1
|
Van den Bos J, Janssens I, Vermeulen M, Dams A, De Reu H, Peeters S, Faghel C, Ouaamari YE, Wens I, Cools N. The Efficiency of Brain-Derived Neurotrophic Factor Secretion by mRNA-Electroporated Regulatory T Cells Is Highly Impacted by Their Activation Status. Eur J Immunol 2025; 55:e202451005. [PMID: 39703060 PMCID: PMC11830389 DOI: 10.1002/eji.202451005] [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: 02/29/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/21/2024]
Abstract
Genetic engineering of regulatory T cells (Tregs) presents a promising avenue for advancing immunotherapeutic strategies, particularly in autoimmune diseases and transplantation. This study explores the modification of Tregs via mRNA electroporation, investigating the influence of T-cell activation status on transfection efficiency, phenotype, and functionality. For this CD45RA+ Tregs were isolated, expanded, and modified to overexpress brain-derived neurotrophic factor (BDNF). Kinetics of BDNF expression and secretion were explored. Treg activation state was assessed by checking the expression of activation markers CD69, CD71, and CD137. Our findings show that only activated Tregs secrete BDNF post-genetic engineering, even though both activated and resting Tregs express BDNF intracellularly. Notably, the mTOR pathway and CD137 are implicated in the regulation of protein secretion in activated Tregs, indicating a complex interplay of signalling pathways. This study contributes to understanding the mechanisms governing protein expression and secretion in engineered Tregs, offering insights for optimizing cell-based therapies and advancing immune regulation strategies.
Collapse
Affiliation(s)
- Jasper Van den Bos
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Ibo Janssens
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Morgane Vermeulen
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Amber Dams
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Hans De Reu
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- Flow Cytometry and Sorting Core Facility (FACSUA)University of AntwerpAntwerpBelgium
| | - Stefanie Peeters
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Carole Faghel
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Yousra El Ouaamari
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Inez Wens
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
| | - Nathalie Cools
- Laboratory of Experimental HematologyVaccine and Infections Disease Institute (VAXINFECTIO)Faculty of Medicine and Health SciencesUniversity of AntwerpAntwerpBelgium
- Flow Cytometry and Sorting Core Facility (FACSUA)University of AntwerpAntwerpBelgium
| |
Collapse
|
2
|
Sprent J, Boyman O. Optimising IL-2 for Cancer Immunotherapy. Immune Netw 2024; 24:e5. [PMID: 38455463 PMCID: PMC10917570 DOI: 10.4110/in.2024.24.e5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/01/2024] [Accepted: 01/08/2024] [Indexed: 03/09/2024] Open
Abstract
The key role of T cells in cancer immunotherapy is well established and is highlighted by the remarkable capacity of Ab-mediated checkpoint blockade to overcome T-cell exhaustion and amplify anti-tumor responses. However, total or partial tumor remission following checkpoint blockade is still limited to only a few types of tumors. Hence, concerted attempts are being made to devise new methods for improving tumor immunity. Currently, much attention is being focused on therapy with IL-2. This cytokine is a powerful growth factor for T cells and optimises their effector functions. When used at therapeutic doses for cancer treatment, however, IL-2 is highly toxic. Nevertheless, recent work has shown that modifying the structure or presentation of IL-2 can reduce toxicity and lead to effective anti-tumor responses in synergy with checkpoint blockade. Here, we review the complex interaction of IL-2 with T cells: first during normal homeostasis, then during responses to pathogens, and finally in anti-tumor responses.
Collapse
Affiliation(s)
- Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst 2010, Australia
- St. Vincent’s Clinical School, University of New South Wales, Sydney 1466, Australia
- Menzies Institute of Medical Research, Hobart 7000, Australia
| | - Onur Boyman
- Department of Immunology, University Hospital Zurich, Zurich 8091, Switzerland
- Faculty of Medicine and Faculty of Science, University of Zurich, Zurich 8057, Switzerland
| |
Collapse
|
3
|
Kushwaha A, Agarwal V. Pseudomonas aeruginosa quorum-sensing molecule N-(3-oxododecanoyl)-L-homoserine lactone mediates Ca +2 dysregulation, mitochondrial dysfunction, and apoptosis in human peripheral blood lymphocytes. Heliyon 2023; 9:e21462. [PMID: 38027911 PMCID: PMC10660034 DOI: 10.1016/j.heliyon.2023.e21462] [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: 05/30/2022] [Revised: 09/01/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023] Open
Abstract
N-(3-oxododecanoyl)-l-homoserine lactone is a Pseudomonas aeruginosa secreted quorum-sensing molecule that mediates the secretion of virulence factors, biofilm formation and plays a pivotal role in proliferation and persistence in the host. Apart from regulating quorum-sensing, the autoinducer signal molecule N-(3-oxododecanoyl)-l-homoserine lactone (3O-C12-HSL or C12) of a LasI-LasR circuit exhibits immunomodulatory effects and induces apoptosis in various host cells. However, the precise pathophysiological impact of C12 on human peripheral blood lymphocytes and its involvement in mitochondrial dysfunction remained largely elusive. In this study, the results suggest that C12 (100 μM) induces upregulation of cytosolic and mitochondrial Ca+2 levels and triggers mitochondrial dysfunction through the generation of mitochondrial ROS (mROS), disruption of mitochondrial transmembrane potential (ΔΨm), and opening of the mitochondrial permeability transition pore (mPTP). Additionally, it was observed that C12 induces phosphatidylserine (PS) exposure and promotes apoptosis in human peripheral blood lymphocytes. However, apoptosis plays a critical role in the homeostasis and development of lymphocytes, whereas enhanced apoptosis can cause immunodeficiency through cell loss. These findings suggest that C12 exerts a detrimental effect on lymphocytes by mediating mitochondrial dysfunction and enhancing apoptosis, which might further impair the effective mounting of immune responses during Pseudomonas aeruginosa-associated infections.
Collapse
Affiliation(s)
- Ankit Kushwaha
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| | - Vishnu Agarwal
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Prayagraj, Uttar Pradesh, 211004, India
| |
Collapse
|
4
|
Alvanou M, Lysandrou M, Christophi P, Psatha N, Spyridonidis A, Papadopoulou A, Yannaki E. Empowering the Potential of CAR-T Cell Immunotherapies by Epigenetic Reprogramming. Cancers (Basel) 2023; 15:1935. [PMID: 37046597 PMCID: PMC10093039 DOI: 10.3390/cancers15071935] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 04/14/2023] Open
Abstract
T-cell-based, personalized immunotherapy can nowadays be considered the mainstream treatment for certain blood cancers, with a high potential for expanding indications. Chimeric antigen receptor T cells (CAR-Ts), an ex vivo genetically modified T-cell therapy product redirected to target an antigen of interest, have achieved unforeseen successes in patients with B-cell hematologic malignancies. Frequently, however, CAR-T cell therapies fail to provide durable responses while they have met with only limited success in treating solid cancers because unique, unaddressed challenges, including poor persistence, impaired trafficking to the tumor, and site penetration through a hostile microenvironment, impede their efficacy. Increasing evidence suggests that CAR-Ts' in vivo performance is associated with T-cell intrinsic features that may be epigenetically altered or dysregulated. In this review, we focus on the impact of epigenetic regulation on T-cell differentiation, exhaustion, and tumor infiltration and discuss how epigenetic reprogramming may enhance CAR-Ts' memory phenotype, trafficking, and fitness, contributing to the development of a new generation of potent CAR-T immunotherapies.
Collapse
Affiliation(s)
- Maria Alvanou
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, 570 10 Thessaloniki, Greece
- Bone Marrow Transplantation Unit, Institute of Cell Therapy, University of Patras, 265 04 Rio, Greece
| | - Memnon Lysandrou
- Bone Marrow Transplantation Unit, Institute of Cell Therapy, University of Patras, 265 04 Rio, Greece
| | - Panayota Christophi
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, 570 10 Thessaloniki, Greece
- Bone Marrow Transplantation Unit, Institute of Cell Therapy, University of Patras, 265 04 Rio, Greece
| | - Nikoleta Psatha
- Department of Genetics, Development and Molecular Biology, School of Biology, Aristotle University of Thessaloniki, 570 10 Thessaloniki, Greece
| | - Alexandros Spyridonidis
- Bone Marrow Transplantation Unit, Institute of Cell Therapy, University of Patras, 265 04 Rio, Greece
| | - Anastasia Papadopoulou
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, 570 10 Thessaloniki, Greece
| | - Evangelia Yannaki
- Hematology Department-Hematopoietic Cell Transplantation Unit, Gene and Cell Therapy Center, George Papanikolaou Hospital, 570 10 Thessaloniki, Greece
- Department of Medicine, University of Washington, Seattle, WA 98195-2100, USA
| |
Collapse
|
5
|
Cai Y, Ye LP, Song YQ, Mao XL, Wang L, Jiang YZ, Que WT, Li SW. Liver injury in COVID-19: Detection, pathogenesis, and treatment. World J Gastroenterol 2021; 27:3022-3036. [PMID: 34168405 PMCID: PMC8192279 DOI: 10.3748/wjg.v27.i22.3022] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/24/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
In the early December 2019, a novel coronavirus named severe acute respiratory syndrome coronavirus 2 was first reported in Wuhan, China, followed by an outbreak that spread around the world. Numerous studies have shown that liver injury is common in patients with coronavirus disease 2019 (COVID-19), and may aggravate the severity of the disease. However, the exact cause and specific mechanism of COVID-associated liver injury needs to be elucidated further. In this review, we present an analysis of the clinical features, potential mechanisms, and treatment strategies for liver injury associated with COVID-19. We hope that this review would benefit clinicians in devising better strategies for management of such patients.
Collapse
Affiliation(s)
- Yue Cai
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Li-Ping Ye
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- School of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Ya-Qi Song
- School of Medicine, Zhejiang University, Hangzhou 310000, Zhejiang Province, China
| | - Xin-Li Mao
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Li Wang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot 010000, Inner Mongolia Autonomous Region, China
| | - Yan-Zhi Jiang
- Department of Gastroenterology and Hepatology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Wei-Tao Que
- Department of Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China
| | - Shao-Wei Li
- Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
- Department of Gastroenterology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| |
Collapse
|
6
|
Fernandez MF, Qiao G, Tulla K, Prabhakar BS, Maker AV. Combination Immunotherapy With LIGHT and Interleukin-2 Increases CD8 Central Memory T-Cells In Vivo. J Surg Res 2021; 263:44-52. [PMID: 33631377 DOI: 10.1016/j.jss.2021.01.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/30/2020] [Accepted: 01/15/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND The generation of long-term durable tumor immunity and prolonged disease-free survival depends on the ability to generate and support CD8+ central memory T-cells. Microsatellite-stable colon cancer is resistant to currently available immunotherapies; thus, development of novel mechanisms to increase both lymphocyte infiltration and central memory formation are needed to improve outcomes in these patients. We have previously demonstrated that both interleukin-2 (IL-2) and LIGHT (TNFSF14) independently enhance antitumor immune responses and hypothesize that combination immunotherapy may increase the CD8+ central memory T-cell response. METHODS Murine colorectal cancer tumors were established in syngeneic mice. Tumors were treated with control, soluble, or liposomal IL-2 at established intervals. A subset of animal tumors overexpressed tumor necrosis superfamily factor LIGHT (TNFSF14). Peripheral blood, splenic, and tumor-infiltrating lymphocytes were isolated for phenotypic studies and flow cytometry. RESULTS Tumors exposed to a combination of LIGHT and IL-2 experienced a decrease in tumor size compared with IL-2 alone that was not demonstrated in wild-type tumors or between other treatment groups. Combination exposure also increased splenic central memory CD8+ cells compared with IL-2 administration alone, while not increasing tumor-infiltrating lymphocytes. In the periphery, the combination enhanced levels of circulating CD8 T-cells and central memory T-cells, while also increasing circulating T-regulatory cells. CONCLUSIONS Combination of IL-2, whether soluble or liposomal, with exposure to LIGHT results in increased CD8+ central memory cells in the spleen and periphery. New combination immunotherapy strategies that support both effector and memory T-cell functions are critical to enhancing durable antitumor responses and warrant further investigation.
Collapse
Affiliation(s)
- Manuel F Fernandez
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
| | - Guilin Qiao
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
| | - Kiara Tulla
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
| | - Bellur S Prabhakar
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois
| | - Ajay V Maker
- Division of Surgical Oncology, Department of Surgery, University of Illinois at Chicago, Chicago, Illinois.
| |
Collapse
|
7
|
COVID-19-associated gastrointestinal and liver injury: clinical features and potential mechanisms. Signal Transduct Target Ther 2020; 5:256. [PMID: 33139693 PMCID: PMC7605138 DOI: 10.1038/s41392-020-00373-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/08/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease-2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The infection is spreading globally and poses a huge threat to human health. Besides common respiratory symptoms, some patients with COVID-19 experience gastrointestinal symptoms, such as diarrhea, nausea, vomiting, and loss of appetite. SARS-CoV-2 might infect the gastrointestinal tract through its viral receptor angiotensin-converting enzyme 2 (ACE2) and there is increasing evidence of a possible fecal–oral transmission route. In addition, there exist multiple abnormalities in liver enzymes. COVID-19-related liver injury may be due to drug-induced liver injury, systemic inflammatory reaction, and hypoxia–ischemia reperfusion injury. The direct toxic attack of SARS-CoV-2 on the liver is still questionable. This review highlights the manifestations and potential mechanisms of gastrointestinal and hepatic injuries in COVID-19 to raise awareness of digestive system injury in COVID-19.
Collapse
|
8
|
Tough DF, Rioja I, Modis LK, Prinjha RK. Epigenetic Regulation of T Cell Memory: Recalling Therapeutic Implications. Trends Immunol 2019; 41:29-45. [PMID: 31813765 DOI: 10.1016/j.it.2019.11.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 02/06/2023]
Abstract
Memory T cells possess functional differences from naïve T cells that powerfully contribute to the efficiency of secondary immune responses. These abilities are imprinted during the primary response, linked to the acquisition of novel patterns of gene expression. Underlying this are alterations at the chromatin level (epigenetic modifications) that regulate constitutive and inducible gene transcription. T cell epigenetic memory can persist long-term, contributing to long-lasting immunity after infection or vaccination. However, acquired epigenetic states can also hinder effective tumor immunity or contribute to autoimmunity. The growing understanding of epigenetic gene regulation as it relates to both the stability and malleability of T cell memory may offer the potential to selectively modify T cell memory in disease by targeting epigenetic mechanisms.
Collapse
Affiliation(s)
- David F Tough
- Epigenetics Research Unit, Oncology, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Inma Rioja
- Epigenetics Research Unit, Oncology, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Louise K Modis
- Adaptive Immunity Research Unit, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK
| | - Rab K Prinjha
- Epigenetics Research Unit, Oncology, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK; Adaptive Immunity Research Unit, GlaxoSmithKline, Gunnels Wood Road, Stevenage, Herts, SG1 2NY, UK.
| |
Collapse
|
9
|
Behr FM, Chuwonpad A, Stark R, van Gisbergen KPJM. Armed and Ready: Transcriptional Regulation of Tissue-Resident Memory CD8 T Cells. Front Immunol 2018; 9:1770. [PMID: 30131803 PMCID: PMC6090154 DOI: 10.3389/fimmu.2018.01770] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/17/2018] [Indexed: 11/13/2022] Open
Abstract
A fundamental benefit of immunological memory is the ability to respond in an enhanced manner upon secondary encounter with the same pathogen. Tissue-resident memory CD8 T (TRM) cells contribute to improved protection against reinfection through the generation of immediate effector responses at the site of pathogen entry. Key to the potential of TRM cells to develop rapid recall responses is their location within the epithelia of the skin, lungs, and intestines at prime entry sites of pathogens. TRM cells are among the first immune cells to respond to pathogens that have been previously encountered in an antigen-specific manner. Upon recognition of invading pathogens, TRM cells release IFN-γ and other pro-inflammatory cytokines and chemokines. These effector molecules activate the surrounding epithelial tissue and recruit other immune cells including natural killer (NK) cells, B cells, and circulating memory CD8 T cells to the site of infection. The repertoire of TRM effector functions also includes the direct lysis of infected cells through the release of cytotoxic molecules such as perforin and granzymes. The mechanisms enabling TRM cells to respond in such a rapid manner are gradually being uncovered. In this review, we will address the signals that instruct TRM generation and maintenance as well as the underlying transcriptional network that keeps TRM cells in a deployment-ready modus. Furthermore, we will discuss how TRM cells respond to reinfection of the tissue and how transcription factors may control immediate and proliferative TRM responses.
Collapse
Affiliation(s)
- Felix M Behr
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| | - Ammarina Chuwonpad
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands
| | - Regina Stark
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| | - Klaas P J M van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory AMC/UvA, Amsterdam, Netherlands.,Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| |
Collapse
|
10
|
Li X, Wang X, Ito A. Tailoring inorganic nanoadjuvants towards next-generation vaccines. Chem Soc Rev 2018; 47:4954-4980. [PMID: 29911725 DOI: 10.1039/c8cs00028j] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Vaccines, one of the most effective and powerful public health measures, have saved countless lives over the past century and still have a tremendous global impact. As an indispensable component of modern vaccines, adjuvants play a critical role in strengthening and/or shaping a specific immune response against infectious diseases as well as malignancies. The application of nanotechnology provides the possibility of precisely tailoring the building blocks of nanoadjuvants towards modern vaccines with the desired immune response. The last decade has witnessed great academic progress in inorganic nanomaterials for vaccine adjuvants in terms of nanometer-scale synthesis, structure control, and functionalization design. Inorganic adjuvants generally facilitate the delivery of antigens, allowing them to be released in a sustained manner, enhance immunogenicity, deliver antigens efficiently to specific targets, and induce a specific immune response. In particular, the recent discovery of the intrinsic immunomodulatory function of inorganic nanomaterials further allows us to shape the immune response towards the desired type and increase the efficacy of vaccines. In this article, we comprehensively review state-of-the-art research on the use of inorganic nanomaterials as vaccine adjuvants. Attention is focused on the physicochemical properties of versatile inorganic nanoadjuvants, such as composition, size, morphology, shape, hydrophobicity, and surface charge, to effectively stimulate cellular immunity, considering that the clinically used alum adjuvants can only induce strong humoral immunity. In addition, the efforts made to date to expand the application of inorganic nanoadjuvants in cancer vaccines are summarized. Finally, we discuss the future prospects and our outlook on tailoring inorganic nanoadjuvants towards next-generation vaccines.
Collapse
Affiliation(s)
- Xia Li
- Health Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan.
| | | | | |
Collapse
|
11
|
Condotta SA, Richer MJ. The immune battlefield: The impact of inflammatory cytokines on CD8+ T-cell immunity. PLoS Pathog 2017; 13:e1006618. [PMID: 29073270 PMCID: PMC5658174 DOI: 10.1371/journal.ppat.1006618] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Stephanie A. Condotta
- Department of Microbiology and Immunology, Microbiome and Disease Tolerance Centre, Rosalind and Morris Goodman Cancer Research Centre McGill University, Montreal, Quebec, Canada
| | - Martin J. Richer
- Department of Microbiology and Immunology, Microbiome and Disease Tolerance Centre, Rosalind and Morris Goodman Cancer Research Centre McGill University, Montreal, Quebec, Canada
| |
Collapse
|
12
|
Exploiting Cell Death Pathways for Inducible Cell Elimination to Modulate Graft-versus-Host-Disease. Biomedicines 2017; 5:biomedicines5020030. [PMID: 28613269 PMCID: PMC5489816 DOI: 10.3390/biomedicines5020030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/05/2017] [Accepted: 06/08/2017] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cell transplantation is a potent form of immunotherapy, potentially life-saving for many malignant hematologic diseases. However, donor lymphocytes infused with the graft while exerting a graft versus malignancy effect can also cause potentially fatal graft versus host disease (GVHD). Our group has previously validated the inducible caspase-9 suicide gene in the haploidentical stem cell transplant setting, which proved successful in reversing signs and symptoms of GVHD within hours, using a non-therapeutic dimerizing agent. Cellular death pathways such as apoptosis and necroptosis are important processes in maintaining healthy cellular homeostasis within the human body. Here, we review two of the most widely investigated cell death pathways active in T-cells (apoptosis and necroptosis), as well as the emerging strategies that can be exploited for the safety of T-cell therapies. Furthermore, such strategies could be exploited for the safety of other cellular therapeutics as well.
Collapse
|
13
|
Histone deacetylase inhibitors suppress immature dendritic cell's migration by regulating CC chemokine receptor 1 expression. Cell Immunol 2017; 316:11-20. [PMID: 28341057 DOI: 10.1016/j.cellimm.2017.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/25/2017] [Accepted: 02/27/2017] [Indexed: 11/23/2022]
Abstract
The modulation of immature dendritic cells (iDCs), which involves processes such as phagocytosis, migration, and maturation, is considered a beneficial research theme. Once activated by an antigen, iDCs turn to mature DCs (mDCs) and migrate towards secondary lymphoid organs, and initiate the progress of cellular immunity. Histone deacetylase inhibitors (HDACis) are also thought to be a major modulator of cellular immunity. Herein, we demonstrate that HDACis (trichostatin-A (TSA), sodium butylate (SB), scriptaid (ST)) play a central regulatory role in the migratory activity of iDCs. In our results, TSA, SB and ST showed the potent inhibitory effect on the migration of iDCs stimulated by MIP-1α. The inhibitory activities of HDACis were found to be caused by reduction of CCR1 expression on the cell surface, and by the inhibition of phosphorylation of p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinases 1 and 2 (ERK 1/2), and c-Jun N-terminal kinase (JNK).
Collapse
|
14
|
Regulation of effector and memory CD8(+) T cell function by inflammatory cytokines. Cytokine 2015; 82:16-23. [PMID: 26688544 DOI: 10.1016/j.cyto.2015.11.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 11/05/2015] [Accepted: 11/12/2015] [Indexed: 01/03/2023]
Abstract
Cells communicate with each other through the production and secretion of cytokines, which are integral to the host response to infection. Once recognized by specific cytokine receptors expressed on the cell surface, these exogenous signals direct the biological function of a cell in order to adapt to their microenvironment. CD8(+) T cells are critical immune cells that play an important role in the control and elimination of intracellular pathogens. Current findings have demonstrated that cytokines influence all aspects of the CD8(+) T cell response to infection or immunization. The cytokine milieu induced at the time of activation impacts the overall magnitude and function of the effector CD8(+) T cell response and the generation of functional memory CD8(+) T cells. This review will focus on the impact of inflammatory cytokines on different aspects of CD8(+) T cell biology.
Collapse
|
15
|
Abstract
Human periodontitis is associated with a wide range of bacteria and viruses and with complex innate and adaptive immune responses. Porphyromonas gingivalis, Tannerella forsythia, Aggregatibacter actinomycetemcomitans, Treponema denticola, cytomegalovirus and other herpesviruses are major suspected pathogens of periodontitis, and a combined herpesvirus–bacterial periodontal infection can potentially explain major clinical features of the disease. Cytomegalovirus infects periodontal macrophages and T‐cells and elicits a release of interleukin‐1β and tumor necrosis factor‐α. These proinflammatory cytokines play an important role in the host defense against the virus, but they also have the potential to induce alveolar bone resorption and loss of periodontal ligament. Gingival fibroblasts infected with cytomegalovirus also exhibit diminished collagen production and release of an increased level of matrix metalloproteinases. This article reviews innate and adaptive immunity to cytomegalovirus and suggests that immune responses towards cytomegalovirus can play roles in controlling, as well as in exacerbating, destructive periodontal disease.
Collapse
|
16
|
Kalyanasundaram Bhanumathy K, Zhang B, Xie Y, Xu A, Tan X, Xiang J. Potent immunotherapy against well-established thymoma using adoptively transferred transgeneIL-6-engineered dendritic cell-stimulated CD8+T-cells with prolonged survival and enhanced cytotoxicity. J Gene Med 2015. [DOI: 10.1002/jgm.2836] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
| | - Bei Zhang
- Cancer Research Cluster, Saskatchewan Cancer Agency, Division of Oncology; University of Saskatchewan; Saskatoon Saskatchewan Canada
| | - Yufeng Xie
- Cancer Research Cluster, Saskatchewan Cancer Agency, Division of Oncology; University of Saskatchewan; Saskatoon Saskatchewan Canada
| | - Aizhang Xu
- Cancer Research Cluster, Saskatchewan Cancer Agency, Division of Oncology; University of Saskatchewan; Saskatoon Saskatchewan Canada
| | - Xin Tan
- College of Life Science; Beijing Institute of Technology; Beijing China
| | - Jim Xiang
- Cancer Research Cluster, Saskatchewan Cancer Agency, Division of Oncology; University of Saskatchewan; Saskatoon Saskatchewan Canada
| |
Collapse
|
17
|
Sikora E. Activation-induced and damage-induced cell death in aging human T cells. Mech Ageing Dev 2015; 151:85-92. [PMID: 25843236 DOI: 10.1016/j.mad.2015.03.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 03/04/2015] [Accepted: 03/30/2015] [Indexed: 12/20/2022]
Abstract
In multicellular organisms the proper system functionality is ensured by the balance between cell division, differentiation, senescence and death. This balance is changed during aging. Immunosenescence plays a crucial role in aging and leads to the shrinkage of T cell repertoire and the propensity to apoptosis. The elimination of expanded T cells at the end of immune response is crucial to maintain homeostasis and avoid any uncontrolled inflammation. Resting mature T lymphocytes, when activated via their antigen-specific receptor (TCR) and CD28 co-receptor, start to proliferate and then undergo the so called activation induced cell death (AICD), which mechanistically is triggered by the death receptor and leads to apoptosis. T lymphocytes, like other cells, are also exposed to damage, which can trigger the so called damage-induced cell death (DICD). It was hypothesized that oxidative stress and chronic antigenic load increasing with age reduced lymphocyte susceptibility to DICD and enhanced a proinflamatory status leading to increased AICD. However, data collected so far are inconsistent and does not support this assumption. Systematic and comprehensive studies are still needed for conclusive elucidation of the role of AICD and DICD in human immunosenescence, including the role of autophagy and necroptosis in the processes.
Collapse
Affiliation(s)
- Ewa Sikora
- Molecular Bases of Aging Laboratory, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, Warsaw, Poland.
| |
Collapse
|
18
|
Zhang H, Wu H, Gao L, Qiu Y, Xiao J, Zhang Y. Identification, expression and immunological responses to bacterial challenge following vaccination of BLT1 gene from turbot, Scophthalmus maximus. Gene 2014; 557:229-35. [PMID: 25541026 DOI: 10.1016/j.gene.2014.12.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2014] [Revised: 11/26/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
Abstract
Leukotriene B4 (LTB4) is well known as a chemoattractant for leucocytes, recent studies also showed its involvement in adaptive immunity. The purpose of this work is to report the cloning, characterization and gene expression of leukotriene B4 receptor (BLT1) in turbot (Scophthalmus maximus), as well as the immunological response to challenge following vaccination with a live attenuated vaccine Vibrio anguillarum MVAV6203. The full cDNA sequence of turbot BLT1 was cloned. The open reading frame consists of 1119bp nucleotides, which translate into 372 amino acid protein. A high conservation of amino acid sequence was found in the seven transmembrane (TM) domains and intracellular loops. The intracellular loop 3 consisting of a unique cluster of basic amino acid residues might be associated with signal transduction. High amino acid similarity and a phylogenetic tree confirmed it as a leukotriene B4 receptor member. The BLT1 gene is expressed in a wide range of tissues with the highest expression in kidney followed by spleen. The expression of turbot BLT1 was significantly up-regulated in spleen, gut and gill after vaccination and in kidney and skin after challenge. These results suggest a potential role of turbot BLT1 in protection against infection.
Collapse
Affiliation(s)
- Hua Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Center for Translational Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Haizhen Wu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Liang Gao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Ying Qiu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Jingfan Xiao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
19
|
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]
|
20
|
Gong C, Linderman JJ, Kirschner D. Harnessing the heterogeneity of T cell differentiation fate to fine-tune generation of effector and memory T cells. Front Immunol 2014; 5:57. [PMID: 24600448 PMCID: PMC3928592 DOI: 10.3389/fimmu.2014.00057] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 01/31/2014] [Indexed: 11/13/2022] Open
Abstract
Recent studies show that naïve T cells bearing identical T cell receptors experience heterogeneous differentiation and clonal expansion processes. The factors controlling this outcome are not well characterized, and their contributions to immune cell dynamics are similarly poorly understood. In this study, we develop a computational model to elaborate mechanisms occurring within and between two important physiological compartments, lymph nodes and blood, to determine how immune cell dynamics are controlled. Our multi-organ (multi-compartment) model integrates cellular and tissue level events and allows us to examine the heterogeneous differentiation of individual precursor cognate naïve T cells to generate both effector and memory T lymphocytes. Using this model, we simulate a hypothetical immune response and reproduce both primary and recall responses to infection. Increased numbers of antigen-bearing dendritic cells (DCs) are predicted to raise production of both effector and memory T cells, and distinct “sweet spots” of peptide-MHC levels on those DCs exist that favor CD4+ or CD8+ T cell differentiation toward either effector or memory cell phenotypes. This has important implications for vaccine development and immunotherapy.
Collapse
Affiliation(s)
- Chang Gong
- Department of Computational Medicine and Bioinformatics, University of Michigan , Ann Arbor, MI , USA
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan , Ann Arbor, MI , USA
| | - Denise Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School , Ann Arbor, MI , USA
| |
Collapse
|
21
|
Predicting lymph node output efficiency using systems biology. J Theor Biol 2013; 335:169-84. [PMID: 23816876 DOI: 10.1016/j.jtbi.2013.06.016] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Revised: 06/11/2013] [Accepted: 06/11/2013] [Indexed: 11/20/2022]
Abstract
Dendritic cells (DCs) capture pathogens and foreign antigen (Ag) in peripheral tissues and migrate to secondary lymphoid tissues, such as lymph nodes (LNs), where they present processed Ag as MHC-bound peptide (pMHC) to naïve T cells. Interactions between DCs and T cells result, over periods of hours, in activation, clonal expansion and differentiation of antigen-specific T cells, leading to primed cells that can now participate in immune responses. Two-photon microscopy (2PM) has been widely adopted to analyze lymphocyte dynamics and can serve as a powerful in vivo assay for cell trafficking and activation over short length and time scales. Linking biological phenomena between vastly different spatiotemporal scales can be achieved using a systems biology approach. We developed a 3D agent-based cellular model of a LN that allows for the simultaneous in silico simulation of T cell trafficking, activation and production of effector cells under different antigen (Ag) conditions. The model anatomy is based on in situ analysis of LN sections (from primates and mice) and cell dynamics based on quantitative measurements from 2PM imaging of mice. Our simulations make three important predictions. First, T cell encounters by DCs and T cell receptor (TCR) repertoire scanning are more efficient in a 3D model compared with 2D, suggesting that a 3D model is needed to analyze LN function. Second, LNs are able to produce primed CD4+T cells at the same efficiency over broad ranges of cognate frequencies (from 10(-5) to 10(-2)). Third, reducing the time that naïve T cells are required to bind DCs before becoming activated will increase the rate at which effector cells are produced. This 3D model provides a robust platform to study how T cell trafficking and activation dynamics relate to the efficiency of T cell priming and clonal expansion. We envision that this systems biology approach will provide novel insights for guiding vaccine development and understanding immune responses to infection.
Collapse
|
22
|
Renault TT, Chipuk JE. Getting away with murder: how does the BCL-2 family of proteins kill with immunity? Ann N Y Acad Sci 2013; 1285:59-79. [PMID: 23527542 DOI: 10.1111/nyas.12045] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The adult human body produces approximately one million white blood cells every second. However, only a small fraction of the cells will survive because the majority is eliminated through a genetically controlled form of cell death known as apoptosis. This review places into perspective recent studies pertaining to the BCL-2 family of proteins as critical regulators of the development and function of the immune system, with particular attention on B cell and T cell biology. Here we discuss how elegant murine model systems have revealed the major contributions of the BCL-2 family in establishing an effective immune system. Moreover, we highlight some key regulatory pathways that influence the expression, function, and stability of individual BCL-2 family members, and discuss their role in immunity. From lethal mechanisms to more gentle ones, the final portion of the review discusses the nonapoptotic functions of the BCL-2 family and how they pertain to the control of immunity.
Collapse
Affiliation(s)
- Thibaud T Renault
- Department of Oncological Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
23
|
Tzelepis F, Joseph J, Haddad EK, Maclean S, Dudani R, Agenes F, Peng SL, Sekaly RP, Sad S. Intrinsic role of FoxO3a in the development of CD8+ T cell memory. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:1066-75. [PMID: 23277488 PMCID: PMC3815477 DOI: 10.4049/jimmunol.1200639] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD8(+) T cells undergo rapid expansion during infection with intracellular pathogens, which is followed by swift and massive culling of primed CD8(+) T cells. The mechanisms that govern the massive contraction and maintenance of primed CD8(+) T cells are not clear. We show in this study that the transcription factor, FoxO3a, does not influence Ag presentation and the consequent expansion of CD8(+) T cell response during Listeria monocytogenes infection, but plays a key role in the maintenance of memory CD8(+) T cells. The effector function of primed CD8(+) T cells as revealed by cytokine secretion and CD107a degranulation was not influenced by inactivation of FoxO3a. Interestingly, FoxO3a-deficient CD8(+) T cells displayed reduced expression of proapoptotic molecules BIM and PUMA during the various phases of response, and underwent reduced apoptosis in comparison with wild-type cells. A higher number of memory precursor effector cells and memory subsets was detectable in FoxO3a-deficient mice compared with wild-type mice. Furthermore, FoxO3a-deficient memory CD8(+) T cells upon transfer into normal or RAG1-deficient mice displayed enhanced survival. These results suggest that FoxO3a acts in a cell-intrinsic manner to regulate the survival of primed CD8(+) T cells.
Collapse
Affiliation(s)
- Fanny Tzelepis
- National Research Council of Canada, Institute for Biological Sciences, Ottawa, Ontario K1A 0R6, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Purushothaman D, Marcel N, Garg M, Venkataraman R, Sarin A. Apoptotic programs are determined during lineage commitment of CD4+ T effectors: selective regulation of T effector-memory apoptosis by inducible nitric oxide synthase. THE JOURNAL OF IMMUNOLOGY 2012; 190:97-105. [PMID: 23225886 DOI: 10.4049/jimmunol.1103694] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lineage-committed T effectors generated in response to Ag during the inflammatory phase are destined to die during termination of the immune response. We present evidence to suggest that molecular signatures of lineage commitment are reflected in apoptotic cascades activated in CD4(+) T effectors. Exemplifying this, ablation of inducible NO synthase (iNOS) protected effector-memory T (TEM) cells, but not T(Naive) or central-memory T cells, activated in vitro, from apoptosis triggered by cytokine deprivation. Furthermore, attrition of T effectors generated in the secondary, but not the primary, response to Ag was substantially reduced in mice, which received iNOS inhibitors. Distinct patterns of iNOS expression were revealed in wild-type TEM effectors undergoing apoptosis, and ablation of iNOS protein in primary and TEM wild-type effectors confirmed observations made in iNOS(-/-) cells. Describing molecular correlates of this dependence, mitochondrial damage, activation of the protein Bax, and release from mitochondria of the apoptosis-inducing factor were selectively abrogated in iNOS(-/-) TEM effectors. Suggesting that iNOS dependence was linked to the functional identity of T cell subsets, both iNOS induction and apoptosis were compromised in IFN-γ(-/-) TEM effectors, which mirrored the response patterns of iNOS(-)(/)(-) TEM. Collectively, these observations suggest that programs regulating deletion and differentiation are closely integrated and likely encoded during lineage commitment of T effectors.
Collapse
Affiliation(s)
- Divya Purushothaman
- National Centre for Biological Sciences, Bangalore 560065, Karnataka, India.
| | | | | | | | | |
Collapse
|
25
|
Garrod KR, Moreau HD, Garcia Z, Lemaître F, Bouvier I, Albert ML, Bousso P. Dissecting T cell contraction in vivo using a genetically encoded reporter of apoptosis. Cell Rep 2012; 2:1438-47. [PMID: 23159042 DOI: 10.1016/j.celrep.2012.10.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 10/01/2012] [Accepted: 10/22/2012] [Indexed: 12/11/2022] Open
Abstract
Contraction is a critical phase of immunity whereby the vast majority of effector T cells die by apoptosis, sparing a population of long-lived memory cells. Where, when, and why contraction occurs has been difficult to address directly due in large part to the rapid clearance of apoptotic T cells in vivo. To circumvent this issue, we introduced a genetically encoded reporter for caspase-3 activity into naive T cells to identify cells entering the contraction phase. Using two-photon imaging, we found that caspase-3 activity in T cells was maximal at the peak of the response and was associated with loss of motility followed minutes later by cell death. We demonstrated that contraction is a widespread process occurring uniformly in all organs tested and targeting phenotypically diverse T cells. Importantly, we identified a critical window of time during which antigen encounters act to antagonize T cell apoptosis, supporting a causal link between antigen clearance and T cell contraction. Our results offer insight into a poorly explored phase of immunity and provide a versatile methodology to study apoptosis during the development or function of a variety of immune cells in vivo.
Collapse
Affiliation(s)
- Kym R Garrod
- Institut Pasteur, Dynamics of Immune Responses Unit, 75015 Paris, France; INSERM U668, 75015 Paris, France
| | | | | | | | | | | | | |
Collapse
|
26
|
Zhang P, Wu J, Deoliveira D, Chao NJ, Chen BJ. Allospecific CD4(+) effector memory T cells do not induce graft-versus-host disease in mice. Biol Blood Marrow Transplant 2012; 18:1488-99. [PMID: 22809867 DOI: 10.1016/j.bbmt.2012.07.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 07/09/2012] [Indexed: 11/29/2022]
Abstract
We studied whether allospecific CD4(+) effector memory T cells (T(EM)) could induce graft-versus-host disease (GVHD) using a novel GVHD model induced solely by CD4(+) T cell receptor transgenic TEa cells. Allospecific T(EM) generated in a lymphopenic host bore a typical memory phenotype. Moreover, these cells were able to elicit a faster and more effective proliferative response on challenge with alloantigen in vitro and to mediate "second-set" skin graft rejection in vivo. However, these allospecific T(EM) were unable to induce GVHD. Allospecific T(EM) recipients became tolerant to alloantigen as a result of clonal deletion. Even though allospecific T(EM) were able to respond to alloantigen initially, the expansion of these cells and inflammatory cytokine production during GVHD were dramatically decreased. The inability of allospecific T(EM) to sustain the alloresponse may be a result of enhanced activation-induced cell death. These observations provide insight into how allospecific CD4(+) T(EM) respond to alloantigen during GVHD and underscore the fundamental differences in alloresponses mediated by allospecific T(EM) in graft rejection and GVHD settings.
Collapse
Affiliation(s)
- Ping Zhang
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | |
Collapse
|
27
|
Sekine Y, Yamamoto C, Kakisaka M, Muromoto R, Kon S, Ashitomi D, Fujita N, Yoshimura A, Oritani K, Matsuda T. Signal-Transducing Adaptor Protein-2 Modulates Fas-Mediated T Cell Apoptosis by Interacting with Caspase-8. THE JOURNAL OF IMMUNOLOGY 2012; 188:6194-204. [DOI: 10.4049/jimmunol.1103467] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
28
|
von Gunten S, Simon HU. Granulocyte death regulation by naturally occurring autoantibodies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 750:157-72. [PMID: 22903673 DOI: 10.1007/978-1-4614-3461-0_12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Programmed cell death (PCD) plays a central role in the regulation of granulocytes that are key effector cells of the innate immune system. Granulocytes are produced in high amounts in the bone marrow. A safe elimination of granulocytes by cell death (apoptosis) is essential to maintain the numbers of these cells balanced. In many acute and chronic inflammatory diseases, delayed apoptosis is one mechanism that contributes to accumulation of neutrophil and eosinophil granulocytes at the site of inflammation. On the other hand, a safe elimination of granulocytes by cell death is required to avoid unwanted tissue damage for instance by secretion of toxic products from these cells. Recent evidence shows that humans produce an array of naturally occurring autoantibodies (NAbs) with the capacity to regulate granulocyte death, including agonistic and antagonistic NAbs that bind to the receptors Fas, Siglec-8, and Siglec-9. Together with other factors, these various NAbs exhibit different properties in terms of the form of cell death they induce, the molecular signaling pathways they engage, as well as the efficacy or potency by which they induce cell death. Moreover, several regulatory mechanisms seem to exist that control their biological activity. Novel insights support the concept of granulocyte death regulation by NAbs, which might have important implications for our understanding of the pathogenesis and treatment of inflammatory diseases, including many autoimmune and allergic disorders.
Collapse
|
29
|
Young KG, Maclean S, Dudani R, Krishnan L, Sad S. CD8+ T cells primed in the periphery provide time-bound immune-surveillance to the central nervous system. THE JOURNAL OF IMMUNOLOGY 2011; 187:1192-200. [PMID: 21715683 DOI: 10.4049/jimmunol.1100695] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
After vaccination, memory CD8(+) T cells migrate to different organs to mediate immune surveillance. In most nonlymphoid organs, following an infection, CD8(+) T cells differentiate to become long-lived effector-memory cells, thereby providing long-term protection against a secondary infection. In this study, we demonstrated that Ag-specific CD8(+) T cells that migrate to the mouse brain following a systemic Listeria infection do not display markers reminiscent of long-term memory cells. In contrast to spleen and other nonlymphoid organs, none of the CD8(+) T cells in the brain reverted to a memory phenotype, and all of the cells were gradually eliminated. These nonmemory phenotype CD8(+) T cells were found primarily within the choroid plexus, as well as in the cerebrospinal fluid-filled spaces. Entry of these CD8(+) T cells into the brain was governed primarily by CD49d/VCAM-1, with the majority of entry occurring in the first week postinfection. When CD8(+) T cells were injected directly into the brain parenchyma, cells that remained in the brain retained a highly activated (CD69(hi)) phenotype and were gradually lost, whereas those that migrated out to the spleen were CD69(low) and persisted long-term. These results revealed a mechanism of time-bound immune surveillance to the brain by CD8(+) T cells that do not reside in the parenchyma.
Collapse
Affiliation(s)
- Kevin G Young
- National Research Council of Canada-Institute for Biological Sciences, Ottawa, Ontario K1A 0R6, Canada
| | | | | | | | | |
Collapse
|
30
|
Zhao Y, Shan Z, Li Q, Zhou Y, Zeng X, Fan Q, Liao C, Zhu Y, Zhao Y, Lu X, Liu J. Effect of CD4+ Memory T Cells on Rejection Response of Ectopic Heart Transplantation in Mice. Transplant Proc 2011; 43:1989-93. [DOI: 10.1016/j.transproceed.2010.12.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 11/05/2010] [Accepted: 12/13/2010] [Indexed: 11/29/2022]
|
31
|
Wolkers MC, Bensinger SJ, Green DR, Schoenberger SP, Janssen EM. Interleukin-2 rescues helpless effector CD8+ T cells by diminishing the susceptibility to TRAIL mediated death. Immunol Lett 2011; 139:25-32. [PMID: 21621553 DOI: 10.1016/j.imlet.2011.04.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Revised: 04/21/2011] [Accepted: 04/21/2011] [Indexed: 11/28/2022]
Abstract
CD8(+) T cells primed in the absence of CD4(+) T cell help are programmed to produce TRAIL, which results in Death receptor (DR5) mediated apoptosis upon restimulation. Here, we studied whether these 'helpless' effector CD8(+) T cells are consigned to an apoptotic fate or whether their helpless program can be altered by inflammatory or growth cytokines. We found that helpless CD8(+) T cells regained their full proliferative and functional capacity only when IL-2 was added to cell cultures, while IL-7 and IL-15, two common gamma chain cytokines associated with CD8(+) T cell homeostasis and memory, could only partly restore secondary expansion in helpless CD8(+) T cells. Recovery of functional CD8(+) T cell immunity by IL-2 was concomitant with induction of IL2Rα (CD25) expression, downregulation of TRAIL, and the upregulation of anti-apoptotic molecules Bcl-2 and FLIP. The addition of IL-2 to helpless CD8(+) T cells also interfered with DR5-mediated apoptosis induction, indicating that IL-2 affects several components of the TRAIL-DR5 pathway. Collectively, these data demonstrate that the helpless phenotype is not fixed, and that IL-2R signaling at the time of reactivation can play an important role in restoring CD8(+) T cell function.
Collapse
Affiliation(s)
- Monika C Wolkers
- Department of Cellular Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
32
|
Schuldt K, Kretz CC, Timmann C, Sievertsen J, Ehmen C, Esser C, Loag W, Ansong D, Dering C, Evans J, Ziegler A, May J, Krammer PH, Agbenyega T, Horstmann RD. A -436C>A polymorphism in the human FAS gene promoter associated with severe childhood malaria. PLoS Genet 2011; 7:e1002066. [PMID: 21625619 PMCID: PMC3098189 DOI: 10.1371/journal.pgen.1002066] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 03/18/2011] [Indexed: 11/29/2022] Open
Abstract
Human genetics and immune responses are considered to critically influence the outcome of malaria infections including life-threatening syndromes caused by Plasmodium falciparum. An important role in immune regulation is assigned to the apoptosis-signaling cell surface receptor CD95 (Fas, APO-1), encoded by the gene FAS. Here, a candidate-gene association study including variant discovery at the FAS gene locus was carried out in a case-control group comprising 1,195 pediatric cases of severe falciparum malaria and 769 unaffected controls from a region highly endemic for malaria in Ghana, West Africa. We found the A allele of c.−436C>A (rs9658676) located in the promoter region of FAS to be significantly associated with protection from severe childhood malaria (odds ratio 0.71, 95% confidence interval 0.58–0.88, pempirical = 0.02) and confirmed this finding in a replication group of 1,412 additional severe malaria cases and 2,659 community controls from the same geographic area. The combined analysis resulted in an odds ratio of 0.71 (95% confidence interval 0.62–0.80, p = 1.8×10−7, n = 6035). The association applied to c.−436AA homozygotes (odds ratio 0.47, 95% confidence interval 0.36–0.60) and to a lesser extent to c.−436AC heterozygotes (odds ratio 0.73, 95% confidence interval 0.63–0.84), and also to all phenotypic subgroups studied, including severe malaria anemia, cerebral malaria, and other malaria complications. Quantitative FACS analyses assessing CD95 surface expression of peripheral blood mononuclear cells of naïve donors showed a significantly higher proportion of CD69+CD95+ cells among persons homozygous for the protective A allele compared to AC heterozygotes and CC homozygotes, indicating a functional role of the associated CD95 variant, possibly in supporting lymphocyte apoptosis. Severe malaria caused by infection with the protozoan parasite Plasmodium falciparum is a major health burden, causing approximately one million fatalities annually, predominantly among young children in Sub-Saharan Africa. The occurrence of severe malaria may depend on a complex interplay of transmission dynamics and the development of a protective immune response but also on heritable differences in the susceptibility to the disease. In two large studies including a total of 2,607 affected children and 3,428 apparently healthy individuals from Ghana, West Africa, we investigated genetic variants of the FAS gene, which encodes CD95, a molecule critically involved in the programmed cell death of lymphocytes. We found that a single nucleotide variant in the FAS promoter was associated with a 29%–reduced risk of developing severe malaria. In individuals carrying two copies of the protective allele, a higher proportion of activated lymphocytes was found to express CD95. These findings indicate that a predisposition to an increased expression of CD95 may help to protect from severe malaria, possibly by rendering activated T-lymphocytes more susceptible to programmed cell death.
Collapse
Affiliation(s)
- Kathrin Schuldt
- Department of Molecular Medicine, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Mueller P, Liu X, Pieters J. Migration and homeostasis of naive T cells depends on coronin 1-mediated prosurvival signals and not on coronin 1-dependent filamentous actin modulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:4039-4050. [PMID: 21339362 DOI: 10.4049/jimmunol.1003352] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Coronins are WD repeat-containing proteins highly conserved in the eukaryotic kingdom implicated in the regulation of F-actin. Mammalian coronin 1, one of the most conserved isoforms expressed in leukocytes, regulates survival of T cells, which has been suggested to be due to its role in preventing F-actin-induced apoptosis. In this study, we come to a different conclusion. We show that coronin 1 does not modulate F-actin and that induction of F-actin failed to induce apoptosis. Instead, coronin 1 was required for providing prosurvival signals, in the absence of which T cells rapidly underwent apoptosis. These results argue against a role for coronin 1 in F-actin-mediated T cell apoptosis and establish coronin 1 as an essential regulator of the balance between prosurvival and proapoptotic signals in naive T cells.
Collapse
|
34
|
Cush SS, Flaño E. KLRG1+NKG2A+ CD8 T cells mediate protection and participate in memory responses during γ-herpesvirus infection. THE JOURNAL OF IMMUNOLOGY 2011; 186:4051-8. [PMID: 21346231 DOI: 10.4049/jimmunol.1003122] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Functional CD8 T cell effector and memory responses are generated and maintained during murine γ-herpesvirus 68 (γHV68) persistent infection despite continuous presentation of viral lytic Ags. However, the identity of the CD8 T cell subpopulations that mediate effective recall responses and that can participate in the generation of protective memory to a γ-herpesvirus infection remains unknown. During γHV68 persistence, ∼75% of γHV68-specific CD8 T cells coexpress the NK receptors killer cell lectin-like receptor G1 (KLRG1) and NKG2A. In this study, we take advantage of this unique phenotype to analyze the capacity of CD8 T cells expressing or not expressing KLRG1 and NKG2A to mediate effector and memory responses. Our results show that γHV68-specific KLRG1(+)NKG2A(+) CD8 T cells have an effector memory phenotype as well as characteristics of polyfunctional effector cells such us IFN-γ and TNF-α production, killing capacity, and are more efficient at protecting against a γHV68 challenge than their NKG2A(-)KLRG1(-) counterparts. Nevertheless, γHV68-specific NKG2A(+)KLRG1(+) CD8 T cells express IL-7 and IL-15 receptors, can survive long-term without cognate Ag, and subsequently mount a protective response during antigenic recall. These results highlight the plasticity of the immune system to generate protective effector and proliferative memory responses during virus persistence from a pool of KLRG1(+)NKG2A(+) effector memory CD8 T cells.
Collapse
Affiliation(s)
- Stephanie S Cush
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | |
Collapse
|
35
|
Fousteri G, Dave A, Juedes A, Juntti T, Morin B, Togher L, Farber DL, von Herrath M. Increased memory conversion of naïve CD8 T cells activated during late phases of acute virus infection due to decreased cumulative antigen exposure. PLoS One 2011; 6:e14502. [PMID: 21253594 PMCID: PMC3017078 DOI: 10.1371/journal.pone.0014502] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 12/16/2010] [Indexed: 11/28/2022] Open
Abstract
Background Memory CD8 T cells form an essential part of protective immunity against viral infections. Antigenic load, costimulation, CD4-help, cytokines and chemokines fluctuate during the course of an antiviral immune response thus affecting CD8 T cell activation and memory conversion. Methodology/Principal Findings In the present study, naïve TCR transgenic LCMV-specific P14 CD8 T cells engaged at a late stage during the acute antiviral LCMV response showed reduced expansion kinetics but greater memory conversion in the spleen. Such late activated cells displayed a memory precursor effector phenotype already at the peak of the systemic antiviral response, suggesting that the environment determined their fate during antigen encounter. In the spleen, the majority of late transferred cells exhibited a central memory phenotype compared to the effector memory displayed by the early transferred cells. Increasing the inflammatory response by exogenous administration of IFNγ, PolyI:C or CpG did not affect memory conversion in the late transferred group, suggesting that the diverging antigen load early versus later during acute infection had determined their fate. In agreement, reduction in the LCMV antigenic load after ribavirin treatment enhanced the contribution of early transferred cells to the long lasting memory pool. Conclusions/Significance Our results show that naïve CD8 cells, exposed to reduced duration or concentration of antigen during viral infection convert into memory more efficiently, an observation that could have significant implications for vaccine design.
Collapse
Affiliation(s)
- Georgia Fousteri
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Amy Dave
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Amy Juedes
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Therese Juntti
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Bret Morin
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Lisa Togher
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
| | - Donna L. Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, New York, United States of America
| | - Matthias von Herrath
- Diabetes Center, La Jolla Institute for Allergy and Immunology, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
36
|
McComb S, Mulligan R, Sad S. Caspase-3 is transiently activated without cell death during early antigen driven expansion of CD8(+) T cells in vivo. PLoS One 2010; 5:e15328. [PMID: 21203525 PMCID: PMC3008739 DOI: 10.1371/journal.pone.0015328] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/08/2010] [Indexed: 12/18/2022] Open
Abstract
Background CD8+ T cell responses develop rapidly during infection and are swiftly reduced during contraction, wherein >90% of primed CD8+ T cells are eliminated. The role of apoptotic mechanisms in controlling this rapid proliferation and contraction of CD8+ T cells remains unclear. Surprisingly, evidence has shown non-apoptotic activation of caspase-3 to occur during in vitro T-cell proliferation, but the relevance of these mechanisms to in vivo CD8+ T cell responses has yet to be examined. Methods and Findings We have evaluated the activity of caspase-3, a key downstream inducer of apoptosis, throughout the entirety of a CD8+ T cell response. We utilized two infection models that differ in the intensity, onset and duration of antigen-presentation and inflammation. Expression of cleaved caspase-3 in antigen specific CD8+ T cells was coupled to the timing and strength of antigen presentation in lymphoid organs. We also observed coordinated activation of additional canonical apoptotic markers, including phosphatidylserine exposure. Limiting dilution analysis directly showed that in the presence of IL7, very little cell death occurred in both caspase-3hi and caspase-3low CD8+ T cells. The expression of active caspase-3 peaked before effector phenotype (CD62Llow) CD8+ T cells emerged, and was undetectable in effector-phenotype cells. In addition, OVA-specific CD8+ cells remained active caspase-3low throughout the contraction phase. Conclusions Our results specifically implicate antigen and not inflammation in driving activation of apoptotic mechanisms without cell death in proliferating CD8+ T cells. Furthermore, the contraction of CD8+ T cell response following expansion is likely not mediated by the key downstream apoptosis inducer, caspase-3.
Collapse
Affiliation(s)
- Scott McComb
- NRC-Institute for Biological Sciences, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Rebecca Mulligan
- NRC-Institute for Biological Sciences, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Subash Sad
- NRC-Institute for Biological Sciences, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
- * E-mail:
| |
Collapse
|
37
|
Shen T, Zheng J, Xu C, Liu J, Zhang W, Lu F, Zhuang H. PD-1 expression on peripheral CD8+ TEM/TEMRA subsets closely correlated with HCV viral load in chronic hepatitis C patients. Virol J 2010; 7:310. [PMID: 21070674 PMCID: PMC2989324 DOI: 10.1186/1743-422x-7-310] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Accepted: 11/12/2010] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Tight correlation between host circulating CD8+ T cell-mediated immune response and control of viral replication is classical characteristic of long-term HCV infection. CD8+ T cell maturation/activation markers are expected to be associated with viral replication and disease progression in chronic HCV infection. The aim of the present study was to explore novel markers on CD8+ T cells with ability to evaluate HCV viral replication and disease progression. METHODS PBMCs were isolated from 37 chronic HCV-infected patients and 17 healthy controls. Distributed pattern of CD8+ T cells subsets and expression of PD-1, CD38, HLA-DR and CD127 were analyzed by flow cytometry. The correlation between expression of surface markers and HCV viral load or ALT was studied. RESULTS Declined naïve and increased TEMRA CD8+ T subsets were found in HCV-infected individuals compared with healthy controls. Percentage and MFI of PD-1, CD38 and HLA-DR on all CD8+ T cell subsets were higher in HCV-infected patients than healthy controls. In contrast, CD127 expression on CD8+ TCM showed an opposite trend as PD-1, CD38 and HLA-DR did. In chronic HCV infection, MFI of PD-1 on CD8+ TEM (p < 0.0001) and TEMRA (p = 0.0015) was positively correlated with HCV viral load while HLA-DR expression on non-naive CD8+ T cell subsets (p < 0.05) was negatively correlated with HCV viral load. CONCLUSION PD-1 level on peripheral CD8+ TEM/TEMRA was highly correlated with HCV viral load in chronic HCV-infected patients, which made PD-1 a novel indicator to evaluate HCV replication and disease progression in chronic hepatitis C patients.
Collapse
Affiliation(s)
- Tao Shen
- Department of Microbiology, Peking University Health Science Center, Beijing 100191, PR China
| | | | | | | | | | | | | |
Collapse
|
38
|
Gorbachev AV, Fairchild RL. CD4+CD25+ regulatory T cells utilize FasL as a mechanism to restrict DC priming functions in cutaneous immune responses. Eur J Immunol 2010; 40:2006-15. [PMID: 20405474 DOI: 10.1002/eji.200939387] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Recent studies have suggested Fas-mediated elimination of antigen-presenting cells as an important mechanism down-regulating the induction of autoimmune responses. It remains unknown whether this mechanism restricts the magnitude of immune responses to non-self antigens. We used a mouse model of a cutaneous CD8(+) T-cell-mediated immune response (contact hypersensitivity, CHS) to test if CD4(+)CD25(+) T cells expressing FasL regulate hapten-specific effector CD8(+) T cell expansion through the elimination of Fas-expressing hapten-presenting DC. In WT mice, attenuation of CD4(+)CD25(+) T regulatory cell activity by anti-CD25 mAb increased hapten-presenting DC numbers in skin-draining LN, which led to increased effector CD8(+) T-cell priming for CHS responses. In contrast, CD4(+)CD25(+) T cells did not regulate hapten-specific CD8(+) T-cell priming and CHS responses initiated by Fas-defective (lpr) DC. Thus, restricting DC priming functions through Fas-FasL interactions is a potent mechanism employed by CD4(+)CD25(+) regulatory cells to restrict CD8(+) T-cell-mediated allergic immune responses in the skin.
Collapse
Affiliation(s)
- Anton V Gorbachev
- Department of Immunology, Cleveland Clinic Foundation, Cleveland, OH, USA.
| | | |
Collapse
|
39
|
Abstract
The induction of cell death in immune cells by naturally occurring antibodies specific for death receptors may present an important antiinflammatory mechanism of intravenous immunoglobulin (IVIG). Conversely, the protection of tissue cells from death receptor-mediated apoptosis by blocking antibodies is thought to contribute to the beneficial effects of IVIG in certain inflammatory disorders such as toxic epidermal necrolysis, also known as Lyell's syndrome. In this review, we focus on recent insights into the role of functional antibodies against Fas, sialic acid-binding immunoglobulin-like lectin (Siglec)-8, and Siglec-9 receptors in IVIG-mediated cell survival or death effects. In addition, we examine a variety of factors in inflammatory disease that may interplay with these cellular events and influence the therapeutic efficacy or potency of IVIG. These involve activation status of the target cell, cytokine microenvironment, pathogenesis and stage of disease, individual genetic determinants, species characteristics, and batch-to-batch variations of IVIG preparations.
Collapse
Affiliation(s)
- Stephan von Gunten
- Institute of Pharmacology, University of Bern, Friedbühlstrasse 49, Bern, Switzerland
| | | |
Collapse
|
40
|
Regulation of memory CD8 T-cell differentiation by cyclin-dependent kinase inhibitor p27Kip1. Mol Cell Biol 2010; 30:5145-59. [PMID: 20805358 DOI: 10.1128/mcb.01045-09] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Induction of potent T-cell memory is the goal of vaccinations, but the molecular mechanisms that regulate the formation of memory CD8 T cells are not well understood. Despite the recognition that controls of cellular proliferation and apoptosis govern the number of memory T cells, the cell cycle regulatory mechanisms that control these key cellular processes in CD8 T cells during an immune response are poorly defined. Here, we have identified the cyclin-dependent kinase inhibitor p27(Kip1) as a critical regulator of the CD8 T-cell homeostasis at all phases of the T-cell response to an acute viral infection in mice. By acting as a timer for cell cycle exit, p27(Kip1) curtailed the programmed expansion of interleukin-2-producing memory precursors and markedly limited the magnitude and quality of CD8 T-cell memory. In the absence of p27(Kip1), CD8 T cells showed superior recall responses shortly after vaccination with recombinant Listeria monocytogenes. Additionally, we show that p27(Kip1) constrains proliferative renewal of memory CD8 T cells, especially of the effector memory subset. These findings provide critical insights into the cell cycle regulation of CD8 T-cell homeostasis and suggest that modulation of p27(Kip1) could bolster vaccine-induced T-cell memory and protective immunity.
Collapse
|
41
|
Almolda B, González B, Castellano B. Activated microglial cells acquire an immature dendritic cell phenotype and may terminate the immune response in an acute model of EAE. J Neuroimmunol 2010; 223:39-54. [PMID: 20451260 DOI: 10.1016/j.jneuroim.2010.03.021] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 03/17/2010] [Accepted: 03/31/2010] [Indexed: 12/23/2022]
Abstract
Antigen presentation, a key mechanism in immune responses, involves two main signals: the first is provided by the engagement of a major histocompatibility complex (MHC), class I or class II, with their TCR receptor in lymphocytes, whereas the second demands the participation of different co-stimulatory molecules, such as CD28, CTLA-4 and their receptors B7.1 and B7.2. Specific T-cell activation and deactivation are achieved through this signalling. The aim of our study is to characterise, in the acute experimental autoimmune encephalomyelitis (EAE) model in Lewis rat, the temporal expression pattern of these molecules as well as the cells responsible for their expression. To accomplish that, MBP-immunised female Lewis rats were daily examined for the presence of clinical symptoms and sacrificed, according to their clinical score, at different phases during EAE. Spinal cords were cut with a cryostat and processed for immunohistochemistry: MHC-class I and MHC-class II, co-stimulatory molecules (B7.1, B7.2, CD28, CTLA-4) and markers of dendritic cells (CD1 for immature cells and fascin for mature cells). Our results show that microglial cells are activated in the inductive phase and, during this phase and peak, they are able to express MHC-class I, MHC-class II and CD1, but not B7.1 and B7.2. This microglial phenotype may induce the apoptosis or anergy of infiltrated CD28+ lymphocytes observed around blood vessels and in the parenchyma. During the recovery phase, microglial cells express high MHC-class I and class II and, those located in the surroundings of blood vessels, displayed the B7.2 co-stimulatory molecule. These cells are competent to interact with CTLA-4+ cells, which indicate an active role of microglial cells in modulating the ending of the immune response by inducing lymphocyte activity inhibition and Treg activation. Once clinical symptomatology disappeared, some foci of activated microglial cells (MHC-class II+/B7.2+) were still present in concomitance with CTLA-4+ cells, suggesting a prolonged involvement of microglia in lymphocyte inhibition and tolerance promotion. In addition to microglia, during the inductive and recovery phases, we also found perivascular ED2+ cells and fascin+ cells which are able to migrate to the parenchyma and may play a role in lymphocytic regulation. Further studies to understand the specific function played by these cells are warranted.
Collapse
Affiliation(s)
- Beatriz Almolda
- Department of Cellular Biology, Physiology and Immunology, Autonomous University of Barcelona, Bellaterra, Spain.
| | | | | |
Collapse
|
42
|
Tsai S, Shameli A, Yamanouchi J, Clemente-Casares X, Wang J, Serra P, Yang Y, Medarova Z, Moore A, Santamaria P. Reversal of Autoimmunity by Boosting Memory-like Autoregulatory T Cells. Immunity 2010; 32:568-80. [DOI: 10.1016/j.immuni.2010.03.015] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2008] [Revised: 12/18/2009] [Accepted: 02/09/2010] [Indexed: 10/19/2022]
|
43
|
Russell MS, Dudani R, Krishnan L, Sad S. IFN-gamma expressed by T cells regulates the persistence of antigen presentation by limiting the survival of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2010; 183:7710-8. [PMID: 19923462 DOI: 10.4049/jimmunol.0901274] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ag presentation to T cells orchestrates the development of acquired immune response. Although it is considered that Ag presentation may persist at high levels during chronic infections, we have previously reported that in mice infected with bacillus Calmette-Guérin, Ag presentation gets drastically curtailed during the chronic stage of infection despite antigenic persistence. In this report we evaluated the mechanism of this curtailment. Ag presentation declined precipitously as the T cell response developed, and Ag presentation was not curtailed in mice that were deficient in CD8(+) T cells or MHC class II, suggesting that T cells regulate Ag presentation. Curtailment of Ag presentation was reduced in IFN-gamma-deficient mice, but not in mice with a deficiency/mutation in inducible NOS2, perforin, or Fas ligand. In hosts with no T cells (Rag1(-/-)), Ag presentation was not curtailed during the chronic stage of infection. However, adoptive transfer of wild-type, but not IFN-gamma(-/-), CD4(+) and CD8(+) T cells into Rag1-deficient hosts strongly curtailed Ag presentation. Increased persistence of Ag presentation in IFN-gamma-deficient hosts correlated to increased survival of dendritic cells, but not of macrophages, and was not due to increased stimulatory capacity of IFN-gamma-deficient dendritic cells. These results reveal a novel mechanism indicating how IFN-gamma prevents the persistence of Ag presentation, thereby preventing memory T cells from going into exhaustion.
Collapse
Affiliation(s)
- Marsha S Russell
- National Research Council Institute for Biological Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | |
Collapse
|
44
|
Linderman JJ, Riggs T, Pande M, Miller M, Marino S, Kirschner DE. Characterizing the dynamics of CD4+ T cell priming within a lymph node. THE JOURNAL OF IMMUNOLOGY 2010; 184:2873-85. [PMID: 20154206 DOI: 10.4049/jimmunol.0903117] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Generating adaptive immunity postinfection or immunization requires physical interaction within a lymph node T zone between Ag-bearing dendritic cells (DCs) and rare cognate T cells. Many fundamental questions remain regarding the dynamics of DC-CD4+ T cell interactions leading to priming. For example, it is not known how the production of primed CD4+ T cells relates to the numbers of cognate T cells, Ag-bearing DCs, or peptide-MHCII level on the DC. To address these questions, we developed an agent-based model of a lymph node to examine the relationships among cognate T cell frequency, DC density, parameters characterizing DC-T cell interactions, and the output of primed T cells. We found that the output of primed CD4+ T cells is linearly related to cognate frequency, but nonlinearly related to the number of Ag-bearing DCs present during infection. This addresses the applicability of two photon microscopy studies to understanding actual infection dynamics, because these types of experiments increase the cognate frequency by orders of magnitude compared with physiologic levels. We found a trade-off between the quantity of peptide-major histocompatibility class II on the surface of individual DCs and number of Ag-bearing DCs present in the lymph node in contributing to the production of primed CD4+ T cells. Interestingly, peptide-major histocompatibility class II t(1/2) plays a minor, although still significant, role in determining CD4+ T cell priming, unlike the primary role that has been suggested for CD8+ T cell priming. Finally, we identify several pathogen-targeted mechanisms that, if altered in their efficiency, can significantly effect the generation of primed CD4+ T cells.
Collapse
Affiliation(s)
- Jennifer J Linderman
- Department of Chemical Engineering, Center for Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
45
|
Boyman O, Cho JH, Sprent J. The Role of Interleukin-2 in Memory CD8 Cell Differentiation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 684:28-41. [DOI: 10.1007/978-1-4419-6451-9_3] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
Sener A, Tang AL, Farber DL. Memory T-cell predominance following T-cell depletional therapy derives from homeostatic expansion of naive T cells. Am J Transplant 2009; 9:2615-23. [PMID: 19775313 DOI: 10.1111/j.1600-6143.2009.02820.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
T-cell depletion reportedly leads to alterations in the T-cell compartment with predominant survival of memory phenotype CD4 T cells. Here, we asked whether the prevalence of memory T cells postdepletion results from their inherent resistance to depletion and/or to the homeostatic expansion of naive T cells and their phenotypic conversion to memory, which is known to occur in lymphopenic conditions. Using a 'mosaic memory' mouse model with trackable populations of alloreactive memory T cells, we found that treatment with murine antithymocyte globulin (mATG) or antilymphocyte serum (ALS) effectively depleted alloreactive memory CD4 T cells, followed by rapid homeostatic proliferation of endogenous CD4 T cells peaking at 4 days postdepletion, with no homeostatic advantage to the antigen-specific memory population. Interestingly, naive (CD44lo) CD4 T cells exhibited the greatest increase in homeostatic proliferation following mATG treatment, divided more extensively compared to memory (CD44hi) CD4 T cells and converted to a memory phenotype. Our results provide novel evidence that memory CD4 T cells are susceptible to lymphodepletion and that the postdepletional T-cell compartment is repopulated to a significant extent by homeostatically expanded naive T cells in a mouse model, with important important implications for immune alterations triggered by induction therapy.
Collapse
Affiliation(s)
- A Sener
- Division of Transplantation, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
47
|
Albaghdadi H, Robinson N, Finlay B, Krishnan L, Sad S. Selectively reduced intracellular proliferation of Salmonella enterica serovar typhimurium within APCs limits antigen presentation and development of a rapid CD8 T cell response. THE JOURNAL OF IMMUNOLOGY 2009; 183:3778-87. [PMID: 19692639 DOI: 10.4049/jimmunol.0900843] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ag presentation to CD8(+) T cells commences immediately after infection, which facilitates their rapid expansion and control of pathogen. This paradigm is not followed during infection with virulent Salmonella enterica serovar Typhimurium (ST), an intracellular bacterium that causes mortality in susceptible C57BL/6J mice within 7 days and a chronic infection in resistant mice (129 x 1SvJ). Infection of mice with OVA-expressing ST results in the development of a CD8(+) T cell response that is detectable only after the second week of infection despite the early detectable bacterial burden. The mechanism behind the delayed CD8(+) T cell activation was evaluated, and it was found that dendritic cells/macrophages or mice infected with ST-OVA failed to present Ag to OVA-specific CD8(+) T cells. Lack of early Ag presentation was not rescued when mice or dendritic cells/macrophages were infected with an attenuated aroA mutant of ST or with mutants having defective Salmonella pathogenicity island I/II genes. Although extracellular ST proliferated extensively, the replication of ST was highly muted once inside macrophages. This muted intracellular proliferation of ST resulted in the generation of poor levels of intracellular Ag and peptide-MHC complex on the surface of dendritic cells. Additional experiments revealed that ST did not actively inhibit Ag presentation, rather it inhibited the uptake of another intracellular pathogen, Listeria monocytogenes, thereby causing inhibition of Ag presentation against L. monocytogenes. Taken together, this study reveals a dichotomy in the proliferation of ST and indicates that selectively reduced intracellular proliferation of virulent pathogens may be an important mechanism of immune evasion.
Collapse
Affiliation(s)
- Homam Albaghdadi
- National Research Council Institute for Biological Sciences, Ottawa, Ontario, Canada
| | | | | | | | | |
Collapse
|
48
|
B7-H1 blockade increases survival of dysfunctional CD8(+) T cells and confers protection against Leishmania donovani infections. PLoS Pathog 2009; 5:e1000431. [PMID: 19436710 PMCID: PMC2674929 DOI: 10.1371/journal.ppat.1000431] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2008] [Accepted: 04/15/2009] [Indexed: 12/20/2022] Open
Abstract
Experimental visceral leishmaniasis (VL) represents an exquisite model to study CD8+ T cell responses in a context of chronic inflammation and antigen persistence, since it is characterized by chronic infection in the spleen and CD8+ T cells are required for the development of protective immunity. However, antigen-specific CD8+ T cell responses in VL have so far not been studied, due to the absence of any defined Leishmania-specific CD8+ T cell epitopes. In this study, transgenic Leishmania donovani parasites expressing ovalbumin were used to characterize the development, function, and fate of Leishmania-specific CD8+ T cell responses. Here we show that L. donovani parasites evade CD8+ T cell responses by limiting their expansion and inducing functional exhaustion and cell death. Dysfunctional CD8+ T cells could be partially rescued by in vivo B7-H1 blockade, which increased CD8+ T cell survival but failed to restore cytokine production. Nevertheless, B7-H1 blockade significantly reduced the splenic parasite burden. These findings could be exploited for the design of new strategies for immunotherapeutic interventions against VL. The protozoan parasite Leishmania donovani is the cause of visceral leishmaniasis, a chronic disease that currently affects 12 million people worldwide. We are interested in understanding the immune mechanisms that can control infection. Preliminary studies suggested that CD8+ T cells can kill parasites and limit disease; however, studying these important killer cells has been hindered, because we do not know what parasite molecules they recognize. To overcome this, we engineered parasites to express ovalbumin. Since many tools exist to track and measure immune cells targeted at ovalbumin, we can now track the specific CD8+ T cell responses that develop upon infection with Leishmania. We found that Leishmania initially induced CD8+ T cells to divide and produce molecules such as IFN-gamma that may help them to kill parasites. However, the CD8+ T cells rapidly lost their effector function and died off as infection progressed. More encouragingly, though, we were able to recover some CD8+ T cell function by blocking immune inhibitory molecules that are induced by parasite infection. The recovered T cells killed parasites and controlled infection. These results are important as they could be exploited for the design of new therapeutic vaccine strategies aimed at inducing protective CD8+ T cells.
Collapse
|
49
|
Cush SS, Flaño E. Protective antigen-independent CD8 T cell memory is maintained during {gamma}-herpesvirus persistence. THE JOURNAL OF IMMUNOLOGY 2009; 182:3995-4004. [PMID: 19299697 DOI: 10.4049/jimmunol.0803625] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ag persistence during high-titer chronic viral infections induces CD8 T cell dysfunction and lack of Ag-independent CD8 T cell memory formation. However, we have a poor understanding of the generation and maintenance of CD8 T cell memory during asymptomatic persistent viral infections, particularly gamma-herpesvirus infections. In this study, we demonstrate that the continuous presence of cognate Ag in the host is not required for the maintenance of CD8 T cell memory during a persistent gamma-herpesvirus infection. Importantly, the Ag-independent CD8 T cell memory that is maintained during gamma-herpesvirus persistence has the capacity to survive long-term under homeostatic conditions and to mount a protective recall response to a secondary encounter with the pathogen. These data highlight the ability of the immune system to maintain a population of protective memory CD8 T cells with capacity for long-term Ag-independent survival in the presence of systemic virus persistence.
Collapse
Affiliation(s)
- Stephanie S Cush
- Center for Vaccines and Immunity, Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | |
Collapse
|
50
|
Abstract
FAS belongs to the subgroup of the tumor necrosis factor receptor (TNF-R) family that contains an intracellular "death domain" and triggers apoptosis. Its physiological ligand FASL is a member of the TNF cytokine family. Studies with mutant mice and cells from human patients have shown that FAS plays critical roles in the immune system, including the killing of pathogen-infected cells and the death of obsolete and potentially dangerous lymphocytes. Fas thereby functions as a guardian against autoimmunity and tumor development. FAS triggers apoptosis through FADD-mediated recruitment and activation of caspase-8. In certain cells such as hepatocytes, albeit not lymphocytes, FAS-induced apoptosis requires amplification through proteolytic activation of the proapoptotic BCL-2 family member BID. Curiously, several components of the FAS signaling machinery have been implicated in nonapoptotic processes, including cellular activation, differentiation, and proliferation. This review describes current understanding of Fas-induced apoptosis signaling and proposes experimental strategies for future advances.
Collapse
Affiliation(s)
- Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.
| | | | | |
Collapse
|