1
|
Napodano C, Ciasca G, Chiusolo P, Pocino K, Gragnani L, Stefanile A, Gulli F, Lorini S, Minnella G, Fosso F, Di Santo R, Romanò S, Basile V, De Stefano V, Rapaccini GL, Zignego AL, Di Stasio E, Marino M, Basile U. Serological and Molecular Characterization of Hepatitis C Virus-Related Cryoglobulinemic Vasculitis in Patients without Cryoprecipitate. Int J Mol Sci 2023; 24:11602. [PMID: 37511357 PMCID: PMC10380893 DOI: 10.3390/ijms241411602] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Prolonged B cells stimulation due to the Hepatitis C virus (HCV) can result in autoimmunity, stigmatized by rising levels of cryoglobulins (CGs), the rheumatoid factor (RF), and free light chains (FLC) of immunoglobulins (Ig) associated with a range of symptoms, from their absence to severe cryoglobulinemic vasculitis and lymphoma. Here, we aimed to identify an immunological signature for the earliest stages of vasculitis when cryoprecipitate is still not detectable. We firstly analyzed the IgG subclasses, FLC, and RF in 120 HCV-RNA-positive patients divided into four groups according to the type of cryoprecipitate and symptoms: 30 asymptomatic without cryoprecipitate (No Cryo), 30 with vasculitis symptoms but without CGs that we supposed were circulating but still not detectable (Circulating), 30 type II and 30 type III mixed cryoglobulinemia (Cryo II and Cryo III, respectively). Our results revealed that patients with supposed circulating CGs displayed a pattern of serological parameters that closely resembled Cryo II and Cryo III, with a stronger similarity to Cryo II. Accordingly, we analyzed the groups of Circulating and Cryo II for their immunoglobulin heavy chain (IgH) and T-cell receptor (TCR) gene rearrangements, finding a similar mixed distribution of monoclonal, oligoclonal, and polyclonal responses compared to a control group of ten HCV-RNA-negative patients recovered from infection, who displayed a 100% polyclonal response. Our results strengthened the hypothesis that circulating CGs are the origin of symptoms in HCV-RNA-positive patients without cryoprecipitate and demonstrated that an analysis of clonal IGH and TCR rearrangements is the best option for the early diagnosis of extrahepatic complications.
Collapse
Affiliation(s)
- Cecilia Napodano
- Department of Laboratory Medicine and Pathology, S. Agostino Estense Hospital, 41126 Modena, Italy;
| | - Gabriele Ciasca
- Sezione di Fisica, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (R.D.S.); (S.R.)
- Fondazione Policlinico Universitario “A. Gemelli” I.R.C.C.S., 00168 Rome, Italy; (P.C.); (G.M.); (F.F.); (V.D.S.); (G.L.R.)
| | - Patrizia Chiusolo
- Fondazione Policlinico Universitario “A. Gemelli” I.R.C.C.S., 00168 Rome, Italy; (P.C.); (G.M.); (F.F.); (V.D.S.); (G.L.R.)
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Krizia Pocino
- Unità Operativa Complessa di Patologia Clinica, Ospedale Generale di Zona San Pietro Fatebenefratelli, 00189 Rome, Italy; (K.P.); (A.S.)
| | - Laura Gragnani
- Department of Translation Research and New Technologies in Medicine and Surgery, Università di Pisa, 56126 Pisa, Italy;
| | - Annunziata Stefanile
- Unità Operativa Complessa di Patologia Clinica, Ospedale Generale di Zona San Pietro Fatebenefratelli, 00189 Rome, Italy; (K.P.); (A.S.)
| | - Francesca Gulli
- Unit of Clinical Pathology, Bambino Gesù Children’s Hospital I.R.C.C.S., 00165 Rome, Italy;
| | - Serena Lorini
- Department of Experimental and Clinical Medicine, Interdepartmental Centre MASVE, University of Florence, 50121 Florence, Italy; (S.L.); (A.L.Z.)
| | - Gessica Minnella
- Fondazione Policlinico Universitario “A. Gemelli” I.R.C.C.S., 00168 Rome, Italy; (P.C.); (G.M.); (F.F.); (V.D.S.); (G.L.R.)
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Federica Fosso
- Fondazione Policlinico Universitario “A. Gemelli” I.R.C.C.S., 00168 Rome, Italy; (P.C.); (G.M.); (F.F.); (V.D.S.); (G.L.R.)
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Riccardo Di Santo
- Sezione di Fisica, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (R.D.S.); (S.R.)
| | - Sabrina Romanò
- Sezione di Fisica, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (G.C.); (R.D.S.); (S.R.)
| | - Valerio Basile
- Clinical Pathology Unit and Cancer Biobank, Department of Research and Advanced Technologies, I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Valerio De Stefano
- Fondazione Policlinico Universitario “A. Gemelli” I.R.C.C.S., 00168 Rome, Italy; (P.C.); (G.M.); (F.F.); (V.D.S.); (G.L.R.)
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, 00168 Roma, Italy
| | - Gian Ludovico Rapaccini
- Fondazione Policlinico Universitario “A. Gemelli” I.R.C.C.S., 00168 Rome, Italy; (P.C.); (G.M.); (F.F.); (V.D.S.); (G.L.R.)
| | - Anna Linda Zignego
- Department of Experimental and Clinical Medicine, Interdepartmental Centre MASVE, University of Florence, 50121 Florence, Italy; (S.L.); (A.L.Z.)
| | - Enrico Di Stasio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
| | - Mariapaola Marino
- Fondazione Policlinico Universitario “A. Gemelli” I.R.C.C.S., 00168 Rome, Italy; (P.C.); (G.M.); (F.F.); (V.D.S.); (G.L.R.)
- Sezione di Patologia Generale, Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Umberto Basile
- Dipartimento di Patologia Clinica, Ospedale Santa Maria Goretti, A.U.S.L. Latina, 04100 Latina, Italy;
| |
Collapse
|
2
|
He Y, Dang S, Ma W, Chen L, Zhang R, Mei S, Wei X, Lv Q, Peng B, Sun Y, Kong D, Chen J, Li S, Tang X, Lu Q, Zhu C, Chen Z, Wan J, Zou X, Li M, Feng T, Ren L, Wang J. Temporal dynamics of SARS-CoV-2 genome mutations that occurred in vivo on an aircraft. BIOSAFETY AND HEALTH 2023; 5:62-67. [PMID: 36320662 PMCID: PMC9613807 DOI: 10.1016/j.bsheal.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
We analyzed variations in the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genome during a flight-related cluster outbreak of coronavirus disease 2019 (COVID-19) in Shenzhen, China, to explore the characteristics of SARS-CoV-2 transmission and intra-host single nucleotide variations (iSNVs) in a confined space. Thirty-three patients with COVID-19 were sampled, and 14 were resampled 3-31 days later. All 47 nasopharyngeal swabs were deep-sequenced. iSNVs and similarities in the consensus genome sequence were analyzed. Three SARS-CoV-2 variants of concern, Delta (n = 31), Beta (n = 1), and C.1.2 (n = 1), were detected among the 33 patients. The viral genome sequences from 30 Delta-positive patients had similar SNVs; 14 of these patients provided two successive samples. Overall, the 47 sequenced genomes contained 164 iSNVs. Of the 14 paired (successive) samples, the second samples (T2) contained more iSNVs (median: 3; 95% confidence interval [95% CI]: 2.77-10.22) than did the first samples (T1; median: 2; 95% CI: 1.63-3.74; Wilcoxon test, P = 0.021). 38 iSNVs were detected in T1 samples, and only seven were also detectable in T2 samples. Notably, T2 samples from two of the 14 paired samples had additional mutations than the T1 samples. The iSNVs of the SARS-CoV-2 genome exhibited rapid dynamic changes during a flight-related cluster outbreak event. Intra-host diversity increased gradually with time, and new site mutations occurred in vivo without a population transmission bottleneck. Therefore, we could not determine the generational relationship from the mutation site changes alone.
Collapse
Affiliation(s)
- Yaqing He
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Shengyuan Dang
- National Health Commission of the People's Republic of China Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Wentai Ma
- University of Chinese Academy of Sciences, Beijing 100190, China
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing 100101, China
| | - Long Chen
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Renli Zhang
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Shujiang Mei
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xinyi Wei
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Qiuying Lv
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Bo Peng
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Ying Sun
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Dongfeng Kong
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Jiancheng Chen
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Shimin Li
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xiujuan Tang
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Qingju Lu
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Can Zhu
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Zhigao Chen
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Jia Wan
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Xuan Zou
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Mingkun Li
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing 100101, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Tiejiang Feng
- Shenzhen Research Center for Communicable Disease Control and Prevention, Chinese Academy of Medical Sciences, Shenzhen 518055, China
- Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Lili Ren
- National Health Commission of the People's Republic of China Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jianwei Wang
- National Health Commission of the People's Republic of China Key Laboratory of Systems Biology of Pathogens and Christophe Mérieux Laboratory, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- Key Laboratory of Respiratory Disease Pathogenomics, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
3
|
Weigand K, Peschel G, Grimm J, Luu K, Schacherer D, Wiest R, Müller M, Schwarz H, Buechler C. Soluble CD137 is a novel serum marker of liver cirrhosis in patients with hepatitis C and alcohol-associated disease etiology. Eur J Immunol 2021; 52:633-645. [PMID: 34914098 DOI: 10.1002/eji.202149488] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/24/2021] [Accepted: 12/10/2021] [Indexed: 11/06/2022]
Abstract
Defective T-cell functions play a role in the persistence of HCV infection. Activated T cells express CD137, which costimulates antivirus T-cell responses, and this activity is antagonized by soluble CD137 (sCD137). Here, we show that in sera of 81 patients with chronic HCV, sCD137 levels did not correlate with measures of viral infection, and did not decline after virus eradication using direct-acting antivirals. Thus, serum sCD137 was similar in patients infected with HCV and in uninfected controls. Of note, in HCV patients with liver cirrhosis and patients with mostly alcohol-associated liver cirrhosis, sCD137 was increased. A negative association of sCD137 and albumin existed in both cohorts. sCD137 concentrations were similar in hepatic and portal vein blood excluding the liver as the origin of higher levels. Recombinant sCD137 reduced Th1 and Th2 but not Th17 cell polarization in vitro, and accordingly lowered IFN-γ, TNF, and IL-13 in cell media. Serum sCD137 is associated with inflammatory states, and positively correlated with serum TNF in cirrhotic HCV patients following virus eradication. Our study argues against a role of sCD137 in HCV infection and suggests a function of sCD137 in liver cirrhosis, which yet has to be defined.
Collapse
Affiliation(s)
- Kilian Weigand
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Georg Peschel
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Jonathan Grimm
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Khang Luu
- Department of Physiology and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Doris Schacherer
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Reiner Wiest
- Department of Visceral Surgery and Medicine, University Inselspital, Bern, Switzerland
| | - Martina Müller
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - Herbert Schwarz
- Department of Physiology and Immunology Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christa Buechler
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
4
|
Duru AD, Sun R, Allerbring EB, Chadderton J, Kadri N, Han X, Peqini K, Uchtenhagen H, Madhurantakam C, Pellegrino S, Sandalova T, Nygren PÅ, Turner SJ, Achour A. Tuning antiviral CD8 T-cell response via proline-altered peptide ligand vaccination. PLoS Pathog 2020; 16:e1008244. [PMID: 32365082 PMCID: PMC7224568 DOI: 10.1371/journal.ppat.1008244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/14/2020] [Accepted: 04/11/2020] [Indexed: 12/16/2022] Open
Abstract
Viral escape from CD8+ cytotoxic T lymphocyte responses correlates with disease progression and represents a significant challenge for vaccination. Here, we demonstrate that CD8+ T cell recognition of the naturally occurring MHC-I-restricted LCMV-associated immune escape variant Y4F is restored following vaccination with a proline-altered peptide ligand (APL). The APL increases MHC/peptide (pMHC) complex stability, rigidifies the peptide and facilitates T cell receptor (TCR) recognition through reduced entropy costs. Structural analyses of pMHC complexes before and after TCR binding, combined with biophysical analyses, revealed that although the TCR binds similarly to all complexes, the p3P modification alters the conformations of a very limited amount of specific MHC and peptide residues, facilitating efficient TCR recognition. This approach can be easily introduced in peptides restricted to other MHC alleles, and can be combined with currently available and future vaccination protocols in order to prevent viral immune escape.
Collapse
Affiliation(s)
- Adil Doganay Duru
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
- NSU Cell Therapy Institute & Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, Florida, United States of America
| | - Renhua Sun
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Eva B. Allerbring
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Jesseka Chadderton
- Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, Australia
| | - Nadir Kadri
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Xiao Han
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Kaliroi Peqini
- DISFARM, Dipartimento di Scienze Farmaceutiche, Sezinone Chimica Generale e Organica, Università degli Studi, Milano, Italy
| | - Hannes Uchtenhagen
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Chaithanya Madhurantakam
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
- Structural and Molecular Biology Laboratory, Department of Biotechnology, TERI, School of Advanced Studies, New Delhi, India
| | - Sara Pellegrino
- DISFARM, Dipartimento di Scienze Farmaceutiche, Sezinone Chimica Generale e Organica, Università degli Studi, Milano, Italy
| | - Tatyana Sandalova
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Per-Åke Nygren
- Division of Protein Engineering, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Center, Royal Institute of Technology, Stockholm, Sweden
| | - Stephen J. Turner
- Department of Microbiology, Biomedical Discovery Institute, Monash University, Clayton, Australia
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institute, and Division of Infectious Diseases, Karolinska University Hospital, Solna, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
5
|
Verma NK, Chalasani MLS, Scott JD, Kelleher D. CG-NAP/Kinase Interactions Fine-Tune T Cell Functions. Front Immunol 2019; 10:2642. [PMID: 31781123 PMCID: PMC6861388 DOI: 10.3389/fimmu.2019.02642] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/24/2019] [Indexed: 01/04/2023] Open
Abstract
CG-NAP, also known as AKAP450, is an anchoring/adaptor protein that streamlines signal transduction in various cell types by localizing signaling proteins and enzymes with their substrates. Great efforts are being devoted to elucidating functional roles of this protein and associated macromolecular signaling complex. Increasing understanding of pathways involved in regulating T lymphocytes suggests that CG-NAP can facilitate dynamic interactions between kinases and their substrates and thus fine-tune T cell motility and effector functions. As a result, new binding partners of CG-NAP are continually being uncovered. Here, we review recent advances in CG-NAP research, focusing on its interactions with kinases in T cells with an emphasis on the possible role of this anchoring protein as a target for therapeutic intervention in immune-mediated diseases.
Collapse
Affiliation(s)
- Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | | | - John D Scott
- Department of Pharmacology, University of Washington School of Medicine, Seattle, WA, United States
| | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore.,Departments of Medicine and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
6
|
Boyd A, Duchesne L, Lacombe K. Research gaps in viral hepatitis. J Int AIDS Soc 2019; 21 Suppl 2:e25054. [PMID: 29633564 PMCID: PMC5978714 DOI: 10.1002/jia2.25054] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/20/2017] [Indexed: 12/21/2022] Open
Abstract
Introduction The World Health Organization has aimed for global elimination of both hepatitis B virus (HBV) and hepatitis C virus (HCV) by 2030. Treatments available to cure HCV and control HBV, as well as vaccination to prevent HBV infection, have certainly allowed for such bold goals, yet the final steps to usher in elimination require further evidence. Discussion We broadly discuss the needs for three major public health approaches. First, an effective vaccine exists for HBV and mass‐vaccination campaigns have resulted in decreases in hepatitis B surface antigen seroprevalence and overall rates of liver‐related morality. Still, HBV vaccination coverage is poor in certain regions of the world, while the reasons for such low coverage require further study. A prophylactic vaccine is probably needed to eliminate HCV, but is not being readily developed. Second, identifying HBV/HCV infected individuals remains a priority to increase awareness of disease status, particularly for key populations. Research evaluating large‐scale implementation of novel, rapid and mobile point‐of‐care tests would be helpful to determine whether increased awareness is achievable in these settings. Third, antiviral therapy allows for strong HBV suppression and HCV cure, while its access depends on financial factors among many others. Although there is strong evidence to treat key populations and specific groups with progressed disease, as stated in current guidelines, the advantages of extending treatment eligibility to decrease onward spread of HBV/HCV infection and prevent further burden of disease are lacking “real world” evidence. Novel anti‐HBV treatments are being developed to target intrahepatocellular HBV replication, but are still in the early phases of clinical development. Each of the strategies mentioned above has specific implications for HIV infection. Conclusions There are certainly effective tools to combat the spread of viral hepatitis and treat infected individuals – yet how they are able to reach key populations, and the infrastructure required to do so, continue to represent the largest research gap when evaluating the progress towards elimination. Continuously adapted and informed research is required to establish the priorities in achieving elimination goals.
Collapse
Affiliation(s)
- Anders Boyd
- INSERM, UMR_S1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France.,Department of Infectious Diseases, Research and Prevention, Public Health Service of Amsterdam, Amsterdam, Netherlands
| | - Léa Duchesne
- INSERM, UMR_S1136, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Paris, France
| | - Karine Lacombe
- Department of Infectious and Tropical Diseases, Saint-Antoine Hospital, AP-HP, Paris, France.,Sorbonne Universités, INSERM, UPMC Univ Paris 06, Institut Pierre Louis d'épidémiologie et de Santé Publique (IPLESP UMRS 1136), Paris, France
| |
Collapse
|
7
|
Ray RB, Ray R. Hepatitis C Virus Manipulates Humans as its Favorite Host for a Long-Term Relationship. Hepatology 2019; 69:889-900. [PMID: 30102776 PMCID: PMC6351149 DOI: 10.1002/hep.30214] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/07/2018] [Indexed: 02/06/2023]
Abstract
Chronic hepatitis C virus (HCV) infection-associated liver disease is a global health problem. HCV often causes silent disease, and eventually progresses to end-stage liver disease. HCV infects hepatocytes; however, initial manifestation of liver disease is mostly displayed in hepatic stellate cells (HSCs), causing fibrosis/cirrhosis, and is believed to occur from inflammation in the liver. It remains unclear why HCV is not spontaneously cleared from infected liver in the majority of individuals and develops chronic infection with progressive liver disease. Direct-acting antivirals (DAAs) show excellent results in controlling viremia, although beneficial consequence in advanced liver disease remains to be understood. In this review, we highlight the current knowledge that has contributed to our understanding of the role of HCV in inflammation, immune evasion, metabolic disorders, liver pathogeneses, and efforts in vaccine development.
Collapse
Affiliation(s)
- Ratna B. Ray
- Department of Pathology, Saint Louis University, Saint Louis, Missouri 63104, USA,Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri 63104, USA
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, Saint Louis, Missouri 63104, USA
| |
Collapse
|
8
|
Krishna S, Ulrich P, Wilson E, Parikh F, Narang P, Yang S, Read AK, Kim-Schulze S, Park JG, Posner M, Wilson Sayres MA, Sikora A, Anderson KS. Human Papilloma Virus Specific Immunogenicity and Dysfunction of CD8 + T Cells in Head and Neck Cancer. Cancer Res 2018; 78:6159-6170. [PMID: 30154146 DOI: 10.1158/0008-5472.can-18-0163] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 06/26/2018] [Accepted: 08/20/2018] [Indexed: 11/16/2022]
Abstract
Human papillomavirus subtype 16 (HPV16) is the primary cause of an increasing number of head and neck squamous cell carcinomas (HNSCC), providing strong rationale for T-cell immune therapies against HPV+ HNSCC. Here we assess immunogenicity of HPV16-specific CD8+ T cells (CTL) and characterize HPV-specific mechanisms of T-cell dysfunction. We identified 16 strong and 29 moderately immunogenic CTL-epitopes from HPV16 E2, E6, and E7 antigens restricted by 12 common HLA class I alleles. E2-specific CTL-reactivity was higher in patients with HPV+ HNSCC than in healthy controls (>3-fold; P = 0.026). Patient-derived E2, E6, and E7 peripheral CTLs exhibited heterogeneity in dysfunctional phenotypes. Immunogenomic analyses of 119 HNSCC transcriptomes revealed high T-cell infiltration and dysfunction in HPV+ HNSCC and correlation of HPV antigen expression with T-cell exhaustion gene signatures. Indoleamine 2,3-dioxygenase (IDO-1) was strongly expressed in HPV+ HNSCC versus HPV- HNSCC (P = 0.001) and correlated with E7 expression (R 2 = 0.84; P = 0.033). Combination treatment with PD-1 blockade and IDO-1 inhibition overcame profound CTL-dysfunction, enhancing HPV+ HNSCC sensitivity to CTL-cytotoxicity in vitro (up to 10-fold in E7-CTLs, P = 0.011). Our findings implicate mechanisms of T-cell escape in HPV+ HNSCC, wherein high tumoral HPV-antigen load results in high expression of immune dysfunction genes on tumor cells (e.g., IDO-1), and dysfunction of HPV-specific CTLs (e.g., E7, E2-CTLs). The HPV16 CTL-epitopes identified in this study, in combination with blockade of HPV+ HNSCC-specific PD-1/IDO-1 checkpoints, may be useful for targeted immunotherapy.Significance: This study evaluates the HPV antigen T-cell immunogenicity role of inhibitory receptors and other exhaustion markers in the cytotoxic function of HPV antigen-specific CTLs and identifies combined inhibition of PD-1/IDO-1 as a strategy to enhance CTL targeting of HPV+ HNSCC. Cancer Res; 78(21); 6159-70. ©2018 AACR.
Collapse
Affiliation(s)
- Sri Krishna
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, Arizona
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona
| | - Peaches Ulrich
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, Arizona
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Eric Wilson
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, Arizona
| | - Falguni Parikh
- Department of Otolaryngology and Head and Neck Surgery, Baylor College of Medicine, Houston, Texas
| | - Pooja Narang
- School of Life Sciences, Arizona State University, Tempe, Arizona
| | - Shanshan Yang
- Biodesign Institute Bioinformatics Core Facility, Arizona State University, Tempe, Arizona
| | - Amelia K Read
- Department of Otolaryngology, Stanford University, Stanford, California
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Jin G Park
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, Arizona
| | - Marshall Posner
- Tisch Cancer institute, Icahn School of Medicine at Mount Sinai Medical Center, New York, New York
| | - Melissa A Wilson Sayres
- School of Life Sciences, Arizona State University, Tempe, Arizona
- Center for Evolution and Medicine, Arizona State University, Tempe, Arizona
| | - Andrew Sikora
- Department of Otolaryngology and Head and Neck Surgery, Baylor College of Medicine, Houston, Texas
| | - Karen S Anderson
- Biodesign Center for Personalized Diagnostics, Arizona State University, Tempe, Arizona.
- School of Life Sciences, Arizona State University, Tempe, Arizona
| |
Collapse
|
9
|
Hofmann M, Wieland D, Pircher H, Thimme R. Memory vs memory-like: The different facets of CD8+T-cell memory in HCV infection. Immunol Rev 2018; 283:232-237. [DOI: 10.1111/imr.12642] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Maike Hofmann
- Department of Medicine II; University Hospital Freiburg; Faculty of Medicine; University of Freiburg; Freiburg Germany
| | - Dominik Wieland
- Department of Medicine II; University Hospital Freiburg; Faculty of Medicine; University of Freiburg; Freiburg Germany
| | - Hanspeter Pircher
- Institute for Immunology; Medical Center; Faculty of Medicine; University of Freiburg; Freiburg Germany
| | - Robert Thimme
- Department of Medicine II; University Hospital Freiburg; Faculty of Medicine; University of Freiburg; Freiburg Germany
| |
Collapse
|
10
|
Escape of Hepatitis C Virus from Epitope I Neutralization Increases Sensitivity of Other Neutralization Epitopes. J Virol 2018; 92:JVI.02066-17. [PMID: 29467319 PMCID: PMC5899191 DOI: 10.1128/jvi.02066-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/05/2018] [Indexed: 01/01/2023] Open
Abstract
The hepatitis C virus (HCV) E2 glycoprotein is a major target of the neutralizing antibody (nAb) response, with multiple type-specific and broadly neutralizing antibody (bnAb) epitopes identified. The 412-to-423 region can generate bnAbs that block interaction with the cell surface receptor CD81, with activity toward multiple HCV genotypes. In this study, we reveal the structure of rodent monoclonal antibody 24 (MAb24) with an extensive contact area toward a peptide spanning the 412-to-423 region. The crystal structure of the MAb24–peptide 412-to-423 complex reveals the paratope bound to a peptide hairpin highly similar to that observed with human MAb HCV1 and rodent MAb AP33, but with a different angle of approach. In viral outgrowth experiments, we demonstrated three distinct genotype 2a viral populations that acquired resistance to MAb24 via N415D, N417S, and N415D/H386R mutations. Importantly, the MAb24-resistant viruses exhibited significant increases in sensitivity to the majority of bnAbs directed to epitopes within the 412-to-423 region and in additional antigenic determinants located within E2 and the E1E2 complex. This study suggests that modification of N415 causes a global change in glycoprotein structure that increases its vulnerability to neutralization by other antibodies. This finding suggests that in the context of an antibody response to viral infection, acquisition of escape mutations in the 412-to-423 region renders the virus more susceptible to neutralization by other specificities of nAbs, effectively reducing the immunological fitness of the virus. A vaccine for HCV that generates polyspecific humoral immunity with specificity for the 412-to-423 region and at least one other region of E2 is desirable. IMPORTANCE Understanding how antibodies neutralize hepatitis C virus (HCV) is essential for vaccine development. This study reveals for the first time that when HCV develops resistance to a major class of bnAbs targeting the 412-to-423 region of E2, this results in a concomitant increase in sensitivity to neutralization by a majority of other bnAb specificities. Vaccines for the prevention of HCV infection should therefore generate bnAbs directed toward the 412-to-423 region of E2 and additional bnAb epitopes within the viral glycoproteins.
Collapse
|
11
|
Cabral MS, Santos TPS, Santos PL, Schinoni MI, Oliveira IS, Pereira AB, Atta AM, Sousa-Atta MLB. Immune response of Th17-associated cytokines by peripheral blood mononuclear cells from patients with chronic hepatitis C virus infection. Cytokine 2018; 102:200-205. [PMID: 28969940 DOI: 10.1016/j.cyto.2017.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 09/08/2017] [Accepted: 09/12/2017] [Indexed: 02/07/2023]
Abstract
Hepatitis C virus (HCV) chronic infection causes severe cellular immune dysfunction. Here, we investigated the production of Th17-associated cytokines by peripheral blood mononuclear cells (PBMCs) of untreated patients with HCV, patients presenting an early virologic response (EVR) after 12weeks of treatment with interferon-α plus ribavirin with or without HCV protease inhibitors, and patients who were nonresponders to HCV therapy. PBMCs were stimulated with HCV core and nonstructural antigens, and the production of Th17-associated cytokines was measured with a Milliplex MAP immunoassay. Core-stimulated PBMCs from both untreated and nonresponder patients produced interleukin (IL)-17A, and vigorous production of IL-17A in response to NS3 antigen was only verified in the untreated group. Nonresponder patients also produced IL-17F after core antigen stimulation. IL-21 production was unaltered in the three groups of patients, whereas IL-17E and IL-22 were not detected. The production of Th17 cytokines by cells from patients showing an EVR was insignificant. IL-17A and IL-17F levels were not correlated with alanine aminotransferase levels or viremia. However, advanced fibrosis was associated with higher IL-17A production in T0 cells stimulated with core antigen. Untreated patients with HCV and patients who were nonresponders to antiviral treatment differed in their PBMC immune responses of Th17-associated cytokines. The early virological response to antiviral treatment dramatically decreased Th17 immune responses to HCV antigens.
Collapse
Affiliation(s)
- Milena S Cabral
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Brazil
| | - Taciana P S Santos
- Programa de Pós-Graduação em Imunologia, Universidade Federal da Bahia, Brazil
| | - Priscila L Santos
- Laboratório de Biologia Molecular, Universidade Federal de Sergipe, Brazil
| | | | - Isabela S Oliveira
- Laboratório de Pesquisa em Imunologia, Universidade Federal da Bahia, Brazil
| | - Ariana B Pereira
- Laboratório de Pesquisa em Imunologia, Universidade Federal da Bahia, Brazil
| | - Ajax M Atta
- Laboratório de Pesquisa em Imunologia, Universidade Federal da Bahia, Brazil
| | | |
Collapse
|
12
|
Pollmann J, Rölle A, Hofmann M, Cerwenka A. Hepatitis C Virus and Human Cytomegalovirus-Natural Killer Cell Subsets in Persistent Viral Infections. Front Immunol 2017; 8:566. [PMID: 28567042 PMCID: PMC5434107 DOI: 10.3389/fimmu.2017.00566] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 04/27/2017] [Indexed: 12/17/2022] Open
Abstract
Hepatitis C virus (HCV) and human cytomegalovirus (HCMV) are prominent examples of RNA and DNA viruses, respectively, that establish a persistent infection in their host. HCV affects over 185 million patients worldwide, who are at high risk for developing liver fibrosis, liver cirrhosis, and ultimately hepatocellular carcinoma. Recent breakthroughs in HCV therapy, using direct-acting antivirals have provided the opportunity to monitor natural killer (NK) cells after clearance of a chronic infection. There is now increasing evidence that the individual NK cell repertoire before infection is predictive for the course of disease. HCMV affects the majority of the global population. While being asymptomatic in healthy individuals, HCMV represents a severe clinical challenge in immunocompromised patients. Both viral infections, HCV and HCMV, lead to long-lasting and profound alterations within the entire NK cell compartment. This review article, will discuss the diverse range of changes in the NK cell compartment as well as potential consequences for the course of disease.
Collapse
Affiliation(s)
- Julia Pollmann
- Research Group Innate Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Rölle
- Clinical Cooperation Unit Applied Tumor-Immunity, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Maike Hofmann
- Faculty of Medicine, Department of Medicine II, University Hospital Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Adelheid Cerwenka
- Research Group Innate Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty Mannheim, Division of Immunbiochemistry, University Heidelberg, Heidelberg, Germany
| |
Collapse
|
13
|
Lythgoe KA, Gardner A, Pybus OG, Grove J. Short-Sighted Virus Evolution and a Germline Hypothesis for Chronic Viral Infections. Trends Microbiol 2017; 25:336-348. [PMID: 28377208 PMCID: PMC5405858 DOI: 10.1016/j.tim.2017.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/03/2017] [Accepted: 03/03/2017] [Indexed: 12/24/2022]
Abstract
With extremely short generation times and high mutability, many viruses can rapidly evolve and adapt to changing environments. This ability is generally beneficial to viruses as it allows them to evade host immune responses, evolve new behaviours, and exploit ecological niches. However, natural selection typically generates adaptation in response to the immediate selection pressures that a virus experiences in its current host. Consequently, we argue that some viruses, particularly those characterised by long durations of infection and ongoing replication, may be susceptible to short-sighted evolution, whereby a virus' adaptation to its current host will be detrimental to its onward transmission within the host population. Here we outline the concept of short-sighted viral evolution and provide examples of how it may negatively impact viral transmission among hosts. We also propose that viruses that are vulnerable to short-sighted evolution may exhibit strategies that minimise its effects. We speculate on the various mechanisms by which this may be achieved, including viral life history strategies that result in low rates of within-host evolution, or the establishment of a 'germline' lineage of viruses that avoids short-sighted evolution. These concepts provide a new perspective on the way in which some viruses have been able to establish and maintain global pandemics.
Collapse
Affiliation(s)
| | - Andy Gardner
- School of Biology, University of St Andrews, St Andrews, KY16 9TH, UK
| | - Oliver G Pybus
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, UK
| | - Joe Grove
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, WC1E 6BT, UK
| |
Collapse
|
14
|
da Silva RA, de Carvalho IMVG, de Matos RPA, Yamasaki LHT, Bittar C, Rahal P, Jardim ACG. Evidence of bottleneck effect on hepatitis C virus transmission between a couple under interferon based therapy. INFECTION GENETICS AND EVOLUTION 2016; 47:87-93. [PMID: 27888038 DOI: 10.1016/j.meegid.2016.11.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/20/2016] [Accepted: 11/11/2016] [Indexed: 01/06/2023]
Abstract
Issues on the correlation of viral genetic diversity and treatment response to the hepatitis C infection remain uncertain. The bottleneck effect dictates the characteristics of the viral population that will establish the infection in a new host and is related to how the immune system and treatment will be effective against the virus. Here we evaluated the phylogenetic characteristics of quasispecies population and the treatment response pattern of a HCV infected couple. We also analyzed whether the viral population of these patients indicated that they were exposed to the same source for primer infection. This study included two patients (P10 and P11) HCV genotype 1b infected. The couple presented horizontal transmission. Viral RNA was isolated from serum samples collected before, during and after treatment, at specific time points. The HCV NS5A gene sequence was amplified, cloned and sequenced. Genetic and evolutionary analyses were performed to compare the quasispecies population of these two patients and local control patients. Genetic distance and diversity were calculated. Phylogenetic analyses were performed by using maximum likelihood and Bayesian methodologies. The analysis of the baseline samples showed that the genetic distance of the viral populations of patients P10 and P11 was significantly lower than when these patients and the control group based on sequences from local patients were analyzed, supporting the horizontal transmission hypothesis. Phylogenetic analysis with sequences from all the time point samples also demonstrated two patterns of evolution depending on the treatment response. The Bayesian analysis showed that one isolate corresponding to the baseline sample of P10 was grouped into the P11 clade, suggesting a way of infection and a bottleneck effect. Our data suggests that the patient P11 viral population may be originated from variants from P10 patient and consequently showing that clinical differences between treatment responses can emerge from the bottleneck effect on viral populations.
Collapse
Affiliation(s)
- Rafael Alves da Silva
- Laboratório de Hepatologia Molecular Aplicada, LHEMA, Disciplina de Gastroenterologia, Departamento de Medicina, Universidade Federal de São Paulo, Av. Pedro de Toledo n° 669, 5° Andar, SP, Brazil; Laboratório de Parasitologia, Instituto Butantan, Av. Vital Brazil, n° 1500, SP, Brazil
| | | | | | | | - Cíntia Bittar
- Laboratório de Estudos Genômicos, Ibilce, UNESP, São José do Rio Preto, SP, Brazil
| | - Paula Rahal
- Laboratório de Estudos Genômicos, Ibilce, UNESP, São José do Rio Preto, SP, Brazil
| | - Ana Carolina Gomes Jardim
- Laboratório de Estudos Genômicos, Ibilce, UNESP, São José do Rio Preto, SP, Brazil; Laboratório de Virologia, Instituto de Ciências Biomédicas, Universidade Federal de Uberlândia - UFU, Uberlândia, MG, Brazil
| |
Collapse
|
15
|
Moise L, Beseme S, Tassone R, Liu R, Kibria F, Terry F, Martin W, De Groot AS. T cell epitope redundancy: cross-conservation of the TCR face between pathogens and self and its implications for vaccines and autoimmunity. Expert Rev Vaccines 2016; 15:607-17. [PMID: 26588466 DOI: 10.1586/14760584.2016.1123098] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
T cells are extensively trained on 'self' in the thymus and then move to the periphery, where they seek out and destroy infections and regulate immune response to self-antigens. T cell receptors (TCRs) on T cells' surface recognize T cell epitopes, short linear strings of amino acids presented by antigen-presenting cells. Some of these epitopes activate T effectors, while others activate regulatory T cells. It was recently discovered that T cell epitopes that are highly conserved on their TCR face with human genome sequences are often associated with T cells that regulate immune response. These TCR-cross-conserved or 'redundant epitopes' are more common in proteins found in pathogens that have co-evolved with humans than in other non-commensal pathogens. Epitope redundancy might be the link between pathogens and autoimmune disease. This article reviews recently published data and addresses epitope redundancy, the "elephant in the room" for vaccine developers and T cell immunologists.
Collapse
Affiliation(s)
- Leonard Moise
- a EpiVax, Inc ., Providence , RI , USA.,b Institute for Immunology and Informatics , University of Rhode Island , Providence , RI , USA
| | | | - Ryan Tassone
- b Institute for Immunology and Informatics , University of Rhode Island , Providence , RI , USA
| | - Rui Liu
- b Institute for Immunology and Informatics , University of Rhode Island , Providence , RI , USA
| | | | | | | | - Anne S De Groot
- a EpiVax, Inc ., Providence , RI , USA.,b Institute for Immunology and Informatics , University of Rhode Island , Providence , RI , USA
| |
Collapse
|
16
|
Abdelwahab SF. Cellular immune response to hepatitis-C-virus in subjects without viremia or seroconversion: is it important? Infect Agent Cancer 2016; 11:23. [PMID: 27186234 PMCID: PMC4867533 DOI: 10.1186/s13027-016-0070-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 03/30/2016] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C Virus (HCV) causes chronic infection and represents a global health burden. To date, there is no licensed vaccine for HCV. The high viral replication rate and the existence of several HCV genotypes and quasispecies hamper the development of an effective universal vaccine. In this regard, the current HCV vaccine candidates show genotype-specific protection or narrow cross reactivity against other genotypes. Importantly, HCV spontaneous clearance occurs in 15-50 % of infected subjects, indicating that natural resistance to chronic infection exists. This phenomenon was demonstrated among humans and chimpanzees and continues to motivate researchers attempting to develop an effective HCV vaccine. However, what constitutes a protective immune response or correlate of protection against HCV infection is still vague. Additionally, the mechanisms behind successful HCV clearance suggest the coordination of several arms of the immune system, with cell-mediated immunity (CMI) playing a crucial role in this process. By contrast, although neutralizing antibodies have been identified, they are isolate-specific and poorly correlate with viral clearance. Antigen-specific CD4 T cells, instead, correlate with transient decline in HCV viremia and long-lasting control of the infection. Unfortunately, HCV has been very successful in evading host immune mechanisms, leading to complications such as liver fibrosis, cirrhosis and hepatocellular carcinoma. Interestingly, CMI to HCV antigens were shown among exposed individuals without viremia or seroconversion, suggesting the clearance of prior HCV infection(s). These individuals include family members living with HCV-infected subjects, healthcare workers, IV drug users, and sexual contacts. The correlates of protection could be closely monitored among these individuals. This review provides a summary of HCV-specific immune responses in general and of CMI in particular in these cohorts. The importance of these CMI responses are discussed.
Collapse
Affiliation(s)
- Sayed F. Abdelwahab
- Departement of Microbiology and Immunology, Faculty of Medicine, Minia University, Minia, 61511 Egypt
- Department of Microbiology, College of Pharmacy, Taif University, Taif, 21974 Kingdom of Saudi Arabia
| |
Collapse
|
17
|
Mosaad YM. Clinical Role of Human Leukocyte Antigen in Health and Disease. Scand J Immunol 2015; 82:283-306. [PMID: 26099424 DOI: 10.1111/sji.12329] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Revised: 06/02/2015] [Accepted: 06/12/2015] [Indexed: 12/19/2022]
Abstract
Most of the genes in the major histocompatibility complex (MHC) region express high polymorphism that is fundamental for their function. The most important function of human leukocyte antigen (HLA) molecule is in the induction, regulation of immune responses and the selection of the T cell repertoire. A clinician's attention is normally drawn to a system only when it malfunctions. The HLA system is no exception in this regard, but in contrast to other systems, it also arouses interest when it functions well - too well, in fact. Population studies carried out over the last several decades have identified a long list of human diseases that are significantly more common among individuals that carry particular HLA alleles including inflammatory, autoimmune and malignant disorders. HLA-disease association is the name of this phenomenon, and the mechanism underlying is still a subject of hot debate. Social behaviours are affected by HLA genes and preference for HLA disparate mates may provide 'good genes' for an individual's offspring. Also, certain HLA genes may be associated with shorter life and others with longer lifespan, but the effects depend both on the genetic background and on the environmental conditions. The following is a general overview of the important functional aspects of HLA in health and diseases.
Collapse
Affiliation(s)
- Y M Mosaad
- Clinical Immunology Unit, Clinical Pathology Department & Mansoura Research Center for Cord Stem Cell (MARC_CSC), Mansoura Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
18
|
Abdel-Hady KM, Gutierrez AH, Terry F, Desrosiers J, De Groot AS, Azzazy HME. Identification and retrospective validation of T-cell epitopes in the hepatitis C virus genotype 4 proteome: an accelerated approach toward epitope-driven vaccine development. Hum Vaccin Immunother 2015; 10:2366-77. [PMID: 25424944 DOI: 10.4161/hv.29177] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
With over 150 million people chronically infected worldwide and millions more infected annually, hepatitis C continues to pose a burden on the global healthcare system. The standard therapy of hepatitis C remains expensive, with severe associated side effects and inconsistent cure rates. Vaccine development against the hepatitis C virus has been hampered by practical and biological challenges posed by viral evasion mechanisms. Despite these challenges, HCV vaccine research has presented a number of candidate vaccines that progressed to phase II trials. However, those efforts focused mainly on HCV genotypes 1 and 2 as vaccine targets and barely enough attention was given to genotype 4, the variant most prevalent in the Middle East and central Africa. We describe herein the in silico identification of highly conserved and immunogenic T-cell epitopes from the HCV genotype 4 proteome, using the iVAX immunoinformatics toolkit, as targets for an epitope-driven vaccine. We also describe a fast and inexpensive approach for results validation using the empirical data on the Immune Epitope Database (IEDB) as a reference. Our analysis identified 90 HLA class I epitopes of which 20 were found to be novel and 19 more had their binding predictions retrospectively validated; empirical data for the remaining 51 epitopes was insufficient to validate their binding predictions. Our analysis also identified 14 HLA class II epitopes, of which 8 had most of their binding predictions validated. Further investigation is required regarding the efficacy of the identified epitopes as vaccine targets in populations where HCV genotype 4 is most prevalent.
Collapse
Affiliation(s)
- Karim M Abdel-Hady
- a Department of Chemistry; School of Sciences and Engineering; The American University in Cairo; New Cairo, Egypt
| | | | | | | | | | | |
Collapse
|
19
|
Zehn D, Wherry EJ. Immune Memory and Exhaustion: Clinically Relevant Lessons from the LCMV Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 850:137-52. [PMID: 26324351 DOI: 10.1007/978-3-319-15774-0_10] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The development of dysfunctional or exhausted T cells is characteristic of immune responses to chronic viral infections and cancer. Exhausted T cells are defined by reduced effector function, sustained upregulation of multiple inhibitory receptors, an altered transcriptional program and perturbations of normal memory development and homeostasis. This review focuses on (a) illustrating milestone discoveries that led to our present understanding of T cell exhaustion, (b) summarizing recent developments in the field, and (c) identifying new challenges for translational research. Exhausted T cells are now recognized as key therapeutic targets in human infections and cancer. Much of our knowledge of the clinically relevant process of exhaustion derives from studies in the mouse model of Lymphocytic choriomeningitis virus (LCMV) infection. Studies using this model have formed the foundation for our understanding of human T cell memory and exhaustion. We will use this example to discuss recent advances in our understanding of T cell exhaustion and illustrate the value of integrated mouse and human studies and will emphasize the benefits of bi-directional mouse-to-human and human-to-mouse research approaches.
Collapse
Affiliation(s)
- D Zehn
- Division of Immunology and Allergy, Lausanne University Hospital, Lausanne, Switzerland,
| | | |
Collapse
|
20
|
Gededzha MP, Mphahlele MJ, Selabe SG. Prediction of T-cell epitopes of hepatitis C virus genotype 5a. Virol J 2014; 11:187. [PMID: 25380768 PMCID: PMC4289306 DOI: 10.1186/1743-422x-11-187] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 10/14/2014] [Indexed: 12/26/2022] Open
Abstract
Background Hepatitis C virus (HCV) is a public health problem with almost 185 million people estimated to be infected worldwide and is one of the leading causes of hepatocellular carcinoma. Currently, there is no vaccine for HCV infection and the current treatment does not clear the infection in all patients. Because of the high diversity of HCV, protective vaccines will have to overcome significant viral antigenic diversities. The objective of this study was to predict T-cell epitopes from HCV genotype 5a sequences. Methods HCV near full-length protein sequences were analyzed to predict T-cell epitopes that bind human leukocyte antigen (HLA) class I and HLA class II in HCV genotype 5a using Propred I and Propred, respectively. The Antigenicity score of all the predicted epitopes were analysed using VaxiJen v2.0. All antigenic predicted epitopes were analysed for conservation using the IEDB database in comparison with 406, 221, 98, 33, 45, 45 randomly selected sequences from each of the HCV genotypes 1a, 1b, 2, 3, 4 and 6 respectively, downloaded from the GenBank. For epitope prediction binding to common HLA alleles found in South Africa, the IEDB epitope analysis tool was used. Results A total of 24 and 77 antigenic epitopes that bind HLA class I and HLA class II respectively were predicted. The highest number of HLA class I binding epitopes were predicted within the NS3 (63%), followed by NS5B (21%). For the HLA class II, the highest number of epitopes were predicted in the NS3 (30%) followed by the NS4B (23%) proteins. For conservation analysis, 8 and 31 predicted epitopes were conserved in different genotypes for HLA class I and HLA class II alleles respectively. Several epitopes bind with high affinity for both HLA class I alleles and HLA class II common in South Africa. Conclusion The predicted conserved T-cell epitopes analysed in this study will contribute towards the future design of HCV vaccine candidates which will avoid variation in genotypes, which in turn will be capable of inducing broad HCV specific immune responses.
Collapse
Affiliation(s)
| | | | - Selokela G Selabe
- HIV and Hepatitis Research Unit, Department of Virology, University of Limpopo, Medunsa Campus/National Health Laboratory Service, Pretoria, South Africa.
| |
Collapse
|
21
|
Moise L, Terry F, Gutierrez AH, Tassone R, Losikoff P, Gregory SH, Bailey-Kellogg C, Martin WD, De Groot AS. Smarter vaccine design will circumvent regulatory T cell-mediated evasion in chronic HIV and HCV infection. Front Microbiol 2014; 5:502. [PMID: 25339942 PMCID: PMC4186478 DOI: 10.3389/fmicb.2014.00502] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/08/2014] [Indexed: 01/17/2023] Open
Abstract
Despite years of research, vaccines against HIV and HCV are not yet available, due largely to effective viral immunoevasive mechanisms. A novel escape mechanism observed in viruses that cause chronic infection is suppression of viral-specific effector CD4(+) and CD8(+) T cells by stimulating regulatory T cells (Tregs) educated on host sequences during tolerance induction. Viral class II MHC epitopes that share a T cell receptor (TCR)-face with host epitopes may activate Tregs capable of suppressing protective responses. We designed an immunoinformatic algorithm, JanusMatrix, to identify such epitopes and discovered that among human-host viruses, chronic viruses appear more human-like than viruses that cause acute infection. Furthermore, an HCV epitope that activates Tregs in chronically infected patients, but not clearers, shares a TCR-face with numerous human sequences. To boost weak CD4(+) T cell responses associated with persistent infection, vaccines for HIV and HCV must circumvent potential Treg activation that can handicap efficacy. Epitope-driven approaches to vaccine design that involve careful consideration of the T cell subsets primed during immunization will advance HIV and HCV vaccine development.
Collapse
Affiliation(s)
- Leonard Moise
- EpiVax, Inc., Providence, RI, USA
- Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | | | - Andres H. Gutierrez
- Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Ryan Tassone
- Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| | - Phyllis Losikoff
- Department of Medicine, Rhode Island Hospital and the Warren Alpert Medical School at Brown University, Providence, RI, USA
| | | | | | | | - Anne S. De Groot
- EpiVax, Inc., Providence, RI, USA
- Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, USA
| |
Collapse
|
22
|
Doolan DL, Apte SH, Proietti C. Genome-based vaccine design: the promise for malaria and other infectious diseases. Int J Parasitol 2014; 44:901-13. [PMID: 25196370 DOI: 10.1016/j.ijpara.2014.07.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 07/30/2014] [Accepted: 07/31/2014] [Indexed: 01/08/2023]
Abstract
Vaccines are one of the most effective interventions to improve public health, however, the generation of highly effective vaccines for many diseases has remained difficult. Three chronic diseases that characterise these difficulties include malaria, tuberculosis and HIV, and they alone account for half of the global infectious disease burden. The whole organism vaccine approach pioneered by Jenner in 1796 and refined by Pasteur in 1857 with the "isolate, inactivate and inject" paradigm has proved highly successful for many viral and bacterial pathogens causing acute disease but has failed with respect to malaria, tuberculosis and HIV as well as many other diseases. A significant advance of the past decade has been the elucidation of the genomes, proteomes and transcriptomes of many pathogens. This information provides the foundation for new 21st Century approaches to identify target antigens for the development of vaccines, drugs and diagnostic tests. Innovative genome-based vaccine strategies have shown potential for a number of challenging pathogens, including malaria. We advocate that genome-based rational vaccine design will overcome the problem of poorly immunogenic, poorly protective vaccines that has plagued vaccine developers for many years.
Collapse
Affiliation(s)
- Denise L Doolan
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia.
| | - Simon H Apte
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| | - Carla Proietti
- Infectious Diseases Programme, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4029, Australia
| |
Collapse
|
23
|
Ashraf W, Manzoor S, Ashraf J, Ahmed QL, Khalid M, Tariq M, Imran M, Aziz H. Transcript analysis of P2X receptors in PBMCs of chronic HCV patients: an insight into antiviral treatment response and HCV-induced pathogenesis. Viral Immunol 2014; 26:343-50. [PMID: 24116708 DOI: 10.1089/vim.2013.0044] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND After invasion of hepatocytes and immune cells, hepatitis C virus has the ability to escape from the host immune system, leading to the progression of disease into chronic infection with associated liver morbidities. Adenosine 5'triphosphate (ATP) is released in most of the pathological events from the affected cells and acts as a signaling molecule by binding to P2X receptors expressed on the host's immune cells and activates the immune system for pro-inflammatory response. Therefore, the present study was designed to analyze the transcript expression of the ionotropic purinergic P2X receptors on peripheral blood mononuclear cells (PBMCs) of chronic HCV patients to have study the immune responses mediated by P2X receptors in chronic HCV infections. METHODS PBMCs were isolated from the collected blood samples. Transcript analysis of P2X receptors in PBMCs was done. The identity of amplified product was confirmed by sequencing PCR, while the quantification of the transcript expression was done by real time PCR. The relative expression of the P2X receptors was analyzed by unpaired Student's t test using GraphPad Prims 5 software. RESULTS We found that out of seven isoforms of P2X receptors, P2X1, P2X4, P2X5, and P2X7 receptors are expressed on the PBMCs. P2X1 and P2X7 are significantly upregulated in treatment-naïve chronic HCV patients by 2.2- and 2.5-fold, respectively. However, only P2X7 expression is found increased by 2.7-fold in patients achieving sustained virological response (SVR) after antiviral treatment compared to healthy controls. The expression of P2X receptors remained unaltered in chronic HCV patients not responding to the treatment. CONCLUSION The present study confirms the significant involvement of P2X receptors in the immune responses mediated by the PBMCs in the chronic HCV infection, which should be further investigated to devise strategies to augment the immune system against this chronic viral disease.
Collapse
Affiliation(s)
- Waseem Ashraf
- 1 Atta ur Rahman School of Applied Bio-Sciences, National University of Sciences & Technology, Islamabad
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Kim H, Meyer K, Di Bisceglie AM, Ray R. Inhibition of c3 convertase activity by hepatitis C virus as an additional lesion in the regulation of complement components. PLoS One 2014; 9:e101422. [PMID: 24983375 PMCID: PMC4077819 DOI: 10.1371/journal.pone.0101422] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 06/06/2014] [Indexed: 01/25/2023] Open
Abstract
We have previously reported that in vitro HCV infection of cells of hepatocyte origin attenuates complement system at multiple steps, and attenuation also occurs in chronically HCV infected liver, irrespective of the disease stage. However, none of these regulations alone completely impaired complement pathways. Modulation of the upstream proteins involved in proteolytic processing of the complement cascade prior to convertase formation is critical in promoting the function of the complement system in response to infection. Here, we examined the regulation of C2 complement expression in hepatoma cells infected in vitro with cell culture grown virus, and validated our observations using randomly selected chronically HCV infected patient liver biopsy specimens. C2 mRNA expression was significantly inhibited, and classical C3 convertase (C4b2a) decreased. In separate experiments for C3 convertase function, C3b deposition onto bacterial membrane was reduced using HCV infected patient sera as compared to uninfected control, suggesting impaired C3 convertase. Further, iC3b level, a proteolytically inactive form of C3b, was lower in HCV infected patient sera, reflecting impairment of both C3 convertase and Factor I activity. The expression level of Factor I was significantly reduced in HCV infected liver biopsy specimens, while Factor H level remained unchanged or enhanced. Together, these results suggested that inhibition of C3 convertase activity is an additional cumulative effect for attenuation of complement system adopted by HCV for weakening innate immune response.
Collapse
Affiliation(s)
- Hangeun Kim
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, United States of America
| | - Keith Meyer
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, United States of America
| | - Adrian M. Di Bisceglie
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, United States of America
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
25
|
Type I interferon suppresses de novo virus-specific CD4 Th1 immunity during an established persistent viral infection. Proc Natl Acad Sci U S A 2014; 111:7409-14. [PMID: 24799699 DOI: 10.1073/pnas.1401662111] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
CD4 T cells are central to orchestrate, sustain, and potentially regenerate antiviral immunity throughout persistent viral infections. Although the evolving immune environment during persistent infection reshapes established CD4 T-cell responses, the fate of naïve CD4 T cells primed in the midst of persistent infection is unclear. We demonstrate that, in marked contrast to the onset of infection, virus-specific CD4 T cells primed during an established persistent infection have diminished ability to develop Th1 responses, to efficiently accumulate in peripheral tissues, and almost exclusively differentiate into T follicular helper cells. Consistent with suppressed Th1 and heightened Tfh differentiation, virus-specific CD4 T cells primed during the established persistent infection provide help to B cells, but only limited help to CD8 T cells. The suppression of de novo Th1 generation and tissue distribution was mediated by chronic type I IFN (IFN-I) production and was effectively restored by blocking IFN-I signaling during CD4 T-cell priming. Thus, we establish a suppressive function of chronic IFN-I signaling and mechanism of immunoregulation during an established persistent virus infection.
Collapse
|
26
|
Statistical linkage analysis of substitutions in patient-derived sequences of genotype 1a hepatitis C virus nonstructural protein 3 exposes targets for immunogen design. J Virol 2014; 88:7628-44. [PMID: 24760894 DOI: 10.1128/jvi.03812-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED Chronic hepatitis C virus (HCV) infection is one of the leading causes of liver failure and liver cancer, affecting around 3% of the world's population. The extreme sequence variability of the virus resulting from error-prone replication has thwarted the discovery of a universal prophylactic vaccine. It is known that vigorous and multispecific cellular immune responses, involving both helper CD4(+) and cytotoxic CD8(+) T cells, are associated with the spontaneous clearance of acute HCV infection. Escape mutations in viral epitopes can, however, abrogate protective T-cell responses, leading to viral persistence and associated pathologies. Despite the propensity of the virus to mutate, there might still exist substitutions that incur a fitness cost. In this paper, we identify groups of coevolving residues within HCV nonstructural protein 3 (NS3) by analyzing diverse sequences of this protein using ideas from random matrix theory and associated methods. Our analyses indicate that one of these groups comprises a large percentage of residues for which HCV appears to resist multiple simultaneous substitutions. Targeting multiple residues in this group through vaccine-induced immune responses should either lead to viral recognition or elicit escape substitutions that compromise viral fitness. Our predictions are supported by published clinical data, which suggested that immune genotypes associated with spontaneous clearance of HCV preferentially recognized and targeted this vulnerable group of residues. Moreover, mapping the sites of this group onto the available protein structure provided insight into its functional significance. An epitope-based immunogen is proposed as an alternative to the NS3 epitopes in the peptide-based vaccine IC41. IMPORTANCE Despite much experimental work on HCV, a thorough statistical study of the HCV sequences for the purpose of immunogen design was missing in the literature. Such a study is vital to identify epistatic couplings among residues that can provide useful insights for designing a potent vaccine. In this work, ideas from random matrix theory were applied to characterize the statistics of substitutions within the diverse publicly available sequences of the genotype 1a HCV NS3 protein, leading to a group of sites for which HCV appears to resist simultaneous substitutions possibly due to deleterious effect on viral fitness. Our analysis leads to completely novel immunogen designs for HCV. In addition, the NS3 epitopes used in the recently proposed peptide-based vaccine IC41 were analyzed in the context of our framework. Our analysis predicts that alternative NS3 epitopes may be worth exploring as they might be more efficacious.
Collapse
|
27
|
Morrow MP, Yan J, Sardesai NY. Human papillomavirus therapeutic vaccines: targeting viral antigens as immunotherapy for precancerous disease and cancer. Expert Rev Vaccines 2013; 12:271-83. [PMID: 23496667 DOI: 10.1586/erv.13.23] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Infections with oncogenic HPV types have the potential to lead to the induction of several types of cancer, notably cervical, vulvar, anal, and head and neck cancer. While prophylactic vaccines are currently available and show high efficacy against the establishment of HPV infection, low rates of initiation and lower rates of completion of the vaccination regimen, as well as the lack of an opportunity to be vaccinated prior to infection, has lead to the development of a patient population for whom no immune-based therapy for infection is available. In the current review the authors examine clinical approaches to HPV-targeted immune therapies, the bulk of which target the regulatory proteins E6 and E7 that are constitutively expressed in HPV-associated cancer cells. Early studies demonstrate a correlation between induction of T-cell responses and clearance of HPV-associated precancerous lesions. The clinical data corroborates these findings and highlight the importance of Th1 skewing. Improvements in our understanding of tumor immunology and development of more potent Th1-directed vaccine platforms make it feasible to foresee a HPV therapeutic vaccine in the coming years.
Collapse
Affiliation(s)
- Matthew P Morrow
- Inovio Pharmaceuticals, Inc., 1787 Sentry Parkway West, Blue Bell, PA 19422, USA
| | | | | |
Collapse
|
28
|
Lassmann B, Arumugaswami V, Chew KW, Lewis MJ. A new system to measure and compare hepatitis C virus replication capacity using full-length, replication competent viruses. J Virol Methods 2013; 194:82-8. [PMID: 23973740 DOI: 10.1016/j.jviromet.2013.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 08/06/2013] [Accepted: 08/09/2013] [Indexed: 11/16/2022]
Abstract
Measuring the in vitro replication capacity of viruses is an important tool for assessing the effects of selective pressure of immune responses and drug therapy. Measuring hepatitis C virus (HCV) replication capacity utilizing primarily sub-genomic reporter constructs is limited. To overcome some of these limitations a quantitative reverse transcriptase PCR (RT-qPCR) was designed to measure simultaneously the growth rate of 2 whole genome HCV variants under identical culture conditions. The assay demonstrates 100% specificity of detection of each variant and a linear detection range from 200 to 2×10(8) copies. The system was validated using a panel of HCV mutants, including the NS3 protease inhibitor drug resistance mutants R155K and T54A. The creation of a unique sequence tag results in highly sensitive and specific discrimination of parental JFH-FNX and modified clones using distinct probes in a RT-qPCR allowing for comparison of the effect of drug resistance or immune escape mutations on HCV replication. This system has advantages over existing methods both by permitting direct comparison of the replication capacity of fully replication-competent HCV mutants under identical culture conditions and by measuring effects on replication capacity due to mutations affecting all stages of the viral life cycle including entry and egress.
Collapse
Affiliation(s)
- Britta Lassmann
- Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | | | | | | |
Collapse
|
29
|
The role of chemokines in acute and chronic hepatitis C infection. Cell Mol Immunol 2013; 11:25-40. [PMID: 23954947 DOI: 10.1038/cmi.2013.37] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Revised: 07/08/2013] [Accepted: 07/14/2013] [Indexed: 12/12/2022] Open
Abstract
Hepatitis C imposes a significant burden on global healthcare. Chronic infection is associated with progressive inflammation of the liver which typically manifests in cirrhosis, organ failure and cancer. By virtue of elaborate evasion strategies, hepatitis C virus (HCV) succeeds as a persistent human virus. It has an extraordinary capacity to subvert the immune response enabling it to establish chronic infections and associated liver disease. Chemokines are low molecular weight chemotactic peptides that mediate the recruitment of inflammatory cells into tissues and back into the lymphatics and peripheral blood. Thus, they are central to the temporal and spatial distribution of effector and regulatory immune cells. The interactions between chemokines and their cognate receptors help shape the immune response and therefore, have a major influence on the outcome of infection. However, chemokines represent a target for modulation by viruses including the HCV. HCV is known to modulate chemokine expression in vitro and may therefore enable its survival by subverting the immune response in vivo through altered leukocyte chemotaxis resulting in impaired viral clearance and the establishment of chronic low-grade inflammation. In this review, the roles of chemokines in acute and chronic HCV infection are described with a particular emphasis placed on chemokine modulation as a means of immune subversion. We provide an in depth discussion of the part played by chemokines in mediating hepatic fibrosis while addressing the potential applications for these chemoattractants in prognostic medicine.
Collapse
|
30
|
Cell-cell contact-mediated hepatitis C virus (HCV) transfer, productive infection, and replication and their requirement for HCV receptors. J Virol 2013; 87:8545-58. [PMID: 23720720 DOI: 10.1128/jvi.01062-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hepatitis C virus (HCV) infection is believed to begin with interactions between cell-free HCV and cell receptors that include CD81, scavenger receptor B1 (SR-B1), claudin-1 (CLDN1), and occludin (OCLN). In this study, we have demonstrated that HCV spreading from infected hepatocytes to uninfected hepatocytes leads to the transfer of HCV and the formation of infection foci and is cell density dependent. This cell-cell contact-mediated (CCCM) HCV transfer occurs readily and requires all these known HCV receptors and an intact actin cytoskeleton. With a fluorescently labeled replication-competent HCV system, the CCCM transfer process was further dissected by live-cell imaging into four steps: donor cell-target cell contact, formation of viral puncta-target cell conjugation, transfer of viral puncta, and posttransfer. Importantly, the CCCM HCV transfer leads to productive infection of target cells. Taken together, these results show that CCCM HCV transfer constitutes an important and effective route for HCV infection and dissemination. These findings will aid in the development of new and novel strategies for preventing and treating HCV infection.
Collapse
|
31
|
Hepatitis C virus suppresses C9 complement synthesis and impairs membrane attack complex function. J Virol 2013; 87:5858-67. [PMID: 23487461 DOI: 10.1128/jvi.00174-13] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hepatitis C virus (HCV) proteins inhibit complement component expression, which may attenuate immunity against infection. In this study, we examined whether HCV regulates the membrane attack complex (MAC) via complement component C9. MAC is composed of C5b to C9 (C5b-9) and mediates cell lysis of invaded pathogens. Liver biopsy specimens from chronically HCV-infected patients exhibited a lower level of C9 mRNA expression than liver biopsy specimens from unrelated disease or healthy control human liver RNA. Hepatocytes infected with cell culture-grown HCV or expressing HCV core protein also displayed significant repression of C9 mRNA and protein levels. Promoter analysis suggested that the T cell factor-4 (TCF-4E) transcription factor is responsible for HCV core-mediated C9 promoter regulation. Sera from chronically HCV-infected patients displayed a lower level of C5b-9 and a reduced antimicrobial effect on model organisms compared to unrelated patient sera or sera from healthy volunteers. Together, these results for C9 regulation by HCV core protein coupled with functional impairment of the membrane attack complex underscore HCV-mediated attenuation of immune mechanisms.
Collapse
|