1
|
Kock KH, Tan LM, Han KY, Ando Y, Jevapatarakul D, Chatterjee A, Lin QXX, Buyamin EV, Sonthalia R, Rajagopalan D, Tomofuji Y, Sankaran S, Park MS, Abe M, Chantaraamporn J, Furukawa S, Ghosh S, Inoue G, Kojima M, Kouno T, Lim J, Myouzen K, Nguantad S, Oh JM, Rayan NA, Sarkar S, Suzuki A, Thungsatianpun N, Venkatesh PN, Moody J, Nakano M, Chen Z, Tian C, Zhang Y, Tong Y, Tan CTY, Tizazu AM, Loh M, Hwang YY, Ho RC, Larbi A, Ng TP, Won HH, Wright FA, Villani AC, Park JE, Choi M, Liu B, Maitra A, Pithukpakorn M, Suktitipat B, Ishigaki K, Okada Y, Yamamoto K, Carninci P, Chambers JC, Hon CC, Matangkasombut P, Charoensawan V, Majumder PP, Shin JW, Park WY, Prabhakar S. Asian diversity in human immune cells. Cell 2025; 188:2288-2306.e24. [PMID: 40112801 DOI: 10.1016/j.cell.2025.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/03/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
The relationships of human diversity with biomedical phenotypes are pervasive yet remain understudied, particularly in a single-cell genomics context. Here, we present the Asian Immune Diversity Atlas (AIDA), a multi-national single-cell RNA sequencing (scRNA-seq) healthy reference atlas of human immune cells. AIDA comprises 1,265,624 circulating immune cells from 619 donors, spanning 7 population groups across 5 Asian countries, and 6 controls. Though population groups are frequently compared at the continental level, we found that sub-continental diversity, age, and sex pervasively impacted cellular and molecular properties of immune cells. These included differential abundance of cell neighborhoods as well as cell populations and genes relevant to disease risk, pathogenesis, and diagnostics. We discovered functional genetic variants influencing cell-type-specific gene expression, which were under-represented in non-Asian populations, and helped contextualize disease-associated variants. AIDA enables analyses of multi-ancestry disease datasets and facilitates the development of precision medicine efforts in Asia and beyond.
Collapse
Affiliation(s)
- Kian Hong Kock
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Le Min Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Yoshinari Ando
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Laboratory for Transcriptome Technology, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Damita Jevapatarakul
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Ankita Chatterjee
- John C. Martin Centre for Liver Research and Innovations, Sonarpur, Kolkata 700150, India
| | - Quy Xiao Xuan Lin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Eliora Violain Buyamin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Radhika Sonthalia
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Deepa Rajagopalan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Yoshihiko Tomofuji
- Laboratory for Systems Genetics, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Statistical Genetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shvetha Sankaran
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Mi-So Park
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Mai Abe
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Juthamard Chantaraamporn
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Seiko Furukawa
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Supratim Ghosh
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India
| | - Gyo Inoue
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Miki Kojima
- Laboratory for Transcriptome Technology, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Tsukasa Kouno
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Jinyeong Lim
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Keiko Myouzen
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Sarintip Nguantad
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Jin-Mi Oh
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Nirmala Arul Rayan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Sumanta Sarkar
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Narita Thungsatianpun
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Prasanna Nori Venkatesh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Jonathan Moody
- Laboratory for Genome Information Analysis, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Masahiro Nakano
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan
| | - Ziyue Chen
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore
| | - Chi Tian
- Department of Pharmacy, Faculty of Science, National University of Singapore (NUS), Singapore 117543, Singapore
| | - Yuntian Zhang
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine (YLLSoM), NUS, Singapore 119228, Singapore
| | - Yihan Tong
- Department of Pharmacy, Faculty of Science, National University of Singapore (NUS), Singapore 117543, Singapore
| | - Crystal T Y Tan
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Anteneh Mehari Tizazu
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Marie Loh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Nanyang Technological University (NTU), Lee Kong Chian School of Medicine (LKCMedicine), 11 Mandalay Road, Singapore 308232, Singapore
| | - You Yi Hwang
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Roger C Ho
- Department of Psychological Medicine, YLLSoM, NUS, 1E Kent Ridge Road, Singapore 119228, Singapore; Institute for Health Innovation & Technology, NUS, 14 Medical Drive, Singapore 117599, Singapore
| | - Anis Larbi
- Singapore Immunology Network (SIgN), A(∗)STAR, 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Tze Pin Ng
- Department of Geriatric Medicine, Khoo Teck Puat Hospital, Singapore 768828, Singapore; St Luke's Hospital, Singapore 659674, Singapore; Geriatric Education and Research Institute, Singapore 768024, Singapore
| | - Hong-Hee Won
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea; Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University, Seoul 06351, Republic of Korea
| | - Fred A Wright
- Department of Biological Sciences, Bioinformatics Research Center, and Department of Statistics, North Carolina State University, Raleigh, NC 27695, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, and Mass General Cancer Center, Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon 34051, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Boxiang Liu
- Department of Pharmacy, Faculty of Science, National University of Singapore (NUS), Singapore 117543, Singapore; Department of Biomedical Informatics, Yong Loo Lin School of Medicine (YLLSoM), NUS, Singapore 119228, Singapore; Precision Medicine Translational Research Programme, NUS Centre for Cancer Research, and Cardiovascular-Metabolic Disease Translational Research Programme, YLLSoM, NUS, Singapore 119228, Singapore
| | - Arindam Maitra
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics, Kalyani, West Bengal 741251, India
| | - Manop Pithukpakorn
- Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Bhoom Suktitipat
- Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand; Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yukinori Okada
- Laboratory for Systems Genetics, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Department of Statistical Genetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8654, Japan; Laboratory of Statistical Immunology, Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Premium Research Institute for Human Metaverse Medicine, Osaka University, Suita 565-0871, Japan
| | - Kazuhiko Yamamoto
- Laboratory for Autoimmune Diseases, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Piero Carninci
- Laboratory for Transcriptome Technology, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Genomics Research Center, Fondazione Human Technopole, Viale Rita Levi-Montalcini, 1 - Area MIND, Milano, Lombardy 20157, Italy
| | - John C Chambers
- Nanyang Technological University (NTU), Lee Kong Chian School of Medicine (LKCMedicine), 11 Mandalay Road, Singapore 308232, Singapore
| | - Chung-Chau Hon
- Laboratory for Genome Information Analysis, RIKEN Center for IMS, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan; Graduate School of Integrated Sciences for Life, Hiroshima University, 1-3-3-2 Kagamiyama, Higashihiroshima, Hiroshima 739-0046, Japan
| | - Ponpan Matangkasombut
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Varodom Charoensawan
- Single-cell omics and Systems Biology of Diseases (scSyBiD) Research Unit, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Integrative Computational BioScience Center, Mahidol University, Nakhon Pathom 73170, Thailand; Siriraj Genomics, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand; School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand
| | - Partha P Majumder
- John C. Martin Centre for Liver Research and Innovations, Sonarpur, Kolkata 700150, India; Indian Statistical Institute, 203 B.T. Road, Kolkata 700108, India
| | - Jay W Shin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul 06351, Republic of Korea.
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A(∗)STAR), 60 Biopolis Street, Genome, Singapore 138672, Singapore; Nanyang Technological University (NTU), Lee Kong Chian School of Medicine (LKCMedicine), 11 Mandalay Road, Singapore 308232, Singapore; Cancer Science Institute of Singapore, NUS, 14 Medical Drive, Singapore 117599, Singapore.
| |
Collapse
|
2
|
Pizzarello CR, Jackson CM, Herman K, Seppo AE, Rebhahn J, Scherzi T, Berin MC, Looney RJ, Mosmann TR, Järvinen KM. A Phenotypically Distinct Human Th2 Cell Subpopulation Is Associated With Development of Allergic Disorders in Infancy. Allergy 2025; 80:949-964. [PMID: 39899007 PMCID: PMC11971024 DOI: 10.1111/all.16489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 11/18/2024] [Accepted: 12/26/2024] [Indexed: 02/04/2025]
Abstract
BACKGROUND Little is known about the ontogeny of T cell immunity during infancy in farming and urban lifestyles due to the lack of immunophenotyping in such birth cohorts. METHODS Two birth cohorts (farming and urban) at differing risks and rates of allergic diseases were compared. Blood mononuclear cells were collected from infants at birth, and 6 and 12 months of age. Full spectrum flow cytometry, followed by traditional gating and the Scalable Weighted Iterative Flow-clustering Technique (SWIFT) high-dimensional analysis, were used to identify cell populations that differed between farming and urban infants. Additionally, single-cell RNAseq and multiplex cytokine assays were used to assess the function of cell populations of interest. RESULTS Several regulatory T cell (Treg) subpopulations were elevated in farming lifestyles and in non-atopic infants. A unique effector memory CD25+CD127+CD161-CD49d+CCR4+CRTH2+ Th2 population was elevated at 6 months in urban infants and in those who developed atopic dermatitis and/or food allergy and allergic sensitization. Although this population shared Th2 and IL-9 skewing with Th2A cells, the population uniquely failed to express CD161, produced more IL-2 and TNF-α, and upregulated the differentially expressed genes (DEGs), FOXP3 and the cytokine inducible SH2-containing protein gene (CISH) relative to Th2A cells. This population has been termed Th2B cells. CONCLUSION We describe a unique effector memory Th2 population elevated in urban high-risk infants, potentially implicated in the development of allergic disease.
Collapse
Affiliation(s)
- Catherine R Pizzarello
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Courtney M Jackson
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
| | - Katherine Herman
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
| | - Jonathan Rebhahn
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Tyler Scherzi
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - M Cecilia Berin
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - R John Looney
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Tim R Mosmann
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| |
Collapse
|
3
|
Ali AA, Tabll AA. Unlocking potential: Virus-like particles as a promising strategy for effective HCV vaccine development. Virology 2025; 602:110307. [PMID: 39580887 DOI: 10.1016/j.virol.2024.110307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/01/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024]
Abstract
Hepatitis C virus (HCV) is a leading cause of liver disease worldwide. The development of prophylactic vaccine is essential for HCV global eradication. Despite over three decades of research, no effective vaccine for HCV has been developed, primarily due to the virus's genetic diversity, immune evasion mechanisms, and incomplete understanding of protective immunity. However, Virus-Like Particles (VLPs) offer a promising approach to overcoming these challenges. VLPs mimic the structure of native virus but without the infectious genome, making them safe and non-infectious vaccines candidates. The capability of VLPs to incorporate neutralizing and conformational epitopes, and engage humoral and cellular immune responses, positions them as a promising tool for overcoming challenges associated with the HCV vaccine development. This review examines the challenges and immunological considerations for HCV vaccine development and provides an overview of the VLPs-based vaccines development. It also discusses future directions and public health implications of HCV vaccine development.
Collapse
Affiliation(s)
- Ahmed A Ali
- Molecular Biology Department, Biotechnology Research Institute, National Research Centre, (NRC), 12622, Cairo, Egypt.
| | - Ashraf A Tabll
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, 12622, Cairo, Egypt; Egyptian Centre for Research and Regenerative Medicine (ECRRM), 11517, Cairo, Egypt.
| |
Collapse
|
4
|
Dejanović B, Barak O, Čolović P, Janjić N, Savić Ž, Gvozdanović N, Ružić M. Hospital Mortality in Acute Decompensation of Alcoholic Liver Cirrhosis: Can Novel Survival Markers Outperform Traditional Ones? J Clin Med 2024; 13:6208. [PMID: 39458158 PMCID: PMC11508931 DOI: 10.3390/jcm13206208] [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: 09/21/2024] [Revised: 10/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Background: There is a strong correlation between systemic inflammation intensity and clinical presentation, disease progression, and survival during liver cirrhosis decompensation. This study aimed to evaluate the prognostic performance of blood-based biomarkers as meta-inflammation markers, including NLR, PLR, LMR, INPR, MPR, ALBI, FIB4, and APRI, in predicting hospital mortality in patients with acute decompensation of alcohol-related liver cirrhosis. Methods: Data from 411 patients with their first onset of acute decompensation were analyzed, forming two groups: deceased and survived during hospitalization. Generalized partial least squares regression analysis was applied to explore the effects of surrogate indicators on mortality rates, using mortality rate as the dependent variable. Root Mean Square Error, Akaike's, and Bayesian information criteria determined that four components accounted for most of the variance. Results: Variables with significant negative contributions to the outcome prediction (ranked by standardized regression coefficients) were encephalopathy grade, total bilirubin, Child-Turcotte-Pugh score, MELD, NLR, MPV, FIB4, INR, PLR, and ALT. Coefficient sizes ranged from -0.63 to -0.09, with p-values from 0 to 0.018. Conclusions: NLR, PLR, and FIB4 significantly contribute to hospital mortality prediction in patients with acute decompensation of alcohol-related liver cirrhosis. Conversely, some variables used to predict liver disease severity, including INPR, APRI, LMR, and ALBI score, did not significantly contribute to hospital mortality prediction in this patient population.
Collapse
Affiliation(s)
- Božidar Dejanović
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (O.B.); (N.J.); (Ž.S.); (N.G.); (M.R.)
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Otto Barak
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (O.B.); (N.J.); (Ž.S.); (N.G.); (M.R.)
| | - Petar Čolović
- Department of Psychology, Faculty of Philosophy, University of Novi Sad, 21000 Novi Sad, Serbia;
| | - Nebojša Janjić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (O.B.); (N.J.); (Ž.S.); (N.G.); (M.R.)
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Željka Savić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (O.B.); (N.J.); (Ž.S.); (N.G.); (M.R.)
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Nikola Gvozdanović
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (O.B.); (N.J.); (Ž.S.); (N.G.); (M.R.)
- Clinic of Gastroenterology and Hepatology, University Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| | - Maja Ružić
- Faculty of Medicine, University of Novi Sad, 21000 Novi Sad, Serbia; (O.B.); (N.J.); (Ž.S.); (N.G.); (M.R.)
- Clinic of Infectious Disease, University Clinical Center of Vojvodina, 21000 Novi Sad, Serbia
| |
Collapse
|
5
|
Doyle EH, Aloman C, El-Shamy A, Eng FJ, Kim-Schulze S, Rahman A, Schiano T, Heeger P, Branch AD. Imprinted immune abnormalities in liver transplant patients cured of hepatitis C with antiviral drugs. Liver Transpl 2024; 30:728-741. [PMID: 38315053 DOI: 10.1097/lvt.0000000000000342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/11/2024] [Indexed: 02/07/2024]
Abstract
Chronic HCV infection induces interferon and dysregulates immune responses through inflammation and chronic antigenic stimulation. Antiviral drugs can cure HCV, providing a unique opportunity to examine the immunological restoration that does and does not occur when a chronic viral infection is eradicated. We quantified blood cytokines levels and used mass cytometry to immunophenotype peripheral blood mononuclear cells before and after HCV cure in 2 groups of patients and controls. At baseline, serum interferon α and soluble CD163 (a macrophage product) were elevated in both liver transplant and nonliver transplant patients compared to controls; the frequencies of several peripheral blood mononuclear cell populations differed from controls; and programmed death protein 1-positivity was increased in nearly all T cell subsets. Many abnormalities persisted after HCV cure, including elevated programmed death protein 1 expression on CD4 naïve and central memory T cells, elevated soluble CD163, and expansion of the plasmablast/plasma cell compartment. Several myeloid-lineage subsets, including Ag-presenting dendritic cells, remained dysregulated. In mechanistic studies, interferon α treatment increased programmed death protein 1 on human T cells and increased T cell receptor signaling. The data identify immunological abnormalities that persist after curative HCV treatment. Before cure, high levels of interferon α may stimulate programmed death protein 1 expression on human T cells, causing persistent functional changes.
Collapse
MESH Headings
- Humans
- Liver Transplantation/adverse effects
- Male
- Antiviral Agents/therapeutic use
- Middle Aged
- Female
- Antigens, CD/immunology
- Antigens, CD/blood
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/blood
- Antigens, Differentiation, Myelomonocytic/immunology
- Hepatitis C, Chronic/immunology
- Hepatitis C, Chronic/drug therapy
- Hepatitis C, Chronic/blood
- Hepatitis C, Chronic/surgery
- Interferon-alpha/therapeutic use
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Cell Surface/blood
- Receptors, Cell Surface/immunology
- Adult
- Case-Control Studies
- Aged
- Hepacivirus/immunology
- Hepacivirus/drug effects
- Leukocytes, Mononuclear/immunology
- Cytokines/blood
- Immunophenotyping
- Treatment Outcome
Collapse
Affiliation(s)
- Erin H Doyle
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Costica Aloman
- Department of Surgery, Westchester Medical Center, New York Medical College, Valhalla, New York, USA
| | - Ahmed El-Shamy
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- College of Graduate Studies, Master of Pharmaceutical Sciences Program
| | - Francis J Eng
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Seunghee Kim-Schulze
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Adeeb Rahman
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Thomas Schiano
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Peter Heeger
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrea D Branch
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
6
|
Acute Systemic White Blood Cell Changes following Degenerative Cervical Myelopathy (DCM) in a Mouse Model. Int J Mol Sci 2022; 23:ijms231911496. [PMID: 36232808 PMCID: PMC9570488 DOI: 10.3390/ijms231911496] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022] Open
Abstract
Degenerative cervical myelopathy (DCM) is caused by age-related degeneration of the cervical spine, causing chronic spinal cord compression and inflammation. The aim of this study was to assess whether the natural progression of DCM is accompanied by hematological changes in the white blood cell composition. If so, these changes can be used for diagnosis complementing established imaging approaches and for the development of treatment strategies, since peripheral immunity affects the progression of DCM. Gradual compression of the spinal cord was induced in C57B/L mice at the C5-6 level. The composition of circulating white blood cells was analyzed longitudinally at four time points after induction of DCM using flow cytometry. At 12 weeks, serum cytokine levels were measured using a Luminex x-MAP assay. Neurological impairment in the mouse model was also assessed using the ladder walk test and CatWalk. Stepping function (* p < 0.05) and overground locomotion (*** p < 0.001) were impaired in the DCM group. Importantly, circulating monocytes and T cells were affected primarily at 3 weeks following DCM. T cells were two-fold lower in the DCM group (*** p < 0.0006), whereas monocytes were four-fold increased (*** p < 0.0006) in the DCM compared with the sham group. Our data suggest that changes in white blood cell populations are modest, which is unique to other spinal cord pathologies, and precede the development of neurobehavioral symptoms.
Collapse
|
7
|
Willuweit K, Frey A, Passenberg M, Korth J, Saka N, Anastasiou OE, Möhlendick B, Schütte A, Schmidt H, Rashidi-Alavijeh J. Patients with Liver Cirrhosis Show High Immunogenicity upon COVID-19 Vaccination but Develop Premature Deterioration of Antibody Titers. Vaccines (Basel) 2022; 10:vaccines10030377. [PMID: 35335009 PMCID: PMC8949848 DOI: 10.3390/vaccines10030377] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/15/2022] [Accepted: 02/22/2022] [Indexed: 02/04/2023] Open
Abstract
SARS-CoV-2 infection is known to lead to severe morbidity and mortality in patients with liver cirrhosis. For this reason, vaccination of these patients against COVID-19 is widely recommended. However, data regarding immunogenicity in patients with liver cirrhosis is limited and even less is known about the kinetics of antibody response, as well as the optimal timing of booster immunization. We analyzed immunogenicity in 110 patients with liver cirrhosis after receiving two doses of the mRNA-based vaccine BNT162b2 following the standard protocol and compared these results to a control group consisting of 80 healthcare workers. One hundred and six patients with liver cirrhosis (96%) developed antibodies against SARS-CoV-2, compared to 79 (99%) in the control group (p = 0.400). Still, the median SARS-CoV-2 IgG titer was significantly lower in patients with liver cirrhosis compared to the control group (939 vs. 1905 BAU/mL, p = 0.0001). We also analyzed the strength of the antibody response in relation to the time between the second dose and antibody detection. Antibody titers remained relatively stable in the control group while showing a rapid and significant decrease in patients with liver cirrhosis. In conclusion, our data reveals a favorable initial outcome after vaccination with the COVID-19 vaccine BNT162b2 in cirrhotic patients but show a rapid deterioration of the antibody response after time, thereby giving a strong hint towards the importance of early booster immunization for this group of patients.
Collapse
Affiliation(s)
- Katharina Willuweit
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (K.W.); (A.F.); (M.P.); (N.S.); (A.S.); (H.S.)
| | - Alexandra Frey
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (K.W.); (A.F.); (M.P.); (N.S.); (A.S.); (H.S.)
| | - Moritz Passenberg
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (K.W.); (A.F.); (M.P.); (N.S.); (A.S.); (H.S.)
| | - Johannes Korth
- Department of Nephrology, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany;
| | - Nissrin Saka
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (K.W.); (A.F.); (M.P.); (N.S.); (A.S.); (H.S.)
| | - Olympia E. Anastasiou
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 179, 45147 Essen, Germany;
| | - Birte Möhlendick
- Institute of Pharmacogenetics, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany;
| | - Andreas Schütte
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (K.W.); (A.F.); (M.P.); (N.S.); (A.S.); (H.S.)
| | - Hartmut Schmidt
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (K.W.); (A.F.); (M.P.); (N.S.); (A.S.); (H.S.)
| | - Jassin Rashidi-Alavijeh
- Department of Gastroenterology, Hepatology and Transplant Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany; (K.W.); (A.F.); (M.P.); (N.S.); (A.S.); (H.S.)
- Correspondence: ; Tel.: +49-201-723-84001
| |
Collapse
|
8
|
Auma AWN, Shive CL, Kostadinova L, Anthony DD. Variable Normalization of Naïve CD4+ Lymphopenia and Markers of Monocyte and T Cell Activation over the Course of Direct-Acting Anti-Viral Treatment of Chronic Hepatitis C Virus Infection. Viruses 2021; 14:50. [PMID: 35062255 PMCID: PMC8780994 DOI: 10.3390/v14010050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic hepatitis C virus (HCV) infection is associated with naïve CD4+ T cell lymphopenia and long-standing/persistent elevation of cellular and soluble immune activation parameters, the latter heightened in the setting of HIV co-infection. The underlying mechanisms are not completely understood. However, we recently reported that accelerated peripheral cell death may contribute to naïve CD4+ T cell loss and that mechanistic relationships between monocyte activation, T cell activation, and soluble inflammatory mediators may also contribute. Chronic HCV infection can be cured by direct-acting anti-viral (DAA) therapy, and success is defined as sustained virological response (SVR, undetectable HCV RNA (ribonucleic acid) at 12 weeks after DAA treatment completion). However, there is no general consensus on the short-term and long-term immunological outcomes of DAA therapy. Here, we consolidate previous reports on the partial normalization of naïve CD4+ lymphopenia and T cell immune activation and the apparent irreversibility of monocyte activation following DAA therapy in HCV infected and HCV/HIV co-infected individuals. Further, advanced age and cirrhosis are associated with delayed or abrogation of immune reconstitution after DAA therapy, an indication that non-viral factors also likely contribute to host immune dysregulation in HCV infection.
Collapse
Affiliation(s)
- Ann W. N. Auma
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (A.W.N.A.); (C.L.S.)
| | - Carey L. Shive
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (A.W.N.A.); (C.L.S.)
- Cleveland VA Medical Center, Cleveland, OH 44106, USA;
| | | | - Donald D. Anthony
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA; (A.W.N.A.); (C.L.S.)
- Cleveland VA Medical Center, Cleveland, OH 44106, USA;
- Metro Health Medical Center, Division of Rheumatology, Cleveland, OH 44106, USA
| |
Collapse
|
9
|
A 56-Year-Old Woman with Chronic Hepatitis C Liver Disease and Meningitis due to Streptococcus equi subsp. Zooepidemicus. Case Rep Crit Care 2021; 2021:7227054. [PMID: 34631173 PMCID: PMC8500762 DOI: 10.1155/2021/7227054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 11/25/2022] Open
Abstract
Streptococcus equi subsp. zooepidemicus (S. equi subsp. zooepidemicus), which carries the Lancefield group C antigen, is an uncommon human pathogen. It is considered an opportunistic commensal of the equine upper respiratory tract and causes invasive infections in immunocompromised hosts, following close contact to infected horses. Meningitis caused by S. equi subsp. zooepidemicus is a rare infectious disease with high rates of complications. We present the case of a 56-year-old female with acutely altered mental status following three days of fever and vomiting. For several months, she was taking care of horses. The most relevant preexisting illnesses were chronic hepatitis C infection and traumatic paraplegia due to spinal cord injury 30 years ago. Laboratory evaluation on admission revealed leukocytosis, hyponatremia, and elevated C-reactive protein. Cerebral CT scan showed diffuse cerebral edema. Whereas cerebrospinal fluid real-time PCR assay for common pathogens was negative, cultures showed S. equi subsp. zooepidemicus. She recovered fully after intravenous administration of ceftriaxone for four weeks. This is one of only few reported cases of S. equi subsp. zooepidemicus meningitis and the first case in chronic hepatitis C infection. Our case supports the necessity for extended microbiological examination especially in immunocompromised patients if PCR examination for common pathogens is negative.
Collapse
|
10
|
Abstract
Hepatitis C virus (HCV) is a small positive-sense, single-stranded RNA virus, the causal organism for chronic hepatitis. Chronic hepatitis leads to inflammation of liver, causing cirrhosis, fibrosis and steatosis, which may ultimately lead to liver cancer in a few cases. Innate and adaptive immune responses play an important role in the pathogenesis of HCV infection, thus acting as an important component in deciding the fate of the disease. Numerous studies have indicated that the derangement of these immune responses results in the persistence of infection leading to chronic state of the disease. Interactions between virus and host immune system generally result in the elimination of virus, but as the virus evolves with different evading mechanisms, it makes environment favourable for its survival and replication. It has been reported that HCV impairs the immune system by functional modulation of the cells of innate as well as adaptive immune responses, resulting in chronic state of the disease, influencing the response to antiviral therapy in these patients. These defects in the immune system lead to suboptimal immune responses and therefore, impaired effector functions. This review highlights the involvement or association of different immune cells such as natural killer cells, B cells, dendritic cells and T cells in HCV infection and how the virus plays a role in manipulating certain regulatory mechanisms to make these cells dysfunctional for its own persistence and survival.
Collapse
Affiliation(s)
- Shallu Tomer
- Department of Immunopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Sunil K Arora
- Department of Immunopathology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
11
|
Auma AWN, Shive CL, Lange A, Damjanovska S, Kowal C, Zebrowski E, Pandiyan P, Wilson B, Kalayjian RC, Canaday DH, Anthony DD. Naïve CD4+ T Cell Lymphopenia and Apoptosis in Chronic Hepatitis C Virus Infection Is Driven by the CD31+ Subset and Is Partially Normalized in Direct-Acting Antiviral Treated Persons. Front Immunol 2021; 12:641230. [PMID: 33912168 PMCID: PMC8075159 DOI: 10.3389/fimmu.2021.641230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 03/24/2021] [Indexed: 12/29/2022] Open
Abstract
Background The mechanisms underlying naïve CD4+ lymphopenia during chronic Hepatitis C Virus (HCV) infection are unclear. Whether direct-acting antiviral (DAA) therapy restores peripheral naïve CD4+ T cell numbers and function is unknown. Methods We enumerated frequencies and counts of peripheral naïve CD4+, CD4+CD31+ and CD4+CD31- T cells by flow cytometry in a cross sectional analysis comparing chronic HCV infected (n=34), DAA-treated(n=29), and age-range matched controls (n=25), as well as in a longitudinal cohort of HCV DAA treated persons (n=16). The cross-sectional cohort was stratified by cirrhosis state. Cell apoptosis/survival (AnnexinV+7AAD+/BCL-2 labeling) and cell cycle entry (Ki67 expression) of CD31+ and CD31- naïve CD4+ T cells was analyzed directly ex vivo and following 3 and 5 days of in vitro culture with media, interleukin (IL) -7 or CD3/CD28 activator. Results In the cross-sectional cohort, naïve CD4+ proportions were lower in chronic HCV infected persons compared to controls and DAA-treated persons, an effect in part attributed to cirrhosis. Age was associated with naïve cell counts and proportions in HCV infected and treated persons as well. Naïve CD4+ cell proportions negatively correlated with plasma levels of soluble CD14 following therapy in DAA-treated persons. Naïve CD4+ cells from HCV infected persons exhibited greater direct ex vivo apoptosis and cell-cycling compared to cells from DAA-treated persons and controls, and this was localized to the CD4+CD31+ subset. On the other hand, no remarkable differences in expression of BCL-2 or IL-7 Receptor (CD127) at baseline or following in vitro media or IL7 containing culture were observed. In the longitudinal cohort, naïve CD4+CD31+/CD31- ratio tended to increase 24 weeks after DAA therapy initiation. Conclusions Activation and apoptosis of peripheral naïve CD4+CD31+ T cells appear to contribute to naïve CD4+ lymphopenia in chronic HCV infection, and this defect is partially reversible with HCV DAA therapy. Age and cirrhosis -associated naïve CD4+ lymphopenia is present both before and after HCV DAA therapy. These findings have implications for restoration of host immune function after DAA therapy.
Collapse
Affiliation(s)
- Ann W N Auma
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Carey L Shive
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States.,GRECC, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | - Alyssa Lange
- GRECC, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | - Sofi Damjanovska
- GRECC, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | - Corinne Kowal
- GRECC, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | | | - Pushpa Pandiyan
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States
| | - Brigid Wilson
- GRECC, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | - Robert C Kalayjian
- Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, United States
| | - David H Canaday
- GRECC, VA Northeast Ohio Healthcare System, Cleveland, OH, United States
| | - Donald D Anthony
- Department of Pathology, Case Western Reserve University, Cleveland, OH, United States.,GRECC, VA Northeast Ohio Healthcare System, Cleveland, OH, United States.,Department of Medicine, MetroHealth Medical Center, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
12
|
Rueschenbaum S, Ciesek S, Queck A, Widera M, Schwarzkopf K, Brüne B, Welsch C, Wedemeyer H, Zeuzem S, Weigert A, Lange CM. Dysregulated Adaptive Immunity Is an Early Event in Liver Cirrhosis Preceding Acute-on-Chronic Liver Failure. Front Immunol 2021; 11:534731. [PMID: 33574809 PMCID: PMC7870861 DOI: 10.3389/fimmu.2020.534731] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Introduction Acute-on-chronic liver failure (ACLF) is characterized by high levels of systemic inflammation and parallel suppression of innate immunity, whereas little is known about adaptive immune immunity in ACLF. We therefore aimed to characterize the development of the adaptive immune system during the progression of liver cirrhosis to ACLF. Patients with compensated/stable decompensated liver cirrhosis, acute decompensation of liver cirrhosis, or ACLF were recruited from a prospective cohort study. Comprehensive immunophenotyping was performed using high dimensional flow cytometry. Replication of Torque teno (TT) virus was quantified as a marker of immunosuppression. High frequencies of detectable TT virus were observed already in patients with compensated/stable decompensated liver cirrhosis compared to healthy controls (>50% vs. 19%), suggesting relatively early occurrence of immunosuppression in cirrhosis. In line, profoundly reduced numbers of distinct innate and adaptive immune cell populations were observed before ACLF development. These changes were accompanied by parallel upregulation of co-stimulatory (e.g. CD40L, OX40, CD69, GITR, TIM-1) and inhibitory immune checkpoints (e.g. PDPN, PROCR, 2B4, TIGIT) on CD4+ and CD8+ T cells, which again preceded the development of ACLF. On a functional basis, the capacity of CD4+ and CD8+ T cells to produce pro-inflammatory cytokines upon stimulation was strongly diminished in patients with acute decompensation of liver cirrhosis and ACLF. Conclusion Impaired innate and—in particular—adaptive cellular immunity occurs relatively early in the pathogenesis of liver cirrhosis and precedes ACLF. This may contribute to the development of ACLF by increasing the risk of infections in patients with liver cirrhosis.
Collapse
Affiliation(s)
- Sabrina Rueschenbaum
- Department of Gastroenterology and Hepatology, University Hospital and University of Duisburg-Essen, Essen, Germany.,Department of Internal Medicine 1, Goethe-University Hospital Frankfurt, Frankfurt, Germany
| | - Sandra Ciesek
- Institute of Virology, University Hospital Essen, Essen, Germany
| | - Alexander Queck
- Department of Internal Medicine 1, Goethe-University Hospital Frankfurt, Frankfurt, Germany
| | - Marek Widera
- Institute of Virology, University Hospital Essen, Essen, Germany
| | - Katharina Schwarzkopf
- Department of Internal Medicine 1, Goethe-University Hospital Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Faculty of Medicine, Institute of Biochemistry 1, Goethe-University Frankfurt, Frankfurt, Germany
| | - Christoph Welsch
- Department of Internal Medicine 1, Goethe-University Hospital Frankfurt, Frankfurt, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology and Hepatology, University Hospital and University of Duisburg-Essen, Essen, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine 1, Goethe-University Hospital Frankfurt, Frankfurt, Germany
| | - Andreas Weigert
- Faculty of Medicine, Institute of Biochemistry 1, Goethe-University Frankfurt, Frankfurt, Germany
| | - Christian M Lange
- Department of Gastroenterology and Hepatology, University Hospital and University of Duisburg-Essen, Essen, Germany.,Department of Internal Medicine 1, Goethe-University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
13
|
Nakamura A, Yoshimura T, Deguchi M, Hosokawa Y, Someya G, Satoh T, Ichikawa T, Okuyama K, Yosioka Y, Asakura H. The significance of measuring peripheral blood lymphocytosis in liver cirrhosis-Diagnosis of cirrhosis-associated immune dysfunction using hemogram. KANZO 2020; 61:552-563. [DOI: 10.2957/kanzo.61.552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Astushi Nakamura
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | | | - Manami Deguchi
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | - Yuei Hosokawa
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | - Go Someya
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | - Tomomi Satoh
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | - Takeshi Ichikawa
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | - Keiji Okuyama
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | - Yosihiro Yosioka
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| | - Hitoshi Asakura
- Nippon Koukan Hospital Center for Gastrointestinal and Hepatic Diseases
| |
Collapse
|
14
|
Romani S, Stafford K, Nelson A, Bagchi S, Kottilil S, Poonia B. Peripheral PD-1 + T Cells Co-expressing Inhibitory Receptors Predict SVR With Ultra Short Duration DAA Therapy in HCV Infection. Front Immunol 2019; 10:1470. [PMID: 31316516 PMCID: PMC6610534 DOI: 10.3389/fimmu.2019.01470] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
Direct acting antiviral (DAA) regimens of 12 weeks result in HCV clearance in vast majority of patients across genotypes. We previously demonstrated an ultra-short regimen of 4 weeks DAA cleared HCV in a subset of patients. Here, we hypothesized that individual level of antiviral immunity differentially influenced viral clearance and investigated biomarkers of a successful response. Cohorts of HCV patients treated for 4 weeks with DAA therapy who either achieved sustained virologic response (SVR) or relapsed were compared at baseline and at end of therapy (EOT) for immune cell phenotypes and HCV specific immunity. Higher levels of PD-1+ CD8+ and CD4+ T lymphocytes co-expressing inhibitory receptors (IR) were present at baseline and at EOT in HCV patients who eventually achieved SVR compared with those who relpased. HCV specific CD8+ T cells were predominantly contained within these IR expressing PD-1+ subsets. Patients in the SVR group had significantly higher CD8+ T cell degranulation in response to HCV peptides at baseline and higher levels of cytokine producing T cells at EOT time-point, relative to those who relapsed. In ex vivo cultures, PD-1+CD160+ CD8+ T cells had higher HCV specific degranulation and PD-1+2B4+ CD8+ T cells had higher cytokine expression (IFNγ+TNFα+ or IFNγ+CD107a+) compared with single or no IR expressing subsets, indicating higher virus specific functional capacity of these subsets. Receiver operating characteristics curve (ROC) for baseline circulating frequencies of PD-1+CD160+, PD-1+Tim-3+ CD8+ T cells and PD-1+CD160+, PD-1+Blimp-1+, PD-1-CTLA4+ CD4+ T cells respectively, had associated C-statistics of 0.8214 and 0.9451 for discriminatin of patients who successfully cleared HCV with 4 weeks treatment. Thus, PD-1+ virus-specific CD8+ T cell subsets with cytotoxic capacity are present in a subset of chronic HCV infected individuals that associate with ability to achieve SVR, indicating role of immunity in DAA mediated viral clearance with short duration therapy.
Collapse
Affiliation(s)
- Sara Romani
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Kristen Stafford
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Amy Nelson
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shashwatee Bagchi
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shyam Kottilil
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Bhawna Poonia
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
15
|
Cereceda-Monteoliva N, Lewis H, Al-Himdani S, Stone C. Periorbital necrotising fasciitis with underlying undiagnosed hepatitis C infection. BMJ Case Rep 2019; 12:e223720. [PMID: 31015232 PMCID: PMC6506045 DOI: 10.1136/bcr-2017-223720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2019] [Indexed: 11/04/2022] Open
Abstract
We report the case of a 56-year-old man, previously well, who presented with a spontaneous right-sided periorbital necrotising soft tissue infection and subsequently found to have undiagnosed hepatitis C and liver cirrhosis. The patient presented with rapid onset right eye pain, periorbital swelling and septic shock. CT scan revealed diffuse inflammatory changes to the soft tissue anterior to the right eye. The initial treatment included intravenous antibiotics, emergency debridement of necrotic tissue and admission to intensive care. Group A streptococcus was cultured from the debrided tissue. The patient developed decompensated liver failure and life-threatening haematemesis. Liver screening detected hepatitis C positive serology, the only risk factor for which was an old tattoo. The patient was effectively managed by early involvement of multiple clinical teams. We review the literature surrounding periorbital necrotising fasciitis, discuss the evidence for hepatic disorders as a potential cause and make recommendations for managing these patients.
Collapse
Affiliation(s)
| | - Hannah Lewis
- Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | | | | |
Collapse
|
16
|
Abstract
The syndrome of decreased immunity caused by cirrhosis is a combination of different immunological mechanisms and reactions which result from an advanced stage of the liver disease. The synthesis of proteins of the acute phase becomes impaired, there develop different deficiencies of the complement system, and there ensues a decrease of receptors that are meant to recognize antigens. The negative changes become apparent in the field of cell responses, e.g. there are changes in the amounts of generated monocytes and macrophages, and their phagocytic capabilities and chemotaxic reactions are impacted as well. The humoral response results in distorted synthesis of particular antigen categories. The risk of detrimental immunoresponses with the end result of endotoxemia is not rarely coupled with both local and global infections. The combination of the aforesaid immunodeficiencies worsens the healing chances of cirrhosis sufferers and more often than not it increases the mortality of the affected patients.
Collapse
|
17
|
Nguyen LN, Zhao J, Cao D, Dang X, Wang L, Lian J, Zhang Y, Jia Z, Wu XY, Morrison Z, Xie Q, Ji Y, Zhang Z, El Gazzar M, Ning S, Moorman JP, Yao ZQ. Inhibition of TRF2 accelerates telomere attrition and DNA damage in naïve CD4 T cells during HCV infection. Cell Death Dis 2018; 9:900. [PMID: 30185784 PMCID: PMC6125360 DOI: 10.1038/s41419-018-0897-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/02/2018] [Accepted: 07/09/2018] [Indexed: 12/14/2022]
Abstract
T cells play a crucial role in viral clearance and vaccine responses; however, the mechanisms that regulate their homeostasis during viral infections remain unclear. In this study, we investigated the machineries of T-cell homeostasis and telomeric DNA damage using a human model of hepatitis C virus (HCV) infection. We found that naïve CD4 T cells in chronically HCV-infected patients (HCV T cells) were significantly reduced due to apoptosis compared with age-matched healthy subjects (HSs). These HCV T cells were not only senescent, as demonstrated by overexpression of aging markers and particularly shortened telomeres; but also DNA damaged, as evidenced by increased dysfunctional telomere-induced foci (TIF). Mechanistically, the telomere shelterin protein, in particular telomeric repeat binding factor 2 (TRF2) that functions to protect telomeres from DNA damage, was significantly inhibited posttranscriptionally via the p53-dependent Siah-1a ubiquitination. Importantly, knockdown of TRF2 in healthy T cells resulted in increases in telomeric DNA damage and T-cell apoptosis, whereas overexpression of TRF2 in HCV T cells alleviated telomeric DNA damage and T-cell apoptosis. To the best of our knowledge, this is the first report revealing that inhibition of TRF2 promotes T-cell telomere attrition and telomeric DNA damage that accelerates T-cell senescent and apoptotic programs, which contribute to naïve T-cell loss during viral infection. Thus, restoring the impaired T-cell telomeric shelterin machinery may offer a new strategy to improve immunotherapy and vaccine response against human viral diseases.
Collapse
Affiliation(s)
- Lam Nhat Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Dechao Cao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Xindi Dang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Ling Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Jianqi Lian
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhansheng Jia
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xiao Y Wu
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Zheng Morrison
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Qian Xie
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Yingjie Ji
- Research Center for Clinical and Translational Medicine, Beijing 302 Hospital, Beijing, China
| | - Zheng Zhang
- Research Center for Clinical and Translational Medicine, Beijing 302 Hospital, Beijing, China
| | - Mohamed El Gazzar
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Shunbin Ning
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
| | - Jonathan P Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA
- Department of Veterans Affairs, Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Johnson City, TN, 37614, USA
| | - Zhi Q Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
- Division of Infectious, Inflammatory and Immunologic Diseases, Department of Internal Medicine, Quillen College of Medicine, ETSU, Johnson City, TN, 37614, USA.
- Department of Veterans Affairs, Hepatitis (HCV/HBV/HIV) Program, James H. Quillen VA Medical Center, Johnson City, TN, 37614, USA.
| |
Collapse
|
18
|
Brill L, Lavon I, Vaknin-Dembinsky A. Reduced expression of the IL7Ra signaling pathway in Neuromyelitis optica. J Neuroimmunol 2018; 324:81-89. [PMID: 30248528 DOI: 10.1016/j.jneuroim.2018.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/19/2018] [Accepted: 08/19/2018] [Indexed: 12/13/2022]
Abstract
Neuromyelitis optica (NMO) is a chronic inflammatory demyelinating autoimmune disease of the central nervous system that most commonly affects the optic nerves and spinal cord. To characterize the immunological pathways involved in NMO, whole blood RNA expression array was performed using Nanostring nCounter technology. Two major clusters of genes were found associated with NMO: T cell-associated genes and the TNF/NF-kB signaling pathway. Analysis of the genes within the first cluster confirmed significantly reduced expression of IL7Ra (CD127) in the peripheral blood of NMO patients vs that in healthy controls. IL7Ra upstream transcription factors and its downstream survival signaling pathway were also markedly reduced. In line with the essential role of IL7Ra in T cell maturation and survival, a significantly lower number of naïve T cells, and reduced T cell survival signaling mediated by increased BID (BH3-interacting domain death agonist) expression and increased apoptosis was observed. Cumulatively, these findings indicate that the IL7Ra signaling pathway may play a role in the autoimmune process in NMO.
Collapse
Affiliation(s)
- Livnat Brill
- Department of Neurology, the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Israel
| | - Iris Lavon
- Department of Neurology, the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Israel; Leslie and Michael Center for Neuro-oncology, Hadassah-Medical Center, Israel
| | - Adi Vaknin-Dembinsky
- Department of Neurology, the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Hebrew University of Jerusalem, The Faculty of Medicine, Israel.
| |
Collapse
|
19
|
Zhao J, Dang X, Zhang P, Nguyen LN, Cao D, Wang L, Wu X, Morrison ZD, Zhang Y, Jia Z, Xie Q, Wang L, Ning S, EL Gazzar M, Moorman JP, Yao ZQ. Insufficiency of DNA repair enzyme ATM promotes naive CD4 T-cell loss in chronic hepatitis C virus infection. Cell Discov 2018; 4:16. [PMID: 29644094 PMCID: PMC5891503 DOI: 10.1038/s41421-018-0015-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/21/2017] [Indexed: 12/23/2022] Open
Abstract
T cells have a crucial role in viral clearance and vaccine response; however, the mechanisms regulating their responses to viral infections or vaccinations remain elusive. In this study, we investigated T-cell homeostasis, apoptosis, DNA damage, and repair machineries in a large cohort of subjects with hepatitis C virus (HCV) infection. We found that naive CD4 T cells in chronically HCV-infected individuals (HCV T cells) were significantly reduced compared with age-matched healthy subjects. In addition, HCV T cells were prone to apoptosis and DNA damage, as evidenced by increased 8-oxoguanine expression and γH2AX/53BP1-formed DNA damage foci-hallmarks of DNA damage responses. Mechanistically, the activation of DNA repair enzyme ataxia telangiectasia mutated (ATM) was dampened in HCV T cells. ATM activation was also diminished in healthy T cells exposed to ATM inhibitor or to HCV (core protein) that inhibits the phosphoinositide 3 kinase pathway, mimicking the biological effects in HCV T cells. Importantly, ectopic expression of ATM was sufficient to repair the DNA damage, survival deficit, and cell dysfunctions in HCV T cells. Our results demonstrate that insufficient DNA repair enzyme ATM leads to increased DNA damage and renders HCV T cells prone to apoptotic death, which contribute to the loss of naive T cells in HCV infection. Our study reveals a novel mechanism for T-cell dysregulation and viral persistence, providing a new strategy to improve immunotherapy and vaccine responses against human viral diseases.
Collapse
Affiliation(s)
- Juan Zhao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Xindi Dang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Peixin Zhang
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi’an 710038, China
| | - Lam Nhat Nguyen
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Dechao Cao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Lin Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Xiaoyuan Wu
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Zheng D Morrison
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi’an 710038, China
| | - Zhansheng Jia
- Department of Infectious Diseases, Tangdu Hospital, the Fourth Military Medical University, Xi’an 710038, China
| | - Qian Xie
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Biomedical Science, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
| | - Ling Wang
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Shunbin Ning
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Mohamed EL Gazzar
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
| | - Jonathan P Moorman
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
- Department of Veterans Affairs, Hepatitis (HCV/HIV) Program, James H Quillen VA Medical Center, Johnson City, TN 37614 USA
| | - Zhi Q Yao
- Center of Excellence in Inflammation, Infectious Disease and Immunity, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614 USA
- Department of Internal Medicine, Division of Infectious, Inflammatory and Immunologic Diseases, Quillen College of Medicine, ETSU, Johnson City, TN 37614 USA
- Department of Veterans Affairs, Hepatitis (HCV/HIV) Program, James H Quillen VA Medical Center, Johnson City, TN 37614 USA
| |
Collapse
|
20
|
Pre-vaccine plasma levels of soluble inflammatory indices negatively predict responses to HAV, HBV, and tetanus vaccines in HCV and HIV infection. Vaccine 2017; 36:453-460. [PMID: 29254840 DOI: 10.1016/j.vaccine.2017.12.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/29/2017] [Accepted: 12/07/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic hepatitis C virus (HCV) and HIV infections are associated with impaired responses to neo-antigens contained in hepatitis A virus (HAV)/hepatitis B virus (HBV) vaccines, yet responsible mechanisms are unclear. METHODS ACTG 5232 and CFAR0910 were clinical trials where pre-vaccine levels of plasma IP10, IL-6, sCD163 and sCD14 were measured in viremic HCV- (n = 15) or HIV-infected participants (n = 24) and uninfected controls (n = 10). Accelerated dosing HAV/HBV vaccine and tetanus booster were administered and antibody response was measured at 0, 1, 3, 8, and 24 weeks. RESULTS Pre-vaccine plasma IP10, IL-6, and sCD14 levels were elevated in both HCV and HIV-infected participants, while sCD163 was also elevated in HCV-infected participants. Pre-immunization tetanus antibody levels were lower in HIV-infected than in uninfected participants, while vaccine induced antibody responses were intact in HCV and HIV-infected participants. After HAV/HBV vaccination, HCV and HIV-infected participants had lower and less durable HAV and HBV antibody responses than uninfected controls. Among HCV-infected participants, pre-vaccine plasma IP10, IL-6, sCD14, and sCD163 levels inversely correlated with HAV, HBV and tetanus antibody responses after vaccine. Low HAV/HBV vaccine responses in HIV-infected participants prohibited assessment of immune correlates. CONCLUSIONS During HCV and HIV infection markers of systemic inflammation reflect immune dysfunction as demonstrated by poor response to HAV/HBV neo-antigen vaccine.
Collapse
|
21
|
Abstract
Alcohol-related liver disease (ALD) remains the most important cause of death due to alcohol. Infections, particularly bacterial infections, are one of the most frequent and severe complications of advanced ALDs, such as alcoholic cirrhosis and severe alcoholic hepatitis (sAH). The specific mechanisms responsible for this altered host defence are yet to be deciphered. The aim of the present study is to review the current knowledge of infectious complications in ALD and its pathophysiological mechanisms, distinguishing the role of alcohol consumption and the contribution of different forms of ALD. To date, corticosteroids are the only treatment with proven efficacy in sAH, but their impact on the occurrence of infections remains controversial. The combination of an altered host defence and corticosteroid treatment in sAH has been suggested as a cause of opportunistic fungal and viral infections. A high level of suspicion with systematic screening and prompt, adequate treatment are warranted to improve outcomes in these patients. Prophylactic or preemptive strategies in this high-risk population might be a preferable option, because of the high short-term mortality rate despite adequate therapies. However, these strategies should be assessed in well-designed trials before clinical implementation.
Collapse
|
22
|
Di Profio B, Villar CC, Saraiva L, Ortega KL, Pannuti CM. Is periodontitis a risk factor for infections in cirrhotic patients? Med Hypotheses 2017; 106:19-22. [PMID: 28818265 DOI: 10.1016/j.mehy.2017.06.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/09/2017] [Accepted: 06/26/2017] [Indexed: 02/08/2023]
Affiliation(s)
- B Di Profio
- Department of Estomatology, School of Dentistry, University of São Paulo, Brazil
| | - C C Villar
- Department of Estomatology, School of Dentistry, University of São Paulo, Brazil
| | - L Saraiva
- Department of Estomatology, School of Dentistry, University of São Paulo, Brazil
| | - K L Ortega
- Department of Estomatology, School of Dentistry, University of São Paulo, Brazil
| | - C M Pannuti
- Department of Estomatology, School of Dentistry, University of São Paulo, Brazil.
| |
Collapse
|
23
|
Brief Report: CD14brightCD16- monocytes and sCD14 level negatively associate with CD4-memory T-cell frequency and predict HCV-decline on therapy. J Acquir Immune Defic Syndr 2017; 73:258-262. [PMID: 27258231 DOI: 10.1097/qai.0000000000001104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During HIV+ hepatitis C virus (HCV)+ coinfection CD14CD16 monocytes produce soluble immune-activation markers that predict disease progression and poor response to interferon (IFN)-α treatment. We evaluated relationships among immune activation, monocyte phenotype, CD4-memory T cells, and HCV-, cytomegalovirus-, and cytomegalovirus/Epstein-Barr virus/influenza-specific IFN-γ-response before and during IFN-α treatment. Effector-memory and central-memory CD4 T-cell frequencies were lower in HCV+ HIV+ donors than in uninfected donors and correlated negatively with HCV level, CD14CD16 monocytes, and plasma sCD14. sCD14 and CD14CD16 monocytes negatively correlated with IFN-α-dependent HCV decline. CD4 effector-memory T cells positively associated with cytomegalovirus/Epstein-Barr virus/influenza(CEF)-specific IFN-γ response, while sCD14 negatively associated with both CD4 effector-memory T cells and CEF-specific IFN-γ response. These data support a role for memory-CD4 T cells in HCV containment and link immune activation and CD14CD16-monocyte frequency to the failure of IFN-dependent HCV clearance.
Collapse
|
24
|
Vogelaar L, de Haar C, Aerts BR, Peppelenbosch MP, Timman R, Hanssen BE, van der Woude CJ. Fatigue in patients with inflammatory bowel disease is associated with distinct differences in immune parameters. Clin Exp Gastroenterol 2017; 10:83-90. [PMID: 28496351 PMCID: PMC5422327 DOI: 10.2147/ceg.s123942] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Although it is well recognized that fatigue is an important problem in many of the quiescent inflammatory bowel disease (IBD) patients, it is unknown whether the immune status is different in fatigued versus non-fatigued patients. In this study, we contrasted various characteristics of the immune system in fatigued against non-fatigued patients with IBD in clinical remission. Patients and methods Patients with IBD in clinical remission were phenotyped according to the Montreal classification, and the checklist individual strength-fatigue (CIS-fatigue) was used to assess fatigue (CIS-fatigue ≥ 35). Flow cytometry on peripheral blood samples was used to investigate differences in leukocyte subsets. The expression of various cytokines was determined in stimulated whole blood and serum samples using enzyme-linked immunosorbent assay. Differences between fatigued and non-fatigued patients with IBD were assessed. Results In total, 55 patients were included in the fatigue group (FG) and 29 patients in the non-fatigue group (NFG). No differences in demographic and clinical characteristics were observed between the groups. Flow cytometry data showed a significantly lower percentage of monocytes (p = 0.011) and a higher percentage of memory T-cells (p = 0.005) and neutrophils (p = 0.033) in the FG compared with the NFG. Whole blood stimulation showed increased TNF-α (p = 0.022) and IFN-γ (p = 0.047) in the FG. The median serum level was significantly higher for IL-12 (p < 0.001) and IL-10 (p = 0.005) and lower for IL-6 (p = 0.002) in the FG compared with NFG. Conclusion Significant differences in immune profile between fatigued and non-fatigued patients with IBD in clinical remission were found, which point out to a chronically active and Th1-skewed immune system in patients with fatigue. Whether these immune differences are directly involved in the fatigue complaints via immune-to-brain communication pathways remains to be determined. As such, further exploration of the underlying immune effects associated with fatigue is warranted to determine potential treatment options.
Collapse
Affiliation(s)
- Lauran Vogelaar
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam
| | - Colin de Haar
- Applied Tumor Immunology, Laboratory of Translational Immunology, UMC Utrecht, Utrecht
| | - Bas Rj Aerts
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam
| | | | - Reinier Timman
- Department of Psychiatry, Section of Medical Psychology and Psychotherapy, Erasmus MC, Rotterdam, the Netherlands
| | - Bettina E Hanssen
- Department of Gastroenterology and Hepatology, Erasmus MC, Rotterdam
| | | |
Collapse
|
25
|
Tanaskovic S, Price P, French MA, Fernandez S. Impaired Upregulation of the Costimulatory Molecules, CD27 and CD28, on CD4 + T Cells from HIV Patients Receiving ART Is Associated with Poor Proliferative Responses. AIDS Res Hum Retroviruses 2017; 33:101-109. [PMID: 27701900 DOI: 10.1089/aid.2015.0327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
HIV patients beginning antiretroviral therapy (ART) with advanced immunodeficiency often retain low CD4+ T cell counts despite virological control. We examined proliferative responses and upregulation of costimulatory molecules, following anti-CD3 stimulation, in HIV patients with persistent CD4+ T cell deficiency on ART. Aviremic HIV patients with nadir CD4+ T cell counts <100 cells/μL and who had received ART for a median time of 7 (range 1-11) years were categorized into those achieving low (<350 cells/μL; n = 13) or normal (>500 cells/μL; n = 20) CD4+ T cell counts. Ten healthy controls were also recruited. CD4+ T cell proliferation (Ki67) and upregulation of costimulatory molecules (CD27 and CD28) after anti-CD3 stimulation were assessed by flow cytometry. Results were related to proportions of CD4+ T cells expressing markers of T cell senescence (CD57), activation (HLA-DR), and apoptotic potential (Fas). Expression of CD27 and/or CD28 on uncultured CD4+ T cells was similar in patients with normal CD4+ T cell counts and healthy controls, but lower in patients with low CD4+ T cell counts. Proportions of CD4+ T cells expressing CD27 and/or CD28 correlated inversely with CD4+ T cell expression of CD57, HLA-DR, and Fas. After anti-CD3 stimulation, induction of CD27hiCD28hi expression was independent of CD4+ T cell counts, but lower in HIV patients than in healthy controls. Induction of CD27hiCD28hi expression correlated with induction of Ki67 expression in total, naïve, and CD31+ naïve CD4+ T cells from patients. In HIV patients responding to ART, impaired induction of CD27 and CD28 on CD4+ T cells after stimulation with anti-CD3 is associated with poor proliferative responses as well as greater CD4+ T cell activation and immunosenescence.
Collapse
Affiliation(s)
- Sara Tanaskovic
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Patricia Price
- School of Biomedical Science, Curtin University of Technology, Perth, Australia
| | - Martyn A. French
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
- Department of Clinical Immunology, Royal Perth Hospital and PathWest Laboratory Medicine, Perth, Australia
| | - Sonia Fernandez
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| |
Collapse
|
26
|
Kugler DG, Flomerfelt FA, Costa DL, Laky K, Kamenyeva O, Mittelstadt PR, Gress RE, Rosshart SP, Rehermann B, Ashwell JD, Sher A, Jankovic D. Systemic toxoplasma infection triggers a long-term defect in the generation and function of naive T lymphocytes. J Exp Med 2016; 213:3041-3056. [PMID: 27849554 PMCID: PMC5154934 DOI: 10.1084/jem.20151636] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 08/05/2016] [Accepted: 10/18/2016] [Indexed: 12/12/2022] Open
Abstract
Kugler et al. show that systemic infection with Toxoplasma gondii triggers a long-term impairment in thymic function, which leads to an immunodeficient state reflected in decreased antimicrobial resistance. Because antigen-stimulated naive T cells either die as effectors or enter the activated/memory pool, continuous egress of new T lymphocytes from thymus is essential for maintenance of peripheral immune homeostasis. Unexpectedly, we found that systemic infection with the protozoan Toxoplasma gondii triggers not only a transient increase in activated CD4+ Th1 cells but also a persistent decrease in the size of the naive CD4+ T lymphocyte pool. This immune defect is associated with decreased thymic output and parasite-induced destruction of the thymic epithelium, as well as disruption of the overall architecture of that primary lymphoid organ. Importantly, the resulting quantitative and qualitative deficiency in naive CD4+ T cells leads to an immunocompromised state that both promotes chronic toxoplasma infection and leads to decreased resistance to challenge with an unrelated pathogen. These findings reveal that systemic infectious agents, such as T. gondii, can induce long-term immune alterations associated with impaired thymic function. When accumulated during the lifetime of the host, such events, even when occurring at low magnitude, could be a contributing factor in immunological senescence.
Collapse
Affiliation(s)
- David G Kugler
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Francis A Flomerfelt
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Diego L Costa
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Karen Laky
- T Cell Development Section, Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Olena Kamenyeva
- Biological Imaging, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Paul R Mittelstadt
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ronald E Gress
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Stephan P Rosshart
- Immunology Section, Liver Diseases Branch, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute for Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jonathan D Ashwell
- Laboratory of Immune Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Alan Sher
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Dragana Jankovic
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
27
|
Desai SN, Dodge JL, Landay AL, Glesby MJ, Latham PS, Villacres MC, French AL, Gange SJ, Greenblatt RM, Peters MG. Hepatic fibrosis and immune phenotype vary by HCV viremia in HCV/HIV co-infected subjects: A Women's interagency HIV study. Medicine (Baltimore) 2016; 95:e4483. [PMID: 27537569 PMCID: PMC5370796 DOI: 10.1097/md.0000000000004483] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
HCV and HIV independently lead to immune dysregulation. The mechanisms leading to advanced liver disease progression in HCV/HIV coinfected subjects remain unclear.In this cross-sectional study, we assessed the association of HCV viremia, liver fibrosis, and immune response patterns in well-characterized HIV phenotypes: Elite controllers (Elites), HIV controlled (ARTc), and HIV uncontrolled (ARTuc) matched by age and race. Groups were stratified by HCV RNA status. Regulatory T-cell frequencies, T-cell activation (HLADR+CD38+), apoptosis (Caspase-3+), and intracellular cytokines (interferon-γ, IL-2, IL-17) were assessed using multiparametric flow-cytometry. Liver fibrosis was scored by AST to platelet ratio index (APRI).We found liver fibrosis (APRI) was 50% lower in Elites and ARTc compared to ARTuc. Higher liver fibrosis was associated with significantly low CD4+ T cell counts (P < 0.001, coefficient r = -0.463). Immune activation varied by HIV phenotype but was not modified by HCV viremia. HCV viremia was associated with elevated CD8 T-cell Caspase-3 in Elites, ARTuc, and HIV- except ARTc. CD8 T-cell Caspase-3 levels were significantly higher in HCV RNA+ Elites (P = 0.04) and ARTuc (P = 0.001) and HIV- groups (P = 0.02) than ARTc. Importantly, ARTuc HCV RNA+ had significantly higher CD4 T-cell interleukin-17 levels than ARTuc HCV RNA- (P = 0.005).HIV control was associated with lower liver fibrosis in HCV/HIV co-infected women. HCV viremia is associated with an inflammatory CD4 TH-17 phenotype in absence of HIV control and higher frequency of pro-apoptosis CD8 T-cells critical to avert progression of HIV and HCV disease that is attenuated in ART controllers. Elite controllers with HCV viremia are more prone to CD8 T-cell apoptosis than ART controllers, which could have negative consequences over time, highlighting the importance of ART control in HCV/HIV coinfected individuals.
Collapse
Affiliation(s)
- Seema N. Desai
- Rush University Medical Center, Chicago, IL
- Correspondence: Seema N. Desai, PhD, Assistant Professor, Department of Immunology/Microbiology, Rush University Medical Center, 1735 W. Harrison Street, Chicago, IL 60612 (e-mail: )
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Shmagel KV, Saidakova EV, Shmagel NG, Korolevskaya LB, Chereshnev VA, Robinson J, Grivel JC, Douek DC, Margolis L, Anthony DD, Lederman MM. Systemic inflammation and liver damage in HIV/hepatitis C virus coinfection. HIV Med 2016; 17:581-9. [PMID: 27187749 DOI: 10.1111/hiv.12357] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2015] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Chronic hepatitis C virus (HCV) and HIV viral infections are characterized by systemic inflammation. Yet the relative levels, drivers and correlates of inflammation in these settings are not well defined. METHODS Seventy-nine HIV-infected patients who had been receiving antiretroviral therapy (ART) for more than 2 years and who had suppressed plasma HIV levels (< 50 HIV-1 RNA copies/mL) were included in the study. Two patient groups, HCV-positive/HIV-positive and HCV-negative/HIV-positive, and a control group comprised of healthy volunteers (n = 20) were examined. Markers of systemic inflammation [interleukin (IL)-6, interferon gamma-induced protein (IP)-10, soluble tumour necrosis factor receptor-I (sTNF-RI) and sTNF-RII], monocyte/macrophage activation [soluble CD163 (sCD163), soluble CD14 and neopterin], intestinal epithelial barrier loss [intestinal fatty acid binding protein (I-FABP) and lipopolysaccharide (LPS)] and coagulation (d-dimers) were analysed. CD4 naïve T cells and CD4 recent thymic emigrants (RTEs) were enumerated. RESULTS Plasma levels of IP-10, neopterin and sCD163 were higher in HCV/HIV coinfection than in HIV monoinfection and were positively correlated with indices of hepatic damage [aspartate aminotransferase (AST), alanine aminotransferase (ALT) and the AST to platelet ratio index (APRI)]. Levels of I-FABP were comparably increased in HIV monoinfection and HIV/HCV coinfection but LPS concentrations were highest in HCV/HIV coinfection, suggesting impaired hepatic clearance of LPS. Plasma HCV levels were not related to any inflammatory indices except sCD163. In coinfected subjects, a previously recognized relationship of CD4 naïve T-cell and RTE counts to hepatocellular injury was defined more mechanistically by an inverse relationship to sCD163. CONCLUSIONS Hepatocellular injury in HCV/HIV coinfection is linked to elevated levels of certain inflammatory cytokines and an apparent failure to clear systemically translocated microbial products. A related decrease in CD4 naïve T cells and RTEs also merits further exploration.
Collapse
Affiliation(s)
- K V Shmagel
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm, Russia.,Perm State University, Perm, Russia
| | - E V Saidakova
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm, Russia.,Perm State University, Perm, Russia
| | - N G Shmagel
- Perm State University, Perm, Russia.,Perm Regional Center for Protection against AIDS and Infectious Diseases, Perm, Russia
| | - L B Korolevskaya
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm, Russia.,Perm State University, Perm, Russia
| | - V A Chereshnev
- Institute of Ecology and Genetics of Microorganisms UB RAS, Perm, Russia.,Perm State University, Perm, Russia.,Institute of Immunology and Physiology UB RAS, Yekaterinburg, Russia
| | - J Robinson
- Case Western Reserve University, Cleveland, OH, USA
| | - J-C Grivel
- National Institute of Child Health and Development, Bethesda, MD, USA
| | - D C Douek
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | - L Margolis
- National Institute of Child Health and Development, Bethesda, MD, USA
| | - D D Anthony
- Case Western Reserve University, Cleveland, OH, USA
| | - M M Lederman
- Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
29
|
Grady BPX, Nanlohy NM, van Baarle D. HCV monoinfection and HIV/HCV coinfection enhance T-cell immune senescence in injecting drug users early during infection. IMMUNITY & AGEING 2016; 13:10. [PMID: 27034702 PMCID: PMC4815107 DOI: 10.1186/s12979-016-0065-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/22/2016] [Indexed: 01/09/2023]
Abstract
Background Injecting drug users (IDU) are at premature risk of developing multimorbidity and mortality from causes commonly observed in the elderly. Ageing of the immune system (immune-senescence) can lead to premature morbidity and mortality and can be accelerated by chronic viral infections. Here we investigated the impact of HCV monoinfection and HIV/HCV coinfection on immune parameters in (ex-) IDU. We analyzed telomere length and expression of activation, differentiation and exhaustion markers on T cells at baseline (t = 1) and at follow-up (t = 2) (median interval 16.9 years) in IDU who were: HCV mono-infected (n = 21); HIV/HCV coinfected (n = 23) or multiple exposed but uninfected (MEU) (n = 8). Results The median time interval between t = 1 and t = 2 was 16.9 years. Telomere length within CD4+ and CD8+ T cells decreased significantly over time in all IDU groups (p ≤ 0.012). CD4+ T-cell telomere length in HCV mono-infected IDU was significantly reduced compared to healthy donors at t = 1 (p < 0.008). HIV/HCV coinfected IDU had reduced CD4+ and CD8+ T-cell telomere lengths (p ≤ 0.002) to healthy donors i at t = 1. This was related to persistent levels of immune activation but not due to increased differentiation of T cells over time. Telomere length decrease was observed within all T-cell subsets, but mainly found in immature T cells (CD27+CD57+) (p ≤ 0.015). Conclusions HCV mono-infection and HIV/HCV coinfection enhance T-cell immune-senescence. Our data suggest that this occurred early during infection, which warrants early treatment for both HCV and HIV to reduce immune senescence in later life. Electronic supplementary material The online version of this article (doi:10.1186/s12979-016-0065-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bart P X Grady
- Department of Research, Cluster Infectious Diseases, Public Health Service, Amsterdam, The Netherlands ; Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center, Amsterdam, The Netherlands
| | - Nening M Nanlohy
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Debbie van Baarle
- Department of Immunology, University Medical Center Utrecht, Utrecht, The Netherlands ; Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands ; Present address: Department of Immune Mechanisms, Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| |
Collapse
|
30
|
Burchill MA, Golden-Mason L, Wind-Rotolo M, Rosen HR. Memory re-differentiation and reduced lymphocyte activation in chronic HCV-infected patients receiving direct-acting antivirals. J Viral Hepat 2015; 22:983-91. [PMID: 26482547 DOI: 10.1111/jvh.12465] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 08/12/2015] [Indexed: 12/14/2022]
Abstract
Recently, the treatment of HCV has advanced significantly due to the introduction of direct-acting antivirals (DAAs). Studies using interferon (IFN)-containing regimens failed to consistently show restoration of immunologic responses. Therefore, IFN-free DAA formulations provide a unique opportunity to dissect the immunologic effect of HCV cure. This study investigates the restoration of the immune compartment as a consequence of rapid viral clearance in patients successfully treated with DAAs and in the absence of IFN and ribavirin. Here, we evaluate the immunologic changes that occurred following DAA-mediated HCV cure. Peripheral blood from nineteen previously treatment-naïve patients with chronic HCV genotype 1a/1b who received an IFN and ribavirin-free regimen of daclatasvir, asunaprevir and BMS-791325 was evaluated. Immune reconstitution occurs in patients in whom HCV was successfully eradicated via DAA therapy. Restoration of the CD4(+) T-cell compartment in the peripheral blood and a re-differentiation of the T lymphocyte memory compartment resulted in a more effector memory cell population and a reduction in expression in the co-inhibitory molecule TIGIT in bulk T lymphocytes. Furthermore, we observed a partial reversal of the exhausted phenotype in HCV-specific CD8(+) T cells and a dampening of the activation state in peripheral NK cells. Collectively, our data provide the groundwork for dissecting the effect of DAA therapy on the immune system and identifying novel mechanisms by which chronic HCV infection exerts immunosuppressive effects on T cells through the recently described co-inhibitory molecule TIGIT.
Collapse
Affiliation(s)
- M A Burchill
- Department of Medicine, Division of GI/Heaptology, University of Colorado-Denver, Aurora, CO, USA
| | - L Golden-Mason
- Department of Medicine, Division of GI/Heaptology, University of Colorado-Denver, Aurora, CO, USA
| | - M Wind-Rotolo
- Bristol-Myers Squibb, Exploratory Clinical and Translational Research, Lawrenceville, NJ, USA
| | - H R Rosen
- Department of Medicine, Division of GI/Heaptology, University of Colorado-Denver, Aurora, CO, USA
| |
Collapse
|
31
|
Changes in HIV RNA and CD4 cell count after acute HCV infection in chronically HIV-infected individuals. J Acquir Immune Defic Syndr 2015; 68:536-42. [PMID: 25559595 DOI: 10.1097/qai.0000000000000514] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVE Little is known about the impact of acute hepatitis C virus (HCV) co-infection on HIV-1 disease progression. We investigated CD4 cell count and HIV RNA concentration changes after HCV infection in individuals chronically infected with HIV-1. METHODS We selected individuals that had the last negative and first positive HCV RNA test less than 1 year apart. Bivariate linear mixed-effects regression was used to model trends in HIV RNA level and CD4 cell count from 2 years before the last negative HCV RNA test until the first of the following dates: start of anti-HCV medication, change in combination antiretroviral therapy (cART) status, and end of follow-up. RESULTS At the estimated time of HCV co-infection, of 89 individuals, 63 (71%) were cART-treated and 26 (29%) were not on cART. In persons on cART, median CD4 cell count declined from 587 to 508 cells per cubic millimeter (P < 0.0001) during the first 5 months after HCV infection and returned to 587 cells per cubic millimeter after 2.2 years. Also, the probability of an HIV RNA >50 copies per milliliter peaked to 18.6% at HCV co-infection, with lower probabilities 6 months before (3.5%, P = 0.006 compared with peak probability) and after (2.9%, P = 0.009). In persons not on cART, no significant impact of HCV co-infection on trends in the HIV RNA level or CD4 cell count was observed. CONCLUSIONS Acute HCV infection in cART-treated, chronically HIV-infected patients was associated with a temporary decrease in CD4 cell counts and increased risk of HIV viremia >50 copies per milliliter. This may increase the risk of further HIV transmission.
Collapse
|
32
|
Jain MK, Adams-Huet B, Terekhova D, Kushner LE, Bedimo R, Li X, Holodniy M. Acute and chronic immune biomarker changes during interferon/ribavirin treatment in HIV/HCV co-infected patients. J Viral Hepat 2015; 22:25-36. [PMID: 24506344 PMCID: PMC4127161 DOI: 10.1111/jvh.12226] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 12/04/2013] [Indexed: 12/14/2022]
Abstract
Chronic viral infections lead to persistent immune activation, which is alleviated by eradicating or suppressing the infection. To understand the effects of interferon treatment on immune system activation by chronic infections, we evaluated kinetic patterns of a broad spectrum of serum biomarkers during HCV treatment in HIV/HCV co-infected patients. HCV viral load and 50 biomarkers were analysed at baseline and 27 time points during pegylated interferon-alpha and ribavirin (IFN/RBV) treatment of 12 HIV/HCV co-infected patients. We evaluated biomarker changes from baseline for each time point and biomarker correlations with clinical parameters, treatment response and liver histopathology. IL-1α, IL-12p40, IL-1RA, IP-10, MIG, MIP-1α/1β, HGF, sCD40L, TRAIL and leptin increased in the first day. IL-12p70, IL-17A, IL-10, GROα, IL-8, MCP-3, IL-4 and M-CSF peaked later during week 1. IL-1α, HGF, IP-10, MIP-1α, TRAIL, sCD40L, IL-10, IL-12p70, MCP-3, FGFb, ENA-78, TGF-β, IL-2, IFN-γ, IL-6, IL-15, IL-7 and PDGF-BB decreased below baseline over the course of treatment. Higher BMI, baseline HCV viral load and leptin levels were associated with lack of sustained virologic response. ENA-78 was associated with sustained viral response. Positive correlations were found between liver inflammation and baseline CD4 count, sVCAM and HGF; fibrosis stage and HGF; liver steatosis, BMI and leptin. Our findings suggest IFN/RBV treatment initially increases levels of several biomarkers, but eventually leads to a decline in many immune markers. These findings shed light on the relationship between IFN treatment and immune activation by chronic viral infections, such as HCV.
Collapse
Affiliation(s)
- Mamta K. Jain
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Beverley Adams-Huet
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX
| | - Darya Terekhova
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Lauren E. Kushner
- AIDS Research Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | - Roger Bedimo
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
,Department of Internal Medicine, North Texas VA Medical Center, Dallas, TX
| | - Xilong Li
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, TX
| | - Mark Holodniy
- AIDS Research Center, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
,Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
33
|
Association of chronic hepatitis C infection with T-cell phenotypes in HIV-negative and HIV-positive women. J Acquir Immune Defic Syndr 2014; 67:295-303. [PMID: 25314250 DOI: 10.1097/qai.0000000000000310] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Hepatitis C virus (HCV) viremia is thought to have broad systemic effects on the cellular immune system that go beyond its impact on just those T cells that are HCV specific. However, previous studies of chronic HCV and circulating T-cell subsets (activation and differentiation phenotypes) in HIV negatives used general population controls, rather than a risk-appropriate comparison group. Studies in HIV positives did not address overall immune status (total CD4⁺ count). METHODS We used fresh blood from HIV-positive and at-risk HIV-negative women, with and without chronic HCV, to measure percentages of activated CD4⁺ and CD8⁺ T cells, Tregs, and T-cell differentiation phenotypes (naive, central memory, effector memory (EM), and terminally differentiated effector). This included 158 HIV negatives and 464 HIV positives, of whom 18 and 63, respectively, were HCV viremic. RESULTS In multivariate models of HIV negatives, HCV viremia was associated with 25% fewer naive CD4⁺ (P = 0.03), 33% more EM CD4⁺ (P = 0.0002), and 37% fewer central memory CD8⁺ (P = 0.02) T cells. Among HIV positives, we observed only 1 of these 3 relationships: higher percentage of EM CD4⁺ among HCV viremic women. Furthermore, the association with EM CD4⁺ among HIV positives was limited to individuals with diminished immune status (total CD4⁺ count ≤500 cells/μL), as were associations of HCV viremia with higher percentages of activated CD4⁺ and Tregs. Among HIV positives with high CD4⁺ count, no significant associations were observed. CONCLUSIONS These data suggest that HCV viremia in HIV negatives is associated with accelerated T-cell differentiation, but among HIV positives, the impact of HCV viremia is less straightforward and varies by total CD4v count.
Collapse
|
34
|
Lambracht-Washington D, Rosenberg RN. Co-stimulation with TNF receptor superfamily 4/25 antibodies enhances in-vivo expansion of CD4+CD25+Foxp3+ T cells (Tregs) in a mouse study for active DNA Aβ42 immunotherapy. J Neuroimmunol 2014; 278:90-9. [PMID: 25595257 DOI: 10.1016/j.jneuroim.2014.12.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/05/2014] [Accepted: 12/08/2014] [Indexed: 01/09/2023]
Abstract
The study was designed to test DNA Aβ42 immunization in mice as alternative approach for possible active immunotherapy in Alzheimer patients. As results, we found polarized Th2 immune responses, efficient Aβ42 antibody levels, and disappearance of antigen specific T cells. In-vivo TNFRSF4/25 antibody co-stimulation enhanced Aβ42 specific T cell responses with initial Th2 expansion and subsequent development of Aβ42 specific CD4+CD25+Foxp3+ cells. It showed that Th2 biased responses due to gene gun immunizations propagate the development of regulatory T cells. In conclusion, full-length DNA Aβ42 immunization into skin results in a regulatory response with minimal risk of inflammation and autoimmunity.
Collapse
Affiliation(s)
- Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, Alzheimer's Disease Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
35
|
Albillos A, Lario M, Álvarez-Mon M. Cirrhosis-associated immune dysfunction: distinctive features and clinical relevance. J Hepatol 2014; 61:1385-96. [PMID: 25135860 DOI: 10.1016/j.jhep.2014.08.010] [Citation(s) in RCA: 825] [Impact Index Per Article: 75.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 07/27/2014] [Accepted: 08/09/2014] [Indexed: 02/06/2023]
Abstract
The term cirrhosis-associated immune dysfunction refers to the main syndromic abnormalities of immune function, immunodeficiency and systemic inflammation that are present in cirrhosis. The course of advanced cirrhosis, regardless of its aetiology, is complicated by cirrhosis-associated immune dysfunction and this constitutes the pathophysiological hallmark of an increased susceptibility to bacterial infection, distinctive of the disease. Cirrhosis impairs the homeostatic role of the liver in the systemic immune response. Damage to the reticulo-endothelial system compromises the immune surveillance function of the organ and the reduced hepatic synthesis of proteins, involved in innate immunity and pattern recognition, hinders the bactericidal ability of phagocytic cells. Systemic inflammation, in form of activated circulating immune cells and increased serum levels of pro-inflammatory cytokines, is the result of persistent episodic activation of circulating immune cells from damage-associated molecular patterns, released from necrotic liver cells and, as cirrhosis progresses, from pathogen-associated molecular patterns, released from the leaky gut. Cirrhosis-associated immune dysfunction phenotypes switch from predominantly "pro-inflammatory" to predominantly "immunodeficient" in patients with stable ascitic cirrhosis and in patients with severely decompensated cirrhosis and extra-hepatic organ failure (e.g. acute-on-chronic liver failure), respectively. These cirrhosis-associated immune dysfunction phenotypes represent the extremes of a spectrum of reversible dynamic events that take place during the course of cirrhosis. Systemic inflammation can affect the functions of tissue somatic cells and modify the clinical manifestation of cirrhosis. The best characterized example is the contribution of systemic inflammation to the haemodynamic derangement of cirrhosis, which correlates negatively with prognosis.
Collapse
Affiliation(s)
- Agustín Albillos
- Department of Medicine, Universidad de Alcalá, Madrid, Spain; CIBERehd, Instituto de Salud Carlos III, Madrid, Spain; Service of Gastroenterology and Hepatology, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain.
| | - Margaret Lario
- Department of Medicine, Universidad de Alcalá, Madrid, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine, Universidad de Alcalá, Madrid, Spain; CIBERehd, Instituto de Salud Carlos III, Madrid, Spain; Service of Immune Diseases and Oncology, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
36
|
Influence of hepatitis C virus coinfection on CD4⁺ T cells of HIV-infected patients receiving HAART. AIDS 2014; 28:2381-8. [PMID: 25111083 DOI: 10.1097/qad.0000000000000418] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
OBJECTIVE The effects of hepatitis C virus (HCV) coinfection on immune homeostasis and immune restoration in treated HIV infection are not well understood. METHODS We studied 79 HIV-infected patients who had been receiving HAART for more than 2 years and who had HIV viral load below 50 copies/ml. Four patient groups were studied: HIV/HCV, CD4⁺ cells above 350/μl; HIV/HCV, CD4 cells below 350/μl; HIV/HCV, CD4 cells above 350/μl; HIV/HCV, CD4⁺ cells below 350/μl. Controls comprised 20 healthy volunteers. Naive, central memory, effector memory, and terminal effector CD4⁺ T cells were enumerated. Naive CD4CD31 T cells were counted as recent thymic emigrants (RTEs). Activation state and ex-vivo apoptosis of CD4⁺ T cells, levels of liver enzymes, and aspartate aminotransferase-to-platelet ratio index were evaluated. RESULTS CD4⁺ T-cell counts and the numbers of all circulating CD4 T-cell maturation subsets were diminished in HIV infection; CD4⁺ T-cell activation and apoptosis were increased in HIV infection, but none of these indices was affected by HCV coinfection. RTE numbers were diminished in HIV infection, were inversely related to age, and were increased in women and lower in HIV/HCV patients than in singly HIV-infected patients. In coinfected patients, RTE numbers were inversely related to levels of liver enzymes, but not to HCV viral load. CONCLUSION Whereas we could find no relationship between HCV infection and most indices of CD4⁺ T-cell homeostasis or activation, CD4⁺ RTEs are diminished in the circulation of HCV coinfected persons and appear to be related to indices of ongoing hepatic damage or inflammation.
Collapse
|
37
|
Emerging clinical issues related to management of multiorgan comorbidities and polypharmacy. Curr Opin HIV AIDS 2014; 9:371-8. [DOI: 10.1097/coh.0000000000000068] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
38
|
Sanchez Rodriguez R, Pauli ML, Neuhaus IM, Yu SS, Arron ST, Harris HW, Yang SHY, Anthony BA, Sverdrup FM, Krow-Lucal E, MacKenzie TC, Johnson DS, Meyer EH, Löhr A, Hsu A, Koo J, Liao W, Gupta R, Debbaneh MG, Butler D, Huynh M, Levin EC, Leon A, Hoffman WY, McGrath MH, Alvarado MD, Ludwig CH, Truong HA, Maurano MM, Gratz IK, Abbas AK, Rosenblum MD. Memory regulatory T cells reside in human skin. J Clin Invest 2014; 124:1027-36. [PMID: 24509084 DOI: 10.1172/jci72932] [Citation(s) in RCA: 269] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Accepted: 11/21/2013] [Indexed: 01/07/2023] Open
Abstract
Regulatory T cells (Tregs), which are characterized by expression of the transcription factor Foxp3, are a dynamic and heterogeneous population of cells that control immune responses and prevent autoimmunity. We recently identified a subset of Tregs in murine skin with properties typical of memory cells and defined this population as memory Tregs (mTregs). Due to the importance of these cells in regulating tissue inflammation in mice, we analyzed this cell population in humans and found that almost all Tregs in normal skin had an activated memory phenotype. Compared with mTregs in peripheral blood, cutaneous mTregs had unique cell surface marker expression and cytokine production. In normal human skin, mTregs preferentially localized to hair follicles and were more abundant in skin with high hair density. Sequence comparison of TCRs from conventional memory T helper cells and mTregs isolated from skin revealed little homology between the two cell populations, suggesting that they recognize different antigens. Under steady-state conditions, mTregs were nonmigratory and relatively unresponsive; however, in inflamed skin from psoriasis patients, mTregs expanded, were highly proliferative, and produced low levels of IL-17. Taken together, these results identify a subset of Tregs that stably resides in human skin and suggest that these cells are qualitatively defective in inflammatory skin disease.
Collapse
|
39
|
Lario M, Muñoz L, Ubeda M, Borrero MJ, Martínez J, Monserrat J, Díaz D, Alvarez-Mon M, Albillos A. Defective thymopoiesis and poor peripheral homeostatic replenishment of T-helper cells cause T-cell lymphopenia in cirrhosis. J Hepatol 2013; 59:723-30. [PMID: 23742913 DOI: 10.1016/j.jhep.2013.05.042] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 05/09/2013] [Accepted: 05/19/2013] [Indexed: 12/18/2022]
Abstract
BACKGROUND & AIMS Depletion of circulating CD4(+) T-helper (Th) lymphocytes, especially naive Th cells, is common in cirrhosis. Little is known about the pathogenetic mechanisms involved in Th-cell depletion in cirrhosis. We investigated the mechanisms involved in circulating Th-cell lymphopenia in cirrhosis. METHODS Circulating naive and memory Th cells were analyzed by flow cytometry in 60 patients with cirrhosis and 40 sex- and age-matched healthy controls. Thymopoiesis, apoptosis, cell activation, and proliferation were assessed through CD31, annexin-V, HLA-DR and Ki-67 expression, respectively. Lipopolysaccharide (LPS)-binding protein (LBP) and spleen size were measured as indicators of bacterial translocation and splenic pooling, respectively. RESULTS Compared to controls, patients showed reduced numbers of Th cells involving a greater depletion of the naive than memory Th-cell compartment (2.7- vs. 1.5-fold, respectively). Recent thymic emigrants were diminished (p < 0.01), and each patient had a lower number of CD31(+) naive Th cells than the matched-control. Spontaneous and induced apoptosis (Annexin-V(+)) of Th cells was increased in patients. Activated (HLA-DR(+)) and proliferating (Ki-67(+)) memory Th cells were increased in patients (p < 0.01), and they directly correlated with plasma LBP (p < 0.05) and negatively with naive Th cells (p < 0.01), respectively. Naive Th cells were inversely correlated (p < 0.01) with their frequencies of apoptosis and of activated memory Th cells, LBP, and spleen size. On multivariate analysis, defective thymic generation of naive Th cells, increased memory Th-cell activation, and splenomegaly were independently associated with Th-cell depletion. CONCLUSIONS Th-cell immunodeficiency in cirrhosis is explained by a universal defect in thymopoiesis exacerbated by splenic pooling and activation-driven cell-death induced by bacterial translocation.
Collapse
Affiliation(s)
- Margaret Lario
- Departamento de Medicina, Universidad de Alcalá, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Miedema F, Hazenberg MD, Tesselaar K, van Baarle D, de Boer RJ, Borghans JAM. Immune activation and collateral damage in AIDS pathogenesis. Front Immunol 2013; 4:298. [PMID: 24133492 PMCID: PMC3783946 DOI: 10.3389/fimmu.2013.00298] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 09/09/2013] [Indexed: 12/17/2022] Open
Abstract
In the past decade, evidence has accumulated that human immunodeficiency virus (HIV)-induced chronic immune activation drives progression to AIDS. Studies among different monkey species have shown that the difference between pathological and non-pathological infection is determined by the response of the immune system to the virus, rather than its cytopathicity. Here we review the current understanding of the various mechanisms driving chronic immune activation in HIV infection, the cell types involved, its effects on HIV-specific immunity, and how persistent inflammation may cause AIDS and the wide spectrum of non-AIDS related pathology. We argue that therapeutic relief of inflammation may be beneficial to delay HIV-disease progression and to reduce non-AIDS related pathological side effects of HIV-induced chronic immune stimulation.
Collapse
Affiliation(s)
- Frank Miedema
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Mette D. Hazenberg
- Department of Internal Medicine and Hematology, Academic Medical Center, Amsterdam, Netherlands
| | - Kiki Tesselaar
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Debbie van Baarle
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Rob J. de Boer
- Theoretical Biology and Bioinformatics, Utrecht University, Utrecht, Netherlands
| | - José A. M. Borghans
- Department of Immunology, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
41
|
Hartling HJ, Gaardbo JC, Ronit A, Salem M, Laye M, Clausen MR, Skogstrand K, Gerstoft J, Ullum H, Nielsen SD. Impaired Thymic Output in Patients with Chronic Hepatitis C Virus Infection. Scand J Immunol 2013; 78:378-86. [PMID: 23841696 DOI: 10.1111/sji.12096] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/04/2013] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | - M. Salem
- Viro-Immunology Research Unit; Department of Infectious Diseases, Rigshospitalet; Copenhagen University Hospital; Copenhagen; Denmark
| | - M. Laye
- Centre of Inflammation and Metabolism, Rigshospitalet; Copenhagen University Hospital; Copenhagen; Denmark
| | - M. R. Clausen
- Department of Hepatology, Rigshospitalet; Copenhagen University Hospital; Copenhagen; Denmark
| | - K. Skogstrand
- Department of Clinical Biochemistry and Immunology; Statens Serum Institute; Copenhagen; Denmark
| | - J. Gerstoft
- Viro-Immunology Research Unit; Department of Infectious Diseases, Rigshospitalet; Copenhagen University Hospital; Copenhagen; Denmark
| | - H. Ullum
- Department of Clinical Immunology, Rigshospitalet; Copenhagen University Hospital; Copenhagen; Denmark
| | - S. D. Nielsen
- Viro-Immunology Research Unit; Department of Infectious Diseases, Rigshospitalet; Copenhagen University Hospital; Copenhagen; Denmark
| |
Collapse
|
42
|
CD4+ CD31+ recent thymic emigrants in CHD7 haploinsufficiency (CHARGE syndrome): A case. Hum Immunol 2013; 74:1047-50. [DOI: 10.1016/j.humimm.2013.06.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 05/24/2013] [Accepted: 06/01/2013] [Indexed: 11/17/2022]
|
43
|
Altered T-cell subsets in HIV-1 natural viral suppressors (elite controllers) with hepatitis C infection. AIDS 2013; 27:1989-92. [PMID: 23660578 DOI: 10.1097/qad.0b013e328362dea4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We have established a cohort of 64 Natural Viral Suppressors (NVS) (similar to Elite Controllers/Elite Suppressors), 30 of which have chronic hepatitis C virus (HCV). We investigated T-cell phenotypic changes in association with HCV infection. NVS without HCV and normal controls had similar T-cell phenotypes. However, NVS with HCV had lower naive cell proportions (CD4 and CD8) compared with NVS without HCV (P = 0.0008 and P = 0.02) or normal controls (P = 0.0163 and P = 0.017). These results and previously reported data suggest that HCV coinfection increases immune activation and T-cell disturbances. Any associated T-cell functional changes or potential clinical consequences need further study.
Collapse
|
44
|
Kuniholm MH, Parrinello CM, Anastos K, Augenbraun M, Plankey M, Nowicki M, Peters M, Golub ET, Lurain N, Landay AL, Strickler HD, Kaplan RC. Hepatitis C viremia is associated with cytomegalovirus IgG antibody levels in HIV-infected women. PLoS One 2013; 8:e61973. [PMID: 23613990 PMCID: PMC3629158 DOI: 10.1371/journal.pone.0061973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 03/15/2013] [Indexed: 11/19/2022] Open
Abstract
Background Individuals with HIV infection exhibit high cytomegalovirus (CMV) IgG levels, but there are few data regarding the association of hepatitis C virus (HCV) with the immune response against CMV. Methods Associations of HCV with CMV seropositivity and CMV IgG levels were studied in 635 HIV-infected women, 187 of whom were HCV-seropositive, with adjustment in multivariable models for age, race/ethnicity, and HIV disease characteristics. Eighty one percent of the women reported receipt of highly active antiretroviral therapy (HAART) prior to or at CMV testing. Results In adjusted models women with chronic HCV had higher CMV IgG levels than those without HCV RNA (β = 2.86, 95% CI:0.89 – 4.83; P = 0.004). The association of HCV RNA with CMV IgG differed by age (Pinteraction = 0.0007), with a strong association observed among women in the low and middle age tertiles (≤45.3 years of age; β = 6.21, 95% CI:3.30 – 9.11, P<0.0001) but not among women in the high age tertile. CMV IgG levels were not associated with non-invasive measures of liver disease, APRI and FIB-4, or with HCV RNA level and adjustment for Epstein-Barr virus (EBV) IgG levels did not affect the association between HCV and CMV. Conclusions CMV IgG levels are higher in HCV/HIV co-infected women than in HIV mono-infected women. Further research on the association of HCV with CMV IgG is indicated because prior studies have found CMV IgG to be associated with morbidity and mortality in the general population and subclinical carotid artery disease in HIV-infected patients.
Collapse
Affiliation(s)
- Mark H Kuniholm
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Hull MW, Rollet K, Klein MB. Reply to Mandorfer et al. Clin Infect Dis 2012; 56:905-6. [PMID: 23243174 DOI: 10.1093/cid/cis1036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
46
|
Saunders JAH, Estes KA, Kosloski LM, Allen HE, Dempsey KM, Torres-Russotto DR, Meza JL, Santamaria PM, Bertoni JM, Murman DL, Ali HH, Standaert DG, Mosley RL, Gendelman HE. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson's disease. J Neuroimmune Pharmacol 2012; 7:927-38. [PMID: 23054369 PMCID: PMC3515774 DOI: 10.1007/s11481-012-9402-z] [Citation(s) in RCA: 236] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 08/27/2012] [Indexed: 02/06/2023]
Abstract
Animal models and clinical studies have linked the innate and adaptive immune system to the pathology of Parkinson's disease (PD). Despite such progress, the specific immune responses that influence disease progression have eluded investigators. Herein, we assessed relationships between T cell phenotype and function with PD progression. Peripheral blood lymphocytes from two separate cohorts, a discovery cohort and a validation cohort, totaling 113 PD patients and 96 age- and environment-matched caregivers were examined by flow cytometric analysis and T cell proliferation assays. Increased effector/memory T cells (Tem), defined as CD45RO+ and FAS+ CD4+ T cells and decreased CD31+ and α4β7+ CD4+ T cells were associated with progressive Unified Parkinson's Disease Rating Scale III scores. However, no associations were seen between immune biomarkers and increased age or disease duration. Impaired abilities of regulatory T cells (Treg) from PD patients to suppress effector T cell function was observed. These data support the concept that chronic immune stimulation, notably Tem activation and Treg dysfunction is linked to PD pathobiology and disease severity, but not disease duration. The association of T cell phenotypes with motor symptoms provides fresh avenues for novel biomarkers and therapeutic designs.
Collapse
Affiliation(s)
- Jessica A. Hutter Saunders
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Katherine A. Estes
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Lisa M. Kosloski
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Heather E. Allen
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL USA
| | - Kathryn M. Dempsey
- College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE USA
| | | | - Jane L. Meza
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE USA
| | | | - John M. Bertoni
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE USA
| | - Daniel L. Murman
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE USA
| | - Hesham H. Ali
- College of Information Science and Technology, University of Nebraska at Omaha, Omaha, NE USA
| | - David G. Standaert
- Department of Neurology, University of Alabama at Birmingham, Birmingham, AL USA
| | - R. Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
- Department of Pharmacology and Experimental Neuroscience Movement Disorders Program, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| | - Howard E. Gendelman
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880 USA
| |
Collapse
|
47
|
Rasmussen AL, Wang IM, Shuhart MC, Proll SC, He Y, Cristescu R, Roberts C, Carter VS, Williams CM, Diamond DL, Bryan JT, Ulrich R, Korth MJ, Thomassen LV, Katze MG. Chronic immune activation is a distinguishing feature of liver and PBMC gene signatures from HCV/HIV coinfected patients and may contribute to hepatic fibrogenesis. Virology 2012; 430:43-52. [PMID: 22608059 DOI: 10.1016/j.virol.2012.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 04/06/2012] [Accepted: 04/17/2012] [Indexed: 02/06/2023]
Abstract
Hepatitis C virus/human immunodeficiency virus (HCV/HIV) coinfected patients demonstrate accelerated progression to severe liver injury in comparison to HCV monoinfected patients, although the underlying mechanisms are unclear owing to infection of separate tissue compartments with two distinct viral pathogens. Microarray analysis of paired liver biopsy and peripheral blood mononuclear cell (PBMC) specimens from HCV/HIV coinfected and HCV monoinfected patients identified a gene expression signature associated with increased inflammation and immune activation that was present only in liver and PBMC samples from coinfected patients. We also identified in these samples liver- and PBMC-specific signatures enriched with fibrogenic/hepatic stellate activation and proinflammatory genes, respectively. Finally, Bayesian networks were constructed by assimilating these data with existing data from liver and PBMC samples from other cohorts, augmenting enrichment of biologically important pathways and further indicating that chronic immune activation in HCV/HIV coinfection may exacerbate liver disease progression in coinfected patients.
Collapse
Affiliation(s)
- Angela L Rasmussen
- Department of Microbiology, University of Washington, Box 358070, Seattle, WA 98195, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Maniar A, Ellis C, Asmuth D, Pollard R, Rutledge J. HIV infection and atherosclerosis: evaluating the drivers of inflammation. Eur J Prev Cardiol 2012; 20:720-8. [DOI: 10.1177/2047487312447843] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Archana Maniar
- Department of Medicine, University of California Davis, USA
| | - Collin Ellis
- Department of Medicine, University of California Davis, USA
| | - David Asmuth
- Department of Medicine, University of California Davis, USA
| | | | - John Rutledge
- Department of Medicine, University of California Davis, USA
| |
Collapse
|
49
|
Steiner KL, Malhotra I, Mungai PL, Muchiri EM, Dent AE, King CL. In utero activation of fetal memory T cells alters host regulatory gene expression and affects HIV susceptibility. Virology 2012; 425:23-30. [PMID: 22280894 DOI: 10.1016/j.virol.2012.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Revised: 10/16/2011] [Accepted: 01/04/2012] [Indexed: 11/26/2022]
Abstract
In utero priming to malaria antigens renders cord blood mononuclear cells (CBMC) more susceptible to productive HIV infection in vitro in the absence of exogenous stimulation. This provides a unique model to better understand mechanisms affecting lymphocyte susceptibility to HIV infection in vivo. Effector memory CD3(+)CD4(+) T cells (T(EM)) were the exclusive initial targets of HIV with rapid spread to central memory cells. HIV susceptibility correlated with increased expression of CD25 and HLA-DR on T(EM). Virus entered all samples equally, however gag/pol RNA was only detected in HIV susceptible samples, suggesting regulation of proviral gene transcription. Targeted analysis of human genes in memory T cells showed greater expression of IFNG, NFATc1, IRF1, FOS, and PPIA and decreased expression YY1 and TFCP2 in HIV susceptible samples. Thus fetal priming to exogenous antigens enhances specific proviral gene transcription pathways in effector memory cells that may increase risk of vertical transmission of HIV.
Collapse
Affiliation(s)
- Kevin L Steiner
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH 44106-7286, USA
| | | | | | | | | | | |
Collapse
|
50
|
Roohvand F, Kossari N. Advances in hepatitis C virus vaccines, Part one: Advances in basic knowledge for hepatitis C virus vaccine design. Expert Opin Ther Pat 2011; 21:1811-30. [PMID: 22022980 DOI: 10.1517/13543776.2011.630662] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Around 3% of the world population is infected with HCV, with 3 - 4 million newly infected subjects added to this reservoir every year. At least 10% of these people will develop liver cirrhosis or cancer over time, while no approved vaccine against HCV infection is available to date. AREAS COVERED This paper includes a detailed and correlated patent (selected by HCAPLUS search database) and literature (searched by PubMed) review on the HCV genome, proteins and key epitopes (including underestimated HCV proteins, alternate reading frame proteins), HCV immunology, immunosuppressive mechanisms and protective correlations of immunity in acute and chronic states of infection (features for prophylactic and therapeutic HCV vaccine design), recent HCV cell culture systems (HCV/JFH1) and animal models. In part two of this review, advances in HCV vaccine formulations and modalities as well as a detailed list of the current trials for HCV vaccine and discussion of the pros and cones of different strategies will be provided. EXPERT OPINION By using the advanced basic knowledge and tools obtained about HCV vaccinology in recent years and the application of novel formulations and modalities, at least partially effective vaccines will become available in the near future to prevent (or treat) the chronic (if not the acute) state of HCV infection. A few of such vaccines are already in clinical trials.
Collapse
Affiliation(s)
- Farzin Roohvand
- Pasteur Institute of Iran, Hepatitis & AIDS Department, Pasteur Ave., Tehran, Iran.
| | | |
Collapse
|