1
|
Leukocyte cell-derived chemotaxin 2 is an antiviral regulator acting through the proto-oncogene MET. Nat Commun 2022; 13:3176. [PMID: 35676290 PMCID: PMC9177837 DOI: 10.1038/s41467-022-30879-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
Retinoic acid-inducible gene (RIG)-I is an essential innate immune sensor that recognises pathogen RNAs and induces interferon (IFN) production. However, little is known about how host proteins regulate RIG-I activation. Here, we show that leukocyte cell-derived chemotaxin 2 (LECT2), a hepatokine and ligand of the MET receptor tyrosine kinase is an antiviral regulator that promotes the RIG-I-mediated innate immune response. Upon binding to MET, LECT2 induces the recruitment of the phosphatase PTP4A1 to MET and facilitates the dissociation and dephosphorylation of phosphorylated SHP2 from MET, thereby protecting RIG-I from SHP2/c-Cbl-mediated degradation. In vivo, LECT2 overexpression enhances RIG-I-dependent IFN production and inhibits lymphocytic choriomeningitis virus (LCMV) replication in the liver, whereas these changes are reversed in LECT2 knockout mice. Forced suppression of MET abolishes IFN production and antiviral activity in vitro and in vivo. Interestingly, hepatocyte growth factor (HGF), an original MET ligand, inhibits LECT2-mediated anti-viral signalling; conversely, LECT2-MET signalling competes with HGF-MET signalling. Our findings reveal previously unrecognized crosstalk between MET-mediated proliferation and innate immunity and suggest that targeting LECT2 may have therapeutic value in infectious diseases and cancer. The innate antiviral immune response is an important defense against infection. Here, the authors show that leukocyte cell-derived chemotaxin 2 (LECT2) promotes RIG-I-mediated innate immune responses by preventing its degradation, and inhibits lymphocytic choriomeningitis virus replication in the liver.
Collapse
|
2
|
Rauff B, Amar A, Chudhary SA, Mahmood S, Tayyab GUN, Hanif R. Interferon-λ rs12979860 genotype association with liver fibrosis in chronic hepatitis C (CHC) patients in the Pakistani population. Arch Virol 2021; 166:1047-1056. [PMID: 33528661 DOI: 10.1007/s00705-020-04901-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/15/2020] [Indexed: 12/18/2022]
Abstract
Risk and progression of liver fibrosis and cirrhosis in chronic hepatitis C (CHC) patients is significantly influenced by host genetic factors in a polygenic manner. The rs12979860 genetic polymorphism in the interferon-λ3-interferon-λ4 (IFNL3-IFNL4) region has been found to be a major determinant of hepatic inflammatory and fibrotic progression in CHC patients of mainly Caucasian origin; however, it is not known if this association applies to other ethnicities, including Pakistani CHC patients. Here, we genotyped IFNL3-IFNL4 rs12979860 genetic variants in a sample set of 502 Pakistani patients with CHC and used logistic regression analysis to determine its association with the risk and progression of HCV-related fibrosis and cirrhosis. We demonstrate that the rs12979860 major (CC) genotype, despite not determining the risk of stage-specific hepatic fibrosis independently, is associated with a marginally significant risk of liver cirrhosis (OR: 1.64, p = 0.049) after an adjustment for age, gender, body mass index, HCV viral load, and liver enzymes. In a subgroup of CHC patients with sustained ALT levels of <60 IU/L, a more pronounced impact of the IFNL3-IFNL4 rs12979860 major (CC) genotype on advanced liver fibrosis (OR: 4.99, p = 0.017) and cirrhosis (OR: 3.34, p = 0.005) was seen. The present study suggests that IFNL3-IFNL4 rs12979860 polymorphism may also be a significant predictor of hepatic fibrosis and cirrhosis in Pakistani CHC patients, especially in those with normal or near-normal liver enzyme levels.
Collapse
Affiliation(s)
- Bisma Rauff
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney and Westmead Hospital, Sydney, Australia
- Institute of Biomedical and Allied Health Sciences (IBAHS), University of Health Sciences (UHS), Lahore, Pakistan
| | - Ali Amar
- Department of Human Genetics and Molecular Biology, University of Health Sciences (UHS), Lahore, Pakistan
| | - Shafiq Ahmad Chudhary
- Institute of Biomedical and Allied Health Sciences (IBAHS), University of Health Sciences (UHS), Lahore, Pakistan
| | - Saqib Mahmood
- Institute of Biomedical and Allied Health Sciences (IBAHS), University of Health Sciences (UHS), Lahore, Pakistan
| | | | - Rumeza Hanif
- Department of Healthcare Biotechnology, Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan.
| |
Collapse
|
3
|
Alonso A, Van der Elst W, Sanchez L, Luaces P, Molenberghs G. Identifying individual predictive factors for treatment efficacy. Biometrics 2020; 78:35-45. [PMID: 33128231 DOI: 10.1111/biom.13398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/12/2020] [Indexed: 11/29/2022]
Abstract
Given the heterogeneous responses to therapy and the high cost of treatments, there is an increasing interest in identifying pretreatment predictors of therapeutic effect. Clearly, the success of such an endeavor will depend on the amount of information that the patient-specific variables convey about the individual causal treatment effect on the response of interest. In the present work, using causal inference and information theory, a strategy is proposed to evaluate individual predictive factors for cancer immunotherapy efficacy. In a first step, the methodology proposes a causal inference model to describe the joint distribution of the pretreatment predictors and the individual causal treatment effect. Further, in a second step, the so-called predictive causal information (PCI), a metric that quantifies the amount of information the pretreatment predictors convey on the individual causal treatment effects, is introduced and its properties are studied. The methodology is applied to identify predictors of therapeutic success for a therapeutic vaccine in advanced lung cancer. A user-friendly R library EffectTreat is provided to carry out the necessary calculations.
Collapse
Affiliation(s)
- Ariel Alonso
- I-BioStat, Catholic University of Leuven, Leuven, Belgium
| | | | | | | | - Geert Molenberghs
- I-BioStat, Catholic University of Leuven, Leuven, Belgium.,I-BioStat, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
4
|
Alharthi J, Latchoumanin O, George J, Eslam M. Macrophages in metabolic associated fatty liver disease. World J Gastroenterol 2020; 26:1861-1878. [PMID: 32390698 PMCID: PMC7201150 DOI: 10.3748/wjg.v26.i16.1861] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 03/10/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023] Open
Abstract
Metabolic associated fatty liver disease (MAFLD), formerly named non-alcoholic fatty liver disease is the most common liver disorder in many countries. The inflammatory subtype termed steatohepatitis is a driver of disease progression to cirrhosis, hepatocellular carcinoma, liver transplantation, and death, but also to extrahepatic complications including cardiovascular disease, diabetes and chronic kidney disease. The plasticity of macrophages in response to various environmental cues and the fact that they can orchestrate cross talk between different cellular players during disease development and progression render them an ideal target for drug development. This report reviews recent advances in our understanding of macrophage biology during the entire spectrum of MAFLD including steatosis, inflammation, fibrosis, and hepatocellular carcinoma, as well as for the extra-hepatic manifestations of MAFLD. We discuss the underlying molecular mechanisms of macrophage activation and polarization as well as cross talk with other cell types such as hepatocytes, hepatic stellate cells, and adipose tissue. We conclude with a discussion on the potential translational implications and challenges for macrophage based therapeutics for MAFLD.
Collapse
Affiliation(s)
- Jawaher Alharthi
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney 2145, NSW, Australia
| | - Olivier Latchoumanin
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney 2145, NSW, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney 2145, NSW, Australia
| | - Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney 2145, NSW, Australia
| |
Collapse
|
5
|
Grzegorzewska AE. Genetic Polymorphisms within Interferon-λ Region and Interferon-λ3 in the Human Pathophysiology: Their Contribution to Outcome, Treatment, and Prevention of Infections with Hepatotropic Viruses. Curr Med Chem 2019; 26:4832-4851. [DOI: 10.2174/0929867325666180719121142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 03/21/2018] [Accepted: 07/09/2018] [Indexed: 12/16/2022]
Abstract
:
Genetic polymorphisms within the interferon λ (IFN-λ) chromosomal region,
mainly rs12979860 of IFN-λ4 gene (IFNL4), are known as associated with spontaneous hepatitis
C virus (HCV) resolution and sustained viral response to therapy with pegylated interferon-
α and ribavirin. Strong linkage disequilibrium of IFNL4 rs12979860 with IFNL4
rs368234815, which is casually associated with HCV spontaneous and therapeutical eradication,
at least partially explains favorable HCV outcomes attributed to major homozygosity in
rs12979860. Effects of IFN-based antiviral treatment are associated with pretreatment expression
of the IFN-λ1 receptor, expression of hepatic IFN-stimulated genes, production of IFN-
λ4, and preactivation of the JAK-STAT signaling. Nowadays direct-acting antivirals (DAAs)
became a potent tool in the treatment of hepatitis C, but IFN-λs are still under investigation as
potential antivirals and might be an option in HCV infection (DAA resistance, recurrent viremia,
adverse effects).
:
Patients with altered immunocompetence are especially prone to infections. In uremic subjects,
polymorphisms within the IFN-λ chromosomal region associate with spontaneous HCV
clearance, similarly like in the non-uremic population. Circulating IFN-λ3 shows a positive
correlation with plasma titers of antibodies to surface antigen of hepatitis B virus (anti-HBs),
which are crucial for protection against hepatitis B virus. More efficient anti-HBs production
in the presence of higher IFN-λ3 levels might occur due to IFN-λ3-induced regulation of indoleamine
2,3-dioxygenase (IDO) expression. IFN-stimulated response element is a part of
IDO gene promoter. It is worth further investigation whether IDO gene, circulating IDO, genetic
polymorphisms within the IFN-λ region, and circulating IFN-λ3 act in concordance in
immunological response to hepatotropic viruses.
Collapse
Affiliation(s)
- Alicja E. Grzegorzewska
- Chair and Department of Nephrology, Transplantology and Internal Diseases, Poznan University of Medical Sciences, Poznan, Poland
| |
Collapse
|
6
|
Abstract
Fibrosis is a highly conserved and coordinated wound healing response to injury. In the liver, injury is promoted by immune effector mechanisms that are common across various disease etiologies and even between organs such as lungs, kidneys, heart, and other organs. Thus, the liver represents a useful model to study inflammation and repair, particularly as it is frequently biopsied in clinical contexts. Currently, strong evidence implicates IFNL3/4 polymorphisms and interferon (IFN)-λ3 levels as determinants of the extent of hepatic inflammation and fibrosis in viral and nonviral liver diseases, as well as in governing the severity of nonhepatotropic viral diseases. Interestingly, IFNL3/4 polymorphisms and IFN-λ3 levels correlate with fibrosis extent in other organs such as the lung and kidney. In this review, we discuss the association between IFN-λ and tissue inflammation and fibrosis in human disease and the potential clinical utility of the findings.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, Australia
| | - Golo Ahlenstiel
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, Australia
| |
Collapse
|
7
|
Laser capture microdissection: techniques and applications in liver diseases. Hepatol Int 2019; 13:138-147. [DOI: 10.1007/s12072-018-9917-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 11/28/2018] [Indexed: 02/07/2023]
|
8
|
Bibert S, Wójtowicz A, Taffé P, Tarr PE, Bernasconi E, Furrer H, Günthard HF, Hoffmann M, Kaiser L, Osthoff M, Fellay J, Cavassini M, Bochud PY. Interferon lambda 3/4 polymorphisms are associated with AIDS-related Kaposi's sarcoma. AIDS 2018; 32:2759-2765. [PMID: 30234607 DOI: 10.1097/qad.0000000000002004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Kaposi's sarcoma, the most common AIDS-related cancer, represents a major public concern in resource-limited countries. Single nucleotide polymorphisms within the Interferon lambda 3/4 region (IFNL3/4) determine the expression, function of IFNL4, and influence the clinical course of an increasing number of viral infections. OBJECTIVES To analyze whether IFNL3/4 variants are associated with susceptibility to AIDS-related Kaposi's sarcoma among MSM enrolled in the Swiss HIV Cohort Study (SHCS). METHODS The risk of developing Kaposi's sarcoma according to the carriage of IFNL3/4 SNPs rs8099917 and rs12980275 and their haplotypic combinations was assessed by using cumulative incidence curves and Cox regression models, accounting for relevant covariables. RESULTS Kaposi's sarcoma was diagnosed in 221 of 2558 MSM Caucasian SHCS participants. Both rs12980275 and rs8099917 were associated with an increased risk of Kaposi's sarcoma (cumulative incidence 15 versus 10%, P = 0.01 and 16 versus 10%, P = 0.009, respectively). Diplotypes predicted to produce the active P70 form (cumulative incidence 16 versus 10%, P = 0.01) but not the less active S70 (cumulative incidence 11 versus 10%, P = 0.7) form of IFNL4 were associated with an increased risk of Kaposi's sarcoma, compared with those predicted not to produce IFNL4. The associations remained significant in a multivariate Cox regression model after adjustment for age at infection, combination antiretroviral therapy, median CD4+ T-cell count nadir and CD4+ slopes (hazard ratio 1.42, 95% confidence interval 1.06-1.89, P = 0.02 for IFLN P70 versus no IFNL4). CONCLUSION This study reports for the first time an association between IFNL3/4 polymorphisms and susceptibility to AIDS-related Kaposi's sarcoma.
Collapse
Affiliation(s)
| | | | - Patrick Taffé
- Institute for Social and Preventive Medicine, University (IUMSP), Lausanne University Hospital, Lausanne
| | - Philip E Tarr
- Department of Medicine, Kantonspital Baselland, University of Basel, Bruderholz
| | - Enos Bernasconi
- Division of Infectious diseases, Regional hospital of Lugano, Lugano
| | - Hansjakob Furrer
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern
| | - Huldrych F Günthard
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich
- Institute of Medical Virology, University of Zurich, Zurich
| | - Matthias Hoffmann
- Division of Infectious Diseases and Hospital Epidemiology, Department of Internal Medicine, Cantonal Hospital St. Gallen, St. Gallen
| | - Laurent Kaiser
- Laboratory of Virology, Division of Infectious Diseases and Division of Laboratory Medicine, University Hospital of Geneva and Medical School, University of Geneva, Geneva
| | - Michael Osthoff
- Division of Infectious Diseases and Hospital Epidemiology and Department of Internal Medicine, University Hospital Basel, Basel
| | - Jacques Fellay
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne
- Precision Medicine unit, Lausanne University Hospital, Lausanne, Switzerland
| | | | | |
Collapse
|
9
|
Ramamurthy N, Marchi E, Ansari MA, Pedergnana V, Mclean A, Hudson E, STOP HCV consortium, Bowden R, Spencer CC, Barnes E, Klenerman P. Impact of Interferon Lambda 4 Genotype on Interferon-Stimulated Gene Expression During Direct-Acting Antiviral Therapy for Hepatitis C. Hepatology 2018; 68:859-871. [PMID: 29534310 PMCID: PMC6207923 DOI: 10.1002/hep.29877] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/23/2018] [Accepted: 03/08/2018] [Indexed: 12/19/2022]
Abstract
New directly acting antivirals (DAAs) provide very high cure rates in most patients infected by hepatitis C virus (HCV). However, some patient groups have been relatively harder to treat, including those with cirrhosis or infected with HCV genotype 3. In the recent BOSON trial, genotype 3, patients with cirrhosis receiving a 16-week course of sofosbuvir and ribavirin had a sustained virological response (SVR) rate of around 50%. In patients with cirrhosis, interferon lambda 4 (IFNL4) CC genotype was significantly associated with SVR. This genotype was also associated with a lower interferon-stimulated gene (ISG) signature in peripheral blood and in liver at baseline. Unexpectedly, patients with the CC genotype showed a dynamic increase in ISG expression between weeks 4 and 16 of DAA therapy, whereas the reverse was true for non-CC patients. Conclusion: These data provide an important dynamic link between host genotype and phenotype in HCV therapy also potentially relevant to naturally acquired infection. (Hepatology 2018; 00:000-000).
Collapse
Affiliation(s)
- Narayan Ramamurthy
- Peter Medawar Building for Pathogen Research and Translational Gastroeneterology Unit, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Emanuele Marchi
- Peter Medawar Building for Pathogen Research and Translational Gastroeneterology Unit, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - M. Azim Ansari
- Peter Medawar Building for Pathogen Research and Translational Gastroeneterology Unit, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUnited Kingdom
- Oxford Martin SchoolUniversity of OxfordOxfordUnited Kingdom
| | - Vincent Pedergnana
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUnited Kingdom
| | - Angela Mclean
- Department of ZoologyUniversity of OxfordOxfordUnited Kingdom
| | - Emma Hudson
- Peter Medawar Building for Pathogen Research and Translational Gastroeneterology Unit, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | | | - Rory Bowden
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUnited Kingdom
| | - Chris C.A. Spencer
- Wellcome Trust Centre for Human GeneticsUniversity of OxfordOxfordUnited Kingdom
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research and Translational Gastroeneterology Unit, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research and Translational Gastroeneterology Unit, Nuffield Department of MedicineUniversity of OxfordOxfordUnited Kingdom
| |
Collapse
|
10
|
Annibali O, Piccioni L, Tomarchio V, Circhetta E, Sarlo C, Franceschini L, Cantonetti M, Rizzo E, Angeletti S, Tirindelli MC, Scagnolari C, Statzu M, Avvisati G, Riva E. Impact of IFN lambda 3/4 single nucleotide polymorphisms on the cytomegalovirus reactivation in autologous stem cell transplant patients. PLoS One 2018; 13:e0200221. [PMID: 30036376 PMCID: PMC6056038 DOI: 10.1371/journal.pone.0200221] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/21/2018] [Indexed: 12/14/2022] Open
Abstract
Cytomegalovirus (CMV) infection represents one of the main cause mortality after Stem Cell Transplantation. Recently, a protective effect of the T allele of rs12979860 IL28B Single Nucleotide Polymorphisms (SNPs) against CMV infection in the allogenic stem cell transplantation was suggested. We investigate whether the rs12979860 IL28B SNP and the relative rs368234815 (IFNλ4) genotype may affect the incidence of active CMV infection in Autologous stem cell transplantation (Auto-SCT) setting. The study included 99 patients who underwent to Auto-SCT. IL28 and IFNΔ4 SNPs were correlated with CMV reactivation along with other clinical and treatment parameters. CMV reactivation by CMV DNAemia was evaluated once a week until day 100 from Auto-SCT. CMV reactivation was documented in 50% (TT-ΔG/ΔG), 35% (CC-TT/TT) and 29.2% (CT-TT/ΔG) of the patients respectively. No differences in CMV copies number were recorded at reactivation between different IL28/IFNλ4 genotypes. The analysis of patients older than 60 years showed a significantly higher incidence of active CMV infection in the TT-ΔG/ΔG (83%) population with respect to CC-TT/TT (21%) and CT-TT/ΔG (40%) patients. Our data suggest a negative role of TT-ΔG/ΔG genotype in the CMV reactivation in Auto-SCT. The exposure to rituximab and the pre-infusion presence of anti CMV IgG also significantly influenced CMV reactivation.
Collapse
Affiliation(s)
- Ombretta Annibali
- Unit of Hematology, Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Livia Piccioni
- Laboratory of Virology, Campus Bio-Medico University, Rome, Italy
| | - Valeria Tomarchio
- Unit of Hematology, Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Erika Circhetta
- Unit of Hematology, Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Chiara Sarlo
- Unit of Hematology, Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Luca Franceschini
- Hematology and Stem Cell Transplant Unit, Tor Vergata University, Rome, Italy
| | - Maria Cantonetti
- Hematology and Stem Cell Transplant Unit, Tor Vergata University, Rome, Italy
| | - Emanuela Rizzo
- Hematology and Stem Cell Transplant Unit, Tor Vergata University, Rome, Italy
| | - Silvia Angeletti
- Laboratory of Pathology and Microbiology, Campus Bio-Medico University, Rome, Italy
| | | | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, “Sapienza” University, Rome, Italy
| | - Maura Statzu
- Department of Molecular Medicine, Laboratory of Virology, “Sapienza” University, Rome, Italy
| | - Giuseppe Avvisati
- Unit of Hematology, Stem Cell Transplantation, Campus Bio-Medico University, Rome, Italy
| | - Elisabetta Riva
- Laboratory of Virology, Campus Bio-Medico University, Rome, Italy
| |
Collapse
|
11
|
Wang L, Yao M, Fang M, Zheng WJ, Dong ZZ, Pan LH, Zhang HJ, Yao DF. Expression of hepatic Wnt5a and its clinicopathological features in patients with hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2018; 17:227-232. [PMID: 29709351 DOI: 10.1016/j.hbpd.2018.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 12/27/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUD Wingless-type MMTV integration site family member 5a (Wnt5a) is involved in carcinogenesis. However, little data are available in Wnt5a signaling with hepatocellular carcinoma (HCC). In the present study, we investigated the expression of hepatic Wnt5a in HCC and the role of Wnt5a in HCC progression and outcome. METHODS Wnt5a expression and cellular distribution in HCCs and their matched paracancerous tissues from 87 patients were analyzed with tissue microarray and immunohistochemistry and compared with hepatic Wnt3a signaling. Wnt5a expression was categorized into low or high based on immunohistochemistry. Overall survival rate of HCC patients was estimated in correlation with the hepatic Wnt5a level using Kaplan-Meier method; the survival difference between patients with low and those with high Wnt5a was compared with log-rank test; and prognostic analysis was carried out with Cox regression. RESULTS Total incidence of Wnt5a expression in the HCC tissues was 70.1%, which was significantly lower (χ2 = 13.585, P < 0.001) than that in their paracancerous tissues (88.5%). Significant difference of Wnt5a intensity was found between HCC and their paracancerous tissues (Z = 8.463, P < 0.001). Wnt5a intensity was inversely correlated with Wnt3a signaling (r = -0.402, P < 0.001) in HCC tissues. A decrease of Wnt5a expression in relation to the clinical staging from stage I to IV and low or no staining at advanced HCC were observed. Wnt5a level was related to periportal embolus (χ2 = 11.069, P < 0.001), TNM staging (χ2 = 8.852, P < 0.05), 5-year survival (χ2 = 4.961, P < 0.05), and confirmed as an independent prognosis factor of HCC patients (hazard ratio: 1.957; 95% confidence interval: 1.109-3.456; P < 0.05). CONCLUSIONS The decrease of hepatic Wnt5a signaling is associated with HCC progression and poor prognosis.
Collapse
Affiliation(s)
- Li Wang
- Department of Medical Informatics, Medical College of Nantong University, Nantong, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Min Yao
- Department of Immunology, Medical College of Nantong University, Nantong, China
| | - Miao Fang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Wen-Jie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Zhi-Zhen Dong
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Liu-Hong Pan
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Hai-Jian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China
| | - Deng-Fu Yao
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, No. 20 West Temple Rd, Nantong 226001, China.
| |
Collapse
|
12
|
Srinidhi BV, Fletcher GJ, Sachidanantham J, Rupali P, Ramalingam VV, Demosthenes JP, Abraham OC, Pulimood SA, Rebekah G, Kannangai R. Effect of Interleukin-28B polymorphism on Interleukin-28 expression and immunological recovery amongst HIV-1-infected individuals following antiretroviral therapy. Indian J Med Microbiol 2017; 35:580-584. [PMID: 29405153 DOI: 10.4103/ijmm.ijmm_17_299] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Type III interferon is well known to have diverse antiviral and immunomodulatory activities. Studies describing the association of interleukin (IL)-28 polymorphisms in treatment-experienced HIV participants are limited. This study was aimed to determine the association of IL-28B gene polymorphisms with immunological recovery in HIV patients on 6-9 months of antiretroviral therapy (ART). METHODS Eighty treatment-naive HIV patients were recruited, of which 48 patients were followed up after 6-9 months of ART. Whole blood samples were collected before and after 6-9 months of ART. CD4, CD8 and CD3 counts were enumerated flow cytometry. IL-28B polymorphisms (rs12979860 and rs8099917) were profiled by polymerase chain reaction (PCR)-restriction fragment length polymorphism. The IL-28 mRNA and plasma HIV-1 viral load were estimated using real-time PCR and plasma IL-28 level by ELISA. RESULTS The CD4, CD4/CD3%, IL-28 mRNA and reversal of CD4/CD8 ratio were significantly increased following 6-9 months of ART (P < 0.01). The rs12979860 CC genotype and rs12979860:rs8099917 (CC: TT) haplotype showed significant association with higher CD4+ T-cell count amongst treatment-naive HIV-infected individuals (P < 0.05). In addition, there was a significant association of rs12979860 CC genotype with increase in CD4/CD3% following 6-9 months of ART. IL-28 mRNA showed correlation with the HIV-1 viral load, and there was a significant increase in the IL-28 mRNA expression following 6-9 months of ART. CONCLUSION Our preliminary findings suggest that IL-28 polymorphisms could influence both immunological recovery and therapeutic response in HIV infection. Hence, functional studies are warranted to understand the mechanistic basis of IL-28-mediated host genetic influence on HIV therapeutic response.
Collapse
Affiliation(s)
- B V Srinidhi
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, India
| | - G John Fletcher
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, India
| | | | - Priscilla Rupali
- Department of Medicine and Infectious Diseases, Christian Medical College, Vellore, Tamil Nadu, India
| | | | - J P Demosthenes
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, India
| | - O C Abraham
- Department of Medicine and Infectious Diseases, Christian Medical College, Vellore, Tamil Nadu, India
| | - Susanne A Pulimood
- Department of Dermatology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Grace Rebekah
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, India
| | - Rajesh Kannangai
- Department of Clinical Virology, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
13
|
Kah J, Volz T, Lütgehetmann M, Groth A, Lohse AW, Tiegs G, Sass G, Dandri M. Haem oxygenase-1 polymorphisms can affect HCV replication and treatment responses with different efficacy in humanized mice. Liver Int 2017; 37:1128-1137. [PMID: 27992676 DOI: 10.1111/liv.13347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 11/22/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND & AIMS Enhancement of host anti-oxidant enzymes, such as haemoxygenase-1, may attenuate virus-mediated hepatocyte injury, while the induction of HO-1 by cobalt-protoporphyrin-IX (CoPP) administration, as the application of its haem degradation product biliverdin (BV), was shown to hinder HCV replication in vitro. In addition, (GT)n -repeats length in the polymorphic region of the HO-1 promoter may affect HO-1 expression and responsiveness to infection and disease severity. Aim of this study was to investigate the antiviral and hepatoprotective effects of CoPP-mediated HO-1 induction, alone or in combination with interferon alpha (peg-IFNα), in HCV-infected mice harbouring hepatocytes from donors with different HO-1-promoter polymorphisms. METHODS Upon establishment of HCV infection, CoPP, BV and peg-IFNα were given alone or in combination. Viraemia changes and intrahepatic human gene expression were determined by qRT-PCR and immunohistochemistry. RESULTS CoPP administration increased human HO-1 expression and significantly reduced viraemia, although changes correlated with promoter length (Δ0.5log and Δ2log reduction with medium- and short-polymorphism respectively). Polymorphisms did not influence BV-mediated antiviral effects (Δ1log). Notably, HO-1 induction attenuated basal HCV-driven enhancement of interferon genes and pro-inflammatory cytokines, both in cells with short- or medium-polymorphisms. Moreover, simultaneous administration of CoPP and peg-IFNα reduced viraemia even stronger (median 3log), whereas 1log viraemia reduction was determined in mice receiving peg-IFNα monotherapy. CONCLUSIONS Although the protective function of HO-1 could be elicited in vivo with both host polymorphisms, the strength of HO-1 induction and suppression of HCV occurred in a polymorphism-dependent manner, indicating that host-genetic determinants may affect disease progression and infection outcome.
Collapse
Affiliation(s)
- Janine Kah
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tassilo Volz
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marc Lütgehetmann
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Institute of Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Groth
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ansgar W Lohse
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, Hamburg, Germany
| | - Gisa Tiegs
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriele Sass
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Maura Dandri
- I. Department of Medicine, Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, Hamburg, Germany
| |
Collapse
|
14
|
Eslam M, McLeod D, Kelaeng KS, Mangia A, Berg T, Thabet K, Irving WL, Dore GJ, Sheridan D, Grønbæk H, Abate ML, Hartmann R, Bugianesi E, Spengler U, Rojas A, Booth DR, Weltman M, Mollison L, Cheng W, Riordan S, Mahajan H, Fischer J, Nattermann J, Douglas MW, Liddle C, Powell E, Romero-Gomez M, George J. IFN-λ3, not IFN-λ4, likely mediates IFNL3-IFNL4 haplotype-dependent hepatic inflammation and fibrosis. Nat Genet 2017; 49:795-800. [PMID: 28394349 DOI: 10.1038/ng.3836] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/13/2017] [Indexed: 12/15/2022]
Abstract
Genetic variation in the IFNL3-IFNL4 (interferon-λ3-interferon-λ4) region is associated with hepatic inflammation and fibrosis. Whether IFN-λ3 or IFN-λ4 protein drives this association is not known. We demonstrate that hepatic inflammation, fibrosis stage, fibrosis progression rate, hepatic infiltration of immune cells, IFN-λ3 expression, and serum sCD163 levels (a marker of activated macrophages) are greater in individuals with the IFNL3-IFNL4 risk haplotype that does not produce IFN-λ4, but produces IFN-λ3. No difference in these features was observed according to genotype at rs117648444, which encodes a substitution at position 70 of the IFN-λ4 protein and reduces IFN-λ4 activity, or between patients encoding functionally defective IFN-λ4 (IFN-λ4-Ser70) and those encoding fully active IFN-λ4-Pro70. The two proposed functional variants (rs368234815 and rs4803217) were not superior to the discovery SNP rs12979860 with respect to liver inflammation or fibrosis phenotype. IFN-λ3 rather than IFN-λ4 likely mediates IFNL3-IFNL4 haplotype-dependent hepatic inflammation and fibrosis.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Duncan McLeod
- Department of Anatomical Pathology, Institute of Clinical Pathology and Medical Research (ICPMR), Westmead Hospital, Sydney, New South Wales, Australia
| | - Kebitsaone Simon Kelaeng
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Alessandra Mangia
- Division of Hepatology, Ospedale Casa Sollievo della Sofferenza, IRCCS, San Giovanni Rotondo, Italy
| | - Thomas Berg
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Clinic Leipzig, Leipzig, Germany
| | - Khaled Thabet
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
- Biochemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - William L Irving
- NIHR Biomedical Research Unit in Gastroenterology and the Liver, University of Nottingham, Nottingham, UK
| | - Gregory J Dore
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - David Sheridan
- Institute of Translational and Stratified Medicine, Plymouth University, Plymouth, UK
| | - Henning Grønbæk
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
| | - Maria Lorena Abate
- Division of Gastroenterology and Hepatology, Department of Medical Science, University of Turin, Turin, Italy
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Elisabetta Bugianesi
- Division of Gastroenterology and Hepatology, Department of Medical Science, University of Turin, Turin, Italy
| | - Ulrich Spengler
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Angela Rojas
- UCM IC Digestive Diseases and ciberehd. University Hospital Virgen del Rocio, Institute of Biomedicine of Seville, Seville, Spain
| | - David R Booth
- Institute of Immunology and Allergy Research, Westmead Hospital and Westmead Millennium Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Martin Weltman
- Department of Gastroenterology and Hepatology, Nepean Hospital, Sydney, New South Wales, Australia
| | - Lindsay Mollison
- Department of Gastroenterology and Hepatology, Fremantle Hospital, Fremantle, Western Australia, Australia
| | - Wendy Cheng
- Department of Gastroenterology and Hepatology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Stephen Riordan
- Gastrointestinal and Liver Unit, Prince of Wales Hospital and University of New South Wales, Sydney, New South Wales, Australia
| | - Hema Mahajan
- Department of Anatomical Pathology, Institute of Clinical Pathology and Medical Research (ICPMR), Westmead Hospital, Sydney, New South Wales, Australia
| | - Janett Fischer
- Section of Hepatology, Clinic for Gastroenterology and Rheumatology, University Clinic Leipzig, Leipzig, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Mark W Douglas
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
- Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney at Westmead Hospital, Westmead, New South Wales, Australia
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Elizabeth Powell
- University of Queensland, School of Medicine, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Manuel Romero-Gomez
- UCM IC Digestive Diseases and ciberehd. University Hospital Virgen del Rocio, Institute of Biomedicine of Seville, Seville, Spain
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Syedbasha M, Egli A. Interferon Lambda: Modulating Immunity in Infectious Diseases. Front Immunol 2017; 8:119. [PMID: 28293236 PMCID: PMC5328987 DOI: 10.3389/fimmu.2017.00119] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 01/25/2017] [Indexed: 12/22/2022] Open
Abstract
Interferon lambdas (IFN-λs; IFNL1-4) modulate immunity in the context of infections and autoimmune diseases, through a network of induced genes. IFN-λs act by binding to the heterodimeric IFN-λ receptor (IFNLR), activating a STAT phosphorylation-dependent signaling cascade. Thereby hundreds of IFN-stimulated genes are induced, which modulate various immune functions via complex forward and feedback loops. When compared to the well-characterized IFN-α signaling cascade, three important differences have been discovered. First, the IFNLR is not ubiquitously expressed: in particular, immune cells show significant variation in the expression levels of and susceptibilities to IFN-λs. Second, the binding affinities of individual IFN-λs to the IFNLR varies greatly and are generally lower compared to the binding affinities of IFN-α to its receptor. Finally, genetic variation in the form of a series of single-nucleotide polymorphisms (SNPs) linked to genes involved in the IFN-λ signaling cascade has been described and associated with the clinical course and treatment outcomes of hepatitis B and C virus infection. The clinical impact of IFN-λ signaling and the SNP variations may, however, reach far beyond viral hepatitis. Recent publications show important roles for IFN-λs in a broad range of viral infections such as human T-cell leukemia type-1 virus, rotaviruses, and influenza virus. IFN-λ also potentially modulates the course of bacterial colonization and infections as shown for Staphylococcus aureus and Mycobacterium tuberculosis. Although the immunological processes involved in controlling viral and bacterial infections are distinct, IFN-λs may interfere at various levels: as an innate immune cytokine with direct antiviral effects; or as a modulator of IFN-α-induced signaling via the suppressor of cytokine signaling 1 and the ubiquitin-specific peptidase 18 inhibitory feedback loops. In addition, the modulation of adaptive immune functions via macrophage and dendritic cell polarization, and subsequent priming, activation, and proliferation of pathogen-specific T- and B-cells may also be important elements associated with infectious disease outcomes. This review summarizes the emerging details of the IFN-λ immunobiology in the context of the host immune response and viral and bacterial infections.
Collapse
Affiliation(s)
- Mohammedyaseen Syedbasha
- Applied Microbiology Research, Department of Biomedicine, University of Basel , Basel , Switzerland
| | - Adrian Egli
- Applied Microbiology Research, Department of Biomedicine, University of Basel, Basel, Switzerland; Clinical Microbiology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
16
|
Yamagiwa Y, Asano M, Kawasaki Y, Korenaga M, Murata K, Kanto T, Mizokami M, Masaki N. Pretreatment serum levels of interferon-gamma-inducible protein-10 are associated with virologic response to telaprevir-based therapy. Cytokine 2016; 88:29-36. [PMID: 27541605 DOI: 10.1016/j.cyto.2016.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 07/03/2016] [Accepted: 07/05/2016] [Indexed: 01/16/2023]
Abstract
AIM Telaprevir (TVR) remarkably improves the efficacy of interferon treatment for chronic hepatitis C. Interleukin-28B (IL28B) genotype and interferon-gamma-inducible protein-10 (IP-10) level predict virologic response to peg-interferon (Peg-IFN)/ribavirin (RBV) therapy. We aimed to investigate the usefulness of pretreatment serum IP-10 levels and IL28B genotyping in predicting sustained virologic response (SVR) to TVR-based triple therapy. METHODS In this multi-center study, patients infected with hepatitis C virus genotype 1 with high viral load (⩾5.0logIU/mL) were treated with TVR for 12weeks and Peg-IFN/RBV for 24weeks in Japan. IL28B genotype, serum IP-10 levels, other clinical parameters, and drug dosages were assessed before treatment. RESULTS We included 121 patients who were treated with TVR for at least 8weeks and Peg-IFN/RBV for 24weeks. The median IP-10 levels were significantly lower in rapid virologic response (RVR) or SVR in the IL28B non-TT genotype group, with no significant difference in the TT genotype group. RVR rates were significantly lower in the group with higher serum IP-10 levels (>450pg/mL). In the non-TT IL28B genotype group, RVR and SVR rates were significantly lower in the group with higher IP-10 levels. SVR rates in the group with lower IP-10 levels (<450pg/mL) increased to 82% for those showing RVR, but reduced to 27% in the group with higher IP-10 levels for those not showing RVR. CONCLUSIONS Determination of serum IP-10 levels before treatment could be useful for predicting favorable virologic response to TVR-based triple therapy, especially in patients with IL28B non-TT genotype.
Collapse
Affiliation(s)
- Yoko Yamagiwa
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan.
| | - Mai Asano
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Youhei Kawasaki
- Department of Drug Evaluation & Informatics, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Shizuoka 422-8526, Japan
| | - Masaaki Korenaga
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Kazumoto Murata
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Tatsuya Kanto
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Masashi Mizokami
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Naohiko Masaki
- The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Japan
| |
Collapse
|
17
|
O'Connor KS, Read SA, Wang M, Schibeci S, Eslam M, Ong A, Weltman MD, Douglas MW, Mazzola A, Craxì A, Petta S, Stewart GJ, Liddle C, George J, Ahlenstiel G, Booth DR. IFNL3/4 genotype is associated with altered immune cell populations in peripheral blood in chronic hepatitis C infection. Genes Immun 2016; 17:328-334. [PMID: 27307212 PMCID: PMC5399140 DOI: 10.1038/gene.2016.27] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022]
Abstract
Single-nucleotide polymorphisms near the interferon lambda 3 (IFNL3) gene predict outcomes to infection and anti-viral treatment in hepatitis C virus (HCV) infection. To identify IFNL3 genotype effects on peripheral blood, we collected phenotype data on 400 patients with genotype 1 chronic hepatitis C (CHC). The IFNL3 responder genotype predicted significantly lower white blood cells (WBCs), as well as lower absolute numbers of monocytes, neutrophils and lymphocytes for both rs8099917 and rs12979860. We sought to define the WBC subsets driving this association using flow cytometry of 67 untreated CHC individuals. Genotype-associated differences were seen in the ratio of CD4CD45RO+ to CD4CD45RO-; CD8CD45RO+ to CD8CD45RO-, NK CD56 dim to bright and monocyte numbers and percentages. Whole blood expression levels of IFNL3, IFNLR1 (interferon lambda receptor 1), IFNLR1-mem (a membrane-associated receptor), IFNLR1-sol (a truncated soluble receptor), MxA and T- and NK (natural killer) cell transcription factors TBX21, GATA3, RORC, FOXP3 and EOMES in two subjects were also determined. CHC patients demonstrated endogenous IFN activation with higher levels of MxA, IFNLR1, IFNLR1-mem and IFNLR1-sol, and IFNL3 genotype-associated differences in transcription factors. Taken together, these data provide evidence of an IFNL3 genotype association with differences in monocyte, T- and NK cell levels in the peripheral blood of patients with CHC. This could underpin genotype associations with spontaneous and treatment-induced HCV clearance and hepatic necroinflammation.
Collapse
Affiliation(s)
- K S O'Connor
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - S A Read
- Storr Liver Centre, Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - M Wang
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - S Schibeci
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - M Eslam
- Storr Liver Centre, Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - A Ong
- Storr Liver Centre, Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
- Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney and Westmead Hospital, Sydney, NSW, Australia
| | - M D Weltman
- Department of Gastroenterology and Hepatology, Nepean Hospital, Sydney, NSW, Australia
| | - M W Douglas
- Storr Liver Centre, Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
- Centre for Infectious Diseases and Microbiology, Marie Bashir Institute for Infectious Diseases and Biosecurity, University of Sydney and Westmead Hospital, Sydney, NSW, Australia
| | - A Mazzola
- Sezione di Gastroenterologia, Di.Bi.M.I.S., University of Palermo, Palermo, Italy
| | - A Craxì
- Sezione di Gastroenterologia, Di.Bi.M.I.S., University of Palermo, Palermo, Italy
| | - S Petta
- Sezione di Gastroenterologia, Di.Bi.M.I.S., University of Palermo, Palermo, Italy
| | - G J Stewart
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| | - C Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - J George
- Storr Liver Centre, Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - G Ahlenstiel
- Storr Liver Centre, Westmead Institute for Medical Research and Westmead Hospital, University of Sydney, Sydney, NSW, Australia
| | - D R Booth
- Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
18
|
Krämer B, Finnemann C, Sastre B, Lutz P, Glässner A, Wolter F, Goeser F, Kokordelis P, Kaczmarek D, Nischalke HD, Strassburg CP, Spengler U, Nattermann J. IL-28B Genetic Variants Determine the Extent of Monocyte-Induced Activation of NK Cells in Hepatitis C. PLoS One 2016; 11:e0162068. [PMID: 27583440 PMCID: PMC5008784 DOI: 10.1371/journal.pone.0162068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 08/17/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Immuno-genetic studies suggest a functional link between NK cells and λ-IFNs. We recently showed that NK cells are negative for the IFN-λ receptor IFN-λR1 and do not respond to IFN-λ, suggesting a rather indirect association between IL-28B genotype and NK cell activity. METHODS A total of 75 HCV(+) patients and 67 healthy controls were enrolled into this study. IL-28B (rs12979860) and IFNL-4 (rs368234815) genotypes were determined by rtPCR. Total PBMC, monocytes, and NK cells were stimulated with IL-29, the TLR-7/8 agonist R848, or a combination of both. NK cell IFN-γ response was analysed by FACS. IL-12 and IL-18 secretion of monocytes was studied by ELISA. In blocking experiments anti-IL-12/anti-IL-18 were used. RESULTS Following stimulation of total PBMCs with R848 we found NK cell IFN- γ responses to vary with the IL-28B genotype, with carriers of a T/T genotype displaying the lowest frequency of IFN-γ(+)NK cells. When isolated NK cells were studied no such associations were observed, indicating an indirect association between IL-28B genotype and NK cell activity. Accordingly, we found R848-stimulated monocytes of patients with a T/T genotype to be significantly less effective in triggering NK cell IFN- γ production than monocytes from carriers of a non-T/T genotype. In line with these findings we observed monocytes from T/T patients to secrete significantly lower concentrations of IL-12 than monocytes from non-T/T individuals. CONCLUSIONS Our data indicate that monocytes from carriers of an IL-28B T/T genotype display a reduced ability to stimulate NK cell activity and, thus, provide a link between IL-28B genotype and NK functions.
Collapse
Affiliation(s)
- Benjamin Krämer
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Claudia Finnemann
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Beatriz Sastre
- Department of Infectious Diseases, Institute for Health Research (IRYCIS), University Hospital Ramón y Cajal, Madrid, Spain
- AIDS Research Network (RIS-RETICS), Madrid, Spain
| | - Philipp Lutz
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Andreas Glässner
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Franziska Wolter
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Felix Goeser
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Pavlos Kokordelis
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Dominik Kaczmarek
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Hans-Dieter Nischalke
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Christian P. Strassburg
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Ulrich Spengler
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| |
Collapse
|
19
|
Chinnaswamy S. Gene-disease association with human IFNL locus polymorphisms extends beyond hepatitis C virus infections. Genes Immun 2016; 17:265-75. [PMID: 27278127 PMCID: PMC7091887 DOI: 10.1038/gene.2016.24] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 04/01/2016] [Accepted: 05/06/2016] [Indexed: 12/25/2022]
Abstract
Interferon (IFN) lambda (IFN-λ or type III IFN) gene polymorphisms were discovered in the year 2009 to have a strong association with spontaneous and treatment-induced clearance of hepatitis C virus (HCV) infection in human hosts. This landmark discovery also brought renewed interest in type III IFN biology. After more than half a decade since this discovery, we now have reports that show that genetic association of IFNL gene polymorphisms in humans is not limited only to HCV infections but extends beyond, to include varied diseases such as non-alcoholic fatty liver disease, allergy and several other viral diseases including that caused by the human immunodeficiency virus. Notably, all these conditions have strong involvement of host innate immune responses. After the discovery of a deletion polymorphism that leads to the expression of a functional IFN-λ4 as the prime 'functional' variant, the relevance of other polymorphisms regulating the expression of IFN-λ3 is in doubt. Herein, I seek to critically address these issues and review the current literature to provide a framework to help further understanding of IFN-λ biology.
Collapse
Affiliation(s)
- S Chinnaswamy
- National Institute of Biomedical Genomics, Kalyani, West Bengal India
- Department of Clinical Immunology, Rheumatology and Allergy, Healthy Ageing Research Centre, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
20
|
Ferraris P, Chandra PK, Panigrahi R, Aboulnasr F, Chava S, Kurt R, Pawlotsky JM, Wilkens L, Osterlund P, Hartmann R, Balart LA, Wu T, Dash S. Cellular Mechanism for Impaired Hepatitis C Virus Clearance by Interferon Associated with IFNL3 Gene Polymorphisms Relates to Intrahepatic Interferon-λ Expression. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:938-51. [PMID: 26896692 PMCID: PMC5807932 DOI: 10.1016/j.ajpath.2015.11.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 11/13/2015] [Accepted: 11/24/2015] [Indexed: 12/12/2022]
Abstract
The single nucleotide polymorphism located within the IFNL3 (also known as IL28B) promoter is one of the host factors associated with hepatitis C virus (HCV) clearance by interferon (IFN)-α therapy; however the mechanism remains unknown. We investigated how IL28B gene polymorphism influences HCV clearance with infected primary human hepatocytes, liver biopsies, and hepatoma cell lines. Our study confirms that the rs12979860-T/T genotype has a strong correlation with ss469415590-ΔG/ΔG single nucleotide polymorphism that produces IFN-λ4 protein. We found that IFN-α and IFN-λ1 antiviral activity against HCV was impaired in IL28B T/T infected hepatocytes compared with C/C genotype. Western blot analysis showed that IL28B TT genotype hepatocytes expressed higher levels of IFN-λ proteins (IL28B, IL-29), preactivated IFN-stimulated gene (ISG) expression, and impaired Stat phosphorylation when stimulated with either IFN-α or IFN-λ1. Furthermore, we showed that silencing IFN-λ1 in T/T cell line reduced basal ISG expression and improved antiviral activity. Likewise, overexpression of IFN-λ (1 to 4) in C/C cells induced basal ISG expression and prevented IFN-α antiviral activity. We showed that IFN-λ4, produced at low level only in T/T cells induced expression of IL28B and IL-29 and prevented IFN-α antiviral activity in HCV cell culture. Our results suggest that IFN-λ4 protein expression associated with the IL28B-T/T variant preactivates the Janus kinase-Stat signaling, leading to impaired HCV clearance by both IFN-α and IFN-λ.
Collapse
Affiliation(s)
- Pauline Ferraris
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Partha K Chandra
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Rajesh Panigrahi
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Fatma Aboulnasr
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Srinivas Chava
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Ramazan Kurt
- Department of Medicine, Gastroenterology, and Hepatology, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Jean-Michel Pawlotsky
- Department of Molecular Virology and Immunology, Institut Mondor de la Recherche, Creteil, France
| | - Ludwig Wilkens
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Pamela Osterlund
- Department of Vaccination and Immune Protection Viral Infections, National Institute for Health and Welfare, Helsinki, Finland
| | - Rune Hartmann
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Luis A Balart
- Department of Medicine, Gastroenterology, and Hepatology, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Tong Wu
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana
| | - Srikanta Dash
- Department of Pathology and Laboratory Medicine, Tulane University Health Sciences Center, New Orleans, Louisiana; Department of Medicine, Gastroenterology, and Hepatology, Tulane University Health Sciences Center, New Orleans, Louisiana.
| |
Collapse
|
21
|
Resistance-Associated NS5A Variants of Hepatitis C Virus Are Susceptible to Interferon-Based Therapy. PLoS One 2015; 10:e0138060. [PMID: 26368554 PMCID: PMC4569333 DOI: 10.1371/journal.pone.0138060] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 08/25/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND & AIMS The presence of resistance-associated variants (RAVs) of hepatitis C virus (HCV) attenuates the efficacy of direct acting antivirals (DAAs). The objective of this study was to characterize the susceptibility of RAVs to interferon-based therapy. METHODS Direct and deep sequencing were performed to detect Y93H RAV in the NS5A region. Twenty nine genotype 1b patients with detectable RAV at baseline were treated by a combination of simeprevir, pegylated interferon and ribavirin. The longitudinal changes in the proportion of Y93H RAV during therapy and at breakthrough or relapse were determined. RESULTS By direct sequencing, Y93H RAV became undetectable or decreased in proportion at an early time point during therapy (within 7 days) in 57% of patients with both the Y93H variant and wild type virus at baseline when HCV RNA was still detectable. By deep sequencing, the proportion of Y93H RAV against Y93 wild type was 52.7% (5.8%- 97.4%) at baseline which significantly decreased to 29.7% (0.16%- 98.3%) within 7 days of initiation of treatment (p = 0.023). The proportion of Y93H RAV was reduced in 21 of 29 cases (72.4%) and a marked reduction of more than 10% was observed in 14 cases (48.7%). HCV RNA reduction was significantly greater for Y93H RAV (-3.65±1.3 logIU/mL/day) than the Y93 wild type (-3.35±1.0 logIU/mL/day) (p<0.001). CONCLUSION Y93H RAV is more susceptible to interferon-based therapy than the Y93 wild type.
Collapse
|
22
|
Zhang Y, Zhang D, Tu C, Zhou P, Zheng Y, Peng Z, Feng Y, Xiao S, Li Z. Wnt5a is involved in the pathogenesis of cutaneous lichen planus. Clin Exp Dermatol 2015; 40:659-664. [PMID: 25581355 DOI: 10.1111/ced.12561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2014] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cutaneous lichen planus (CLP) is a chronic inflammatory and immune-mediated disease. Wnt5a is one of the most extensively studied Wnt proteins, and has important functions in stimulating inflammation, cell proliferation, cell fate determination and cell differentiation. Wnt5a expression in CLP has not been comprehensively studied to date. AIM To determine the expression and distribution of Wnt5a in CLP. METHODS Skin samples were obtained from patients with CLP and healthy controls (HCs). The WNT5A gene was detected by real-time quantitative PCR, and Wnt5a protein by immunohistochemical analysis and western blotting. RESULTS WNT5A mRNA was upregulated in CLP samples compared with the HC skin samples (P < 0.001). Wnt5a protein was overexpressed in all layers of the epidermis and dermis in CLP lesions compared with HC skin (all P < 0.001). These results were confirmed by western blotting. CONCLUSIONS The data presented in this study suggest that Wnt5a pathway may play an important role in the pathogenesis of CLP.
Collapse
Affiliation(s)
- Y Zhang
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - D Zhang
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - C Tu
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - P Zhou
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - Y Zheng
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - Z Peng
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - Y Feng
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - S Xiao
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| | - Z Li
- Department of Dermatology, Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, PR China
| |
Collapse
|
23
|
Abad AA, Elst WVD, Molenberghs G. Validating predictors of therapeutic success: A causal inference approach. STAT MODEL 2015. [DOI: 10.1177/1471082x15586286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In personalized medicine medical decisions, practices and/or products are tailored to the individual patient. The idea is to provide the right patient with the right drug at the right dose at the right time. However, our current lack of ability to predict an individual patient's treatment success for most diseases and conditions is a major challenge to achieve the goal of personalized medicine. In the present work, we argue that many of the techniques often used to evaluate predictors of therapeutic success may not be able to answer the relevant scientific questions and we propose a new validation strategy based on causal inference. The methodology is illustrated using data from a clinical trial in opiate/heroin addiction. The user-friendly R library EffectTreat is provided to carry out the necessary calculations.
Collapse
|
24
|
Iijima S, Matsuura K, Watanabe T, Onomoto K, Fujita T, Ito K, Iio E, Miyaki T, Fujiwara K, Shinkai N, Kusakabe A, Endo M, Nojiri S, Joh T, Tanaka Y. Influence of genes suppressing interferon effects in peripheral blood mononuclear cells during triple antiviral therapy for chronic hepatitis C. PLoS One 2015; 10:e0118000. [PMID: 25706116 PMCID: PMC4338062 DOI: 10.1371/journal.pone.0118000] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/03/2015] [Indexed: 01/16/2023] Open
Abstract
The levels of expression of interferon-stimulated genes (ISGs) in liver are associated with response to treatment with pegylated interferon (PEG-IFN) plus ribavirin (RBV). However, associations between the responses of ISGs to IFN-based therapy and treatment efficacy or interleukin-28B (IL28B) genotype have not yet been determined. Therefore, we investigated the early responses of ISGs and interferon-lambdas (IFN-λs) in peripheral blood mononuclear cells (PBMCs) during PEG-IFN/RBV plus NS3/4 protease inhibitor (PI) therapy. We prospectively enrolled 50 chronic hepatitis C patients with HCV genotype 1, and collected PBMCs at baseline, 8 and 24 h after the initial administration of PEG-IFN/RBV/PI. Levels of mRNAs for selected ISGs and IFN-λs were evaluated by real-time PCR. All 31 patients with a favorable IL28B genotype and 13 of 19 with an unfavorable genotype achieved sustained virological responses (SVR). Levels of mRNA for A20, SOCS1, and SOCS3, known to suppress antiviral activity by interfering with the IFN signaling pathway, as well as IRF1 were significantly higher at 8 h in patients with an unfavorable IL28B genotype than in those with a favorable one (P = 0.007, 0.026, 0.0004, 0.0006, respectively), especially in the non-SVR group. Particularly, the fold-change of IRF1 at 8 h relative to baseline was significantly higher in non-SVR than in SVR cases with an unfavorable IL28B genotype (P = 0.035). In conclusion, levels of several mRNAs of genes suppressing antiviral activity in PBMCs during PEG-IFN/RBV/PI differed according to IL28B genotypes, paralleling treatment efficacy.
Collapse
Affiliation(s)
- Sayuki Iijima
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kentaro Matsuura
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Infectious Disease and Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD, United States of America
| | - Tsunamasa Watanabe
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Koji Onomoto
- Laboratory of Molecular Genetics, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Takashi Fujita
- Laboratory of Molecular Genetics, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Kyoko Ito
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Etsuko Iio
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Tomokatsu Miyaki
- Division of Internal Medicine, Toyokawa City Hospital, Toyokawa, Japan
| | - Kei Fujiwara
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Noboru Shinkai
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Atsunori Kusakabe
- Division of Gastroenterology, Nagoya Daini Red Cross Hospital, Nagoya, Japan
| | - Mio Endo
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shunsuke Nojiri
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Joh
- Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yasuhito Tanaka
- Department of Virology and Liver Unit, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
25
|
Griffiths SJ, Dunnigan CM, Russell CD, Haas JG. The Role of Interferon-λ Locus Polymorphisms in Hepatitis C and Other Infectious Diseases. J Innate Immun 2015; 7:231-42. [PMID: 25634147 PMCID: PMC6738896 DOI: 10.1159/000369902] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 11/15/2014] [Indexed: 12/19/2022] Open
Abstract
Since its discovery in 2003, the type III interferon-λ (IFN-λ) family has been found to contribute significantly to the host response to infection. Whilst IFN-λ shares many features with type I IFN induction and signalling pathways, the tissue-specific restricted expression of its receptor, IL28RA, makes IFN-λ a major mediator of host innate immunity in tissues and organs with a high epithelial cell content. Host susceptibility and responses to infection are known to be heterogeneous, and the identification of common genetic variants linked to disease outcome by genome-wide association studies (GWAS) has underscored the significance of host polymorphisms in responses to infection. Several such GWAS have highlighted the IFN-λ locus on chromosome 19q13 as an area of genetic variation significantly associated with hepatitis C virus (HCV) infection, and the rs12979860 genotype can be used in clinical practice as a biomarker for predicting a successful response to treatment with pegylated IFN and ribavarin. Here, we discuss IFN-λ genetic polymorphisms and their role in HCV and other infectious diseases as well as their potential impact on clinical diagnostics, patient stratification and therapy. Finally, the broader role of IFN-λ in the immunopathogenesis of non-infectious inflammatory diseases is considered.
Collapse
Affiliation(s)
- Samantha J Griffiths
- Division of Infection and Pathway Medicine, University of Edinburgh Medical School, Edinburgh, UK
| | | | | | | |
Collapse
|
26
|
O’Connor KS, George J, Booth D, Ahlenstiel G. Dendritic cells in hepatitis C virus infection: key players in the IFNL3-genotype response. World J Gastroenterol 2014; 20:17830-8. [PMID: 25548481 PMCID: PMC4273133 DOI: 10.3748/wjg.v20.i47.17830] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 07/14/2014] [Accepted: 07/24/2014] [Indexed: 02/06/2023] Open
Abstract
Recently, single nucleotide polymorphisms, in the vicinity of the interferon lambda 3 (IFNL3) gene have been identified as the strongest predictor of spontaneous and treatment induced clearance of hepatitis C virus (HCV) infection. Since then, increasing evidence has implicated the innate immune response in mediating the IFNL3 genotype effect. Dendritic cells (DCs) are key to the host immune response in HCV infection and their vital role in the IFNL3 genotype effect is emerging. Reports have identified subclasses of DCs, particularly myeloid DC2s and potentially plasmacytoid DCs as the major producers of IFNL3 in the setting of HCV infection. Given the complexities of dendritic cell biology and the conflicting current available data, this review aims to summarize what is currently known regarding the role of dendritic cells in HCV infection and to place it into context of what is know about lambda interferons and dendritic cells in general.
Collapse
|
27
|
Egli A, Santer DM, O'Shea D, Barakat K, Syedbasha M, Vollmer M, Baluch A, Bhat R, Groenendyk J, Joyce MA, Lisboa LF, Thomas BS, Battegay M, Khanna N, Mueller T, Tyrrell DLJ, Houghton M, Humar A, Kumar D. IL-28B is a key regulator of B- and T-cell vaccine responses against influenza. PLoS Pathog 2014; 10:e1004556. [PMID: 25503988 PMCID: PMC4263767 DOI: 10.1371/journal.ppat.1004556] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 11/05/2014] [Indexed: 12/11/2022] Open
Abstract
Influenza is a major cause of morbidity and mortality in immunosuppressed persons, and vaccination often confers insufficient protection. IL-28B, a member of the interferon (IFN)-λ family, has variable expression due to single nucleotide polymorphisms (SNPs). While type-I IFNs are well known to modulate adaptive immunity, the impact of IL-28B on B- and T-cell vaccine responses is unclear. Here we demonstrate that the presence of the IL-28B TG/GG genotype (rs8099917, minor-allele) was associated with increased seroconversion following influenza vaccination (OR 1.99 p = 0.038). Also, influenza A (H1N1)-stimulated T- and B-cells from minor-allele carriers showed increased IL-4 production (4-fold) and HLA-DR expression, respectively. In vitro, recombinant IL-28B increased Th1-cytokines (e.g. IFN-γ), and suppressed Th2-cytokines (e.g. IL-4, IL-5, and IL-13), H1N1-stimulated B-cell proliferation (reduced 70%), and IgG-production (reduced>70%). Since IL-28B inhibited B-cell responses, we designed antagonistic peptides to block the IL-28 receptor α-subunit (IL28RA). In vitro, these peptides significantly suppressed binding of IFN-λs to IL28RA, increased H1N1-stimulated B-cell activation and IgG-production in samples from healthy volunteers (2-fold) and from transplant patients previously unresponsive to vaccination (1.4-fold). Together, these findings identify IL-28B as a key regulator of the Th1/Th2 balance during influenza vaccination. Blockade of IL28RA offers a novel strategy to augment vaccine responses.
Collapse
Affiliation(s)
- Adrian Egli
- Infection Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Deanna M. Santer
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Daire O'Shea
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- Division of Infectious Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Khaled Barakat
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Pharmacy, University of Alberta, Canada
| | | | - Madeleine Vollmer
- Infection Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Aliyah Baluch
- Division of Infectious Diseases, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Rakesh Bhat
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Jody Groenendyk
- Department of Biochemistry, School of Translational Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Michael A. Joyce
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Luiz F. Lisboa
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Brad S. Thomas
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Manuel Battegay
- Infection Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Basel, Switzerland
| | - Nina Khanna
- Infection Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Basel, Switzerland
| | - Thomas Mueller
- Division of Nephrology, University Hospital of Zurich, Zurich, Switzerland
| | - D. Lorne J. Tyrrell
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Michael Houghton
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Alberta, Canada
| | - Atul Humar
- Department of Medicine and Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Deepali Kumar
- Department of Medicine and Multi-Organ Transplant Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
28
|
Heim MH, Thimme R. Innate and adaptive immune responses in HCV infections. J Hepatol 2014; 61:S14-25. [PMID: 25443342 DOI: 10.1016/j.jhep.2014.06.035] [Citation(s) in RCA: 205] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 06/29/2014] [Accepted: 06/30/2014] [Indexed: 12/21/2022]
Abstract
Hepatitis C virus has been identified a quarter of a decade ago as a leading cause of chronic viral hepatitis that can lead to cirrhosis and hepatocellular carcinoma. Only a minority of patients can clear the virus spontaneously during acute infection. Elimination of HCV during acute infection correlates with a rapid induction of innate, especially interferon (IFN) induced genes, and a delayed induction of adaptive immune responses. However, the majority of patients is unable to clear the virus and develops viral persistence in face of an ongoing innate and adaptive immune response. The virus has developed several strategies to escape these immune responses. For example, to escape innate immunity, the HCV NS3/4A protease can efficiently cleave and inactivate two important signalling molecules in the sensory pathways that react to HCV pathogen-associated molecular patterns (PAMPs) to induce IFNs, i.e., the mitochondrial anti-viral signalling protein (MAVS) and the Toll-IL-1 receptor-domain-containing adaptor-inducing IFN-β (TRIF). Despite these escape mechanisms, IFN-stimulated genes (ISGs) are induced in a large proportion of patients with chronic infection. Of note, chronically HCV infected patients with constitutive IFN-stimulated gene (ISG) expression have a poor response to treatment with pegylated IFN-α (PegIFN-α) and ribavirin. The mechanisms that protect HCV from IFN-mediated innate immune reactions are not entirely understood, but might involve blockade of ISG protein translation at the ribosome, localization of viral replication to cell compartments that are not accessible to anti-viral IFN-stimulated effector systems, or direct antagonism of effector systems by viral proteins. Escape from adaptive immune responses can be achieved by emergence of viral escape mutations that avoid recognition by antibodies and T cells. In addition, chronic infection is characterized by the presence of functionally and phenotypically altered NK and T cell responses that are unable to clear the virus but most likely contribute to the ongoing liver disease. In this review, we will summarize current knowledge about the role of innate and adaptive immune responses in determining the outcome of HCV infection.
Collapse
Affiliation(s)
- Markus H Heim
- Division of Gastroenterology and Hepatology, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Department of Biomedicine, University of Basel, Hebelstrasse 20, 4031 Basel, Switzerland.
| | - Robert Thimme
- Department of Medicine, Clinic for Gastroenterology, Hepatology, Endocrinology, Infectious Diseases, University Hospital Freiburg, Freiburg, Germany.
| |
Collapse
|
29
|
Shirasaki T, Honda M, Shimakami T, Murai K, Shiomoto T, Okada H, Takabatake R, Tokumaru A, Sakai Y, Yamashita T, Lemon SM, Murakami S, Kaneko S. Impaired interferon signaling in chronic hepatitis C patients with advanced fibrosis via the transforming growth factor beta signaling pathway. Hepatology 2014; 60:1519-30. [PMID: 24962339 DOI: 10.1002/hep.27277] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2014] [Accepted: 06/20/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED Malnutrition in the advanced fibrosis stage of chronic hepatitis C (CH-C) impairs interferon (IFN) signaling by inhibiting mammalian target of rapamycin complex 1 (mTORC1) signaling. However, the effect of profibrotic signaling on IFN signaling is not known. Here, the effect of transforming growth factor (TGF)-β signaling on IFN signaling and hepatitis C virus (HCV) replication was examined in Huh-7.5 cells by evaluating the expression of forkhead box O3A (Foxo3a), suppressor of cytokine signaling 3 (Socs3), c-Jun, activating transcription factor 2, ras homolog enriched in brain, and mTORC1. The findings were confirmed in liver tissue samples obtained from 91 patients who received pegylated-IFN and ribavirin combination therapy. TGF-β signaling was significantly up-regulated in the advanced fibrosis stage of CH-C. A significant positive correlation was observed between the expression of TGF-β2 and mothers against decapentaplegic homolog 2 (Smad2), Smad2 and Foxo3a, and Foxo3a and Socs3 in the liver of CH-C patients. In Huh-7.5 cells, TGF-β1 activated the Foxo3a promoter through an AP1 binding site; the transcription factor c-Jun was involved in this activation. Foxo3a activated the Socs3 promoter and increased HCV replication. TGF-β1 also inhibited mTORC1 and IFN signaling. Interestingly, c-Jun and TGF-β signaling was up-regulated in treatment-resistant IL28B minor genotype patients (TG/GG at rs8099917), especially in the early fibrosis stage. Branched chain amino acids or a TGF-β receptor inhibitor canceled these effects and showed an additive effect on the anti-HCV activity of direct-acting antiviral drugs (DAAs). CONCLUSION Blocking TGF-β signaling could potentiate the antiviral efficacy of IFN- and/ or DAA-based treatment regimens and would be useful for the treatment of difficult-to-cure CH-C patients.
Collapse
Affiliation(s)
- Takayoshi Shirasaki
- Department of Gastroenterology, Kanazawa University Graduate School of Medicine, Kanazawa, Japan; Department of Advanced Medical Technology, Kanazawa University Graduate School of Health Medicine, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Meissner EG, Wu D, Osinusi A, Bon D, Virtaneva K, Sturdevant D, Porcella S, Wang H, Herrmann E, McHutchison J, Suffredini AF, Polis M, Hewitt S, Prokunina-Olsson L, Masur H, Fauci AS, Kottilil S. Endogenous intrahepatic IFNs and association with IFN-free HCV treatment outcome. J Clin Invest 2014; 124:3352-63. [PMID: 24983321 PMCID: PMC4109554 DOI: 10.1172/jci75938] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 05/19/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND. Hepatitis C virus (HCV) infects approximately 170 million people worldwide and may lead to cirrhosis and hepatocellular carcinoma in chronically infected individuals. Treatment is rapidly evolving from IFN-α-based therapies to IFN-α-free regimens that consist of directly acting antiviral agents (DAAs), which demonstrate improved efficacy and tolerability in clinical trials. Virologic relapse after DAA therapy is a common cause of treatment failure; however, it is not clear why relapse occurs or whether certain individuals are more prone to recurrent viremia. METHODS. We conducted a clinical trial using the DAA sofosbuvir plus ribavirin (SOF/RBV) and performed detailed mRNA expression analysis in liver and peripheral blood from patients who achieved either a sustained virologic response (SVR) or relapsed. RESULTS. On-treatment viral clearance was accompanied by rapid downregulation of IFN-stimulated genes (ISGs) in liver and blood, regardless of treatment outcome. Analysis of paired pretreatment and end of treatment (EOT) liver biopsies from SVR patients showed that viral clearance was accompanied by decreased expression of type II and III IFNs, but unexpectedly increased expression of the type I IFN IFNA2. mRNA expression of ISGs was higher in EOT liver biopsies of patients who achieved SVR than in patients who later relapsed. CONCLUSION. These results suggest that restoration of type I intrahepatic IFN signaling by EOT may facilitate HCV eradication and prevention of relapse upon withdrawal of SOF/RBV. TRIAL REGISTRATION. ClinicalTrials.gov NCT01441180.
Collapse
Affiliation(s)
- Eric G. Meissner
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - David Wu
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Anu Osinusi
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Dimitra Bon
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Kimmo Virtaneva
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Dan Sturdevant
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Steve Porcella
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Honghui Wang
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Eva Herrmann
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - John McHutchison
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Anthony F. Suffredini
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Michael Polis
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Stephen Hewitt
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Ludmila Prokunina-Olsson
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Henry Masur
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Anthony S. Fauci
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| | - Shyamasundaran Kottilil
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, Maryland, USA. Division of Infectious Diseases, Institute of Human Virology, University of Maryland Medical School, Baltimore, Maryland, USA. Institute of Biostatistics and Mathematical Modeling, Johann Wolfgang Goethe University, Frankfurt, Germany. Genomics Unit, Research Technologies Section, Rocky Mountain Laboratories, NIAID, NIH, Hamilton, Montana, USA. Critical Care Medicine Department, Clinical Center, NIH, Bethesda, Maryland, USA. Gilead Sciences, Foster City, California, USA. Department of Pathology, NCI, NIH, Bethesda, Maryland, USA. Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, NCI, NIH, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Egli A, Santer DM, O'Shea D, Tyrrell DL, Houghton M. The impact of the interferon-lambda family on the innate and adaptive immune response to viral infections. Emerg Microbes Infect 2014; 3:e51. [PMID: 26038748 PMCID: PMC4126180 DOI: 10.1038/emi.2014.51] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/06/2014] [Accepted: 05/20/2014] [Indexed: 12/12/2022]
Abstract
Type-III interferons (IFN-λ, IFNL) are the most recently described family of IFNs. This family of innate cytokines are increasingly being ascribed pivotal roles in host-pathogen interactions. Herein, we will review the accumulating evidence detailing the immune biology of IFNL during viral infection, and the implications of this novel information on means to advance the development of therapies and vaccines against existing and emerging pathogens. IFNLs exert antiviral effects via induction of IFN-stimulated genes. Common single nucleotide polymorphisms (SNPs) in the IFNL3, IFNL4 and the IFNL receptor α-subunit genes have been strongly associated with IFN-α-based treatment of chronic hepatitis C virus infection. The clinical impact of these SNPs may be dependent on the status of viral infection (acute or chronic) and the potential to develop viral resistance. Another important function of IFNLs is macrophage and dendritic cell polarization, which prime helper T-cell activation and proliferation. It has been demonstrated that IFNL increase Th1- and reduce Th2-cytokines. Therefore, can such SNPs affect the IFNL signaling and thereby modulate the Th1/Th2 balance during infection? In turn, this may influence the subsequent priming of cytotoxic T cells versus antibody-secreting B cells, with implications for the breadth and durability of the host response.
Collapse
Affiliation(s)
- Adrian Egli
- Infection Biology, Department of Biomedicine, University Hospital of Basel , 4031 Basel, Switzerland ; Clinical Microbiology, University Hospital of Basel , 4031 Basel, Switzerland
| | - Deanna M Santer
- Department of Medical Microbiology and Immunology, and Li Ka Shing Institute of Virology, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - Daire O'Shea
- Department of Medical Microbiology and Immunology, and Li Ka Shing Institute of Virology, University of Alberta , Edmonton, Alberta T6G 2E1, Canada ; Division of Infectious Diseases, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - D Lorne Tyrrell
- Department of Medical Microbiology and Immunology, and Li Ka Shing Institute of Virology, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| | - Michael Houghton
- Department of Medical Microbiology and Immunology, and Li Ka Shing Institute of Virology, University of Alberta , Edmonton, Alberta T6G 2E1, Canada
| |
Collapse
|
32
|
Stättermayer AF, Scherzer T, Beinhardt S, Rutter K, Hofer H, Ferenci P. Review article: genetic factors that modify the outcome of viral hepatitis. Aliment Pharmacol Ther 2014; 39:1059-1070. [PMID: 24654629 PMCID: PMC7159786 DOI: 10.1111/apt.12717] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 12/10/2013] [Accepted: 03/01/2014] [Indexed: 12/16/2022]
Abstract
BACKGROUND Genetic factors can play an important role for treatment response and disease progression in chronic viral hepatitis. AIM To review the influence of host genetic factors on the clinical course as well as on treatment response in patients with viral hepatitis. METHODS Review of the literature. RESULTS A landmark genome-wide association study (GWAS) identified polymorphisms in the IL28B gene on chromosome 19 (19q13.13) associated with response to therapy with pegylated interferon-α (PEG-IFN) and ribavirin (RBV) and spontaneous viral clearance in acute hepatitis C. Furthermore, IL28B genotype is associated with changes of lipid metabolism and insulin resistance. A further GWAS demonstrated that ITPA genetic variants protect HCV genotype 1 patients from RBV-induced anaemia. Another polymorphism in the patatin-like phospholipase domain containing 3 (PNPLA3) is associated with hepatic steatosis. Difficult-to-treat hepatitis C patients homozygous for GG had an up to five-fold lower chance of viral clearance on PEG/RBV than non-GG patients. In chronic hepatitis B patients treated with PEG-IFN several retrospective analyses of IL28B rs12980275 and rs12979860 genotypes yielded conflicting results which can be explained by the heterogeneity between the study populations. Some variants of the HLA-DP locus (HLA-DPA1 A allele and HLA-DPB1) protect against progression of chronic hepatitis B infection. CONCLUSIONS The determination of IL28B polymorphisms may be useful to individualise treatment options when using PEG/RBV based therapies for chronic hepatitis C infection. In contrast, so far identified genetic factors play only a minor role in chronic hepatitis B infection.
Collapse
Affiliation(s)
- A. F. Stättermayer
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - T. Scherzer
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - S. Beinhardt
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - K. Rutter
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - H. Hofer
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria
| | - P. Ferenci
- Division of Gastroenterology and HepatologyDepartment of Internal Medicine IIIMedical University of ViennaViennaAustria
| |
Collapse
|
33
|
O'Brien TR, Prokunina-Olsson L, Donnelly RP. IFN-λ4: the paradoxical new member of the interferon lambda family. J Interferon Cytokine Res 2014; 34:829-38. [PMID: 24786669 DOI: 10.1089/jir.2013.0136] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Interferons (IFNs) are generally considered antiviral cytokines, yet the newly discovered IFN-λ4 is linked with the failure to clear hepatitis C virus (HCV) infection either spontaneously or in response to treatment. IFN-λ4 can be generated only by individuals who carry the IFNL4-ΔG allele (rs368234815), which is the strongest known host factor for predicting clearance of HCV. The ancestral IFNL4-ΔG allele is the major variant in Africans while the minor variant in Asians, suggesting very strong negative genetic selection for this allele-most likely driven by an infectious agent other than HCV. IFN-λ4 most closely resembles IFN-λ3, but these proteins share only 29% amino-acid identity, and, in contrast to IFN-λ3, IFN-λ4 is only weakly secreted. Nevertheless, IFN-λ4 signals through the IFN-λ receptor complex and induces expression of IFN-stimulated genes via the Janus kinase-signal transducer and activator of transcription signaling pathway. Although the IFNL4-ΔG variant is strongly associated with the failure to clear HCV infection, HCV-infected patients who carry this allele have lower baseline HCV RNA levels in the absence of treatment. Resolving the paradoxical functions of IFN-λ4, which appears to induce antiviral activity yet impair effective clearance of HCV, may yield critical new insights into the immunologic response to HCV infection and IFN biology.
Collapse
Affiliation(s)
- Thomas R O'Brien
- 1 Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute , National Institutes of Health, Bethesda, Maryland
| | | | | |
Collapse
|
34
|
Hepatitis C virus and human miR-122: insights from the bench to the clinic. Curr Opin Virol 2014; 7:11-8. [PMID: 24721497 DOI: 10.1016/j.coviro.2014.03.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 03/07/2014] [Accepted: 03/10/2014] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that function as part of RNA-induced silencing complexes that repress the expression of target genes. Over the past few years, miRNAs have been found to mediate complex regulation of a wide variety of mammalian viral infections, including Hepatitis C virus (HCV) infection. Here, we focus on a highly abundant, liver-specific miRNA, miR-122. In a unique and unusual interaction, miR-122 binds to two sites in the 5' untranslated region (UTR) of the HCV genome and promotes viral RNA accumulation. We will discuss what has been learned about this important interaction to date, provide insights into how miR-122 is able to modulate HCV RNA accumulation, and how miR-122 might be exploited for antiviral intervention.
Collapse
|