scholarly journals Role of Interferon-gamma and Immune Response Biomarkers in Predicting IFN-alpha Responsiveness and Treatment Outcome in Patients with Hepatitis C Virus

2012 ◽  
Vol 8 (4) ◽  
pp. 288-298 ◽  
Author(s):  
Amira F. Barakat ◽  
Asmaa Hegazy ◽  
Raghda E. Farag ◽  
Azza Abdul Baky ◽  
Lamiaa F. Arafa ◽  
...  
Cells ◽  
2019 ◽  
Vol 8 (6) ◽  
pp. 603 ◽  
Author(s):  
Sunil Gupta ◽  
Scott A. Read ◽  
Nicholas A. Shackel ◽  
Lionel Hebbard ◽  
Jacob George ◽  
...  

Micronutrient deficiencies develop for a variety of reasons, whether geographic, socioeconomic, nutritional, or as a result of disease pathologies such as chronic viral infection. As micronutrients are essential for a strong immune response, deficiencies can significantly dampen both the innate and the adaptive arms of antiviral immunity. The innate immune response in particular is crucial to protect against hepatitis C virus (HCV), a hepatotropic virus that maintains chronic infection in up to 80% of individuals if left untreated. While many micronutrients are required for HCV replication, an overlapping group of micronutrients are also necessary to enact a potent immune response. As the liver is responsible for the storage and metabolism of many micronutrients, HCV persistence can influence the micronutrients’ steady state to benefit viral persistence both directly and by weakening the antiviral response. This review will focus on common micronutrients such as zinc, iron, copper, selenium, vitamin A, vitamin B12, vitamin D and vitamin E. We will explore their role in the pathogenesis of HCV infection and in the response to antiviral therapy. While chronic hepatitis C virus infection drives deficiencies in micronutrients such as zinc, selenium, vitamin A and B12, it also stimulates copper and iron excess; these micronutrients influence antioxidant, inflammatory and immune responses to HCV.


2018 ◽  
Author(s):  
Ayesha Obaid ◽  
Anam Naz ◽  
Shifa Tariq Ashraf ◽  
Faryal Mehwish Awan ◽  
Aqsa Ikram ◽  
...  

Background. Hepatitis C Virus (HCV) is a major causative agent of liver infection leading to critical liver damage. In response to HCV, the improper regulation of host immune system leads to chronic infection. The host immune system employs multiple cell types, diverse variety of cytokine mediators and interacting signaling networks to neutralize the HCV infection. To understand the complexity of the interactions within the immune signaling networks, systems biology provides an efficient alternative approach. Integrating such approaches with immunology and virology helps to study highly complex immune regulatory networks within the host and presents a concise view of the whole system. Methods. Initially, a logic-based diagram is generated based on multiple reported interactions between immune cells and cytokines during host immune response to HCV. Furthermore, an abstracted sub-network is modeled qualitatively which consists of both the key cellular and cytokine components of the HCV induced immune system. Rene’ Thomas formalism is applied in the study to generate a qualitative model which requires only the qualitative thresholds and associated logical parameters generated via SMBioNet software in accordance with biological observations. Furthermore, the continuous dynamics of the model have been studied via Petri nets based analysis. Results. In the presence of NS5A protein of HCV, the behaviors of the Natural Killer (NK) and T regulatory (Tregs) cells along with cytokines such as IFN-γ, IL-10, IL-12 are predicted. The model also attempts to consider the viral strategies to circumvent immune response mediated by viral proteins. The state graph analysis enabled the prediction of paths leading to disease state. The most probable cycle is predicted based on maximum betweenness centrality. Furthermore, to study the continuous dynamics of the modeled network, a Petri net (PN) model was generated. The predictive ability of the model implicates the critical role of IL-12 over-expression in pathogenesis. This observation speculates that IL-12 has a dual role under varying circumstances and leads to varying disease outcomes. Conclusion. This model attempts to reduce the noisy biological data and captures a holistic view of the regulations amongst the key determinants of HCV induced adaptive immune responses. The observations warrant for further studies to elucidate the role of IL-12 under varying external and internal stimuli. Also, introducing diversion by therapeutic perturbation may divert the system from diseased paths to recovery by stabilizing the activation of IFN-γ producing NK cells. The modeling approach employed in this study can be extended to include real-time experimental data to propose new therapeutic interventions.


2018 ◽  
Author(s):  
Ayesha Obaid ◽  
Anam Naz ◽  
Shifa Tariq Ashraf ◽  
Faryal Mehwish Awan ◽  
Aqsa Ikram ◽  
...  

Background. Hepatitis C Virus (HCV) is a major causative agent of liver infection leading to critical liver damage. In response to HCV, the improper regulation of host immune system leads to chronic infection. The host immune system employs multiple cell types, diverse variety of cytokine mediators and interacting signaling networks to neutralize the HCV infection. To understand the complexity of the interactions within the immune signaling networks, systems biology provides an efficient alternative approach. Integrating such approaches with immunology and virology helps to study highly complex immune regulatory networks within the host and presents a concise view of the whole system. Methods. Initially, a logic-based diagram is generated based on multiple reported interactions between immune cells and cytokines during host immune response to HCV. Furthermore, an abstracted sub-network is modeled qualitatively which consists of both the key cellular and cytokine components of the HCV induced immune system. Rene’ Thomas formalism is applied in the study to generate a qualitative model which requires only the qualitative thresholds and associated logical parameters generated via SMBioNet software in accordance with biological observations. Furthermore, the continuous dynamics of the model have been studied via Petri nets based analysis. Results. In the presence of NS5A protein of HCV, the behaviors of the Natural Killer (NK) and T regulatory (Tregs) cells along with cytokines such as IFN-γ, IL-10, IL-12 are predicted. The model also attempts to consider the viral strategies to circumvent immune response mediated by viral proteins. The state graph analysis enabled the prediction of paths leading to disease state. The most probable cycle is predicted based on maximum betweenness centrality. Furthermore, to study the continuous dynamics of the modeled network, a Petri net (PN) model was generated. The predictive ability of the model implicates the critical role of IL-12 over-expression in pathogenesis. This observation speculates that IL-12 has a dual role under varying circumstances and leads to varying disease outcomes. Conclusion. This model attempts to reduce the noisy biological data and captures a holistic view of the regulations amongst the key determinants of HCV induced adaptive immune responses. The observations warrant for further studies to elucidate the role of IL-12 under varying external and internal stimuli. Also, introducing diversion by therapeutic perturbation may divert the system from diseased paths to recovery by stabilizing the activation of IFN-γ producing NK cells. The modeling approach employed in this study can be extended to include real-time experimental data to propose new therapeutic interventions.


2021 ◽  
Author(s):  
Batkhishig Munkhjargal ◽  
Bilguun Enkhtuvshin ◽  
Uranbileg Ulziisaikhan ◽  
Baljinnyam Tuvdenjamts ◽  
Khulan Unurbuyan ◽  
...  

AbstractObjectiveHepatitis C virus (HCV) is a single-stranded RNA virus that causes chronic hepatitis, cirrhosis, and liver cancer. Approximately 170 million individuals are infected with HCV worldwide. The pathogenesis of HCV-associated liver injury is thought to be due to the host antiviral immune response, including the T cell response, and excessive production of proinflammatory cytokines, reactive oxygen species, and nitric oxide (NO).Interferon-γ (IFN-γ) is a key cytokine in the adaptive immune response that is primarily secreted from CD4+ T helper cells to induce cytotoxic T lymphocyte (CTL) cell response against HCV infection. Another important role of IFN-γ is the activation of macrophages in the liver resulting in inhibition of viral replication and increased NO production.Enhanced inducible nitric oxide synthase (iNOS) expression and NO production observed in the liver of HCV-infected patients is positively correlated with viral load and hepatic inflammation. HCV-infected macrophages are major producers of NO in the liver. It is not completely understood how HCV proteins affect iNOS expression and what the role of IFN-γ is in HCV protein expression in HCV-infected macrophages. In this study, we examined the effect of INF-γ and HCV proteins on iNOS expression in the Raw264.7 cell line.ResultsConsistent with other studies, HCV core and NS5A proteins induced iNOS expression in macrophages. Moreover, HCV E1 protein-enhanced iNOS expression is highest in the presence and absence of IFN-γ activation.ConclusionThese results indicate that hepatitis C virus core, NS5A, E1 protein regulates iNOS protein expression in IFN-γ-activated and resting macrophage cell lines. These findings points to a future research direction for understanding the pathogenesis of HCV-related liver inflammation.


2018 ◽  
Vol 92 (17) ◽  
Author(s):  
Qinya Xie ◽  
Shengwen Chen ◽  
Renyun Tian ◽  
Xiang Huang ◽  
Rilin Deng ◽  
...  

ABSTRACT Emerging evidence indicates that long noncoding RNAs (lncRNAs) regulate various biological processes, especially innate and adaptive immunity. However, the relationship between lncRNAs and the interferon (IFN) pathway remains largely unknown. Here, we report that lncRNA ITPRIP-1 (lncITPRIP-1) is involved in viral infection and plays a crucial role in the virus-triggered IFN signaling pathway through the targeting of melanoma differentiation-associated gene 5 (MDA5). LncITPRIP-1 can be induced by viral infection, which is not entirely dependent on the IFN signal. Besides, there is no coding potential found in the lncITPRIP-1 transcript. LncITPRIP-1 binds to the C terminus of MDA5, and it possesses the ability to boost the oligomerization of both the full length and the 2 caspase activation and recruitment domains of MDA5 in a K63-linked polyubiquitination-independent manner. Amazingly, we also found that MDA5 can suppress hepatitis C virus (HCV) replication independently of IFN signaling through its C-terminal-deficient domain bound to viral RNA, in which lncITPRIP-1 plays a role as an assistant. In addition, the expression of lncITPRIP-1 is highly consistent with MDA5 expression, indicating that lncITPRIP-1 may function as a cofactor of MDA5. All the data suggest that lncITPRIP-1 enhances the innate immune response to viral infection through the promotion of oligomerization and activation of MDA5. Our study discovers the first lncRNA ITPRIP-1 involved in MDA5 activation. IMPORTANCE Hepatitis C virus infection is a global health issue, and there is still no available vaccine, which makes it urgent to reveal the underlying mechanisms of HCV and host factors. Although RIG-I has been recognized as the leading cytoplasmic sensor against HCV for a long time, recent findings that MDA5 regulates the IFN response to HCV have emerged. Our work validates the significant role of MDA5 in IFN signaling and HCV infection and proposes the first lncRNA inhibiting HCV replication by promoting the activation of MDA5 and mediating the association between MDA5 and HCV RNA, the study of which may shed light on the MDA5 function and treatment for hepatitis C patients. Our suggested model of how lncITPRIP-1 orchestrates signal transduction for IFN production illustrates the essential role of lncRNAs in virus elimination.


2019 ◽  
Author(s):  
WI Twu ◽  
K Tabata ◽  
D Paul ◽  
R Bartenschlager

Sign in / Sign up

Export Citation Format

Share Document