:310
  161. 161. Bui JK, Mellors JW. Reversal of T-cell exhaustion as a strategy to improve immune control of HIV-1. AIDS. 2015;29(15):1911-1915
  162. 162. Aberg JA. Aging, inflammation, and HIV infection. Topics in Antiviral Medicine. 2012;20(3):101-105
  163. 163. Chou JP, Effros RB. T cell replicative senescence in human aging. Current Pharmaceutical Design. 2013;19(9):1680-1698
  164. 164. Tsoukas C. Immunosenescence and aging in HIV. Current Opinion in HIV and AIDS. 2014;9(4):398-404
  165. 165. Bai F, Bellistri GM, Tincati C, Savoldi A, Pandolfo A, Bini T, et al. Reduced CD127 expression on peripheral CD4+ T cells impairs immunological recovery in course of suppressive highly active antiretroviral therapy. AIDS. 2010;24(16):2590-2593
  166. 166. Hartling HJ, Jespersen S, Gaardbo JC, Sambleben C, Thorsteinsson K, Gerstoft J, et al. Reduced IL-7R T cell expression and increased plasma sCD127 in late presenting HIV-infected individuals. Journal of Acquired Immune Deficiency Syndromes. 2017 Jan 1;74(1):81-90

Written By

Konstantin Shmagel and Evgeniya Saidakova

Submitted: 31 January 2018 Reviewed: 27 March 2018 Published: 05 November 2018