Skip to main content
The Journal of Infectious Diseases logoLink to The Journal of Infectious Diseases
. 2020 Nov 27;222(Suppl 9):S802–S813. doi: 10.1093/infdis/jiaa279

Insights Into the Pathophysiology of Liver Disease in HCV/HIV: Does it End With HCV Cure?

Andre J Jeyarajan 1,, Raymond T Chung 1
PMCID: PMC7693973  PMID: 33245355

Abstract

HCV-HIV coinfected patients exhibit rapid progression of liver damage relative to HCV monoinfected patients. The availability of new directly acting antiviral agents has dramatically improved outcomes for coinfected patients as sustained virologic response rates now exceed 95% and fibrosis-related parameters are improved. Nevertheless, coinfected patients still have a higher mortality risk and more severe hepatocellular carcinoma compared to HCV monoinfected patients, implying the existence of pathways unique to people living with HIV that continue to promote accelerated liver disease. In this article, we review the pathobiology of liver disease in HCV-HIV coinfected patients in the directly acting antiviral era and explore the mechanisms through which HIV itself induces liver damage. Since liver disease is one of the leading causes of non-AIDS-related mortality in HIV-positive patients, enhancing our understanding of HIV-associated fibrotic pathways will remain important for new diagnostic and therapeutic strategies to slow or reverse liver disease progression, even after HCV cure.

Keywords: HCV, HIV, direct-acting antivirals, fibrogenesis, HCC


As of 2015, there were approximately 1.6 million persons in North America living with human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome (AIDS) [1]. The introduction of potent HIV antiretroviral therapy (ART) has substantially prolonged the survival of HIV-infected individuals, but this extension of life expectancy has allowed other comorbid conditions to manifest. In a recent Canadian population-based retrospective cohort study, the prevalence of liver disease was 7 and 10 times greater in HIV-positive men and women, respectively, than in their HIV-negative counterparts, and was in large part attributed to coinfection with the hepatitis C virus (HCV) [2]. Most people who acquire acute hepatitis C are unable to spontaneously clear the virus and develop chronic infection, which considerably increases their risk for cirrhosis [3] and thus hepatocellular carcinoma (HCC). Recent meta-analyses estimate that 20% of HIV-infected persons in North America are coinfected with HCV [1]. Coinfection is a major public health concern owing to similar methods of transmission—parenteral, perinatal, and sexual. Furthermore, HIV enhances HCV transmission and accelerates the course of HCV-related liver disease [4, 5]. While the effect of HCV on the natural history of HIV is unclear, it is evident that HCV coinfection categorically increases the risk of all-cause and liver-related mortality for HIV-positive persons [6].

Available evidence suggests that sustained virologic response (SVR)—undetectable HCV RNA—is associated with reduced incidence of hepatic decompensation and HCC and a lower mortality risk in HCV-infected patients [7]. In the era when pegylated interferon-alfa and ribavirin (PEG-IFN-alfa/RBV) was the standard of care for treating HCV, HCV-HIV coinfected patients did not respond as well as those infected with HCV alone [8, 9]. However, following the development and implementation of directly acting antiviral (DAAs) agents in 2011, HCV-HIV coinfected patients now achieve SVR rates on par with those of HCV monoinfected patients [10] that are also accompanied by improved measures of fibrosis, as assessed by transient elastography [11]. Indeed, the guidelines established by the American Association for the Study of Liver Diseases and the Infectious Diseases Society of America for the management of HCV-HIV coinfected patients recommend the same treatment regimens as those prescribed for patients with chronic hepatitis C [12].

In the pre-DAA era, hepatic fibrosis progressed more rapidly in HCV-HIV coinfected patients than in HCV monoinfected patients [13]. Recent data modeling using the Swiss HIV Cohort Study, however, predicts that this will not be the case if coinfected patients are identified and treated with DAAs at METAVIR F0 or F1 stage fibrosis [14]. Furthermore, a recent prospective study of 2 French national cohorts revealed that, in the context of ART and DAAs, HCV-HIV patients with compensated cirrhosis experience similar risks for further liver events and HCC as HCV monoinfected patients [15]. Therefore, it may seem that research into the pathogenesis of liver disease in HCV-HIV coinfected patients is of little consequence at present. However, coinfected patients still have a greater mortality risk and earlier onset and more aggressive HCC [15], suggesting that DAAs produce incomplete resolution of inflammatory and profibrogenic stimuli. In addition, it is becomingly increasingly evident that HIV infection itself can promote liver damage. For example, hepatic steatosis (fatty liver) is present in up to 36% of HIV-positive patients and portends an increased risk of developing advanced fibrosis [16]. Thus, in the absence of approved antifibrotic drugs, improving our understanding of mechanisms of hepatic fibrogenesis in HCV-HIV coinfection may reveal unique pathways that can be exploited to retard or reverse liver disease progression.

In this article, we will first review the pathogenesis of HCV-HIV related liver disease and elaborate on mechanisms through which HIV infection impacts fibrogenesis. We will then ultimately evaluate whether HCV clearance is sufficient to prevent further liver damage and progression to HCC.

HEPATIC FIBROSIS

Hepatic fibrosis is a reversible wound-healing response to sustained hepatocellular injury that results in increased extracellular matrix (ECM) deposition in the perisinusoidal region (space of Disse) by hepatic myofibroblasts. Progressive ECM accumulation disrupts blood flow through the sinusoids as the normal parenchymal architecture becomes dissected by fibrous septa. While an array of hepatic cells are involved in fibrogenesis, hepatic stellate cells (HSCs) are the primary drivers [17]. Chronic liver injury foments an inflammatory milieu that promotes the transdifferentiation of quiescent HSCs into activated, chemotactic, ECM-producing myofibroblasts. Activation is regulated by a variety of extracellular signals from both immune and parenchymal cells; of particular interest are hepatocytes, macrophages, and natural killer (NK) cells. Free oxygen radicals (reactive oxygen species) and apoptotic bodies released from damaged hepatocytes may directly or indirectly activate HSCs [18]. Macrophages, including both liver-resident Kupffer cells (KCs) and bone marrow monocyte-derived macrophages, release cytokines, such as transforming growth factor-β1 (TGF-β1) and tumor necrosis factor (TNF), that promote HSC activation. Binding of TGF-β to its cell surface receptors initiates phosphorylation of cytoplasmic SMAD proteins, which then shuttle into the nucleus to regulate transcription of type 1 collagen, one of the primary components of the ECM [19, 20]. In mice, TNF secreted by hepatic macrophages does not directly activate HSCs but instead facilitates their survival in a nuclear factor κ-light-chain–enhancer of activated B cells (NF-κB)-dependent manner [21]. In contrast to hepatocytes and profibrotic macrophages, which activate HSCs, NK cells have distinct antifibrotic activities, such as clearing senescent activated HSCs [22] and initiating HSC apoptosis through production of IFN-γ [23]. In the following sections we will review how HCV and HIV directly regulate these processes to promote fibrosis (Figure 1).

Figure 1.

Figure 1.

HIV-associated profibrotic pathways. HIV induces ROS production and phosphorylation of NRF2 and SMAD3 in hepatocytes, which in turn activate AREs and TGF-β1 expression, respectively. HIV infection can also lead to hepatocyte apoptosis and ensuing efferocytosis by macrophages and HSCs, which ultimately promotes HSC transdifferentiation into ECM-producing myofibroblasts. HIV can directly activate HSCs and macrophages and initiate the release of cytokines that amplify the immune response. Importantly, macrophages and HSCs also express TLR4 and are sensitive to activation by LPS, circulating levels of which are elevated in HIV-infected patients due to microbial translocation. The intrahepatic accumulation of LPS also increases CXCL16 secretion by myeloid dendritic cells, which functions to attract cytotoxic NK cells to the liver. However, NK cell death increases, thus permitting the survival of activated HSCs. CD4+ T cells may also regulate NK cell activity through secretion of IL-2; however, in HIV-infected patients these cells are defective, preventing clearance of activated HSCs. Abbreviations: ARE, antioxidant response element; CCL2, C-C motif chemokine 2; CCR, C-C chemokine receptor; COL1A1, collagen type I α1; CXCL16, C-X-C motif chemokine 16; CXCR, C-X-C chemokine receptor; DR, death receptor; ECM, extracellular matrix; HIV, human immunodeficiency virus; HSC, hepatic stellate cell; IL-2, interleukin-2; LPS, lipopolysaccharide; mDC, myeloid dendritic cell; NK cell, natural killer cell; P-NRF2, phosphorylated nuclear factor erythroid 2-related factor 2; P-RELA, phosphorylated transcription factor p65; P-SMAD3, phosphorylated mothers against decapentaplegic homolog 3; ROS, reactive oxygen species; TGF-β1, transforming growth factor-β1; TIMP1, TIMP metallopeptidase inhibitor 1; TLR4, toll-like receptor 4; TNF, tumor necrosis factor; TRAIL, TNF-related apoptosis-inducing ligand receptor.

DIRECT VIRAL EFFECTS ON HEPATIC CELLS

Profibrogenic Cytokine Release and Oxidative Stress

CD4+ T lymphocytes, macrophages, and dendritic cells are well-characterized reservoirs for HIV. By contrast, HIV does not infect hepatocytes, but hepatocytes do express the canonical HIV coreceptors C-C motif chemokine receptor 5 (CCR5) and C-X-C motif chemokine receptor 4 (CXCR4) [24]. In Huh7.5.1 hepatoma cells, HCV JFH1 (Japanese patient with fulminant hepatitis; genotype 2a) and HIV, independently and cooperatively, induced oxidative stress and TGF-β1 signaling, evidenced by increased nuclear factor erythroid 2-related factor 2 (NRF2) and mothers against decapentaplegic homolog 3 (SMAD3) phosphorylation (activation), respectively [25]. When Huh7.5.1 cells were cocultured with LX-2 human HSCs in a transwell system, to facilitate the exchange of secreted soluble factors, HCV infection and HIV exposure similarly promoted reactive oxygen species generation and increased SMAD3 activation, resulting in the upregulation of the ECM-related genes collagen type 1 α 1 (COL1A1) and TIMP metallopeptidase inhibitor 1 (TIMP1) in HSCs [25]. The profibrogenic effects of HCV and HIV have been further buttressed by a 2019 study that modified the coculture system to incorporate HIV-infected primary human macrophages [26]. In this work, HCV (genotype 1a) and HIV induced LX-2 cell transdifferentiation and chemotaxis [26].

Hepatocyte Apoptosis

Phagocytosis of apoptotic hepatocytes by macrophages or HSCs (efferocytosis) may contribute to liver fibrosis [18, 27]. It has previously been shown that in vitro exposure of HCV JFH1 and CCR5- and CXCR4-tropic HIV to Huh7.5.1 cells induces caspase-dependent apoptosis through both the extrinsic and intrinsic (mitochondrial) pathways [28]. As with the synergistic interactions between HCV and HIV in activating profibrogenic pathways, both viruses also cooperatively induce Huh7.5.1 apoptosis [28]. These findings have clinical significance as plasma levels of the TNF superfamily member 10 (TNFSF10 or TRAIL) ligand that binds death receptors 4/5 (also known as TRAIL receptors 1/2), upon which activation of the extrinsic apoptotic pathway is dependent, are increased in HIV-infected individuals [29]. Notably, however, the HIV envelope can also activate caspase-independent apoptotic pathways in Huh7 cells [24].

Direct HSC Activation

In contrast to hepatocytes, in vitro experiments have suggested that HIV can infect HSCs, although through pathways separate from those involving CD4, CCR5, and CXCR4 [30]. HIV exposure to HSCs resulted in the propagation of infectious viral particles that could be transmitted to human MT4 T cells in coculture through physical interactions [30]. Moreover, HIV upregulated expression of type I collagen and monocyte chemoattractant protein-1 (MCP-1, also known as CCL2) in stellate cells [30]. Like hepatocytes, HSCs express low levels of CCR5 and CXCR4 and HIV gp120 can activate HSCs in a CCR5-dependent fashion as well [31]. HIV gp120-induced HSC activation is also, in part, regulated by toll-like receptor 4 (TLR4) as incubation with either a chemical inhibitor (CLI-095) or neutralizing antibody suppressed CCL2 secretion and chemotaxis [32].

Because CCR5 is a key entry receptor for HIV, a number of HIV antiretroviral drugs targeting CCR5 have been developed. One such drug, maraviroc, appears to have antifibrotic potential. A 2014 retrospective study found that maraviroc use over 2 years in HCV-HIV coinfected patients was associated with fibrosis stagnation (in 59.38%) and improvement (in 29.62%) [33]. Furthermore, in vitro treatment of LX-2 cells with maraviroc reduced TGF-β1 signaling and ECM production [34]. Although maraviroc is now not commonly used to treat HIV, the dual CCR2-CCR5 receptor antagonist, cenicriviroc, is currently under clinical trial for both HIV treatment [35] and for patients with nonalcoholic steatohepatitis and liver fibrosis [36]. Interestingly, Sherman and colleagues recently revealed that cenicriviroc treatment was associated with improvement in noninvasive markers of fibrosis in HIV-infected patients [37]. However, the mechanism through which CCR2-CCR5 neutralization ameliorates fibrosis in HIV-infected patients remains to be elucidated.

Macrophage Activation

As in HSCs, HIV infection in macrophages can drive immune activation through stimulation of TLRs. In vitro experiments have demonstrated that suppression of TLR4 signaling in monocyte-derived macrophages reduces HIV gp120-effected CCL2 secretion [32]. TLR4 and CCR5 may constitute a “receptor cluster” that transmits and disseminates HIV gp120 signals because CCR5 antagonism in macrophages mitigated lipopolysaccharide (TLR4 ligand)-induced CCL2 upregulation [32]. In KCs isolated from healthy patients or HBV/HCV-positive patients, in vitro infection of HIV also increased expression of proinflammatory cytokines, such as TNF and interleukin-6 (IL-6), when challenged with lipopolysaccharide (LPS) in a TLR4-dependent fashion [38]. Alternatively, HIV infection in primary human KCs can further amplify the immune response through upregulation of triggering receptor expressed on myeloid cells 1 (TREM1) [39]. Notably, the percentage of CD68+ (macrophage marker)/TREM1+ peripheral blood mononuclear cells (PBMCs) is greater in HIV-infected patients than in HCV-monoinfected and uninfected individuals [39].

HIV-associated macrophage activation and fibrogenesis have been confirmed in vivo using simian immunodeficiency virus (SIV)-infected rhesus macaques, a well-defined and representative model for studying HIV pathogenesis. In this animal model, SIV infection increased hepatic trafficking of macrophages expressing CCR2, the corresponding receptor for CCL2 [40]. In addition, hepatic macrophage invasion paralleled hepatic CCL3 (CCR5 ligand), TGF-β, and TNF upregulation [40], thus advancing a potential mechanism for HIV/SIV-induced liver fibrosis.

Although hepatic macrophages are important reservoirs for HIV, viral replication is blocked by ART [38, 41]. One plausible basis for liver disease in HIV-diagnosed patients may be elaboration of the macrophage activation marker soluble CD163 (sCD163), which is cleaved from CD163 scavenger receptors expressed on monocyte lineage cells, perhaps by disintegrin and metalloproteinase domain-containing protein 17 (ADAM17) [42, 43]. sCD163 levels are higher in HCV-HIV coinfected patients when compared to either HCV or HIV monoinfected patients and are associated with inflammation and fibrosis [44]. Successful ART reduces sCD163 levels; however, ART produces incomplete reduction of sCD163 levels to those observed in HIV-uninfected persons, suggesting that fibrogenesis may persist even with well controlled HIV [44].

IMMUNE SYSTEM DYSFUNCTION

T-Cell Depletion and Death

Previous studies have attributed accelerated fibrosis progression in HCV-HIV coinfected patients to increased HCV persistence as a result of dysregulated immune responses, notably a reduction in CD4+ T lymphocyte sensitivity to HCV peptides [45]. The chronic phase of HIV infection is characterized by gradual CD4+ T-cell depletion and robust activation of CD4+ and CD8+ T cells, eventually culminating in T-cell death [46, 47]. Consequently, a high CD4 count has been identified as one of a number of factors that can be utilized to predict spontaneous eradication of acute hepatitis C in HIV-diagnosed men [48]. Interestingly, persons who are able to control both HIV and HCV have reduced T-cell exhaustion and activation and their CD4+ T cells produce more cytokines than those of patients who can neither control HIV nor spontaneously clear HCV [49]. As previously suggested by Sherman and colleagues, exhaustion of both HCV- and HIV-specific CD8+ T cells may be regulated by CD39, which is upregulated in HCV- and HIV-infected patients [50, 51]. CD39 is involved in the hydrolysis of adenosine triphosphate to adenosine [52], which can then inhibit T-cell proliferation through adenosine a2a receptor engagement [53]. HIV can also induce CD8+ T-cell death through Fas cell surface death receptor (FAS)-mediated activation of the extrinsic apoptotic pathway [54].

Interleukin Activation

Mechanistically, it has been proposed that CD4+ and CD8+ T-cell activation drive fibrosis via IL-15, as expression levels parallel HSC activation in HCV-HIV coinfected patients [55]. Interestingly, an IL15 polymorphism (rs10833AA genotype) in coinfected patients is associated with significant liver fibrosis, but patients harboring this genetic variant are also more likely to achieve PEG-IFN-alfa/RBV-induced SVR [56]. The rs12979860 single nucleotide polymorphisms near the IFN-λ3 (IFNL3 or IL28B) gene have also been associated with patient responsiveness to IFN-based HCV treatment; the CC genotype confers a higher SVR rate relative to the TC and TT genotypes [57]. However, in a prospective Canadian cohort of HCV-HIV coinfected patients without fibrosis at baseline, researchers found that the rs12979860CC genotype was associated with increased liver fibrosis [58]. Eslam and colleagues provide congruent support for the profibrogenic effects of rs12979860 genotypes; in a large cohort of chronic hepatitis C patients, nearly 60% possessing the rs12979860CC genotype exhibited mild or advanced steatosis and fibrosis [59]. To reconcile the ostensibly disparate roles of the IFNL genotype in regulating both viral eradication and fibrogenesis, the authors suggest the IFNL genotype mediates activation of inflammatory and apoptotic pathways thereby allowing hepatocytes to clear HCV, yet also permitting sustained inflammation in nonresponders to treatment [59].

CD4+ T cells can also influence fibrogenesis by regulating NK cell degranulation (cytotoxicity) via induction of IL-2 [60]. Glässner et al demonstrated that defective CD4+ T cells from HCV-HIV coinfected patients were unable to activate NK cell cytotoxicity, observing a diminished number of IL-2–positive CD4+ T cells in coinfected patients than in healthy controls or persons with HCV monoinfection [60]. However, a more recent study found no significant difference in NK cell degranulation when cells were cultured in CD4+ T-cell conditioned media from HCV-HIV coinfected patients with variegated grades of fibrosis [61]. Instead, NK cell activation may be regulated by dendritic cells. In SIV-infected rhesus macaques, C-X-C chemokine ligand 16 (CXCL16) produced by myeloid dendritic cells increased shuttling of CXCR6-positive NK cells to the liver [62]. There, a cytotoxic subset was preferentially preserved to perhaps mitigate HSC activation, but NK cell death increased, thus maintaining transformed HSCs [62].

Gut Microbiome

Despite suppression of HIV replication with ART, immune activation biomarkers are still upregulated in many HIV-positive patients [63] and intestinal mucosal CD4+ T-cell counts do not wholly recoup from nadir [64]. Gastrointestinal tract analyses revealed that T helper 17 cells (TH17)—a distinct CD4+ T-cell effector lineage that produce IL-17 and IL-22—are depleted in HIV infection [65]. Because TH17 cells play an important role in host antimicrobial defense [66], their loss can foster microbial translocation and gut dysbiosis, as is observed in patients with HIV [67]. As previously discussed, bacterial endotoxin (LPS) can activate HSCs and macrophages through TLRs. Direct LPS treatment in murine HSCs increased their reactivity to activation by KCs and TGF-β [68]. Furthermore, in vivo administration of LPS in mice induced HSC NF-κB transcription factor p65 (RELA) transcriptional activity [68], thus promoting cell survival. A number of studies have also observed that the intestinal microbiota of HIV-diagnosed persons are enriched in Prevotella enterotype species [69], which may be related to sexual practices [70, 71], although the biological implications of this observation are as yet undefined.

STEATOSIS AND STEATOHEPATITIS

Although hepatic fibrosis is the dominant manifestation of liver disease progression in HCV-HIV coinfected patients, hepatic steatosis, the accumulation of triglycerides in hepatocytes, is also a prevalent histologic finding. The reported prevalence ranges from 13% to 60% [16, 72, 73] and is largely dependent on patient characteristics, such as drinking habits or underlying metabolic disorders, and the genotype of HCV infection; for example, HCV genotype 3 has been directly implicated in steatosis development [74].

Insulin resistance is the major metabolic defect underlying steatosis development, as excess carbohydrates increase hepatocyte triglyceride synthesis—through activation of the de novo lipogenesis transcription factors sterol regulatory element-binding protein 1 (SREBP-1c) and carbohydrate-response element binding protein (ChREBP)—and insulin-sensitive adipose triglyceride lipase (ATGL) catalyzes the lipolysis of peripheral adipose tissue and thus increases the hepatic influx of free fatty acids (FFAs). Excessive alcohol consumption can also contribute to steatosis development through SREBP-1c and ChREBP activation [75]. While steatosis itself is generally benign and indolent, in some patients defective β-oxidation and triglyceride synthesis can ultimately lead to the accumulation of hepatotoxic FFA metabolites, including ceramides, diacylglycerols, and lysophosphatidylcholine, that can induce inflammation and hepatocyte death via activation of apoptotic pathways and stress-inducible kinases (eg, c-Jun N-terminal kinases) [76, 77]. These processes contribute to the hallmarks of steatohepatitis (fatty liver with concomitant necroinflammatory activity), which confers a significantly worse prognosis [78, 79]. In the following sections, we will review how HIV and antiretroviral drugs impact steatosis and steatohepatitis development (Figure 2).

Figure 2.

Figure 2.

Mechanisms of HIV-associated steatosis and steatohepatitis development. HIV ARVs, including NRTIs, NNRTIs, and PIs, have been associated with metabolic dysfunction and impaired mitochondrial activity, which can contribute to the development of steatosis and steatohepatitis. HIV accessory protein Vpr can directly regulate lipogenesis through transcriptional activation of SREBP-1c and ChREBP. Moreover, Vpr can inhibit mitochondrial β-oxidation, evinced by decreased PPARα expression. Consequently, expression of the PPARα target MTTP is also reduced, which thus impairs VLDL export and permits hepatocyte lipid droplet accumulation (macrovesicular steatosis). Dysfunctional β-oxidation and triglyceride synthesis eventually leads to the production of lipotoxic intermediates that can induce inflammation, oxidative stress, and apoptosis, processes characteristic of steatohepatitis. Abbreviations: ARV, antiretroviral; ChREBP, carbohydrate-responsive element-binding protein; FFA, free fatty acid; HIV, human immunodeficiency virus; HSC, hepatic stellate cell; LXRα, liver X nuclear receptor α; MTTP, microsomal triglyceride transfer protein; NNRTI, nonnucleoside reverse transcriptase inhibitor; NRTI, nucleoside reverse transcriptase inhibitor; PI, protease inhibitor; PPARα, peroxisome proliferator-activated receptor α; SREBP-1c, sterol regulatory element-binding protein 1; TG, triglyceride; VLDL, very low-density lipoprotein.

Drug-Related Liver Injury

Early antiretroviral regimens, particularly those based on nucleoside reverse transcriptase inhibitors (NRTIs) and protease inhibitors (PIs), were associated with mitochondrial toxicity [80] and metabolic derangements, notably insulin resistance and lipodystrophy [81–83]. In human hepatoma cells, it has been shown that efavirenz and nevirapine—nonnucleoside reverse transcriptase inhibitors (NNRTIs)—also disrupt mitochondrial activity and increase intracellular lipid accumulation [84, 85]. These findings appear to have clinical relevance as efavirenz use is also associated with the progression of steatosis [73]. As previously reported by Sherman et al., contemporary antiretroviral regimens may also be associated with steatosis [51]. New integrase strand transfer inhibitors can contribute to the development of fatty liver as dolutegravir has been implicated in weight gain [51, 86, 87].

Lipogenesis and Impaired Mitochondrial β-Oxidation

Hitherto, there have been few studies describing direct links between HIV viral effects and steatosis. While more than half of patients with nonalcoholic fatty liver disease (NAFLD) are obese [88], HIV-positive patients with NAFLD appear to have a lower body mass index and self-report to be more physically active [89], implying a unique role for HIV in the development of steatosis. In a 2017 study, Agarwal et al demonstrated that transgenic mice expressing HIV accessory protein Vpr, under a tetracycline-repressible construct, developed hepatic steatosis through increased de novo lipogenesis, evinced by enhanced SREBP-1c and ChREBP expression [90]. Mechanistically, the authors proposed that Vpr binds to liver X receptor-α (LXRα) to promote LXRα-regulated Srebp1c and Chrebp gene expression [90]. Furthermore, HIV Vpr induced dysfunctional β-oxidation, which consequently reduced very low-density lipoprotein (VLDL) secretion via downregulated microsomal triglyceride transfer protein (MTTP) expression [90]. The HIV matrix protein p17 also activates LXRalpha-mediated transcription and increases intracellular lipid accumulation in HepG2 cells [91]. Studies in pathogenic and nonpathogenic SIV-infected nonhuman primates further suggest a role for HIV in the development of NAFLD. He et al observed that a number of subjects developed NAFLD during late-stage SIV infection and the authors contend that a synergistic relationship between high-fat diet feeding and SIV infection inhibits intestinal CD4+ T-cell recovery and promotes CD4+ regulatory T-cell depletion [92].

Alcohol Metabolism

Alcohol use is another major determinant of steatosis progression in HCV-HIV coinfected patients [72]. Heavy alcohol use is associated with hastened fibrosis progression while concurrent HIV suppression with ART actually increases HCV RNA [93, 94]. As with HIV-associated immune activation, ethanol metabolites can also deplete gut mucosal integrity thus increasing intestinal permeability and circulating levels of LPS which can, in turn, activate macrophages and HSCs [95]. In vitro experiments utilizing both primary human hepatocytes and Huh7.5 cells demonstrated that enhanced ethanol metabolism, like HCV, augments HIV-induced apoptosis [96]. The effects of ethanol and HIV on hepatocyte death have also been confirmed in mice with humanized livers [96].

EFFECT OF HCV ERADICATION ON MECHANISMS OF LIVER DISEASE PROGRESSION

While HCV clearance has tangible clinical benefits with respect to liver-related complications and death, studies report that inflammatory and fibrotic biomarkers remain elevated in coinfected patients even after achieving SVR (Table 1). As with ART, IFN-based therapies reduce sCD163 levels but do not return them to baseline, implying that latent macrophage activation may continue to promote HSC activation and ECM accumulation [44]. This also appears to be the case when IFN-free regimens are used as levels did not markedly change after DAA-induced viral eradication in chronic hepatitis C patients [97]. In contrast, plasma TGF-β levels do decrease with DAA therapy in coinfected patients, but this difference is not statistically significant and levels are actually higher in patients with hepatic inflammation (METAVIR A1) post-SVR [98]. Likewise, levels of the proinflammatory cytokines TNF and IL-6 remain unaffected by PEG-IFN/RBV treatment in HCV patients with cirrhosis [99]. SVR also has a negligible effect on gut dysbiosis and the elevated serum endotoxin levels observed in HCV patients with cirrhosis [99]. While the aforementioned studies suggest that an aberrant cytokine milieu persists post-SVR, other studies have reported that DAA regimens reduce circulating levels of chemokines such as IFN-γ–induced protein 10 (IP-10), CXCL11, and macrophage inflammatory protein-1β (MIP-1β) in HCV-HIV coinfected patients [100].

Table 1.

Effect of SVR on Mechanisms of Accelerated Liver Disease in HCV-HIV Coinfected Patients

Mechanism Influence of SVR Overall Effect
Altered cytokine profile Persistently elevated serum IL-6 and TNF levels [99]
No significant effect on TGF-β levels [98]
Decreases IP-10, CXCL11 and MIP-1β [100]
Unclear
Immune system dysfunction Maintains elevated kynurenine/tryptophan ratio [101] None
Microbial translocation and gut dysbiosis No effect on microbiota composition and endotoxin levels [99] None
Profibrotic macrophage activation PEG-IFN-alfa/RBV reduces sCD163 levels [44]
DAAs reduce sCD163 levels in chronic hepatitis C patients relative to baseline, but difference is not significant [97]
Positive
Steatosis Improves HOMA-IR and diabetes [102, 103]
Increases triglyceride and cholesterol export via VLDL [104]
Positive

Abbreviations: DAA, directly acting antiviral; HCV, hepatitis C virus; HIV, human immunodeficiency virus; HOMA-IR, homeostatic model assessment of insulin resistance; IL-6, interleukin-6; IP-10, interferon-γ–induced protein 10; MIP-1β, macrophage inflammatory protein-1β; PEG-IFN-alfa, pegylated interferon alfa; RBV, ribavirin; SVR, sustained virologic response; TGF-β, transforming growth factor-β; TNF, tumor necrosis factor; VLDL, very low-density lipoprotein.

In addition to its profibrogenic activities, TGF-β has an immunosuppressive effect by restricting HCV-reactive T-cell proliferation. Indeed, this effect continues following SVR as inhibition of TGF-β in PBMCs isolated from patients 4 years after viral clearance increased T-cell proliferation following exposure to HCV antigens [105]. Immunosuppression post-SVR in HCV-HIV coinfected patients may also be ascribed to increased tryptophan catabolism, evinced by the maintenance of elevated kynurenine levels and kynurenine/tryptophan ratios [101].

In regard to steatosis development, it has been shown that SVR improves Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) scores and measures of diabetes in HCV-infected patients [102]. DAAs also increase triglyceride and cholesterol release through VLDL, thus normalizing hepatic lipid homeostasis [104]. Most importantly, magnetic resonance imaging revealed that DAAs reduce steatosis in patients with chronic hepatitis C [103].

Overall, however, there appears to be no unequivocal consensus on how DAA-induced SVR affects these mechanistic derangements. Therefore, until more definitive explanations have been realized, HCV-cured, HIV coinfected patients should continue to be monitored for fibrosis progression.

EFFECT OF DAAS ON HCC DEVELOPMENT

Recent studies have revealed that, in HCV monoinfected patients, there remains a residual risk for HCC occurrence even after DAA therapy, especially among those with a history of HCC [106, 107]. This may be somewhat attributable to an altered cytokine milieu that persists after DAA treatment [108]. Debes et al analyzed 12 biomarkers that were elevated prior to DAA initiation and were associated with HCC development in HCV-infected patients [108]. Notable cytokines identified included IL-6, TNF, and TRAIL [108]. In many patients who developed HCC, TNF levels increased after 12 weeks of DAA treatment [108]. IL-6 also increased but only in patients who developed recurrent HCC [108]. Indeed, genetic polymorphisms in TNF are linked with HCC incidence [109] and in mouse HCC models, IL-6 transsignaling regulates HCC development by activating β-catenin and precluding hepatocyte apoptosis through inhibition of the tumor suppressor p53 [108, 110]. By contrast, TRAIL levels decreased in most patients who developed HCC, thus perhaps inhibiting apoptosis and permitting the survival of malignant hepatocytes [108].

As a further matter, it has recently been shown that HCV infection induces an epigenetic signature associated with HCC development that also remains unaltered by DAA therapy [111]. Interestingly, the authors found that the TGF and WNT signaling pathways were significantly enhanced in both differentially modified and expressed genes post-SVR [111]. However, in vitro treatment of HCV-cured (HCV-infected and DAA-treated) Huh7.5 cells with C646 (histone acetyltransferase inhibitor) or erlotinib (epidermal growth factor receptor inhibitor) could redress these epigenetic differences [111]. Therefore, further examination of this epigenetic signature and how HCV treatment could be made more efficacious to eliminate the risk of future tumor development post-SVR is necessary. Until then, continued surveillance of HCC in HCV-cured, HIV-positive patients, especially in those with advanced fibrosis, is recommended [112].

CONCLUSIONS

Because HCV-HIV coinfected patients now routinely achieve SVR rates comparable to those of HCV-monoinfected patients, it would appear that the contribution of HCV to liver disease in HIV-positive patients will be of limited import. However, despite improved liver function with DAAs, many HCV-infected patients with decompensated cirrhosis still die or require liver transplantation [113] and the incidence of HCC among HCV-HIV coinfected patients continues to rise [114]. As a result, future studies examining the longer-term impacts of DAA therapy on HCV-HIV patients with advanced liver disease should be conducted, especially among those with superimposed conditions that can contribute to further liver damage, such as heavy alcohol use and those related to the metabolic syndrome. In addition, it is apparent that SVR does not ameliorate all immune activation and fibrotic mediators and even suppressed HIV infection itself contributes to hepatic inflammation and fibrosis. Further insight into how HIV precisely influences hepatic pathophysiology will allow us to appropriately manage and improve clinical outcomes for these patients. Until more definitive antifibrotic approaches can be developed, continued clinical follow-up of HCV-cured patients, including assessment of fibrosis progression and HCC surveillance, particularly those with established fibrosis, would appear to be warranted.

Notes

Financial support. This work was supported by the National Institutes of Health (grant number AI136715-01 to R. T. C.).

Supplement sponsorship. This supplement is sponsored by educational grants from Gilead Sciences Inc. and Abbott Laboratories.

Potential conflicts of interest. Both authors: No reported conflicts of interest. Both authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

  • 1. Platt L, Easterbrook P, Gower E, et al.  Prevalence and burden of HCV co-infection in people living with HIV: a global systematic review and meta-analysis. Lancet Infect Dis 2016; 16:797–808. [DOI] [PubMed] [Google Scholar]
  • 2. Hogg RS, Eyawo O, Collins AB, et al. ; Comparative Outcomes and Service Utilization Trends (COAST) Study Health-adjusted life expectancy in HIV-positive and HIV-negative men and women in British Columbia, Canada: a population-based observational cohort study. Lancet HIV 2017; 4:e270–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Thein HH, Yi Q, Dore GJ, Krahn MD. Estimation of stage-specific fibrosis progression rates in chronic hepatitis C virus infection: a meta-analysis and meta-regression. Hepatology 2008; 48:418–31. [DOI] [PubMed] [Google Scholar]
  • 4. Polis CB, Shah SN, Johnson KE, Gupta A. Impact of maternal HIV coinfection on the vertical transmission of hepatitis C virus: a meta-analysis. Clin Infect Dis 2007; 44:1123–31. [DOI] [PubMed] [Google Scholar]
  • 5. Fierer DS, Dieterich DT, Fiel MI, et al.  Rapid progression to decompensated cirrhosis, liver transplant, and death in HIV-infected men after primary hepatitis C virus infection. Clin Infect Dis 2013; 56:1038–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Hernando V, Perez-Cachafeiro S, Lewden C, et al. ; CoRIS All-cause and liver-related mortality in HIV positive subjects compared to the general population: differences by HCV co-infection. J Hepatol 2012; 57:743–51. [DOI] [PubMed] [Google Scholar]
  • 7. Nahon P, Bourcier V, Layese R, et al.  Eradication of hepatitis C virus infection in patients with cirrhosis reduces risk of liver and non-liver complications. Gastroenterology 2017; 152:142–56.e2. [DOI] [PubMed] [Google Scholar]
  • 8. 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. [DOI] [PubMed] [Google Scholar]
  • 9. Torriani FJ, Rodriguez-Torres M, Rockstroh JK, et al. ; APRICOT Study Group Peginterferon alfa-2a plus ribavirin for chronic hepatitis C virus infection in HIV-infected patients. N Engl J Med 2004; 351:438–50. [DOI] [PubMed] [Google Scholar]
  • 10. Rockstroh JK, Lacombe K, Viani RM, et al.  Efficacy and safety of glecaprevir/pibrentasvir in patients coinfected with hepatitis C virus and human immunodeficiency virus type 1: the EXPEDITION-2 Study. Clin Infect Dis 2018; 67:1010–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Rial-Crestelo D, Sepúlveda MA, González-Gasca FJ, et al.  Does fibrosis really regress in HIV/HCV co-infected patients after treatment with direct-antiviral agents?  AIDS  2019; 34:427–32. [DOI] [PubMed] [Google Scholar]
  • 12. American Association for the Study of Liver Disease-Infectious Diseases Society of America. HCV guidance: recommendations for testing, managing, and treating hepatitis C http://www.hcvguidelines.org. Accessed 27 May 2020. [DOI] [PMC free article] [PubMed]
  • 13. Konerman MA, Mehta SH, Sutcliffe CG, et al.  Fibrosis progression in human immunodeficiency virus/hepatitis C virus coinfected adults: prospective analysis of 435 liver biopsy pairs. Hepatology 2014; 59: 767–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Zahnd C, Salazar-Vizcaya L, Dufour JF, et al. ; Swiss HIV ; Swiss Hepatitis C Cohort Studies. Modelling the impact of deferring HCV treatment on liver-related complications in HIV coinfected men who have sex with men. J Hepatol 2016; 65:26–32. [DOI] [PubMed] [Google Scholar]
  • 15. Salmon-Ceron D, Nahon P, Layese R, et al. ; ANRS CO12 CirVir and ANRS CO13 HEPAVIH Study Groups Human immunodeficiency virus/hepatitis C virus (HCV) co-infected patients with cirrhosis are no longer at higher risk for hepatocellular carcinoma or end-stage liver disease as compared to HCV mono-infected patients. Hepatology 2019; 70:939–54. [DOI] [PubMed] [Google Scholar]
  • 16. Pembroke T, Deschenes M, Lebouché B, et al.  Hepatic steatosis progresses faster in HIV mono-infected than HIV/HCV co-infected patients and is associated with liver fibrosis. J Hepatol 2017; 67:801–8. [DOI] [PubMed] [Google Scholar]
  • 17. Mederacke I, Hsu CC, Troeger JS, et al.  Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat Commun 2013; 4:2823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Zhan SS, Jiang JX, Wu J, et al.  Phagocytosis of apoptotic bodies by hepatic stellate cells induces NADPH oxidase and is associated with liver fibrosis in vivo. Hepatology 2006; 43:435–43. [DOI] [PubMed] [Google Scholar]
  • 19. Hellerbrand C, Stefanovic B, Giordano F, Burchardt ER, Brenner DA. The role of TGFbeta1 in initiating hepatic stellate cell activation in vivo. J Hepatol 1999; 30: 77–87. [DOI] [PubMed] [Google Scholar]
  • 20. Inagaki Y, Okazaki I. Emerging insights into transforming growth factor beta Smad signal in hepatic fibrogenesis. Gut 2007; 56:284–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Pradere JP, Kluwe J, De Minicis S, et al.  Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology 2013; 58:1461–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Krizhanovsky V, Yon M, Dickins RA, et al.  Senescence of activated stellate cells limits liver fibrosis. Cell 2008; 134:657–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Jeong W, Park O, Suh Y, et al.  Suppression of innate immunity (natural killer cell/interferon-γ) in the advanced stages of liver fibrosis in mice. Hepatology 2011; 53:1373–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Vlahakis SR, Villasis-Keever A, Gomez TS, Bren GD, Paya CV. Human immunodeficiency virus-induced apoptosis of human hepatocytes via CXCR4. J Infect Dis 2003; 188:1455–60. [DOI] [PubMed] [Google Scholar]
  • 25. Salloum S, Holmes JA, Jindal R, et al.  Exposure to human immunodeficiency virus/hepatitis C virus in hepatic and stellate cell lines reveals cooperative profibrotic transcriptional activation between viruses and cell types. Hepatology 2016; 64:1951–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Akil A, Endsley M, Shanmugam S, et al.  Fibrogenic gene expression in hepatic stellate cells induced by HCV and HIV replication in a three cell co-culture model system. Sci Rep 2019; 9:568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Schwabe RF, Tabas I, Pajvani UB.  Mechanisms of fibrosis development in nonalcoholic steatohepatitis.  Gastroenterology  2020; 158(7):1913–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Jang JY, Shao RX, Lin W, et al.  HIV infection increases HCV-induced hepatocyte apoptosis. J Hepatol 2011; 54:612–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Herbeuval JP, Boasso A, Grivel JC, et al.  TNF-related apoptosis-inducing ligand (TRAIL) in HIV-1-infected patients and its in vitro production by antigen-presenting cells. Blood 2005; 105:2458–64. [DOI] [PubMed] [Google Scholar]
  • 30. Tuyama AC, Hong F, Saiman Y, et al.  Human immunodeficiency virus (HIV)-1 infects human hepatic stellate cells and promotes collagen I and monocyte chemoattractant protein-1 expression: implications for the pathogenesis of HIV/hepatitis C virus-induced liver fibrosis. Hepatology 2010; 52:612–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Bruno R, Galastri S, Sacchi P, et al.  gp120 modulates the biology of human hepatic stellate cells: a link between HIV infection and liver fibrogenesis. Gut 2010; 59:513–20. [DOI] [PubMed] [Google Scholar]
  • 32. Del Cornò M, Cappon A, Donninelli G, Varano B, Marra F, Gessani S. HIV-1 gp120 signaling through TLR4 modulates innate immune activation in human macrophages and the biology of hepatic stellate cells. J Leukoc Biol 2016; 100:599–606. [DOI] [PubMed] [Google Scholar]
  • 33. Gonzalez EO, Boix V, Deltoro MG, et al.  The effects of maraviroc on liver fibrosis in HIV/HCV co-infected patients. J Int AIDS Soc 2014; 17:19643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Coppola N, Perna A, Lucariello A, et al.  Effects of treatment with maraviroc a CCR5 inhibitor on a human hepatic stellate cell line. J Cell Physiol 2018; 233:6224–31. [DOI] [PubMed] [Google Scholar]
  • 35. Tobira Therapeutics, Inc. Efficacy, safety, and tolerability of cenicriviroc (CVC) in combination with truvada or sustiva plus truvada in HIV 1-infected, antiretroviral treatment-naïve, adult patients infected with only CCR5-tropic virus.ClinicalTrials.gov., US National Library of Medicine; https://clinicaltrials.gov/ct2/show/NCT01338883. Accessed 27 May 2020. [Google Scholar]
  • 36. Tobira Therapeutics, Inc. Efficacy and safety study of cenicriviroc for the treatment of nonalcoholic steatohepatitis (NASH) in adult participants with liver fibrosis (CENTAUR).ClinicalTrials.gov., US National Library of Medicine; https://clinicaltrials.gov/ct2/show/NCT02217475. Accessed 27 May 2020. [Google Scholar]
  • 37. Sherman KE, Abdel-Hameed E, Rouster SD, et al.  Improvement in hepatic fibrosis biomarkers associated with chemokine receptor inactivation through mutation or therapeutic blockade. Clin Infect Dis 2019; 68:1911–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Mosoian A, Zhang L, Hong F, et al.  Frontline science: HIV infection of Kupffer cells results in an amplified proinflammatory response to LPS. J Leukoc Biol 2017; 101:1083–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Hyun J, McMahon RS, Lang AL, et al.  HIV and HCV augments inflammatory responses through increased TREM-1 expression and signaling in Kupffer and myeloid cells. PLoS Pathog 2019; 15:e1007883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Fisher BS, Green RR, Brown RR, et al.  Liver macrophage-associated inflammation correlates with SIV burden and is substantially reduced following cART. PLoS Pathog 2018; 14:e1006871. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Kandathil AJ, Sugawara S, Goyal A, et al.  No recovery of replication-competent HIV-1 from human liver macrophages. J Clin Invest 2018; 128:4501–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Weaver LK, Hintz-Goldstein KA, Pioli PA, et al.  Pivotal advance: activation of cell surface Toll-like receptors causes shedding of the hemoglobin scavenger receptor CD163. J Leukoc Biol 2006; 80:26–35. [DOI] [PubMed] [Google Scholar]
  • 43. Etzerodt A, Maniecki MB, Møller K, Møller HJ, Moestrup SK. Tumor necrosis factor α-converting enzyme (TACE/ADAM17) mediates ectodomain shedding of the scavenger receptor CD163. J Leukoc Biol 2010; 88:1201–5. [DOI] [PubMed] [Google Scholar]
  • 44. Lidofsky A, Holmes JA, Feeney ER, et al.  Macrophage activation marker soluble CD163 is a dynamic marker of liver fibrogenesis in human immunodeficiency virus/hepatitis C virus coinfection. J Infect Dis 2018; 218:1394–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Danta M, Semmo N, Fabris P, et al.  Impact of HIV on host-virus interactions during early hepatitis C virus infection. J Infect Dis 2008; 197:1558–66. [DOI] [PubMed] [Google Scholar]
  • 46. Douek DC, Picker LJ, Koup RA. T cell dynamics in HIV-1 infection. Annu Rev Immunol 2003; 21:265–304. [DOI] [PubMed] [Google Scholar]
  • 47. Núñez M, Soriano V, López M, et al.  Coinfection with hepatitis C virus increases lymphocyte apoptosis in HIV-infected patients. Clin Infect Dis 2006; 43:1209–12. [DOI] [PubMed] [Google Scholar]
  • 48. Thomson EC, Fleming VM, Main J, et al.  Predicting spontaneous clearance of acute hepatitis C virus in a large cohort of HIV-1-infected men. Gut 2011; 60:837–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Dominguez-Molina B, Ferrando-Martinez S, Tarancon-Diez L, et al.  Immune correlates of natural HIV elite control and simultaneous HCV clearance-supercontrollers. Front Immunol 2018; 9:2897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Gupta PK, Godec J, Wolski D, et al.  CD39 expression identifies terminally exhausted CD8+ T cells. PLoS Pathog 2015; 11:e1005177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Sherman KE, Peters MG, Thomas DL.  HIV and the liver. Top Antivir Med. 2019; 27:101–110. [PMC free article] [PubMed] [Google Scholar]
  • 52. Junger WG. Immune cell regulation by autocrine purinergic signalling. Nat Rev Immunol 2011; 11:201–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Huang S, Apasov S, Koshiba M, Sitkovsky M. Role of A2a extracellular adenosine receptor-mediated signaling in adenosine-mediated inhibition of T-cell activation and expansion. Blood 1997; 90:1600–10. [PubMed] [Google Scholar]
  • 54. Mueller YM, De Rosa SC, Hutton JA, et al.  Increased CD95/Fas-induced apoptosis of HIV-specific CD8+ T cells. Immunity 2001; 15:871–82. [DOI] [PubMed] [Google Scholar]
  • 55. Allison RD, Katsounas A, Koziol DE, et al.  Association of interleukin-15-induced peripheral immune activation with hepatic stellate cell activation in persons coinfected with hepatitis C virus and HIV. J Infect Dis 2009; 200:619–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Jiménez-Sousa MA, Berenguer J, Rallón N, et al.  IL15 polymorphism is associated with advanced fibrosis, inflammation-related biomarkers and virological response in human immunodeficiency virus/hepatitis C virus coinfection. Liver Int 2016; 36:1258–66. [DOI] [PubMed] [Google Scholar]
  • 57. Ge D, Fellay J, Thompson AJ, et al.  Genetic variation in IL28B predicts hepatitis C treatment-induced viral clearance. Nature 2009; 461:399–401. [DOI] [PubMed] [Google Scholar]
  • 58. Moqueet N, Cooper C, Gill J, Hull M, Platt RW, Klein MB; Canadian Co-infection Cohort Responder interferon λ genotypes are associated with higher risk of liver fibrosis in HIV-hepatitis C virus coinfection. J Infect Dis 2016; 214:80–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Eslam M, Hashem AM, Leung R, et al.  Interferon-λ rs12979860 genotype and liver fibrosis in viral and non-viral chronic liver disease. Nat Commun 2015; 6:6422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Glässner A, Eisenhardt M, Kokordelis P, et al.  Impaired CD4⁺ T cell stimulation of NK cell anti-fibrotic activity may contribute to accelerated liver fibrosis progression in HIV/HCV patients. J Hepatol 2013; 59:427–33. [DOI] [PubMed] [Google Scholar]
  • 61. Polo ML, Ghiglione YA, Salido JP, et al.  Liver cirrhosis in HIV/HCV-coinfected individuals is related to NK cell dysfunction and exhaustion, but not to an impaired NK cell modulation by CD4+ T-cells. J Int AIDS Soc 2019; 22:e25375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Evans TI, Li H, Schafer JL, et al.  SIV-induced translocation of bacterial products in the liver mobilizes myeloid dendritic and natural killer cells associated with liver damage. J Infect Dis 2016; 213:361–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. French MA, King MS, Tschampa JM, da Silva BA, Landay AL. Serum immune activation markers are persistently increased in patients with HIV infection after 6 years of antiretroviral therapy despite suppression of viral replication and reconstitution of CD4+ T cells. J Infect Dis 2009; 200:1212–5. [DOI] [PubMed] [Google Scholar]
  • 64. Chun TW, Nickle DC, Justement JS, et al.  Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy. J Infect Dis 2008; 197:714–20. [DOI] [PubMed] [Google Scholar]
  • 65. Brenchley JM, Paiardini M, Knox KS, et al.  Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood 2008; 112:2826–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Kolls JK, Lindén A. Interleukin-17 family members and inflammation. Immunity 2004; 21:467–76. [DOI] [PubMed] [Google Scholar]
  • 67. Vujkovic-Cviji I, Dunham RM, Iwai S, et al.  Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism. Sci Transl Med 2013; 5:193ra91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Seki E, De Minicis S, Osterreicher CH, et al.  TLR4 enhances TGF-beta signaling and hepatic fibrosis. Nat Med 2007; 13:1324–32. [DOI] [PubMed] [Google Scholar]
  • 69. Maurice JB, Garvey L, Tsochatzis EA, et al.  Monocyte-macrophage activation is associated with nonalcoholic fatty liver disease and liver fibrosis in HIV monoinfection independently of the gut microbiome and bacterial translocation. AIDS 2019; 33:805–14. [DOI] [PubMed] [Google Scholar]
  • 70. Noguera-Julian M, Rocafort M, Guillén Y, et al.  Gut microbiota linked to sexual preference and HIV infection. EBioMedicine 2016; 5:135–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Kelley CF, Kraft CS, de Man TJ, et al.  The rectal mucosa and condomless receptive anal intercourse in HIV-negative MSM: implications for HIV transmission and prevention. Mucosal Immunol 2017; 10:996–1007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Woreta TA, Sutcliffe CG, Mehta SH, et al.  Incidence and risk factors for steatosis progression in adults coinfected with HIV and hepatitis C virus. Gastroenterology 2011; 140:809–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Macías J, Berenguer J, Japón MA, et al.  Hepatic steatosis and steatohepatitis in human immunodeficiency virus/hepatitis C virus-coinfected patients. Hepatology 2012; 56:1261–70. [DOI] [PubMed] [Google Scholar]
  • 74. Adinolfi LE, Gambardella M, Andreana A, Tripodi MF, Utili R, Ruggiero G. Steatosis accelerates the progression of liver damage of chronic hepatitis C patients and correlates with specific HCV genotype and visceral obesity. Hepatology 2001; 33:1358–64. [DOI] [PubMed] [Google Scholar]
  • 75. You M, Arteel GE.  Effect of ethanol on lipid metabolism. J Hepatol 2019; 70:237–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Hirsova P, Ibrahim SH, Gores GJ, Malhi H. Lipotoxic lethal and sublethal stress signaling in hepatocytes: relevance to NASH pathogenesis. J Lipid Res 2016; 57:1758–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med 2018; 24:908–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Matteoni CA, Younossi ZM, Gramlich T, Boparai N, Liu YC, McCullough AJ. Nonalcoholic fatty liver disease: a spectrum of clinical and pathological severity. Gastroenterology 1999; 116:1413–9. [DOI] [PubMed] [Google Scholar]
  • 79. Deleuran T, Grønbaek H, Vilstrup H, Jepsen P. Cirrhosis and mortality risks of biopsy-verified alcoholic pure steatosis and steatohepatitis: a nationwide registry-based study. Aliment Pharmacol Ther 2012; 35:1336–42. [DOI] [PubMed] [Google Scholar]
  • 80. Lewis W, Day BJ, Copeland WC. Mitochondrial toxicity of NRTI antiviral drugs: an integrated cellular perspective. Nat Rev Drug Discov 2003; 2:812–22. [DOI] [PubMed] [Google Scholar]
  • 81. Carr A, Samaras K, Burton S, et al.  A syndrome of peripheral  lipodystrophy, hyperlipidaemia and insulin resistance in patients receiving HIV protease inhibitors. AIDS 1998; 12:51–58. [DOI] [PubMed] [Google Scholar]
  • 82. Noor MA, Parker RA, O’Mara E, et al.  The effects of HIV protease inhibitors atazanavir and lopinavir/ritonavir on insulin sensitivity in HIV-seronegative healthy adults. AIDS 2004; 18:2137–44. [DOI] [PubMed] [Google Scholar]
  • 83. Guaraldi G, Squillace N, Stentarelli C, et al.  Nonalcoholic fatty liver disease in HIV-infected patients referred to a metabolic clinic: prevalence, characteristics, and predictors. Clin Infect Dis 2008; 47:250–7. [DOI] [PubMed] [Google Scholar]
  • 84. Blas-García A, Apostolova N, Ballesteros D, et al.  Inhibition of mitochondrial function by efavirenz increases lipid content in hepatic cells. Hepatology 2010; 52:115–25. [DOI] [PubMed] [Google Scholar]
  • 85. Paemanee A, Sornjai W, Kittisenachai S, et al.  Nevirapine induced mitochondrial dysfunction in HepG2 cells. Sci Rep 2017; 7:9194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Menard A, Meddeb L, Tissot-Dupont H, et al.  Dolutegravir and weight gain: an unexpected bothering side effect?  AIDS  2017; 31:1499–500. [DOI] [PubMed] [Google Scholar]
  • 87. Norwood J, Turner M, Bofill C, et al.  Weight gain in persons with HIV switched from efavirenz-based to integrase strand transfer inhibitor-based regimens. J Acquir Immune Defic Syndr 2017; 76:527–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 2016; 64:73–84. [DOI] [PubMed] [Google Scholar]
  • 89. Mohammed SS, Aghdassi E, Salit IE, et al.  HIV-positive patients with nonalcoholic fatty liver disease have a lower body mass index and are more physically active than HIV-negative patients. J Acquir Immune Defic Syndr 2007; 45:432–8. [DOI] [PubMed] [Google Scholar]
  • 90. Agarwal N, Iyer D, Gabbi C, et al.  HIV-1 viral protein R (Vpr) induces fatty liver in mice via LXRα and PPARα dysregulation: implications for HIV-specific pathogenesis of NAFLD. Sci Rep 2017; 7:13362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Renga B, Francisci D, Carino A, et al.  The HIV matrix protein p17 induces hepatic lipid accumulation via modulation of nuclear receptor transcriptoma. Sci Rep 2015; 5:15403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  •  92. He T, Xu C, Krampe N, et al.  High-fat diet exacerbates SIV pathogenesis and accelerates disease progression. J Clin Invest 2019; 129:5474–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  •  93. Cooper CL, Cameron DW. Effect of alcohol use and highly active antiretroviral therapy on plasma levels of hepatitis C virus (HCV) in patients coinfected with HIV and HCV. Clin Infect Dis 2005; 41(suppl1):S105–9. [DOI] [PubMed] [Google Scholar]
  •  94. Lo Re V., 3rd Is moderate alcohol consumption safe for human immunodeficiency virus/hepatitis C virus-coinfected women? Clin Infect Dis 2017; 65:2057–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  •  95. Rao R. Endotoxemia and gut barrier dysfunction in alcoholic liver disease. Hepatology 2009; 50:638–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  •  96. Ganesan M, New-Aaron M, Dagur RS, et al.  Alcohol metabolism potentiates HIV-induced hepatotoxicity: contribution to end-stage liver disease. Biomolecules 2019; 9:851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  •  97. Gorin JB, Malone DFG, Strunz B, et al.  Plasma FABP4 is associated with liver disease recovery during treatment-induced clearance of chronic HCV infection. Sci Rep 2020; 10:2081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  •  98. Schwabl P, Mandorfer M, Steiner S, et al.  Interferon-free regimens improve portal hypertension and histological necroinflammation in HIV/HCV patients with advanced liver disease. Aliment Pharmacol Ther 2017; 45:139–49. [DOI] [PubMed] [Google Scholar]
  •  99. Bajaj JS, Sterling RK, Betrapally NS, et al.  HCV eradication does not impact gut dysbiosis or systemic inflammation in cirrhotic patients. Aliment Pharmacol Ther 2016; 44:638–43. [DOI] [PubMed] [Google Scholar]
  • 100. Carlton-Smith C, Holmes JA, Naggie S, et al.  IFN-free therapy is associated with restoration of type I IFN response in HIV-1 patients with acute HCV infection who achieve SVR. J Viral Hepat 2018; 25:465–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Jenabian MA, Mehraj V, Costiniuk CT, et al.  Influence of hepatitis C virus sustained virological response on immunosuppressive tryptophan catabolism in ART-treated HIV/HCV coinfected patients. J Acquir Immune Defic Syndr 2016; 71:254–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Cacoub P, Desbois AC, Comarmond C, Saadoun D. Impact of sustained virological response on the extrahepatic manifestations of chronic hepatitis C: a meta-analysis. Gut 2018; 67:2025–34. [DOI] [PubMed] [Google Scholar]
  • 103. Tada T, Kumada T, Toyoda H, et al.  Viral eradication reduces both liver stiffness and steatosis in patients with chronic hepatitis C virus infection who received direct-acting anti-viral therapy. Aliment Pharmacol Ther 2018; 47:1012–22. [DOI] [PubMed] [Google Scholar]
  • 104. Sun HY, Cheng PN, Tseng CY, Tsai WJ, Chiu YC, Young KC. Favouring modulation of circulating lipoproteins and lipid loading capacity by direct antiviral agents grazoprevir/elbasvir or ledipasvir/sofosbuvir treatment against chronic HCV infection. Gut 2018; 67:1342–50. [DOI] [PubMed] [Google Scholar]
  • 105. Spaan M, Claassen MAA, Hou J, Janssen HLA, De Knegt RJ, Boonstra A. The intrahepatic T cell compartment does not normalize years after therapy-induced hepatitis C virus eradication. J Infect Dis 2015; 212:386–90. [DOI] [PubMed] [Google Scholar]
  • 106. Reig M, Mariño Z, Perelló C, et al.  Unexpected high rate of early tumor recurrence in patients with HCV-related HCC undergoing interferon-free therapy. J Hepatol 2016; 65:719–26. [DOI] [PubMed] [Google Scholar]
  • 107. Yang JD, Aqel BA, Pungpapong S, Gores GJ, Roberts LR, Leise MD. Direct acting antiviral therapy and tumor recurrence after liver transplantation for hepatitis C-associated hepatocellular carcinoma. J Hepatol 2016; 65:859–60. [DOI] [PubMed] [Google Scholar]
  • 108. Debes JD, van Tilborg M, Groothuismink ZMA, et al.  Levels of cytokines in serum associate with development of hepatocellular carcinoma in patients with HCV infection treated with direct-acting antivirals. Gastroenterology 2018; 154:515–7.e3. [DOI] [PubMed] [Google Scholar]
  • 109. Tarhuni A, Guyot E, Rufat P, et al.  Impact of cytokine gene variants on the prediction and prognosis of hepatocellular carcinoma in patients with cirrhosis. J Hepatol 2014; 61:342–50. [DOI] [PubMed] [Google Scholar]
  • 110. Bergmann J, Müller M, Baumann N, et al.  IL-6 trans-signaling is essential for the development of hepatocellular carcinoma in mice. Hepatology 2017; 65:89–103. [DOI] [PubMed] [Google Scholar]
  • 111. Perez S, Kaspi A, Domovitz T, et al.  Hepatitis C virus leaves an epigenetic signature post cure of infection by direct-acting antivirals. PLoS Genet. 2019; 15:e1008181. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112. Galle PR, Forner A, Llovet JM, et al.  EASL clinical practice guidelines: management of hepatocellular carcinoma. J Hepatol 2018; 69:182–236. [DOI] [PubMed] [Google Scholar]
  • 113. Belli LS, Berenguer M, Cortesi PA, et al. ; European Liver and Intestine Association (ELITA) Delisting of liver transplant candidates with chronic hepatitis C after viral eradication: a European study. J Hepatol 2016; 65:524–31. [DOI] [PubMed] [Google Scholar]
  • 114. Gjærde LI, Shepherd L, Jablonowska E, et al.  Trends in incidences and risk factors for hepatocellular carcinoma and other liver events in HIV and hepatitis C virus-coinfected individuals from 2001 to 2014: a Multicohort Study. Clin Infect Dis 2016; 63:821–9. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of Infectious Diseases are provided here courtesy of Oxford University Press

RESOURCES