1
|
Démoulins T, Baron ML, Gauchat D, Kettaf N, Reed SJ, Charpentier T, Kalinke U, Lamarre A, Ahmed R, Sékaly RP, Sarkar S, Kalia V. Induction of thymic atrophy and loss of thymic output by type-I interferons during chronic viral infection. Virology 2022; 567:77-86. [PMID: 35032866 DOI: 10.1016/j.virol.2021.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 11/30/2021] [Accepted: 12/20/2021] [Indexed: 01/30/2023]
Abstract
Type-I interferon (IFN-I) signals exert a critical role in disease progression during viral infections. However, the immunomodulatory mechanisms by which IFN-I dictates disease outcomes remain to be fully defined. Here we report that IFN-I signals mediate thymic atrophy in viral infections, with more severe and prolonged loss of thymic output and unique kinetics and subtypes of IFN-α/β expression in chronic infection compared to acute infection. Loss of thymic output was linked to inhibition of early stages of thymopoiesis (DN1-DN2 transition, and DN3 proliferation) and pronounced apoptosis during the late DP stage. Notably, infection-associated thymic defects were largely abrogated upon ablation of IFNαβR and partially mitigated in the absence of CD8 T cells, thus implicating direct as well as indirect effects of IFN-I on thymocytes. These findings provide mechanistic underpinnings for immunotherapeutic strategies targeting IFN-1 signals to manipulate disease outcomes during chronic infections and cancers.
Collapse
Affiliation(s)
- Thomas Démoulins
- Institute of Virology and Immunology, Bern, Switzerland; Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Dominique Gauchat
- Centre Hospitalier de l'Université de Montréal (CHUM), 1000, rue Saint-Denis, Montréal, Québec, H2X 0C1, Canada
| | - Nadia Kettaf
- Laboratoire d'immunologie, Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Saint-Luc, Montréal, QC, H2X 1P1, Canada
| | - Steven James Reed
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Tania Charpentier
- Centre INRS-Institut Armand-Frappier, 531, Boulevard des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Alain Lamarre
- Centre INRS-Institut Armand-Frappier, 531, Boulevard des Prairies, Laval, Québec, H7V 1B7, Canada
| | - Rafi Ahmed
- Department of Microbiology & Immunology, School of Medicine, Emory University, 1510 Clifton Road, Atlanta, GA, USA
| | - Rafick-Pierre Sékaly
- Department of Pathology, Emory University Winship Cancer Center, Atlanta, GA, USA
| | - Surojit Sarkar
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Pathology, University of Washington School of Medicine, Seattle, WA, 98195, USA; Department of Pediatrics, Division of Hematology and Oncology, University of Washington, Seattle, WA, 98195, USA.
| | - Vandana Kalia
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, 98101, USA; Department of Pediatrics, Division of Hematology and Oncology, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
2
|
Zhang S, Asquith B, Szydlo R, Tregoning JS, Pollock KM. Peripheral T cell lymphopenia in COVID-19: potential mechanisms and impact. IMMUNOTHERAPY ADVANCES 2021; 1:ltab015. [PMID: 35965490 PMCID: PMC9364037 DOI: 10.1093/immadv/ltab015] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/10/2021] [Accepted: 06/30/2021] [Indexed: 12/23/2022] Open
Abstract
Immunopathogenesis involving T lymphocytes, which play a key role in defence against viral infection, could contribute to the spectrum of COVID-19 disease and provide an avenue for treatment. To address this question, a review of clinical observational studies and autopsy data in English and Chinese languages was conducted with a search of registered clinical trials. Peripheral lymphopenia affecting CD4 and CD8 T cells was a striking feature of severe COVID-19 compared with non-severe disease. Autopsy data demonstrated infiltration of T cells into organs, particularly the lung. Seventy-four clinical trials are on-going that could target T cell-related pathogenesis, particularly IL-6 pathways. SARS-CoV-2 infection interrupts T cell circulation in patients with severe COVID-19. This could be due to redistribution of T cells into infected organs, activation induced exhaustion, apoptosis, or pyroptosis. Measuring T cell dynamics during COVID-19 will inform clinical risk-stratification of hospitalised patients and could identify those who would benefit most from treatments that target T cells.
Collapse
Affiliation(s)
- Sifan Zhang
- Department of Infectious Disease, Imperial College London, London, UK
| | - Becca Asquith
- Department of Infectious Disease, Imperial College London, London, UK
| | - Richard Szydlo
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, UK
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Katrina M Pollock
- Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
3
|
Akagi J, Baba H. CD57 ratio as a convenient and useful immunological and prognostic parameter for stage IV carcinoma. Oncol Lett 2018; 15:9257-9263. [PMID: 29928332 DOI: 10.3892/ol.2018.8451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 11/30/2017] [Indexed: 11/06/2022] Open
Abstract
Cluster of differentiation (CD)8+CD57+ T cells are derived through the CD8+ T cell-differentiation signaling pathway from early differentiated CD27+CD8+CD57-T cells (early-CD8+ T cells) to terminal-differentiated CD27-CD8+CD57+ T cells (terminal-CD8+ T cells) via intermediate-differentiated CD27+CD8+CD57+ T cells (intermediate-CD8+ T cells). The increase of CD8+CD57+ T cells in the peripheral blood of patients with cancer has been associated with prognosis, which suggests their suitability as a candidate immunological marker. The present study investigated the association of these CD57-related CD8+ T cell populations in the peripheral blood of 100 Stage IV cancer patients with progression-free survival (PFS), using a Cox regression model. Univariate analysis indicated that early- and intermediate-CD8+ T cells were associated with shorter PFS, whereas terminal-CD8+ T cells were associated with longer PFS. A strong inverse correlation was observed between early- and terminal-CD8+ T cells, and multivariate analysis demonstrated that the CD57 ratio (terminal-CD8+ T cells/early-CD8+ T cells) was a more significant independent prognostic factor compared with early- or terminal-CD8+ T cells. Patients with a higher CD57 ratio had a significantly longer PFS compared with those with a lower CD57 ratio, in whom terminal-CD8+ T cells were supposed to be predominant. Conversely, results indicated inhibition of the CD8+ T cell differentiation signaling pathway in patients with a low CD57 ratio, which lead to a predominance of early-CD8+ T cells, a characteristic of immunosuppressive cells. The present findings suggested that the CD57 ratio appears to be a powerful immunological prognostic parameter obtained from the peripheral blood, precisely reflecting the state of CD8+ T cell-differentiation.
Collapse
Affiliation(s)
- Junji Akagi
- Department of Surgery, Tamana Regional Health Medical Center, Tamana, Kumamoto 865-0005, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
4
|
Akagi J, Baba H, Sekine T, Ogawa K. Terminally differentiated CD8 + T cells and CD57 -FOXP3 +CD8 + T cells are highly associated with the efficacy of immunotherapy using activated autologous lymphocytes. Oncol Lett 2018; 15:9529-9536. [PMID: 29805674 DOI: 10.3892/ol.2018.8512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 09/07/2017] [Indexed: 11/06/2022] Open
Abstract
Treatment with activated autologous lymphocytes (AALs) has demonstrated mixed results for cancer treatment. Preliminary results revealed that the proportion of cluster of differentiation (CD)8+CD57+ T cells is significantly increased in AALs, indicating that they are able to determine treatment outcome. Therefore, the role of CD8+CD57+ T cells in AAL efficacy was investigated. T lymphocytes were isolated from 35 patients with stage IV gastric carcinomas (17 men and 18 women; aged 41-84 years) receiving immunotherapy using AALs (IAAL). Using fluorescence activated cell sorting, CD8, CD27, CD57, and forkhead box protein 3 (FOXP3) expression was investigated on CD8+ T cell populations in CD8+ T cell differentiation prior to and following in vitro culture. The association between these populations and progression-free survival (PFS) was analyzed using Cox univariate, and multivariate analyses and Kaplan-Meier survival analysis. CD57 expression was negative in early-differentiated CD8+ T cells (CD27+CD8+CD57-), and positive in intermediate- (CD27+CD8+CD57+) and terminal- (CD27-CD8+CD57+) differentiated CD8+ T cells. Univariate analysis revealed a significant association between terminal-CD8+ T cells and longer PFS times (P=0.035), whereas CD57-FOXP3+CD8+ T cells were associated with shorter PFS times. Multivariate analysis revealed that CD57-FOXP3+CD8+ T cells was an independent poor prognostic factor, whereas CD57+FOXP3+CD8+ T cells were not associated with PFS. Although IAAL increased the proportion of terminal-CD8+ T cells relative to the pre-culture proportions, patients with a high CD57-FOXP3+CD8+ T cell percentage exhibited repressed terminal-CD8+ T cell induction, leading to poor patient prognosis. Terminally differentiated CD27-CD8+CD57+ T cells were responsible for the effectiveness of AALs; however, CD57-FOXP3+CD8+ T cells abrogated their efficacy, possibly by inhibiting their induction.
Collapse
Affiliation(s)
- Junji Akagi
- Department of Surgery, Tamana Regional Health Medical Center, Tamana, Kumamoto 865-0005, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | | | | |
Collapse
|
5
|
Shrimali RK, Ahmad S, Verma V, Zeng P, Ananth S, Gaur P, Gittelman RM, Yusko E, Sanders C, Robins H, Hammond SA, Janik JE, Mkrtichyan M, Gupta S, Khleif SN. Concurrent PD-1 Blockade Negates the Effects of OX40 Agonist Antibody in Combination Immunotherapy through Inducing T-cell Apoptosis. Cancer Immunol Res 2018; 5:755-766. [PMID: 28848055 DOI: 10.1158/2326-6066.cir-17-0292] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 06/21/2017] [Accepted: 07/18/2017] [Indexed: 11/16/2022]
Abstract
Combination therapies that depend on checkpoint inhibitor antibodies (Abs) such as for PD-1 or its ligand (PD-L1) together with immune stimulatory agonist Abs like anti-OX40 are being tested in the clinic to achieve improved antitumor effects. Here, we studied the potential therapeutic and immune effects of one such combination: Ab to PD-1 with agonist Ab to OX40/vaccine. We tested the antitumor effects of different treatment sequencing of this combination. We report that simultaneous addition of anti-PD-1 to anti-OX40 negated the antitumor effects of OX40 Ab. Antigen-specific CD8+ T-cell infiltration into the tumor was diminished, the resultant antitumor response weakened, and survival reduced. Although we observed an increase in IFNγ-producing E7-specifc CD8+ T cells in the spleens of mice treated with the combination of PD-1 blockade with anti-OX40/vaccine, these cells underwent apoptosis both in the periphery and the tumor. These results indicate that anti-PD-1 added at the initiation of therapy exhibits a detrimental effect on the positive outcome of anti-OX40 agonist Ab. These findings have important implications on the design of combination immunotherapy for cancer, demonstrating the need to test treatment combination and sequencing before moving to the clinic. Cancer Immunol Res; 5(9); 755-66. ©2017 AACR.
Collapse
Affiliation(s)
| | - Shamim Ahmad
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Vivek Verma
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Peng Zeng
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Sudha Ananth
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Pankaj Gaur
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | | | - Erik Yusko
- Adaptive Biotechnologies, Seattle, Washington
| | | | - Harlan Robins
- Adaptive Biotechnologies, Seattle, Washington.,Computational Biology Program, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | - John E Janik
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | | | - Seema Gupta
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University, Augusta, Georgia.
| |
Collapse
|
6
|
George J, Mattapallil JJ. Interferon-α Subtypes As an Adjunct Therapeutic Approach for Human Immunodeficiency Virus Functional Cure. Front Immunol 2018; 9:299. [PMID: 29520278 PMCID: PMC5827157 DOI: 10.3389/fimmu.2018.00299] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/02/2018] [Indexed: 01/12/2023] Open
Abstract
Human immunodeficiency virus (HIV) establishes life-long latency in infected individuals. Although highly active antiretroviral therapy (HAART) has had a significant impact on the course of HIV infection leading to a better long-term outcome, the pool of latent reservoir remains substantial even under HAART. Numerous approaches have been under development with the goal of eradicating the latent HIV reservoir though with limited success. Approaches that combine immune-mediated control of HIV to activate both the innate and the adaptive immune system under suppressive therapy along with "shock and kill" drugs may lead to a better control of the reactivated virus. Interferon-α (IFN-α) is an innate cytokine that has been shown to activate intracellular defenses capable of restricting and controlling HIV. IFN-α, however, harbors numerous functional subtypes that have been reported to display different binding affinities and potency. Recent studies have suggested that certain subtypes such as IFN-α8 and IFN-α14 have potent anti-HIV activity with little or no immune activation, whereas other subtypes such as IFN-α4, IFN-α5, and IFN-α14 activate NK cells. Could these subtypes be used in combination with other strategies to reduce the latent viral reservoir? Here, we review the role of IFN-α subtypes in HIV infection and discuss the possibility that certain subtypes could be potential adjuncts to a "shock and kill" or therapeutic vaccination strategy leading to better control of the latent reservoir and subsequent functional cure.
Collapse
Affiliation(s)
- Jeffy George
- Uniformed Services University, Bethesda, MD, United States
| | | |
Collapse
|
7
|
Khouri R, Silva-Santos G, Dierckx T, Menezes SM, Decanine D, Theys K, Silva AC, Farré L, Bittencourt A, Mangino M, Roederer M, Vandamme AM, Van Weyenbergh J. A genetic IFN/STAT1/FAS axis determines CD4 T stem cell memory levels and apoptosis in healthy controls and Adult T-cell Leukemia patients. Oncoimmunology 2018; 7:e1426423. [PMID: 29721391 PMCID: PMC5927537 DOI: 10.1080/2162402x.2018.1426423] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 01/04/2018] [Accepted: 01/07/2018] [Indexed: 11/22/2022] Open
Abstract
Adult T-cell leukemia (ATL) is an aggressive, chemotherapy-resistant CD4+CD25+ leukemia caused by HTLV-1 infection, which usually develops in a minority of patients several decades after infection. IFN + AZT combination therapy has shown clinical benefit in ATL, although its mechanism of action remains unclear. We have previously shown that an IFN-responsive FAS promoter polymorphism in a STAT1 binding site (rs1800682) is associated to ATL susceptibility and survival. Recently, CD4 T stem cell memory (TSCM) Fashi cells have been identified as the hierarchical cellular apex of ATL, but a possible link between FAS, apoptosis, proliferation and IFN response in ATL has not been studied. In this study, we found significant ex vivo antiproliferative, antiviral and immunomodulatory effects of IFN-α treatment in short-term culture of primary mononuclear cells from ATL patients (n = 25). Bayesian Network analysis allowed us to integrate ex vivo IFN-α response with clinical, genetic and immunological data from ATL patients, thereby revealing a central role for FAS -670 polymorphism and apoptosis in the coordinated mechanism of action of IFN-α. FAS genotype-dependence of IFN-induced apoptosis was experimentally validated in an independent cohort of healthy controls (n = 20). The same FAS -670 polymorphism also determined CD4 TSCM levels in a genome-wide twin study (p = 7 × 10-11, n = 460), confirming a genetic link between apoptosis and TSCM levels. Transcriptomic analysis and cell type deconvolution confirmed the FAS genotype/TSCM link and IFN-α-induced downregulation of CD4 TSCM-specific genes in ATL patient cells. In conclusion, ex vivo IFN-α treatment exerts a pleiotropic effect on primary ATL cells, with a genetic IFN/STAT1/Fas axis determining apoptosis vs. proliferation and underscoring the CD4 TSCM model of ATL leukemogenesis.
Collapse
Affiliation(s)
- Ricardo Khouri
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | | | - Tim Dierckx
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Soraya Maria Menezes
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Daniele Decanine
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | - Kristof Theys
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | - Aline Clara Silva
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | - Lourdes Farré
- Instituto Gonçalo Moniz (IGM) - Fundação Oswaldo Cruz (FIOCRUZ), Salvador-Bahia, Brazil
| | - Achiléa Bittencourt
- Department of Pathology, Complexo Hospitalar Universitário Prof Edgard Santos, Universidade Federal da Bahia (UFBA), Salvador-Bahia, Brazil
| | - Massimo Mangino
- Department of Twin Research & Genetic Epidemiology, King's College, London, UK
| | - Mario Roederer
- Immunotechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda-MD, USA
| | - Anne-Mieke Vandamme
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
- Center for Global Health and Tropical Medicine, Unidade de Microbiologia, Instituto de Highne e Medicina Tropical, Universidade, Nova de Lisboa, Lisbon, Portugal
| | - Johan Van Weyenbergh
- KU Leuven – University of Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| |
Collapse
|
8
|
Abstract
The human gut is in constant complex interaction with the external environment. Although much is understood about the composition and function of the microbiota, much remains to be learnt about the mechanisms by which these organisms interact with the immune system in health and disease. Type 1 interferon (T1IFN), a ubiquitous and pleiotropic family of cytokines, is a critical mediator of the response to viral, bacterial, and other antigens sampled in the intestine. Although inflammation is enhanced in mouse model of colitis when T1IFN signaling is lost, the action of T1IFN is context specific and can be pro- or anti-inflammatory. In humans, T1IFN has been used to treat inflammatory diseases, including multiple sclerosis and inflammatory bowel disease but intestinal inflammation can also develop after the administration of T1IFN. Recent findings indicate that "tonic" or "endogenous" T1IFN, induced by signals from the commensal microbiota, modulates the local signaling environment to prime the intestinal mucosal immune system to determine later responses to pathogens and commensal organisms. This review will summarize the complex immunological effects of T1IFN and recent the role of T1IFN as a mediator between the microbiota and the mucosal immune system, highlighting human data wherever possible. It will discuss what we can learn from clinical experiences with T1IFN and how the T1IFN pathway may be manipulated in the future to maintain mucosal homeostasis.
Collapse
|
9
|
Peripheral Leukocyte Migration in Ferrets in Response to Infection with Seasonal Influenza Virus. PLoS One 2016; 11:e0157903. [PMID: 27315117 PMCID: PMC4912066 DOI: 10.1371/journal.pone.0157903] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 06/07/2016] [Indexed: 12/31/2022] Open
Abstract
In order to better understand inflammation associated with influenza virus infection, we measured cell trafficking, via flow cytometry, to various tissues in the ferret model following infection with an A(H3N2) human seasonal influenza virus (A/Perth/16/2009). Changes in immune cells were observed in the blood, bronchoalveolar lavage fluid, and spleen, as well as lymph nodes associated with the site of infection or distant from the respiratory system. Nevertheless clinical symptoms were mild, with circulating leukocytes exhibiting rapid, dynamic, and profound changes in response to infection. Each of the biological compartments examined responded differently to influenza infection. Two days after infection, when infected ferrets showed peak fever, a marked, transient lymphopenia and granulocytosis were apparent in all infected animals. Both draining and distal lymph nodes demonstrated significant accumulation of T cells, B cells, and granulocytes at days 2 and 5 post-infection. CD8+ T cells significantly increased in spleen at days 2 and 5 post-infection; CD4+ T cells, B cells and granulocytes significantly increased at day 5. We interpret our findings as showing that lymphocytes exit the peripheral blood and differentially home to lymph nodes and tissues based on cell type and proximity to the site of infection. Monitoring leukocyte homing and trafficking will aid in providing a more detailed view of the inflammatory impact of influenza virus infection.
Collapse
|
10
|
Jiang J, Wang M, Liang B, Shi Y, Su Q, Chen H, Huang J, Su J, Pan P, Li Y, Wang H, Chen R, Liu J, Zhao F, Ye L, Liang H. In vivo effects of methamphetamine on HIV-1 replication: A population-based study. Drug Alcohol Depend 2016; 159:246-54. [PMID: 26790825 DOI: 10.1016/j.drugalcdep.2015.12.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 11/18/2022]
Abstract
BACKGROUND Although a number of in vitro studies have shown that methamphetamine (METH) can increase HIV-1 replication in human immune cells, a direct link between METH use and HIV-1 pathogenesis remains to be determined among HIV-1 patients. METHODS According to the status of METH use and HIV-1 infection, we enrolled participants and divided them into four groups: METH+HIV+, METH-HIV+, METH+HIV-, and METH-HIV-. HIV viral loads and HIV-1-related cellular factors were measured and compared among different groups. RESULTS A total of 60 participants were enrolled into this study, 15 within each group. HIV viral loads in METH+HIV+ group were significantly higher than those in METH-HIV+ group, while CD4+ T cell counts had an inverse trend between the two groups (p<0.05). METH users or HIV-1 infected patients had lower CCR5+, CXCR4+ percentages in CD4+ T cells than METH-HIV- subjects (p<0.01). However, METH use had little effect on CD3 expression in PBMCs and the levels of MIP-1α, MIP-1β and IL-6 in PBMCs or plasma, which were increased by HIV-1 infection with or without METH. TLR-9 and IFN-α levels in PBMCs of METH users with or without HIV infection were higher than non-METH users (p<0.05). CONCLUSIONS METH use is associated with higher viral loads and lower CD4+ T cell counts in HIV-infected individuals. This finding may be mediated by activation of innate immunity (TLR-9, IFN-α) by METH use.
Collapse
Affiliation(s)
- Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Minlian Wang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Bingyu Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Yi Shi
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Qijian Su
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Hui Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jinming Su
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Peijiang Pan
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Yu Li
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Hong Wang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Rongfeng Chen
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Jie Liu
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Fangning Zhao
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical Research Center, Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment & Guangxi Universities Key Laboratory of Prevention and Control of Highly Prevalent Disease, School of Public Health, Guangxi Medical University, Nanning 530021, Guangxi, China; Guangxi Collaborative Innovation Center for Biomedicine, Guangxi Medical Research Center, Guangxi Medical University, Nanning 530021, Guangxi, China.
| |
Collapse
|
11
|
Banks HT, Flores KB, Hu S, Rosenberg E, Buzon M, Yu X, Lichterfeld M. Immuno-modulatory strategies for reduction of HIV reservoir cells. J Theor Biol 2015; 372:146-58. [PMID: 25701451 DOI: 10.1016/j.jtbi.2015.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 02/04/2015] [Accepted: 02/05/2015] [Indexed: 11/18/2022]
Abstract
Antiretroviral therapy is able to suppress the viral load to below the detection limit, but it is not able to eradicate HIV reservoirs. Thus, there is a critical need for a novel treatment to eradicate (or reduce) the reservoir in order to eliminate the need for a lifelong adherence to antiretroviral therapy, which is expensive and potentially toxic. In this paper, we investigate the possible pharmacological strategies or combinations of strategies that may be beneficial to reduce or possibly eradicate the latent reservoir. We do this via studies with a validated mathematical model, where the parameter values are obtained with newly acquired clinical data for HIV patients. Our findings indicate that the strategy of reactivating the reservoir combined with enhancement of the killing rate of HIV-specific CD8+ T cells is able to eradicate the reservoir. In addition, our analysis shows that a targeted suppression of the immune system is also a possible strategy to eradicate the reservoir.
Collapse
Affiliation(s)
- H T Banks
- Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC 27695-8212, USA.
| | - Kevin B Flores
- Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC 27695-8212, USA
| | - Shuhua Hu
- Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC 27695-8212, USA
| | - Eric Rosenberg
- Harvard Medical School and Mass General Hospital, Ragon Institute, Boston, MA, USA
| | - Maria Buzon
- Harvard Medical School and Mass General Hospital, Ragon Institute, Boston, MA, USA
| | - Xu Yu
- Harvard Medical School and Mass General Hospital, Ragon Institute, Boston, MA, USA
| | - Matthias Lichterfeld
- Harvard Medical School and Mass General Hospital, Ragon Institute, Boston, MA, USA
| |
Collapse
|
12
|
Martins KAO, Bavari S, Salazar AM. Vaccine adjuvant uses of poly-IC and derivatives. Expert Rev Vaccines 2014; 14:447-59. [PMID: 25308798 DOI: 10.1586/14760584.2015.966085] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pathogen-associated molecular patterns (PAMPs) are stand-alone immunomodulators or 'danger signals,' that are increasingly recognized as critical components of many modern vaccines. Polyinosinic-polycytidylic acid (poly-IC) is a synthetic dsRNA that can activate multiple elements of the host defense in a pattern that parallels that of a viral infection. When properly combined with an antigen, it can be utilized as a PAMP-adjuvant, resulting in modulation and optimization of the antigen-specific immune response. We briefly review the preclinical and clinical uses of poly-IC and two poly-IC derivatives, poly-IC12U (Ampligen) and poly-ICLC (Hiltonol), as vaccine adjuvants.
Collapse
|
13
|
Cha L, de Jong E, French MA, Fernandez S. IFN-α exerts opposing effects on activation-induced and IL-7-induced proliferation of T cells that may impair homeostatic maintenance of CD4+ T cell numbers in treated HIV infection. THE JOURNAL OF IMMUNOLOGY 2014; 193:2178-86. [PMID: 25063872 DOI: 10.4049/jimmunol.1302536] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
To determine whether IFN-α is a cause of the T cell hyperactivation and IL-7 signaling pathway defects that are observed in some HIV patients receiving antiretroviral therapy, we have investigated the effect of IFN-α on the proliferation of CD4(+) and CD8(+) T cells from healthy donors (n = 30) and treated HIV(+) donors (n = 20). PBMC were cultured for 7 d with staphylococcal enterotoxin B or IL-7 in the absence or presence of 100 U/ml IFN-α8. Total and naive CD4(+) and CD8(+) T cells were assessed for proliferation (via Ki67 expression), CD127 expression, and phosphorylated STAT5 levels using flow cytometry. IFN-α significantly enhanced activation-induced proliferation (via staphylococcal enterotoxin B stimulation) but inhibited homeostatic proliferation (IL-7 induced) of CD4(+) and CD8(+) T cells. Both of these effects may adversely affect CD4(+) T cell homeostasis in HIV patients. CD127 expression was increased in both healthy and HIV(+) donors following culture with IFN-α8, and levels of IL-7-induced phosphorylated STAT5 were increased by IFN-α8 in healthy donors only. Hence, the inhibitory effects of IFN-α on IL-7-induced proliferation of CD4(+) T cells are unlikely to be mediated by downregulation of CD127 expression or inhibition of STAT5 phosphorylation. These data suggest that increased IFN-α activity may promote the loss of T cells by accelerating cell turnover and activation-induced cell death while decreasing the renewal of T cells by inhibiting the proliferative effect of IL-7.
Collapse
Affiliation(s)
- Lilian Cha
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia; and
| | - Emma de Jong
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia; and
| | - Martyn A French
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia; and Department of Clinical Immunology, Royal Perth Hospital and PathWest Laboratory Medicine, Perth, Western Australia 6000, Australia
| | - Sonia Fernandez
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia 6009, Australia; and
| |
Collapse
|
14
|
Music N, Reber AJ, Lipatov AS, Kamal RP, Blanchfield K, Wilson JR, Donis RO, Katz JM, York IA. Influenza vaccination accelerates recovery of ferrets from lymphopenia. PLoS One 2014; 9:e100926. [PMID: 24968319 PMCID: PMC4072694 DOI: 10.1371/journal.pone.0100926] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 06/01/2014] [Indexed: 01/06/2023] Open
Abstract
Ferrets are a useful animal model for human influenza virus infections, since they closely mimic the pathogenesis of influenza viruses observed in humans. However, a lack of reagents, especially for flow cytometry of immune cell subsets, has limited research in this model. Here we use a panel of primarily species cross-reactive antibodies to identify ferret T cells, cytotoxic T lymphocytes (CTL), B cells, and granulocytes in peripheral blood. Following infection with seasonal H3N2 or H1N1pdm09 influenza viruses, these cell types showed rapid and dramatic changes in frequency, even though clinically the infections were mild. The loss of B cells and CD4 and CD8 T cells, and the increase in neutrophils, were especially marked 1–2 days after infection, when about 90% of CD8+ T cells disappeared from the peripheral blood. The different virus strains led to different kinetics of leukocyte subset alterations. Vaccination with homologous vaccine reduced clinical symptoms slightly, but led to a much more rapid return to normal leukocyte parameters. Assessment of clinical symptoms may underestimate the effectiveness of influenza vaccine in restoring homeostasis.
Collapse
Affiliation(s)
- Nedzad Music
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Adrian J. Reber
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Aleksandr S. Lipatov
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ram P. Kamal
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Kristy Blanchfield
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jason R. Wilson
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ruben O. Donis
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jacqueline M. Katz
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ian A. York
- Influenza Division, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
15
|
Moraga I, Spangler J, Mendoza JL, Garcia KC. Multifarious determinants of cytokine receptor signaling specificity. Adv Immunol 2014; 121:1-39. [PMID: 24388212 DOI: 10.1016/b978-0-12-800100-4.00001-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytokines play crucial roles in regulating immune homeostasis. Two important characteristics of most cytokines are pleiotropy, defined as the ability of one cytokine to exhibit diverse functionalities, and redundancy, defined as the ability of multiple cytokines to exert overlapping activities. Identifying the determinants for unique cellular responses to cytokines in the face of shared receptor usage, pleiotropy, and redundancy will be essential in order to harness the potential of cytokines as therapeutics. Here, we discuss the biophysical (ligand-receptor geometry and affinity) and cellular (receptor trafficking and intracellular abundance of signaling molecules) parameters that contribute to the specificity of cytokine bioactivities. Whereas the role of extracellular ternary complex geometry in cytokine-induced signaling is still not completely elucidated, cytokine-receptor affinity is known to impact signaling through modulation of the stability and kinetics of ternary complex formation. Receptor trafficking also plays an important and likely underappreciated role in the diversification of cytokine bioactivities but it has been challenging to experimentally probe trafficking effects. We also review recent efforts to quantify levels of intracellular signaling components, as second messenger abundance can affect cytokine-induced bioactivities both quantitatively and qualitatively. We conclude by discussing the application of protein engineering to develop therapeutically relevant cytokines with reduced pleiotropy and redirected biological functionalities.
Collapse
Affiliation(s)
- Ignacio Moraga
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, California, USA; Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Jamie Spangler
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, California, USA; Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - Juan L Mendoza
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, California, USA; Program in Immunology, Stanford University School of Medicine, Stanford, California, USA
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California, USA; Department of Structural Biology, Stanford University School of Medicine, Stanford, California, USA; Program in Immunology, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
16
|
Type I interferon upregulates Bak and contributes to T cell loss during human immunodeficiency virus (HIV) infection. PLoS Pathog 2013; 9:e1003658. [PMID: 24130482 PMCID: PMC3795023 DOI: 10.1371/journal.ppat.1003658] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Accepted: 08/12/2013] [Indexed: 11/19/2022] Open
Abstract
The role of Type I interferon (IFN) during pathogenic HIV and SIV infections remains unclear, with conflicting observations suggesting protective versus immunopathological effects. We therefore examined the effect of IFNα/β on T cell death and viremia in HIV infection. Ex vivo analysis of eight pro- and anti-apoptotic molecules in chronic HIV-1 infection revealed that pro-apoptotic Bak was increased in CD4+ T cells and correlated directly with sensitivity to CD95/Fas-mediated apoptosis and inversely with CD4+ T cell counts. Apoptosis sensitivity and Bak expression were primarily increased in effector memory T cells. Knockdown of Bak by RNA interference inhibited CD95/Fas-induced death of T cells from HIV-1-infected individuals. In HIV-1-infected patients, IFNα-stimulated gene expression correlated positively with ex vivo T cell Bak levels, CD95/Fas-mediated apoptosis and viremia and negatively with CD4+ T cell counts. In vitro IFNα/β stimulation enhanced Bak expression, CD95/Fas expression and CD95/Fas-mediated apoptosis in healthy donor T cells and induced death of HIV-specific CD8+ T cells from HIV-1-infected patients. HIV-1 in vitro sensitized T cells to CD95/Fas-induced apoptosis and this was Toll-like receptor (TLR)7/9- and Type I IFN-dependent. This sensitization by HIV-1 was due to an indirect effect on T cells, as it occurred in peripheral blood mononuclear cell cultures but not purified CD4+ T cells. Finally, peak IFNα levels and viral loads correlated negatively during acute SIV infection suggesting a potential antiviral effect, but positively during chronic SIV infection indicating that either the virus drives IFNα production or IFNα may facilitate loss of viral control. The above findings indicate stage-specific opposing effects of Type I IFNs during HIV-1 infection and suggest a novel mechanism by which these cytokines contribute to T cell depletion, dysregulation of cellular immunity and disease progression.
Collapse
|
17
|
Corre B, Perrier J, El Khouri M, Cerboni S, Pellegrini S, Michel F. Type I interferon potentiates T-cell receptor mediated induction of IL-10-producing CD4⁺ T cells. Eur J Immunol 2013; 43:2730-40. [PMID: 23839924 DOI: 10.1002/eji.201242977] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Revised: 04/26/2013] [Accepted: 06/18/2013] [Indexed: 12/24/2022]
Abstract
Type I interferons (IFNs) have the dual ability to promote the development of the immune response and exert an anti-inflammatory activity. We analyzed the integrated effect of IFN-α, TCR signal strength, and CD28 costimulation on human CD4⁺ T-cell differentiation into cell subsets producing the anti- and proinflammatory cytokines IL-10 and IFN-γ. We show that IFN-α boosted TCR-induced IL-10 expression in activated peripheral CD45RA⁺CD4⁺ T cells and in whole blood cultures. The functional cooperation between TCR and IFN-α efficiently occurred at low engagement of receptors. Moreover, IFN-α rapidly cooperated with anti-CD3 stimulation alone. IFN-α, but not IL-10, drove the early development of type I regulatory T cells that were mostly IL-10⁺ Foxp3⁻ IFN-γ⁻ and favored IL-10 expression in a fraction of Foxp3⁺ T cells. Our data support a model in which IFN-α costimulates TCR toward the production of IL-10 whose level can be amplified via an autocrine feedback loop.
Collapse
Affiliation(s)
- Béatrice Corre
- Department of Immunology, Unit of Cytokine Signaling, Institut Pasteur, Paris, France
| | | | | | | | | | | |
Collapse
|
18
|
Beq S, Rozlan S, Pelletier S, Willems B, Bruneau J, Lelievre JD, Levy Y, Shoukry NH, Cheynier R. Altered thymic function during interferon therapy in HCV-infected patients. PLoS One 2012; 7:e34326. [PMID: 22529911 PMCID: PMC3328332 DOI: 10.1371/journal.pone.0034326] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 02/25/2012] [Indexed: 12/19/2022] Open
Abstract
Interferon alpha (IFNα) therapy, despite good efficacy in curing HCV infection, leads to major side effects, in particular inducement of a strong peripheral T-cell lymphocytopenia. We here analyze the early consequences of IFNα therapy on both thymic function and peripheral T-cell homeostasis in patients in the acute or chronic phase of HCV-infection as well as in HIV/HCV co-infected patients. The evolution of T-cell subsets and T-cell homeostasis were estimated by flow cytometry while thymic function was measured through quantification of T-cell receptor excision circles (TREC) and estimation of intrathymic precursor T-cell proliferation during the first four months following the initiation of IFNα therapy. Beginning with the first month of therapy, a profound lymphocytopenia was observed for all T-cell subsets, including naïve T-cells and recent thymic emigrants (RTE), associated with inhibition of intrathymic precursor T-cell proliferation. Interleukin (IL)-7 plasma concentration rapidly dropped while lymphocytopenia progressed. This was neither a consequence of higher consumption of the cytokine nor due to its neutralization by soluble CD127. Decrease in IL-7 plasma concentration under IFNα therapy correlated with the decline in HCV viral load, thymic activity and RTE concentration in blood. These data demonstrate that IFNα-based therapy rapidly impacts on thymopoiesis and, consequently, perturbs T-cell homeostasis. Such a side effect might be detrimental for the continuation of IFNα therapy and may lead to an increased level of infectious risk, in particular in HIV/HCV co-infected patients. Altogether, this study suggests the therapeutic potential of IL-7 in the maintenance of peripheral T-cell homeostasis in IFNα-treated patients.
Collapse
Affiliation(s)
- Stephanie Beq
- Département de Virologie, Institut Pasteur, Paris, France
| | - Sandra Rozlan
- Département de Virologie, Institut Pasteur, Paris, France
| | - Sandy Pelletier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Hôpital St-Luc, Montréal, Québec, Canada
- Département de microbiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Bernard Willems
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Hôpital St-Luc, Montréal, Québec, Canada
- Département de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Julie Bruneau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Hôpital St-Luc, Montréal, Québec, Canada
- Département de médecine familiale, Université de Montréal, Montréal, Québec, Canada
| | - Jean-Daniel Lelievre
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Henri-Mondor Albert-Chenevier, Immunologie Clinique, INSERM, U955, Université Paris 12, Créteil, France
| | - Yves Levy
- Assistance Publique-Hôpitaux de Paris (AP-HP), Groupe Henri-Mondor Albert-Chenevier, Immunologie Clinique, INSERM, U955, Université Paris 12, Créteil, France
| | - Naglaa H. Shoukry
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Hôpital St-Luc, Montréal, Québec, Canada
- Département de médecine, Université de Montréal, Montréal, Québec, Canada
| | - Rémi Cheynier
- Département de Virologie, Institut Pasteur, Paris, France
- Inserm U1016, Département Immunologie-Hématologie, Institut Cochin, Paris, France
- CNRS, UMR 8104, Paris, France
- Faculté de Médecine René Descartes, Université Paris Descartes, UMR-S 8104, Paris, France
- * E-mail:
| |
Collapse
|
19
|
Yue C, Soboloff J, Gamero AM. Control of type I interferon-induced cell death by Orai1-mediated calcium entry in T cells. J Biol Chem 2011; 287:3207-16. [PMID: 22144678 DOI: 10.1074/jbc.m111.269068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Store-operated Ca(2+) entry (SOCE) is an essential process in T cell activation. SOCE is controlled by the Ca(2+) release-activated Ca(2+) (CRAC) channel encoded by the gene Orai1 that is expressed on the plasma membrane and activated by STIM1 when ER Ca(2+) stores are depleted. Our earlier work showed that a somatic T-cell line Jurkat mutant H123 bearing a defect in Ca(2+) signaling was susceptible to the apoptotic effects of type I interferons (IFN-α/β). The nature of the mutation and whether this mutation was linked to IFN-α/β apoptotic susceptibility was unknown. Here we show that H123 cells lacked Orai1 and exhibit reduced STIM1 protein. Reconstitution of both Orai1 and STIM1 in H123 cells rescued SOCE in response to thapsigargin and ionomycin and abrogated IFN-α/β-induced apoptosis. Reciprocally, overexpression of the dominant negative Orai1-E106A in either parental Jurkat cells or an unrelated human T cell line (CEM391) inhibited SOCE and led to sensitization to IFN-α/β-induced apoptosis. Furthermore, we showed that the Ca(2+) response pathway antagonized the IFN-α/β -induced transcriptional responses; in the absence of SOCE, this negative regulatory effect was lost. However, the inhibitory effect of Ca(2+) on type I IFN-induced gene transcription was diminished by pharmacological inhibition of NF-κB in cells with intact SOCE. Our findings reveal an unexpected and novel regulatory crosstalk mechanism between type I IFNs and store-operated Ca(2+) signaling pathways mediated at least in part by NF-κB activity with significant clinical implications to both viral and tumor immunology.
Collapse
Affiliation(s)
- Chanyu Yue
- Department of Biochemistry, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | |
Collapse
|
20
|
Bacher N, Graulich E, Jonuleit H, Grabbe S, Steinbrink K. Interferon-α abrogates tolerance induction by human tolerogenic dendritic cells. PLoS One 2011; 6:e22763. [PMID: 21818385 PMCID: PMC3144929 DOI: 10.1371/journal.pone.0022763] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 06/29/2011] [Indexed: 02/08/2023] Open
Abstract
Background Administration of interferon-α (IFN-α) represents an approved adjuvant therapy as reported for malignancies like melanoma and several viral infections. In malignant diseases, tolerance processes are critically involved in tumor progression. In this study, the effect of IFN-α on tolerance induction by human tolerogenic dendritic cells (DC) was analyzed. We focussed on tolerogenic IL-10-modulated DC (IL-10 DC) that are known to induce anergic regulatory T cells (iTregs). Methodology/Principal Findings IFN-α promoted an enhanced maturation of IL-10 DC as demonstrated by upregulation of the differentiation marker CD83 as well as costimulatory molecules. IFN-α treatment resulted in an increased capacity of DC to stimulate T cell activation compared to control tolerogenic DC. We observed a strengthened T cell proliferation and increased IFN-γ production of CD4+ and CD8+ T cells stimulated by IFN-α-DC, demonstrating a restoration of the immunogenic capacity of tolerogenic DC in the presence of IFN-α. Notably, restimulation experiments revealed that IFN-α treatment of tolerogenic DC abolished the induction of T cell anergy and suppressor function of iTregs. In contrast, IFN-α neither affected the priming of iTregs nor converted iTregs into effector T cells. Conclusions/Significance IFN-α inhibits the induction of T cell tolerance by reversing the tolerogenic function of human DC.
Collapse
Affiliation(s)
- Nicole Bacher
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
- * E-mail: (KS); (NB)
| | - Edith Graulich
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
| | - Helmut Jonuleit
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
| | - Kerstin Steinbrink
- Department of Dermatology, University Medical Center Mainz, University of Mainz, Mainz, Germany
- * E-mail: (KS); (NB)
| |
Collapse
|
21
|
Quetglas JI, Fioravanti J, Ardaiz N, Medina-Echeverz J, Baraibar I, Prieto J, Smerdou C, Berraondo P. A Semliki forest virus vector engineered to express IFNα induces efficient elimination of established tumors. Gene Ther 2011; 19:271-8. [PMID: 21734727 DOI: 10.1038/gt.2011.99] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Semliki Forest virus (SFV) represents a promising gene therapy vector for tumor treatment, because it produces high levels of recombinant therapeutic proteins while inducing apoptosis in infected cells. In this study, we constructed a SFV vector expressing murine interferon alpha (IFNα). IFNα displays antitumor activity mainly by enhancing an antitumor immune response, as well as by a direct antiproliferative effect. In spite of the antiviral activity of IFNα, SFV-IFN could be produced in BHK cells at high titers. This vector was able to infect TC-1 cells, a tumor cell line expressing E6 and E7 proteins of human papillomavirus, leading to high production of IFNα both in vitro and in vivo. When injected into subcutaneous TC-1 tumors implanted in mice, SFV-IFN was able to induce an E7-specific cytotoxic T lymphocyte response, and to modify tumor infiltrating immune cells, reducing the percentage of T regulatory cells and activating myeloid cells. As a consequence, SFV-IFN was able to eradicate 58% of established tumors treated 21 days after implantation with long-term tumor-free survival and very low toxicity. SFV-IFN was also able to induce significant antitumor responses in a subcutaneous tumor model of murine colon adenocarcimoma. These data suggest that local production of IFNα by intratumoral injection of recombinant SFV-IFN could represent a potent new strategy to treat tumors in patients.
Collapse
Affiliation(s)
- J I Quetglas
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Strioga M, Pasukoniene V, Characiejus D. CD8+ CD28- and CD8+ CD57+ T cells and their role in health and disease. Immunology 2011; 134:17-32. [PMID: 21711350 DOI: 10.1111/j.1365-2567.2011.03470.x] [Citation(s) in RCA: 369] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic antigenic stimulation leads to gradual accumulation of late-differentiated, antigen-specific, oligoclonal T cells, particularly within the CD8(+) T-cell compartment. They are characterized by critically shortened telomeres, loss of CD28 and/or gain of CD57 expression and are defined as either CD8(+) CD28(-) or CD8(+) CD57(+) T lymphocytes. There is growing evidence that the CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell population plays a significant role in various diseases or conditions, associated with chronic immune activation such as cancer, chronic intracellular infections, chronic alcoholism, some chronic pulmonary diseases, autoimmune diseases, allogeneic transplantation, as well as has a great influence on age-related changes in the immune system status. CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell population is heterogeneous and composed of various functionally competing (cytotoxic and immunosuppressive) subsets thus the overall effect of CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell-mediated immunity depends on the predominance of a particular subset. Many articles claim that CD8(+) CD28(-) (CD8(+) CD57(+)) T cells have lost their proliferative capacity during process of replicative senescence triggered by repeated antigenic stimulation. However recent data indicate that CD8(+) CD28(-) (CD8(+) CD57(+)) T cells can transiently up-regulate telomerase activity and proliferate under certain stimulation conditions. Similarly, conflicting data is provided regarding CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell sensitivity to apoptosis, finally leading to the conclusion that this T-cell population is also heterogeneous in terms of its apoptotic potential. This review provides a comprehensive approach to the CD8(+) CD28(-) (CD8(+) CD57(+)) T-cell population: we describe in detail its origins, molecular and functional characteristics, subsets, role in various diseases or conditions, associated with persistent antigenic stimulation.
Collapse
Affiliation(s)
- Marius Strioga
- Laboratory of Immunology, Institute of Oncology, Vilnius University, Vilnius Faculty of Medicine, Vilnius University, Vilnius, Lithuania.
| | | | | |
Collapse
|
23
|
Fioravanti J, González I, Medina-Echeverz J, Larrea E, Ardaiz N, González-Aseguinolaza G, Prieto J, Berraondo P. Anchoring interferon alpha to apolipoprotein A-I reduces hematological toxicity while enhancing immunostimulatory properties. Hepatology 2011; 53:1864-73. [PMID: 21425312 DOI: 10.1002/hep.24306] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2010] [Accepted: 03/10/2011] [Indexed: 12/07/2022]
Abstract
UNLABELLED Interferon alpha (IFNα) is widely used for the treatment of viral hepatitis but substantial toxicity hampers its clinical use. In this work, we aimed at improving the efficacy of IFNα therapy by increasing the IFNα half-life and providing liver tropism. We selected apolipoprotein A-I (ApoA-I) as the stabilizing and targeting moiety. We generated plasmids encoding IFNα, albumin bound to IFNα (ALF), or IFNα linked to ApoA-I (IA) and mice were treated either by hydrodynamic administration of the plasmids or by injection of the corresponding recombinant proteins or high-density lipoproteins containing IA. The plasma half-life of IA was intermediate between IFNα and ALF. IA was targeted to the liver and induced higher hepatic expression of interferon-stimulated genes than IFNα or even ALF. IA exhibits stronger in vivo antiviral activity than IFNα and the hematologic cytopenic effects of IA are milder than those observed when using IFNα or ALF. In contrast to IFNα, IA does not cause activation-dependent cell death of lymphocytes in vitro. Accordingly, in vivo studies showed that IA boosts T-cell immune responses more efficiently than IFNα or ALF. The difference in immunostimulatory activity between IFNα and IA disappears in scavenger receptor class B type I (SR-BI) knockout mice, suggesting that crosstalk between SR-BI and IFNα receptor is essential for enhanced induction of cytotoxic T cells by IA. CONCLUSION Anchoring IFNα to ApoA-I prolongs the half-life of IFNα and promotes targeting to the liver. Importantly, the fusion protein shows increased immunostimulatory properties and lower hematological toxicity.
Collapse
Affiliation(s)
- Jessica Fioravanti
- Division of Hepatology and Gene Therapy, Center for Applied Medical Research, University of Navarra, Pamplona, Navarra, Spain
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Simons DL, Lee G, Kirkwood JM, Lee PP. Interferon signaling patterns in peripheral blood lymphocytes may predict clinical outcome after high-dose interferon therapy in melanoma patients. J Transl Med 2011; 9:52. [PMID: 21545749 PMCID: PMC3114759 DOI: 10.1186/1479-5876-9-52] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 05/05/2011] [Indexed: 02/08/2023] Open
Abstract
Background High-dose Interferon (HDI) therapy produces a clinical response and achieves relapse-free survival in 20-33% of patients with operable high risk or metastatic melanoma. However, patients may develop significant side effects frequently necessitating dose reduction or discontinuation of therapy. We recently showed that peripheral blood lymphocytes (PBL) from some melanoma patients have impaired interferon (IFN) signaling which could be restored with high concentrations of IFN. This exploratory study evaluated IFN signaling in PBL of melanoma patients to assess whether the restoration of PBL IFN signaling may predict a beneficial effect for HDI in melanoma patients. Methods PBL from 14 melanoma patients harvested on Day 0 and Day 29 of neoadjuvant HDI induction therapy were analyzed using phosflow to assess their interferon signaling patterns through IFN-α induced phosphorylation of STAT1-Y701. Results Patients who had a clinical response to HDI showed a lower PBL interferon signaling capacity than non-responders at baseline (Day 0). Additionally, clinical responders and patients with good long-term outcome showed a significant increase in their PBL interferon signaling from Day 0 to Day 29 compared to clinical non-responders and patients that developed metastatic disease. The differences in STAT1 activation from pre- to post- HDI treatment could distinguish between patients who were inclined to have a favorable or unfavorable outcome. Conclusion While the sample size is small, these results suggest that interferon signaling patterns in PBL correlate with clinical responses and may predict clinical outcome after HDI in patients with melanoma. A larger confirmatory study is warranted, which may yield a novel approach to select patients for HDI therapy.
Collapse
Affiliation(s)
- Diana L Simons
- Dept of Medicine, Stanford University, Stanford, CA, USA.
| | | | | | | |
Collapse
|
25
|
Changes in glucose and glutamine lymphocyte metabolisms induced by type I interferon α. Mediators Inflamm 2010; 2010:364290. [PMID: 21234393 PMCID: PMC3017935 DOI: 10.1155/2010/364290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Accepted: 12/08/2010] [Indexed: 01/16/2023] Open
Abstract
In lymphocytes (LY), the well-documented antiproliferative effects of IFN-α are associated with inhibition of protein synthesis, decreased amino acid incorporation, and cell cycle arrest. However, the effects of this cytokine on the metabolism of glucose and glutamine in these cells have not been well investigated. Thus, mesenteric and spleen LY of male Wistar rats were cultured in the presence or absence of IFN-α, and the changes on glucose and glutamine metabolisms were investigated. The reduced proliferation of mesenteric LY was accompanied by a reduction in glucose total consumption (35%), aerobic glucose metabolism (55%), maximal activity of glucose-6-phosphate dehydrogenase (49%), citrate synthase activity (34%), total glutamine consumption (30%), aerobic glutamine consumption (20.3%) and glutaminase activity (56%). In LY isolated from spleen, IFNα also reduced the proliferation and impaired metabolism. These data demonstrate that in LY, the antiproliferative effects of IFNα are associated with a reduction in glucose and glutamine metabolisms.
Collapse
|
26
|
Mattei F, Schiavoni G, Tough DF. Regulation of immune cell homeostasis by type I interferons. Cytokine Growth Factor Rev 2010; 21:227-36. [DOI: 10.1016/j.cytogfr.2010.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 04/29/2010] [Accepted: 05/07/2010] [Indexed: 12/16/2022]
|
27
|
Pilz A, Kratky W, Stockinger S, Simma O, Kalinke U, Lingnau K, von Gabain A, Stoiber D, Sexl V, Kolbe T, Rülicke T, Müller M, Decker T. Dendritic cells require STAT-1 phosphorylated at its transactivating domain for the induction of peptide-specific CTL. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 183:2286-93. [PMID: 19620292 DOI: 10.4049/jimmunol.0901383] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Phosphorylation of transcription factor STAT-1 on Y701 regulates subcellular localization whereas phosphorylation of the transactivating domain at S727 enhances transcriptional activity. In this study, we investigate the impact of STAT-1 and the importance of transactivating domain phosphorylation on the induction of peptide-specific CTL in presence of the TLR9-dependent immune adjuvant IC31. STAT-1 deficiency completely abolished CTL induction upon immunization, which was strongly reduced in animals carrying the mutation of the S727 phospho-acceptor site. A comparable reduction of CTL was found in mice lacking the type I IFN (IFN-I) receptor, whereas IFN-gamma-deficient mice behaved like wild-type controls. This finding suggests that S727-phosphorylated STAT-1 supports IFN-I-dependent induction of CTL. In adoptive transfer experiments, IFN-I- and S727-phosphorylated STAT-1 were critical for the activation and function of dendritic cells. Mice with a T cell-specific IFN-I receptor ablation did not show impaired CTL responses. Unlike the situation observed for CTL development S727-phosphorylated STAT-1 restrained proliferation of naive CD8(+) T cells both in vitro and following transfer into Rag-deficient mice. In summary, our data reveal a dual role of S727-phosphorylated STAT-1 for dendritic cell maturation as a prerequisite for the induction of CTL activity and for T cell autonomous control of activation-induced or homeostatic proliferation.
Collapse
Affiliation(s)
- Andreas Pilz
- Department of Genetics, Max F Perutz Laboratories, University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Receptor density is key to the alpha2/beta interferon differential activities. Mol Cell Biol 2009; 29:4778-87. [PMID: 19564411 DOI: 10.1128/mcb.01808-08] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Multiple type I interferons (IFN-alpha/beta) elicit Jak/Stat activation, rapid gene induction, and pleiotropic effects, such as differentiation, antiviral protection, and blocks in proliferation, which are dependent on the IFN subtype and the cellular context. To date, ligand- and receptor-specific molecular determinants underlying IFN-alpha/beta differential activities or potencies have been well characterized. To analyze cellular determinants that impact subtype-specific potency, human fibrosarcoma U5A-derived clones, exhibiting a gradient of IFN sensitivity by virtue of increasing receptor levels, were monitored for Jak/Stat signaling, gene induction, cell cycle lengthening, and apoptosis. In cells with scarce receptors, IFN-beta was more potent than IFN-alpha2 in antiproliferative activity, while the two subtypes were equipotent in all other readouts. Conversely, in cells with abundant receptors, IFN-alpha2 matched or even surpassed IFN-beta in all readouts tested. Our results suggest that the differential activities of the IFN subtypes are dictated not only by the intrinsic ligand/receptor binding kinetics but also by the density of cell surface receptor components.
Collapse
|
29
|
Impaired interferon signaling is a common immune defect in human cancer. Proc Natl Acad Sci U S A 2009; 106:9010-5. [PMID: 19451644 DOI: 10.1073/pnas.0901329106] [Citation(s) in RCA: 197] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Immune dysfunction develops in patients with many cancer types and may contribute to tumor progression and failure of immunotherapy. Mechanisms underlying cancer-associated immune dysfunction are not fully understood. Efficient IFN signaling is critical to lymphocyte function; animals rendered deficient in IFN signaling develop cancer at higher rates. We hypothesized that altered IFN signaling may be a key mechanism of immune dysfunction common to cancer. To address this, we assessed the functional responses to IFN in peripheral blood lymphocytes from patients with 3 major cancers: breast cancer, melanoma, and gastrointestinal cancer. Type-I IFN (IFN-alpha)-induced signaling was reduced in T cells and B cells from all 3 cancer-patient groups compared to healthy controls. Type-II IFN (IFN-gamma)-induced signaling was reduced in B cells from all 3 cancer patient groups, but not in T cells or natural killer cells. Impaired-IFN signaling was equally evident in stage II, III, and IV breast cancer patients, and downstream functional defects in T cell activation were identified. Taken together, these findings indicate that defects in lymphocyte IFN signaling arise in patients with breast cancer, melanoma, and gastrointestinal cancer, and these defects may represent a common cancer-associated mechanism of immune dysfunction.
Collapse
|
30
|
Démoulins T, Abdallah A, Kettaf N, Baron ML, Gerarduzzi C, Gauchat D, Gratton S, Sékaly RP. Reversible blockade of thymic output: an inherent part of TLR ligand-mediated immune response. THE JOURNAL OF IMMUNOLOGY 2008; 181:6757-69. [PMID: 18981093 DOI: 10.4049/jimmunol.181.10.6757] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
TLRs constitute a first set of sensors that detect viral nucleic acids including dsRNA which triggers TLR3. We report the early, direct, and detrimental effect of polyinosine-polycytidilic acid treatment on T cell development. Inhibition of thymopoiesis was targeted to several thymocyte subpopulations. First, both a blockade of the double negative (DN)1-DN2 transition and a severe down-regulation of DN3-DN4 thymocyte proliferation were observed. In addition, an important decrease in the absolute numbers of double-positive thymocytes, concomitant with an increase in frequencies of apoptotic cells in this population were shown. This inhibition of thymopoiesis resulted in a reduced thymic output, as evidenced by a drop of the absolute numbers of naive T cells and TCR excision circles levels. The decrease in thymic cellularity and defects in thymic development were severely reduced, but not completely abolished in IFN-alpha/betaR(-/-) mice, showing a direct contribution of type I IFNs, known to be massively up-regulated in viral infections, to the inhibition of T cell development. Strikingly, the TCR repertoire in treated mice was biased toward shorter CDR3 lengths as a result of a decreased expression of TdT and Rag2. However, thymic integrity remained intact since thymopoiesis was restored both quantitatively and qualitatively 14 days after the cessation of polyinosine-polycytidilic acid treatment. These results demonstrate a novel immunomodulatory role for virally encoded TLR ligands and RNA sensors; they further illustrate the diversity of mechanisms that viruses use to interfere with the development of a pathogen-specific immune responses.
Collapse
Affiliation(s)
- Thomas Démoulins
- Laboratoire d'Immunologie, Centre de Recherches du Centre Hospitalier de l'Université de Montréal, Saint-Luc, Canada
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Kocic G, Bjelakovic G, Pavlovic D, Jevtovic T, Pavlovic V, Sokolovic D, Basic J, Cekic S, Cvetkovic T, Kocic R, Stojanovic S. Protective effect of interferon-alpha on the DNA- and RNA-degrading pathway in anti-Fas-antibody induced apoptosis. Hepatol Res 2007; 37:637-46. [PMID: 17517072 DOI: 10.1111/j.1872-034x.2007.00090.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
AIM Fas membrane-associated polypeptide antigen is a receptor molecule responsible for apoptosis-mediated signals. In animal models of acute viral hepatitis, apoptosis of hepatocytes is mediated by Fas-death receptors; therefore, the aim of this study was to evaluate the effect of interferon (IFN)-alpha on apoptotic markers and nuclease activity against different coding and non-coding single and double stranded RNAs during Fas-induced liver apoptosis. METHODS An in vivo experiment was performed with simultaneous administration of anti-Fas (CD95) antibodies and IFN-alpha, and an in vitro experiment was performed in hepatocyte cultures treated with anti-Fas antibodies and IFN-alpha. RESULTS Detection of apoptosis using Annexin V-FITC/propidium iodide, Bcl-2 and Bax expression in hepatocyte cultures confirmed the appearance of early apoptotic events and progression toward late apoptosis after anti-Fas antibody treatment. IFN-alpha had a tendency to retard the apoptosis process in Fas-induced apoptosis by increasing the number of viable cells and decreasing the number of cells in late apoptosis, by increasing the percentage of Bcl-2 positive cells, by decreasing the percentage of Bax positive cells, and by decreasing the nuclease activity compared to the anti-Fas antibody treated group. Total DNA and RNA concentration was much reduced in the Fas group and DNA fragmentation assay provided evidence for increased DNA degradation. Enhanced nuclease activity against DNA, rRNA, poly(A), poly(C), poly(U), poly(I:C), and poly(A:U) was manifested in the anti-Fas antibody treated group, except for the inhibitory-bound alkaline RNase. CONCLUSIONS The results demonstrate that the RNA-degrading pathway in Fas-induced apoptosis can accelerate the liberation of the latent enzyme from the inhibitor complex. IFN-alpha prevented enormous, Fas-ligand induced degradation of all the substrates used in this experimental study, most probably due to similarities in the signal transduction pathways. Investigations of death receptor-induced apoptosis may lead to novel treatment combinations for patients with acute or chronic liver diseases.
Collapse
Affiliation(s)
- Gordana Kocic
- Institute of Biochemistry and Institute of Physiology, Medical Faculty, University of Nis, Serbia and Montenegro
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Djavani MM, Crasta OR, Zapata JC, Fei Z, Folkerts O, Sobral B, Swindells M, Bryant J, Davis H, Pauza CD, Lukashevich IS, Hammamieh R, Jett M, Salvato MS. Early blood profiles of virus infection in a monkey model for Lassa fever. J Virol 2007; 81:7960-73. [PMID: 17522210 PMCID: PMC1951294 DOI: 10.1128/jvi.00536-07] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Acute arenavirus disease in primates, like Lassa hemorrhagic fever in humans, begins with flu-like symptoms and leads to death approximately 2 weeks after infection. Our goal was to identify molecular changes in blood that are related to disease progression. Rhesus macaques (Macaca mulatta) infected intravenously with a lethal dose of lymphocytic choriomeningitis virus (LCMV) provide a model for Lassa virus infection of humans. Blood samples taken before and during the course of infection were used to monitor gene expression changes that paralleled disease onset. Changes in blood showed major disruptions in eicosanoid, immune response, and hormone response pathways. Approximately 12% of host genes alter their expression after LCMV infection, and a subset of these genes can discriminate between virulent and non-virulent LCMV infection. Major transcription changes have been given preliminary confirmation by quantitative PCR and protein studies and will be valuable candidates for future validation as biomarkers for arenavirus disease.
Collapse
Affiliation(s)
- Mahmoud M Djavani
- Institute of Human Virology, University of Maryland Biotechnology Institute, 725 West Lombard St., Baltimore, MD 21201, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Critchley-Thorne RJ, Yan N, Nacu S, Weber J, Holmes SP, Lee PP. Down-regulation of the interferon signaling pathway in T lymphocytes from patients with metastatic melanoma. PLoS Med 2007; 4:e176. [PMID: 17488182 PMCID: PMC1865558 DOI: 10.1371/journal.pmed.0040176] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Accepted: 03/26/2007] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Dysfunction of the immune system has been documented in many types of cancers. The precise nature and molecular basis of immune dysfunction in the cancer state are not well defined. METHODS AND FINDINGS To gain insights into the molecular mechanisms of immune dysfunction in cancer, gene expression profiles of pure sorted peripheral blood lymphocytes from 12 patients with melanoma were compared to 12 healthy controls. Of 25 significantly altered genes in T cells and B cells from melanoma patients, 17 are interferon (IFN)-stimulated genes. These microarray findings were further confirmed by quantitative PCR and functional responses to IFNs. The median percentage of lymphocytes that phosphorylate STAT1 in response to interferon-alpha was significantly reduced (Delta = 16.8%; 95% confidence interval, 0.98% to 33.35%) in melanoma patients (n = 9) compared to healthy controls (n = 9) in Phosflow analysis. The Phosflow results also identified two subgroups of patients with melanoma: IFN-responsive (33%) and low-IFN-response (66%). The defect in IFN signaling in the melanoma patient group as a whole was partially overcome at the level of expression of IFN-stimulated genes by prolonged stimulation with the high concentration of IFN-alpha that is achievable only in IFN therapy used in melanoma. The lowest responders to IFN-alpha in the Phosflow assay also showed the lowest gene expression in response to IFN-alpha. Finally, T cells from low-IFN-response patients exhibited functional abnormalities, including decreased expression of activation markers CD69, CD25, and CD71; TH1 cytokines interleukin-2, IFN-gamma, and tumor necrosis factor alpha, and reduced survival following stimulation with anti-CD3/CD28 antibodies compared to controls. CONCLUSIONS Defects in interferon signaling represent novel, dominant mechanisms of immune dysfunction in cancer. These findings may be used to design therapies to counteract immune dysfunction in melanoma and to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Rebecca J Critchley-Thorne
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California, United States of America
| | - Ning Yan
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California, United States of America
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Serban Nacu
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Jeffrey Weber
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Peter P Lee
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California, United States of America
| |
Collapse
|
34
|
Herbeuval JP, Shearer GM. HIV-1 immunopathogenesis: how good interferon turns bad. Clin Immunol 2007; 123:121-8. [PMID: 17112786 PMCID: PMC1930161 DOI: 10.1016/j.clim.2006.09.016] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Accepted: 09/27/2006] [Indexed: 01/29/2023]
Abstract
The hallmark of acquired immunodeficiency syndrome (AIDS) is the progressive loss of CD4+ T cells that results from infection with human immunodeficiency virus type-1 (HIV-1). Despite 25 years of AIDS research, questions remain concerning the mechanisms responsible for HIV-induced CD4+ T cell depletion. Here we briefly review the in vitro and in vivo literature concerning the protective role of interferon-alpha (IFN-alpha) in HIV/AIDS. We then develop a laboratory- and clinically supported model of CD4+ T cell apoptosis in which either infectious or noninfectious HIV-1 induces the production of type I interferon by plasmacytoid dendritic cells (pDC). The interferon produced binds to its receptor on primary CD4+ T cells resulting in membrane expression of the TNF-related apoptosis-inducing ligand (TRAIL) death molecule. The binding of infectious or noninfectious HIV-1 to CD4 on these T cells results in expression of the TRAIL death receptor 5 (DR5), leading to the selective death of HIV-exposed CD4+ T cells.
Collapse
|
35
|
Wiegand J, Cornberg M, Aslan N, Schlaphoff V, Sarrazin C, Kubitschke A, Buggisch P, Ciner A, Jaeckel E, Manns MP, Wedemeyer H. Fate and Function of Hepatitis-C-Virus-Specific T-Cells during Peginterferon-α2b therapy for Acute Hepatitis C. Antivir Ther 2007. [DOI: 10.1177/135965350701200306] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Strong hepatitis C virus (HCV)-specific T-cell responses are associated with spontaneous clearance of acute hepatitis C. However, recent studies described a decline in HCV-specific CD8+ T-cells during interferon treatment, suggesting that the success of acute HCV therapy might be independent of adaptive immunity. Methods T-cell responses of eight human leukocyte antigen (HLA)-A2-positive, acutely infected patients treated with peginterferon-α2b were studied by ELISPOT and proliferation assays and flow cytometry analysis using HCV-specific tetramers. Results HCV-specific T-cells predominately declined during therapy. However, diverse patterns of CD4+ and CD8+ T-cell kinetics were observed. In patients with sustained virological response chemokine receptor 3 (CXCR-3) expression of HCV-specific CD8+ T-cells was upregulated, indicating homing to the liver. Low levels of T-cells remained detectable throughout treatment and follow up. In contrast, T-cells of a relapse patient did not upregulate CXCR-3 but displayed a higher staining for annexin-V, followed by a complete loss of peripheral virus-specific CD8+ T-cells by week 12. Conclusions Kinetics of HCV-specific T-cell responses are heterogeneous in interferon-treated patients with acute hepatitis C. The decline of T-cells might be a consequence of both apoptosis and homing. The balance between cell death and regulation of chemokine receptors might lead to different long-term outcomes.
Collapse
Affiliation(s)
- Johannes Wiegand
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Nuray Aslan
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Verena Schlaphoff
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Christoph Sarrazin
- Saarland University Hospital, Klinik für Innere Medizin II, Homburg/Saar, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Anne Kubitschke
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Peter Buggisch
- Universitätsklinikum Eppendorf, University of Hamburg, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Ayse Ciner
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Michael P Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Germany
- German Network of Competence on Viral Hepatitis (HepNet)
| |
Collapse
|
36
|
Rodriguez B, Lederman MM, Jiang W, Bazdar DA, Gàrate K, Harding CV, Sieg SF. Interferon-alpha differentially rescues CD4 and CD8 T cells from apoptosis in HIV infection. AIDS 2006; 20:1379-89. [PMID: 16791012 DOI: 10.1097/01.aids.0000233571.51899.ab] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To examine the effects of interferon-alpha (IFN-alpha) on T cell survival and activation in HIV infection. DESIGN The effects of IFN-alpha on spontaneous apoptosis and CD38 expression among T cell subsets were determined in vitro and studied in relation to CD4 cell counts, plasma HIV RNA levels and the age of the subjects. METHODS Peripheral blood mononuclear cells from 48 HIV-infected persons and 17 healthy donors were incubated in vitro overnight with or without the addition of IFN-alpha. Percentages of apoptotic cells (positive for annexin V) and CD38 cells were determined among T cell subsets. RESULTS IFN-alpha inhibited spontaneous apoptosis of CD4 and CD8 T lymphocytes. This protective activity was impaired in CD4 T cells from HIV-infected persons. The reduced protection of IFN-alpha among CD4 cells from HIV-infected persons was not related to the percentages of activated (CD38 or CD45RO+CD38+) cells. Surprisingly, IFN-alpha induced CD38 expression among CD8 T cells from HIV-infected persons, and the magnitude of this effect was directly related to circulating CD4 T cell count. The CD8 T cell subset that expressed CD38 in response to IFN-alpha was defined as CD28 negative, CD62 ligand (CD62L) intermediate/negative. CONCLUSIONS Heightened expression of IFN-alpha in HIV infection may contribute to the phenotypic activation state that characterizes chronic infection while a diminished responsiveness of CD4 T cells to the protective effect of this cytokine may contribute to differential survival of CD4 and CD8 T cells in HIV disease.
Collapse
Affiliation(s)
- Benigno Rodriguez
- Division of Infectious Diseases, Case Western Reserve University, Center foe AIDS Research, University Hospitals of Cleveland, Cleveland, Ohio, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Herbeuval JP, Nilsson J, Boasso A, Hardy AW, Kruhlak MJ, Anderson SA, Dolan MJ, Dy M, Andersson J, Shearer GM. Differential expression of IFN-alpha and TRAIL/DR5 in lymphoid tissue of progressor versus nonprogressor HIV-1-infected patients. Proc Natl Acad Sci U S A 2006; 103:7000-5. [PMID: 16632604 PMCID: PMC1444883 DOI: 10.1073/pnas.0600363103] [Citation(s) in RCA: 153] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2006] [Indexed: 12/16/2022] Open
Abstract
Loss of CD4+ T cells, the hallmark of HIV pathogenesis, was suggested to be partly due to apoptosis. We recently reported that IFN-alpha produced by HIV-1-activated plasmacytoid dendritic cells (pDCs) contributes to CD4+ T cell apoptosis by the TNF-related apoptosis-inducing ligand (TRAIL)/death receptor (DR)5 pathway. Here, we show that HIV-1-induced intracellular expression of IFN-alpha in pDCs is coupled to increased expression of IFN regulatory factor 7 and MyD88 by pDCs in vivo and in vitro. Expression of IFN-alpha was increased in lymphoid tonsillar tissue (LT) of patients with progressive (HIV(prog)) compared with nonprogressive (HIV(NP)) HIV-1 disease and to uninfected controls. LT from HIV(prog) exhibited higher TRAIL and DR5 mRNA levels than LT from HIV(NP) or controls. TRAIL mRNA levels in LT correlated with plasma viral load. We show that HIV-1 induces IFN-alpha and the TRAIL/DR5 apoptotic pathway in LT, suggesting a role for these cytokines in HIV-1 immunopathogenesis.
Collapse
Affiliation(s)
- Jean-Philippe Herbeuval
- *Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20092
| | - Jakob Nilsson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital, SE-171 77 Stockholm, Sweden
| | - Adriano Boasso
- *Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20092
| | - Andrew W. Hardy
- *Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20092
| | - Michael J. Kruhlak
- *Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20092
| | | | - Matthew J. Dolan
- Infectious Diseases Service, Wilford Hall Medical Center, Lackland Air Force Base, Defense Institute for Medical Operations, Brooks City-Base, TX 78235; and
| | - Michel Dy
- Centre National pour la Recherche Scientifique, Unité Mixte Recherche 8147, Faculté de Médecine René Descartes Paris V, 75270 Paris Cedex 06, France
| | - Jan Andersson
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital, SE-171 77 Stockholm, Sweden
| | - Gene M. Shearer
- *Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20092
- Center for Infectious Medicine, Department of Medicine, Karolinska Institute, Karolinska University Hospital, SE-171 77 Stockholm, Sweden
| |
Collapse
|
38
|
Chi B, Dickensheets HL, Spann KM, Alston MA, Luongo C, Dumoutier L, Huang J, Renauld JC, Kotenko SV, Roederer M, Beeler JA, Donnelly RP, Collins PL, Rabin RL. Alpha and lambda interferon together mediate suppression of CD4 T cells induced by respiratory syncytial virus. J Virol 2006; 80:5032-40. [PMID: 16641294 PMCID: PMC1472058 DOI: 10.1128/jvi.80.10.5032-5040.2006] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Accepted: 02/21/2006] [Indexed: 12/15/2022] Open
Abstract
The mechanism by which respiratory syncytial virus (RSV) suppresses T-cell proliferation to itself and other antigens is poorly understood. We used monocyte-derived dendritic cells (MDDC) and CD4 T cells and measured [(3)H]thymidine incorporation to determine the factors responsible for RSV-induced T-cell suppression. These two cell types were sufficient for RSV-induced suppression of T-cell proliferation in response to cytomegalovirus or Staphylococcus enterotoxin B. Suppressive activity was transferable with supernatants from RSV-infected MDDC and was not due to transfer of live virus or RSV F (fusion) protein. Supernatants from RSV-infected MDDC, but not MDDC exposed to UV-killed RSV or mock conditions, contained alpha interferon (IFN-alpha; median, 43 pg/ml) and IFN-lambda (approximately 1 to 20 ng/ml). Neutralization of IFN-alpha with monoclonal antibody (MAb) against one of its receptor chains, IFNAR2, or of IFN-lambda with MAb against either of its receptor chains, IFN-lambdaR1 (interleukin 28R [IL-28R]) or IL-10R2, had a modest effect. In contrast, blocking the two receptors together markedly reduced or completely blocked the RSV-induced suppression of CD4 T-cell proliferation. Defining the mechanism of RSV-induced suppression may guide vaccine design and provide insight into previously uncharacterized human T-cell responses and activities of interferons.
Collapse
Affiliation(s)
- Bo Chi
- Center for Biologics Evaluation and Research, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Ordway D, Henao-Tamayo M, Orme IM, Gonzalez-Juarrero M. Foamy macrophages within lung granulomas of mice infected with Mycobacterium tuberculosis express molecules characteristic of dendritic cells and antiapoptotic markers of the TNF receptor-associated factor family. THE JOURNAL OF IMMUNOLOGY 2005; 175:3873-81. [PMID: 16148133 DOI: 10.4049/jimmunol.175.6.3873] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Highly vacuolated or foamy macrophages are a distinct characteristic of granulomas in the lungs of animals infected with Mycobacterium tuberculosis. To date these have usually been considered to represent activated macrophages derived from monocytes entering the lesions from the blood. However, we demonstrate in this study that foamy macrophages express high levels of DEC-205, a marker characteristic of dendritic cells (DCs). In addition to high expression of the DEC-205 marker, these cells were characterized as CD11b(+)CD11c(high)MHC class II(high), and CD40(high), which are additional markers typically expressed by DCs. Up-regulation of CD40 was seen only during the early chronic stage of the lung disease, and both the expression of CD40 and MHC class II markers were down-regulated as the disease progressed into the late chronic phase. Foamy cells positive for the DEC-205 marker also expressed high levels of TNFR-associated factor-1 (TRAF-1), TRAF-2, and TRAF-3, markers associated with resistance to apoptosis. These data indicate that in addition to the central role of DCs in initiating the acquired immune response against M. tuberculosis infection, they also participate in the granulomatous response.
Collapse
Affiliation(s)
- Diane Ordway
- Mycobacteria Research Laboratories, Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO 80523, USA.
| | | | | | | |
Collapse
|
40
|
Kawamura K, Kadowaki N, Kitawaki T, Uchiyama T. Virus-stimulated plasmacytoid dendritic cells induce CD4+ cytotoxic regulatory T cells. Blood 2005; 107:1031-8. [PMID: 16219801 DOI: 10.1182/blood-2005-04-1737] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Immune responses to pathogens need to be maintained within appropriate levels to minimize tissue damage, whereas such controlled immunity may allow persistent infection of certain types of pathogens. Interleukin 10 (IL-10) plays an important role in such immune regulation. We previously showed that HSV-stimulated human plasmacytoid dendritic cells (pDCs) induced naive CD4+ T cells to differentiate into interferon gamma (IFN-gamma)/IL-10-producing T cells. Here we show that HSV-stimulated pDCs induce allogeneic naive CD4+ T cells to differentiate into cytotoxic regulatory T cells that poorly proliferate on restimulation and inhibit proliferation of coexisting naive CD4+ T cells. IL-3-stimulated pDCs or myeloid DCs did not induce such regulatory T cells. Both IFN-alpha and IL-10 were responsible for the induction of anergic and regulatory properties. High percentages of CD4+ T cells cocultured with HSV-stimulated pDCs, and to a lesser extent those cocultured with IL-3-stimulated pDCs, expressed granzyme B and perforin in an IL-10-dependent manner. CD4+ T cells cocultured with HSV-stimulated pDCs accordingly exhibited cytotoxic activity. The finding that virus-stimulated pDCs are capable of inducing CD4+ cytotoxic regulatory T cells suggests that this DC subset may play an important role in suppressing excessive inflammatory responses and also in inducing persistent viral infection.
Collapse
Affiliation(s)
- Kazuko Kawamura
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | |
Collapse
|