1
|
Ma X, Ma R, Zhang M, Qian B, Wang B, Yang W. Recent Progress in Multiple Sclerosis Treatment Using Immune Cells as Targets. Pharmaceutics 2023; 15:pharmaceutics15030728. [PMID: 36986586 PMCID: PMC10057470 DOI: 10.3390/pharmaceutics15030728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/02/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune-mediated demyelinating disease of the central nervous system. The main pathological features are inflammatory reaction, demyelination, axonal disintegration, reactive gliosis, etc. The etiology and pathogenesis of the disease have not been clarified. The initial studies believed that T cell-mediated cellular immunity is the key to the pathogenesis of MS. In recent years, more and more evidence has shown that B cells and their mediated humoral immune and innate immune cells (such as microglia, dendritic cells, macrophages, etc.) also play an important role in the pathogenesis of MS. This article mainly reviews the research progress of MS by targeting different immune cells and analyzes the action pathways of drugs. The types and mechanisms of immune cells related to the pathogenesis are introduced in detail, and the mechanisms of drugs targeting different immune cells are discussed in depth. This article aims to clarify the pathogenesis and immunotherapy pathway of MS, hoping to find new targets and strategies for the development of therapeutic drugs for MS.
Collapse
Affiliation(s)
- Xiaohong Ma
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
- The First Clinical Medical School, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Rong Ma
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Mengzhe Zhang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
| | - Baicheng Qian
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Baoliang Wang
- Department of Neuroscience, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
- Correspondence: (B.W.); (W.Y.)
| | - Weijing Yang
- School of Pharmaceutical Sciences, Henan Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China
- Correspondence: (B.W.); (W.Y.)
| |
Collapse
|
2
|
Zhao J, Jung S, Li X, Li L, Kasinath V, Zhang H, Movahedi SN, Mardini A, Sabiu G, Hwang Y, Saxena V, Song Y, Ma B, Acton SE, Kim P, Madsen JC, Sage PT, Tullius SG, Tsokos GC, Bromberg JS, Abdi R. Delivery of costimulatory blockade to lymph nodes promotes transplant acceptance in mice. J Clin Invest 2022; 132:e159672. [PMID: 36519543 PMCID: PMC9754003 DOI: 10.1172/jci159672] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 10/11/2022] [Indexed: 12/15/2022] Open
Abstract
The lymph node (LN) is the primary site of alloimmunity activation and regulation during transplantation. Here, we investigated how fibroblastic reticular cells (FRCs) facilitate the tolerance induced by anti-CD40L in a murine model of heart transplantation. We found that both the absence of LNs and FRC depletion abrogated the effect of anti-CD40L in prolonging murine heart allograft survival. Depletion of FRCs impaired homing of T cells across the high endothelial venules (HEVs) and promoted formation of alloreactive T cells in the LNs in heart-transplanted mice treated with anti-CD40L. Single-cell RNA sequencing of the LNs showed that anti-CD40L promotes a Madcam1+ FRC subset. FRCs also promoted the formation of regulatory T cells (Tregs) in vitro. Nanoparticles (NPs) containing anti-CD40L were selectively delivered to the LNs by coating them with MECA-79, which binds to peripheral node addressin (PNAd) glycoproteins expressed exclusively by HEVs. Treatment with these MECA-79-anti-CD40L-NPs markedly delayed the onset of heart allograft rejection and increased the presence of Tregs. Finally, combined MECA-79-anti-CD40L-NPs and rapamycin treatment resulted in markedly longer allograft survival than soluble anti-CD40L and rapamycin. These data demonstrate that FRCs are critical to facilitating costimulatory blockade. LN-targeted nanodelivery of anti-CD40L could effectively promote heart allograft acceptance.
Collapse
Affiliation(s)
- Jing Zhao
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Sungwook Jung
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaofei Li
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Lushen Li
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Vivek Kasinath
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hengcheng Zhang
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Said N. Movahedi
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ahmad Mardini
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Gianmarco Sabiu
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Yoonha Hwang
- IVIM Technology, Daejeon, South Korea
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Vikas Saxena
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Bing Ma
- Institute for Genome Sciences and
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Sophie E. Acton
- Stromal Immunology Group, Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Pilhan Kim
- IVIM Technology, Daejeon, South Korea
- Graduate School of Nanoscience and Technology and
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Joren C. Madsen
- Center for Transplantation Sciences, Department of Surgery
- Division of Cardiac Surgery, Department of Surgery, and
| | - Peter T. Sage
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Stefan G. Tullius
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - George C. Tsokos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan S. Bromberg
- Department of Surgery and
- Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Reza Abdi
- Transplantation Research Center and
- Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
de Leve S, Wirsdörfer F, Jendrossek V. The CD73/Ado System-A New Player in RT Induced Adverse Late Effects. Cancers (Basel) 2019; 11:cancers11101578. [PMID: 31623231 PMCID: PMC6827091 DOI: 10.3390/cancers11101578] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 10/11/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) is a central component of standard treatment for many cancer patients. RT alone or in multimodal treatment strategies has a documented contribution to enhanced local control and overall survival of cancer patients, and cancer cure. Clinical RT aims at maximizing tumor control, while minimizing the risk for RT-induced adverse late effects. However, acute and late toxicities of IR in normal tissues are still important biological barriers to successful RT: While curative RT may not be tolerable, sub-optimal tolerable RT doses will lead to fatal outcomes by local recurrence or metastatic disease, even when accepting adverse normal tissue effects that decrease the quality of life of irradiated cancer patients. Technical improvements in treatment planning and the increasing use of particle therapy have allowed for a more accurate delivery of IR to the tumor volume and have thereby helped to improve the safety profile of RT for many solid tumors. With these technical and physical strategies reaching their natural limits, current research for improving the therapeutic gain of RT focuses on innovative biological concepts that either selectively limit the adverse effects of RT in normal tissues without protecting the tumor or specifically increase the radiosensitivity of the tumor tissue without enhancing the risk of normal tissue complications. The biology-based optimization of RT requires the identification of biological factors that are linked to differential radiosensitivity of normal or tumor tissues, and are amenable to therapeutic targeting. Extracellular adenosine is an endogenous mediator critical to the maintenance of homeostasis in various tissues. Adenosine is either released from stressed or injured cells or generated from extracellular adenine nucleotides by the concerted action of the ectoenzymes ectoapyrase (CD39) and 5′ ectonucleotidase (NT5E, CD73) that catabolize ATP to adenosine. Recent work revealed a role of the immunoregulatory CD73/adenosine system in radiation-induced fibrotic disease in normal tissues suggesting a potential use as novel therapeutic target for normal tissue protection. The present review summarizes relevant findings on the pathologic roles of CD73 and adenosine in radiation-induced fibrosis in different organs (lung, skin, gut, and kidney) that have been obtained in preclinical models and proposes a refined model of radiation-induced normal tissue toxicity including the disease-promoting effects of radiation-induced activation of CD73/adenosine signaling in the irradiated tissue environment. However, expression and activity of the CD73/adenosine system in the tumor environment has also been linked to increased tumor growth and tumor immune escape, at least in preclinical models. Therefore, we will discuss the use of pharmacologic inhibition of CD73/adenosine-signaling as a promising strategy for improving the therapeutic gain of RT by targeting both, malignant tumor growth and adverse late effects of RT with a focus on fibrotic disease. The consideration of the therapeutic window is particularly important in view of the increasing use of RT in combination with various molecularly targeted agents and immunotherapy to enhance the tumor radiation response, as such combinations may result in increased or novel toxicities, as well as the increasing number of cancer survivors.
Collapse
Affiliation(s)
- Simone de Leve
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen, 45122 Essen, Germany.
| |
Collapse
|
4
|
Yang WC, Hwang YS, Chen YY, Liu CL, Shen CN, Hong WH, Lo SM, Shen CR. Interleukin-4 Supports the Suppressive Immune Responses Elicited by Regulatory T Cells. Front Immunol 2017; 8:1508. [PMID: 29184551 PMCID: PMC5694475 DOI: 10.3389/fimmu.2017.01508] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 10/25/2017] [Indexed: 11/13/2022] Open
Abstract
Interleukin-4 (IL-4) has been considered as one of the tolerogenic cytokines in many autoimmune animal models and clinical settings. Despite its role in antagonizing pathogenic Th1 responses, little is known about whether IL-4 possesses functions that affect regulatory T cells (Tregs). Tregs are specialized cells responsible for the maintenance of peripheral tolerance through their immune modulatory capabilities. Interestingly, it has been suggested that IL-4 supplement at a high concentration protects responder T cells (Tresps) from Treg-mediated immune suppression. In addition, such supplement also impedes TGF-β-induced Treg differentiation in vitro. However, these phenomena may contradict the tolerogenic role of IL-4, and the effects of IL-4 on Tregs are therefore needed to be further elucidated. In this study, we utilized IL-4 knockout (KO) mice to validate the role of IL-4 on Treg-mediated immune suppression. Although IL-4 KO and control animals harbor similar frequencies of Tregs, Tregs from IL-4 KO mice weakly suppressed autologous Tresp activation. In addition, IL-4 deprivation impaired the ability of Tregs to modulate immune response, whereas IL-4 supplementation reinforced IL-4 KO Tregs in their function in suppressing Tresps. Finally, the presence of IL-4 was associated with increased cell survival and granzyme expression of Tregs. These results suggest the essential role of IL-4 in supporting Treg-mediated immune suppression, which may benefit the development of therapeutic strategies for autoimmune diseases.
Collapse
Affiliation(s)
- Wei-Cheng Yang
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Yih-Shiou Hwang
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Ophthalmology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| | - Ying-Yu Chen
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chao-Lin Liu
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.,College of Engineering, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Wei-Hsin Hong
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Sheng-Min Lo
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chia-Rui Shen
- Department and Graduate Institute of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan.,Department of Ophthalmology, Lin-Kou Chang Gung Memorial Hospital, Taoyuan City, Taiwan
| |
Collapse
|
5
|
Ventura J, Lobachevsky PN, Palazzolo JS, Forrester H, Haynes NM, Ivashkevich A, Stevenson AW, Hall CJ, Ntargaras A, Kotsaris V, Pollakis GC, Potsi G, Skordylis K, Terzoudi G, Pateras IS, Gorgoulis VG, Georgakilas AG, Sprung CN, Martin OA. Localized Synchrotron Irradiation of Mouse Skin Induces Persistent Systemic Genotoxic and Immune Responses. Cancer Res 2017; 77:6389-6399. [PMID: 29113972 DOI: 10.1158/0008-5472.can-17-1066] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/07/2017] [Accepted: 09/19/2017] [Indexed: 11/16/2022]
Abstract
The importance of nontargeted (systemic) effects of ionizing radiation is attracting increasing attention. Exploiting synchrotron radiation generated by the Imaging and Medical Beamline at the Australian Synchrotron, we studied radiation-induced nontargeted effects in C57BL/6 mice. Mice were locally irradiated with a synchrotron X-ray broad beam and a multiplanar microbeam radiotherapy beam. To assess the influence of the beam configurations and variations in peak dose and irradiated area in the response of normal tissues outside the irradiated field at 1 and 4 days after irradiation, we monitored oxidatively induced clustered DNA lesions (OCDL), DNA double-strand breaks (DSB), apoptosis, and the local and systemic immune responses. All radiation settings induced pronounced persistent systemic effects in mice, which resulted from even short exposures of a small irradiated area. OCDLs were elevated in a wide variety of unirradiated normal tissues. In out-of-field duodenum, there was a trend for elevated apoptotic cell death under most irradiation conditions; however, DSBs were elevated only after exposure to lower doses. These genotoxic events were accompanied by changes in plasma concentrations of macrophage-derived cytokine, eotaxin, IL10, TIMP1, VEGF, TGFβ1, and TGFβ2, along with changes in tissues in frequencies of macrophages, neutrophils, and T lymphocytes. Overall, our findings have implications for the planning of therapeutic and diagnostic radiation treatments to reduce the risk of radiation-related adverse systemic effects. Cancer Res; 77(22); 6389-99. ©2017 AACR.
Collapse
Affiliation(s)
- Jessica Ventura
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Obstetrics and Gynaecology, Royal Women's Hospital, University of Melbourne, Melbourne, Victoria, Australia
| | - Pavel N Lobachevsky
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jason S Palazzolo
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Helen Forrester
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| | - Nicole M Haynes
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alesia Ivashkevich
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia.,Radiation Oncology, Canberra Hospital, Garran, Australian Capital Territory, Australia
| | - Andrew W Stevenson
- CSIRO, Clayton, Victoria, Australia.,Australian Synchrotron, Clayton, Victoria, Australia
| | | | - Andreas Ntargaras
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Vasilis Kotsaris
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gerasimos Ch Pollakis
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Gianna Potsi
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Konstantinos Skordylis
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Georgia Terzoudi
- Laboratory of Health Physics, Radiobiology and Cytogenetics, Institute of Nuclear and Radiological Sciences and Technology, Energy and Safety, National Center for Scientific Research 'Demokritos', Athens, Greece
| | - Ioannis S Pateras
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, United Kingdom
| | - Alexandros G Georgakilas
- Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens, Athens, Greece
| | - Carl N Sprung
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research and Monash University, Clayton, Victoria, Australia
| | - Olga A Martin
- Molecular Radiation Biology Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. .,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
6
|
Bieńkowska A, Kiernozek E, Kozlowska E, Bugajski Ł, Drela N. A new approach to the role of IL-7 and TGF-ß in the in vitro generation of thymus-derived CD4+CD25+Foxp3+ regulatory T cells. Cytokine 2017; 102:107-116. [PMID: 28826649 DOI: 10.1016/j.cyto.2017.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/28/2017] [Accepted: 07/24/2017] [Indexed: 12/28/2022]
Abstract
Thymus-derived regulatory T cells of CD4+CD25+Foxp3+ phenotype develop as a functional, mature population playing an essential role in self-tolerance and immune homeostasis, and exhibiting therapeutic potential to inhibit adverse immune response. Despite intensive research on thymus-derived Tregs, the knowledge about agents involved in their generation, survival, proliferation, and biological functions is still insufficient. In this research we have focused on the role of selected cytokines in previously developed in vitro model based on the application of anti-CD3 monoclonal antibodies. We have demonstrated an essential role of IL-7 and TGF-β in the generation of thymus-derived Tregs in the co-culture of thymocytes and JAWS II cells. In addition, in vitro generated Tregs exhibited their suppressive function similarly to Tregs sorted from freshly isolated thymus.
Collapse
Affiliation(s)
- Anna Bieńkowska
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland.
| | - Ewelina Kiernozek
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Ewa Kozlowska
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Łukasz Bugajski
- The Laboratory of Cytometry, Nencki Institute of Experimental Biology PAS, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Nadzieja Drela
- University of Warsaw, Faculty of Biology, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| |
Collapse
|
7
|
Wirsdörfer F, Jendrossek V. The Role of Lymphocytes in Radiotherapy-Induced Adverse Late Effects in the Lung. Front Immunol 2016; 7:591. [PMID: 28018357 PMCID: PMC5155013 DOI: 10.3389/fimmu.2016.00591] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 11/29/2016] [Indexed: 12/31/2022] Open
Abstract
Radiation-induced pneumonitis and fibrosis are dose-limiting side effects of thoracic irradiation. Thoracic irradiation triggers acute and chronic environmental lung changes that are shaped by the damage response of resident cells, by the resulting reaction of the immune system, and by repair processes. Although considerable progress has been made during the last decade in defining involved effector cells and soluble mediators, the network of pathophysiological events and the cellular cross talk linking acute tissue damage to chronic inflammation and fibrosis still require further definition. Infiltration of cells from the innate and adaptive immune systems is a common response of normal tissues to ionizing radiation. Herein, lymphocytes represent a versatile and wide-ranged group of cells of the immune system that can react under specific conditions in various ways and participate in modulating the lung environment by adopting pro-inflammatory, anti-inflammatory, or even pro- or anti-fibrotic phenotypes. The present review provides an overview on published data about the role of lymphocytes in radiation-induced lung disease and related damage-associated pulmonary diseases with a focus on T lymphocytes and B lymphocytes. We also discuss the suspected dual role of specific lymphocyte subsets during the pneumonitic phase and fibrotic phase that is shaped by the environmental conditions as well as the interaction and the intercellular cross talk between cells from the innate and adaptive immune systems and (damaged) resident epithelial cells and stromal cells (e.g., endothelial cells, mesenchymal stem cells, and fibroblasts). Finally, we highlight potential therapeutic targets suited to counteract pathological lymphocyte responses to prevent or treat radiation-induced lung disease.
Collapse
Affiliation(s)
- Florian Wirsdörfer
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| | - Verena Jendrossek
- Institute of Cell Biology (Cancer Research), University Hospital Essen , Essen , Germany
| |
Collapse
|
8
|
van der Merwe M, Abdelsamed HA, Seth A, Ong T, Vogel P, Pillai AB. Recipient myeloid-derived immunomodulatory cells induce PD-1 ligand-dependent donor CD4+Foxp3+ regulatory T cell proliferation and donor-recipient immune tolerance after murine nonmyeloablative bone marrow transplantation. THE JOURNAL OF IMMUNOLOGY 2013; 191:5764-76. [PMID: 24190658 DOI: 10.4049/jimmunol.1302191] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We showed previously that nonmyeloablative total lymphoid irradiation/rabbit anti-thymocyte serum (TLI/ATS) conditioning facilitates potent donor-recipient immune tolerance following bone marrow transplantation (BMT) across MHC barriers via recipient invariant NKT (iNKT) cell-derived IL-4-dependent expansion of donor Foxp3(+) naturally occurring regulatory T cells (nTregs). In this study, we report a more specific mechanism. Wild-type (WT) BALB/c (H-2(d)) hosts were administered TLI/ATS and BMT from WT or STAT6(-/-) C57BL/6 (H-2(b)) donors. Following STAT6(-/-) BMT, donor nTregs demonstrated no loss of proliferation in vivo, indicating that an IL-4-responsive population in the recipient, rather than the donor, drives donor nTreg proliferation. In graft-versus-host disease (GVHD) target organs, three recipient CD11b(+) cell subsets (Gr-1(high)CD11c(-), Gr-1(int)CD11c(-), and Gr-1(low)CD11c(+)) were enriched early after TLI/ATS + BMT versus total body irradiation/ATS + BMT. Gr-1(low)CD11c(+) cells induced potent H-2K(b+)CD4(+)Foxp3(+) nTreg proliferation in vitro in 72-h MLRs. Gr-1(low)CD11c(+) cells were reduced significantly in STAT6(-/-) and iNKT cell-deficient Jα18(-/-) BALB/c recipients after TLI/ATS + BMT. Depletion of CD11b(+) cells resulted in severe acute GVHD, and adoptive transfer of WT Gr-1(low)CD11c(+) cells to Jα18(-/-) BALB/c recipients of TLI/ATS + BMT restored day-6 donor Foxp3(+) nTreg proliferation and protection from CD8 effector T cell-mediated GVHD. Blockade of programmed death ligand 1 and 2, but not CD40, TGF-β signaling, arginase 1, or iNOS, inhibited nTreg proliferation in cocultures of recipient-derived Gr-1(low)CD11c(+) cells with donor nTregs. Through iNKT-dependent Th2 polarization, myeloid-derived immunomodulatory dendritic cells are expanded after nonmyeloablative TLI/ATS conditioning and allogeneic BMT, induce PD-1 ligand-dependent donor nTreg proliferation, and maintain potent graft-versus-host immune tolerance.
Collapse
Affiliation(s)
- Marie van der Merwe
- Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | | | | | | | | |
Collapse
|
9
|
Quan L, Gong Z, Yao S, Bandera EV, Zirpoli G, Hwang H, Roberts M, Ciupak G, Davis W, Sucheston L, Pawlish K, Bovbjerg DH, Jandorf L, Cabasag C, Coignet JG, Ambrosone CB, Hong CC. Cytokine and cytokine receptor genes of the adaptive immune response are differentially associated with breast cancer risk in American women of African and European ancestry. Int J Cancer 2013; 134:1408-21. [PMID: 23996684 DOI: 10.1002/ijc.28458] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 07/18/2013] [Accepted: 08/13/2013] [Indexed: 12/21/2022]
Abstract
Disparities in breast cancer biology are evident between American women of African ancestry (AA) and European ancestry (EA) and may be due, in part, to differences in immune function. To assess the potential role of constitutional host immunity on breast carcinogenesis, we tested associations between breast cancer risk and 47 single nucleotide polymorphisms (SNPs) in 26 cytokine-related genes of the adaptive immune system using 650 EA (n = 335 cases) and 864 AA (n = 458 cases) women from the Women's Circle of Health Study (WCHS). With additional participant accrual to the WCHS, promising SNPs from the initial analysis were evaluated in a larger sample size (1,307 EAs and 1,365 AAs). Multivariate logistic regression found SNPs in genes important for T helper type 1 (Th1) immunity (IFNGR2 rs1059293, IL15RA rs2296135, LTA rs1041981), Th2 immunity (IL4R rs1801275), and T regulatory cell-mediated immunosuppression (TGFB1 rs1800469) associated with breast cancer risk, mainly among AAs. The combined effect of these five SNPs was highly significant among AAs (P-trend = 0.0005). When stratified by estrogen receptor (ER) status, LTA rs1041981 was associated with ER-positive breast cancers among EAs and marginally among AAs. Only among AA women, IL15 rs10833 and IL15RA rs2296135 were associated with ER-positive tumors, and IL12RB1 rs375947, IL15 rs10833 and TGFB1 rs1800469 were associated with ER-negative tumors. Our study systematically identified genetic variants in the adaptive immune response pathway associated with breast cancer risk, which appears to differ by ancestry groups, menopausal status and ER status.
Collapse
Affiliation(s)
- Lei Quan
- Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, NY
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Conte E, Gili E, Fruciano M, Fagone E, Vancheri C. Human lung fibroblasts increase CD4(+)CD25(+)Foxp3(+) T cells in co-cultured CD4(+) lymphocytes. Cell Immunol 2013; 285:55-61. [PMID: 24076465 DOI: 10.1016/j.cellimm.2013.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 09/02/2013] [Accepted: 09/03/2013] [Indexed: 12/29/2022]
Abstract
Aim of this study was to evaluate functional modifications induced by human lung fibroblasts in co-cultured CD4(+) T lymphocytes. CD4(+) T cells, resting or stimulated with ionomycin/PMA for 6h, were co-cultured with fibroblasts isolated from pulmonary biopsies, in contact or separated by a semi-permeable membrane. The expression of CD25, CTLA-4, TGF-β, IFNγ, IL-2, IL-4, IL-10 and Foxp3 was evaluated by flow cytometric analysis. Fibroblasts induced a significant increment in CD25(+) cells in co-cultured activated CD4(+) T lymphocytes separated by a membrane. Moreover, fibroblasts treatment with a COX2 inhibitor abrogated the increment in CD25(+) cells whereas exogenous PGE2 restored it. The CD25(+) subpopulation was characterized by increased presence of Fox-P3, CTLA-4, IL-10 and TGF-β positive cells while IFN-γ and IL-2 positive cells were diminished. Proliferative response of CD4(+) to the anti CD3/CD28-Abs was abrogated in CD4(+) co-cultured with fibroblasts thus demonstrating a suppressive feature of the expanded CD25(+) subpopulation.
Collapse
Affiliation(s)
- Enrico Conte
- Department of Clinical and MolecularBiomedicine, University of Catania, Catania, Italy.
| | | | | | | | | |
Collapse
|
11
|
Activation-induced FoxP3 expression regulates cytokine production in conventional T cells stimulated with autologous dendritic cells. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 19:1583-92. [PMID: 22855393 DOI: 10.1128/cvi.00308-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A defining feature of dendritic cells (DCs) is their ability to induce the proliferation of autologous T cells in the absence of foreign antigen-a process termed the "autologous mixed leukocyte reaction" (AMLR). We report that equine monocyte-derived DCs, but not macrophages, are potent inducers of the AMLR. The response is contact dependent and major histocompatibility complex class II dependent and primarily involves CD3(+) CD4(+) CD8(-) T cells. Upon stimulation with DCs or the mitogen concanavalin A, a subset of the proliferating T cells expresses the regulatory T-cell (Treg) transcription factor FoxP3. Although many of these FoxP3(+) T cells are capable of producing the effector cytokines interleukin-4 (IL-4) and gamma interferon (IFN-γ), they are more likely to produce IL-10 and less likely to produce IFN-γ than equivalent FoxP3(-) cells. Therefore, FoxP3 expression is an inherent component of equine T cell activation and is associated with a more immunosuppressive cytokine profile. These results confirm that FoxP3 expression in the horse, in contrast to the mouse, is regulated similarly to FOXP3 expression in humans and provide evidence that FoxP3 expression by conventional T cells may help regulate the developing immune response.
Collapse
|
12
|
IL-12 inhibits the TGF-β-dependent T cell developmental programs and skews the TGF-β-induced differentiation into a Th1-like direction. Immunobiology 2012; 217:74-82. [DOI: 10.1016/j.imbio.2011.07.032] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Revised: 07/19/2011] [Accepted: 07/29/2011] [Indexed: 01/05/2023]
|
13
|
Regulation of non-classical major histocompatability complex class I mRNA expression in bovine embryos. J Reprod Immunol 2011; 91:31-40. [DOI: 10.1016/j.jri.2011.05.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 05/10/2011] [Accepted: 05/17/2011] [Indexed: 01/28/2023]
|
14
|
Svobodova E, Krulova M, Zajicova A, Pokorna K, Prochazkova J, Trosan P, Holan V. The role of mouse mesenchymal stem cells in differentiation of naive T-cells into anti-inflammatory regulatory T-cell or proinflammatory helper T-cell 17 population. Stem Cells Dev 2011; 21:901-10. [PMID: 21663543 DOI: 10.1089/scd.2011.0157] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (MSCs) modulate immune response and can produce significant levels of transforming growth factor-β (TGF-β) and interleukin-6 (IL-6). These 2 cytokines represent the key factors that reciprocally regulate the development and polarization of naive T-cells into regulatory T-cell (Treg) population or proinflammatory T helper 17 (Th17) cells. In the present study we demonstrate that MSCs and their products effectively regulate expression of transcription factors Foxp3 and RORγt and control the development of Tregs and Th17 cells in a population of alloantigen-activated mouse spleen cells or purified CD4(+)CD25(-) T-cells. The immunomodulatory effects of MSCs were more pronounced when these cells were stimulated to secrete TGF-β alone or TGF-β together with IL-6. Unstimulated MSCs produce TGF-β, but not IL-6, and the production of TGF-β can be further enhanced by the anti-inflammatory cytokines IL-10 or TGF-β. In the presence of proinflammatory cytokines, MSCs secrete significant levels of IL-6, in addition to a spontaneous production of TGF-β. MSCs producing TGF-β induced preferentially expression of Foxp3 and activation of Tregs, whereas MSC supernatants containing TGF-β together with IL-6 supported RORγt expression and development of Th17 cells. The effects of MSC supernatants were blocked by the inclusion of neutralization monoclonal antibody anti-TGF-β or anti-IL-6 into the culture system. The results showed that MSCs represent important players that reciprocally regulate the development and differentiation of uncommitted naive T-cells into anti-inflammatory Foxp3(+) Tregs or proinflammatory RORγt(+) Th17 cell population and thereby can modulate autoimmune, immunopathological, and transplantation reactions.
Collapse
Affiliation(s)
- Eliska Svobodova
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
15
|
Hamza E, Steinbach F, Marti E. CD4+CD25+ T cells expressing FoxP3 in Icelandic horses affected with insect bite hypersensitivity. Vet Immunol Immunopathol 2011; 148:139-44. [PMID: 21700344 DOI: 10.1016/j.vetimm.2011.05.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 03/15/2011] [Accepted: 05/30/2011] [Indexed: 02/01/2023]
Abstract
Insect bite hypersensitivity (IBH) is an IgE-mediated dermatitis caused by bites of midges from the genus Culicoides. We have shown previously that peripheral blood mononuclear cells (PBMC) from IBH-affected horses produce higher levels of IL-4 and lower levels of IL-10 and TGF-β1 than those from healthy horses, suggesting that IBH is associated with a reduced regulatory immune response. FoxP3 is a crucial marker of regulatory T cells (Tregs). Here we have determined the proportion of CD4(+)CD25(+)FoxP3(+) T cells by flow cytometry in PBMC directly after isolation or after stimulation with Culicoides extract or a control antigen (Tetanus Toxoid). There were no differences between healthy and IBH horses either in the proportion of FoxP3(+)CD4(+)CD25(+) cells in freshly isolated PBMC or in the following stimulation with Tetanus Toxoid. However, upon stimulation of PBMC with the allergen, expression of FoxP3 by CD4(+)CD25(+high) and CD4(+)CD25(+dim) cells was significantly higher in healthy than in IBH horses. Addition of recombinant IL-4 to PBMC from healthy horses stimulated with the allergen significantly decreased the proportion of FoxP3 expressing cells within CD4(+)CD25(+high). These results suggest that IBH is associated with a decreased number of allergen-induced Tregs. This could be a consequence of the increased IL-4 production by PBMC of IBH-affected horses.
Collapse
Affiliation(s)
- Eman Hamza
- Department of Clinical Research and Veterinary Public Health, Vetsuisse Faculty, University of Bern, Bern, Switzerland.
| | | | | |
Collapse
|
16
|
Garden O, Pinheiro D, Cunningham F. All creatures great and small: regulatory T cells in mice, humans, dogs and other domestic animal species. Int Immunopharmacol 2011; 11:576-88. [DOI: 10.1016/j.intimp.2010.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 11/01/2010] [Indexed: 12/12/2022]
|
17
|
Nonredundant roles of IL-10 and TGF-β in suppression of immune responses to hepatic AAV-factor IX gene transfer. Mol Ther 2011; 19:1263-72. [PMID: 21386826 DOI: 10.1038/mt.2011.33] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hepatic gene transfer using adeno-associated viral (AAV) vectors has been shown to efficiently induce immunological tolerance to a variety of proteins. Regulatory T-cells (Treg) induced by this route suppress humoral and cellular immune responses against the transgene product. In this study, we examined the roles of immune suppressive cytokines interleukin-10 (IL-10) and transforming growth factor-β (TGF-β) in the development of tolerance to human coagulation factor IX (hF.IX). Interestingly, IL-10 deficient C57BL/6 mice receiving gene transfer remained tolerant to hF.IX and generated Treg that suppressed anti-hF.IX formation. Effects of TGF-β blockade were also minor in this strain. In contrast, in C3H/HeJ mice, a strain known to have stronger T-cell responses against hF.IX, IL-10 was specifically required for the suppression of CD8(+) T-cell infiltration of the liver. Furthermore, TGF-β was critical for tipping the balance toward an regulatory immune response. TGF-β was required for CD4(+)CD25(+)FoxP3(+) Treg induction, which was necessary for suppression of effector CD4(+) and CD8(+) T-cell responses as well as antibody formation. These results demonstrate the crucial, nonredundant roles of IL-10 and TGF-β in prevention of immune responses against AAV-F.IX-transduced hepatocytes.
Collapse
|
18
|
Neutralization of IL-4 reverses the nonresponsiveness of CD4+ T cells to regulatory T-cell induction in non-responder mouse strains. Mol Immunol 2010; 48:137-46. [PMID: 20869773 DOI: 10.1016/j.molimm.2010.08.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 08/09/2010] [Accepted: 08/31/2010] [Indexed: 01/08/2023]
Abstract
It is well established that naive cells can be converted by TGF-β into CD4(+)CD25(+) regulatory T (Treg) cells with therapeutic potentials. Likewise, it is shown that all-trans retinoic acid (ATRA) can greatly enhance TGF-β-induced Treg conversion, a phenomenon which has mainly been studied in C57BL/6 mice. Here we show that, although purified naive cells are highly susceptible to Treg generation, total CD4(+) T-cell populations from different mouse strains display significantly different sensitivities to TGF-β/ATRA-induced Treg conversion. The resistance of "non-responder" strains is associated with an enhanced production of IL-4 by memory T cells as well as an increased sensitivity of naive T cells to the action of IL-4. Importantly, neutralization of IL-4 overcomes the differences, thereby enabling TGF-β/ATRA to generate large numbers of functional Treg cells from total CD4(+) T cells in a consistent manner across different mouse strains. Likewise, blockade of IL-4 significantly enhances TGF-β/ATRA-induced Treg conversion from human naive T cells in the presence of memory cells. These results show that the inherent resistance of "non-responder" mouse strains to Treg conversion induced by TGF-β and ATRA can be reverted by neutralization of IL-4 and that inhibiting the action of IL-4 is beneficial or even necessary for consistent inducible Treg generation.
Collapse
|
19
|
Kasten-Jolly J, Heo Y, Lawrence DA. Impact of developmental lead exposure on splenic factors. Toxicol Appl Pharmacol 2010; 247:105-15. [PMID: 20542052 DOI: 10.1016/j.taap.2010.06.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 05/25/2010] [Accepted: 06/03/2010] [Indexed: 01/09/2023]
Abstract
Lead (Pb) is known to alter the functions of numerous organ systems, including the hematopoietic and immune systems. Pb can induce anemia and can lower host resistance to bacterial and viral infections. The anemia is due to Pb's inhibition of hemoglobin synthesis and Pb's induction of membrane changes, leading to early erythrocyte senescence. Pb also increases B-cell activation/proliferation and skews T-cell help (Th) toward Th2 subset generation. The specific mechanisms for many of the Pb effects are, as yet, not completely understood. Therefore, we performed gene expression analysis, via microarray, on RNA from the spleens of developmentally Pb-exposed mice, in order to gain further insight into these Pb effects. Splenic RNA microarray analysis indicated strong up-regulation of genes coding for proteolytic enzymes, lipases, amylase, and RNaseA. The data also showed that Pb affected the expression of many genes associated with innate immunity. Analysis of the microarray results via GeneSifter software indicated that Pb increased apoptosis, B-cell differentiation, and Th2 development. Direct up-regulation by Pb of expression of the gene encoding the heme-regulated inhibitor (HRI) suggested that Pb can decrease erythropoiesis by blocking globin mRNA translation. Pb's high elevation of digestive/catabolizing enzymes could generate immunogenic self peptides. With Pb's potential to induce new self-peptides and to enhance the expression of caspases, cytokines, and other immunomodulators, further evaluation of Pb's involvement in autoimmune phenomena, especially Th2-mediated autoantibody production, and alteration of organ system activities is warranted.
Collapse
Affiliation(s)
- Jane Kasten-Jolly
- Laboratory of Clinical and Experimental Endocrinology and Immunology, Wadsworth Center, Albany, NY 12201-0509, USA
| | | | | |
Collapse
|
20
|
Holan V, Pokorna K, Prochazkova J, Krulova M, Zajicova A. Immunoregulatory properties of mouse limbal stem cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:2124-9. [PMID: 20065115 DOI: 10.4049/jimmunol.0903049] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Stem cells have been demonstrated in nearly all adult mammalian tissues and play a vital role in their physiological renewal and healing after injury. Due to their irreplaceable role in tissue repair, these cells had to develop mechanisms protecting them from deleterious inflammatory immune reactions and ensuring their increased resistance to various apoptosis-inducing agents. In this study, we demonstrate that a population of mouse limbal cells highly enriched for cells expressing markers and characteristics of limbal stem cells (LSCs) suppresses in a dose-dependent manner the proliferation of lymphocytes elicited by mitogens or TCR-triggering and significantly inhibits the production of proinflammatory cytokines by activated T cells. The suppression was mediated by soluble factor(s) and did not affect early cell activation. LSCs were even more suppressive than mesenchymal stem cells or natural regulatory T cells. In addition, the cells expressing markers and characteristics of LSC had significantly higher levels of mRNA for Fas ligand and for the antiapoptotic molecules Mcl-1, XIAP, and survivin than other limbal cell populations. LSCs were also more resistant to staurosporin-induced apoptotic cell death and to cell-mediated cytotoxic reaction than other limbal cells. Collectively, these results suggest that SC isolated from fresh adult limbal tissue possess immunomodulatory properties and inhibit proinflammatory immune reactions. Simultaneously, these cells express high levels of mRNA for antiapoptotic molecules, which can protect them against cell-mediated cytotoxic reactions and various apoptosis-inducing agents.
Collapse
Affiliation(s)
- Vladimir Holan
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic.
| | | | | | | | | |
Collapse
|