851
|
Abstract
Recent years have seen a significant increase in understanding of the host genetic and genomic determinants of susceptibility to HIV-1 infection and disease progression, driven in large part by candidate gene studies, genome-wide association studies, genome-wide transcriptome analyses, and large-scale in vitro genome screens. These studies have identified common variants in some host loci that clearly influence disease progression, characterized the scale and dynamics of gene and protein expression changes in response to infection, and provided the first comprehensive catalogs of genes and pathways involved in viral replication. Experimental models of AIDS and studies in natural hosts of primate lentiviruses have complemented and in some cases extended these findings. As the relevant technology continues to progress, the expectation is that such studies will increase in depth (e.g., to include host whole exome and whole genome sequencing) and in breadth (in particular, by integrating multiple data types).
Collapse
Affiliation(s)
- Amalio Telenti
- Institute of Microbiology, University Hospital and University of Lausanne, 1011 Lausanne, Switzerland.
| | | |
Collapse
|
852
|
Li JZ, Brumme CJ, Lederman MM, Brumme ZL, Wang H, Spritzler J, Carrington M, Medvik K, Walker BD, Schooley RT, Kuritzkes DR. Characteristics and outcomes of initial virologic suppressors during analytic treatment interruption in a therapeutic HIV-1 gag vaccine trial. PLoS One 2012; 7:e34134. [PMID: 22479542 PMCID: PMC3316607 DOI: 10.1371/journal.pone.0034134] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 02/22/2012] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND In the placebo-controlled trial ACTG A5197, a trend favoring viral suppression was seen in the HIV-1-infected subjects who received a recombinant Ad5 HIV-1 gag vaccine. OBJECTIVE To identify individuals with initial viral suppression (plasma HIV-1 RNA set point <3.0 log(10) copies/ml) during the analytic treatment interruption (ATI) and evaluate the durability and correlates of virologic control and characteristics of HIV sequence evolution. METHODS HIV-1 gag and pol RNA were amplified and sequenced from plasma obtained during the ATI. Immune responses were measured by flow cytometric analysis and intracellular cytokine expression assays. Characteristics of those with and without initial viral suppression were compared using the Wilcoxon rank sum and Fisher's exact tests. RESULTS Eleven out of 104 participants (10.6%) were classified as initial virologic suppressors, nine of whom had received the vaccine. Initial virologic suppressors had significantly less CD4+ cell decline by ATI week 16 as compared to non-suppressors (median 7 CD4+ cell gain vs. 247 CD4+ cell loss, P = 0.04). However, of the ten initial virologic suppressors with a pVL at ATI week 49, only three maintained pVL <3.0 log(10) copies/ml. HIV-1 Gag-specific CD4+ interferon-γ responses were not associated with initial virologic suppression and no evidence of vaccine-driven HIV sequence evolution was detected. Participants with initial virologic suppression were found to have a lower percentage of CD4+ CTLA-4+ cells prior to treatment interruption, but a greater proportion of HIV-1 Gag-reactive CD4+ TNF-α+ cells expressing either CTLA-4 or PD-1. CONCLUSIONS Among individuals participating in a rAd5 therapeutic HIV-1 gag vaccine trial, initial viral suppression was found in a subset of patients, but this response was not sustained. The association between CTLA-4 and PD-1 expression on CD4+ T cells and virologic outcome warrants further study in trials of other therapeutic vaccines in development. TRIAL REGISTRATION ClinicalTrials.gov NCT00080106.
Collapse
Affiliation(s)
- Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, United States of America.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
853
|
The colocalization potential of HIV-specific CD8+ and CD4+ T-cells is mediated by integrin β7 but not CCR6 and regulated by retinoic acid. PLoS One 2012; 7:e32964. [PMID: 22470433 PMCID: PMC3314661 DOI: 10.1371/journal.pone.0032964] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 02/08/2012] [Indexed: 12/22/2022] Open
Abstract
CD4+ T-cells from gut-associated lymphoid tissues (GALT) are major targets for HIV-1 infection. Recruitment of excess effector CD8+ T-cells in the proximity of target cells is critical for the control of viral replication. Here, we investigated the colocalization potential of HIV-specific CD8+ and CD4+ T-cells into the GALT and explored the role of retinoic acid (RA) in regulating this process in a cohort of HIV-infected subjects with slow disease progression. The expression of the gut-homing molecules integrin β7, CCR6, and CXCR3 was identified as a “signature” for HIV-specific but not CMV-specific CD4+ T-cells thus providing a new explanation for their enhanced permissiveness to infection in vivo. HIV-specific CD8+ T-cells also expressed high levels of integrin β7 and CXCR3; however CCR6 was detected at superior levels on HIV-specific CD4+versus CD8+ T-cells. All trans RA (ATRA) upregulated the expression of integrin β7 but not CCR6 on HIV-specific T-cells. Together, these results suggest that HIV-specific CD8+ T-cells may colocalize in excess with CD4+ T-cells into the GALT via integrin β7 and CXCR3, but not via CCR6. Considering our previous findings that CCR6+CD4+ T-cells are major cellular targets for HIV-DNA integration in vivo, a limited ability of CD8+ T-cells to migrate in the vicinity of CCR6+CD4+ T-cells may facilitate HIV replication and dissemination at mucosal sites.
Collapse
|
854
|
Fairfax BP, Makino S, Radhakrishnan J, Plant K, Leslie S, Dilthey A, Ellis P, Langford C, Vannberg FO, Knight JC. Genetics of gene expression in primary immune cells identifies cell type-specific master regulators and roles of HLA alleles. Nat Genet 2012; 44:502-10. [PMID: 22446964 PMCID: PMC3437404 DOI: 10.1038/ng.2205] [Citation(s) in RCA: 379] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 01/31/2012] [Indexed: 12/15/2022]
Abstract
Trans-acting genetic variants have a substantial, albeit poorly characterized, role in the heritable determination of gene expression. Using paired purified primary monocytes and B cells, we identify new predominantly cell type-specific cis and trans expression quantitative trait loci (eQTLs), including multi-locus trans associations to LYZ and KLF4 in monocytes and B cells, respectively. Additionally, we observe a B cell-specific trans association of rs11171739 at 12q13.2, a known autoimmune disease locus, with IP6K2 (P = 5.8 × 10(-15)), PRIC285 (P = 3.0 × 10(-10)) and an upstream region of CDKN1A (P = 2 × 10(-52)), suggesting roles for cell cycle regulation and peroxisome proliferator-activated receptor γ (PPARγ) signaling in autoimmune pathogenesis. We also find that specific human leukocyte antigen (HLA) alleles form trans associations with the expression of AOAH and ARHGAP24 in monocytes but not in B cells. In summary, we show that mapping gene expression in defined primary cell populations identifies new cell type-specific trans-regulated networks and provides insights into the genetic basis of disease susceptibility.
Collapse
Affiliation(s)
- Benjamin P Fairfax
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
855
|
Host-pathogen interactions revealed by human genome-wide surveys. Trends Genet 2012; 28:233-43. [PMID: 22445588 DOI: 10.1016/j.tig.2012.02.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2011] [Revised: 02/02/2012] [Accepted: 02/02/2012] [Indexed: 12/21/2022]
Abstract
Genome-wide association studies (GWAS) have now convincingly shown that the diverse outcomes (such as the resolution of infection, clinical deterioration to severe disease, or progression from acute infection to persistent infection) that occur following microbial infection can be at least partly explained by human genetic variation. Unbiased whole-genome approaches have revealed unprecedentedly robust associations between genetic markers and susceptibility to disease, providing clear insights into our understanding of infectious disease biology by revealing the crucial host-pathogen interaction sites. Further work characterizing both the host causative variations and pathogenic microbial strains with distinct host interactions and disease outcomes is now required to provide potential new intervention strategies.
Collapse
|
856
|
Saxena R, Elbers C, Guo Y, Peter I, Gaunt T, Mega J, Lanktree M, Tare A, Castillo B, Li Y, Johnson T, Bruinenberg M, Gilbert-Diamond D, Rajagopalan R, Voight B, Balasubramanyam A, Barnard J, Bauer F, Baumert J, Bhangale T, Böhm B, Braund P, Burton P, Chandrupatla H, Clarke R, Cooper-DeHoff R, Crook E, Davey-Smith G, Day I, de Boer A, de Groot M, Drenos F, Ferguson J, Fox C, Furlong C, Gibson Q, Gieger C, Gilhuijs-Pederson L, Glessner J, Goel A, Gong Y, Grant S, Grobbee D, Hastie C, Humphries S, Kim C, Kivimaki M, Kleber M, Meisinger C, Kumari M, Langaee T, Lawlor D, Li M, Lobmeyer M, Maitland-van der Zee AH, Meijs M, Molony C, Morrow D, Murugesan G, Musani S, Nelson C, Newhouse S, O'Connell J, Padmanabhan S, Palmen J, Patel S, Pepine C, Pettinger M, Price T, Rafelt S, Ranchalis J, Rasheed A, Rosenthal E, Ruczinski I, Shah S, Shen H, Silbernagel G, Smith E, Spijkerman A, Stanton A, Steffes M, Thorand B, Trip M, van der Harst P, van der A D, van Iperen E, van Setten J, van Vliet-Ostaptchouk J, Verweij N, Wolffenbuttel B, Young T, Zafarmand M, Zmuda J, Boehnke M, Altshuler D, McCarthy M, Kao W, Pankow J, Cappola T, Sever P, et alSaxena R, Elbers C, Guo Y, Peter I, Gaunt T, Mega J, Lanktree M, Tare A, Castillo B, Li Y, Johnson T, Bruinenberg M, Gilbert-Diamond D, Rajagopalan R, Voight B, Balasubramanyam A, Barnard J, Bauer F, Baumert J, Bhangale T, Böhm B, Braund P, Burton P, Chandrupatla H, Clarke R, Cooper-DeHoff R, Crook E, Davey-Smith G, Day I, de Boer A, de Groot M, Drenos F, Ferguson J, Fox C, Furlong C, Gibson Q, Gieger C, Gilhuijs-Pederson L, Glessner J, Goel A, Gong Y, Grant S, Grobbee D, Hastie C, Humphries S, Kim C, Kivimaki M, Kleber M, Meisinger C, Kumari M, Langaee T, Lawlor D, Li M, Lobmeyer M, Maitland-van der Zee AH, Meijs M, Molony C, Morrow D, Murugesan G, Musani S, Nelson C, Newhouse S, O'Connell J, Padmanabhan S, Palmen J, Patel S, Pepine C, Pettinger M, Price T, Rafelt S, Ranchalis J, Rasheed A, Rosenthal E, Ruczinski I, Shah S, Shen H, Silbernagel G, Smith E, Spijkerman A, Stanton A, Steffes M, Thorand B, Trip M, van der Harst P, van der A D, van Iperen E, van Setten J, van Vliet-Ostaptchouk J, Verweij N, Wolffenbuttel B, Young T, Zafarmand M, Zmuda J, Boehnke M, Altshuler D, McCarthy M, Kao W, Pankow J, Cappola T, Sever P, Poulter N, Caulfield M, Dominiczak A, Shields D, Bhatt DL, Zhang L, Curtis S, Danesh J, Casas J, van der Schouw Y, Onland-Moret N, Doevendans P, Dorn G, Farrall M, FitzGerald G, Hamsten A, Hegele R, Hingorani A, Hofker M, Huggins G, Illig T, Jarvik G, Johnson J, Klungel O, Knowler W, Koenig W, März W, Meigs J, Melander O, Munroe P, Mitchell B, Bielinski S, Rader D, Reilly M, Rich S, Rotter J, Saleheen D, Samani N, Schadt E, Shuldiner A, Silverstein R, Kottke-Marchant K, Talmud P, Watkins H, Asselbergs FW, de Bakker P, McCaffery J, Wijmenga C, Sabatine M, Wilson J, Reiner A, Bowden D, Hakonarson H, Siscovick D, Keating B. Large-scale gene-centric meta-analysis across 39 studies identifies type 2 diabetes loci. Am J Hum Genet 2012; 90:410-25. [PMID: 22325160 PMCID: PMC3309185 DOI: 10.1016/j.ajhg.2011.12.022] [Show More Authors] [Citation(s) in RCA: 195] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 12/06/2011] [Accepted: 12/31/2011] [Indexed: 01/12/2023] Open
Abstract
To identify genetic factors contributing to type 2 diabetes (T2D), we performed large-scale meta-analyses by using a custom ∼50,000 SNP genotyping array (the ITMAT-Broad-CARe array) with ∼2000 candidate genes in 39 multiethnic population-based studies, case-control studies, and clinical trials totaling 17,418 cases and 70,298 controls. First, meta-analysis of 25 studies comprising 14,073 cases and 57,489 controls of European descent confirmed eight established T2D loci at genome-wide significance. In silico follow-up analysis of putative association signals found in independent genome-wide association studies (including 8,130 cases and 38,987 controls) performed by the DIAGRAM consortium identified a T2D locus at genome-wide significance (GATAD2A/CILP2/PBX4; p = 5.7 × 10(-9)) and two loci exceeding study-wide significance (SREBF1, and TH/INS; p < 2.4 × 10(-6)). Second, meta-analyses of 1,986 cases and 7,695 controls from eight African-American studies identified study-wide-significant (p = 2.4 × 10(-7)) variants in HMGA2 and replicated variants in TCF7L2 (p = 5.1 × 10(-15)). Third, conditional analysis revealed multiple known and novel independent signals within five T2D-associated genes in samples of European ancestry and within HMGA2 in African-American samples. Fourth, a multiethnic meta-analysis of all 39 studies identified T2D-associated variants in BCL2 (p = 2.1 × 10(-8)). Finally, a composite genetic score of SNPs from new and established T2D signals was significantly associated with increased risk of diabetes in African-American, Hispanic, and Asian populations. In summary, large-scale meta-analysis involving a dense gene-centric approach has uncovered additional loci and variants that contribute to T2D risk and suggests substantial overlap of T2D association signals across multiple ethnic groups.
Collapse
Affiliation(s)
- Richa Saxena
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Clara C. Elbers
- Department of Genetics, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA
- Complex Genetics Section, Department of Medical Genetics, University Medical Center Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Yiran Guo
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- BGI Shenzhen, Beishan Industrial Zone, Yantian District, Shenzhen 518083, China
| | - Inga Peter
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, NY 10029 USA
| | - Tom R. Gaunt
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, Department of Social Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Jessica L. Mega
- Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 021155 USA
| | - Matthew B. Lanktree
- Department of Biochemistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Archana Tare
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Berta Almoguera Castillo
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Servicio de Genética Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Avda. Reyes Católicos 228040, Madrid, Spain
| | - Yun R. Li
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Toby Johnson
- Clinical Pharmacology, Barts and the London Genome Centre, Queen Mary University of London, London EC1M 6BQ, UK
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Marcel Bruinenberg
- LifeLines Cohort Study and Biobank, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Diane Gilbert-Diamond
- Children's Environmental Health and Disease Prevention Center at Dartmouth, Hanover, NH 03755, USA
- Section of Biostatistics and Epidemiology, Department of Community and Family Medicine, Dartmouth Medical School, Hanover, NH 03756, USA
| | | | - Benjamin F. Voight
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - Ashok Balasubramanyam
- Translational Metabolism Unit, Division of Diabetes, Endocrinology and Metabolism, Baylor College of Medicine, Houston, TX 77030, USA
| | - John Barnard
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Florianne Bauer
- Complex Genetics Section, Department of Medical Genetics, University Medical Center Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Jens Baumert
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Tushar Bhangale
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Bernhard O. Böhm
- Cardiology Group Frankfurt-Sachsenhausen, Frankfurt 60598, Germany
| | - Peter S. Braund
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Paul R. Burton
- Department of Health Sciences, University of Leicester, University Rd, Leicester LE1 7RH, UK
| | - Hareesh R. Chandrupatla
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Robert Clarke
- Clinical Trial Service Unit, Richard Doll Building, Old Road Campus, Roosevelt Drive, Oxford OX37LF, UK
| | - Rhonda M. Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL 32610, USA
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | | | - George Davey-Smith
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, Department of Social Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Ian N. Day
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, Department of Social Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Anthonius de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Mark C.H. de Groot
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Fotios Drenos
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Jane Ferguson
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Caroline S. Fox
- Framingham Heart Study, Boston University School of Medicine, Boston, MA 02118, USA
| | - Clement E. Furlong
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA 98195, USA
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Quince Gibson
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Lisa A. Gilhuijs-Pederson
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Joseph T. Glessner
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Anuj Goel
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Yan Gong
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Struan F.A. Grant
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Diederick E. Grobbee
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Claire Hastie
- British Heart Foundation Glasgow Cardiovascular Research Centre, Division of Cardiovascular and Medical Sciences, Western Infirmary, University of Glasgow, Glasgow G12 8TA, UK
| | - Steve E. Humphries
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Cecilia E. Kim
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, University College London, London, UK
- Genetic Epidemiology Group, Department of Epidemiology and Public Health, University College London, London WC1E 6BT, UK
| | - Marcus Kleber
- LURIC Study, Freiburg im Breisgau 79098, Germany
- Synlab Center of Laboratory Diagnostics Heidelberg, Heidelberg 69037, Germany
| | - Christa Meisinger
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Meena Kumari
- Genetic Epidemiology Group, Department of Epidemiology and Public Health, University College London, London WC1E 6BT, UK
| | - Taimour Y. Langaee
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL 32610, USA
| | - Debbie A. Lawlor
- Medical Research Council Centre for Causal Analyses in Translational Epidemiology, Department of Social Medicine, University of Bristol, Oakfield House, Oakfield Grove, Bristol BS8 2BN, UK
| | - Mingyao Li
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Maximilian T. Lobmeyer
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Anke-Hilse Maitland-van der Zee
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Matthijs F.L. Meijs
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cliona M. Molony
- Department of Genetics, Rosetta Inpharmatics, Seattle, WA 98109, USA
| | - David A. Morrow
- Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 021155 USA
| | - Gurunathan Murugesan
- Department of Clinical Pathology, Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Solomon K. Musani
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Christopher P. Nelson
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Stephen J. Newhouse
- Clinical Pharmacology, Barts and the London Genome Centre, Queen Mary University of London, London EC1M 6BQ, UK
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jeffery R. O'Connell
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sandosh Padmanabhan
- British Heart Foundation Glasgow Cardiovascular Research Centre, Division of Cardiovascular and Medical Sciences, Western Infirmary, University of Glasgow, Glasgow G12 8TA, UK
| | - Jutta Palmen
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Sanjey R. Patel
- Division of Sleep Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Carl J. Pepine
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Mary Pettinger
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Thomas S. Price
- Medical Research Council Social, Genetic, and Developmental Psychiatry Centre, Institute of Psychiatry, King's College London, London WC2R 2LS, UK
| | - Suzanne Rafelt
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, LE3 9QP, UK
| | - Jane Ranchalis
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA 98195, USA
| | - Asif Rasheed
- Center for Non-Communicable Diseases, Karachi, Pakistan
| | - Elisabeth Rosenthal
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA 98195, USA
| | - Ingo Ruczinski
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Sonia Shah
- University College Genetics Institute, University College London, 5 University St London, WC1E 6BT, UK
| | - Haiqing Shen
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Günther Silbernagel
- Division of Endocrinology, Diabetology, Nephrology, Vascular Disease, and Clinical Chemistry, Department of Internal Medicine, Eberhard-Karls-University Tübingen, Tübingen 72074, Germany
| | | | | | - Alice Stanton
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Michael W. Steffes
- Department of Laboratory Medicine and Pathology, Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - Barbara Thorand
- Institute of Epidemiology II, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Mieke Trip
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen and Groningen University, 9700 RB Groningen, The Netherlands
| | - Daphne L. van der A
- National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | | - Jessica van Setten
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jana V. van Vliet-Ostaptchouk
- Molecular Genetics, Department of Pathology and Medical Biology, University Medical Center Groningen and University of Groningen, The Netherlands
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Niek Verweij
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bruce H.R. Wolffenbuttel
- Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Taylor Young
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
| | - M. Hadi Zafarmand
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joseph M. Zmuda
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, 130 DeSoto St, Pittsburgh, PA 15261, USA
| | | | | | - Michael Boehnke
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - David Altshuler
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Mark McCarthy
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford OX3 7LJ, UK
| | - W.H. Linda Kao
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21287, USA
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN 55454, USA
| | - Thomas P. Cappola
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Peter Sever
- International Centre for Circulatory Health, Imperial College London, London W2 1PG, UK
| | - Neil Poulter
- International Centre for Circulatory Health, Imperial College London, London W2 1PG, UK
| | - Mark Caulfield
- Clinical Pharmacology, Barts and the London Genome Centre, Queen Mary University of London, London EC1M 6BQ, UK
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Anna Dominiczak
- British Heart Foundation Glasgow Cardiovascular Research Centre, Division of Cardiovascular and Medical Sciences, Western Infirmary, University of Glasgow, Glasgow G12 8TA, UK
| | - Denis C. Shields
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | | | - Li Zhang
- Department of Quantitative Health Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Sean P. Curtis
- Merck Research Laboratories, P.O. Box 2000, Rahway, NJ 07065, USA
| | - John Danesh
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, CB1 8RN, UK
| | - Juan P. Casas
- Department of Epidemiology and Public Health, University College London, London, UK
- Department of Non-communicable Disease Epidemiology, Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Yvonne T. van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - N. Charlotte Onland-Moret
- Complex Genetics Section, Department of Medical Genetics, University Medical Center Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Pieter A. Doevendans
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerald W. Dorn
- Washington University Center for Pharmacogenetics, 660 S. Euclid Ave, Campus Box 8220, St. Louis, MO 63110, USA
| | - Martin Farrall
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
- Department of Cardiovascular Medicine, University of Oxford, Level 6 West Wing, John Radcliffe Hospital, Headley Way, Headington, Oxford OX3 9DU, UK
| | - Garret A. FitzGerald
- The Institute for Translational Medicine and Therapeutics, School of Medicine, University of Pennsylvania, Philadelphia, PA 19146, USA
| | - Anders Hamsten
- Cardiovascular Genetics Group, Atherosclerosis Research Unit, Department of Medicine Solna, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Robert Hegele
- Department of Biochemistry, University of Western Ontario, London, ON N6A 5C1, Canada
| | - Aroon D. Hingorani
- Centre for Clinical Pharmacology, Department of Medicine, University College London, London WC1E 6JF, UK
| | - Marten H. Hofker
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Gordon S. Huggins
- Molecular Cardiology Research Institute, Center for Translational Genomics, Tufts Medical Center and Tufts University, Boston, MA 02114, USA
| | - Thomas Illig
- Institute of Genetic Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Hannover Unified Biobank, Hannover Medical School, 30625 Hannover, Germany
| | - Gail P. Jarvik
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA 98195, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research, University of Florida College of Pharmacy, Gainesville, FL 32610, USA
- Division of Cardiovascular Medicine, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Olaf H. Klungel
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - William C. Knowler
- National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85104, USA
| | - Wolfgang Koenig
- Department of Internal Medicine II-Cardiology, University of Ulm Medical Center, Ulm, Germany
| | - Winfried März
- Synlab Center of Laboratory Diagnostics Heidelberg, Heidelberg 69037, Germany
- Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, University of Heidelberg D-68167 Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8010 Graz, Austria
| | - James B. Meigs
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
- General Medicine Division, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Olle Melander
- Clinical Research Center, Malmö University Hospital, Malmö SE-205 02, Sweden
| | - Patricia B. Munroe
- Clinical Pharmacology, Barts and the London Genome Centre, Queen Mary University of London, London EC1M 6BQ, UK
- William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London EC1M 6BQ, UK
| | - Braxton D. Mitchell
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Susan J. Bielinski
- Division of Epidemiology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Daniel J. Rader
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Muredach P. Reilly
- Cardiovascular Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22902, USA
| | - Jerome I. Rotter
- Medical Genetics Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Danish Saleheen
- Center for Non-Communicable Diseases, Karachi, Pakistan
- Merck Research Laboratories, P.O. Box 2000, Rahway, NJ 07065, USA
| | - Nilesh J. Samani
- Department of Cardiovascular Sciences, University of Leicester, Glenfield Hospital, Leicester, LE3 9QP, UK
| | | | - Alan R. Shuldiner
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Roy Silverstein
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation, Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Avenue Cleveland, OH 44195, USA
| | | | - Philippa J. Talmud
- Centre for Cardiovascular Genetics, Department of Medicine, University College London, 5 University Street, London, WC1E 6JF, UK
| | - Hugh Watkins
- Washington University Center for Pharmacogenetics, 660 S. Euclid Ave, Campus Box 8220, St. Louis, MO 63110, USA
| | - Folkert W. Asselbergs
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
- Department of Cardiology, Division Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Paul I.W. de Bakker
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA
- Complex Genetics Section, Department of Medical Genetics, University Medical Center Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeanne McCaffery
- Weight Control and Diabetes Research Center, The Miriam Hospital and Warren Alpert School of Medicine at Brown University, Providence, RI 02906, USA
| | - Cisca Wijmenga
- Department of Genetics, University Medical Center Groningen and Groningen University, 9700 RB Groningen, The Netherlands
| | - Marc S. Sabatine
- Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 021155 USA
| | - James G. Wilson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Alex Reiner
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Donald W. Bowden
- Center for Human Genomics, Wake Forest University School of Medicine, Winston-Salem, NC 27106, USA
| | - Hakon Hakonarson
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - David S. Siscovick
- Cardiovascular Health Research Unit, Departments of Medicine and Epidemiology, University of Washington, Seattle, WA 98101, USA
| | - Brendan J. Keating
- Center for Applied Genomics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Human Genetics, Abramson Research Center, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
857
|
Varadarajan N, Kwon DS, Law KM, Ogunniyi AO, Anahtar MN, Richter JM, Walker BD, Love JC. Rapid, efficient functional characterization and recovery of HIV-specific human CD8+ T cells using microengraving. Proc Natl Acad Sci U S A 2012; 109:3885-90. [PMID: 22355106 PMCID: PMC3309713 DOI: 10.1073/pnas.1111205109] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The nature of certain clinical samples (tissue biopsies, fluids) or the subjects themselves (pediatric subjects, neonates) often constrain the number of cells available to evaluate the breadth of functional T-cell responses to infections or therapeutic interventions. The methods most commonly used to assess this functional diversity ex vivo and to recover specific cells to expand in vitro usually require more than 10(6) cells. Here we present a process to identify antigen-specific responses efficiently ex vivo from 10(4)-10(5) single cells from blood or mucosal tissues using dense arrays of subnanoliter wells. The approach combines on-chip imaging cytometry with a technique for capturing secreted proteins--called "microengraving"--to enumerate antigen-specific responses by single T cells in a manner comparable to conventional assays such as ELISpot and intracellular cytokine staining. Unlike those assays, however, the individual cells identified can be recovered readily by micromanipulation for further characterization in vitro. Applying this method to assess HIV-specific T-cell responses demonstrates that it is possible to establish clonal CD8(+) T-cell lines that represent the most abundant specificities present in circulation using 100- to 1,000-fold fewer cells than traditional approaches require and without extensive genotypic analysis a priori. This rapid (<24 h), efficient, and inexpensive process should improve the comparative study of human T-cell immunology across ages and anatomic compartments.
Collapse
Affiliation(s)
- Navin Varadarajan
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Douglas S. Kwon
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Charlestown, MA 02129; Divisions of
- Infectious Diseases and
| | - Kenneth M. Law
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Charlestown, MA 02129; Divisions of
| | - Adebola O. Ogunniyi
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Melis N. Anahtar
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Charlestown, MA 02129; Divisions of
| | - James M. Richter
- Gastroenterology, Massachusetts General Hospital, Boston, MA 02114; and
| | - Bruce D. Walker
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Charlestown, MA 02129; Divisions of
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - J. Christopher Love
- Department of Chemical Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Charlestown, MA 02129; Divisions of
| |
Collapse
|
858
|
Abstract
Low- to middle-income countries bear the overwhelming burden of the human immunodeficiency virus type 1 (HIV-1) epidemic in terms of the numbers of their citizens living with HIV/AIDS (acquired immunodeficiency syndrome), the high degrees of viral diversity often involving multiple HIV-1 clades circulating within their populations, and the social and economic factors that compromise current control measures. Distinct epidemics have emerged in different geographical areas. These epidemics differ in their severity, the population groups they affect, their associated risk behaviors, and the viral strains that drive them. In addition to inflicting great human cost, the high burden of HIV infection has a major impact on the social and economic development of many low- to middle-income countries. Furthermore, the high degrees of viral diversity associated with multiclade HIV epidemics impacts viral diagnosis and pathogenicity and treatment and poses daunting challenges for effective vaccine development.
Collapse
Affiliation(s)
- Yiming Shao
- State Key Laboratory for Infectious Disease Control and Prevention, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Changping District, Beijing 102206, China
| | | |
Collapse
|
859
|
Bagasra O, Bagasra AU, Sheraz M, Pace DG. Potential utility of GB virus type C as a preventive vaccine for HIV-1. Expert Rev Vaccines 2012; 11:335-347. [PMID: 22380825 DOI: 10.1586/erv.11.191] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Recent flurries of literature on the beneficial effects of GB virus type C (GBV-C), a hepatitis C-related virus, in HIV-1 coinfected individuals have raised the possibility of its potential use as a preventive vaccine in people with a high risk for HIV-1. However, these findings are still controversial, and the mechanisms contributing to the apparent beneficial effects of GBV-C are still unresolved. Researchers debate whether the beneficial effects of coinfection of GBV-C in HIV-1-infected individuals are due to GBV-C viremia or rather the presence of GBV-C anti-E2 antibodies. We review the strengths and weaknesses of various aspects of the GBV-C debate and propose a new perspective involving intracellular molecular events that attempts to synthesize numerous contrasting perspectives and ideas, while suggesting new directions for future research in this area.
Collapse
Affiliation(s)
- Omar Bagasra
- South Carolina Center for Biotechnology, Claflin University, Orangeburg, SC, USA.
| | | | | | | |
Collapse
|
860
|
Carter CJ. Extensive viral mimicry of 22 AIDS-related autoantigens by HIV-1 proteins and pathway analysis of 561 viral/human homologues suggest an initial treatable autoimmune component of AIDS. ACTA ACUST UNITED AC 2012; 63:254-68. [PMID: 22077229 DOI: 10.1111/j.1574-695x.2011.00848.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
HIV-1 viral proteins, particularly the env protein, are homologous to 22 AIDS autoantigens, suggesting their creation by antiviral antibodies subsequently targeting human homologues. They include antibodies to T-cell receptors, CD4 and CD95, complement components, IgG, TNF and other immune-related proteins. Autoantibodies may compromise the immune system via knockdown of these key proteins, and autoimmune attack on the immune system itself, as supported by immune activation in early stages of infection and during the transition to AIDS. Over 500 human proteins contain pentapeptides or longer consensi, identical to viral peptides. Such homology explains the extensive viral/human interactome, likely related to the ability of viral homologues to compete with human counterparts as binding partners. Pathway analysis of these homologous proteins revealed their involvement in immune-related networks (e.g. natural killer cell toxicity/toll, T-cell/B-cell receptor signalling/antigen processing) and viral and bacterial entry and defence pathways (phagosome/lysosome pathways, DNA sensing/NOD/RIG-1 pathways) relevant to AIDS pathogenesis. At its inception, AIDS may have an autoimmune component selectively targeting the immune system. Immunosuppressive therapy or antibody removal, which has already achieved some success, might be therapeutically beneficial, particularly if targeted at removal of the culpable antibodies, via affinity dialysis.
Collapse
Affiliation(s)
- Chris J Carter
- PolygenicPathways, St Leonard's on Sea, East Sussex, UK.
| |
Collapse
|
861
|
|
862
|
Limou S, Delaneau O, van Manen D, An P, Sezgin E, Le Clerc S, Coulonges C, Troyer JL, Veldink JH, van den Berg LH, Spadoni JL, Taing L, Labib T, Montes M, Delfraissy JF, Schachter F, O'Brien SJ, Buchbinder S, van Natta ML, Jabs DA, Froguel P, Schuitemaker H, Winkler CA, Zagury JF. Multicohort genomewide association study reveals a new signal of protection against HIV-1 acquisition. J Infect Dis 2012; 205:1155-62. [PMID: 22362864 DOI: 10.1093/infdis/jis028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND To date, only mutations in CCR5 have been shown to confer resistance to human immunodeficiency virus type 1 (HIV-1) infection, and these explain only a small fraction of the observed variability in HIV susceptibility. METHODS We performed a meta-analysis between 2 independent European genomewide association studies, each comparing HIV-1 seropositive cases with normal population controls known to be HIV uninfected, to identify single-nucleotide polymorphisms (SNPs) associated with the HIV-1 acquisition phenotype. SNPs exhibiting P < 10(-5) in this first stage underwent second-stage analysis in 2 independent US cohorts of European descent. RESULTS After the first stage, a single highly significant association was revealed for the chromosome 8 rs6996198 with HIV-1 acquisition and was replicated in both second-stage cohorts. Across the 4 groups, the rs6996198-T allele was consistently associated with a significant reduced risk of HIV-1 infection, and the global meta-analysis reached genomewide significance: P(combined) = 7.76 × 10(-8). CONCLUSIONS We provide strong evidence of association for a common variant with HIV-1 acquisition in populations of European ancestry. This protective signal against HIV-1 infection is the first identified outside the CCR5 nexus. First clues point to a potential functional role for a nearby candidate gene, CYP7B1, but this locus warrants further investigation.
Collapse
Affiliation(s)
- Sophie Limou
- Laboratoire Génomique, Bioinformatique, et Applications, EA4627, Chaire de Bioinformatique, Conservatoire National des Arts et Métiers, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
863
|
|
864
|
Mandalia S, Westrop SJ, Beck EJ, Nelson M, Gazzard BG, Imami N. Are long-term non-progressors very slow progressors? Insights from the Chelsea and Westminster HIV cohort, 1988-2010. PLoS One 2012; 7:e29844. [PMID: 22363409 PMCID: PMC3282685 DOI: 10.1371/journal.pone.0029844] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 12/06/2011] [Indexed: 02/05/2023] Open
Abstract
Define and identify long-term non-progressors (LTNP) and HIV controllers (HIC), and estimate time until disease progression. LTNP are HIV-1+ patients who maintain stable CD4+ T-cell counts, with no history of opportunistic infection or antiretroviral therapy (ART). HIC are a subset of LTNP who additionally have undetectable viraemia. These individuals may provide insights for prophylactic and therapeutic development. Records of HIV-1+ individuals attending Chelsea and Westminster Hospital (1988–2010), were analysed. LTNP were defined as: HIV-1+ for >7 years; ART-naïve; no history of opportunistic infection and normal, stable CD4+ T-cell counts. MIXED procedure in SAS using random intercept model identified long-term stable CD4+ T-cell counts. Survival analysis estimated time since diagnosis until disease progression. Subjects exhibiting long-term stable CD4+ T-cell counts with history below the normal range (<450 cells/µl blood) were compared to LTNP whose CD4+ T-cell count always remained normal. Within these two groups subjects with HIV-1 RNA load below limit of detection (BLD) were identified. Of 14,227 patients, 1,204 were diagnosed HIV-1+ over 7 years ago and were ART-naïve. Estimated time until disease progression for the 20% (239) whose CD4+ T-cell counts remained within the normal range, was 6.2 years (IQR: 2.0 to 9.6); significantly longer than 4.0 years (IQR: 1.0 to 7.3) for patients with historical CD4+ T-cell count below normal (Logrank chi-squared = 21.26; p<0.001). Within a subpopulation of 312 asymptomatic patients, 50 exhibited long-term stable CD4+ T-cell counts. Of these, 13 were LTNP, one of whom met HIC criteria. Of the remaining 37 patients with long-term stable low CD4+ T-cell counts, 3 controlled HIV-1 RNA load BLD. Individuals with stable, normal CD4+ T-cell counts progressed less rapidly than those with low CD4+ T-cell counts. Few LTNP and HIC identified in this and other studies, endorse the need for universal definitions to facilitate comparison.
Collapse
Affiliation(s)
- Sundhiya Mandalia
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Samantha J. Westrop
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Eduard J. Beck
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Mark Nelson
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Brian G. Gazzard
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
| | - Nesrina Imami
- Department of Medicine, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
- * E-mail:
| |
Collapse
|
865
|
Fadista J, Bendixen C. Genomic position mapping discrepancies of commercial SNP chips. PLoS One 2012; 7:e31025. [PMID: 22363540 PMCID: PMC3281913 DOI: 10.1371/journal.pone.0031025] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 12/30/2011] [Indexed: 11/23/2022] Open
Abstract
The field of genetics has come to rely heavily on commercial genotyping arrays and accompanying annotations for insights into genotype-phenotype associations. However, in order to avoid errors and false leads, it is imperative that the annotation of SNP chromosomal positions is accurate and unambiguous. We report on genomic positional discrepancies of various SNP chips for human, cattle and mouse species, and discuss their causes and consequences.
Collapse
Affiliation(s)
- João Fadista
- Department of Clinical Sciences Malmö, CRC, Lund University, Malmö, Sweden
| | - Christian Bendixen
- Department of Molecular Biology and Genetics, Faculty of Science and Technology, Aarhus University, Tjele, Denmark
- * E-mail:
| |
Collapse
|
866
|
Abstract
HIV elite controllers (EC) are a rare group of HIV-infected patients who are able to maintain undetectable viral loads during a long period of time in the absence of antiretroviral treatment. Adaptive immunity and host genetic factors, although implicated, do not entirely explain this phenomenon. On the other hand, plasmacytoid dendritic cells (pDCs) are the principal type I interferon (IFN) producers in response to viral infection, and it is unknown whether pDCs are involved in the control of HIV infection in EC. In our study, we analyzed peripheral pDC levels and IFN-α production by peripheral blood mononuclear cells (PBMCs) in EC compared to other groups of HIV-infected patients, the ability of pDCs to reduce HIV production in vitro, and the mechanisms potentially involved. We showed preserved pDC counts and IFN-α production in EC. We also observed a higher capacity of pDCs from EC to reduce HIV production and to induce T cell apoptosis, whereas pDCs from viremic patients barely responded without previous Toll-like receptor 9 (TLR-9) stimulus. The preserved functionality of pDCs from EC to reduce viral production may be one of the mechanisms involved in the control of HIV viremia in these subjects. These results demonstrate the importance of innate immunity in HIV pathogenesis, and an understanding of pDC mechanisms would be helpful for the design of new therapies.
Collapse
|
867
|
|
868
|
Chen H, Hayashi G, Lai OY, Dilthey A, Kuebler PJ, Wong TV, Martin MP, Fernandez Vina MA, McVean G, Wabl M, Leslie KS, Maurer T, Martin JN, Deeks SG, Carrington M, Bowcock AM, Nixon DF, Liao W. Psoriasis patients are enriched for genetic variants that protect against HIV-1 disease. PLoS Genet 2012; 8:e1002514. [PMID: 22577363 PMCID: PMC3343879 DOI: 10.1371/journal.pgen.1002514] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 12/11/2011] [Indexed: 02/08/2023] Open
Abstract
An important paradigm in evolutionary genetics is that of a delicate balance between genetic variants that favorably boost host control of infection but which may unfavorably increase susceptibility to autoimmune disease. Here, we investigated whether patients with psoriasis, a common immune-mediated disease of the skin, are enriched for genetic variants that limit the ability of HIV-1 virus to replicate after infection. We analyzed the HLA class I and class II alleles of 1,727 Caucasian psoriasis cases and 3,581 controls and found that psoriasis patients are significantly more likely than controls to have gene variants that are protective against HIV-1 disease. This includes several HLA class I alleles associated with HIV-1 control; amino acid residues at HLA-B positions 67, 70, and 97 that mediate HIV-1 peptide binding; and the deletion polymorphism rs67384697 associated with high surface expression of HLA-C. We also found that the compound genotype KIR3DS1 plus HLA-B Bw4-80I, which respectively encode a natural killer cell activating receptor and its putative ligand, significantly increased psoriasis susceptibility. This compound genotype has also been associated with delay of progression to AIDS. Together, our results suggest that genetic variants that contribute to anti-viral immunity may predispose to the development of psoriasis.
Collapse
Affiliation(s)
- Haoyan Chen
- Department of Dermatology, University of California San Francisco, San Francisco, California, United States of America
| | - Genki Hayashi
- Department of Dermatology, University of California San Francisco, San Francisco, California, United States of America
| | - Olivia Y. Lai
- Department of Dermatology, University of California San Francisco, San Francisco, California, United States of America
| | - Alexander Dilthey
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Peter J. Kuebler
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Tami V. Wong
- Department of Dermatology, University of California San Francisco, San Francisco, California, United States of America
| | - Maureen P. Martin
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, National Cancer Institute, Frederick, Maryland, United States of America
| | | | - Gil McVean
- Department of Statistics, University of Oxford, Oxford, United Kingdom
| | - Matthias Wabl
- Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California, United States of America
| | - Kieron S. Leslie
- Department of Dermatology, University of California San Francisco, San Francisco, California, United States of America
| | - Toby Maurer
- Department of Dermatology, University of California San Francisco, San Francisco, California, United States of America
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, United States of America
| | - Steven G. Deeks
- HIV/AIDS Program, Department of Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Mary Carrington
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, National Cancer Institute, Frederick, Maryland, United States of America
| | - Anne M. Bowcock
- Division of Human Genetics, Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Douglas F. Nixon
- Division of Experimental Medicine, University of California San Francisco, San Francisco, California, United States of America
| | - Wilson Liao
- Department of Dermatology, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
869
|
Abstract
PURPOSE OF REVIEW The goal of this study is to review key recent findings related to the immunopathogenesis of hepatitis C virus (HCV) infection, especially in regards to T lymphocytes. It aims to complement other reviews in this issue on the roles of host genetics (IL-28B), acute HCV infection (when disease outcome is determined) and other factors that may influence fibrosis progression (microbial translocation). The main focus is on specific immunity and T cells in the context of success and failure to control viral infection. RECENT FINDINGS This review focuses on two areas of intense interest in the recent literature: the relationship between the human leukocyte antigen (HLA), class I-restricted T-cell responses and the evolution of the virus and the role of inhibitory markers on T cells in the immunopathogenesis of HCV. When appropriate, we compare findings from studies of HIV-specific immunity. SUMMARY From examining the virus and the mutational changes associated with T-cell responses and from analyzing the markers on T cells, there have been numerous advances in the understanding of immune evasion mechanisms employed by HCV.
Collapse
|
870
|
Huang J, Al-Mozaini M, Rogich J, Carrington MF, Seiss K, Pereyra F, Lichterfeld M, Yu XG. Systemic inhibition of myeloid dendritic cells by circulating HLA class I molecules in HIV-1 infection. Retrovirology 2012; 9:11. [PMID: 22289474 PMCID: PMC3308926 DOI: 10.1186/1742-4690-9-11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 01/30/2012] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND HIV-1 infection is associated with profound dysfunction of myeloid dendritic cells, for reasons that remain ill-defined. Soluble HLA class I molecules can have important inhibitory effects on T cells and NK cells, but may also contribute to reduced functional properties of professional antigen-presenting cells. Here, we investigated the expression of soluble HLA class I isoforms during HIV-1 infection and assessed their functional impact on antigen-presenting characteristics of dendritic cells. RESULTS Soluble HLA class I molecules were highly upregulated in progressive HIV-1 infection as determined by quantitative Western blots. This was associated with strong increases of intracellular expression of HLA class I isoforms in dendritic cells and monocytes. Using mixed lymphocyte reactions, we found that soluble HLA class I molecules effectively inhibited the antigen-presenting properties of dendritic cells, however, there was no significant influence of HLA class I molecules on the cytokine-secretion properties of these cells. The immunomodulatory effects of soluble HLA class I molecules were mediated by interactions with inhibitory myelomonocytic MHC class I receptors from the Leukocyte Immunoglobulin Like Receptor (LILR) family. CONCLUSIONS During progressive HIV-1 infection, soluble HLA class I molecules can contribute to systemic immune dysfunction by inhibiting the antigen-presenting properties of myeloid dendritic cells through interactions with inhibitory myelomonocytic HLA class I receptors.
Collapse
Affiliation(s)
- Jinghe Huang
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
871
|
Raychaudhuri S, Sandor C, Stahl EA, Freudenberg J, Lee HS, Jia X, Alfredsson L, Padyukov L, Klareskog L, Worthington J, Siminovitch KA, Bae SC, Plenge RM, Gregersen PK, de Bakker PI. Five amino acids in three HLA proteins explain most of the association between MHC and seropositive rheumatoid arthritis. Nat Genet 2012; 44:291-6. [PMID: 22286218 PMCID: PMC3288335 DOI: 10.1038/ng.1076] [Citation(s) in RCA: 694] [Impact Index Per Article: 53.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Accepted: 12/12/2011] [Indexed: 12/16/2022]
Abstract
The genetic association of the major histocompatibility complex (MHC) to rheumatoid arthritis risk has commonly been attributed to alleles in HLA-DRB1. However, debate persists about the identity of the causal variants in HLA-DRB1 and the presence of independent effects elsewhere in the MHC. Using existing genome-wide SNP data in 5,018 individuals with seropositive rheumatoid arthritis (cases) and 14,974 unaffected controls, we imputed and tested classical alleles and amino acid polymorphisms in HLA-A, HLA-B, HLA-C, HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA-DQB1 and HLA-DRB1, as well as 3,117 SNPs across the MHC. Conditional and haplotype analyses identified that three amino acid positions (11, 71 and 74) in HLA-DRβ1 and single-amino-acid polymorphisms in HLA-B (at position 9) and HLA-DPβ1 (at position 9), which are all located in peptide-binding grooves, almost completely explain the MHC association to rheumatoid arthritis risk. This study shows how imputation of functional variation from large reference panels can help fine map association signals in the MHC.
Collapse
Affiliation(s)
- Soumya Raychaudhuri
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Partners HealthCare Center for Personalized Genetic Medicine, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Cynthia Sandor
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Partners HealthCare Center for Personalized Genetic Medicine, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Eli A. Stahl
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jan Freudenberg
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, North Shore–Long Island Jewish Health System, Manhasset, NY USA
| | - Hye-Soon Lee
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Xiaoming Jia
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Boston, MA
| | - Lars Alfredsson
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Leonid Padyukov
- Rheumatology Unit, Department of Medicine, Karolinska Institutet at Karolinska University Hospital Solna, Stockholm, Sweden
| | - Lars Klareskog
- Rheumatology Unit, Department of Medicine, Karolinska Institutet at Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jane Worthington
- Arthritis Research UK Epidemiology Unit, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK
| | - Katherine A. Siminovitch
- Department of Medicine, University of Toronto, Mount Sinai Hospital and University Health Network, Toronto, Ontario, Canada
| | - Sang-Cheol Bae
- Department of Rheumatology, Hanyang University Hospital for Rheumatic Diseases, Seoul, South Korea
| | - Robert M. Plenge
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Peter K. Gregersen
- Robert S. Boas Center for Genomics and Human Genetics, Feinstein Institute for Medical Research, North Shore–Long Island Jewish Health System, Manhasset, NY USA
| | - Paul I.W. de Bakker
- Division of Genetics, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
872
|
Guergnon J, Dalmasso C, Broet P, Meyer L, Westrop SJ, Imami N, Vicenzi E, Morsica G, Tinelli M, Zanone Poma B, Goujard C, Potard V, Gotch FM, Casoli C, Cossarizza A, Macciardi F, Debré P, Delfraissy JF, Galli M, Autran B, Costagliola D, Poli G, Theodorou I, Riva A. Single-nucleotide polymorphism-defined class I and class III major histocompatibility complex genetic subregions contribute to natural long-term nonprogression in HIV infection. J Infect Dis 2012; 205:718-24. [PMID: 22238471 DOI: 10.1093/infdis/jir833] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
We performed a genome-wide association study comparing a cohort of 144 human immunodeficiency virus (HIV type 1-infected, untreated white long-term nonprogressors (LTNPs) with a cohort of 605 HIV-1-infected white seroconverters. Forty-seven single-nucleotide polymorphisms (SNPs), located from class I to class III major histocompatibility complex (MHC) subregions, show statistical association (false discovery rate, <0.05) with the LTNP condition, among which 5 reached genome-wide significance after Bonferonni correction. The MHC LTNP-associated SNPs are ordered in ≥4 linkage disequilibrium blocks; interestingly, an MHC class III linkage disequilibrium block (defined by the rs9368699 SNP) seems specific to the LTNP phenotype.
Collapse
|
873
|
Blacklaws BA. Small ruminant lentiviruses: immunopathogenesis of visna-maedi and caprine arthritis and encephalitis virus. Comp Immunol Microbiol Infect Dis 2012; 35:259-69. [PMID: 22237012 DOI: 10.1016/j.cimid.2011.12.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 12/08/2011] [Accepted: 12/09/2011] [Indexed: 10/14/2022]
Abstract
The small ruminant lentiviruses include the prototype for the genus, visna-maedi virus (VMV) as well as caprine arthritis encephalitis virus (CAEV). Infection of sheep or goats with these viruses causes slow, progressive, inflammatory pathology in many tissues, but the most common clinical signs result from pathology in the lung, mammary gland, central nervous system and joints. This review examines replication, immunity to and pathogenesis of these viruses and highlights major differences from and similarities to some of the other lentiviruses.
Collapse
Affiliation(s)
- Barbara A Blacklaws
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK.
| |
Collapse
|
874
|
Ballana E, Ruiz-de Andres A, Mothe B, Ramirez de Arellano E, Aguilar F, Badia R, Grau E, Clotet B, del Val M, Brander C, Esté JA. Differential prevalence of the HLA-C -- 35 CC genotype among viremic long term non-progressor and elite controller HIV+ individuals. Immunobiology 2012; 217:889-94. [PMID: 22333575 DOI: 10.1016/j.imbio.2011.12.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 12/27/2011] [Accepted: 12/30/2011] [Indexed: 01/11/2023]
Abstract
Susceptibility to HIV infection and disease progression are complex traits modulated by environmental and genetic factors, affecting innate and adaptive immune responses, among other cellular processes. A single nucleotide polymorphism (SNP) 35 kb upstream of the HLA-C gene locus (-35C/T) was previously shown to correlate with increased HLA-C expression and improved control of HIV-1. Here, we genotyped the -35C/T SNP in 639 subjects (180 uninfected patients, 304 HIV progressors and 155 LTNP) and confirmed the association of the -35C/T variant with the LTNP phenotype. The genotype frequencies in the general population subjects did not differ significantly from those seen in HIV progressors (p-value=0.472). However, a significant higher frequency of the protective CC genotype was identified when LTNP were compared either with HIV progressors alone (p-value<0.0001) or progressors and uninfected subjects together (p-value<0.0001). When considering aviremic LTNP alone (elite controllers; viral load below 50 copies/ml), the -35 CC genotype was not overrepresented compared to HIV progressors. Conversely, a significant association was found with the viremic LTNP groups (viral loads below 10,000 copies/ml). These results suggest that other factors alone or in combination with the -35 CC genotype may play an important role in differentiating the elite controller status from LTNP. Combination of different genetic variants may have additive or epistatic effects determining the HIV course of infection.
Collapse
Affiliation(s)
- Ester Ballana
- IrsiCaixa-HIVACAT, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
875
|
Raghavan S, Alagarasu K, Selvaraj P. Immunogenetics of HIV and HIV associated tuberculosis. Tuberculosis (Edinb) 2012; 92:18-30. [PMID: 21943869 DOI: 10.1016/j.tube.2011.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 08/08/2011] [Indexed: 11/28/2022]
Affiliation(s)
- S Raghavan
- Department of Immunology, Tuberculosis Research Centre (ICMR), Mayor V.R. Ramanathan Road, Chetput, Chennai 600031, India
| | | | | |
Collapse
|
876
|
|
877
|
Abstract
Host genetic variation is presently estimated to account for about one-fourth of the observed differences in control of HIV across infected individuals. Genome-wide association studies have confirmed that polymorphism within the HLA class I locus is the primary host genetic contributor to determining outcome after infection. Here we progress beyond the genetic associations alone to consider the functional explanations for these correlations. In this process, the complex and multidimensional effects of HLA molecules in viral disease become apparent.
Collapse
Affiliation(s)
- Mary Carrington
- Cancer and Inflammation Program, Laboratory of Experimental Immunology, SAIC–Frederick, Inc., NCI–Frederick, Frederick, Maryland 20882;
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114;
| | - Bruce D. Walker
- Ragon Institute of MGH, MIT and Harvard, Boston, Massachusetts 02114;
| |
Collapse
|
878
|
Quakkelaar ED, Melief CJM. Experience with synthetic vaccines for cancer and persistent virus infections in nonhuman primates and patients. Adv Immunol 2012; 114:77-106. [PMID: 22449779 DOI: 10.1016/b978-0-12-396548-6.00004-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Synthetic vaccines, in particular long synthetic peptides of approximately 25-50 amino acids in length, are attractive for HIV vaccine development and for induction of therapeutic immune responses in patients with (pre-)malignant disorders. In the case of preventive vaccine development against HIV, no major success has been achieved, but the possibilities are by no means exhausted. A long peptide vaccine consisting of 13 overlapping peptides, which together cover the entire length of the two oncogenic proteins E6 and E7 of high-risk human papilloma virus type 16 (HPV16), caused complete regression of all lesions and eradication of virus in 9 out of 20 women with high-grade vulvar intraepithelial neoplasia, a therapy-resistant preneoplastic disorder. The nature and strength of the vaccine-prompted T cell responses were significantly correlated with the clinical response. Synthetic peptide vaccines are attractive, because they allow rational improvement of vaccine design and detailed pharmacokinetic and pharmacodynamic studies not possible with conventional vaccines. Improvements are possible by addition or conjugation of adjuvants, notably TLR ligands, to the synthetic peptides.
Collapse
Affiliation(s)
- Esther D Quakkelaar
- Department of Immunohematology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
879
|
Abstract
RNA viruses, such as human immunodeficiency virus, hepatitis C virus, influenza virus, and poliovirus replicate with very high mutation rates and exhibit very high genetic diversity. The extremely high genetic diversity of RNA virus populations originates that they replicate as complex mutant spectra known as viral quasispecies. The quasispecies dynamics of RNA viruses are closely related to viral pathogenesis and disease, and antiviral treatment strategies. Over the past several decades, the quasispecies concept has been expanded to provide an adequate framework to explain complex behavior of RNA virus populations. Recently, the quasispecies concept has been used to study other complex biological systems, such as tumor cells, bacteria, and prions. Here, we focus on some questions regarding viral and theoretical quasispecies concepts, as well as more practical aspects connected to pathogenesis and resistance to antiviral treatments. A better knowledge of virus diversification and evolution may be critical in preventing and treating the spread of pathogenic viruses.
Collapse
|
880
|
Kloverpris HN, Stryhn A, Harndahl M, van der Stok M, Payne RP, Matthews PC, Chen F, Riddell L, Walker BD, Ndung'u T, Buus S, Goulder P. HLA-B*57 Micropolymorphism shapes HLA allele-specific epitope immunogenicity, selection pressure, and HIV immune control. J Virol 2012; 86:919-29. [PMID: 22090105 PMCID: PMC3255844 DOI: 10.1128/jvi.06150-11] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The genetic polymorphism that has the greatest impact on immune control of human immunodeficiency virus (HIV) infection is expression of HLA-B*57. Understanding of the mechanism for this strong effect remains incomplete. HLA-B*57 alleles and the closely related HLA-B*5801 are often grouped together because of their similar peptide-binding motifs and HIV disease outcome associations. However, we show here that the apparently small differences between HLA-B*57 alleles, termed HLA-B*57 micropolymorphisms, have a significant impact on immune control of HIV. In a study cohort of >2,000 HIV C-clade-infected subjects from southern Africa, HLA-B*5703 is associated with a lower viral-load set point than HLA-B*5702 and HLA-B*5801 (medians, 5,980, 15,190, and 19,000 HIV copies/ml plasma; P = 0.24 and P = 0.0005). In order to better understand these observed differences in HLA-B*57/5801-mediated immune control of HIV, we undertook, in a study of >1,000 C-clade-infected subjects, a comprehensive analysis of the epitopes presented by these 3 alleles and of the selection pressure imposed on HIV by each response. In contrast to previous studies, we show that each of these three HLA alleles is characterized both by unique CD8(+) T-cell specificities and by clear-cut differences in selection pressure imposed on the virus by those responses. These studies comprehensively define for the first time the CD8(+) T-cell responses and immune selection pressures for which these protective alleles are responsible. These findings are consistent with HLA class I alleles mediating effective immune control of HIV through the number of p24 Gag-specific CD8(+) T-cell responses generated that can drive significant selection pressure on the virus.
Collapse
Affiliation(s)
- Henrik N Kloverpris
- Department of Paediatrics, University of Oxford, Peter Medawar Building, Oxford, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
881
|
Paris R, Bejrachandra S, Thongcharoen P, Nitayaphan S, Pitisuttithum P, Sambor A, Gurunathan S, Francis D, Ratto-Kim S, Karnasuta C, de Souza MS, Polonis VR, Brown AE, Kim JH, Stephens HA. HLA class II restriction of HIV-1 clade-specific neutralizing antibody responses in ethnic Thai recipients of the RV144 prime-boost vaccine combination of ALVAC-HIV and AIDSVAX® B/E. Vaccine 2012; 30:832-6. [DOI: 10.1016/j.vaccine.2011.11.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 05/12/2011] [Accepted: 11/01/2011] [Indexed: 01/24/2023]
|
882
|
Bagasra O, Sheraz M, Pace DG. Hepatitis G Virus or GBV-C: A Natural Anti-HIV Interfering Virus. VIRUSES: ESSENTIAL AGENTS OF LIFE 2012:363-388. [DOI: 10.1007/978-94-007-4899-6_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
883
|
Ahn S, Choi HB, Kim TG. HLA and Disease Associations in Koreans. Immune Netw 2011; 11:324-35. [PMID: 22346771 PMCID: PMC3275700 DOI: 10.4110/in.2011.11.6.324] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 10/13/2011] [Accepted: 10/19/2011] [Indexed: 01/07/2023] Open
Abstract
The human leukocyte antigen (HLA), the major histocompatibility complex (MHC) in humans has been known to reside on chromosome 6 and encodes cell-surface antigen-presenting proteins and many other proteins related to immune system function. The HLA is highly polymorphic and the most genetically variable coding loci in humans. In addition to a critical role in transplantation medicine, HLA and disease associations have been widely studied across the populations world-wide and are found to be important in prediction of disease susceptibility, resistance and of evolutionary maintenance of genetic diversity. Because recently developed molecular based HLA typing has several advantages like improved specimen stability and increased resolution of HLA types, the association between HLA alleles and a given disease could be more accurately quantified. Here, in this review, we have collected HLA association data on some autoimmune diseases, infectious diseases, cancers, drug responsiveness and other diseases with unknown etiology in Koreans and attempt to summarize some remarkable HLA alleles related with specific diseases.
Collapse
Affiliation(s)
- Stephen Ahn
- Department of Microbiology and Immunology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | |
Collapse
|
884
|
Vollbrecht T, Eberle J, Roider J, Bühler S, Stirner R, Henrich N, Seybold U, Bogner JR, Draenert R. Control of M184V HIV-1 mutants by CD8 T-cell responses. Med Microbiol Immunol 2011; 201:201-11. [PMID: 22200907 DOI: 10.1007/s00430-011-0222-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2011] [Indexed: 01/10/2023]
Abstract
Antiretroviral treatment directed against HIV is highly effective, yet limited by drug resistance mutations. We hypothesized that CD8 T cells targeting drug-resistant HIV mutants are able to inhibit viral replication in the setting of a failing therapeutic regimen. We evaluated CD8 T-cell responses and mapped epitopes in HIV-infected patients by interferon-gamma Elispot and intracellular cytokine staining. Autologous virus was sequenced by RT-PCR. Viral replication inhibition assays were performed using M184V mutant virus and CD8 T cell lines. CD8 T-cell responses toward the regions of viral drug resistance mutations in Pol are frequent. Focusing on the M184V mutation, A*02:01-YQYVDDLYV and A*02:01-VIYQYVDDLYV were identified as optimal epitopes for the majority of study subjects. Viral replication of M184V HIV mutants was inhibited by CD8 T cell lines in vitro. In case of a failing lamivudine/emtricitabine containing regimen, individuals with a CD8 T-cell response toward M184V had a significant lower viral load than those without a CD8 response (p = 0.005). Two study subjects even achieved an undetectable viral load. Our data suggest that control of M184V mutant virus by CD8 T-cell responses is possible in vitro and in vivo. This control has important implications for therapeutic vaccination strategies.
Collapse
|
885
|
Yu XQ, Li M, Zhang H, Low HQ, Wei X, Wang JQ, Sun LD, Sim KS, Li Y, Foo JN, Wang W, Li ZJ, Yin XY, Tang XQ, Fan L, Chen J, Li RS, Wan JX, Liu ZS, Lou TQ, Zhu L, Huang XJ, Zhang XJ, Liu ZH, Liu JJ. A genome-wide association study in Han Chinese identifies multiple susceptibility loci for IgA nephropathy. Nat Genet 2011; 44:178-82. [PMID: 22197929 DOI: 10.1038/ng.1047] [Citation(s) in RCA: 234] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 12/01/2011] [Indexed: 12/18/2022]
Abstract
We performed a two-stage genome-wide association study of IgA nephropathy (IgAN) in Han Chinese, with 1,434 affected individuals (cases) and 4,270 controls in the discovery phase and follow-up of the top 61 SNPs in an additional 2,703 cases and 3,464 controls. We identified associations at 17p13 (rs3803800, P = 9.40 × 10(-11), OR = 1.21; rs4227, P = 4.31 × 10(-10), OR = 1.23) and 8p23 (rs2738048, P = 3.18 × 10(-14), OR = 0.79) that implicated the genes encoding tumor necrosis factor (TNFSF13) and α-defensin (DEFA) as susceptibility genes. In addition, we found multiple associations in the major histocompatibility complex (MHC) region (rs660895, P = 4.13 × 10(-20), OR = 1.34; rs1794275, P = 3.43 × 10(-13), OR = 1.30; rs2523946, P = 1.74 × 10(-11), OR = 1.21) and confirmed a previously reported association at 22q12 (rs12537, P = 1.17 × 10(-11), OR = 0.78). We also found that rs660895 was associated with clinical subtypes of IgAN (P = 0.003), proteinuria (P = 0.025) and IgA levels (P = 0.047). Our findings show that IgAN is associated with variants near genes involved in innate immunity and inflammation.
Collapse
Affiliation(s)
- Xue-Qing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
886
|
Abstract
Vaccines are the most effective tools to prevent infectious diseases and to minimize their impact on humans or animals. Despite the successful development of vaccines that are able to elicit potent and protective immune responses, the majority of vaccines have been so far developed empirically and mechanistic events leading to protective immune responses are often poorly understood. This hampers the development of new prophylactic as well as therapeutic vaccines for infectious diseases and cancer. Biological correlates of immune‐mediated protection are currently based on standard readout such as antibody titres and ELISPOT assays. The development of successful vaccines for difficult settings, such as infectious agents leading to chronic infection (HIV, HCV. . .) or cancer, calls for novel ‘readout systems’ or ‘correlates’ of immune‐mediated protection that would reliably predict immune responses to novel vaccines in vivo. Systems biology offers a new approach to vaccine design that is based upon understanding the molecular network mobilized by vaccination. Systems vaccinology approaches investigate more global correlates of successful vaccination, beyond the specific immune response to the antigens administered, providing new methods for measuring early vaccine efficacy and ultimately generating hypotheses for understanding the mechanisms that underlie successful immunogenicity. Using functional genomics, specific molecular signatures of individual vaccine can be identified and used as predictors of vaccination efficiency. The immune response to vaccination involves the coordinated induction of master transcription factors that leads to the development of a broad, polyfunctional and persistent immune response integrating all effector cells of the immune systems.
Collapse
Affiliation(s)
- Adrien Six
- UPMC Univ Paris 06, UMR 7211, F-75013 Paris, France.
| | | | | | | |
Collapse
|
887
|
Deepe RN, Kistner-Griffin E, Martin JN, Deeks SG, Pandey JP. Epistatic interactions between Fc (GM) and FcγR genes and the host control of human immunodeficiency virus replication. Hum Immunol 2011; 73:263-6. [PMID: 22213007 DOI: 10.1016/j.humimm.2011.12.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 11/21/2011] [Accepted: 12/06/2011] [Indexed: 10/14/2022]
Abstract
Host genetic factors are thought to contribute to the interindividual differences in the control of human immunodeficiency virus (HIV) replication. The aim of the present investigation was to determine whether genes encoding GM and KM allotypes-genetic markers of immunoglobulin γ and κ chains, respectively-and those encoding Fcγ receptor (FcγR) IIa and IIIa are associated with the host control of HIV replication. A case-control design was employed among HIV-infected subjects, with a group that spontaneously controlled HIV replication ("controllers") as cases (n = 73) and those who did not control replication as controls (n = 100). Genotyping was performed by polymerase chain reaction-restriction fragment length polymorphism, direct DNA sequencing, and TaqMan genotyping assays. In Caucasian Americans, certain combinations of FcγR and GM genotypes were differentially distributed between controllers and noncontrollers. Among the noncarriers of the FcγRIIa arginine allele, GM21 noncarriers had over 7-fold greater odds of being controllers than the carriers of this allele (odds ratio [OR] = 7.47). These GM determinants also interacted with FcγRIIIa alleles. Among the carriers of the FcγRIIIa valine allele, GM21 noncarriers had over 3-fold greater odds of being controllers than the carriers of this allele (OR = 3.26). These results demonstrate epistatic interactions of genes on chromosomes 14 (GM) and 1 (FcγR) in influencing the control of HIV replication.
Collapse
Affiliation(s)
- Raymond N Deepe
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | | | | | |
Collapse
|
888
|
Sui Y, Gagnon S, Dzutsev A, Zhu Q, Yu H, Hogg A, Wang Y, Xia Z, Belyakov IM, Venzon D, Klinman D, Strober W, Kelsall B, Franchini G, Berzofsky JA. TLR agonists and/or IL-15 adjuvanted mucosal SIV vaccine reduced gut CD4⁺ memory T cell loss in SIVmac251-challenged rhesus macaques. Vaccine 2011; 30:59-68. [PMID: 22041305 PMCID: PMC3258186 DOI: 10.1016/j.vaccine.2011.10.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Accepted: 10/18/2011] [Indexed: 01/08/2023]
Abstract
Adjuvant plays an important role in increasing and directing vaccine-induced immune responses. In a previous study, we found that a mucosal SIV vaccine using a combination of IL-15 and TLR agonists as adjuvant mediated partial protection against SIVmac251 rectal challenge, whereas neither IL-15 nor TLR agonists alone as an adjuvant impacted the plasma viral loads. In this study, dissociation of CD4(+) T cell preservation with viral loads was observed in the animals vaccinated with adjuvants. Significantly higher levels of memory CD4(+) T cell numbers were preserved after SIVmac251 infection in the colons of the animals vaccinated with vaccine containing any of these adjuvants compared to no adjuvant. When we measured the viral-specific CD8(+) tetramer responses in the colon lamina propria, we found significantly higher levels of gag, tat, and pol epitope tetramer(+) T cell responses in these animals compared to ones without adjuvant, even if some of the animals had similarly high viral loads. Furthermore, this CD4(+) T preservation was positively correlated with increased levels of gag and Tat, but not pol tetramer(+) T cell responses, and inversely correlated with beta-chemokine expression. The pre-challenged APOBEC3G expression level, which has previously been shown inversely associated with viral loads, was further found positively correlated with CD4(+) T cell number preservation. Overall, these data highlight one unrecognized role of adjuvant in HIV vaccine development, and show that vaccines can produce a surprising discordance between CD4(+) T cell levels and SIV viral load.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Susan Gagnon
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Amiran Dzutsev
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Qing Zhu
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Huifeng Yu
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Alison Hogg
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Yichuan Wang
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Zheng Xia
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Igor M. Belyakov
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - David Venzon
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| | - Dennis Klinman
- Laboratory of Experimental Immunology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Warren Strober
- Laboratory of Host Defenses, National Institutes of Health, Bethesda, MD 20892
| | - Brian Kelsall
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD 20892
| | | | - Jay A. Berzofsky
- Vaccine Branch, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
889
|
Salgado M, Rabi SA, O'Connell KA, Buckheit RW, Bailey JR, Chaudhry AA, Breaud AR, Marzinke MA, Clarke W, Margolick JB, Siliciano RF, Blankson JN. Prolonged control of replication-competent dual- tropic human immunodeficiency virus-1 following cessation of highly active antiretroviral therapy. Retrovirology 2011; 8:97. [PMID: 22141397 PMCID: PMC3293762 DOI: 10.1186/1742-4690-8-97] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Accepted: 12/05/2011] [Indexed: 01/01/2023] Open
Abstract
Background While initiation of highly active antiretroviral therapy (HAART) during primary HIV-1 infection occasionally results in transient control of viral replication after treatment interruption, the vast majority of patients eventually experience a rebound in plasma viremia. Results Here we report a case of a patient who was started on HAART during symptomatic primary infection and who has subsequently maintained viral loads of < 50 copies/mL for more than nine years after the cessation of treatment. This patient had a high baseline viral load and has maintained a relatively high frequency of latently infected CD4+ T cells. In addition, he does not have any known protective HLA alleles. Thus it is unlikely that he was destined to become a natural elite controller or suppressor. The mechanism of control of viral replication is unclear; he is infected with a CCR5/CXCR4 dual-tropic virus that is fully replication-competent in vitro. In addition, his spouse, who transmitted the virus to him, developed AIDS. The patient's CD4+ T cells are fully susceptible to HIV-1 infection, and he has low titers of neutralizing antibodies to heterologous and autologous HIV-1 isolates. Furthermore, his CD8+ T cells do not have potent HIV suppressive activity. Conclusion This report suggests that some patients may be capable of controlling pathogenic HIV-1 isolates for extended periods of time after the cessation of HAART through a mechanism that is distinct from the potent cytotoxic T lymphocyte (CTL) mediated suppression that has been reported in many elite suppressors.
Collapse
Affiliation(s)
- Maria Salgado
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
890
|
Abstract
Advances in genotyping and sequencing technologies have revolutionized the genetics of complex disease by locating rare and common variants that influence an individual's risk for diseases, such as diabetes, cancers, and psychiatric disorders. However, to capitalize on these data for prevention and therapies requires the identification of causal alleles and a mechanistic understanding for how these variants contribute to the disease. After discussing the strategies currently used to map variants for complex diseases, this Primer explores how variants may be prioritized for follow-up functional studies and the challenges and approaches for assessing the contributions of rare and common variants to disease phenotypes.
Collapse
|
891
|
Kaufman DR, Li F, Cruz AN, Self SG, Barouch DH. Focus and breadth of cellular immune responses elicited by a heterologous insert prime-boost vaccine regimen in rhesus monkeys. Vaccine 2011; 30:506-9. [PMID: 22133509 DOI: 10.1016/j.vaccine.2011.11.079] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 11/17/2011] [Accepted: 11/18/2011] [Indexed: 11/24/2022]
Abstract
The global sequence diversity of HIV-1 presents a daunting challenge for vaccine development. We investigated whether a heterologous insert prime-boost regimen could expand global coverage by selectively boosting cellular immune responses to conserved epitopes. Rhesus monkeys were primed and boosted with recombinant adenovirus vectors expressing homologous or heterologous HIV-1 Gag sequences that were optimized to focus responses on highly conserved epitopes. We observed comparable responses directed to specific regions of the Gag protein in all experimental groups without evidence of improved coverage or expanded breadth in the heterologous insert group. These data suggest that antigen-independent factors contribute to the immunodominance patterns of vaccine-elicited cellular immune responses.
Collapse
Affiliation(s)
- David R Kaufman
- Division of Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, United States
| | | | | | | | | |
Collapse
|
892
|
Wu Y, Gao F, Liu J, Qi J, Gostick E, Price DA, Gao GF. Structural Basis of Diverse Peptide Accommodation by the Rhesus Macaque MHC Class I Molecule Mamu-B*17: Insights into Immune Protection from Simian Immunodeficiency Virus. THE JOURNAL OF IMMUNOLOGY 2011; 187:6382-92. [DOI: 10.4049/jimmunol.1101726] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
893
|
Origin and plasticity of MHC I-associated self peptides. Autoimmun Rev 2011; 11:627-35. [PMID: 22100331 DOI: 10.1016/j.autrev.2011.11.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 11/02/2011] [Indexed: 01/13/2023]
Abstract
Endogenous peptides presented by MHC I molecules represent the essence of self for CD8 T lymphocytes. These MHC I peptides (MIPs) regulate all key events that occur during the lifetime of CD8 T cells. CD8 T cells are selected on self-MIPs, sustained by self-MIPs, and activated in the presence of self-MIPs. Recently, large-scale mass spectrometry studies have revealed that the self-MIP repertoire is more complex and plastic than previously anticipated. The composition of the self-MIP repertoire varies from one cell type to another and can be perturbed by cell-intrinsic and -extrinsic factors including dysregulation of cellular metabolism and infection. The complexity and plasticity of the self-MIP repertoire represent a major challenge for the maintenance of self tolerance and can have pervasive effects on the global functioning of the immune system.
Collapse
|
894
|
Abstract
Recently, genome-wide association studies have identified the major histocompatibility complex class I protein HLA-C as an important molecule that affects HIV disease progression. The association between HLA-C and HIV disease outcome was originally determined through a single nucleotide polymorphism (SNP) 35 kb upstream of the HLA-C locus. More recent work has focused on elucidating the functional significance of the -35 SNP, and several groups now have demonstrated HLA-C surface expression to be a key element in control of HIV viral load, with higher surface expression associating with slower disease progression. Most recently, control of HLA-C surface expression has been correlated with the presence of microRNA binding sites that affect HLA-C expression and control of HIV disease. This review highlights these results and explores the ways in which HLA-C surface expression could affect immune system function in the setting of HIV disease.
Collapse
Affiliation(s)
- Deanna A Kulpa
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | | |
Collapse
|
895
|
Augmented HIV-specific interferon-gamma responses, but impaired lymphoproliferation during interruption of antiretroviral treatment initiated in primary HIV infection. J Acquir Immune Defic Syndr 2011; 58:1-8. [PMID: 21637110 DOI: 10.1097/qai.0b013e318224d0c7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Antiretroviral therapy (ART) introduced during primary HIV infection followed by treatment interruption (TI) is postulated to enhance virologic control through induction of HIV-specific CD4 T cells, which foster virus-specific CD8+ T cells that suppress virus replication. This hypothesis was evaluated in 21 subjects enrolled in AIDS Clinical Trials Group 709, a substudy of AIDS Clinical Trials Group 371, which prospectively evaluated subjects who received ≥1 year of ART initiated in acute or recent HIV infection followed by TI. METHODS Lymphoproliferation was assessed by [methyl-H] thymidine incorporation and HIV-specific CD8+ T-cell interferon-gamma responses by enzyme-linked immunospot-forming assays. Virologic success was defined as sustained viral load <5000 copies per milliliter for 24 weeks after TI. RESULTS HIV-specific lymphoproliferative responses were detected at least once in 5 (24%) of 21 subjects, were generally transient, and were unrelated to HIV-specific interferon-gamma responses (P > 0.4). HIV-specific CD8+ interferon-gamma responses increased after 48 weeks of ART (P = 0.03), but failed to predict virologic success (P = 0.18). Compared with seronegative subjects, lymphoproliferation to Candida, cytomegalovirus, and alloantigens was similar in HIV-infected subjects during ART, but lower during TI (P ≤ 0.04). CONCLUSIONS HIV-specific CD8+ T-cell interferon-gamma responses expand during ART following primary HIV infection, but are not related to HIV-specific lymphoproliferative responses nor virologic success. Impaired non-HIV antigen-specific lymphoproliferation associated with TI suggests this strategy could be deleterious.
Collapse
|
896
|
Lichterfeld M, Yu XG. The emerging role of leukocyte immunoglobulin-like receptors (LILRs) in HIV-1 infection. J Leukoc Biol 2011; 91:27-33. [PMID: 22028331 DOI: 10.1189/jlb.0811442] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
LILRs represent a group of immunomodulatory molecules that regulate the functional properties of professional APCs and influence immune activation in a variety of disease contexts. Many members of the LILR family recognize peptide/MHC class I complexes as their physiological ligands, and increasing evidence suggests that such interactions are prominently influenced by polymorphisms in HLA class I alleles or sequence variations in the presented antigenic peptides. Emerging data show that LILRs are involved in multiple, different aspects of HIV-1 disease pathogenesis and may critically influence spontaneous HIV-1 disease progression. Here, we review recent progress in understanding the role of LILR during HIV-1 infection by focusing on the dynamic interplay between LILR and HLA class I molecules in determining HIV-1 disease progression, the effects of HIV-1 mutational escape on LILR-mediated immune recognition, the contribution of LILR to HIV-1-associated immune dysfunction, and the unique expression patterns of LILR on circulating myeloid DCs from elite controllers, a small subset of HIV-1-infected patients with natural control of HIV-1 replication. Obtaining a more complete understanding of LILR-mediated immune regulation during HIV-1 infection may ultimately allow for improved strategies to treat or prevent HIV-1-associated disease manifestations.
Collapse
Affiliation(s)
- Mathias Lichterfeld
- Infectious Disease Division, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
897
|
Varadarajan N, Julg B, Yamanaka YJ, Chen H, Ogunniyi AO, McAndrew E, Porter LC, Piechocka-Trocha A, Hill BJ, Douek DC, Pereyra F, Walker BD, Love JC. A high-throughput single-cell analysis of human CD8⁺ T cell functions reveals discordance for cytokine secretion and cytolysis. J Clin Invest 2011; 121:4322-31. [PMID: 21965332 DOI: 10.1172/jci58653] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 08/10/2011] [Indexed: 11/17/2022] Open
Abstract
CD8+ T cells are a key component of the adaptive immune response to viral infection. An inadequate CD8+ T cell response is thought to be partly responsible for the persistent chronic infection that arises following infection with HIV. It is therefore critical to identify ways to define what constitutes an adequate or inadequate response. IFN-γ production has been used as a measure of T cell function, but the relationship between cytokine production and the ability of a cell to lyse virus-infected cells is not clear. Moreover, the ability to assess multiple CD8+ T cell functions with single-cell resolution using freshly isolated blood samples, and subsequently to recover these cells for further functional analyses, has not been achieved. As described here, to address this need, we have developed a high-throughput, automated assay in 125-pl microwells to simultaneously evaluate the ability of thousands of individual CD8+ T cells from HIV-infected patients to mediate lysis and to produce cytokines. This concurrent, direct analysis enabled us to investigate the correlation between immediate cytotoxic activity and short-term cytokine secretion. The majority of in vivo primed, circulating HIV-specific CD8+ T cells were discordant for cytolysis and cytokine secretion, notably IFN-γ, when encountering cognate antigen presented on defined numbers of cells. Our approach should facilitate determination of signatures of functional variance among individual effector CD8+ T cells, including those from mucosal samples and those induced by vaccines.
Collapse
Affiliation(s)
- Navin Varadarajan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
898
|
An P, Li R, Wang JM, Yoshimura T, Takahashi M, Samudralal R, O'Brien SJ, Phair J, Goedert JJ, Kirk GD, Troyer JL, Sezgin E, Buchbinder SP, Donfield S, Nelson GW, Winkler CA. Role of exonic variation in chemokine receptor genes on AIDS: CCRL2 F167Y association with pneumocystis pneumonia. PLoS Genet 2011; 7:e1002328. [PMID: 22046140 PMCID: PMC3203199 DOI: 10.1371/journal.pgen.1002328] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Accepted: 08/03/2011] [Indexed: 02/04/2023] Open
Abstract
Chromosome 3p21-22 harbors two clusters of chemokine receptor genes, several of which serve as major or minor coreceptors of HIV-1. Although the genetic association of CCR5 and CCR2 variants with HIV-1 pathogenesis is well known, the role of variation in other nearby chemokine receptor genes remain unresolved. We genotyped exonic single nucleotide polymorphisms (SNPs) in chemokine receptor genes: CCR3, CCRL2, and CXCR6 (at 3p21) and CCR8 and CX3CR1 (at 3p22), the majority of which were non-synonymous. The individual SNPs were tested for their effects on disease progression and outcomes in five treatment-naïve HIV-1/AIDS natural history cohorts. In addition to the known CCR5 and CCR2 associations, significant associations were identified for CCR3, CCR8, and CCRL2 on progression to AIDS. A multivariate survival analysis pointed to a previously undetected association of a non-conservative amino acid change F167Y in CCRL2 with AIDS progression: 167F is associated with accelerated progression to AIDS (RH = 1.90, P = 0.002, corrected). Further analysis indicated that CCRL2-167F was specifically associated with more rapid development of pneumocystis pneumonia (PCP) (RH = 2.84, 95% CI 1.28-6.31) among four major AIDS-defining conditions. Considering the newly defined role of CCRL2 in lung dendritic cell trafficking, this atypical chemokine receptor may affect PCP through immune regulation and inducing inflammation.
Collapse
MESH Headings
- Acquired Immunodeficiency Syndrome/complications
- Chromosomes, Human, Pair 3/genetics
- Cohort Studies
- Disease Progression
- Exons
- Genetic Association Studies
- HEK293 Cells
- HIV-1
- Humans
- Linkage Disequilibrium
- Pneumonia, Pneumocystis/etiology
- Pneumonia, Pneumocystis/genetics
- Polymorphism, Single Nucleotide
- Receptors, CCR/chemistry
- Receptors, CCR/genetics
- Receptors, CCR3/genetics
- Receptors, CCR8/genetics
- Receptors, CXCR6
- Receptors, Chemokine/genetics
- Receptors, Virus/genetics
- Survival Analysis
- Treatment Outcome
Collapse
Affiliation(s)
- Ping An
- Basic Research Laboratory, SAIC–Frederick, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - Rongling Li
- Office of Population Genomics, National Human Genome Research Institute, Bethesda, Maryland, United States of America
| | - Ji Ming Wang
- Laboratory of Molecular Immunoregulation, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - Teizo Yoshimura
- Laboratory of Molecular Immunoregulation, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - Munehisa Takahashi
- Laboratory of Molecular Immunoregulation, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - Ram Samudralal
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Stephen J. O'Brien
- Laboratory of Genomic Diversity, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - John Phair
- Division of Infectious Diseases, Feinberg School of Medicine, Northwestern University Medical School, Chicago, Illinois, United States of America
| | - James J. Goedert
- Infections and Immunoepidemiology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Gregory D. Kirk
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Jennifer L. Troyer
- BSP/CCR Genetics Core, SAIC–Frederick, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - Efe Sezgin
- Laboratory of Genomic Diversity, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - Susan P. Buchbinder
- San Francisco Department of Public Health, San Francisco, California, United States of America
| | | | - George W. Nelson
- BSP/CCR Genetics Core, SAIC–Frederick, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| | - Cheryl A. Winkler
- Basic Research Laboratory, SAIC–Frederick, National Cancer Institute–Frederick, Frederick, Maryland, United States of America
| |
Collapse
|
899
|
Yang OO, Lewis MJ, Reed EF, Gjertson DW, Kalilani-Phiri L, Mkandawire J, Helleringer S, Kohler HP. Human leukocyte antigen class I haplotypes of human immunodeficiency virus-1-infected persons on Likoma Island, Malawi. Hum Immunol 2011; 72:877-80. [PMID: 21663781 PMCID: PMC3175254 DOI: 10.1016/j.humimm.2011.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 05/03/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
Abstract
Human leukocyte antigen (HLA) allele frequencies vary between different human populations, with implications both for the evolutionary pressures shaping those populations as well as for the outcome of new infectious epidemics. We defined HLA class I types in a well-described cohort of persons on Likoma Island in Malawi, a population for which there are lacking data on allelic frequencies. The profile of HLA frequencies was similar but phylogenetically distinct from those of other sub-Saharan African populations in neighboring regions. The most common A alleles included A30, A23, A28 (A*68), and A2, and the most common B alleles included B15 (group), B53, B58, and B44. Notably, the frequency of B53, which is protective against malaria, was similar to that of other malaria-endemic African countries, and higher than that in countries with less malaria. This is the first reported significant dataset of HLA class I allelic frequencies in Malawians.
Collapse
Affiliation(s)
- Otto O Yang
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA.
| | | | | | | | | | | | | | | |
Collapse
|
900
|
What did we learn on host's genetics by studying large cohorts of HIV-1-infected patients in the genome-wide association era? Curr Opin HIV AIDS 2011; 6:290-6. [PMID: 21546832 DOI: 10.1097/coh.0b013e3283478449] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Genome-wide association studies (GWASs) performed in large cohorts of HIV-1-infected patients have shown that high throughput genomics can add valuable information in understanding disease progression. We report recent information gathered in the international field during the last few years and revisit the importance of well documented cohorts for genotype-phenotype association studies. RECENT FINDINGS The majority of GWASs in the HIV-1 field found that viral loads and disease progression are under the control of variants located in the major histocompatibility complex (MHC) in untreated patients. Although these experiments brought a new and more objective vision of genotype-phenotype correlations in HIV-1 disease, they also pointed out that less than 15% of the observed phenotypic variability can be explained as common genetic variants. Most of the studies have included mainly white patients and the few studies performed in Africans are underpowered but suggest that MHC is probably not the only genetic determinant influencing disease progression in this population. SUMMARY Although the first results of the GWASs in HIV disease look as a confirmation of previous findings, high throughput agnostic genomics entered the field of chronic infectious diseases and will probably unveil new genotype-phenotype associations in the future. Networks between existing cohorts leading to 'virtual mega-cohorts' will be necessary to increase the probability to discover new genetic pathways important for HIV disease. Finally, predictive models including genetic information for clinical usage is another challenge in HIV disease genetics.
Collapse
|