1
|
Bai Z, Feng B, McClory SE, de Oliveira BC, Diorio C, Gregoire C, Tao B, Yang L, Zhao Z, Peng L, Sferruzza G, Zhou L, Zhou X, Kerr J, Baysoy A, Su G, Yang M, Camara PG, Chen S, Tang L, June CH, Melenhorst JJ, Grupp SA, Fan R. Single-cell CAR T atlas reveals type 2 function in 8-year leukaemia remission. Nature 2024; 634:702-711. [PMID: 39322664 PMCID: PMC11485231 DOI: 10.1038/s41586-024-07762-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 06/27/2024] [Indexed: 09/27/2024]
Abstract
Despite a high response rate in chimeric antigen receptor (CAR) T cell therapy for acute lymphocytic leukaemia (ALL)1-3, approximately 50% of patients relapse within the first year4-6, representing an urgent question to address in the next stage of cellular immunotherapy. Here, to investigate the molecular determinants of ultralong CAR T cell persistence, we obtained a single-cell multi-omics atlas from 695,819 pre-infusion CAR T cells at the basal level or after CAR-specific stimulation from 82 paediatric patients with ALL enrolled in the first two CAR T ALL clinical trials and 6 healthy donors. We identified that elevated type 2 functionality in CAR T infusion products is significantly associated with patients maintaining a median B cell aplasia duration of 8.4 years. Analysis of ligand-receptor interactions revealed that type 2 cells regulate a dysfunctional subset to maintain whole-population homeostasis, and the addition of IL-4 during antigen-specific activation alleviates CAR T cell dysfunction while enhancing fitness at both transcriptomic and epigenomic levels. Serial proteomic profiling of sera after treatment revealed a higher level of circulating type 2 cytokines in 5-year or 8-year relapse-free responders. In a leukaemic mouse model, type 2high CAR T cell products demonstrated superior expansion and antitumour activity, particularly after leukaemia rechallenge. Restoring antitumour efficacy in type 2low CAR T cells was attainable by enhancing their type 2 functionality, either through incorporating IL-4 into the manufacturing process or by priming manufactured CAR T products with IL-4 before infusion. Our findings provide insights into the mediators of durable CAR T therapy response and suggest potential therapeutic strategies to sustain long-term remission by boosting type 2 functionality in CAR T cells.
Collapse
Affiliation(s)
- Zhiliang Bai
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Bing Feng
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland
| | - Susan E McClory
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Caroline Diorio
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Céline Gregoire
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Bo Tao
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Luojia Yang
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Ziran Zhao
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lei Peng
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Giacomo Sferruzza
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Liqun Zhou
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaolei Zhou
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland
| | - Jessica Kerr
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alev Baysoy
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Graham Su
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Mingyu Yang
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Pablo G Camara
- Department of Genetics and Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Li Tang
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- Institute of Materials Science & Engineering, EPFL, Lausanne, Switzerland.
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Parker Institute for Cancer Immunotherapy at University of Pennsylvania, Philadelphia, PA, USA.
| | | | - Stephan A Grupp
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.
- Human and Translational Immunology, Yale University School of Medicine, New Haven, CT, USA.
- Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Kotliar D, Curtis M, Agnew R, Weinand K, Nathan A, Baglaenko Y, Zhao Y, Sabeti PC, Rao DA, Raychaudhuri S. Reproducible single cell annotation of programs underlying T-cell subsets, activation states, and functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592310. [PMID: 38746317 PMCID: PMC11092745 DOI: 10.1101/2024.05.03.592310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
T-cells recognize antigens and induce specialized gene expression programs (GEPs) enabling functions including proliferation, cytotoxicity, and cytokine production. Traditionally, different classes of helper T-cells express mutually exclusive responses - for example, Th1, Th2, and Th17 programs. However, new single-cell RNA sequencing (scRNA-Seq) experiments have revealed a continuum of T-cell states without discrete clusters corresponding to these subsets, implying the need for new analytical frameworks. Here, we advance the characterization of T-cells with T-CellAnnoTator (TCAT), a pipeline that simultaneously quantifies pre-defined GEPs capturing activation states and cellular subsets. From 1,700,000 T-cells from 700 individuals across 38 tissues and five diverse disease contexts, we discover 46 reproducible GEPs reflecting the known core functions of T-cells including proliferation, cytotoxicity, exhaustion, and T helper effector states. We experimentally characterize several novel activation programs and apply TCAT to describe T-cell activation and exhaustion in Covid-19 and cancer, providing insight into T-cell function in these diseases.
Collapse
Affiliation(s)
- Dylan Kotliar
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Harvard-MIT Division of Health Sciences and Technology, Harvard Medical School, Boston, MA 02115, USA
| | - Michelle Curtis
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ryan Agnew
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kathryn Weinand
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Yuriy Baglaenko
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Center for Autoimmune Genetics and Etiology and Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45219, USA
| | - Yu Zhao
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Pardis C. Sabeti
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Organismic and Evolutionary Biology, FAS Center for Systems Biology, Harvard University, Cambridge, MA 02138, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Deepak A. Rao
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
3
|
Canderan G, Muehling LM, Kadl A, Ladd S, Bonham C, Cross CE, Lima SM, Yin X, Sturek JM, Wilson JM, Keshavarz B, Bryant N, Murphy DD, Cheon IS, McNamara CA, Sun J, Utz PJ, Dolatshahi S, Irish JM, Woodfolk JA. Distinct Type 1 Immune Networks Underlie the Severity of Restrictive Lung Disease after COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.03.587929. [PMID: 38617217 PMCID: PMC11014603 DOI: 10.1101/2024.04.03.587929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The variable etiology of persistent breathlessness after COVID-19 have confounded efforts to decipher the immunopathology of lung sequelae. Here, we analyzed hundreds of cellular and molecular features in the context of discrete pulmonary phenotypes to define the systemic immune landscape of post-COVID lung disease. Cluster analysis of lung physiology measures highlighted two phenotypes of restrictive lung disease that differed by their impaired diffusion and severity of fibrosis. Machine learning revealed marked CCR5+CD95+ CD8+ T-cell perturbations in mild-to-moderate lung disease, but attenuated T-cell responses hallmarked by elevated CXCL13 in more severe disease. Distinct sets of cells, mediators, and autoantibodies distinguished each restrictive phenotype, and differed from those of patients without significant lung involvement. These differences were reflected in divergent T-cell-based type 1 networks according to severity of lung disease. Our findings, which provide an immunological basis for active lung injury versus advanced disease after COVID-19, might offer new targets for treatment.
Collapse
|
4
|
Chen DG, Xie J, Choi J, Ng RH, Zhang R, Li S, Edmark R, Zheng H, Solomon B, Campbell KM, Medina E, Ribas A, Khatri P, Lanier LL, Mease PJ, Goldman JD, Su Y, Heath JR. Integrative systems biology reveals NKG2A-biased immune responses correlate with protection in infectious disease, autoimmune disease, and cancer. Cell Rep 2024; 43:113872. [PMID: 38427562 PMCID: PMC10995767 DOI: 10.1016/j.celrep.2024.113872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/19/2024] [Accepted: 02/09/2024] [Indexed: 03/03/2024] Open
Abstract
Infection, autoimmunity, and cancer are principal human health challenges of the 21st century. Often regarded as distinct ends of the immunological spectrum, recent studies hint at potential overlap between these diseases. For example, inflammation can be pathogenic in infection and autoimmunity. T resident memory (TRM) cells can be beneficial in infection and cancer. However, these findings are limited by size and scope; exact immunological factors shared across diseases remain elusive. Here, we integrate large-scale deeply clinically and biologically phenotyped human cohorts of 526 patients with infection, 162 with lupus, and 11,180 with cancer. We identify an NKG2A+ immune bias as associative with protection against disease severity, mortality, and autoimmune/post-acute chronic disease. We reveal that NKG2A+ CD8+ T cells correlate with reduced inflammation and increased humoral immunity and that they resemble TRM cells. Our results suggest NKG2A+ biases as a cross-disease factor of protection, supporting suggestions of immunological overlap between infection, autoimmunity, and cancer.
Collapse
Affiliation(s)
- Daniel G Chen
- Institute of Systems Biology, Seattle, WA, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jingyi Xie
- Institute of Systems Biology, Seattle, WA, USA; Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, USA
| | | | - Rachel H Ng
- Institute of Systems Biology, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Rongyu Zhang
- Institute of Systems Biology, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Sarah Li
- Institute of Systems Biology, Seattle, WA, USA
| | - Rick Edmark
- Institute of Systems Biology, Seattle, WA, USA
| | - Hong Zheng
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ben Solomon
- Department of Pediatrics, Division of Allergy and Immunology, Stanford School of Medicine, Stanford, CA, USA
| | - Katie M Campbell
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Egmidio Medina
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Antoni Ribas
- Department of Medicine, Division of Hematology-Oncology, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, School of Medicine, Stanford University, Stanford, CA, USA; Center for Biomedical Informatics Research, Department of Medicine, School of Medicine, Stanford University, Stanford, CA, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Philip J Mease
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA, USA; Providence St. Joseph Health, Renton, WA, USA
| | - Jason D Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA, USA; Providence St. Joseph Health, Renton, WA, USA; Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Yapeng Su
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - James R Heath
- Institute of Systems Biology, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
5
|
Mensink M, Verleng LJ, Schrama E, Janssen GM, Tjokrodirijo RT, van Veelen PA, Jiang Q, Pascutti MF, van der Hoorn ML, Eikmans M, de Kivit S, Borst J. Tregs from human blood differentiate into nonlymphoid tissue-resident effector cells upon TNFR2 costimulation. JCI Insight 2024; 9:e172942. [PMID: 38341270 PMCID: PMC10972588 DOI: 10.1172/jci.insight.172942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Tregs can facilitate transplant tolerance and attenuate autoimmune and inflammatory diseases. Therefore, it is clinically relevant to stimulate Treg expansion and function in vivo and to create therapeutic Treg products in vitro. We report that TNF receptor 2 (TNFR2) is a unique costimulus for naive, thymus-derived Tregs (tTregs) from human blood that promotes their differentiation into nonlymphoid tissue-resident (NLT-resident) effector Tregs, without Th-like polarization. In contrast, CD28 costimulation maintains a lymphoid tissue-resident (LT-resident) Treg phenotype. We base this conclusion on transcriptome and proteome analysis of TNFR2- and CD28-costimulated CD4+ tTregs and conventional T cells (Tconvs), followed by bioinformatic comparison with published transcriptomic Treg signatures from NLT and LT in health and disease, including autoimmunity and cancer. These analyses illuminate that TNFR2 costimulation promoted tTreg capacity for survival, migration, immunosuppression, and tissue regeneration. Functional studies confirmed improved migratory ability of TNFR2-costimulated tTregs. Flow cytometry validated the presence of the TNFR2-driven tTreg signature in effector/memory Tregs from the human placenta, as opposed to blood. Thus, TNFR2 can be exploited as a driver of NLT-resident tTreg differentiation for adoptive cell therapy or antibody-based immunomodulation in human disease.
Collapse
|
6
|
Jaxybayeva I, Boranbayeva R, Bulegenova M, Urazalieva N, Gerein V, Manzhuova L. Long-term outcomes and immune profiling in children with multisystem inflammatory syndrome (MIS-C). ACTA BIO-MEDICA : ATENEI PARMENSIS 2023; 94:e2023233. [PMID: 38054683 PMCID: PMC10734220 DOI: 10.23750/abm.v94i6.14788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/08/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND AND AIM Existing follow-up data after MIS-C is limited. PURPOSE OF THE STUDY to investigate the long-term consequences in children who have undergone MIS-C. METHODS The retrospective study included 93 children. The identified changes were divided into the following periods: occurred within first 6 months, 1 year, 2 years, and more than 2 years after MIS-C. Besides, 31 children underwent prospective immunophenotyping of peripheral blood and the determination of cytokines during the acute period of the disease and after discharge. RESULTS Outpatient monitoring events included pneumonia (9.6%), somatic disorder syndrome (11.8%), visual impairment (7.5%), joint damage (6.6%), weight changes (2.2%), and MIS-C recurrence (2.2%). A study of the cardiovascular system showed a statistically significant decrease in the frequency of the right and left heart dilatation, left ventricular dysfunction, pericarditis, pulmonary arterial hypertension, coronaritis, mitral regurgitation. But at the same time an increase in pulmonary and tricuspid valve regurgitation and arrhythmias compared with the acute period was detected. Most of the changes took place within first year of observation. Immune profiling showed reconstitution of CD3, CD4 T-lymphocytes, NK-cells, maintenance of a high relative value of CD8, reduction of CD19+ B-cells, expression of CD3-HLA-DR+, CD25, CD279, CD95. CONCLUSIONS After the history of MIS-C, children in the long-term follow-up had various somatic disorders and disease recurrence. Most patients (64.1%) showed subclinical signs of myocardial involvement within first year of observation. Low expression of CD95 may justify an certain role in the pathogenesis of the disease.
Collapse
|
7
|
Heath J, Chen D, Xie J, Choi J, Ng R, Zhang R, Li S, Edmark R, Zheng H, Solomon B, Campbell K, Medina E, Ribas A, Khatri P, Lanier L, Mease P, Goldman J, Su Y. An NKG2A biased immune response confers protection for infection, autoimmune disease, and cancer. RESEARCH SQUARE 2023:rs.3.rs-3413673. [PMID: 37886475 PMCID: PMC10602172 DOI: 10.21203/rs.3.rs-3413673/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Infection, autoimmunity, and cancer are the principal human health challenges of the 21st century and major contributors to human death and disease. Often regarded as distinct ends of the immunological spectrum, recent studies have hinted there may be more overlap between these diseases than appears. For example, pathogenic inflammation has been demonstrated as conserved between infection and autoimmune settings. T resident memory (TRM) cells have been highlighted as beneficial for infection and cancer. However, these findings are limited by patient number and disease scope; exact immunological factors shared across disease remain elusive. Here, we integrate large-scale deeply clinically and biologically phenotyped human cohorts of 526 patients with infection, 162 with lupus, and 11,180 with cancer. We identify an NKG2A+ immune bias as associative with protection against disease severity, mortality, and autoimmune and post-acute chronic disease. We reveal that NKG2A+ CD8+ T cells correlate with reduced inflammation, increased humoral immunity, and resemble TRM cells. Our results suggest that an NKG2A+ bias is a pan-disease immunological factor of protection and thus supports recent suggestions that there is immunological overlap between infection, autoimmunity, and cancer. Our findings underscore the promotion of an NKG2A+ biased response as a putative therapeutic strategy.
Collapse
|
8
|
Xu J, He J, He J, He Y, Zhang D, Kong R, Dan K. The abscopal effect of anti-CD95 and radiotherapy in melanoma. Discov Oncol 2023; 14:68. [PMID: 37191832 DOI: 10.1007/s12672-023-00682-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Radiotherapy (RT) is frequently adopted to control cancer cell proliferation, which is achieved by altering the tumor microenvironment (TME) and immunogenicity. Apoptosis of cancer cells is the major effect of radiation on tumor tissues. Fas/APO-1(CD95) receptors on the cell membrane are death receptors that can be activated by diverse factors, including radiation and integration with CD95L on CD8+ T cells. The abscopal effect is defined as tumor regression out of the local RT field, and it is produced through anti-tumor immunity. The immune response against the radiated tumor is characterized by the cross-presentation between antigen-presenting cells (APCs), which includes cytotoxic T cells (CTLs) and dendritic cells (DCs). METHODS The effect of activation and radiation of CD95 receptors on melanoma cell lines was examined in vivo and in vitro. In vivo, bilateral lower limbs were given a subcutaneous injection of a dual-tumor. Tumors in the right limb were radiated with a single dose of 10 Gy (primary tumor), while tumors in the left limb (secondary tumor) were spared. RESULTS The anti-CD95 treatment plus radiation (combination treatment) reduced growth rates of both primary and secondary tumors relative to the control or radiation groups. In addition, higher degrees of infiltrating CTLs and DCs were detected in the combination treatment compared to the other groups, but the immune response responsible for secondary tumor rejection was not proven to be tumor specific. In vitro, combination treatment combined with radiation resulted in further apoptosis of melanoma cells relative to controls or cells treated with radiation. CONCLUSIONS Targeting CD95 on cancer cells will induce tumor control and the abscopal effect.
Collapse
Affiliation(s)
- Jixiang Xu
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Zhongshan 319, Luzhou, 646000, China
| | - JiangFeng He
- Nursing Department, The Affiliated Hospital of Southwest Medical University, Zhongshan 319, Luzhou, 646000, China
| | - JiaJun He
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Zhongshan 319, Luzhou, 646000, China
| | - Yuanmin He
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Zhongshan 319, Luzhou, 646000, China
| | - DaoJun Zhang
- Department of Dermatology, The Third Affiliated Hospital of ChongQing Medical University, ShuangHuZhiLu 1, Chongqing, 401120, China
| | - Rui Kong
- Department of Oncology, The Third Affiliated Hospital of ChongQing Medical University, ShuangHuZhiLu 1, Chongqing, 401120, China
| | - Kena Dan
- Department of Dermatology, The Third Affiliated Hospital of ChongQing Medical University, ShuangHuZhiLu 1, Chongqing, 401120, China.
| |
Collapse
|
9
|
Carmena Moratalla A, Carpentier Solorio Y, Lemaître F, Farzam-Kia N, Da Cal S, Guimond JV, Haddad E, Duquette P, Girard JM, Prat A, Larochelle C, Arbour N. Specific alterations in NKG2D + T lymphocytes in relapsing-remitting and progressive multiple sclerosis patients. Mult Scler Relat Disord 2023; 71:104542. [PMID: 36716577 DOI: 10.1016/j.msard.2023.104542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 01/19/2023] [Accepted: 01/25/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND T lymphocytes exhibit numerous alterations in relapsing-remitting (RRMS), secondary progressive (SPMS), and primary progressive multiple sclerosis (PPMS). The NKG2D pathway has been involved in MS pathology. NKG2D is a co-activating receptor on subsets of CD4+ and most CD8+ T lymphocytes. The ligands of NKG2D are expressed at low levels in normal tissues but are elevated in MS postmortem brain tissues compared with controls. Whether the NKG2D pathway shows specific changes in different forms of MS remains unclear. METHODS We performed unsupervised and supervised flow cytometry analysis to characterize peripheral blood T lymphocytes from RRMS, SPMS, and PPMS patients and healthy controls (HC). We used an in vitro microscopy approach to assess the role of NKG2D in the interactions between human CD8+T lymphocytes and human astrocytes. RESULTS Specific CD8+, CD4+, and CD4-CD8- T cell populations exhibited altered frequency in MS patients' subgroups. The proportion of NKG2D+ T lymphocytes declined with age in PPMS patients but not in RRMS and HC. This reduced percentage of NKG2D+ cells was due to lower abundance of γδ and αβ CD4-CD8- T lymphocytes in PPMS patients. NKG2D+ T lymphocytes were significantly less abundant in RRMS than in HC; this was caused by a decreased frequency of CD4-CD8- and CD8+ T lymphocytes and was not linked to age. Blocking NKG2D increased the motility of CD8+ T lymphocytes co-cultured with astrocytes expressing NKG2D ligand. Moreover, preventing NKG2D from interacting with its ligands increased the proportion of CD8+ T lymphocytes exhibiting a kinapse-like behavior characterized by short-term interaction while reducing those displaying a long-lasting synapse-like behavior. These results support that NKG2D participates in the establishment of long-term interactions between activated CD8+ T lymphocytes and astrocytes. CONCLUSION Our data demonstrate specific alterations in NKG2D+ T lymphocytes in MS patients' subgroups and suggest that NKG2D contributes to the interactions between human CD8+ T lymphocytes and human astrocytes.
Collapse
Affiliation(s)
- Ana Carmena Moratalla
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Yves Carpentier Solorio
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Florent Lemaître
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Negar Farzam-Kia
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Sandra Da Cal
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9
| | - Jean Victor Guimond
- CLSC des Faubourgs, CIUSSS du Centre-Sud-de-l'Ile-de-Montréal, Montreal, QC, Canada
| | - Elie Haddad
- Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics, Université de Montréal, Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Pierre Duquette
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - J Marc Girard
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - Alexandre Prat
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - Catherine Larochelle
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9; MS-CHUM Clinic 900 St-Denis Street, Montreal, QC, Canada, H2X0A9
| | - Nathalie Arbour
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM) 900 St-Denis Street Montreal, QC, Canada, H2X0A9.
| |
Collapse
|
10
|
Autophagy of naïve CD4 + T cells in aging - the role of body adiposity and physical fitness. Expert Rev Mol Med 2023; 25:e9. [PMID: 36655333 DOI: 10.1017/erm.2023.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Life expectancy has increased exponentially in the last century accompanied by disability, poor quality of life, and all-cause mortality in older age due to the high prevalence of obesity and physical inactivity in older people. Biologically, the aging process reduces the cell's metabolic and functional efficiency, and disrupts the cell's anabolic and catabolic homeostasis, predisposing older people to many dysfunctional conditions such as cardiovascular disease, neurodegenerative disorders, cancer, and diabetes. In the immune system, aging also alters cells' metabolic and functional efficiency, a process known as 'immunosenescence', where cells become more broadly inflammatory and their functionality is altered. Notably, autophagy, the conserved and important cellular process that maintains the cell's efficiency and functional homeostasis may protect the immune system from age-associated dysfunctional changes by regulating cell death in activated CD4+ T cells. This regulatory process increases the delivery of the dysfunctional cytoplasmic material to lysosomal degradation while increasing cytokine production, proliferation, and differentiation of CD4+ T cell-mediated immune responses. Poor proliferation and diminished responsiveness to cytokines appear to be ubiquitous features of aged T cells and may explain the delayed peak in T cell expansion and cytotoxic activity commonly observed in the 'immunosenescence' phenotype in the elderly. On the other hand, physical exercise stimulates the expression of crucial nutrient sensors and inhibits the mechanistic target of the rapamycin (mTOR) signaling cascade which increases autophagic activity in cells. Therefore, in this perspective review, we will first contextualize the overall view of the autophagy process and then, we will discuss how body adiposity and physical fitness may counteract autophagy in naïve CD4+ T cells in aging.
Collapse
|
11
|
Lemaître F, Farzam-Kia N, Carmena Moratalla A, Carpentier Solorio Y, Clenet ML, Tastet O, Cleret-Buhot A, Guimond JV, Haddad E, Duquette P, Girard JM, Prat A, Larochelle C, Arbour N. IL-27 shapes the immune properties of human astrocytes and their impact on encountered human T lymphocytes. J Neuroinflammation 2022; 19:212. [PMID: 36050707 PMCID: PMC9434874 DOI: 10.1186/s12974-022-02572-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/23/2022] [Indexed: 11/10/2022] Open
Abstract
Background Interleukin-27 (IL-27) can trigger both pro- and anti-inflammatory responses. This cytokine is elevated in the central nervous system (CNS) of multiple sclerosis (MS) patients, but how it influences neuroinflammatory processes remains unclear. As astrocytes express the receptor for IL-27, we sought to determine how these glial cells respond to this cytokine and whether such exposure alters their interactions with infiltrating activated T lymphocytes. To determine whether inflammation shapes the impact of IL-27, we compared the effects of this cytokine in non-inflamed and inflamed conditions induced by an IL-1β exposure. Main body Transcriptomic analysis of IL-27-exposed human astrocytes showed an upregulation of multiple immune genes. Human astrocytes increased the secretion of chemokines (CXCL9, CXCL10, and CXCL11) and the surface expression of proteins (PD-L1, HLA-E, and ICAM-1) following IL-27 exposure. To assess whether exposure of astrocytes to IL-27 influences the profile of activated T lymphocytes infiltrating the CNS, we used an astrocyte/T lymphocyte co-culture model. Activated human CD4+ or CD8+ T lymphocytes were co-cultured with astrocytes that have been either untreated or pre-exposed to IL‑27 or IL-1β. After 24 h, we analyzed T lymphocytes by flow cytometry for transcription factors and immune molecules. The contact with IL-27-exposed astrocytes increased the percentages of T-bet, Eomes, CD95, IL-18Rα, ICAM-1, and PD-L1 expressing CD4+ and CD8+ T lymphocytes and reduced the proportion of CXCR3-positive CD8+ T lymphocytes. Human CD8+ T lymphocytes co-cultured with human IL-27-treated astrocytes exhibited higher motility than when in contact with untreated astrocytes. These results suggested a preponderance of kinapse-like over synapse-like interactions between CD8+ T lymphocytes and IL-27-treated astrocytes. Finally, CD8+ T lymphocytes from MS patients showed higher motility in contact with IL-27-exposed astrocytes compared to healthy donors’ cells. Conclusion Our results establish that IL-27 alters the immune functions of human astrocytes and shapes the profile and motility of encountered T lymphocytes, especially CD8+ T lymphocytes from MS patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02572-1.
Collapse
Affiliation(s)
- Florent Lemaître
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada
| | - Negar Farzam-Kia
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada
| | - Ana Carmena Moratalla
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada
| | - Yves Carpentier Solorio
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada
| | - Marie-Laure Clenet
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada
| | - Olivier Tastet
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada
| | - Aurélie Cleret-Buhot
- Centre de Recherche du Centre Hospitalier de L'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Jean Victor Guimond
- CLSC Des Faubourgs, CIUSSS du Centre-Sud-de-L'Ile-de-Montréal, Montréal, QC, Canada
| | - Elie Haddad
- Department of Microbiology, Infectious Diseases, and Immunology and Department of Pediatrics, Centre de Recherche du Centre Hospitalier, Université de Montréal, Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, QC, Canada
| | - Pierre Duquette
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada.,MS-CHUM Clinic, 900 St-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - J Marc Girard
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada.,MS-CHUM Clinic, 900 St-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Alexandre Prat
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada.,MS-CHUM Clinic, 900 St-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Catherine Larochelle
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada.,MS-CHUM Clinic, 900 St-Denis Street, Montreal, QC, H2X 0A9, Canada
| | - Nathalie Arbour
- Department of Neurosciences, Université de Montréal and Centre de Recherche du CHUM (CRCHUM), 900 St-Denis Street, Room R09.464, Montreal, QC, H2X 0A9, Canada.
| |
Collapse
|
12
|
Wang H, Chen L, Qi L, Jiang N, Zhang Z, Guo H, Song T, Li J, Li H, Zhang N, Chen R. A Single-Cell Atlas of Tumor-Infiltrating Immune Cells in Pancreatic Ductal Adenocarcinoma. Mol Cell Proteomics 2022; 21:100258. [PMID: 35718340 PMCID: PMC9294203 DOI: 10.1016/j.mcpro.2022.100258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 11/30/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with limited treatment options. To guide the design of more effective immunotherapy strategies, mass cytometry was employed to characterize the cellular composition of the PDAC-infiltrating immune cells. The expression of 33 protein markers was examined at the single-cell level in more than two million immune cells from four types of clinical samples, including PDAC tumors, normal pancreatic tissues, chronic pancreatitis tissues, and peripheral blood. Based on the analyses, we identified 23 distinct T-cell phenotypes, with some cell clusters exhibiting aberrant frequencies in the tumors. Programmed cell death protein 1 (PD-1) was extensively expressed in CD4+ and CD8+ T cells and coexpressed with both stimulatory and inhibitory immune markers. In addition, we observed elevated levels of functional markers, such as CD137L and CD69, in PDAC-infiltrating immune cells. Moreover, the combination of PD-1 and CD8 was used to stratify PDAC tumors from The Cancer Genome Atlas database into three immune subtypes, with S1 (PD-1+CD8+) exhibiting the best prognosis. Further analysis suggested distinct molecular mechanisms for immune exclusion in different subtypes. Taken together, the single-cell protein expression data depicted a detailed cell atlas of the PDAC-infiltrating immune cells and revealed clinically relevant information regarding useful cell phenotypes and targets for immunotherapy development.
Collapse
Affiliation(s)
- Hao Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China; Department of Clinical Laboratory, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Lu Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Lisha Qi
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Na Jiang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Zhibin Zhang
- Department of General Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Hua Guo
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Tianqiang Song
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China
| | - Jun Li
- Department of Molecular Pathology, Clinical Pathology Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Hongle Li
- Department of Molecular Pathology, Clinical Pathology Center, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| | - Ning Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, China; Peking University First Hospital, Peking University Health Science Center, Beijing, China.
| | - Ruibing Chen
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China.
| |
Collapse
|
13
|
Weng SC, Wen MC, Hsieh SL, Chen NJ, Tarng DC. Decoy Receptor 3 Suppresses T-Cell Priming and Promotes Apoptosis of Effector T-Cells in Acute Cell-Mediated Rejection: The Role of Reverse Signaling. Front Immunol 2022; 13:879648. [PMID: 35720343 PMCID: PMC9201909 DOI: 10.3389/fimmu.2022.879648] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 05/09/2022] [Indexed: 11/16/2022] Open
Abstract
Background Decoy receptor 3 (DcR3) belongs to the tumor necrosis factor (TNF) receptor superfamily and neutralizes TNF ligands, including FasL and TRAIL, to prevent T activation during T-cell priming. However, the cellular mechanisms underlying acute cell-mediated rejection (ACMR) remain unknown. Methods We generated DcR3 transgenic (Tg) mice and mice with high DcR3 expression (HDE) to study both in vivo and in vitro. FasR RNA knockdown in immortalized CD4+CD8+ T-cells was used to survey the role of DcR3 on FasR/Fas-associated protein with death domain (FADD)/caspase 8 pathway and its cross-link to TNF receptor-associated factor 1 (TNFR1)-associated death domain protein (TRADD) in suppressing TNFR1. TNF/TRADD knockout mice were used to show the importance of TNF adaptor protein. Results DcR3.Fc suppressed C57BL/6 female T-cell activation and transformation into CD4+CD69+, CD4+CD44+, and CD4+CD25+Foxp3+ when compared with isotype IgG1 and its co-treatment with FasL/TRAIL after exposing to bone marrow-derived dendritic cells (BMDCs) that carried alloantigen with male H-Y and minor antigenic determinant. Interleukin-17 and interferon-γ productions by BMDC-activated T-cells were lowered after co-treating with DcR3.Fc. DcR3.Fc induced effector T-cells (Teffs) and was susceptible to FasR-mediated apoptosis through the FADD/TRADD/caspase 8 pathway. After exposing to DcR3.Fc, TRADD was silenced, likely turning down the inflammatory response. The systemic effects of DcR3 Tg mice and HDE phenotype induced by the promoter of cytomegalovirus not only attenuated ACMR severity but also ameliorated the high serum creatinine and blood urea nitrogen levels even with high T-cell exposure frequencies. Besides this, DcR3 has minor biological effects on both MHC-matched and MHC-mismatched models. Conclusions High DcR3 doses protect renal tubular epithelial cells from acute T-cell attack during the T-cell priming stage via interfering with TNF ligand-mediated reverse signaling and possibly promoting Teff apoptosis through FasR upregulation. Our findings supported that the decoy receptor is involved in T-cell modulation in kidney transplant rejection.
Collapse
Affiliation(s)
- Shuo-Chun Weng
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan.,Institute of Clinical Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Center for Geriatrics and Gerontology, Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Mei-Chin Wen
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Pathology and Laboratory Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Shie-Liang Hsieh
- Institute of Clinical Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Department of Medical Research and Education, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Clinical Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Nien-Jung Chen
- Institute of Microbiology and Immunology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Inflammation and Immunity Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Der-Cherng Tarng
- Institute of Clinical Medicine, School of Medicine, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Department and Institute of Physiology, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Division of Nephrology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Center for Intelligent Drug Systems and Smart Bio-devices, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
14
|
Risso V, Lafont E, Le Gallo M. Therapeutic approaches targeting CD95L/CD95 signaling in cancer and autoimmune diseases. Cell Death Dis 2022; 13:248. [PMID: 35301281 PMCID: PMC8931059 DOI: 10.1038/s41419-022-04688-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 02/09/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022]
Abstract
Cell death plays a pivotal role in the maintenance of tissue homeostasis. Key players in the controlled induction of cell death are the Death Receptors (DR). CD95 is a prototypic DR activated by its cognate ligand CD95L triggering programmed cell death. As a consequence, alterations in the CD95/CD95L pathway have been involved in several disease conditions ranging from autoimmune diseases to inflammation and cancer. CD95L-induced cell death has multiple roles in the immune response since it constitutes one of the mechanisms by which cytotoxic lymphocytes kill their targets, but it is also involved in the process of turning off the immune response. Furthermore, beyond the canonical pro-death signals, CD95L, which can be membrane-bound or soluble, also induces non-apoptotic signaling that contributes to its tumor-promoting and pro-inflammatory roles. The intent of this review is to describe the role of CD95/CD95L in the pathophysiology of cancers, autoimmune diseases and chronic inflammation and to discuss recently patented and emerging therapeutic strategies that exploit/block the CD95/CD95L system in these diseases.
Collapse
Affiliation(s)
- Vesna Risso
- INSERM U1242, Oncogenesis Stress Signaling, University of Rennes, Rennes, France
- Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Elodie Lafont
- INSERM U1242, Oncogenesis Stress Signaling, University of Rennes, Rennes, France
- Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Matthieu Le Gallo
- INSERM U1242, Oncogenesis Stress Signaling, University of Rennes, Rennes, France.
- Centre de lutte contre le cancer Eugène Marquis, Rennes, France.
| |
Collapse
|
15
|
Aru B, Soltani M, Pehlivanoglu C, Gürlü E, Ganjalikhani-Hakemi M, Yanikkaya Demirel G. Comparison of Laboratory Methods for the Clinical Follow Up of Checkpoint Blockade Therapies in Leukemia: Current Status and Challenges Ahead. Front Oncol 2022; 12:789728. [PMID: 35155232 PMCID: PMC8829140 DOI: 10.3389/fonc.2022.789728] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 01/06/2022] [Indexed: 02/05/2023] Open
Abstract
The development of immune checkpoint inhibitors, the monoclonal antibodies that modulate the interaction between immune checkpoint molecules or their ligands on the immune cells or tumor tissue has revolutionized cancer treatment. While there are various studies proving their efficacy in hematological malignancies, there is also a body of accumulating evidence indicating that immune checkpoint inhibitors’ clinical benefits are limited in such diseases. In addition, due to their regulatory nature that balances the immune responses, blockade of immune checkpoints may lead to toxic side effects and autoimmune responses, and even primary or acquired resistance mechanisms may restrict their success. Thus, the need for laboratory biomarkers to identify and monitor patient populations who are more likely respond to this type of therapy and the management of side effects seem critical. However, guidelines regarding the use of immune checkpoint inhibitors in hematological cancers and during follow-up are limited while there is no consensus on the laboratory parameters to be investigated for safety and efficacy of the treatment. This review aims to provide an insight into recent information on predictive and prognostic value of biomarkers and laboratory tests for the clinical follow up of hematological malignancies, with an emphasis on leukemia.
Collapse
Affiliation(s)
- Basak Aru
- Department of Immunology, Faculty of Medicine, Yeditepe University, Istanbul, Turkey
| | - Mojdeh Soltani
- Department of Immunology, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Cemil Pehlivanoglu
- Department of Emergency Medicine, Hatay Training and Research Hospital, Antakya, Turkey
| | - Ege Gürlü
- Faculty of Medicine 4thYear Student, Yeditepe University, Istanbul, Turkey
| | | | | |
Collapse
|
16
|
Loo K, Smithy JW, Postow MA, Betof Warner A. Factors Determining Long-Term Antitumor Responses to Immune Checkpoint Blockade Therapy in Melanoma. Front Immunol 2022; 12:810388. [PMID: 35087529 PMCID: PMC8787112 DOI: 10.3389/fimmu.2021.810388] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 12/07/2021] [Indexed: 12/13/2022] Open
Abstract
With the increasing promise of long-term survival with immune checkpoint blockade (ICB) therapies, particularly for patients with advanced melanoma, clinicians and investigators are driven to identify prognostic and predictive factors that may help to identify individuals who are likely to experience durable benefit. Several ICB combinations are being actively developed to expand the armamentarium of treatments for patients who may not achieve long-term responses to ICB single therapies alone. Thus, negative predictive markers are also of great interest. This review seeks to deepen our understanding of the mechanisms underlying the durability of ICB treatments. We will discuss the currently available long-term data from the ICB clinical trials and real-world studies describing the survivorship of ICB-treated melanoma patients. Additionally, we explore the current treatment outcomes in patients rechallenged with ICB and the patterns of ICB resistance based on sites of disease, namely, liver or CNS metastases. Lastly, we discuss the landscape in melanoma in the context of prognostic or predictive factors as markers of long-term response to ICB.
Collapse
Affiliation(s)
- Kimberly Loo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Internal Medicine, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, NY, United States
| | - James W. Smithy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Michael A. Postow
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| | - Allison Betof Warner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill Cornell Medical College, New York, NY, United States
| |
Collapse
|
17
|
Lisowska KA, Storoniak H, Dębska-Ślizień A. T cell subpopulations and cytokine levels in hemodialysis patients. Hum Immunol 2021; 83:134-143. [PMID: 34802797 DOI: 10.1016/j.humimm.2021.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/23/2021] [Accepted: 11/08/2021] [Indexed: 12/19/2022]
Abstract
HD patients have impaired adaptive immune responses, which might depend on the primary cause of chronic kidney disease (CKD). We analyzed percentages of T cells subpopulations with the expression of CD69, CD25, CD95, and HLA-DR antigens in HD patients to determine the status of T cell activation. Also, we determined serum levels of cytokines: IL12p70, TNF, IL-10, IL-6, IL-1β, IL-8. HD patients had increased percentages of CD4+CD25+, CD4+CD69+, CD4+HLA-DR+, CD8+CD69+, and CD8+HLA-DR+ cells compared to healthy people. Also, their IL-6 and IL-8 serum levels were higher. Changes in T cell subpopulations were seen in patients with diabetic nephropathy (DN) or ischemic nephropathy (IN) but not with glomerulonephritis (GN). HD patients dialyzed for more than six months had a lower percentage of CD4+CD69+, CD8+HLA-DR+, CD8+CD95+ cells, higher IL-12p70 levels, and lower IL-8 levels. Our results show that HD treatment and CKD cause influence T cell activation status.
Collapse
Affiliation(s)
- Katarzyna A Lisowska
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland.
| | - Hanna Storoniak
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
18
|
Effect of PTPN22, FAS/FASL, IL2RA and CTLA4 genetic polymorphisms on the risk of developing alopecia areata: A systematic review of the literature and meta-analysis. PLoS One 2021; 16:e0258499. [PMID: 34735462 PMCID: PMC8568157 DOI: 10.1371/journal.pone.0258499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 09/28/2021] [Indexed: 12/21/2022] Open
Abstract
Objectives Genetic association studies on alopecia areata (AA) performed in various populations have shown heterogeneous results. The aim of the current review was to synthesize the results of said studies to estimate the impact of FAS, FASL, PTPN22, CTLA4 and IL2RA gene polymorphisms on AA susceptibility. Design A systematic literature search was conducted in the Medline, Web of Science, Scopus, EMBASE and LILACS databases. Studies published up to June 2020 were included. The results available in the grey literature including the Open Grey and Google Scholar databases were also used. The texts of potentially related studies were screened by individual reviewers. Evidence of publication bias was assessed using the Newcastle-Ottawa scale and the quality of evidence was assessed using the GRADE system. The quantitative synthesis was performed using the fixed effect model. Results Out of 1784 articles, we identified 18 relevant articles for the qualitative synthesis and 16 for the quantitative synthesis. In a study of rs2476601 polymorphism of PTPN22 gene, including 1292 cases and 1832 controls, a correlation was found with the risk of developing AA in the allelic model (OR1.49 [95% C:1.13–1.95]), the heterozygous codominant (OR1.44 [95% CI:1:19–1.76]) and dominant model (OR1.43 [95% CI:1.18–1.73]). No association was found between the presence of FASL, PTPN22, CTLA and IL2RA gene polymorphisms with AA susceptibility. Conclusions The results suggest that the T allele of the single nucleoid polymorphism (SNP) rs2476601 in PTPN22 gene is a risk factor for developing alopecia areata. However, more robust studies defining the ethnic background of the population of origin are required, so that the risk identified in the present study can be validated. Additionally, a greater number of studies is necessary to evaluate the role of the FAS, FASL, PTPN22, CTLA4 and IL2RA genetic variants, given the heterogenous results found in the literature.
Collapse
|
19
|
Sarfas C, White AD, Sibley L, Morrison AL, Gullick J, Lawrence S, Dennis MJ, Marsh PD, Fletcher HA, Sharpe SA. Characterization of the Infant Immune System and the Influence and Immunogenicity of BCG Vaccination in Infant and Adult Rhesus Macaques. Front Immunol 2021; 12:754589. [PMID: 34707617 PMCID: PMC8542880 DOI: 10.3389/fimmu.2021.754589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 09/27/2021] [Indexed: 11/13/2022] Open
Abstract
In many countries where tuberculosis (TB) is endemic, the Bacillus Calmette–Guérin (BCG) vaccine is given as close to birth as possible to protect infants and children from severe forms of TB. However, BCG has variable efficacy and is not as effective against adult pulmonary TB. At present, most animal models used to study novel TB vaccine candidates rely on the use of adult animals. Human studies show that the infant immune system is different to that of an adult. Understanding how the phenotypic profile and functional ability of the immature host immune system compares to that of a mature adult, together with the subsequent BCG immune response, is critical to ensuring that new TB vaccines are tested in the most appropriate models. BCG-specific immune responses were detected in macaques vaccinated within a week of birth from six weeks after immunization indicating that neonatal macaques are able to generate a functional cellular response to the vaccine. However, the responses measured were significantly lower than those typically observed following BCG vaccination in adult rhesus macaques and infant profiles were skewed towards the activation and attraction of macrophages and monocytes and the synthesis in addition to release of pro-inflammatory cytokines such as IL-1, IL-6 and TNF-α. The frequency of specific immune cell populations changed significantly through the first three years of life as the infants developed into young adult macaques. Notably, the CD4:CD8 ratio significantly declined as the macaques aged due to a significant decrease in the proportion of CD4+ T-cells relative to a significant increase in CD8+ T-cells. Also, the frequency of both CD4+ and CD8+ T-cells expressing the memory marker CD95, and memory subset populations including effector memory, central memory and stem cell memory, increased significantly as animals matured. Infant macaques, vaccinated with BCG within a week of birth, possessed a significantly higher frequency of CD14+ classical monocytes and granulocytes which remained different throughout the first three years of life compared to unvaccinated age matched animals. These findings, along with the increase in monokines following vaccination in infants, may provide an insight into the mechanism by which vaccination with BCG is able to provide non-specific immunity against non-mycobacterial organisms.
Collapse
Affiliation(s)
- Charlotte Sarfas
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Andrew D White
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Laura Sibley
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Alexandra L Morrison
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Jennie Gullick
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Steve Lawrence
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Mike J Dennis
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Philip D Marsh
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| | - Helen A Fletcher
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sally A Sharpe
- National Infection Service, UK Health Security Agency, Salisbury, United Kingdom
| |
Collapse
|
20
|
Nyambuya TM, Dludla PV, Nkambule BB. Diet-Induced Obesity Promotes the Upregulation of Fas Expression on T-cells. BIOLOGY 2021; 10:biology10030217. [PMID: 33808960 PMCID: PMC8000544 DOI: 10.3390/biology10030217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022]
Abstract
This study was conducted to assess the expression of Fas (CD95) and programmed cell death-1 (PD-1) on circulating T-cells in obesity using a diet-induced obesity mouse model. Furthermore, we aimed to determine if there are any associations between metabolic disorders and the expression of T-cell regulatory markers. A total of 12 male C57BL/6 mice were randomized into either a high-fat diet (HFD) or low-fat diet (LFD) group for 8 weeks (n = 6/group). Changes in body weights were monitored on a weekly basis. The lipid, glucose, and hematological profiles, as well as Fas and PD1 expression on the T-cell immunophenotype, were measured after 8 weeks of feeding. The HFD-fed group had a higher percentage weight gain (29.17%) in comparison with the LFD-fed group (21.74%) after the 8-week period. In addition, the HFD group had increased fasting glucose and glucose excursion following a 2-h postprandial period. The levels of total cholesterol were elevated in the HFD group when compared with the LFD group (p < 0.05). Notably, the absolute white cell count (p = 0.0096), neutrophil count (p = 0.0022, lymphocytes (p = 0.0155), and monocyte count (p = 0.0015) were elevated in the HFD group when compared with the LFD-fed group. However, the platelets (0.0680), red cell counts (0.3575), and their indices (p > 0.05) were comparable between the two groups. Interestingly, HFD feeding was associated with elevated expression of Fas on T-cells (p < 0.0001), which positively correlated with body weights (r = 0.93, p = 0.0333). No associations were found between Fas expression and dyslipidemia or fasting blood glucose levels (p > 0.05). The multivariant regression analysis showed that the association between the levels of Fas on T-cells and body weights (coefficient: -1.00, t-value: 19.27, p = 0.0330) was independent of fasting blood glucose, total cholesterol, and lymphocyte count. Lastly, the expression of PD-1 on T-cells was comparable between the two diet groups (p = 0.1822). In all, immune activation, dyslipidemia, and poor glucose control in the early stages of obesity may drive the pathogenesis of metabolic T-cell disorders. Importantly, T-cell dysfunction in obesity is partially mediated by an upregulation of Fas which is independent of dyslipidemia and hyperglycemia.
Collapse
Affiliation(s)
- Tawanda Maurice Nyambuya
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban 4013, South Africa
- Department of Health Sciences, Faculty of Health and Applied Sciences, Namibia University of Science and Technology, Windhoek 10005, Namibia
- Correspondence: (T.M.N.); (B.B.N.); Tel.: +264-61-207-2914 (T.M.N.); +27-(0)31-260-8964 (B.B.N.)
| | - Phiwayinkosi Vusi Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa;
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Bongani Brian Nkambule
- School of Laboratory Medicine and Medical Sciences (SLMMS), College of Health Sciences, University of KwaZulu-Natal, Durban 4013, South Africa
- Correspondence: (T.M.N.); (B.B.N.); Tel.: +264-61-207-2914 (T.M.N.); +27-(0)31-260-8964 (B.B.N.)
| |
Collapse
|
21
|
Su Y, Chen D, Yuan D, Lausted C, Choi J, Dai CL, Voillet V, Duvvuri VR, Scherler K, Troisch P, Baloni P, Qin G, Smith B, Kornilov SA, Rostomily C, Xu A, Li J, Dong S, Rothchild A, Zhou J, Murray K, Edmark R, Hong S, Heath JE, Earls J, Zhang R, Xie J, Li S, Roper R, Jones L, Zhou Y, Rowen L, Liu R, Mackay S, O'Mahony DS, Dale CR, Wallick JA, Algren HA, Zager MA, Wei W, Price ND, Huang S, Subramanian N, Wang K, Magis AT, Hadlock JJ, Hood L, Aderem A, Bluestone JA, Lanier LL, Greenberg PD, Gottardo R, Davis MM, Goldman JD, Heath JR. Multi-Omics Resolves a Sharp Disease-State Shift between Mild and Moderate COVID-19. Cell 2020; 183:1479-1495.e20. [PMID: 33171100 PMCID: PMC7598382 DOI: 10.1016/j.cell.2020.10.037] [Citation(s) in RCA: 401] [Impact Index Per Article: 80.2] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/16/2020] [Accepted: 10/22/2020] [Indexed: 12/29/2022]
Abstract
We present an integrated analysis of the clinical measurements, immune cells, and plasma multi-omics of 139 COVID-19 patients representing all levels of disease severity, from serial blood draws collected during the first week of infection following diagnosis. We identify a major shift between mild and moderate disease, at which point elevated inflammatory signaling is accompanied by the loss of specific classes of metabolites and metabolic processes. Within this stressed plasma environment at moderate disease, multiple unusual immune cell phenotypes emerge and amplify with increasing disease severity. We condensed over 120,000 immune features into a single axis to capture how different immune cell classes coordinate in response to SARS-CoV-2. This immune-response axis independently aligns with the major plasma composition changes, with clinical metrics of blood clotting, and with the sharp transition between mild and moderate disease. This study suggests that moderate disease may provide the most effective setting for therapeutic intervention.
Collapse
Affiliation(s)
- Yapeng Su
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Daniel Chen
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Dan Yuan
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | | | - Jongchan Choi
- Institute for Systems Biology, Seattle, WA 98109, USA
| | | | - Valentin Voillet
- Cape Town HVTN Immunology Laboratory, Hutchinson Centre Research Institute of South Africa, NPC (HCRISA), Cape Town 8001, South Africa; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | - Guangrong Qin
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Brett Smith
- Institute for Systems Biology, Seattle, WA 98109, USA
| | | | | | - Alex Xu
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shen Dong
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alissa Rothchild
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Jing Zhou
- Isoplexis Corporation, Branford, CT 06405, USA
| | - Kim Murray
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Rick Edmark
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Sunga Hong
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - John E Heath
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - John Earls
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Rongyu Zhang
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Jingyi Xie
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Sarah Li
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Ryan Roper
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Lesley Jones
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Yong Zhou
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Lee Rowen
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Rachel Liu
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Sean Mackay
- Isoplexis Corporation, Branford, CT 06405, USA
| | - D Shane O'Mahony
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA 98109, USA; Providence St. Joseph Health, Renton, WA 98057, USA
| | - Christopher R Dale
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA 98109, USA; Providence St. Joseph Health, Renton, WA 98057, USA
| | - Julie A Wallick
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA 98109, USA; Providence St. Joseph Health, Renton, WA 98057, USA
| | - Heather A Algren
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA 98109, USA; Providence St. Joseph Health, Renton, WA 98057, USA
| | - Michael A Zager
- Center for Data Visualization, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | - Wei Wei
- Institute for Systems Biology, Seattle, WA 98109, USA
| | | | - Sui Huang
- Institute for Systems Biology, Seattle, WA 98109, USA
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Global Heath, and Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA
| | | | | | - Leroy Hood
- Institute for Systems Biology, Seattle, WA 98109, USA; Providence St. Joseph Health, Renton, WA 98057, USA
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA 98109, USA
| | - Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology, University of California, San Francisco, and Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
| | - Philip D Greenberg
- Division of Clinical Research, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Departments of Immunology and Medicine, University of Washington, Seattle, WA 98109, USA
| | - Raphael Gottardo
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Department of Statistics, University of Washington, Seattle, WA 98195, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; The Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jason D Goldman
- Swedish Center for Research and Innovation, Swedish Medical Center, Seattle, WA 98109, USA; Providence St. Joseph Health, Renton, WA 98057, USA; Division of Allergy & Infectious Diseases, University of Washington, Seattle, WA 98109, USA.
| | - James R Heath
- Institute for Systems Biology, Seattle, WA 98109, USA; Department of Bioengineering, University of Washington, Seattle, WA 98105, USA.
| |
Collapse
|
22
|
Zebley CC, Gottschalk S, Youngblood B. Rewriting History: Epigenetic Reprogramming of CD8 + T Cell Differentiation to Enhance Immunotherapy. Trends Immunol 2020; 41:665-675. [PMID: 32624330 PMCID: PMC7395868 DOI: 10.1016/j.it.2020.06.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 12/31/2022]
Abstract
The full potential of T cell-based immunotherapies remains limited by a variety of T cell extrinsic and intrinsic immunosuppressive mechanisms that can become imprinted to stably reduce the antitumor ability of T cells. Here, we discuss recent insights into memory CD8+ T cell differentiation and exhaustion and the association of these differentiation states with clinical outcomes during immune checkpoint blockade and chimeric antigen receptor (CAR) T cell therapeutic modalities. We consider the barriers limiting immunotherapy with a focus on epigenetic regulation impeding efficacy of adoptively transferred T cells and other approaches that augment T cell responses such as immune checkpoint blockade. Furthermore, we outline conceptual and technical breakthroughs that can be applied to existing therapeutic approaches and to the development of novel cutting-edge strategies.
Collapse
Affiliation(s)
- Caitlin C Zebley
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ben Youngblood
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
23
|
El Saftawy EA, Amin NM, Sabry RM, El-Anwar N, Shash RY, Elsebaie EH, Wassef RM. Can Toxoplasma gondii Pave the Road for Dementia? J Parasitol Res 2020; 2020:8859857. [PMID: 32802484 PMCID: PMC7414348 DOI: 10.1155/2020/8859857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/25/2020] [Accepted: 07/03/2020] [Indexed: 01/04/2023] Open
Abstract
Dementia is an ominous neurological disease. Scientists proposed a link between its occurrence and the presence of Toxoplasma gondii (T. gondii). The long-term sequels of anti-Toxoplasma premunition, chiefly dominated by TNF-α, on the neurons and their receptors as the insulin-like growth factor-1 receptor (IGF-1R), which is tangled in cognition and synaptic plasticity, are still not clear. IGF-1R mediates its action via IGF-1, and its depletion is incorporated in the pathogenesis of dementia. The activated TNF-α signaling pathway induces NF-κβ that may induce or inhibit neurogenesis. This study speculates the potential impact of anti-Toxoplasma immune response on the expression of IGF-1R in chronic cerebral toxoplasmosis. The distributive pattern of T. gondii cysts was studied in association with TNF-α serum levels, the in situ expression of NF-κβ, and IGF-1R in mice using the low virulent ME-49 T. gondii strain. There was an elevation of the TNF-α serum level (p value ≤ 0.004) and significant upsurge in NF-κβ whereas IGF-1R was of low abundance (p value < 0.05) compared to the controls. TNF-α had a strong positive correlation with the intracerebral expression of NF-κβ (r value ≈ 0.943, p value ≈ 0.005) and a strong negative correlation to IGF-1R (r value -0.584 and -0.725 for area% and O.D., respectively). This activated TNF-α/NF-κβ keeps T. gondii under control at the expense of IGF-1R expression, depriving neurons of the effect of IGF-1, the receptor's ligand. We therefore deduce that T. gondii immunopathological reaction may be a road paver for developing dementia.
Collapse
Affiliation(s)
- Enas A. El Saftawy
- Medical Parasitology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
- Armed Forces College of Medicine, Cairo, Egypt
| | - Noha M. Amin
- Medical Parasitology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rania M. Sabry
- Pathology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Noha El-Anwar
- Armed Forces College of Medicine, Cairo, Egypt
- Pathology Department, Faculty of Medicine, Tanta University, Egypt
| | - Rania Y. Shash
- Medical Microbiology and Immunology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Eman H. Elsebaie
- Public Health and Community Medicine, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Rita M. Wassef
- Medical Parasitology Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| |
Collapse
|
24
|
Narsale A, Moya R, Ma J, Anderson LJ, Wu D, Garcia JM, Davies JD. Cancer-driven changes link T cell frequency to muscle strength in people with cancer: a pilot study. J Cachexia Sarcopenia Muscle 2019; 10:827-843. [PMID: 30977974 PMCID: PMC6711422 DOI: 10.1002/jcsm.12424] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/19/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Tumour growth can promote the loss of muscle mass and function. This is particularly disturbing because overall survival is significantly reduced in people with weaker and smaller skeletal muscle. The risk of cancer is also greater in people who are immune deficient. Muscle wasting in mice with cancer can be inhibited by infusion of CD4+ precursor T cells that restore balanced ratios of naïve, memory, and regulatory T cells. These data are consistent with the hypothesis that stronger anti-cancer T cell immunity leads to improved muscle mass and function. As a first step to testing this hypothesis, we determined whether levels of circulating T cell subsets correlate with levels of muscle strength in people with cancer. METHODS The frequency of circulating CD4+ and CD8+ naïve, memory, and regulatory T cell subsets was quantified in 11 men with gastrointestinal cancer (aged 59.3 ± 10.1 years) and nine men without cancer (aged 60 ± 13 years), using flow cytometry. T cell marker expression was determined using real-time PCR and western blot analyses in whole blood and peripheral blood mononuclear cells. Handgrip strength, one-repetition maximum chest press, and knee extension tests were used to determine muscle strength. Performance was determined using a stair climb test. Body composition was determined using dual-energy X-ray absorptiometry scan. The Karnofsky and ECOG scales were used to assess functional impairment. Correlations between frequencies of cell subsets with strength, performance, and body composition were determined using regression analyses. RESULTS Our data show significant correlations between (i) higher frequencies of CD8+ naïve (P = 0.02) and effector memory (P = 0.003) T cells and lower frequencies of CD8+ central memory T cells (P = 0.002) with stronger handgrip strength, (ii) lower frequency of regulatory cells with greater lean mass index (P = 0.04), (iii) lower frequency of CD8+ T cells that express CD95 with greater stair climb power (P = 0.003), (iv) higher frequency of T cells that co-express CD197 and CD45RA and greater one-repetition maximum knee extension strength (P = 0.008), and (iv) higher expression of CD4 in whole blood with greater functional impairment (P = 0.004) in people with cancer. CONCLUSIONS We have identified significant correlations between levels of T cell populations and muscle strength, performance, and body composition in people with cancer. These data justify a follow-up study with a larger cohort to test the validity of the findings.
Collapse
Affiliation(s)
- Aditi Narsale
- San Diego Biomedical Research Institute, San Diego, USA
| | - Rosa Moya
- San Diego Biomedical Research Institute, San Diego, USA
| | - Jasmin Ma
- San Diego Biomedical Research Institute, San Diego, USA
| | - Lindsey J Anderson
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA
| | - Daniel Wu
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA.,Oncology Section, VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA
| | - Jose M Garcia
- Geriatric Research, Education and Clinical Center, VA Puget Sound Health Care System, University of Washington, Seattle, WA, USA.,Division of Diabetes, Endocrinology and Metabolism, MCL, Center for Translational Research in Inflammatory Diseases, Michael E DeBakey Veterans Affairs Medical Center, Baylor College of Medicine, Houston, TX, USA
| | | |
Collapse
|
25
|
Kunert A, Basak EA, Hurkmans DP, Balcioglu HE, Klaver Y, van Brakel M, Oostvogels AAM, Lamers CHJ, Bins S, Koolen SLW, van der Veldt AAM, Sleijfer S, Mathijssen RHJ, Aerts JGJV, Debets R. CD45RA +CCR7 - CD8 T cells lacking co-stimulatory receptors demonstrate enhanced frequency in peripheral blood of NSCLC patients responding to nivolumab. J Immunother Cancer 2019; 7:149. [PMID: 31176366 PMCID: PMC6555948 DOI: 10.1186/s40425-019-0608-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 05/02/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Checkpoint inhibitors have become standard care of treatment for non-small cell lung cancer (NSCLC), yet only a limited fraction of patients experiences durable clinical benefit, highlighting the need for markers to stratify patient populations. METHODS To prospectively identify patients showing response to therapy, we have stained peripheral blood samples of NSCLC patients treated with 2nd line nivolumab (n = 71), as well as healthy controls, with multiplex flow cytometry. By doing so, we enumerated 18 immune cell subsets and assessed expression for 28 T cell markers, which was followed by dimensionality reduction as well as rationale-based analyses. RESULTS In patients with a partial response (PR), representing best overall response (BOR) according to RECIST v1.1, the number of CD8 T cells at baseline and during treatment is similar to those of healthy controls, but 2-fold higher than in patients with progressive and stable disease (PD and SD). CD8 T cell populations in PR patients show enhanced frequencies of T effector memory re-expressing CD45RA (TEMRA) cells, as well as T cells that express markers of terminal differentiation (CD95+) and egression from tumor tissue (CD69-). In PR patients, the fraction of CD8 T cells that lacks co-stimulatory receptors (CD28, ICOS, CD40L, 4-1BB, OX40) correlates significantly with the total numbers and differentiated phenotype of CD8 T cells. CONCLUSIONS This study demonstrates that high numbers of peripheral CD8 T cells expressing differentiation markers and lacking co-stimulatory receptors at baseline are associated with response to nivolumab in NSCLC patients.
Collapse
Affiliation(s)
- Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Office Be 430b, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands.
- Department of Pulmonary Diseases, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | - Edwin A Basak
- Laboratory of Translational Pharmacology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Daan P Hurkmans
- Laboratory of Translational Pharmacology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Hayri E Balcioglu
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Office Be 430b, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Yarne Klaver
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Office Be 430b, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Mandy van Brakel
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Office Be 430b, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Astrid A M Oostvogels
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Office Be 430b, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Cor H J Lamers
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Office Be 430b, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands
| | - Sander Bins
- Laboratory of Translational Pharmacology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Laboratory of Translational Pharmacology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ron H J Mathijssen
- Laboratory of Translational Pharmacology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Diseases, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Office Be 430b, Wytemaweg 80, 3015, CN, Rotterdam, The Netherlands.
| |
Collapse
|
26
|
Lafrenie RM, Speigl L, Buckner CA, Pawelec G, Conlon MS, Shipp C. Frequency of Immune Cell Subtypes in Peripheral Blood Correlates With Outcome for Patients With Metastatic Breast Cancer Treated With High-Dose Chemotherapy. Clin Breast Cancer 2019; 19:433-442. [PMID: 31383605 DOI: 10.1016/j.clbc.2019.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/16/2019] [Accepted: 05/09/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND The frequency of circulating leukocytes has been shown to be a prognostic factor in patients being treated for different types of cancer. In breast cancer, tumor-infiltrating leukocytes may predict patient outcome, but few studies have investigated such associations for circulating leukocytes. PATIENTS AND METHODS Multiparametric flow cytometry was used to examine the immunophenotypes of circulating peripheral blood mononuclear cells for 88 patients with metastatic breast cancer, which was then correlated to breast cancer-specific survival. Patients had been treated either with high-dose cyclophosphamide-containing regimens (group 1, n = 51 patients) or high-dose paclitaxel-containing regimens (group 2, n = 37 patients). RESULTS The frequency of peripheral blood CD14+ monocytes indicated prognosis for patients in group 1 (but not group 2), while higher levels of CD11c+ dendritic cells indicated a better prognosis for patients in group 2 (but not group 1). The frequency of a number of different CD4+ or CD8+ T cell subtypes also predicted prognosis for patients in group 2. For example, patients in group 2 with a higher frequency of circulating CD4+ or CD8+ naive T cells (CD45RA+CD95-CD27+CD28+) showed a poorer prognosis. In contrast, T cells were not associated with prognosis for patients in group 1. CONCLUSION Circulating leukocytes can predict clinical outcome for patients with breast cancer. Prediction of clinical outcome in this cohort of metastatic breast cancer patients was specific to the type of chemotherapy, and this finding is likely to apply to other therapies.
Collapse
Affiliation(s)
- Robert M Lafrenie
- Health Sciences North Research Institute, Sudbury, Ontario, Canada; Laurentian University, Sudbury, Ontario, Canada; Northern Ontario School of Medicine, Sudbury, Ontario, Canada.
| | - Lisa Speigl
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Carly A Buckner
- Health Sciences North Research Institute, Sudbury, Ontario, Canada; Laurentian University, Sudbury, Ontario, Canada
| | - Graham Pawelec
- Health Sciences North Research Institute, Sudbury, Ontario, Canada; Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Michael S Conlon
- Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Christopher Shipp
- Department of Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
27
|
Feng Y, Daley-Bauer LP, Roback L, Potempa M, Lanier LL, Mocarski ES. Caspase-8 restricts natural killer cell accumulation during MCMV Infection. Med Microbiol Immunol 2019; 208:543-554. [PMID: 31115653 DOI: 10.1007/s00430-019-00617-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/17/2019] [Indexed: 01/28/2023]
Abstract
Natural killer (NK) cells provide important host defense against herpesvirus infections and influence subsequent T cell control of replication and maintenance of latency. NK cells exhibit phases of expansion, contraction and memory formation in response to the natural mouse pathogen murine cytomegalovirus (MCMV). Innate and adaptive immune responses are tightly regulated in mammals to avoid excess tissue damage while preventing acute and chronic viral disease and assuring resistance to reinfection. Caspase (CASP)8 is an autoactivating aspartate-specific cysteine protease that initiates extrinsic apoptosis and prevents receptor interacting protein (RIP) kinase (RIPK)1-RIPK3-driven necroptosis. CASP8 also promotes death-independent signal transduction. All of these activities make contributions to inflammation. Here, we demonstrate that CASP8 restricts NK cell expansion during MCMV infection but does not influence NK memory. Casp8-/-Ripk3-/- mice mount higher NK response levels than Casp8+/-Ripk3-/- littermate controls or WT C57BL/6 J mice, indicating that RIPK3 deficiency alone does not contribute to NK response patterns. MCMV m157-responsive Ly49H+ NK cells support increased expansion of both Ly49H- NK cells and CD8 T cells in Casp8-/-Ripk3-/- mice. Surprisingly, hyperaccumulation of NK cells depends on the pronecrotic kinase RIPK1. Ripk1-/-Casp8-/-Ripk3-/- mice fail to show the enhanced expansion of lymphocytes observed in Casp8-/-Ripk3-/- mice even though development and homeostasis are preserved in uninfected Ripk1-/-Casp8-/-Ripk3-/- mice. Thus, CASP8 naturally regulates the magnitude of NK cell responses in response to infection where strong activation signals depend on another key regulator of death signaling, RIPK1. In addition, the strong NK cell response promotes survival of effector CD8 T cells during their expansion. Thus, hyperaccumulation of NK cells and crosstalk with T cells becomes amplified in the absence of extrinsic cell death machinery.
Collapse
Affiliation(s)
- Yanjun Feng
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, 1462 Clifton Rd. N.E, Atlanta, GA, 30322, USA
| | - Lisa P Daley-Bauer
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, 1462 Clifton Rd. N.E, Atlanta, GA, 30322, USA
| | - Linda Roback
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, 1462 Clifton Rd. N.E, Atlanta, GA, 30322, USA
| | - Marc Potempa
- Department of Microbiology and Immunology and Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, 94143, USA
| | - Lewis L Lanier
- Department of Microbiology and Immunology and Parker Institute for Cancer Immunotherapy, University of California, San Francisco, CA, 94143, USA
| | - Edward S Mocarski
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, 1462 Clifton Rd. N.E, Atlanta, GA, 30322, USA.
| |
Collapse
|
28
|
Takeuchi Y, Tanemura A, Tada Y, Katayama I, Kumanogoh A, Nishikawa H. Clinical response to PD-1 blockade correlates with a sub-fraction of peripheral central memory CD4+ T cells in patients with malignant melanoma. Int Immunol 2019; 30:13-22. [PMID: 29294043 DOI: 10.1093/intimm/dxx073] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cancer immunotherapy that blocks immune checkpoint molecules, such as PD-1/PD-L1, unleashes dysfunctional antitumor T-cell responses and has durable clinical benefits in various types of cancers. Yet its clinical efficacy is limited to a small proportion of patients, highlighting the need for identifying biomarkers that can predict the clinical response by exploring antitumor responses crucial for tumor regression. Here, we explored comprehensive immune-cell responses associated with clinical benefits using PBMCs from patients with malignant melanoma treated with anti-PD-1 monoclonal antibody. Pre- and post-treatment samples were collected from two different cohorts (discovery set and validation set) and subjected to mass cytometry assays that measured the expression levels of 35 proteins. Screening by high dimensional clustering in the discovery set identified increases in three micro-clusters of CD4+ T cells, a subset of central memory CD4+ T cells harboring the CD27+FAS-CD45RA-CCR7+ phenotype, after treatment in long-term survivors, but not in non-responders. The same increase was also observed in clinical responders in the validation set. We propose that increases in this subset of central memory CD4+ T cells in peripheral blood can be potentially used as a predictor of clinical response to PD-1 blockade therapy in patients with malignant melanoma.
Collapse
Affiliation(s)
- Yoshiko Takeuchi
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo, Japan.,Department of Respiratory Medicine and Clinical Immunology
| | - Atsushi Tanemura
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yasuko Tada
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo, Japan
| | - Ichiro Katayama
- Department of Dermatology, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute/Exploratory Oncology Research & Clinical Trial Center (EPOC), National Cancer Center, Tokyo, Japan.,Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
29
|
Sagrillo-Fagundes L, Bienvenue-Pariseault J, Legembre P, Vaillancourt C. An insight into the role of the death receptor CD95 throughout pregnancy: Guardian, facilitator, or foe. Birth Defects Res 2019; 111:197-211. [PMID: 30702213 DOI: 10.1002/bdr2.1470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 01/16/2019] [Indexed: 12/24/2022]
Abstract
The prototype death receptor CD95 (Fas) and its ligand, CD95L (FasL), have been thoroughly studied due to their role in immune homeostasis and elimination of infected and transformed cells. The fact that CD95 is present in female reproductive cells and modulated during embryogenesis and pregnancy has raised interest in its role in immune tolerance to the fetoplacental unit. CD95 has been shown to be critical for proper embryonic formation and survival. Moreover, altered expression of CD95 or its ligand causes autoimmunity and has also been directly involved in recurrent pregnancy losses and pregnancy disorders. The objective of this review is to summarize studies that evaluate the mechanisms involved in the activation of CD95 to provide an updated global view of its effect on the regulation of the maternal immune system. Modulation of the CD95 system components may be the immune basis of several common pregnancy disorders.
Collapse
Affiliation(s)
- Lucas Sagrillo-Fagundes
- Department of Environmental toxicology and Chemical Pharmacology, INRS - Institut Armand-Frappier and Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Laval, Quebec, Canada
| | - Josianne Bienvenue-Pariseault
- Department of Environmental toxicology and Chemical Pharmacology, INRS - Institut Armand-Frappier and Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Laval, Quebec, Canada
| | - Patrick Legembre
- Oncogenesis, Stress & Signaling Laboratory INSERM ERL440, Centre Eugène Marquis, Inserm U1242, Equipe Ligue Contre Le Cancer, Rennes, France
| | - Cathy Vaillancourt
- Department of Environmental toxicology and Chemical Pharmacology, INRS - Institut Armand-Frappier and Center for Interdisciplinary Research on Well-Being, Health, Society and Environment, Laval, Quebec, Canada
| |
Collapse
|
30
|
Maimela NR, Liu S, Zhang Y. Fates of CD8+ T cells in Tumor Microenvironment. Comput Struct Biotechnol J 2018; 17:1-13. [PMID: 30581539 PMCID: PMC6297055 DOI: 10.1016/j.csbj.2018.11.004] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/14/2018] [Accepted: 11/18/2018] [Indexed: 12/24/2022] Open
Abstract
Studies have reported a positive correlation between elevated CD8+ T cells in the tumor microenvironment (TME) and good prognosis in cancer. However, the mechanisms linking T cell tumor-infiltration and tumor rejection are yet to be fully understood. The cells and factors of the TME facilitate tumor development in various ways. CD8+ T cell function is influenced by a number of factors, including CD8+ T cell trafficking and localization into tumor sites; as well as CD8+ T cell growth and differentiation. This review highlights recent literature as well as currently evolving concepts regarding the fates of CD8+ T cells in the TME from three different aspects CD8+ T cell trafficking, differentiation and function. A thorough understanding of factors contributing to the fates of CD8+ T cells will allow researchers to develop new strategies and improve on already existing strategies to facilitate CD8+ T cell mediated anti-tumor function, impede T cell dysfunction and modulate the TME into a less immunosuppressive TME.
Collapse
Affiliation(s)
| | - Shasha Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou 450052, China
| |
Collapse
|
31
|
Rallón N, García M, García-Samaniego J, Cabello A, Álvarez B, Restrepo C, Nistal S, Górgolas M, Benito JM. Expression of PD-1 and Tim-3 markers of T-cell exhaustion is associated with CD4 dynamics during the course of untreated and treated HIV infection. PLoS One 2018. [PMID: 29518102 PMCID: PMC5843247 DOI: 10.1371/journal.pone.0193829] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Introduction T-cell exhaustion has been involved in the pathogenesis of HIV infection. We have longitudinally analyzed PD1 and Tim3 surrogate markers of T-cells exhaustion, in parallel with other markers of HIV progression, and its potential association with CD4 changes in treated and untreated infection. Patients and methods 96 HIV patients, 49 of them followed in the absence of cART (cART-naïve group) and 47 after initiation of cART (cART group) were included and followed for a median of 43 [IQR: 31–60] months. PD1 and Tim3 expression, CD8 T-cells activation, recent thymic emigrants, activation/apoptosis and turnover of CD4 cells were assessed at baseline and during follow up. Univariate and multivariate associations with CD4 evolution were explored. Results Parameters significantly associated with CD4 depletion in cART-naïve group were: baseline level (p = 0.02) and variation (p = 0.002) of PD1 and Tim3 co-expression on CD8, and variation of CD95 expression on CD4 (p = 0.007). Parameters significantly associated with CD4 restoration in cART group were: baseline level of CD38+HLADR- subset of CD8 (p = 0.01), variation of PD1 expression on CD8 (p = 0.036), variation of Tim3 expression on CD4 (p = 0.039) and variation of CD95 expression on CD4 (p = 0.035). Conclusions Our results suggest that PD1 and Tim3 markers of exhaustion have a pivotal role in CD4 dynamics in HIV patients and its down-regulation would be a desirable effect of immunotherapies aimed to restore CD4 T-cell pool during progression of HIV infection.
Collapse
Affiliation(s)
- Norma Rallón
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
- * E-mail:
| | - Marcial García
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | | | - Alfonso Cabello
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Beatriz Álvarez
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Clara Restrepo
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - Sara Nistal
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| | - Miguel Górgolas
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - José M. Benito
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz, Universidad Autónoma de Madrid (IIS-FJD, UAM), Madrid, Spain
- Hospital Universitario Rey Juan Carlos, Móstoles, Spain
| |
Collapse
|
32
|
Manjili MH. A Theoretical Basis for the Efficacy of Cancer Immunotherapy and Immunogenic Tumor Dormancy: The Adaptation Model of Immunity. Adv Cancer Res 2018; 137:17-36. [PMID: 29405975 DOI: 10.1016/bs.acr.2017.11.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In the past decades, a variety of strategies have been explored to cure cancer by means of immunotherapy, which is less toxic compared with chemotherapy or radiation therapy, and could establish memory for long-lasting protection against tumor recurrence. These endeavors have been successful in offering therapeutic antibodies, vaccines, or cellular immunotherapies, which resulted in prolonging survival of some cancer patients; however, complete cures have not been consistently achieved. The conception, design, and implementation of these promising immunotherapeutic strategies have been influenced by two schools of thought in immunology, which include the "self-nonself" (SNS) model and the "danger" model. Further progress in cancer immunotherapy to achieve consistent cancer cures requires an evolution in our understanding of how the immune system works. The purpose of this review is to revisit premises and limitations of the SNS and danger models based on the outcomes of cancer immunotherapies by suggesting that both models are two sides of the same coin describing how the immune response is induced against cancer. However, neither explains how the immune response succeeds or fails in eliminating the tumor. To this end, the adaptation model has been proposed to explain efficacy of the immune response for achieving cancer cure.
Collapse
Affiliation(s)
- Masoud H Manjili
- VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
33
|
Preclinical evaluation of the efficacy, pharmacokinetics and immunogenicity of JS-001, a programmed cell death protein-1 (PD-1) monoclonal antibody. Acta Pharmacol Sin 2017; 38:710-718. [PMID: 28317872 DOI: 10.1038/aps.2016.161] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/06/2016] [Indexed: 12/15/2022] Open
Abstract
JS-001 is the first monoclonal antibody (mAb) against programmed cell death protein-1 (PD-1) approved by the China Food and Drug Administration (CFDA) into the clinical trails. To date, however, no pre-clinical pharmacological and pharmacokinetic (PK) data are available. In this study, we investigated the efficacy of JS-001 and conducted a preclinical PK study, including the monitoring of anti-drug antibodies (ADAs). We found that JS-001 specifically bound to PD-1 antigen with an EC50 of 21 nmol/L, and competently blocked the binding of PD-1 antigen to PD-L1 and PD-L2 with IC50 of 3.0 and 3.1 nmol/L, respectively. Furthermore, JS-001 displayed distinct species cross-reactivity: it could bind to the PD-1 antigen on the peripheral blood mononuclear cells (PBMCs) of humans and cynomolgus monkeys, but not to those of mice and woodchucks; the Kd values for the interaction between JS-001 and PD-1 antigens on CD8+ T cells of human and cynomolgus monkey were 2.1 nmol/L and 1.2 nmol/L, respectively. In vitro, treatment with JS-001 (0.01-10 μg/mL) dose-dependently stimulated human T cell proliferation, as well as IFN-γ and TNF-α secretion. In HBsAg-vaccinated cynomolgus monkeys, the expression of PD-1+/CD4+ and PD-1+/CD8+ was significantly elevated, intramuscular injection of JS-001 (1 and 10 mg/kg) resulted in dramatic decreases in PD-1+/CD4+ and PD-1+/CD8+ expression in a dose-dependent manner, which was supported by PD-1 receptor occupancy (RO) results. In the PK study, 18 cynomolgus monkeys treated with single, ascending doses of 1, 10, and 75 mg/kg, and another 6 cynomolgus monkeys received 10 mg/kg successive administration. The plasma clearance of JS-001 followed a linear PK profile with single administration in the 1 and 10 mg/kg groups and a non-linear PK profile in the 75 mg/kg group. In the successive 10 mg/kg administration group, no drug accumulation was observed. But the AUC from the last exposure was lower than that of the first administration, which was probably due to the production of ADAs, as demonstrated in immunogenicity study. These non-clinical data are encouraging and provide a basis for the efficacy and safety of JS-001 in clinical trials.
Collapse
|
34
|
Balomenos D, Shokri R, Daszkiewicz L, Vázquez-Mateo C, Martínez-A C. On How Fas Apoptosis-Independent Pathways Drive T Cell Hyperproliferation and Lymphadenopathy in lpr Mice. Front Immunol 2017; 8:237. [PMID: 28344578 PMCID: PMC5344898 DOI: 10.3389/fimmu.2017.00237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 02/20/2017] [Indexed: 01/20/2023] Open
Abstract
Fas induces massive apoptosis in T cells after repeated in vitro T cell receptor (TCR) stimulation and is critical for lymphocyte homeostasis in Fas-deficient (lpr) mice. Although the in vitro Fas apoptotic mechanism has been defined, there is a large conceptual gap between this in vitro phenomenon and the pathway that leads to in vivo development of lymphadenopathy and autoimmunity. A striking abnormality in lpr mice is the excessive proliferation of CD4+ and CD8+ T cells, and more so of the double-negative TCR+CD4−CD8−B220+ T cells. The basis of lpr T cell hyperproliferation remains elusive, as it cannot be explained by Fas-deficient apoptosis. T cell-directed p21 overexpression reduces hyperactivation/hyperproliferation of all lpr T cell subtypes and lymphadenopathy in lpr mice. p21 controls expansion of repeatedly stimulated T cells without affecting apoptosis. These results confirm a direct link between hyperactivation/hyperproliferation, autoreactivity, and lymphadenopathy in lpr mice and, with earlier studies, suggest that Fas apoptosis-independent pathways control lpr T cell hyperproliferation. lpr T cell hyperproliferation could be an indirect result of the defective apoptosis of repeatedly stimulated lpr T cells. Nonetheless, in this perspective, we argue for an alternative setting, in which lack of Fas would directly cause lpr T cell hyperactivation/hyperproliferation in vivo. We propose that Fas/Fas ligand (FasL) acts as an activation inhibitor of recurrently stimulated T cells, and that its disruption causes overexpansion of T cells in lpr mice. Research to define the underlying mechanism of this Fas/FasL effect could resolve the phenotype of lpr mice and lead to therapeutics for related human syndromes.
Collapse
Affiliation(s)
- Dimitrios Balomenos
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Rahman Shokri
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Lidia Daszkiewicz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Cristina Vázquez-Mateo
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| | - Carlos Martínez-A
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB-CSIC), UAM Campus de Cantoblanco , Madrid , Spain
| |
Collapse
|
35
|
Abstract
Two of the most important contemporary emerging viruses that affect human health in Africa are Ebola virus (EBOV) and Lassa virus (LASV). The 2013-2016 West African outbreak of EBOV was responsible for more than 11,000 deaths, primarily in Guinea, Sierra Leone and Liberia. LASV is constantly emerging in these and surrounding West African countries, with an estimate of more than 500,000 cases of Lassa fever, and approximately 5,000 deaths, annually. Both EBOV and LASV are zoonotic, and human infection often results in a severe haemorrhagic fever in both cases. However, the contribution of specific immune responses to disease differs between EBOV and LASV. This Review examines innate and adaptive immune responses to these viruses with the goal of delineating responses that are associated with protective versus pathogenic outcomes.
Collapse
|
36
|
A20 Curtails Primary but Augments Secondary CD8 + T Cell Responses in Intracellular Bacterial Infection. Sci Rep 2016; 6:39796. [PMID: 28004776 PMCID: PMC5177869 DOI: 10.1038/srep39796] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 11/29/2016] [Indexed: 12/26/2022] Open
Abstract
The ubiquitin-modifying enzyme A20, an important negative feedback regulator of NF-κB, impairs the expansion of tumor-specific CD8+ T cells but augments the proliferation of autoimmune CD4+ T cells. To study the T cell-specific function of A20 in bacterial infection, we infected T cell-specific A20 knockout (CD4-Cre A20fl/fl) and control mice with Listeria monocytogenes. A20-deficient pathogen-specific CD8+ T cells expanded stronger resulting in improved pathogen control at day 7 p.i. Imaging flow cytometry revealed that A20-deficient Listeria-specific CD8+ T cells underwent increased apoptosis and necroptosis resulting in reduced numbers of memory CD8+ T cells. In contrast, the primary CD4+ T cell response was A20-independent. Upon secondary infection, the increase and function of pathogen-specific CD8+ T cells, as well as pathogen control were significantly impaired in CD4-Cre A20fl/fl mice. In vitro, apoptosis and necroptosis of Listeria-specific A20-deficient CD8+ T cells were strongly induced as demonstrated by increased caspase-3/7 activity, RIPK1/RIPK3 complex formation and more morphologically apoptotic and necroptotic CD8+ T cells. In vitro, A20 limited CD95L and TNF-induced caspase3/7 activation. In conclusion, T cell-specific A20 limited the expansion but reduced apoptosis and necroptosis of Listeria-specific CD8+ T cells, resulting in an impaired pathogen control in primary but improved clearance in secondary infection.
Collapse
|
37
|
Matou-Nasri S, Rabhan Z, Al-Baijan H, Al-Eidi H, Yahya WB, Al Abdulrahman A, Almobadel N, Alsubeai M, Al Ghamdi S, Alaskar A, AlBalwi M, Alzahrani M, Alabdulkareem I. CD95-mediated apoptosis in Burkitt's lymphoma B-cells is associated with Pim-1 down-regulation. Biochim Biophys Acta Mol Basis Dis 2016; 1863:239-252. [PMID: 27641442 DOI: 10.1016/j.bbadis.2016.09.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 09/09/2016] [Accepted: 09/14/2016] [Indexed: 01/17/2023]
Abstract
B-cells of the high-grade non-Hodgkin lymphoma Burkitt's lymphoma (BL) overexpress survival oncoproteins, including the proviral integration site for Moloney murine leukaemia virus kinase (Pim)-1, and become apoptosis resistant. Activated death receptor CD95 after ligation with anti-CD95 monoclonal antibody (mAb) resulted in the regression of BL via induction of apoptosis, suggesting a decrease of survival protein expression. Here, CD95-mediated apoptotic pathways in BL B-cell lines (Raji and Daudi) following treatment with anti-CD95 mAb was investigated with the cause-and-effects on pim-1 gene expression, in comparison with leukemic cell line (K562) used as CD95-negative cells. Immunohistochemical staining for CD95 and Pim-1 was performed, and the effects of anti-CD95 mAb on apoptotic signalling using western blotting, on caspase activity and cell survival of BL B-cell and leukemic cell lines were determined. We showed that Raji cells expressed more CD95 receptors than Daudi cells. Half of each population underwent apoptosis accompanied by decreased cell viability after anti-CD95 mAb treatment. Distinct extrinsic and intrinsic CD95-mediated apoptotic pathways in Raji and Daudi cells were revealed by high caspase activity and mitochondrial outer membrane permeabilization, respectively. We observed decreased Pim-1 transcript and protein expression levels with increased heat-shock protein (Hsp)70 and decreased Hsp90 expression in anti-CD95 mAb-treated cells. Throughout the study, K562 cells did not undergo apoptosis upon anti-CD95 mAb treatment. Pim-1 knockdown following to stable transfection with plasmid vectors induced apoptosis and decreased viability of BL and K562 cells. Therefore, CD95-mediated apoptosis induces Pim-1 down-regulation in BL B-cells, but Pim-1 down-regulation cannot fully eradicate BL and leukaemia.
Collapse
Affiliation(s)
- Sabine Matou-Nasri
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Zaki Rabhan
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Haya Al-Baijan
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Hamad Al-Eidi
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Wesam Bin Yahya
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Abdelkareem Al Abdulrahman
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Nasser Almobadel
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Mona Alsubeai
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Saleh Al Ghamdi
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia
| | - Ahmed Alaskar
- KAIMRC, King Saud bin-Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), MNGHA, Riyadh 11426, Saudi Arabia
| | - Mohammed AlBalwi
- Pathology and Laboratory Medicine, KAMC, MNGHA, Riyadh 11426, Saudi Arabia
| | | | - Ibrahim Alabdulkareem
- Cell and Gene Therapy Group, Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard - Health Affairs (MNGHA), P.O. Box 22490, Riyadh 11426, Saudi Arabia.
| |
Collapse
|
38
|
Yadav A, Kumar A, Das M, Tripathi A. Sodium benzoate, a food preservative, affects the functional and activation status of splenocytes at non cytotoxic dose. Food Chem Toxicol 2015; 88:40-7. [PMID: 26706697 DOI: 10.1016/j.fct.2015.12.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 12/11/2015] [Accepted: 12/14/2015] [Indexed: 12/31/2022]
Abstract
Sodium benzoate (SB) is a widely used food preservative due to its bacteriostatic and fungistatic properties. The acceptable daily intake of SB is 5 mg/kg-bw, however, it has been found to be used in the food commodities at relatively high levels (2119 mg/kg). Earlier studies on SB have shown its immunosuppressive properties, but comprehensive immunotoxicity data is lacking. Our studies have shown that SB was non cytotoxic in splenocytes up to 1000 μg/ml for 72 h, however at 2500 μg/ml it was found to be cytotoxic. Thus, 1000 μg/ml dose of SB was chosen for the subsequent experiments. SB significantly suppresses the proliferation of Con A and LPS stimulated splenocytes at 72 h, while allogenic response of T cells was significantly decreased after 96 h. SB did not affect the relative expression of CD3e or CD4 molecules following 72 h exposure, however, it downregulated the relative expression of CD8 co-receptor. Further, exposure of splenocytes to SB for 72 h led to reduced expression of CD28 and CD95, which play a vital role in T cell activation. SB also suppresses the relative expression of CD19, CD40 and CD95 receptors on B cells after 72 h. In addition to the functional responses, SB lowered the expression of IL4, IL6, IFNγ and IL17 cytokines in Con A stimulated splenocytes; and IL6, IFNγ and TNFα in LPS stimulated splenocytes following 48 h of exposure. Taken together, the present study is suggestive of the immunomodulatory potential of SB.
Collapse
Affiliation(s)
- Ashish Yadav
- Food Toxicology Lab, Food, Drug and Chemical Toxicology Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), M.G. Marg, Lucknow, 226001, Uttar Pradesh, India; Molecular Immunology Lab, School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Arvind Kumar
- Molecular Immunology Lab, School of Biotechnology, Faculty of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Mukul Das
- Food Toxicology Lab, Food, Drug and Chemical Toxicology Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), M.G. Marg, Lucknow, 226001, Uttar Pradesh, India.
| | - Anurag Tripathi
- Food Toxicology Lab, Food, Drug and Chemical Toxicology Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), M.G. Marg, Lucknow, 226001, Uttar Pradesh, India.
| |
Collapse
|
39
|
Vitkina TI, Yankova VI, Gvozdenko TA, Kuznetsov VL, Krasnikov DV, Nazarenko AV, Chaika VV, Smagin SV, Tsatsakis AΜ, Engin AB, Karakitsios SP, Sarigiannis DA, Golokhvast KS. The impact of multi-walled carbon nanotubes with different amount of metallic impurities on immunometabolic parameters in healthy volunteers. Food Chem Toxicol 2015; 87:138-47. [PMID: 26683310 DOI: 10.1016/j.fct.2015.11.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 11/29/2015] [Accepted: 11/30/2015] [Indexed: 11/15/2022]
Abstract
The impact of two types of multi-walled carbon nanotubes (MWCNTs) (12-14 nm) with different content of metallic impurities (purified and unpurified nanotubes) on peroxidation processes, the status of immune cells in healthy volunteers and gene expression combined to pathway analysis was studied in vitro. From the study it was shown that the main mechanism of action for both types of MWCNTs is induction of oxidative stress, the intensity of which is directly related to the amount of metallic impurities. Unpurified MWCNTs produced twice as high levels of oxidation than the purified CNTs inducing thus more intense mitochondrial dysfunction. All the above were also verified by gene expression analysis of 2 different human cellular cultures (lung epithelium and keratinoma cells) and the respective pathway analysis; modulation of genes activating the NFkB pathway is associated to inflammatory responses. This may cause a perturbation in the IL-6 signaling pathway in order to regulate inflammatory processes and compensate for apoptotic changes. A plausible hypothesis for the immunological effects observed in vivo, are considered as the result of the synergistic effect of systemic (mediated by cells of the routes of exposure) and local inflammation (blood cells).
Collapse
Affiliation(s)
- T I Vitkina
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Research Institute of Medical Climatology and Rehabilitation Treatment, Vladivostok, Russia
| | - V I Yankova
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Research Institute of Medical Climatology and Rehabilitation Treatment, Vladivostok, Russia
| | - T A Gvozdenko
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Research Institute of Medical Climatology and Rehabilitation Treatment, Vladivostok, Russia
| | - V L Kuznetsov
- Boreskov Institute of Catalysis SB RAS, Novosibirsk, Russia
| | - D V Krasnikov
- Boreskov Institute of Catalysis SB RAS, Novosibirsk, Russia
| | - A V Nazarenko
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Research Institute of Medical Climatology and Rehabilitation Treatment, Vladivostok, Russia
| | - V V Chaika
- Far Eastern Federal University, Vladivostok, Russia
| | - S V Smagin
- Far Eastern Federal University, Vladivostok, Russia
| | - A Μ Tsatsakis
- Far Eastern Federal University, Vladivostok, Russia; University of Crete, Heraklion, Greece
| | - A B Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, 06330, Hipodrom, Ankara, Turkey
| | - S P Karakitsios
- Aristotle University of Thessaloniki, Department of Chemical Engineering, Thessaloniki, Greece
| | - D A Sarigiannis
- Aristotle University of Thessaloniki, Department of Chemical Engineering, Thessaloniki, Greece; Chair of Environmental Health Engineering, Institute for Advanced Study, Pavia, Italy.
| | - K S Golokhvast
- Vladivostok Branch of Far Eastern Scientific Center of Physiology and Pathology of Respiration, Research Institute of Medical Climatology and Rehabilitation Treatment, Vladivostok, Russia; Far Eastern Federal University, Vladivostok, Russia
| |
Collapse
|
40
|
Momesso dos Santos CM, Sato FT, Cury-Boaventura MF, Guirado-Rodrigues SH, Caçula KG, Gonçalves Santos CC, Hatanaka E, de Oliveira HH, Santos VC, Murata G, Borges-Silva CN, Hirabara SM, Pithon-Curi TC, Gorjão R. Effect of regular circus physical exercises on lymphocytes in overweight children. PLoS One 2015; 10:e0120262. [PMID: 25826263 PMCID: PMC4380297 DOI: 10.1371/journal.pone.0120262] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/21/2015] [Indexed: 12/26/2022] Open
Abstract
Obesity associated with a sedentary lifestyle can lead to changes in the immune system balance resulting in the development of inflammatory diseases. The aim of this study was to compare lymphocyte activation mechanisms between overweight children practicing regular circus physical exercises with non-exercised children. The study comprised 60 pubescent children randomly divided into 4 groups: Overweight Children (OWC) (10.67 ± 0.22 years old), Overweight Exercised Children (OWE) (10.00 ± 0.41 years old), Eutrophic Children (EC) (11.00 ± 0.29 years old) and Eutrophic Exercised Children (EE) (10.60 ± 0.29 years old). OWE and EE groups practiced circus activities twice a week, for 4.3 ± 0.5 and 4.4 ± 0.5 months, respectively. Percentage of T regulatory cells (Treg) and the expression of CD95 and CD25 in CD4+ lymphocytes were evaluated by flow cytometry. Lymphocyte proliferation capacity was measured by [14C]-thymidine incorporation and mRNA expression of IL-35, TGF-beta, IL-2 and IL-10 by real-time PCR. Lymphocyte proliferation was higher in OWC and OWE groups compared with the EC (3509 ± 887; 2694 ± 560, and 1768 ± 208 cpm, respectively) and EE (2313 ± 111 cpm) groups. CD95 expression on lymphocytes was augmented in the EC (953.9 ± 101.2) and EE groups (736.7 ± 194.6) compared with the OWC (522.1 ± 125.2) and OWE groups (551.6 ± 144.5). CTLA-4 expression was also lower in the OWC and OWE groups compared with the EC and EE groups. Percentage of Treg, IL-35, and IL-10 mRNA expression were lower in the OWC and OWE groups compared with the EC and EE groups. In conclusion, overweight children present altered immune system balance characterized by elevated lymphocyte proliferation due to a decrease in T regulatory cell percentage. These effects were partially reverted by moderate physical exercise, as demonstrated by decreased lymphocyte proliferation.
Collapse
Affiliation(s)
- Cesar Miguel Momesso dos Santos
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Fábio Takeo Sato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Fernanda Cury-Boaventura
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Silvia Helena Guirado-Rodrigues
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Kim Guimaraes Caçula
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Cristiane Cassoni Gonçalves Santos
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Elaine Hatanaka
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Heloisa Helena de Oliveira
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Vinicius Coneglian Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Gilson Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cristina Neves Borges-Silva
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Sandro Massao Hirabara
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Tania Cristina Pithon-Curi
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Renata Gorjão
- Institute of Physical Activity Sciences and Sports, Post-Graduate Program in Human Movement Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| |
Collapse
|
41
|
Aldahlawi AM, Elshal MF, Ashgan FT, Bahlas S. Chemokine receptors expression on peripheral CD4-lymphocytes in rheumatoid arthritis: Coexpression of CCR7 and CD95 is associated with disease activity. Saudi J Biol Sci 2015; 22:453-8. [PMID: 26150752 PMCID: PMC4487268 DOI: 10.1016/j.sjbs.2015.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 02/07/2015] [Accepted: 02/08/2015] [Indexed: 01/11/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial inflammation triggered by infiltrating CD4 lymphocytes. The positioning and activation of lymphocyte in inflamed synovial tissues are dependent on a number of factors including their chemokine receptor expression profile. We aimed to investigate which chemokine receptors pattern correlate with serum cytokine levels and with disease activity. Forty patients with RA (34 female and 6 male) with age range from 21 to 68 years were included. Twenty healthy volunteers (16 female and 4 male) with matched age (range 21-48 years) were served as healthy controls (HCs). Expression of chemokine receptors (CCR5, CX3CR1 and CCR7) together with the apoptosis-related marker (CD95) was analyzed using three-color flow cytometry analysis after gating on CD4(+) peripheral blood lymphocytes. Plasma levels of IL-6, IL-10, IL-12 and TNF-α cytokines were measured in all participants using ELISA. Disease activity score (DAS28-CRP) system was assessed and active disease was defined as DAS28 ⩾3.2. Twenty-five (62.4%) patients were classified as active RA (ARA) and 15 (37.5%) patients with inactive RA (IRA). Percentages of CD4(+) lymphocytes expressing CD95 with either of CCR7 or CCR5 were significantly higher in ARA compared to IRA and HCs groups, while the expression of CX3CR1 on T-cells was found significantly lower in both CD95(-) and CD95(+) T-cells in RA groups than HC. Percentages of CD4(+)CD95(+)CCR7(+) cells correlated positively with IL-6 (r = 0.390). Whereas CD4(+)CD95(+)CX3CR1(+) were negatively correlated with TNF-α (r = -0.261). Correlation of CD4(+)CD95(+)CCR7(+) T cell subset with disease activity and inflammatory cytokines suggests a role for this cell subset in the pathogenesis of RA. Further investigation will be required to fully characterize this cell subset and its role in disease progression.
Collapse
Affiliation(s)
- Alia M. Aldahlawi
- Biological Sciences Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Corresponding author at: Biological Sciences Department, Faculty of Sciences, King Abdulaziz University, P.O. Box 80200, Jeddah 21589, Saudi Arabia. Tel.: +966 505357982.
| | - Mohammed F. Elshal
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Sadat City, Egypt
| | - Fai T. Ashgan
- Biological Sciences Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami Bahlas
- Rheumatic Disease Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
42
|
Impact of recombinant globular adiponectin on early warm ischemia-reperfusion injury in rat bile duct after liver transplantation. Sci Rep 2014; 4:6426. [PMID: 25233838 PMCID: PMC4168283 DOI: 10.1038/srep06426] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 08/29/2014] [Indexed: 12/28/2022] Open
Abstract
Adiponectin (APN) is an adipocyte protein with anti-diabetic properties, which has been recently revealed to have anti-inflammatory activity in organ ischemia- reperfusion injury (IRI). However, little is known about its function in bile duct IRI after liver transplantation. Therefore, we investigated whether APN affects early warm IRI in rat bile duct using a liver autologous transplantation model. In our study, rats were randomly divided into three experimental groups: a sham group, a IRI group, and a APN group. The serum enzyme levels and BDISS scores of bile duct histology associated with bile duct injury, decreased after administration of APN. Subsequently, the expression of proinflammatory cytokines, such as tumor necrosis factor(TNF-α),.interleukin-6(IL-6) and myeloperoxidase (MPO) decreased. Furthermore, pretreatment with APN suppressed the activation of nuclear factor-kappa B (NF-κB) (p65), a transcription factor involved in inflammatory reactions, compared to other two groups. Administration of APN also downregulated the expression of Fas protein and attenuated caspase-3 activity to decrease bile duct apoptosis. Our results illustrate that APN protects the rat bile duct against early warm IRI by suppressing the inflammatory response and hepatocyte apoptosis, and NF-κB (p65) plays an important role in this process.
Collapse
|
43
|
Abstract
Although 'self-nonself' and 'danger' theories have improved our understanding of the immune system, successful immunotherapy of cancer and many autoimmune diseases still remain far from reach. This indicates that our knowledge of how the immune system decides to respond effectively or ineffectively is limited. Emerging evidence suggest that decision-making during the immune response is not solely determined by 'nonself' entity of the antigen or damage-associated 'danger' signals. This article provides an overview of the 'self-nonself' and 'danger' models, and suggests that 'adaptation' signals are needed to guarantee immunological tolerance that has been observed during the immune response toward 'self', 'nonself' or even 'danger'. This should be facilitated by dynamic expression of adapting receptors (ARs) and adapting ligands on cells of the immune system and other somatic cells. Any alterations in the expression of ARs on certain tissues would result in tissue-specific autoimmune diseases or spontaneous regression of cancer. Identification of such ARs and their nominal adapting ligands could lead to the discovery of currently unknown receptors and their implications in the treatment of cancer, solid organ transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Masoud H Manjili
- Department of Microbiology & Immunology, Virginia Commonwealth University, Massey Cancer Center, Box 980035, 401 College Street, Richmond, VA 23298, USA.
| |
Collapse
|
44
|
Torrentes-Carvalho A, Marinho CF, de Oliveira-Pinto LM, de Oliveira DB, Damasco PV, Cunha RV, de Souza LJ, de Azeredo EL, Kubelka CF. Regulation of T lymphocyte apoptotic markers is associated to cell activation during the acute phase of dengue. Immunobiology 2014; 219:329-40. [DOI: 10.1016/j.imbio.2013.11.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 11/14/2013] [Indexed: 12/29/2022]
|
45
|
Differential protein–protein interactions of full length human FasL and FasL fragments generated by proteolysis. Exp Cell Res 2014; 320:290-301. [DOI: 10.1016/j.yexcr.2013.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 10/25/2013] [Accepted: 11/19/2013] [Indexed: 01/14/2023]
|
46
|
Ebsen H, Schröder A, Kabelitz D, Janssen O. Differential surface expression of ADAM10 and ADAM17 on human T lymphocytes and tumor cells. PLoS One 2013; 8:e76853. [PMID: 24130797 PMCID: PMC3793918 DOI: 10.1371/journal.pone.0076853] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 08/26/2013] [Indexed: 12/25/2022] Open
Abstract
A disintegrin and metalloproteases (ADAMs) have been implicated in many processes controlling organismic development and integrity. Important substrates of ADAM proteases include growth factors, cytokines and their receptors and adhesion proteins. The inducible but irreversible cleavage of their substrates alters cell-cell communication and signaling. The crucial role of ADAM proteases (e.g. ADAM10 and 17) for mammalian development became evident from respective knockout mice, that displayed pre- or perinatal lethality with severe defects in many organs and tissues. Although many substrates for these two ADAM proteases were identified over the last decade, the regulation of their surface appearance, their enzymatic activity and their substrate specificity are still not well understood. We therefore analyzed the constitutive and inducible surface expression of ADAM10 and ADAM17 on a variety of human T cell and tumor cell lines. We demonstrate that ADAM10 is constitutively present at comparably high levels on the majority of the tested cell types. Stimulation with phorbol ester and calcium ionophore does not significantly alter the amount of surface ADAM10, except for a slight down-regulation from T cell blasts. Using FasL shedding as a readout for ADAM10 activity, we show that PKC activation and calcium mobilization are both prerequisite for activation of ADAM10 resulting in a production of soluble FasL. In contrast to ADAM10, the close relative ADAM17 is detected at only low levels on unstimulated cells. ADAM17 surface expression on T cell blasts is rapidly induced by stimulation. Since this inducible mobilization of ADAM17 is sensitive to inhibitors of actin filament formation, we propose that ADAM17 but not ADAM10 is prestored in a subcellular compartment that is transported to the cell surface in an activation- and actin-dependent manner.
Collapse
Affiliation(s)
- Henriette Ebsen
- University of Kiel, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Alexandra Schröder
- University of Kiel, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dieter Kabelitz
- University of Kiel, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Ottmar Janssen
- University of Kiel, Institute for Immunology, University Hospital Schleswig-Holstein Campus Kiel, Kiel, Germany
- * E-mail:
| |
Collapse
|
47
|
Su CC, Lin HC, Lin YP, Shan YS, Yang BC. Expression of Th17-related genes in PHA/IL-2-activated human T cells by Fas signaling via caspase-1- and Stat3-dependent pathway. Cell Immunol 2013; 281:101-10. [PMID: 23590971 DOI: 10.1016/j.cellimm.2013.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 11/19/2012] [Accepted: 03/19/2013] [Indexed: 01/06/2023]
Abstract
T helper 17 (Th17) cells, which produce interleukin 17 (IL-17), are involved in the pathogenesis of autoimmune diseases and inflammatory conditions. Th17 cells have been detected in many Fas ligand-positive tumors. This study investigates the expression of Th17-related genes in PHA/IL-2-activated human T cells upon Fas ligation. Activated T cells transiently express RORγt, IL-17A, and IL-17F. A subsequent Fas receptor stimulation or contact with FasL-expressing glioma cells significantly prolongs the induction of RORγt and Th17-related cytokines. Treatments with inhibitors of caspase-1 and Stat3 reduce the Fas-signal-associated induction of RORγt, IL-17A, and IL-17F, as well as the phosphorylation of Stat3. Although the ligation of Fas results in caspase-8 cleavage and ERK1/2 phosphorylation, inhibitors for caspase-8 and MEK have no effect on the expressions of RORγt, IL-17A, and IL-17F. The results suggest that the Fas signal favors the Th17-phenotypic features of human T cells through the caspase-1/Stat3 signaling pathway.
Collapse
Affiliation(s)
- Chung-Chen Su
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | | | | | | |
Collapse
|
48
|
Wang B, Zhang Q, Zhu B, Cui Z, Zhou J. Protective effect of gadolinium chloride on early warm ischemia/reperfusion injury in rat bile duct during liver transplantation. PLoS One 2013; 8:e52743. [PMID: 23341905 PMCID: PMC3544894 DOI: 10.1371/journal.pone.0052743] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 11/21/2012] [Indexed: 01/15/2023] Open
Abstract
Background Activation of Kupffer cell (KC) is acknowledged as a key event in the initiation and perpetuation of bile duct warm ischemia/reperfusion injury. The inhibitory effect of gadolinium chloride (GdCl3) on KC activation shows potential as a protective intervention in liver injury, but there is less research with regard to bile duct injury. Methods Sixty-five male Sprague-Dawley rats (200–250 g) were randomly divided into three experimental groups: a sham group (n = 15), a control group (n = 25), and a GdCl3 group (n = 25). Specimen was collected at 0.5, 2, 6, 12 and 24 h after operation. Alanine aminotransferase (ALT), alkaline phosphatase (ALP) and total bilirubin (TBIL) of serum were measured. Tumor necrosis factor-α (TNF-α), Capase-3 activity and soluble Fas (sFas) were detected. The pathologic changes of bile duct were observed. Immunochemistry for bile duct Fas was performed. Apoptosis of bile duct cells was evaluated by the terminal UDP nick end labeling assay. Results GdCl3 significantly decreased the levels of ALT, ALP and TBIL at 2, 6, 12, and 24 h, and increased serum sFas at 2, 6 and 12 h (P<0.05). TNF-α was lower in the GdCl3 group than in the control group at 2, 6, 12 and 24 h (P<0.05). Preadministration of GdCl3 significantly reduced the Caspase-3 activity and bile duct cell apoptosis at 2, 6, 12 and 24 h. After operation for 2, 6 and 12 h, the expression of Fas protein was lower in the GdCl3 group than in the control group (P<0.05). Conclusions GdCl3 plays an important role in suppressing bile duct cell apoptosis, including decreasing ALT, ALP, TBIL and TNF-α; suppressing Fas-FasL-Caspase signal transduction during transplantation.
Collapse
Affiliation(s)
- Biao Wang
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Qi Zhang
- Liver Transplantation Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Bili Zhu
- Huiqiao Department, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhonglin Cui
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Jie Zhou
- Department of Hepatobiliary Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
- * E-mail:
| |
Collapse
|
49
|
Merlini E, Luzi K, Suardi E, Barassi A, Cerrone M, Martínez JS, Bai F, D'Eril GVM, Monforte AD, Marchetti G. T-cell phenotypes, apoptosis and inflammation in HIV+ patients on virologically effective cART with early atherosclerosis. PLoS One 2012; 7:e46073. [PMID: 23029393 PMCID: PMC3459872 DOI: 10.1371/journal.pone.0046073] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 08/27/2012] [Indexed: 11/21/2022] Open
Abstract
Objective We investigated the potential relationship between T-cell phenotype, inflammation, endotoxemia, and atherosclerosis evaluated by carotid intima-media thickness (IMT) in a cohort of HIV-positive patients undergoing long-term virologically suppressive combination antiretroviral therapy (cART). Design We studied 163 patients receiving virologically suppressive cART. Methods We measured IMT (carotid ultrasound); CD4+/CD8+ T-cell activation (CD38, CD45R0), differentiation (CD127), apoptosis (CD95), and senescence (CD28, CD57) (flow cytometry); plasma sCD14, IL-6, TNF- α, sVCAM-1, hs-CRP, anti-CMV IgG (ELISA); LPS (LAL). The results were compared by Mann-Whitney, Kruskal-Wallis or Chi-square tests, and factors associated with IMT were evaluated by multivariable logistic regression. Results Of 163 patients, 112 demonstrated normal IMT (nIMT), whereas 51 (31.3%) had pathological IMT (pIMT: ≥1 mm). Of the patients with pIMT, 22 demonstrated an increased IMT (iIMT), and 29 were shown to have plaques. These patient groups had comparable nadir and current CD4+, VLs and total length of time on cART. Despite similar proportions of CD38-expressing CD8+ cells (p = .95), pIMT patients exhibited higher activated memory CD8+CD38+CD45R0+ cells (p = .038) and apoptotic CD4+CD95+ (p = .01) and CD8+CD95+ cells (p = .003). In comparison to nIMT patients, iIMT patients tended to have lower numbers of early differentiated CD28+CD57− memory CD4+ (p = .048) and CD28–CD57−CD8+ cells (p = .006), both of which are associated with a higher proliferative potential. Despite no differences in plasma LPS levels, pIMT patients showed significantly higher circulating levels of sCD14 than did nIMT patients (p = .046). No differences in anti-CMV IgG was shown. Although circulating levels of sCD14 seemed to be associated with a risk of ATS in an unadjusted analysis, this effect was lost after adjusting for classical cardiovascular predictors. Conclusions Despite the provision of full viral suppression by cART, a hyperactivated, pro-apoptotic T-cell profile characterizes HIV-infected patients with early vascular damage, for whom the potential contribution of subclinical endotoxemia and anti-CMV immunity should be investigated further.
Collapse
Affiliation(s)
- Esther Merlini
- Clinic of Infectious Diseases and Tropical Medicine, Department of Health Sciences, University of Milan, San Paolo Hospital, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Tiba F, Nauwelaers F, Sangare L, Coulibaly B, Mrosek V, Kräusslich HG, Böhler T. Constitutive activation and accelerated maturation of peripheral blood T cells in healthy adults in Burkina Faso compared to Germany: the case of malaria? Eur J Med Res 2012; 16:519-25. [PMID: 22112357 PMCID: PMC3351894 DOI: 10.1186/2047-783x-16-12-519] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE It is not exactly known how frequent exposure to Plasmodium falciparum shapes the peripheral blood T-cell population in healthy West Africans. METHODS The frequency of peripheral blood CD4(+) lymphocytes responding to Plasmodium falciparum merozoite surface protein 1 (PfMSP-1) by production of interferon-gamma (IFN-γ), interleukin-2 (IL-2) or tumor necrosis factor-alpha (TNF-α) was determined using a commercially available flow cytometric activation assay (FastImmune) in 17 healthy adults in Nouna, Burkina Faso. T-cell activation and maturation in peripheral blood of healthy adults in Burkina Faso (n=40) and Germany (n=20) were compared using immunophenotyping and three-colour flow cytometry. RESULTS Significant numbers of PfMSP-1 -specific CD4(+) lymphocytes producing IFN-γ, IL-2 and/or TNF-α were detected in 14 healthy adults in Nouna. Cytokine profiles showed predominant production of IFN-γ and TNF-α. Compared to Germans, Burkinabé showed markedly lower proportions of CCR7+ CD45RA+ naive CD4(+) cells and slightly higher frequencies of CD95(+)CD4(+) T-cells and of CD38(+) CD8(+) T-cells. The median antibody-binding capacity of CD95(dim) CD4(+) T-cells in Burkinabé was more than twice the value observed in Germans (263 vs. 108 binding sites per cell, p<0.0001). CONCLUSIONS We hypothesize that an IFN-γ-induced increase in the expression level of CD95 on CD4(+) lymphocytes may lower the activation threshold of resting naive CD4(+) T-cells in healthy adults living in Burkina Faso. Bystander activation of these cells deserves further study as a molecular mechanism linking strong IFN-γ responses against Plasmodium falciparum to decreased susceptibility to parasitemia observed in specific ethnic groups in West Africa.
Collapse
Affiliation(s)
- F Tiba
- Department of Infectious Diseases, Virology, University of Heidelberg, INF324, 69120 Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|