1
|
Zhang Y, Fan M, Zhang Y. Revolutionizing bone defect healing: the power of mesenchymal stem cells as seeds. Front Bioeng Biotechnol 2024; 12:1421674. [PMID: 39497791 PMCID: PMC11532096 DOI: 10.3389/fbioe.2024.1421674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/10/2024] [Indexed: 11/07/2024] Open
Abstract
Bone defects can arise from trauma or pathological factors, resulting in compromised bone integrity and the loss or absence of bone tissue. As we are all aware, repairing bone defects is a core problem in bone tissue engineering. While minor bone defects can self-repair if the periosteum remains intact and normal osteogenesis occurs, significant defects or conditions such as congenital osteogenesis imperfecta present substantial challenges to self-healing. As research on mesenchymal stem cell (MSC) advances, new fields of application have emerged; however, their application in orthopedics remains one of the most established and clinically valuable directions. This review aims to provide a comprehensive overview of the research progress regarding MSCs in the treatment of diverse bone defects. MSCs, as multipotent stem cells, offer significant advantages due to their immunomodulatory properties and ability to undergo osteogenic differentiation. The review will encompass the characteristics of MSCs within the osteogenic microenvironment and summarize the research progress of MSCs in different types of bone defects, ranging from their fundamental characteristics and animal studies to clinical applications.
Collapse
Affiliation(s)
- Yueyao Zhang
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| | - Mengke Fan
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| | - Yingze Zhang
- Trauma Emergency Center, The Third Hospital of Hebei Medical University, Shijiazhuang, China
- Key Laboratory of Biomechanics of Hebei Province, Orthopaedic Research Institution of Hebei Province, Shijiazhuang, China
| |
Collapse
|
2
|
Nakao M, Matsui M, Kim K, Nishiyama N, Grainger DW, Okano T, Kanazawa H, Nagase K. Umbilical cord-derived mesenchymal stem cell sheets transplanted subcutaneously enhance cell retention and survival more than dissociated stem cell injections. Stem Cell Res Ther 2023; 14:352. [PMID: 38072920 PMCID: PMC10712142 DOI: 10.1186/s13287-023-03593-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 11/29/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Human umbilical cord-derived mesenchymal stem cell (hUC-MSC) sheets have recently attracted attention as an alternative approach to injected cell suspensions for stem cell therapy. However, cell engraftment and cytokine expression levels between hUC-MSC sheets and their cell suspensions in vivo have not yet been compared. This study compares hUC-MSC in vivo engraftment efficacy and cytokine expression for both hUC-MSC sheets and cell suspensions. METHODS hUC-MSC sheets were prepared using temperature-responsive cell culture; two types of hUC-MSC suspensions were prepared, either by enzymatic treatment (trypsin) or by enzyme-free temperature reduction using temperature-responsive cell cultureware. hUC-MSC sheets and suspensions were transplanted subcutaneously into ICR mice through subcutaneous surgical placement and intravenous injection, respectively. hUC-MSC sheet engraftment after subcutaneous surgical transplantation was investigated by in vivo imaging while intravenously injected cell suspensions were analyzing using in vitro organ imaging. Cytokine levels in both transplant site tissues and blood were quantified by enzyme-linked immunosorbent assay. RESULTS After subcutaneous transplant, hUC-MSC sheets exhibited longer engraftment duration than hUC-MSC suspensions. This was attributed to extracellular matrix (ECM) and cell-cell junctions retained in sheets but enzymatically altered in suspensions. hUC-MSC suspensions harvested using enzyme-free temperature reduction exhibited relatively long engraftment duration after intravenous injection compared to suspensions prepared using trypsin, as enzyme-free harvest preserved cellular ECM. High HGF and TGF-β1 levels were observed in sheet-transplanted sites compared to hUC-MSC suspension sites. However, no differences in human cytokine levels in murine blood were detected, indicating that hUC-MSC sheets might exert local paracrine rather than endocrine effects. CONCLUSIONS hUC-MSC sheet transplantation could be a more effective cell therapeutic approach due to enhanced engraftment and secretion of therapeutic cytokines over injected hUC-MSC suspensions.
Collapse
Affiliation(s)
- Mitsuyoshi Nakao
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8503, Japan
| | - Kyungsook Kim
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Health Sciences, Salt Lake City, UT, 84112, USA
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa, 226-8503, Japan
| | - David W Grainger
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Health Sciences, Salt Lake City, UT, 84112, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Teruo Okano
- Cell Sheet Tissue Engineering Center (CSTEC), Department of Molecular Pharmaceutics, University of Utah, Health Sciences, Salt Lake City, UT, 84112, USA
| | - Hideko Kanazawa
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan
| | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato-ku, Tokyo, 105-8512, Japan.
| |
Collapse
|
3
|
Banimohamad-Shotorbani B, Karkan SF, Rahbarghazi R, Mehdipour A, Jarolmasjed S, Saghati S, Shafaei H. Application of mesenchymal stem cell sheet for regeneration of craniomaxillofacial bone defects. Stem Cell Res Ther 2023; 14:68. [PMID: 37024981 PMCID: PMC10080954 DOI: 10.1186/s13287-023-03309-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
Bone defects are among the most common damages in human medicine. Due to limitations and challenges in the area of bone healing, the research field has turned into a hot topic discipline with direct clinical outcomes. Among several available modalities, scaffold-free cell sheet technology has opened novel avenues to yield efficient osteogenesis. It is suggested that the intact matrix secreted from cells can provide a unique microenvironment for the acceleration of osteoangiogenesis. To the best of our knowledge, cell sheet technology (CST) has been investigated in terms of several skeletal defects with promising outcomes. Here, we highlighted some recent advances associated with the application of CST for the recovery of craniomaxillofacial (CMF) in various preclinical settings. The regenerative properties of both single-layer and multilayer CST were assessed regarding fabrication methods and applications. It has been indicated that different forms of cell sheets are available for CMF engineering like those used for other hard tissues. By tackling current challenges, CST is touted as an effective and alternative therapeutic option for CMF bone regeneration.
Collapse
Affiliation(s)
- Behnaz Banimohamad-Shotorbani
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sonia Fathi Karkan
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Mehdipour
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyedhosein Jarolmasjed
- Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Sepideh Saghati
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Shafaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Li Y, Chen W, Deng H, Li T, Liu Z, Liu X, Zhang Z, Chen X, Sheng J, Li K. TGF-β1 Protects Trauma-injured Murine Cortical Neurons by Upregulating L-type Calcium Channel Ca v1.2 via the p38 Pathway. Neuroscience 2022; 492:47-57. [PMID: 35460836 DOI: 10.1016/j.neuroscience.2022.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/09/2022] [Accepted: 04/12/2022] [Indexed: 02/05/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of disability and death in adolescents, and there is a lack of effective methods of treatment. The neuroprotective effects exerted by TGF-β1 can ameliorate a range of neuronal lesions in multiple central nervous system diseases. In this study, we used an in-vitro TBI model of mechanical injury on murine primary cortical neurons and the neuro-2a cell line to investigate the neuroprotective role played by TGF-β1 in cortical neurons in TBI. Our results showed that TGF-β1 significantly increased neuronal viability and inhibited apoptosis for 24 h after trauma. The expression of Cav1.2, an L-type calcium channel (LTCC) isoform, decreased significantly after trauma injury, and this change was reversed by TGF-β1. Nimodipine, a classic LTCC blocker, abolished the protective effect of TGF-β1 on trauma-induced neuronal apoptosis. The knockdown of Cav1.2 in differentiated neuro-2a cells significantly inhibited the anti-apoptosis effect of TGF-β1 exerted on injured neuro-2a cells. Moreover, TGF-β1 rescued and enhanced the trauma-suppressed neuro-2a intracellular Ca2+ concentration, while the effect of TGF-β1 was partially inhibited by nimodipine. TGF-β1 significantly upregulated the expression of Cav1.2 by activating the p38 MAPK pathway and by inhibiting trauma-induced neuronal apoptosis. In conclusion, TGF-β1 increased trauma-injured murine cortical neuronal activity and inhibited apoptosis by upregulating Cav1.2 channels via activating the p38 MAPK pathway. Therefore, the TGF-β1/p38 MAPK/Cav 1.2 pathway has the potential to be used as a novel therapeutic target for TBI.
Collapse
Affiliation(s)
- Yanlei Li
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Weiqiang Chen
- Department of Neurosurgery, First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Huixiong Deng
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Tian Li
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Zhenning Liu
- Department of Laboratory, Guangzhou Chest Hospital, China
| | - Xueer Liu
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Zelin Zhang
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Xiaoxuan Chen
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Jiangtao Sheng
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| | - Kangsheng Li
- Department of Microbiology and Immunology, Provincial Key Laboratory of Infectious Diseases and Molecular Immunopathology, Shantou University Medical College, Shantou, China
| |
Collapse
|
5
|
Zhu J, Inomata T, Nakamura M, Fujimoto K, Akasaki Y, Fujio K, Yanagawa A, Uchida K, Sung J, Negishi N, Nagino K, Okumura Y, Miura M, Shokirova H, Kuwahara M, Hirosawa K, Midorikawa-Inomata A, Eguchi A, Huang T, Yagita H, Habu S, Okumura K, Murakami A. Anti-CD80/86 antibodies inhibit inflammatory reaction and improve graft survival in a high-risk murine corneal transplantation rejection model. Sci Rep 2022; 12:4853. [PMID: 35318419 PMCID: PMC8941080 DOI: 10.1038/s41598-022-08949-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/15/2022] [Indexed: 12/27/2022] Open
Abstract
We investigated the effects of anti-CD80/86 antibodies in a murine high-risk corneal transplantation rejection model. A mixed lymphocyte reaction (MLR) assay was conducted with anti-CD80/86 antibodies. Inflammatory cytokine levels in the culture supernatant were measured using an enzyme-linked immunosorbent assay. Interferon (IFN)-γ-producing CD4+ T cell frequencies in the MLR were assessed using flow cytometry. In vivo, high-risk corneal allograft survival and IFN-γ-producing CD4+ T cell frequencies in corneal grafts were assessed with intraperitoneal injection of anti-CD80/86 antibodies compared to phosphate-buffered saline (PBS). RNA-sequencing was performed on corneal grafts 2 weeks post-transplantation. Anti-CD80/86 antibodies significantly decreased T-cell proliferation, IFN-γ+-producing CD4+ T cell frequencies, and IFN-γ, interleukin (IL)-1β, IL-2, IL-10, and tumor necrosis factor-α production in the MLR compared to PBS injection. Intraperitoneal injection of anti-CD80/86 antibodies significantly prolonged corneal graft survival and decreased IFN-γ+-producing CD4+ T cell frequencies compared to PBS injection. Gene set enrichment analysis showed that the gene sets mainly enriched in the control group were related to allograft rejection and inflammatory response compared to PBS injection. Anti-CD80/86 antibodies significantly prolonged corneal graft survival by inhibiting T-cell proliferation and inflammatory response.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Ophthalmology, Subei People's Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan. .,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| | - Masahiro Nakamura
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Precision Health, Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Keiichi Fujimoto
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Yasutsugu Akasaki
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kenta Fujio
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ai Yanagawa
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koichiro Uchida
- Center for Immune Therapeutics and Diagnosis, Juntendo University, Tokyo, Japan
| | - Jaemyoung Sung
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Naoko Negishi
- Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Indoor Environment Neurophysiological Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Nagino
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuichi Okumura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Maria Miura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hurramhon Shokirova
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Mizu Kuwahara
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kunihiko Hirosawa
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akie Midorikawa-Inomata
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsuko Eguchi
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tianxiang Huang
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideo Yagita
- Department of Immunology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Sonoko Habu
- Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Center for Immune Therapeutics and Diagnosis, Juntendo University, Tokyo, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
6
|
da Silva AVA, de Souza TL, Figueiredo FB, Mendes AAV, Ferreira LC, Filgueira CPB, Cuervo P, Porrozzi R, Menezes RC, Morgado FN. Detection of amastigotes and histopathological alterations in the thymus of Leishmania infantum-infected dogs. IMMUNITY INFLAMMATION AND DISEASE 2020; 8:127-139. [PMID: 32207879 PMCID: PMC7212199 DOI: 10.1002/iid3.285] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/22/2019] [Accepted: 12/27/2019] [Indexed: 11/21/2022]
Abstract
Introduction In canine visceral leishmaniasis (CVL), lymphopenia, and the disorganization of lymphoid organs such as spleen and lymph nodes have been demonstrated. However, the involvement of thymus in CVL has not been evaluated so far. Herein, we investigated whether the thymus can be colonized by Leishmania infantum in naturally infected dogs. Methods Thymus were obtained from 16 of 58 dogs and samples of this organ were submitted to immunohistochemistry for laminin and fibronectin detection, histopathology, in situ hybridization and polymerase chain reaction (PCR) targeting the gene ITS‐1 for Leishmania and sequenced. Samples of spleen, skin and popliteal lymph nodes were collected and submitted to immunohistochemistry and parasitological culture followed by multilocus enzyme electrophoresis. Results L. infantum was identified in all dogs. DNA and amastigote forms of Leishmania were detected in the thymus from 16 dogs by PCR and in eight by immunohistochemistry. Besides thymus, parasites were detected in spleen, lymph nodes, and skin. A granulomatous or pyogranulomatous thymitis was observed in eight dogs associated to intact amastigotes forms of this parasite. Fibronectin deposition in thymus was higher in dogs with more clinical signs. Conclusions These results demonstrate that the thymus of dogs can be parasitized by L. infantum, which may generate inflammatory reactions leading to alterations in thymic microarchitecture.
Collapse
Affiliation(s)
- Aurea V A da Silva
- Laboratório de Pesquisa em Leishmanioses, IOC/FIOCRUZ, Rio de Janeiro, Brasil
| | - Tainã L de Souza
- Laboratório de Pesquisa em Leishmanioses, IOC/FIOCRUZ, Rio de Janeiro, Brasil
| | - Fabiano B Figueiredo
- Laboratório de Biologia Celular, Instituto Carlos Chagas, Fundação Oswaldo Cruz, Curitiba, Paraná, Brasil
| | - Artur A V Mendes
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, INI/FIOCRUZ, Rio de Janeiro, Brasil
| | - Luiz C Ferreira
- Serviço de Anatomia Patológica, INI/FIOCRUZ, Rio de Janeiro, Brasil
| | | | - Patricia Cuervo
- Laboratório de Pesquisa em Leishmanioses, IOC/FIOCRUZ, Rio de Janeiro, Brasil
| | - Renato Porrozzi
- Laboratório de Pesquisa em Leishmanioses, IOC/FIOCRUZ, Rio de Janeiro, Brasil
| | - Rodrigo C Menezes
- Laboratório de Pesquisa Clínica em Dermatozoonoses em Animais Domésticos, INI/FIOCRUZ, Rio de Janeiro, Brasil
| | - Fernanda N Morgado
- Laboratório de Pesquisa em Leishmanioses, IOC/FIOCRUZ, Rio de Janeiro, Brasil
| |
Collapse
|
7
|
CD163 and CCR7 as markers for macrophage polarization in lung cancer microenvironment. Cent Eur J Immunol 2020; 44:395-402. [PMID: 32140052 PMCID: PMC7050058 DOI: 10.5114/ceji.2019.92795] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/01/2019] [Indexed: 12/23/2022] Open
Abstract
Introduction M2 macrophages are predominant in the immune infiltrates of resected tumours, but little is known about macrophage phenotype in the local lung cancer environment, which may be evaluated by bronchoalveolar lavage fluid (BALF). Aim of the study To find differences between BALF from lung affected by cancer (clBALF) and hlBALF from the opposite, healthy lung, as a control, from the same patient, regarding their individual macrophage polarization and their correlation with IL-10 and TGF-β. Material and methods Eighteen patients with confirmed lung cancer were investigated. Macrophage subtyping was performed by immunofluorescence with antibodies anti-CCR7 and CD163 (M1 and M2, respectively). Results We found five populations of macrophages: cells with a single reaction: only for CCR7+ or CD163+, a double reaction (CCR7+CD163+), cells with a stronger CD163 (CCR7lowCD163+), and cells with a stronger CCR7 (CCR7+CD163low). The main population in the clBALF was composed of cells with a phenotype similar to M2 (CCR7lowCD163+), while in the hlBALF the predominating phenotype was the one similar to M1 (CCR7+CD163low). The median proportion of TGF-β1 concentration was higher in the clBALF and hlBALF supernatant than in the serum. Conclusions In this study we confirmed the usefulness of the immunofluorescence method with CCR7 and CD163 in the evaluation of BALF macrophage polarization in lung cancer.
Collapse
|
8
|
Nakao M, Inanaga D, Nagase K, Kanazawa H. Characteristic differences of cell sheets composed of mesenchymal stem cells with different tissue origins. Regen Ther 2019; 11:34-40. [PMID: 31193157 PMCID: PMC6517796 DOI: 10.1016/j.reth.2019.01.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 12/28/2018] [Accepted: 01/06/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Stem cell therapy with mesenchymal stem cells (MSCs) has been widely used in many clinical trials, and therapy with MSC sheets shows promise for patients. However, there are few reports characterizing MSC sheets. In the present study, the properties of MSC sheets derived from bone marrow, adipose tissue, and umbilical cord were evaluated. METHODS Cell sheets were fabricated with MSCs from different tissue origins in temperature-responsive cell culture dishes with and without pre-coating of fetal bovine serum (FBS). MSC adhesion behavior in the culture dish was observed. Secretion of cytokines related to cell proliferation and immune regulation from MSC sheets was investigated by ELISA. The adhesion properties of the MSC sheets were investigated by time-lapse microscopy. RESULTS Different cell adhesion and proliferation rates in temperature-responsive cell culture dishes were observed among the three types of MSCs. FBS pre-coating of the dishes enhanced cell attachment and proliferation in all cell types. Harvested cell sheets showed high attachment capacity to tissue culture polystyrene dish surfaces. CONCLUSIONS MSC sheets can be fabricated from MSCs from different tissue origins using temperature-responsive cell culture dishes. The fabricated MSC sheets could be useful in cell transplantation therapies by choosing appropriate types of MSCs that secrete therapeutic cytokines for the targeted diseases.
Collapse
Affiliation(s)
| | | | - Kenichi Nagase
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo, 105-8512, Japan
| | - Hideko Kanazawa
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo, 105-8512, Japan
| |
Collapse
|
9
|
Lange-Consiglio A, Stucchi L, Zucca E, Lavoie JP, Cremonesi F, Ferrucci F. Insights into animal models for cell-based therapies in translational studies of lung diseases: Is the horse with naturally occurring asthma the right choice? Cytotherapy 2019; 21:525-534. [PMID: 30929991 DOI: 10.1016/j.jcyt.2019.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 02/21/2019] [Accepted: 02/25/2019] [Indexed: 12/15/2022]
Abstract
Human asthma is a widespread disease associated with chronic inflammation of the airways, leading to loss of quality of life, disability and death. Corticosteroid administration is the mainstream treatment for asthmatic patients. Corticosteroids reduce airway obstruction and improve quality of life, although symptoms persist despite treatment in many patients. Moreover, available therapies failed to reverse the lung pathology present in asthma. Animal models, mostly rats and mice, in which the disease is experimentally induced, have been studied to identify new therapeutic targets for human asthma. Alternative animal models could include horses in which naturally occurring asthma could represent an important step to test therapies, potentially designed around mouse studies, before being translated to human testing. Horses naturally suffer from asthma, which has striking parallels with human asthma. Severe equine asthma (SEA) is characterized by reversible bronchospasms and neutrophil accumulation in the lungs immunologically mediated mainly by Th2. Moreover, the pulmonary remodelling that occurs in SEA closely resembles that of human asthma, making the equine model unique for investigation of tissue repair and new therapies. Cell therapy, consisting on mesenchymal stromal cells (MSCs) and derivatives (conditioned medium and extracellular vesicles), could represent a novel therapeutic contribution for tissue regeneration. Cell therapy may prove advantageous over conventional therapy in that it may repair or regenerate the site of injury and reduce the reaction to allergens, rather than simply modulating the inflammatory process.
Collapse
Affiliation(s)
- Anna Lange-Consiglio
- Department of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy; Reproduction Unit, Centro Clinico-Veterinario e Zootecnico Sperimentale di Ateneo, Università degli Studi di Milano, Lodi, Italy.
| | - Luca Stucchi
- Equine Medicine Unit, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| | - Enrica Zucca
- Equine Medicine Unit, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| | - Jean Pierre Lavoie
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Université de Montréal, St-Hyacinthe, QC, Canada
| | - Fausto Cremonesi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milano, Italy; Reproduction Unit, Centro Clinico-Veterinario e Zootecnico Sperimentale di Ateneo, Università degli Studi di Milano, Lodi, Italy
| | - Francesco Ferrucci
- Equine Medicine Unit, Department of Health, Animal Science and Food Safety, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
10
|
Antifibrosis effects of Shenge Yangfei Capsules on bleomycin-induced pulmonary fibrosis in rats. CHINESE HERBAL MEDICINES 2018. [DOI: 10.1016/j.chmed.2018.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
11
|
Identification of plasma interleukins as biomarkers for deflazacort and omega-3 based Duchenne muscular dystrophy therapy. Cytokine 2018; 102:55-61. [DOI: 10.1016/j.cyto.2017.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023]
|
12
|
Askari FK, Dick R, Mao M, Brewer GJ. Tetrathiomolybdate Therapy Protects Against Concanavalin a and Carbon Tetrachloride Hepatic Damage in Mice. Exp Biol Med (Maywood) 2016; 229:857-63. [PMID: 15337842 DOI: 10.1177/153537020422900820] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Tetrathiomolybdate, an anticopper drug, has been shown to protect mice against pulmonary fibrosis from bleomycin. Our hypothesis is that it does so by inhibiting fibrosis-inducing cytokines. Indeed, we have good evidence, not yet published, that tetrathiomolybdate inhibits pulmonary levels of transforming growth factor–β and tumor necrosis factor-α expression in these bleomycin experiments. Herein, we evaluate tetrathiomolybdate's effectiveness in mitigating hepatitis and fibrosis in mice from the hepatotoxins, concanavalin A and carbon tetrachloride, and its inhibition of cytokines as a possible mechanism. In short-term experiments, concanavalin A elevated serum amino leucine transferase levels several fold, and tetrathiomolybdate completely prevented this increase. In additional experiments, tetrathiomolybdate therapy reversed the elevated serum transaminase levels despite continued concanavalin A injections, with nearly significant serum interleukin-1β inhibition. Concanavalin A given for 12 weeks produced mild fibrosis, whereas concomitant tetrathiomolybdate treatment resulted in normal histology. Carbon tetrachloride given for 12 weeks resulted in very high serum amino leucine transferase levels, high serum transforming growth factor–β levels, cirrhosis as seen histologically, and increase in liver hydroxyproline, a measure of fibrosis. Concomitant tetrathiomolybdate partially and significantly protected against increases in amino leucine transferase and transforming growth factor–β, fully protected against the increase in hydroxyproline, and resulted in normal histology. In conclusion, tetrathiomolybdate protects against the hepatitis and fibrosis produced by these hepatotoxins, probably by inhibiting the excessive increase in inflammatory and fibrotic cytokines.
Collapse
Affiliation(s)
- Fred K Askari
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | | | |
Collapse
|
13
|
Zucca E, Corsini E, Galbiati V, Lange-Consiglio A, Ferrucci F. Evaluation of amniotic mesenchymal cell derivatives on cytokine production in equine alveolar macrophages: an in vitro approach to lung inflammation. Stem Cell Res Ther 2016; 7:137. [PMID: 27651133 PMCID: PMC5028987 DOI: 10.1186/s13287-016-0398-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 01/01/2023] Open
Abstract
Background Data obtained in both animal models and clinical trials suggest that cell-based therapies represent a potential therapeutic strategy for lung repair and remodeling. Recently, new therapeutic approaches based on the use of stem cell derivatives (e.g., conditioned medium (CM) and microvesicles (MVs)) to regenerate tissues and improve their functions were proposed. The aim of this study was to investigate the immunomodulatory effects of equine amniotic mesenchymal cell derivatives on lipopolysaccharide (LPS)-induced cytokine production in equine alveolar macrophages, which may be beneficial in lung inflammatory disorders such as recurrent airway obstruction (RAO) in horses. RAO shares many features with human asthma, including an increased number of cells expressing mRNA for interleukin (IL)-4 and IL-5 and a decreased expression of IFN-γ in bronchoalveolar lavage fluid (BALF) of affected horses. Methods The release of TNF-α, IL-6, and TGF-β1 at different time points (1, 24, 48, and 72 h) was measured in equine alveolar macrophages stimulated or not with LPS (10 and 100 ng/mL) in the presence or absence of 10 % CM or 50 × 106 MVs/mL. Cytokines were measured using commercially available ELISA kits. For multiple comparisons, analysis of variance was used with Tukey post-hoc test. Differences were considered significant at p ≤ 0.05. Results Significant modulatory effects of CM on LPS-induced TNF-α release at 24 h, and of both CM and MVs on TNF-α release at 48 h were observed. A trend toward a modulatory effect of both CM and MVs on the release of TGF-β and possibly IL-6 was visible over time. Conclusions Results support the potential use of CM and MVs in lung regenerative medicine, especially in situations in which TGF-β may be detrimental, such as respiratory allergy. Further studies should evaluate the potential clinical applications of CM and MVs in equine lung diseases, such as RAO and other inflammatory disorders.
Collapse
Affiliation(s)
- Enrica Zucca
- Department of Health, Animal Science and Food Safety (VESPA), Università degli Studi di Milano, Via Celoria 10, 20133, Milano, Italy
| | - Emanuela Corsini
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Valentina Galbiati
- Laboratory of Toxicology, DiSFeB, Università degli Studi di Milano, Milan, Italy
| | - Anna Lange-Consiglio
- Large Animal Hospital, Reproduction Unit, Università degli Studi di Milano, Lodi, Italy.
| | - Francesco Ferrucci
- Department of Health, Animal Science and Food Safety (VESPA), Università degli Studi di Milano, Via Celoria 10, 20133, Milano, Italy
| |
Collapse
|
14
|
Shahzad M, Small DM, Morais C, Wojcikowski K, Shabbir A, Gobe GC. Protection against oxidative stress-induced apoptosis in kidney epithelium by Angelica and Astragalus. JOURNAL OF ETHNOPHARMACOLOGY 2016; 179:412-419. [PMID: 26719285 DOI: 10.1016/j.jep.2015.12.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 12/06/2015] [Accepted: 12/20/2015] [Indexed: 05/21/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Astragalus membranaceus either alone or in combination with Angelica sinensis has been used traditionally for kidney disease in East Asia and China for thousands of years. Previous studies using in vivo animal models have shown the benefits of these medicinal herbs in kidney diseases that involve oxidative stress. However, the mechanisms by which these medicinal herbs protect kidney cells remain largely unknown. AIM OF THE STUDY To investigate the mechanisms by which ethanol, methanol and aqueous crude extracts of roots of A. membranaceus and A. sinensis afford protection to human kidney proximal tubular epithelial cells, using an in vitro model of oxidative stress. MATERIALS AND METHODS Ethanol, methanol and aqueous extracts of roots of A. membranaceus and A. sinensis were prepared by a three-solvent sequential process. HK2 human kidney proximal tubular epithelial cells were treated with H2O2 alone (0.5mM) or in combination with different concentrations of extracts. Cell mitosis and death (microscopy) and cell viability (MTT assay) were compared. Western immunoblot was used to study expression of apoptosis-related proteins (pro-apoptotic Bax andanti-apoptotic Bcl-XL), and cell survival (NFκB subunits p65 and p50), pro-inflammatory (TNF-α) and protective (TGFβ1) proteins. RESULTS H2O2-induced oxidative stress significantly increased apoptosis and reduced cell survival; upregulated pro-apoptotic and down-regulated Bcl-XL; increased NFκB (p65, p50); increased TNFα and decreased TGFβ1. All changes indicated kidney damage and dysfunction. All were modulated by all extracts of both plant species, except for NFκB which was only modulated by extracts of A. membranaceus. CONCLUSIONS In conclusion, in a model of oxidative stress that might occur after nephrotoxicity, the plant extracts were protective via anti-apoptotic and anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Muhammad Shahzad
- Centre for Kidney Disease Research, School of Medicine, University of Queensland, Translational Research Institute, Brisbane, Australia; Department of Pharmacology, University of Health Sciences, Lahore, Pakistan
| | - David M Small
- Centre for Kidney Disease Research, School of Medicine, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Christudas Morais
- Centre for Kidney Disease Research, School of Medicine, University of Queensland, Translational Research Institute, Brisbane, Australia
| | - Ken Wojcikowski
- Department of Natural and Complementary Medicine, Southern Cross University, Lismore, Australia
| | - Arham Shabbir
- Department of Pharmacology, University of Health Sciences, Lahore, Pakistan
| | - Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, University of Queensland, Translational Research Institute, Brisbane, Australia.
| |
Collapse
|
15
|
Chen W, Konkel JE. Development of thymic Foxp3(+) regulatory T cells: TGF-β matters. Eur J Immunol 2015; 45:958-65. [PMID: 25684698 DOI: 10.1002/eji.201444999] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 01/22/2023]
Abstract
CD4(+) regulatory T cells expressing the transcription factor Foxp3 can be generated in the thymus (tTreg cells), but the cellular and molecular pathways driving their development remain incompletely understood. TGF-β is essential for the generation of Foxp3(+) Treg cells converted from peripheral naïve CD4(+) T cells (pTreg cells), yet a role for TGF-β in tTreg-cell development was initially refuted. Nevertheless, recent studies have unmasked a requirement for TGF-β in the generation of tTreg cells. Experimental evidence reveals that TGF-β in the context of TCR stimulation induces Foxp3 gene transcription in thymic Treg precursors, CD4(+) CD8(-) CD25(-) semimature and mature single-positive thymocytes. Intriguingly, thymic apoptosis was found to be intrinsically linked to the generation of tTreg cells, as apoptosis induced expression of TGF-β intrathymically. In this short review, we will highlight key data, discuss the experimental evidence and propose a modified model of tTreg-cell development involving TGF-β. We will also outline the remaining unresolved questions concerning generation of thymic Foxp3(+) Treg cells and provide our personal perspectives on the mechanisms controlling tTreg-cell development.
Collapse
Affiliation(s)
- WanJun Chen
- Mucosal Immunology Section, OPCB, NIDCR, 30 Convent Dr., Bethesda, MD, USA
| | | |
Collapse
|
16
|
Abstract
INTRODUCTION Immune checkpoint inhibitors, such as ipilimumab , are a new class of immunotherapeutic agents that have shown significant efficacy in melanoma. A number of ongoing clinical trials are investigating the role of ipilimumab in prostate cancer, either alone or in combination with immunomodulating agents such as radiation and chemotherapy, and in combination with cancer vaccines. AREAS COVERED This article reviews the molecular basis, preclinical and clinical evidence on the safety and efficacy of ipilimumab in prostate cancer. Medical literature search using MEDLINE and online abstracts database of national meetings form the basis of this article. EXPERT OPINION A number of preliminary clinical studies suggest the potential therapeutic utility of immune checkpoint inhibitors such as ipilimumab in prostate cancer. Pending the results of large-scale studies, the rationale of combining ipilimumab with standard anticancer therapeutics such as radiation, cytotoxic chemotherapy and other immunotherapeutic agents can be of great value in reducing mortality and morbidity in prostate cancer.
Collapse
Affiliation(s)
- Nishith Singh
- National Cancer Institute, National Institutes of Health, Center for Cancer Research, Medical Oncology Branch, Laboratory of Tumor Immunology and Biology, Bethesda, MD, USA
| | - Ravi A Madan
- National Cancer Institute, National Institutes of Health, Center for Cancer Research, Medical Oncology Branch, Laboratory of Tumor Immunology and Biology, Bethesda, MD, USA
| | - James L Gulley
- National Cancer Institute, National Institutes of Health, Center for Cancer Research, Medical Oncology Branch, Laboratory of Tumor Immunology and Biology, Bethesda, MD, USA
| |
Collapse
|
17
|
Yee M, Buczynski BW, Lawrence BP, O'Reilly MA. Neonatal hyperoxia increases sensitivity of adult mice to bleomycin-induced lung fibrosis. Am J Respir Cell Mol Biol 2012; 48:258-66. [PMID: 23258231 DOI: 10.1165/rcmb.2012-0238oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Supplemental oxygen used to treat infants born prematurely constitutes a major risk factor for long-term deficits in lung function and host defense against respiratory infections. Likewise, neonatal oxygen exposure results in alveolar simplification in adult mice, and enhances leukocyte recruitment and fibrosis when adult mice are infected with a sublethal dose of influenza A virus. Because pulmonary fibrosis was not observed in infected adult mice exposed to room air as neonates, previous neonatal oxygen exposure may have reprogrammed how the adult lung responds to epithelial injury. By administering bleomycin to adult mice exposed to room air or hyperoxia as neonates, we tested the hypothesis that neonatal hyperoxia enhances fibrosis when the epithelium is injured by direct fibrotic stimulus. Increased sensitivity to bleomycin-induced lung fibrosis was observed in adult mice exposed to neonatal hyperoxia, and was associated with increased numbers of leukocytes and an accumulation of active transforming growth factor (TGF)-β1 in the lung. Fate mapping of the respiratory epithelium revealed that the epithelial-mesenchymal transition was not a significant source of fibroblasts in room air-exposed or oxygen-exposed mice treated with bleomycin. Instead, the treatment of mice with anti-Gr-1 antibody that depletes neutrophils and myeloid-derived suppressor cells reduced the early activation of TGF-β1 and attenuated hyperoxia-enhanced fibrosis. Because bleomycin and influenza A virus both cause epithelial injury, understanding how neonatal hyperoxia reprograms the epithelial response to these two different injurious agents could lead to new therapeutic opportunities for treating lung diseases attributed to prematurity.
Collapse
Affiliation(s)
- Min Yee
- Department of Pediatrics, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
18
|
Liu Y, Zhao H, Zhang J, Zhang P, Li M, Qi F, Wang Y, Kou S, Zheng Q, Wang L. The Regulatory Effect of Liuwei Dihuang Pills on Cytokines in Mice with Experimental Autoimmune Encephalomyelitis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2012; 40:295-308. [DOI: 10.1142/s0192415x12500231] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The regulatory effect of Liuwei Dihuang Pills (LDP) was studied on cytokines in mice with experimental autoimmune encephalomyelitis (EAE), a model for human multiple sclerosis (MS), induced by immunization with MOG35-55 and complete Freund's adjuvant (CFA) supplemented with pertussis toxin (PTX). LDP was administrated orally for 40 days, and prednisone acetate (PA) was used as a control. The pathological changes in the spinal cords of mice were observed by light microscope with hematoxylin-eosin (HE) staining and transmission electron microscope (TEM). The protein and mRNA expression of tumor necrosis factor-alpha (TNF-α) and transforming growth factor-beta (TGF-β) in the spinal cords were assessed by immunohistochemistry and RT-PCR assay, and the cyclic adenosine monophosphate (cAMP) in mice plasma was measured by radioimmunoassay (RIA) on days 12, 25 and 40 post-immunization (PI). The results showed that inflammatory cells, demyelination and axonal loss were reduced, and that the protein and mRNA expression of TNF-α and the ratio of TNF-α/TGF-β were obviously decreased, to different extents. However, the levels of cAMP were enhanced in LDP-treated groups. These findings suggested that LDP regulates the cytokine balance in favor of T helper 1 (Th1)/regulatory T (Treg) cells, which depend on enhancement of cAMP levels. LDP has a potential role in the treatment of MS and other demyelinating diseases of the central nervous system.
Collapse
Affiliation(s)
- Yan Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
- Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Jie Zhang
- School of Public Health and Family Medicine, Capital Medical University, Beijing 100069, China
| | - Ping Zhang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ming Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Fang Qi
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Yizhou Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Shuang Kou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Qi Zheng
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
19
|
Celis R, Planell N, Fernández-Sueiro JL, Sanmartí R, Ramírez J, González-Álvaro I, Pablos JL, Cañete JD. Synovial cytokine expression in psoriatic arthritis and associations with lymphoid neogenesis and clinical features. Arthritis Res Ther 2012; 14:R93. [PMID: 22541888 PMCID: PMC3446467 DOI: 10.1186/ar3817] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Revised: 03/02/2012] [Accepted: 04/27/2012] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Psoriatic arthritis (PsA) is an autoantibody-negative immune-mediated disease in which synovial lymphoid neogenesis (LN) occurs. We determined whether LN is associated with specific patterns of inflammatory cytokine expression in paired synovial tissue (ST) and fluid (SF) samples and their potential correlation with the clinical characteristics of PsA. METHODS ST and paired SF samples were obtained from the inflamed knee of PsA patients. ST samples were immunostained with CD3 (T cell), CD20 (B cell), and MECA-79 (high endothelial vessels). Total ST mRNA was extracted, and the gene expression of 21 T-cell-derived and proinflammatory cytokines were measured with quantitative real-time PCR. SF concentrations of Th1, Th2, Th17, and proinflammatory cytokines were determined with the Quantibody Human Th17 Array. Clinical and biologic data were collected at inclusion and after a median of 27 months of follow-up. RESULTS Twenty (43.5%) of 46 patients had LN. Only two genes showed differences (Wilcoxon test, P < 0.06) in ST between LN-positive and LN-negative patients: interleukin-23A (IL-23A) (P = 0.058) and transforming growth factor-beta (TGF-β1) (P = 0.050). IL-23A expression was higher, and TGF-β1 expression was lower in LN-positive patients. ST IL-15 mRNA showed a nonsignificant trend toward higher expression in LN-positive patients, and SF IL-15 protein levels were significantly higher in LN-positive patients (P = 0.002). In all PsA patients, IL-23A mRNA expression correlated with C-reactive protein (CRP) (r = 0.471; P = 0.001) and swollen-joint count (SJC) (r = 0.350; P = 0.018), whereas SF levels of IL-6 and CC chemokine-ligand 20 (CCL-20) correlated with CRP levels (r = 0.377; P = 0.014 and r = 0.501; P < 0.0001, respectively). CONCLUSIONS These findings suggest differences in the cytokine profile of PsA patients with LN, with a higher expression of IL-23A and IL-15 and a lower expression of TGF-β1. In the entire group of patients, IL-23 ST expression and CCL20 SF levels strongly correlated with markers of disease activity. This cytokine pattern was not accompanied by gross clinical or biologic differences between LN-positive and -negative patients. Taken together, these results suggest a role of the IL-17/IL-23 cytokine axis in synovial LN in PsA.
Collapse
Affiliation(s)
- Raquel Celis
- Arthritis Unit, Rheumatology Department, Hospital Clinic de Barcelona and IDIBAPS, c/Villarroel, 170, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Walker LSK, Sansom DM. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat Rev Immunol 2011; 11:852-63. [PMID: 22116087 DOI: 10.1038/nri3108] [Citation(s) in RCA: 568] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The T cell protein cytotoxic T lymphocyte antigen 4 (CTLA4) was identified as a crucial negative regulator of the immune system over 15 years ago, but its mechanisms of action are still under debate. It has long been suggested that CTLA4 transmits an inhibitory signal to the cells that express it. However, not all the available data fit with a cell-intrinsic function for CTLA4, and other studies have suggested that CTLA4 functions in a T cell-extrinsic manner. Here, we discuss the data for and against the T cell-intrinsic and -extrinsic functions of CTLA4.
Collapse
Affiliation(s)
- Lucy S K Walker
- MRC Centre for Immune Regulation, University of Birmingham Medical School, Birmingham, UK.
| | | |
Collapse
|
21
|
Gao J, Huang Y, Li P, Xu D, Li J, Liu Y, Huang Z, Wu Q, Shao X. Antifibrosis effects of total glucosides of Danggui-Buxue-Tang in a rat model of bleomycin-induced pulmonary fibrosis. JOURNAL OF ETHNOPHARMACOLOGY 2011; 136:21-26. [PMID: 21421041 DOI: 10.1016/j.jep.2011.03.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 02/28/2011] [Accepted: 03/03/2011] [Indexed: 05/30/2023]
Abstract
AIM OF THE STUDY The present study examined the antifibrosis effects of DBTG (total glucosides of Danggui-Buxue-Tang) on bleomycin-induced pulmonary injury and fibrosis in rats. MATERIALS AND METHODS Animals were randomly divided into six groups: (1) saline control group; (2) Bleomycin group in which rats were endotracheally instillated with bleomycin (5mg/kg); (3-5) Bleomycin and DBTG group, in which DBTG were given to rats daily (16.32 or 64mg/kg/day, i.g.) one day after bleomycin injection for 4 weeks until the end of the treatment; (6) Bleomycin and positive control group. Animals were sacrificed at 7, 14, and 28 days post bleomycin administration and lungs were removed. Lung specimens were stained with hematoxylin and eosin (HE) and Masson trichrome for histological evaluation of lung injury and fibrosis by light microscopy. Body weight and lung index from various groups were measured, as well as TNF-α, TGF-β1 and type I collagen concentrations in lung homogenates. RESULTS DBTG reduced bleomycin-induced weight loss, decreased the lung index and histological evidence supported the ability of DBTG to attenuate bleomycin-induced lung fibrosis and consolidation. DBTG could partly dose-dependently decrease TNF-α and TGF-β1 activity, as well as it decrease type I collagen expression in lung tissues. CONCLUSIONS The findings of the present study provide evidence that DBTG may serve as a novel target for potential therapeutic treatment of lung fibrosis.
Collapse
Affiliation(s)
- Jian Gao
- Pharmaceutical preparation section, Third-Grade Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine (TCM-2009-202), the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Bai L, Yu Z, Wang C, Qian G, Wang G. Dual role of TGF-β1 on Fas-induced apoptosis in lung epithelial cells. Respir Physiol Neurobiol 2011; 177:241-6. [PMID: 21539941 DOI: 10.1016/j.resp.2011.04.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 04/15/2011] [Accepted: 04/17/2011] [Indexed: 10/18/2022]
Abstract
Recent evidence suggests that TGF-β1 has a dual role in regulating cell response to Fas/Fas ligand (FasL)-induced apoptosis. TGF-β1 may play a positive or negative role on cell sensitivity to apoptosis via Fas/FasL system, depending on cell types and their specific environment. TGF-β1 and the Fas/FasL system are also involved in pathological processes of acute lung injury (ALI) and interstitial lung diseases including early lung injury and subsequent tissue repair. However, it is not well understood how TGF-β1 regulates Fas/FasL mediated apoptotic signaling in lung epithelium. In this study, we found that TGF-β1 could affect the sensitivity of lung epithelial A549 cells to Fas/FasL mediated apoptosis in a time-dependent manner. Apoptosis of A549 cells could be enhanced significantly by co-treatment with TGF-β1 and FasL, or pretreatment with TGF-β1 followed by FasL exposure, as evidenced by markedly increased caspase-8 and JNK activities. However, prolonged exposure to TGF-β1 could result in an obvious inhibition of the Fas/FasL-induced apoptosis, accompanied by down-regulation of Fas and up-regulation of c-Flip. Our results also showed that the effect of TGF-β1 on cell sensitivity to Fas-mediated apoptosis was independent of Akt pathway activation. These findings suggest that timely interplay of TGF-β1 and the Fas/FasL system could determine the final outcomes of cell survival/death signaling, for example, switching cell death signaling to survival signaling during early injury and later repair process of lung epithelium.
Collapse
Affiliation(s)
- Li Bai
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, PR China.
| | | | | | | | | |
Collapse
|
23
|
Carlier VA, Vanderelst LP, Jacquemin MG, Saint-Remy JMR. Regulatory CD4+ T cells in allergic asthma. Expert Rev Clin Immunol 2010; 2:737-44. [PMID: 20477629 DOI: 10.1586/1744666x.2.5.737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Active suppression by regulatory T cells (T(regs)) appears to play a key role in the downregulation of T-cell responses to foreign antigens. Several subtypes of T(regs) have been described but their mechanisms of action remain unclear. Recent data demonstrate that the suppressive capacity of natural T(regs) could be associated with cytotoxicity due to the release of granzymes, which are capable of apoptosis induction in target effector T lymphocytes and in antigen-presenting cells, such as dendritic cells. The mechanism of such nonspecific T(regs) is discussed. Peptide immunotherapy is thought to induce regulatory cells capable of suppressing autoimmune and allergic diseases. We have recently optimized a vaccination strategy by which cytotoxic antigen-specific adaptive T(regs) can be elicited towards allergens involved in allergic asthma. Such a strategy could be of value in the treatment of allergic asthma.
Collapse
Affiliation(s)
- Vincent A Carlier
- University of Leuven, Center for Molecular and Vascular Biology, Campus Gasthuisberg, O&N, Herestraat 49, 3000 Leuven, Belgium.
| | | | | | | |
Collapse
|
24
|
Das K, Siebert U, Gillet A, Dupont A, Di-Poï C, Fonfara S, Mazzucchelli G, De Pauw E, De Pauw-Gillet MC. Mercury immune toxicity in harbour seals: links to in vitro toxicity. Environ Health 2008; 7:52. [PMID: 18959786 PMCID: PMC2600635 DOI: 10.1186/1476-069x-7-52] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2008] [Accepted: 10/29/2008] [Indexed: 05/20/2023]
Abstract
BACKGROUND Mercury is known to bioaccumulate and to magnify in marine mammals, which is a cause of great concern in terms of their general health. In particular, the immune system is known to be susceptible to long-term mercury exposure. The aims of the present study were (1) to determine the mercury level in the blood of free-ranging harbour seals from the North Sea and (2) to examine the link between methylmercury in vitro exposure and immune functions using seal and human mitogen-stimulated peripheral blood mononuclear cells (T-lymphocytes). METHODS Total mercury was analysed in the blood of 22 harbour seals. Peripheral blood mononuclear cells were isolated from seals (n = 11) and from humans (n = 9). Stimulated lymphocytes of both species were exposed to functional tests (proliferation, metabolic activity, radioactive precursor incorporation) under increasing doses of methylmercury (0.1 to 10 microM). The expression of cytokines (IL-2, IL-4 and TGF-beta) was investigated in seal lymphocytes by RT-PCR and by real time quantitative PCR (n = 5) at methylmercury concentrations of 0.2 and 1 microM. Finally, proteomics analysis was attempted on human lymphocytes (cytoplasmic fraction) in order to identify biochemical pathways of toxicity at concentration of 1 microM (n = 3). RESULTS The results showed that the number of seal lymphocytes, viability, metabolic activity, DNA and RNA synthesis were reduced in vitro, suggesting deleterious effects of methylmercury concentrations naturally encountered in free-ranging seals. Similar results were found for human lymphocytes. Functional tests showed that a 1 microM concentration was the critical concentration above which lymphocyte activity, proliferation and survival were compromised. The expression of IL-2 and TGF-beta mRNA was weaker in exposed seal lymphocytes compared to control cells (0.2 and 1 microM). Proteomics showed some variation in the protein expression profile (e.g. vimentin). CONCLUSION Our results suggest that seal and human PBMCs react in a comparable way to MeHg in vitro exposure with, however, larger inter-individual variations. MeHg could be an additional cofactor in the immunosuppressive pollutant cocktail generally described in the blood of seals and this therefore raises the possibility of additional additive effects in the marine mammal immune system.
Collapse
Affiliation(s)
- Krishna Das
- Laboratoire d'Océanologie, Centre de Recherche MARE, B6C, Université de Liège, 4000, Liège, Belgium
| | - Ursula Siebert
- Research and Technology Center Westcoast, University of Kiel, 25761 Buesum, Germany
| | - Audrey Gillet
- Laboratoire d'Océanologie, Centre de Recherche MARE, B6C, Université de Liège, 4000, Liège, Belgium
| | - Aurélie Dupont
- Laboratoire d'Océanologie, Centre de Recherche MARE, B6C, Université de Liège, 4000, Liège, Belgium
| | - Carole Di-Poï
- Laboratoire d'Océanologie, Centre de Recherche MARE, B6C, Université de Liège, 4000, Liège, Belgium
| | - Sonja Fonfara
- Research and Technology Center Westcoast, University of Kiel, 25761 Buesum, Germany
- GKSS Research Centre, Institute for Coastal Research, 21502, Geesthacht, Germany
| | - Gabriel Mazzucchelli
- Laboratoire de Spectrométrie de Masse, B6C Liège, Université de Liège, 4000, Liège, Belgium
| | - Edwin De Pauw
- Laboratoire de Spectrométrie de Masse, B6C Liège, Université de Liège, 4000, Liège, Belgium
| | | |
Collapse
|
25
|
Abstract
Advances in understanding the role of transforming growth factor (TGF)-beta in tumorigenesis have led to the development of TGF-beta inhibitors for cancer treatment. Three platforms of TGF-beta inhibitors have evolved: antisense oligonucleotides, monoclonal antibodies and small molecules. In this review, the current stage of development of each known TGF-beta inhibitor will be discussed. As part of the risk/benefit assessment of TGF-beta inhibitors, the known effects of TGF-beta deficiency in mice, non-clinical toxicology studies with TGF-beta inhibitors in rats, and the clinical studies with monoclonal antibodies against TGF-beta will be summarised.
Collapse
Affiliation(s)
- Michael Lahn
- Eli Lilly & Company Research Laboratories, Therapeutic Area Oncology, Lilly Corporate Center, Indianapolis, Indiana 46285, USA.
| | | | | |
Collapse
|
26
|
Duenker N. Transforming growth factor-beta (TGF-beta) and programmed cell death in the vertebrate retina. ACTA ACUST UNITED AC 2006; 245:17-43. [PMID: 16125544 DOI: 10.1016/s0074-7696(05)45002-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Programmed cell death (PCD) is a precisely regulated phenomenon essential for the homeostasis of multicellular organisms. Developmental systems, particularly the nervous system, have provided key observations supporting the physiological role of PCD. We have recently shown that transforming growth factor-beta (TGF-beta) plays an important role in mediating ontogenetic PCD in the nervous system. As part of the central nervous system the developing retina serves as an ideal model system for investigating apoptotic processes during neurogenesis in vivo as it is easily accessible experimentally and less complex due to its limited number of different neurons. This review summarizes data indicating a pivotal role of TGF-beta in mediating PCD in the vertebrate retina. The following topics are discussed: expression of TGF-beta isoforms and receptors in the vertebrate retina, the TGF-beta signaling pathway, functions and molecular mechanisms of PCD in the nervous system, TGF-beta-mediated retinal apoptosis in vitro and in vivo, and interactions of TGF-beta with other pro- and anti-apoptotic factors.
Collapse
Affiliation(s)
- Nicole Duenker
- Institute for Anatomy, Department of Neuroanatomy, University of Duisburg-Essen, 45122 Essen, Germany
| |
Collapse
|
27
|
Zhou L, Porter JD, Cheng G, Gong B, Hatala DA, Merriam AP, Zhou X, Rafael JA, Kaminski HJ. Temporal and spatial mRNA expression patterns of TGF-beta1, 2, 3 and TbetaRI, II, III in skeletal muscles of mdx mice. Neuromuscul Disord 2005; 16:32-8. [PMID: 16373085 DOI: 10.1016/j.nmd.2005.09.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Revised: 08/02/2005] [Accepted: 09/14/2005] [Indexed: 11/19/2022]
Abstract
To address potential regulatory roles of TGF-beta1 in muscle inflammation and fibrosis associated with dystrophin deficiency, we performed quantitative RT-PCR and in situ hybridization to characterize the temporal and spatial mRNA expression patterns of TGF-beta1 and other TGF-beta subfamily members, TGF-beta2 and TGF-beta3, as well as their receptors, in quadriceps and diaphragm muscles of mdx mice. TGF-beta1 mRNA was markedly upregulated in the endomysial inflammatory cells and regenerating fibers of mdx quadriceps and diaphragm, with the mRNA levels correlated with the degree of endomysial inflammation. Upregulation of TGF-beta2, beta3, and their receptors was also appreciated but to a much lesser degree. While high levels of TGF-beta1 mRNA remained in the aging mdx quadriceps but not the diaphragm, progressive fibrosis only occurred in the diaphragm. Our data support a regulatory role for TGF-beta1 in muscle inflammation in mdx mice. It also suggests different susceptibility of quadriceps and diaphragm muscles to fibrosis induced by TGF-beta1 signaling pathway.
Collapse
Affiliation(s)
- Lan Zhou
- Department of Neurology, The Cleveland Clinic Foundation, 9500 Euclid Avenue/S90, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Thomas DA, Massagué J. TGF-beta directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 2005; 8:369-80. [PMID: 16286245 DOI: 10.1016/j.ccr.2005.10.012] [Citation(s) in RCA: 974] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2005] [Revised: 09/28/2005] [Accepted: 10/20/2005] [Indexed: 11/19/2022]
Abstract
Tumors escape from immune surveillance by producing the immunosuppressive cytokine TGF-beta. However, the mechanism by which TGF-beta inhibits T cell-mediated tumor clearance in vivo is unknown. We demonstrate that TGF-beta acts on cytotoxic T lymphocytes (CTLs) to specifically inhibit the expression of five cytolytic gene products-namely, perforin, granzyme A, granzyme B, Fas ligand, and interferon gamma-which are collectively responsible for CTL-mediated tumor cytotoxicity. Repression of granzyme B and interferon-gamma involves binding of TGF-beta-activated Smad and ATF1 transcription factors to their promoter regions, indicating direct and selective regulation by the TGF-beta/Smad pathway. Neutralization of systemic TGF-beta in mice enables tumor clearance with restoration of cytotoxic gene expression in antigen-specific CTLs in vivo. We suggest that TGF-beta suppresses CTL function in vivo through an anticytotoxic program of transcriptional repression.
Collapse
Affiliation(s)
- Dori A Thomas
- Cancer Biology and Genetics Program and Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | |
Collapse
|
29
|
Mansour H, Cheval L, Elalouf JM, Aude JC, Alyanakian MA, Mougenot B, Doucet A, Deschênes G. T-cell transcriptome analysis points up a thymic disorder in idiopathic nephrotic syndrome. Kidney Int 2005; 67:2168-77. [PMID: 15882260 DOI: 10.1111/j.1523-1755.2005.00322.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Idiopathic nephrotic syndrome is a proteinuric disease secondary to the release of a nonidentified circulating glomerular permeability factor by T cells. Because specificities of T-cell activation in idiopathic nephrotic syndrome remain unknown, we evaluated transcriptional activation of T cells in nephrotic patients during proteinuria. METHODS Transcriptomes of CD2+ cells were analyzed by serial analysis of gene expression (SAGE) in a nephrotic child during proteinuria relapse and after remission, away from any immunosuppressive treatment. Expression of specific transcripts overexpressed during proteinuria relapse was compared by reverse transcription-polymerase chain reaction (RT-PCR) in CD2+ cells from 11 nephrotic patients during relapse and remission and 11 non nephrotic patients during infection and after recovery. RESULTS Differential analysis of CD2+ cell transcriptome identified >200 mRNA tags overexpressed during proteinuria relapse, including many T-cell markers. RT-PCR analysis of expression of specific transcripts indicated that (1) under remission conditions, nephrotic children displayed induction of four transcripts, including IKBKB, and repression of NFKBIA as compared to non nephrotic children after recovery, and (2) proteinuria relapse was associated with induction of L-selectin and T-lymphocyte maturation-associated protein, two markers of T-cell differentiation and recent emigrant/naive T cells. CONCLUSION Results indicate that circulating T cells from relapsing nephrotic patients include a significant population of low-mature cells while those from nephrotic patients in remission are characterized by constitutive activation of nuclear factor-kappaB (NF-kappaB), altogether suggesting a thymic dysregulation of apoptosis in nephrotic patients.
Collapse
Affiliation(s)
- Hicham Mansour
- Institut des Cordeliers, CNRS-UPMC UMR7134, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
30
|
McKarns SC, Schwartz RH. Distinct Effects of TGF-β1 on CD4+ and CD8+ T Cell Survival, Division, and IL-2 Production: A Role for T Cell Intrinsic Smad3. THE JOURNAL OF IMMUNOLOGY 2005; 174:2071-83. [PMID: 15699137 DOI: 10.4049/jimmunol.174.4.2071] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
TGF-beta1 is critical for maintaining T cell homeostasis. Smad3 has been implicated in this regulatory process, yet the cellular targets and molecular details remain poorly understood. In this study, we report that TGF-beta1 impairs the entry of CD4+ and CD8+ T cells into the cell cycle as well as their progression through subsequent rounds of division, and show that Smad3 is essential for TGF-beta1 to inhibit TCR-induced division of only CD4+ and not CD8+ T cells. Both CD8+ and CD4+ T cells from Smad3-/- mice were refractory to TGF-beta1-induced inhibition of IL-2 production, thus demonstrating that not all CD8+ T cell responses to TGF-beta1 are Smad3 independent. These TGF-beta1 effects were all T cell intrinsic, as they were reproduced in purified CD4+ and CD8+ T cells. Finally, we found that Smad3 was critical for the survival of CD8+, but not CD4+ T cells following activation ex vivo. The TCR-induced death of Smad3-/- CD8+ T cells was not dependent upon TNF-alpha production. Exogenous TGF-beta1 partially rescued the CD8+ T cells by signaling through a Smad3-independent pathway. TGF-beta1 also enhanced survival of TCR-stimulated CD4+CD44high T cells in a Smad3-independent manner. Collectively, these findings firmly establish for the first time that TGF-beta1 discriminately regulates CD4+ and CD8+ T cell expansion by signaling through distinct intracellular pathways.
Collapse
Affiliation(s)
- Susan C McKarns
- Laboratory of Cellular and Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | |
Collapse
|
31
|
Liu D, Kang JS, Derynck R. TGF-beta-activated Smad3 represses MEF2-dependent transcription in myogenic differentiation. EMBO J 2004; 23:1557-66. [PMID: 15044954 PMCID: PMC391082 DOI: 10.1038/sj.emboj.7600179] [Citation(s) in RCA: 115] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2003] [Accepted: 02/26/2004] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor beta (TGF-beta) inhibits myogenesis and associated gene expression. We previously reported that the TGF-beta signaling effector Smad3 mediates this inhibition, by interfering with the assembly of myogenic bHLH transcription factor heterodimers on E-box sequences in the regulatory regions of muscle-specific genes. We now show that TGF-beta-activated Smad3 suppresses the function of MEF2, a second class of essential myogenic factors. TGF-beta signaling through Smad3 represses myogenin expression independently of E-boxes, and prevents a tethered MyoD-E47 dimer to activate transcription indirectly through MEF2-binding sites. In addition, Smad3 interacts with MEF2C, which requires its MADS domain, and disrupts its association with the SRC-family coactivator GRIP-1, thus diminishing the transcription activity of MEF2C. Consistent with this physical displacement, TGF-beta signaling blocks the GRIP-1-induced redistribution of MEF2C to discrete nuclear subdomains in 10T1/2 cells, and the recruitment of GRIP-1 to the myogenin promoter in differentiating myoblasts. These findings indicate that the TGF-beta/Smad3 pathway targets two critical components of the myogenic transcription machinery to inhibit terminal differentiation.
Collapse
Affiliation(s)
- Dong Liu
- Departments of Growth and Development and Anatomy, Programs in Cell Biology and Developmental Biology, University of California, San Francisco, CA, USA
| | - Jong Seok Kang
- Departments of Growth and Development and Anatomy, Programs in Cell Biology and Developmental Biology, University of California, San Francisco, CA, USA
| | - Rik Derynck
- Departments of Growth and Development and Anatomy, Programs in Cell Biology and Developmental Biology, University of California, San Francisco, CA, USA
| |
Collapse
|
32
|
Grotendorst GR, Rahmanie H, Duncan MR. Combinatorial signaling pathways determine fibroblast proliferation and myofibroblast differentiation. FASEB J 2004; 18:469-79. [PMID: 15003992 DOI: 10.1096/fj.03-0699com] [Citation(s) in RCA: 208] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Fibroblast proliferation, differentiation into myofibroblasts, and increased collagen synthesis are key events during both normal wound repair and fibrotic lesion formation. Here we report that these biological responses to TGF-beta by fibroblasts are regulated via a CTGF-dependent pathway in concert with either EGF or IGF-2. Our studies indicate these responses to TGF-beta are mutually exclusive, and cells that are proliferating do not express alpha-SMA or elevated levels of collagen synthesis. Cells expressing alpha-SMA do not exhibit DNA synthesis but do coexpress higher levels of types I and III collagen mRNA. Thus, fibroblast proliferation and differentiation are controlled by combinatorial signaling pathways involving not only components of the TGF-beta/CTGF pathway, but also signaling events induced by EGF and IGF-2-activated receptors. Collectively, our studies indicate TGF-beta functions as a classic embryonic inducer, initiating a cascade that is controlled by other factors in the cellular environment. We propose a model for this process with regard to wound repair and fibrotic lesion formation that is likely applicable to other instances of CTGF action during embryogenesis.
Collapse
Affiliation(s)
- Gary R Grotendorst
- Department of Cell Biology and Anatomy, University of Miami School of Medicine, Miami, Florida 33136, USA.
| | | | | |
Collapse
|
33
|
Wahl SM, Swisher J, McCartney-Francis N, Chen W. TGF-beta: the perpetrator of immune suppression by regulatory T cells and suicidal T cells. J Leukoc Biol 2004; 76:15-24. [PMID: 14966194 DOI: 10.1189/jlb.1103539] [Citation(s) in RCA: 123] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Innate and adaptive immunity function to eliminate foreign invaders and respond to injury while enabling coexistence with commensal microbes and tolerance against self and innocuous agents. Although most often effective in accomplishing these objectives, immunologic processes are not fail-safe and may underserve or be excessive in protecting the host. Checks and balances to maintain control of the immune system are in place and are becoming increasingly appreciated as targets for manipulating immunopathologic responses. One of the most recognized mediators of immune regulation is the cytokine transforming growth factor-beta (TGF-beta), a product of immune and nonimmune cells. Emerging data have unveiled a pivotal role for TGF-beta as a perpetrator of suppression by CD4(+)CD25(+) regulatory T (Treg) cells and in apoptotic sequelae. Through its immunosuppressive prowess, TGF-beta effectively orchestrates resolution of inflammation and control of autoaggressive immune reactions by managing T cell anergy, defining unique populations of Treg cells, regulating T cell death, and influencing the host response to infections.
Collapse
Affiliation(s)
- Sharon M Wahl
- NIDCR, NIH, Building 30, Rm. 320, 30 Convent Drive, MSC4352, Bethesda, MD 20892-4352, USA.
| | | | | | | |
Collapse
|
34
|
Azhar M, Schultz JEJ, Grupp I, Dorn GW, Meneton P, Molin DGM, Gittenberger-de Groot AC, Doetschman T. Transforming growth factor beta in cardiovascular development and function. Cytokine Growth Factor Rev 2003; 14:391-407. [PMID: 12948523 PMCID: PMC3855389 DOI: 10.1016/s1359-6101(03)00044-3] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Transforming growth factor betas (TGFbetas) are pleiotropic cytokines involved in many biological processes. Genetic engineering and tissue explanation studies have revealed specific non-overlapping roles for TGFbeta ligands and their signaling molecules in development and in normal function of the cardiovascular system in the adult. In the embryo, TGFbetas appear to be involved in epithelial-mesenchymal transformations (EMT) during endocardial cushion formation, and in epicardial epithelial-mesenchymal transformations essential for coronary vasculature, ventricular myocardial development and compaction. In the adult, TGFbetas are involved in cardiac hypertrophy, vascular remodeling and regulation of the renal renin-angiotensin system. The evidence for TGFbeta activities during cardiovascular development and physiologic function will be given and areas which need further investigation will be discussed.
Collapse
Affiliation(s)
- Mohamad Azhar
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bernasconi P, Passerini L, Annoni A, Ubiali F, Marcozzi C, Confalonieri P, Cornelio F, Mantegazza R. Expression of Transforming Growth Factor-β1 in Thymus of Myasthenia Gravis Patients. Ann N Y Acad Sci 2003; 998:278-83. [PMID: 14592886 DOI: 10.1196/annals.1254.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Pia Bernasconi
- Immunology and Muscular Pathology Unit, Istituto Nazionale Neurologico Carlo Besta, 20133 Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Pearson-White S, McDuffie M. Defective T-cell activation is associated with augmented transforming growth factor Beta sensitivity in mice with mutations in the Sno gene. Mol Cell Biol 2003; 23:5446-59. [PMID: 12861029 PMCID: PMC165712 DOI: 10.1128/mcb.23.15.5446-5459.2003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The proto-oncogene Sno has been shown to be a negative regulator of transforming growth factor beta (TGF-beta) signaling in vitro, using overexpression and artificial reporter systems. To examine Sno function in vivo, we made two targeted deletions at the Sno locus: a 5' deletion, with reduced Sno protein (hypomorph), and an exon 1 deletion removing half the protein coding sequence, in which Sno protein is undetectable in homozygotes (null). Homozygous Sno hypomorph and null mutant mice are viable without gross developmental defects. We found that Sno mRNA is constitutively expressed in normal thymocytes and splenic T cells, with increased expression 1 h following T-cell receptor ligation. Although thymocyte and splenic T-cell populations appeared normal in mutant mice, T-cell proliferation in response to activating stimuli was defective in both mutant strains. This defect could be reversed by incubation with either anti-TGF-beta antibodies or exogenous interleukin-2 (IL-2). Together, these findings suggest that Sno-dependent suppression of TGF-beta signaling is required for upregulation of growth factor production and normal T-cell proliferation following receptor ligation. Indeed, both IL-2 and IL-4 levels are reduced in response to anti-CD3 epsilon stimulation of mutant T cells, and transfected Sno activated an IL-2 reporter system in non-T cells. Mutant mouse embryo fibroblasts also exhibited a reduced cell proliferation rate that could be reversed by administration of anti-TGF-beta. Our data provide strong evidence that Sno is a significant negative regulator of antiproliferative TGF-beta signaling in both T cells and other cell types in vivo.
Collapse
Affiliation(s)
- S Pearson-White
- Department of Microbiology, Health Sciences Center, University of Virginia Medical Center, Jordan Hall, Box 800734, Room 7034, Charlottesville, VA 22908, USA.
| | | |
Collapse
|
37
|
Kettering JD, Mohamedali AM, Green LM, Gridley DS. IL-2 gene and antisense TGF-beta1 strategies counteract HSV-2 transformed tumor progression. Technol Cancer Res Treat 2003; 2:211-21. [PMID: 12779352 DOI: 10.1177/153303460300200305] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The H238 tumor cells are Herpes simplex virus type 2-transformed BALB/c mouse fibroblasts that constitutively express transforming growth factor (TGF-beta1). TGF-beta can diminish immune capacity, whereas interleukin 2 (IL-2) is stimulatory to the immune system and can counteract the negative effects of TGF-beta1. The H238-BALB/c system provides a syngeneic model to evaluate new strategies with the potential to ameliorate tumor-induced immune depression. Plasmids expressing either antisense TGF-beta1 or murine Il-2 were constructed and stably transformed cells generated (masH238 and H238-IL2, respectively). In vitro measurements (ELISA and RT-PCR) demonstrated a >70% decrease in TGF-beta1 secretion by the masH238 tumor cells, and significant levels of IL-2 production by the H238-IL2 transfected cells when compared to wild type and control plasmid-transfected H238 cells. BALB/c mice injected subcutaneously with the masH238 cells developed significantly smaller tumors than the controls. Mice injected with H238-IL-2 cells developed tumors that failed to progress relative to control tumor growth. The differences in tumor growth in the mice were associated with enhanced immune reactivity and an increased response to T lymphocyte mitogens. Significant differences were also noted in lymphocyte populations and expression of CD25 and CD71 activation markers in the blood and spleens of mice receiving transfected tumor cells. Collectively, the data demonstrate that strategies employing antisense TGF-beta1 and IL-2 expression by transfected tumor cells can counteract the progression of a TGF-beta1-secreting tumor and enhance immune function involving modulation of T lymphocyte populations.
Collapse
Affiliation(s)
- James D Kettering
- Dept. of Biochemistry & Microbiology, Loma Linda University, Loma Linda, CA 92350, USA.
| | | | | | | |
Collapse
|
38
|
Ruzek MC, Hawes M, Pratt B, McPherson J, Ledbetter S, Richards SM, Garman RD. Minimal effects on immune parameters following chronic anti-TGF-beta monoclonal antibody administration to normal mice. Immunopharmacol Immunotoxicol 2003; 25:235-57. [PMID: 12784916 DOI: 10.1081/iph-120020473] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Mice genetically deficient in TGF-beta1 or TGF-beta signaling capacity in T or B cells demonstrate profound immune dysregulation, as evidenced by increased lymph node size, expression of markers of memory/activation on T cells, inflammation in a variety of tissues and development of autoantibodies. However, this constant and complete lack of TGF-beta1 or TGF-betaR signaling may not reflect effects of TGF-beta neutralization using antibodies in mature animals. Thus, the present studies were designed to determine if administration of an anti-TGF-beta monoclonal antibody (neutralizes TGF-beta1, 2 and 3) to mature, normal mice results in evidence of immune dysregulation or immune-mediated pathology. An initial study examined daily administration of 0.25, 0.75 and 2.5 mg/kg of anti-TGF-beta to mice for three weeks, achieving blood levels of as high as 9 mg/ml. Comprehensive hematological and histopathological evaluation showed no evidence of pathology. A second study was designed to extend the antibody treatment period and further examine the functional status of the immune system. Mice were injected with 1 mg/mouse (approximately 50 mg/kg) of anti-TGF-beta (1D11) three times per week achieving circulating blood levels of 1-2 mg/ml. Many parameters of immune status were assessed, including natural killer (NK) cell activity, lymphocyte proliferative responses, phagocytic activity, phenotypic assessment of leukocyte subsets, and serum measurements of proinflammatory cytokines, autoantibodies and immunoglobulin isotypes. In addition, histopathological assessment of heart, lungs, liver, kidney, salivary glands, skin, spleen and lymph nodes was also performed. Very few of the multiple immune parameters examined showed detectable changes in anti-TGF-beta-treated mice. Changes that were observed were primarily restricted to the spleen and included increased spleen cell recoveries, increased percentages of macrophages, decreased percentages of NK cells, decreased phagocytic activity, decreased proliferative responses to mitogens and slight increases in T and B cells displaying an activated phenotype. Many of these same parameters examined in the lymph nodes were not altered by the anti-TGF-beta treatment. The thymus was decreased in size, but altered only slightly in one population of developing T cells. Most of the changes observed were modest and returned to control levels after discontinuation of treatments. The only serological finding was an increase in IgA levels in anti-TGF-beta-treated mice, but not in any other isotype. Finally, there was no evidence of increased inflammation in any of the peripheral tissues examined in the anti-TGF-beta-treated mice. In conclusion, although there were changes in some of the immunological parameters examined in these studies, they were few and typically reversed following discontinuation of treatment. The modest nature of the changes observed in these studies is particularly evident when compared to published data of those same parameters examined in mice genetically deficient in TGF-beta1 or mice having TGF-beta unresponsive T or B cells. Thus, there does not appear to be any significant immune dysregulation detectable after long-term antibody-mediated neutralization of TGF-beta in normal mice.
Collapse
Affiliation(s)
- Melanie C Ruzek
- Cell and Protein Therapeutics R&D, Genzyme Corporation, Framingham, Massachusetts 01701-9322, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Bommireddy R, Saxena V, Ormsby I, Yin M, Boivin GP, Babcock GF, Singh RR, Doetschman T. TGF-beta 1 regulates lymphocyte homeostasis by preventing activation and subsequent apoptosis of peripheral lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4612-22. [PMID: 12707339 DOI: 10.4049/jimmunol.170.9.4612] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TGF-beta1 plays an important role in the maintenance of immune homeostasis and self-tolerance. To determine the mechanism by which TGF-beta1 prevents autoimmunity we have analyzed T cell activation in splenic lymphocytes from TGF-beta1-deficient mice. Here we demonstrate that unlike wild-type splenic lymphocytes, those from Tgfb1(-/-) mice are hyporesponsive to receptor-mediated mitogenic stimulation, as evidenced by diminished proliferation and reduced IL-2 production. However, they have elevated levels of IFN-gamma and eventually undergo apoptosis. Receptor-independent stimulation of Tgfb1(-/-) T cells by PMA plus ionomycin induces IL-2 production and mitogenic response, and it rescues them from anergy. Tgfb1(-/-) T cells display decreased CD3 expression; increased expression of the activation markers LFA-1, CD69, and CD122; and increased cell size, all of which indicate prior activation. Consistently, mutant CD4(+) T cells have elevated intracellular Ca(2+) levels. However, upon subsequent stimulation in vitro, increases in Ca(2+) levels are less than those in wild-type cells. This is also consistent with the anergic phenotype. Together, these results demonstrate that the ex vivo proliferative hyporesponsiveness of Tgfb1(-/-) splenic lymphocytes is due to prior in vivo activation of T cells resulting from deregulated intracellular Ca(2+) levels.
Collapse
Affiliation(s)
- Ramireddy Bommireddy
- Department of Molecular Genetics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Bommireddy R, Ormsby I, Yin M, Boivin GP, Babcock GF, Doetschman T. TGF beta 1 inhibits Ca2+-calcineurin-mediated activation in thymocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:3645-52. [PMID: 12646629 PMCID: PMC2804068 DOI: 10.4049/jimmunol.170.7.3645] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
TGFbeta1 is a polypeptide growth modulatory and differentiation factor involved in many biological processes including immune homeostasis and self-tolerance. Tgfb1 knockout mice die around weaning age due to severe inflammation in most major organ systems, but the mechanism underlying this disease is not understood. In this study we demonstrate that Tgfb1(-/-) CD4(+)CD8(+) and CD4(+)CD8(-) thymocytes are hyperresponsive to receptor-mediated and receptor-independent mitogenic stimulation. A suboptimal concentration of ionomycin in the presence of PMA fully activates Tgfb1(-/-) thymocytes, whereas the inhibitors of Ca(2+) influx and calcineurin, EGTA and FK506, eliminate the hyperresponsiveness. Hence, the hypersensitivity of Tgfb1(-/-) thymocytes is due to a lowered threshold for Ca(2+)-dependent activation. Further, we demonstrate that the hypersensitivity of thymocytes results from the absence of TGFbeta1 and not from the inflammatory environment because the thymocytes are hyperresponsive in preinflammatory-stage Tgfb1(-/-) mice. Our results suggest for the first time that TGFbeta1 functions to inhibit aberrant T cell expansion by maintaining intracellular calcium concentration levels low enough to prevent a mitogenic response by Ca(2+)-independent stimulatory pathways alone. Consequently, TGFbeta1 prevents autoimmune disease through a Ca(2+) regulatory pathway that maintains the activation threshold above that inducible by self-MHC-TCR interactions.
Collapse
Affiliation(s)
- Ramireddy Bommireddy
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Ilona Ormsby
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Moying Yin
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Gregory P. Boivin
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - George F. Babcock
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267
- Shriners Hospital for Children, Cincinnati, OH 45229
| | - Thomas Doetschman
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
- Address correspondence and reprint requests to Dr. Thomas Doetschman, Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, 231 Albert Sabin Way, Cincinnati, OH 45267-0524.
| |
Collapse
|
41
|
Stewart GA, Hoyne GF, Ahmad SA, Jarman E, Wallace WAH, Harrison DJ, Haslett C, Lamb JR, Howie SEM. Expression of the developmental Sonic hedgehog (Shh) signalling pathway is up-regulated in chronic lung fibrosis and the Shh receptor patched 1 is present in circulating T lymphocytes. J Pathol 2003; 199:488-95. [PMID: 12635140 DOI: 10.1002/path.1295] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
During pulmonary development, Sonic hedgehog (Shh) and transforming growth factor beta1 (TGF-beta1) signalling both contribute to branching morphogenesis. In interstitial lung disease, the complex alveolar structure of the lung is disrupted and remodelled, which leads to fibrosis, loss of respiratory surface, morbidity, and mortality. It is well documented that TGF-beta1 is involved in fibrosis. However, little is known about Shh signalling in damaged epithelia. This study examined whether or not components of the Shh signalling pathway, as well as TGF-beta1, are expressed in human fibrotic lung disease (cryptogenic fibrosing alveolitis and bronchiectasis) and in murine experimental models of fibrotic and non-fibrotic chronic pulmonary inflammation. Using immunohistochemistry, it was observed that Shh, like TGF-beta1, is up-regulated in epithelial cells at sites of fibrotic disease but not non-fibrotic inflammation. The Shh receptor patched was detected in infiltrating mononuclear cells and alveolar macrophages, as well as normal resting peripheral blood T lymphocytes. Neither Shh nor patched is expressed by hyperproliferative goblet cells in inflammatory epithelium. This study demonstrates that patched is present in human peripheral CD4 and CD8 lymphocytes at both protein and mRNA levels. Taken together, these results suggest that components of the highly conserved Shh signalling pathway may play a role in the remodelling of damaged pulmonary epithelium and that damaged epithelium and cells of the immune system may communicate via this pathway.
Collapse
Affiliation(s)
- Gareth A Stewart
- Immunobiology Group, College of Medicine and Veterinary Medicine, University of Edinburgh, Teviot Place, Edinburgh, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Cytokines, Chemokines and Growth Factors in the Pathogenesis and Treatment of Inflammatory Bowel Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2003. [DOI: 10.1007/978-1-4615-0171-8_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Affiliation(s)
- C B Schmidt-Weber
- Swiss Institute of Allergy and Asthma Research, Obere Str. 22, CH-7270 Davos, Switzerland
| | | |
Collapse
|
44
|
Hobson KG, Cho K, Adamson LK, Greenhalgh DG. Burn-induced thymic apoptosis corresponds with altered TGF-beta(1) and Smad 2/3. J Surg Res 2002; 105:4-9. [PMID: 12069494 DOI: 10.1006/jsre.2002.6430] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Immune suppression is a common complication of injury. Transforming growth factor-beta(1) (TGF-beta(1)), a cytokine with diverse anti-inflammatory and anti-apoptotic effects, may play an important role. Smad 2 and Smad 3 are transcription factors that mediate the effects of TGF-beta(1). We hypothesized that burn-induced immunosuppression would be accompanied by increased apoptosis in lymphoid organs, a change likely associated with changes in TGF-beta(1) and Smad 2/3 expression. Mice were subjected to 18% body surface area flame burn. Lymph nodes, spleen, and thymus were harvested at multiple time points after injury. TGF-beta(1) and Smad 2/3 expression and levels of apoptosis were determined in whole tissue and in sorted T-cells by flow cytometry, RT-PCR, ELISA, and Western blot. TGF-beta(1) protein expression in the thymus increased from 1 to 7 days. Smad 2/3 protein expression was decreased at the same time points. This down-regulation was more dramatic in the non-T-cells than in the T-cells themselves. RT-PCR confirmed down-regulation of Smad 3 mRNA in the thymus from 3 to 6 h. Apoptosis in the thymus doubled at 1 day (6.4% control vs 12.8% burned), which corresponded with a marked decrease in thymus mass on subjective assessment. No changes were observed in other lymphoid organs. Burn injury in mice increases TGF-beta(1) expression in the thymus, while suppressing expression of its intracellular mediator, Smad 2/3. This response is most pronounced in the non-T-cell tissue, which suggests the thymic epithelial cells may be responsible for the increased thymic apoptosis. This TGF-beta(1) and Smad 2/3 counterregulation in response to injury may represent a potential mechanism for postinjury immune suppression.
Collapse
|
45
|
Schmidt-Weber CB, Kunzmann S, Blaser K. TGF-beta-mediated control of allergen-specific T-cell responses. Curr Allergy Asthma Rep 2002; 2:259-62. [PMID: 11918869 DOI: 10.1007/s11882-002-0028-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Control of allergen-specific response by suppressive cytokines involves several layers of regulation, including secretion of the cytokine, deviation of cytokine expression by altered T-cell differentiation, immediate (de-) phosphorylation events upon binding of suppressive cytokines, and laterations in susceptibility of suppression.
Collapse
|
46
|
Liversidge J, Dick A, Gordon S. Nitric oxide mediates apoptosis through formation of peroxynitrite and Fas/Fas-ligand interactions in experimental autoimmune uveitis. THE AMERICAN JOURNAL OF PATHOLOGY 2002; 160:905-16. [PMID: 11891189 PMCID: PMC1867184 DOI: 10.1016/s0002-9440(10)64913-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Conflicting reports have led to the description of nitric oxide as a "double-edged sword" in animal models of autoimmunity. In this study we show that tissue damage within the eye during experimental autoimmune uveoretinitis correlates with peroxynitrite formation in infiltrating monocytes/macrophages within the outer retina together with extensive photoreceptor apoptosis and apoptosis of Fas(+) T cells within the retina. Inducible nitric oxide synthase (NOS2) expression was primarily restricted to infiltrating monocytes/macrophages in the outer retina and photoreceptor rod outer segments (target tissue), but despite showing evidence of lipid peroxidation, myeloid cells remained resistant to apoptosis. The protective effect of the NOS inhibitor N(G)-nitro-L-arginine methyl ester could be attributed to dramatically reduced photoreceptor apoptosis, absence of nitrotyrosine formation, and reduced NOS2 protein expression. However, inhibition of NOS by N(G)-nitro-L-arginine methyl ester was accompanied by a sparing of CD3(+) and CD2(+) T cells despite continued expression of Fas and Fas ligand, thus compromising functional inactivation of T cells in the target tissue. These data suggests that in addition to contributing to tissue damage in the retina through generation of reactive oxygen species, nitric oxide also seems to be required for activation-induced cell death and elimination of T cells in the retina.
Collapse
Affiliation(s)
- Janet Liversidge
- Department of Ophthalmology, University ofAberdeen Medical School, Foresterhill, Aberdeen, United Kingdom.
| | | | | |
Collapse
|
47
|
Ling E, Robinson DS. Transforming growth factor-beta1: its anti-inflammatory and pro-fibrotic effects. Clin Exp Allergy 2002; 32:175-8. [PMID: 11929477 DOI: 10.1046/j.1365-2222.2002.01287.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
48
|
Brill A, Franitza S, Lider O, Hershkoviz R. Regulation of T-cell interaction with fibronectin by transforming growth factor-beta is associated with altered Pyk2 phosphorylation. Immunology 2001; 104:149-56. [PMID: 11683954 PMCID: PMC1783298 DOI: 10.1046/j.1365-2567.2001.01283.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although the involvement of transforming growth factor-beta (TGF-beta) in inflammatory reactions has been extensively studied, its mode of action in the context of the extracellular matrix (ECM) is still not fully understood. We undertook this study in an attempt to reveal the putative roles of TGF-beta in T-cell adhesion and migration. We found that a 60-min treatment of T cells with TGF-beta regulates T-cell adhesion to fibronectin (FN), a prototype cell adhesion protein of the ECM, depending on the presence of other activators. At 5 pg/ml to 1 ng/ml, TGF-beta alone induced T-cell adhesion to FN in an integrin alpha4/beta1- and integrin alpha5/beta1-dependent manner. TGF-beta also attenuated T-cell migration on the stromal cell-derived factor (SDF)-1alpha gradients. These effects of TGF-beta were not accompanied by alteration in the expression of very-late activation antigen type 4 (VLA-4) and VLA-5, nor were they mediated by the cyclo-oxygenase pathway. The cellular mechanism underlying the adhesion-regulating activities of TGF-beta involves adhesion-associated cytoskeletal elements. TGF-beta induced the phosphorylation of focal adhesion kinase Pyk2, but not extracellular signal-regulated kinase (ERK), and this effect was markedly increased in the presence of immobilized FN, suggesting a collaborative role for FN-specific integrins. Indeed, TGF-beta-induced Pyk2 phosphorylation was inhibited by monoclonal antibodies against VLA-4, VLA-5 and CD29. Thus, TGF-beta, which may appear at extravascular sites during inflammation, affects the adhesion of T cells to ECM glycoproteins and their migration by its ability to differentially induce or inhibit the phosphorylation of Pyk2.
Collapse
Affiliation(s)
- Alexander Brill
- Department of Immunology, The Weizmann Institute of ScienceRehovot, Israel
| | - Susanne Franitza
- Department of Immunology, The Weizmann Institute of ScienceRehovot, Israel
| | - Ofer Lider
- Department of Immunology, The Weizmann Institute of ScienceRehovot, Israel
| | | |
Collapse
|
49
|
Takahashi I, Kosaka H, Oritani K, Heath WR, Ishikawa J, Okajima Y, Ogawa M, Kawamoto S, Yamada M, Azukizawa H, Itami S, Yoshikawa K, Tomiyama Y, Matsuzawa Y. A new IFN-like cytokine, limitin, modulates the immune response without influencing thymocyte development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3156-63. [PMID: 11544301 DOI: 10.4049/jimmunol.167.6.3156] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
A novel IFN-like molecule, limitin, was recently identified and revealed to suppress B lymphopoiesis through the IFN-alphabeta receptor, although it lacked growth suppression on myeloid and erythroid progenitors. Here we have studied diverse effects of limitin on T lymphocytes and compared limitin with previously known IFNs. Like IFN-alpha and -beta, limitin modified immunity in the following responses. It suppressed mitogen- and Ag-induced T cell proliferation through inhibiting the responsiveness to exogenous IL-2 rather than suppressing the production of IL-2. In contrast, limitin enhanced cytotoxic T lymphocyte activity associated with the perforin-granzyme pathway. To evaluate the effect of limitin in vivo, a lethal graft-versus-host disease assay was established. Limitin-treatment of host mice resulted in the enhancement of graft-versus-host disease. Limitin did not influence thymocyte development either in fetal thymus organ cultures or in newborn mice injected with limitin-Ig, suggesting that limitin is distinguishable from IFN-alpha and -beta. From these findings, it can be speculated that the human homolog of limitin may be applicable for clinical usage because of its IFN-like activities with low adverse effects on, for example, T lymphopoiesis, erythropoiesis, and myelopoiesis.
Collapse
Affiliation(s)
- I Takahashi
- Department of Internal Medicine and Molecular Science, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Wang J, Guan E, Roderiquez G, Norcross MA. Synergistic induction of apoptosis in primary CD4(+) T cells by macrophage-tropic HIV-1 and TGF-beta1. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3360-6. [PMID: 11544326 DOI: 10.4049/jimmunol.167.6.3360] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Depletion of CD4(+) T lymphocytes is a central immunological characteristic of HIV-1 infection. Although the mechanism of such CD4(+) cell loss following macrophage-tropic (R5) HIV-1 infection remains unclear, interactions between viral and host cell factors are thought to play an important role in the pathogenesis of HIV-1 disease. Based on the observation that TGF-beta1 enhanced expression of HIV chemokine coreceptors, the role of this host factor in virus effects was investigated using PBLs cultured in a nonmitogen-added system in the absence or presence of TGF-beta1. Most CD4 cells in such cultures had the phenotype CD25(-)CD69(-)DR(-)Ki67(-) and were CD45RO(bright)CD45RA(dim). Cultured cells had increased expression of CCR5 and CXCR4 and supported both HIV-1 entry and completion of viral reverse transcription. Virus production by cells cultured in the presence of IL-2 was inhibited by TGF-beta1, and this inhibition was accompanied by a loss of T cells from the culture and an increase in CD4(+) T cell apoptosis. Whereas R5X4 and X4 HIV-1 infection was sufficient to induce T cell apoptosis, R5 HIV-1 failed to induce apoptosis of PBLs in the absence of TGF-beta1 despite the fact that R5 HIV-1 depletes CD4(+) T cells in vivo. Increased apoptosis with HIV and TGF-beta1 was associated with reduced levels of Bcl-2 and increased expression of apoptosis-inducing factor, caspase-3, and cleavage of BID, c-IAP-1, and X-linked inhibitor of apoptosis. These results show that TGF-beta1 promotes depletion of CD4(+) T cells after R5 HIV-1 infection by inducing apoptosis and suggest that TGF-beta1 might contribute to the pathogenesis of HIV-1 infection in vivo.
Collapse
Affiliation(s)
- J Wang
- Laboratory of Gene Regulation, Division of Therapeutic Proteins, Center for Biologics Evaluation and Research, Food and Drug Administration, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|