1
|
Lambaren K, Trac N, Fehrenbach D, Madhur M, Chung EJ. T Cell-Targeting Nanotherapies for Atherosclerosis. Bioconjug Chem 2025; 36:332-346. [PMID: 39979082 DOI: 10.1021/acs.bioconjchem.4c00590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Cardiovascular diseases remain the leading cause of mortality worldwide. Specifically, atherosclerosis is a primary cause of acute cardiac events. However, current therapies mainly focus on lipid-lowering versus addressing the underlying inflammatory response that leads to its development and progression. Nanoparticle-mediated drug delivery offers a promising approach for targeting and regulating these inflammatory responses. In atherosclerotic lesions, inflammatory cascades result in increased T helper (Th) 1 and Th17 activity and reduced T regulatory activation. The regulation of T cell responses is critical in preventing the inflammatory imbalance in atherosclerosis, making them a key therapeutic target for nanotherapy to achieve precise atherosclerosis treatment. By functionalizing nanoparticles with targeting modalities, therapeutic agents can be delivered specifically to immune cells in atherosclerotic lesions. In this Review, we outline the role of T cells in atherosclerosis, examine current nanotherapeutic strategies for targeting T cells and modulating their differentiation, and provide perspectives for the development of nanoparticles specifically tailored to target T cells for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Karla Lambaren
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Noah Trac
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
| | - Daniel Fehrenbach
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Meena Madhur
- Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Eun Ji Chung
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California 90089, United States
- Department of Medicine, Division of Nephrology and Hypertension, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
- Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California 90089, United States
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, Keck School of Medicine, University of Southern California, Los Angeles, California 90089, United States
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles, California 90089, United States
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California 90089, United States
- Bridge Institute, University of Southern California, Los Angeles, California 90089, United States
| |
Collapse
|
2
|
Moysi E, Sharma AA, O’Dell S, Georgakis S, Del Rio Estrada PM, Torres-Ruiz F, Navarro MG, Villalobos YAL, Rios SA, Reyes-Teran G, Beddall MH, Ko SH, Belinky F, Orfanakis M, de Leval L, Enriquez AB, Buckner CM, Moir S, Doria-Rose N, Boritz E, Mascola JR, Sekaly RP, Koup RA, Petrovas C. Neutralization activity in chronic HIV infection is characterized by a distinct programming of follicular helper CD4 T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.31.605954. [PMID: 39131331 PMCID: PMC11312598 DOI: 10.1101/2024.07.31.605954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
A subset of people living with HIV (PLWH) can produce broadly neutralizing antibodies (bNAbs) against HIV, but the lymph node (LN) dynamics that promote the generation of these antibodies are poorly understood. Here, we explored LN-associated histological, immunological, and virological mechanisms of bNAb generation in a cohort of anti-retroviral therapy (ART)-naïve PLWH. We found that participants who produce bNAbs, termed neutralizers, have a superior LN-associated B cell follicle architecture compared with PLWH who do not. The latter was associated with a significantly higher in situ prevalence of Bcl-6hi follicular helper CD4 T cells (TFH), expressing a molecular program that favors their differentiation and stemness, and significantly reduced IL-10 follicular suppressor CD4 T cells. Furthermore, our data reveal possible molecular targets mediating TFH- B cell interactions in neutralizers. Together, we identify cellular and molecular mechanisms that contribute to the development of bNAbs in PLWH.
Collapse
Affiliation(s)
- Eirini Moysi
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Ashish A. Sharma
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sijy O’Dell
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Spiros Georgakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Perla Mariana Del Rio Estrada
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Fernanda Torres-Ruiz
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Mauricio González Navarro
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico, Subdireccion de Otorrinolaringologia, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”
| | - Yara Andrea Luna Villalobos
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Santiago Avila Rios
- Centro de Investigación en Enfermedades Infecciosas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Gustavo Reyes-Teran
- Institutos Nacionales de Salud y Hospitales de Alta Especialidad, Secretaría de Salud de México, Mexico City, Mexico
| | - Margaret H. Beddall
- ImmunoTechnology Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Sung-Hee Ko
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Frida Belinky
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laurence de Leval
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana B. Enriquez
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Susan Moir
- Laboratory of Immunoregulation, NIAID, NIH, Bethesda, MD, USA
| | - Nicole Doria-Rose
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Eli Boritz
- Virus Persistence and Dynamics Section, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - John R. Mascola
- Virology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- ModeX Therapeutics, Weston, MA, USA
| | - Rafick-Pierre Sekaly
- Pathology Advanced Translational Research Unit, Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard A. Koup
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Huang SQ, Xia L, Xia YQ, Huang HL, Dong L. Icaritin attenuates recurrent spontaneous abortion in mice by modulating Treg/Th17 imbalance via TGF-β/SMAD signaling pathway. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119574. [PMID: 37689142 DOI: 10.1016/j.bbamcr.2023.119574] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/21/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023]
Abstract
Recurrent spontaneous abortion (RSA) is a challenging global issue. Although the cause is unknown, increasing evidence suggests that immunological factors play a crucial role in RSA development. Icaritin (ICT), a natural compound derived from Epimedium, has demonstrated diverse biological activities, including anti-inflammatory, anti-cancer, and immunomodulatory effects. In this study, we aimed to investigate the potential therapeutic role of ICT in mitigating RSA in a mouse model. Specifically, we sought to determine whether ICT could modulate the Treg/Th17 cell imbalance via the TGF-β/SMAD signaling pathway and contributed to improved pregnancy outcomes. We conducted experiments on a mouse model with RSA and administered ICT orally. We then examined the effects of ICT on various types of immune cells including Treg and Th17 cells, and assessed the pregnancy outcomes. We also investigated the potential signaling pathway ICT exerted its effects. Our findings revealed that treatment with ICT led to an increase in Treg cells and a decrease in Th17 cells, which restored immune homeostasis and contributed to improved pregnancy outcomes. Furthermore, our data demonstrated that ICT's effects were mediated through the activation of TGF-β/SMAD signaling components. In conclusion, our study suggested that ICT ameliorated RSA by modulating Treg/Th17 cell imbalance via the TGF-β/SMAD signaling pathway. GENERAL SIGNIFICANCE: Our results highlighted the potential of ICT as a novel therapeutic agent for RSA, offering new insights into the underlying mechanisms and opening avenues for future research and clinical translation.
Collapse
Affiliation(s)
- Si-Qing Huang
- Department of Gynecology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Lei Xia
- College of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yan-Qiu Xia
- Department of Gynecology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China
| | - Hong-Li Huang
- Department of Gynecology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Li Dong
- Department of Gynecology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| |
Collapse
|
4
|
Xu Y, Su M, Wang Z, Liu Q, Xu X, Gu S, Pan W, Ge W. Dysfunction of Tregs contributes to FGR pathogenesis via regulating Smads signalling pathway. J Cell Mol Med 2020; 24:3647-3655. [PMID: 32057179 PMCID: PMC7131912 DOI: 10.1111/jcmm.15059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/02/2020] [Accepted: 01/19/2020] [Indexed: 12/21/2022] Open
Abstract
Fetal growth restriction (FGR) is ranked number two of most common complication of abnormal pregnancy worldwide. The pathogenesis of FGR is complicated due to multiple aetiologies and the exact mechanism for FGR development is currently unknown. T regulatory cells (Tregs) are proven to play central roles in the maintenance of normal pregnancy. Peripheral blood samples of 102 pregnant human were collected analysed using flow cytometry to identify Tregs. We found that reduced Tregs and down-regulation of Foxp3 were observed in peripheral blood of FGR patients. In FGR mouse model, we have found that Tregs were not only reduced in spleen but also in placenta. In vitro, Foxp3 and its transcription regulatory signalling molecules, including P-Smad2, P-Smad3 and Smad4, were diminished as well. Inhibition on Foxp3 expression was partially reversed by overexpression of Smad2 and Smad4. In FGR patients, Western blot results revealed that Foxp3, P-Smad2, P-Smad3 and Smad4 expression was inhibited in placenta. Our preliminary result suggests that maternal-foetal immune tolerance mediated by Tregs plays an essential role in the development of FGR. The inhibited expression of Foxp3 and down-regulated Smad2/Smad3/Smad4 signalling pathway were involved in the FGR pathogenesis. Targeting maternal-foetal immune tolerance through Tregs might represent a novel therapeutic option for FGR.
Collapse
Affiliation(s)
- Yunzhao Xu
- Department of ObstetricsAffiliated Hospital of Nantong UniversityNantongJiangsuChina
- Department of Obstetrics and GynecologyNantong UniversityNantongJiangsuChina
| | - Min Su
- Department of ObstetricsAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Ziheng Wang
- Department of Clinical BiobankAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| | - Qinqin Liu
- Department of Obstetrics and GynecologyNantong UniversityNantongJiangsuChina
| | - Xiangyu Xu
- Department of Obstetrics and GynecologyNantong UniversityNantongJiangsuChina
| | - Shuting Gu
- Department of Obstetrics and GynecologyNantong UniversityNantongJiangsuChina
| | - Weidong Pan
- Department of Obstetrics and GynecologyNantong UniversityNantongJiangsuChina
| | - Wenliang Ge
- Department of Pediatric SurgeryAffiliated Hospital of Nantong UniversityNantongJiangsuChina
| |
Collapse
|
5
|
Mao R, Liu H, Yi B, Purohit S, Kuczma M, Kraj P, She JX. Cell-based high throughput screening identified a novel compound that promotes regulatory T cells and prevents autoimmune colitis. Biochem Pharmacol 2019; 169:113618. [PMID: 31449782 DOI: 10.1016/j.bcp.2019.08.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 08/21/2019] [Indexed: 10/26/2022]
Abstract
Regulatory T cells (TR) show great promise for treating autoimmune diseases, allergies and preventing transplant rejection; however, their clinical application has been hampered by the lack of efficient ex vivo or in vivo expansion strategies. Here we report screening data on 130,000 low molecular weight (LMW) compounds for their TR promoting potential using a self-developed high-throughput cell-based assay. One of the lead compounds, an isoxazolecarboxamide designated as TRP38, efficiently converts naïve CD4+ T cells to TR cells in vitro and protects mice from autoimmune colitis in vivo. In addition, TRP38 can synergize with other compounds and/or cytokines such as rapamycin and TGFβ for TR conversion, probably via directly inhibiting P70s6 phosphorylation without affecting mTOR expression, underscoring the importance of complementary and coordinated activity of multiple signaling pathways for the increased level of stable TR cell production.
Collapse
Affiliation(s)
- Rui Mao
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, United States
| | - Haitao Liu
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, United States
| | - Bing Yi
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, United States
| | - Sharad Purohit
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, United States; Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States; Department of Interdisciplinary Health Sciences, Augusta University, Augusta, GA 30912, United States
| | - Michal Kuczma
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, United States
| | - Piotr Kraj
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, United States
| | - Jin-Xiong She
- Center for Biotechnology and Genomic Medicine, Augusta University, Augusta, GA 30912, United States; Department of Obstetrics and Gynecology, Medical College of Georgia, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
6
|
NF-κB-driven miR-34a impairs Treg/Th17 balance via targeting Foxp3. J Autoimmun 2019; 102:96-113. [DOI: 10.1016/j.jaut.2019.04.018] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/20/2019] [Accepted: 04/22/2019] [Indexed: 12/11/2022]
|
7
|
Zhu F, Yi G, Liu X, Zhu F, Zhao A, Wang A, Zhu R, Chen Z, Zhao B, Fang S, Yu X, Lin R, Liang R, Li D, Zhao W, Zhang Z, Guo W, Zhang S, Ge S, Fan X, Zhao G, Li B. Ring finger protein 31-mediated atypical ubiquitination stabilizes forkhead box P3 and thereby stimulates regulatory T-cell function. J Biol Chem 2018; 293:20099-20111. [PMID: 30389786 DOI: 10.1074/jbc.ra118.005802] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/21/2018] [Indexed: 01/05/2023] Open
Abstract
The CD4+CD25+FOXP3+ regulatory T (Treg) cells are critical for maintaining immune tolerance in healthy individuals and are reported to restrict anti-inflammatory responses and thereby promote tumor progression, suggesting them as a target in the development of antitumor immunotherapy. Forkhead box P3 (FOXP3) is a key transcription factor governing Treg lineage differentiation and their immune-suppressive function. Here, using Treg cells, as well as HEK-293T and Jurkat T cells, we report that the stability of FOXP3 is directly and positively regulated by the E3 ubiquitin ligase ring finger protein 31 (RNF31), which catalyzes the conjugation of atypical ubiquitin chains to the FOXP3 protein. We observed that shRNA-mediated RNF31 knockdown in human Treg cells decreases FOXP3 protein levels and increases levels of interferon-γ, resulting in a Th1 helper cell-like phenotype. Human Treg cells that ectopically expressed RNF31 displayed stronger immune-suppressive capacity, suggesting that RNF31 positively regulates both FOXP3 stability and Treg cell function. Moreover, we found that RNF31 is up-regulated in Treg cells that infiltrate human gastric tumor tissues compared with their counterparts residing in peripheral and normal tissue. We also found that elevated RNF31 expression in intratumoral Treg cells is associated with poor survival of gastric cancer patients, suggesting that RNF31 supports the immune-suppressive functions of Treg cells. Our results suggest that RNF31 could be a potential therapeutic target in immunity-based interventions against human gastric cancer.
Collapse
Affiliation(s)
- Fuxiang Zhu
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025,; the Unit of Molecular Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Shanghai 200025
| | - Gang Yi
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025,; the Shanghai Key laboratory of Bio-energy Crops, School of Life Science, Shanghai University, Shanghai 200025
| | - Xu Liu
- the Department of Gastrointestinal Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai 200025
| | - Fangming Zhu
- the Shanghai Key laboratory of Bio-energy Crops, School of Life Science, Shanghai University, Shanghai 200025
| | - Anna Zhao
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025
| | - Aiting Wang
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025,; the Unit of Molecular Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Shanghai 200025
| | - Ruihong Zhu
- the Unit of Molecular Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Shanghai 200025
| | - Zuojia Chen
- the Unit of Molecular Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Shanghai 200025
| | - Binbin Zhao
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025,; the Unit of Molecular Immunology, Institut Pasteur of Shanghai, University of Chinese Academy of Sciences, Shanghai 200025
| | - Sijie Fang
- the Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025
| | - Xiao Yu
- the Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Digestive Organ Transplantation, Henan 450052, and
| | - Ruirong Lin
- the Department of Biliary-Pancreatic Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Rui Liang
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025
| | - Dan Li
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025
| | - Wenyi Zhao
- the Department of Gastrointestinal Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai 200025
| | - Zizhen Zhang
- the Department of Gastrointestinal Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai 200025
| | - Wenzhi Guo
- the Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Digestive Organ Transplantation, Henan 450052, and
| | - Shuijun Zhang
- the Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Digestive Organ Transplantation, Henan 450052, and
| | - Shengfang Ge
- the Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025
| | - Xianqun Fan
- the Department of Ophthalmology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025
| | - Gang Zhao
- the Department of Gastrointestinal Surgery, RenJi Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Rd., Shanghai 200025,.
| | - Bin Li
- From the Shanghai Institute of Immunology and Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai 200025,.
| |
Collapse
|
8
|
Do JS, Zhong F, Huang AY, Van't Hof WJ, Finney M, Laughlin MJ. Foxp3 expression in induced T regulatory cells derived from human umbilical cord blood vs. adult peripheral blood. Bone Marrow Transplant 2018; 53:1568-1577. [PMID: 29743573 DOI: 10.1038/s41409-018-0205-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 12/23/2022]
Abstract
Foxp3 is essential for T regulatory cell (Treg) function. Broad complex-Tramtrack-Bric-a-brac domain (BTB) and Cap'n'collar (CNC) homology 1, transcription factor 2 (BACH2) stabilizes Treg immune homeostasis in murine studies. However, little is known regarding what role, if any, BACH2 may have in Foxp3 regulation in human-induced Treg (iTreg). We examined Foxp3 expression and regulation comparing iTreg differentiated from umbilical cord blood (UCB) vs. adult blood (AB) naive CD4+ T-cells. Foxp3 expression was higher in UCB vs. AB-derived iTreg, and was sustained during 21-day expansion in vitro. The number of Foxp3+ iTreg generated from UCB vs. AB naive CD4+ T-cells was higher in iTreg differentiation conditions. In addition, UCB iTreg were more potent in suppressing T-cell proliferation compared to AB iTreg. Naive UCB CD4+ T-cells highly expressed BACH2 protein compared to AB. Putative transcriptional BACH2 binding sites were identified at the Foxp3 promoter, using BACH2 consensus sequence. Cross-linking chromatin immunoprecipitation (ChIP) showed that BACH2 binds to the Foxp3 proximal promoter in UCB iTreg, but not AB iTreg. BACH2 was transcriptionally active, as shRNA-mediated BACH2 knockdown resulted in reduction of Foxp3 gene transcription in UCB CD4+ T-cells. In summary, BACH2 serves to stabilize robust Foxp3 expression in UCB CD4+ T-cell-derived iTreg.
Collapse
Affiliation(s)
- Jeong-Su Do
- Cleveland Cord Blood Center, Cleveland, OH, USA. .,Case Western Reserve University, Cleveland, OH, USA.
| | - Fei Zhong
- Cleveland Cord Blood Center, Cleveland, OH, USA
| | - Alex Y Huang
- Case Western Reserve University, Cleveland, OH, USA
| | | | | | - Mary J Laughlin
- Cleveland Cord Blood Center, Cleveland, OH, USA.,Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
9
|
Cuadrado E, van den Biggelaar M, de Kivit S, Chen YY, Slot M, Doubal I, Meijer A, van Lier RA, Borst J, Amsen D. Proteomic Analyses of Human Regulatory T Cells Reveal Adaptations in Signaling Pathways that Protect Cellular Identity. Immunity 2018; 48:1046-1059.e6. [DOI: 10.1016/j.immuni.2018.04.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 11/02/2017] [Accepted: 04/09/2018] [Indexed: 12/12/2022]
|
10
|
Shen L, Zhang LL, Li H, Liu X, Yu XX, Hu P, Hui H, Guo QL, Zhang S. Oroxylin A inhibits the generation of Tregs in non-small cell lung cancer. Oncotarget 2018; 8:49395-49408. [PMID: 28472762 PMCID: PMC5564777 DOI: 10.18632/oncotarget.17218] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 03/22/2017] [Indexed: 01/11/2023] Open
Abstract
Oroxylin A (OA), a naturally occurring monoflavonoid isolated from Scutellariae radix, has previously been reported to inhibit the proliferation of several cancer cell lines. CD4+CD25+Foxp3+ regulatory T cells (Tregs) play an important role in maintenance of immunologic self-tolerance. Tregs also increase in cancer and take part in suppressing antitumor immune responses. Here, we explored how OA affected the Tregs in lung cancer environment and the involved underlying mechanism. It is found that OA reversed the generation of Tregs induced by H460 lung cancer cells co-culture. Furthermore, in vivo, OA reduced tumor formation rate and attenuated Foxp3 expression in tumor-infiltrating lymphocytes. We also found that transforming growth factor-β1 (TGF-β1) neutralizing antibody reversed the enhancement of Treg number and expression of p-Smad3ˎ p-p38ˎp-JNKˎp-ERK1/2 in the co-culture model. Moreover, OA reduced the secretion of TGF-β1 and down-regulated the activation of NF-κB signaling in H460 cells. OA also inhibited Treg activity by a direct inhibition of the T cells' response to TGF-β1. In conclusion, our study demonstrated that OA inhibits the generation of Tregs in lung cancer environment by inhibiting the T cells' response to TGF-β1 and decreasing the secretion of TGF-β1 in lung cancer cells via NF-κB signaling.
Collapse
Affiliation(s)
- Le Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lu-Lu Zhang
- Department of Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiao Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiao-Xuan Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Po Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Qing-Long Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Shuai Zhang
- Department of Thoracic Surgery, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Cancer Institute of Jiangsu Province, Nanjing 210009, People's Republic of China
| |
Collapse
|
11
|
Peng J, Yu Z, Xue L, Wang J, Li J, Liu D, Yang Q, Lin Y. The effect of foxp3-overexpressing Treg cells on non-small cell lung cancer cells. Mol Med Rep 2018; 17:5860-5868. [PMID: 29436663 PMCID: PMC5866031 DOI: 10.3892/mmr.2018.8606] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/13/2017] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to investigate the novel mechanisms of forkhead box protein P3 (foxp3) in T regulatory (Treg) cells in lung cancer behavior. Treg cells were isolated from the peripheral blood of healthy volunteers and then co-cultured with 95D cells. A plasmid overexpressing foxp3 was constructed and transfected into Treg cells and an MTS assay was performed to assess cell viability. Flow cytometry was performed to evaluate cell apoptosis and reverse transcription-quantitative polymerase chain reaction was used to measure mRNA expression. A Transwell assay was used to assess cell invasion. Treg cells were successfully isolated from peripheral blood with purity of 94.26%. Foxp3 expression in Treg cells was significantly increased following co-culture with 95D cells, while matrix metalloproteinase-9 expression was upregulated in 95D cells co-cultured with Treg cells. The apoptosis, invasion and migration abilities of 95D cells were suppressed by co-culture with Treg cells, whereas the adhesive ability was enhanced. Foxp3 overexpression in Treg cells enhanced the viability and invasiveness of 95D cells, whereas cell adhesion and migration were decreased. The results of the present study demonstrate that the viability and invasiveness of 95D cells are enhanced by foxp3 overexpression in Treg cells, indicating that increased levels of foxp3 in the tumor microenvironment may promote tumor cell growth.
Collapse
Affiliation(s)
- Jiangzhou Peng
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Zigang Yu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Lei Xue
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Jiabin Wang
- Department of Thoracic Surgery, Shanwei People's Hospital, Shanwei, Guangdong 516600, P.R. China
| | - Jun Li
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Degang Liu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Qiang Yang
- Department of Thoracic Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Yihui Lin
- Department of Neurology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| |
Collapse
|
12
|
Oh H, Grinberg-Bleyer Y, Liao W, Maloney D, Wang P, Wu Z, Wang J, Bhatt DM, Heise N, Schmid RM, Hayden MS, Klein U, Rabadan R, Ghosh S. An NF-κB Transcription-Factor-Dependent Lineage-Specific Transcriptional Program Promotes Regulatory T Cell Identity and Function. Immunity 2017; 47:450-465.e5. [PMID: 28889947 DOI: 10.1016/j.immuni.2017.08.010] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/03/2017] [Accepted: 08/17/2017] [Indexed: 01/30/2023]
Abstract
Both conventional T (Tconv) cells and regulatory T (Treg) cells are activated through ligation of the T cell receptor (TCR) complex, leading to the induction of the transcription factor NF-κB. In Tconv cells, NF-κB regulates expression of genes essential for T cell activation, proliferation, and function. However the role of NF-κB in Treg function remains unclear. We conditionally deleted canonical NF-κB members p65 and c-Rel in developing and mature Treg cells and found they have unique but partially redundant roles. c-Rel was critical for thymic Treg development while p65 was essential for mature Treg identity and maintenance of immune tolerance. Transcriptome and NF-κB p65 binding analyses demonstrated a lineage specific, NF-κB-dependent transcriptional program, enabled by enhanced chromatin accessibility. These dual roles of canonical NF-κB in Tconv and Treg cells highlight the functional plasticity of the NF-κB signaling pathway and underscores the need for more selective strategies to therapeutically target NF-κB.
Collapse
Affiliation(s)
- Hyunju Oh
- Department of Microbiology & Immunology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Yenkel Grinberg-Bleyer
- Department of Microbiology & Immunology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Will Liao
- New York Genome Center, New York, NY 10013, USA
| | | | - Pingzhang Wang
- Department of Systems Biology and Department of Biomedical Informatics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Zikai Wu
- Department of Systems Biology and Department of Biomedical Informatics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jiguang Wang
- Department of Systems Biology and Department of Biomedical Informatics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Dev M Bhatt
- Department of Microbiology & Immunology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA
| | - Nicole Heise
- Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Roland M Schmid
- II Medizinische Klinik, Klinikum Rechts der Isar, Technische Universität Munich, Munich, Germany
| | - Matthew S Hayden
- Department of Microbiology & Immunology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA; Section of Dermatology, Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, 03756, USA
| | - Ulf Klein
- Department of Microbiology & Immunology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA; Department of Pathology & Cell Biology, College of Physicians & Surgeons, Columbia University, New York, NY 10032, USA
| | - Raul Rabadan
- Department of Systems Biology and Department of Biomedical Informatics, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Sankar Ghosh
- Department of Microbiology & Immunology, Columbia University College of Physicians & Surgeons, New York, NY 10032, USA.
| |
Collapse
|
13
|
Zhao M, Zhang H, Liu X, Jiang Y, Ren L, Hu X. The Effect of TGF-β on Treg Cells in Adverse Pregnancy Outcome upon Toxoplasma gondii Infection. Front Microbiol 2017; 8:901. [PMID: 28603517 PMCID: PMC5445113 DOI: 10.3389/fmicb.2017.00901] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 05/03/2017] [Indexed: 11/30/2022] Open
Abstract
Toxoplasma gondii (T. gondii) is a ubiquitous intracellular protozoan parasite that causes adverse pregnancy outcomes. Its infection downregulates the Treg cell population and TGF-β level, which may contribute to adverse pregnancy outcomes. TGF-β plays a critical role in Treg cell development, but whether TGF-β treatment can affect the number and function of Treg cells and hence alleviate adverse pregnancy outcomes caused by T. gondii infection remains elusive. In this study, T. gondii-infected pregnant mice were treated with TGF-β or TGF-β-neutralizing antibody. The pregnancy outcomes were observed on gestational day 14. The numbers of Treg cells and pSmad3, programmed death 1 (PD-1), and Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) expression of Treg cells were analyzed by flow cytometry. Histological changes were assessed using HE staining, while IL-10 and TNF-α levels were measured using ELISA. The results indicated that TGF-β treatment improved the T. gondii-induced adverse pregnancy outcomes, with alleviation of hemorrhage, restoration of uterine spiral arteries of the placenta, and increased Treg cell numbers; meanwhile, TGF-β neutralization resulted in more serious adverse pregnancy outcomes, with serious hemorrhage, more dilated uterine spiral arteries, and decreased Treg cell numbers. pSmad3 expression in CD4+ cells and CTLA-4 and PD-1 levels on Treg cells were upregulated by TGF-β treatment, but downregulated by TGF-β neutralization. The ratio of IL-10/TNF-α also increased after TGF-β treatment, but decreased after TGF-β neutralization. Our data indicate that TGF-β treatment could improve adverse pregnancy outcomes caused by T. gondii infection by upregulating Treg cell differentiation and function via the TGF-β/Smad3 signaling pathway, but not the proliferation of Treg cells.
Collapse
Affiliation(s)
- Mingdong Zhao
- Department of Radiology, Affiliated Hospital of Binzhou Medical UniversityBinzhou, China
| | - Haixia Zhang
- Department of Immunology, Binzhou Medical UniversityYantai, China
| | - Xianbing Liu
- Department of Immunology, Binzhou Medical UniversityYantai, China
| | - Yuzhu Jiang
- Department of Immunology, Binzhou Medical UniversityYantai, China
| | - Liqin Ren
- Medicine and Pharmacy Research Center, Binzhou Medical UniversityYantai, China
| | - Xuemei Hu
- Department of Immunology, Binzhou Medical UniversityYantai, China.,Medicine and Pharmacy Research Center, Binzhou Medical UniversityYantai, China
| |
Collapse
|
14
|
Chen J, Huang C, Zhu D, Shen P, Duan Y, Wang J, Yang C, Wu L. Chinese 1 strain of Toxoplasma gondii excreted-secreted antigens negatively modulate Foxp3 via inhibition of the TGFßRII/Smad2/Smad3/Smad4 pathway. J Cell Mol Med 2017; 21:1944-1953. [PMID: 28300338 PMCID: PMC5571543 DOI: 10.1111/jcmm.13115] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 01/02/2017] [Indexed: 11/29/2022] Open
Abstract
Toxoplasma gondii is an opportunistic intracellular parasite and is considered an important aetiological factor in the process of abortion, especially as occurs in early gestation. Chinese 1 strain of T. gondii is a dominant genotype prevalent in China. Although it is known that early foetal resorption triggered by RH strain of T. gondii is attributable to immune mechanisms rather than its direct effect in uterus, the underlying mechanism of the abortion caused by Chinese 1 strain remains unclear. This study was designed to investigate the effect of excreted–secreted antigens (ESA) of Chinese 1 strain of T. gondii on the expression of forkhead box transcription factor (Foxp3) as it pertains to early pregnancy and abortion. ESA caused a marked inhibition in the expression of Foxp3 both in vivo and in vitro. In addition, ESA negatively modulated Smad2 and Smad3 at the posttranslational level. Smad2 siRNA cooperated with ESA to further suppress the level of Foxp3. This inhibitory effect on Foxp3 expression was partially abrogated by overexpression of Smad2, Smad3 and Smad4. Additionally, ESA attenuated the expression of TGFßRII, whereas TGFßRII agonist could profoundly reversed the decreased Foxp3 triggered by ESA. Collectively, the findings suggested that ESA restricted Foxp3 expression by inhibiting TGFßRII/Smad2/Smad3/Smad4 signalling, ultimately resulting in abortion.
Collapse
Affiliation(s)
- Jinling Chen
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Caiqun Huang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Dandan Zhu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Pei Shen
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yinong Duan
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Jianxin Wang
- Laboratory Medicine Center, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Chunzhao Yang
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| | - Liting Wu
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
15
|
Stokes III JA, Mishra MK. Role of Resveratrol (RES) in Regenerative Medicine. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Within the last quarter century, technology has been a major catalyst of the advancement in various fields of scientific knowledge, particularly medical research. This new enlightenment has spurred the exploration of alternative treatment methods to some of society's most problematic diseases. One such innovative treatment is the use of Resveratrol (RES) to treat a number of pathophysiological conditions. RES is a natural polyphenolic compound found in the skin(s) of blueberries, red grapes (a major constituent of red wine), some vegetables, and even peanuts. The compound has a number of potent regenerative properties, which include: anti-aging, anti-inflammatory, and antioxidative. Research has confirmed both in vivo and in vitro RES's beneficial applications to numerous diseases. This chapter centers on its unique healing powers and beneficial applications against myriad debilitating conditions.
Collapse
|
16
|
Chan IH, Wu V, Bilardello M, Jorgenson B, Bal H, McCauley S, Van Vlasselaer P, Mumm JB. PEG-rIL-10 treatment decreases FoxP3(+) Tregs despite upregulation of intratumoral IDO. Oncoimmunology 2016; 5:e1197458. [PMID: 27622052 DOI: 10.1080/2162402x.2016.1197458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/24/2016] [Accepted: 05/29/2016] [Indexed: 10/21/2022] Open
Abstract
IL-10 has been classically defined as a broad-spectrum immunosuppressant and is thought to facilitate the development of regulatory CD4(+) T cells. IL-10 is believed to represent one of the major suppressive factors secreted by IDO(+)FoxP3(+)CD4(+) Tregs. Contrary to this view, we have previously reported that PEGylated recombinant IL-10 (PEG-rIL-10) treatment of mice induces potent IFNγ and CD8(+) T-cell-dependent antitumor immunity. This hypothesis is currently being tested in clinical trials and we have reported that treatment of cancer patients with PEG-rHuIL-10 results in inhibition and regression of tumor growth as well as increased serum IFNγ. We have continued to assess PEG-rIL-10's pleiotropic effects and report that treatment of tumor-bearing mice and humans with PEG-rIL-10 increases intratumoral indoleamine 2, 3-dioxygenase (IDO) in an IFNγ-dependent manner. This should result in an increase in Tregs, but paradoxically our data illustrate that PEG-rIL-10 treatment of mice reduces intratumoral FoxP3(+)CD4(+) T cells in an IDO-independent manner. Additional investigation indicates that PEG-rIL-10 inhibits TGFβ/IL-2-dependent in vitro polarization of FoxP3(+)CD4(+) Tregs and potentiates IFNγ(+)T-bet(+)CD4(+) T cells. These data suggest that rather than acting as an immunosuppressant, PEG-rIL-10 may counteract the FoxP3(+)CD4(+) Treg suppressive milieu in tumor-bearing mice and humans, thereby further facilitating PEG-rIL-10's potent antitumor immunity.
Collapse
|
17
|
Liu X, Berry CT, Ruthel G, Madara JJ, MacGillivray K, Gray CM, Madge LA, McCorkell KA, Beiting DP, Hershberg U, May MJ, Freedman BD. T Cell Receptor-induced Nuclear Factor κB (NF-κB) Signaling and Transcriptional Activation Are Regulated by STIM1- and Orai1-mediated Calcium Entry. J Biol Chem 2016; 291:8440-52. [PMID: 26826124 DOI: 10.1074/jbc.m115.713008] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Indexed: 12/18/2022] Open
Abstract
T cell activation following antigen binding to the T cell receptor (TCR) involves the mobilization of intracellular Ca(2+) to activate the key transcription factors nuclear factor of activated T lymphocytes (NFAT) and NF-κB. The mechanism of NFAT activation by Ca(2+) has been determined. However, the role of Ca(2+) in controlling NF-κB signaling is poorly understood, and the source of Ca(2+) required for NF-κB activation is unknown. We demonstrate that TCR- but not TNF-induced NF-κB signaling upstream of IκB kinase activation absolutely requires the influx of extracellular Ca(2+) via STIM1-dependent Ca(2+) release-activated Ca(2+)/Orai channels. We further show that Ca(2+) influx controls phosphorylation of the NF-κB protein p65 on Ser-536 and that this posttranslational modification controls its nuclear localization and transcriptional activation. Notably, our data reveal that this role for Ca(2+) is entirely separate from its upstream control of IκBα degradation, thereby identifying a novel Ca(2+)-dependent distal step in TCR-induced NF-κB activation. Finally, we demonstrate that this control of distal signaling occurs via Ca(2+)-dependent PKCα-mediated phosphorylation of p65. Thus, we establish the source of Ca(2+) required for TCR-induced NF-κB activation and define a new distal Ca(2+)-dependent checkpoint in TCR-induced NF-κB signaling that has broad implications for the control of immune cell development and T cell functional specificity.
Collapse
Affiliation(s)
| | - Corbett T Berry
- From the Departments of Pathobiology and the School of Biomedical Engineering, Drexel University, Philadelphia, Pennsylvania 19104
| | | | | | | | - Carolyn M Gray
- Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Lisa A Madge
- Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Kelly A McCorkell
- Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | | | - Uri Hershberg
- the School of Biomedical Engineering, Drexel University, Philadelphia, Pennsylvania 19104
| | - Michael J May
- Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | | |
Collapse
|
18
|
Lv Q, Qiao SM, Xia Y, Shi C, Xia YF, Chou GX, Wang ZT, Dai Y, Wei ZF. Norisoboldine ameliorates DSS-induced ulcerative colitis in mice through induction of regulatory T cells in colons. Int Immunopharmacol 2015; 29:787-797. [PMID: 26363976 DOI: 10.1016/j.intimp.2015.08.040] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 08/26/2015] [Accepted: 08/31/2015] [Indexed: 12/13/2022]
Abstract
Norisoboldine (NOR), the main active constituent of Radix Linderae, was previously demonstrated to ameliorate collagen-induced arthritis in rats through regulating the imbalance of T cells in intestines, which implied its therapeutic potential in inflammatory bowel disease. Here, we investigated the effect of NOR on ulcerative colitis (UC) induced by dextran sulfate sodium (DSS) in mice. Results showed that NOR (20, 40mg/kg) markedly reduced the symptoms of colitis, the levels of IL-1β and TNF-α, and the activation of ERK, p38 MAPK and NF-κB-p65. NOR only slightly decreased the levels of IFN-γ and IL-17A in mouse colons, but it dramatically increased the level of IL-10 at both protein and mRNA grades. Consistently, NOR increased the number of CD4(+)CD25(+)Foxp3(+) Treg cells more obviously than it decreased that of CD4(+)IL-17(+) Th17 cells in mesenteric lymph nodes (MLNs) and colonic lamina proprias (LPs) of colitis mice, and promoted the expression of Foxp3 mRNA in colon tissues. It could facilitate the in vitro differentiation of Treg cells from naive T cells and promote the phosphorylations of Smad2/3 in colon tissues of colitis mice. On the other hand, NOR did not affect the expressions of homing receptors CCR9 and α4β7 in SPs, and homing ligands CCL25 and Madcam-1 in MLNs and colonic LPs, suggesting that the increase of Treg cells in colons by NOR was not due to gut homing. In conclusion, NOR can ameliorate DSS-induced UC in mice, and the mechanisms involve reduction of pro-inflammatory cytokines and selective induction of Treg cells in colons.
Collapse
Affiliation(s)
- Qi Lv
- State Key Laboratory of Natural Medicine, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Si-Miao Qiao
- State Key Laboratory of Natural Medicine, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Ying Xia
- State Key Laboratory of Natural Medicine, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Can Shi
- State Key Laboratory of Natural Medicine, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yu-Feng Xia
- State Key Laboratory of Natural Medicine, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Gui-Xin Chou
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zheng-Tao Wang
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yue Dai
- State Key Laboratory of Natural Medicine, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| | - Zhi-Feng Wei
- State Key Laboratory of Natural Medicine, Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
19
|
Kyurkchiev D, Bochev I, Ivanova-Todorova E, Mourdjeva M, Oreshkova T, Belemezova K, Kyurkchiev S. Secretion of immunoregulatory cytokines by mesenchymal stem cells. World J Stem Cells 2014; 6:552-570. [PMID: 25426252 PMCID: PMC4178255 DOI: 10.4252/wjsc.v6.i5.552] [Citation(s) in RCA: 462] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 08/20/2014] [Accepted: 09/10/2014] [Indexed: 02/06/2023] Open
Abstract
According to the minimal criteria of the International Society of Cellular Therapy, mesenchymal stem cells (MSCs) are a population of undifferentiated cells defined by their ability to adhere to plastic surfaces when cultured under standard conditions, express a certain panel of phenotypic markers and can differentiate into osteogenic, chondrogenic and adipogenic lineages when cultured in specific inducing media. In parallel with their major role as undifferentiated cell reserves, MSCs have immunomodulatory functions which are exerted by direct cell-to-cell contacts, secretion of cytokines and/or by a combination of both mechanisms. There are no convincing data about a principal difference in the profile of cytokines secreted by MSCs isolated from different tissue sources, although some papers report some quantitative but not qualitative differences in cytokine secretion. The present review focuses on the basic cytokines secreted by MSCs as described in the literature by which the MSCs exert immunodulatory effects. It should be pointed out that MSCs themselves are objects of cytokine regulation. Hypothetical mechanisms by which the MSCs exert their immunoregulatory effects are also discussed in this review. These mechanisms may either influence the target immune cells directly or indirectly by affecting the activities of predominantly dendritic cells. Chemokines are also discussed as participants in this process by recruiting cells of the immune systems and thus making them targets of immunosuppression. This review aims to present and discuss the published data and the personal experience of the authors regarding cytokines secreted by MSCs and their effects on the cells of the immune system.
Collapse
|
20
|
MiRNome and transcriptome aided pathway analysis in human regulatory T cells. Genes Immun 2014; 15:303-12. [PMID: 24848933 DOI: 10.1038/gene.2014.20] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 03/24/2014] [Accepted: 03/27/2014] [Indexed: 12/15/2022]
Abstract
Owing to their manifold immune regulatory functions, regulatory T cells (Treg) have received tremendous interest as targets for therapeutic intervention of diverse immunological pathologies or cancer. Directed manipulation of Treg will only be achievable with extensive knowledge about the intrinsic programs that define their regulatory function. We simultaneously analyzed miR and mRNA transcript levels in resting and activated human Treg cells in comparison with non-regulatory conventional T cells (Tcon). Based on experimentally validated miR-target information, both transcript levels were integrated into a comprehensive pathway analysis. This strategy revealed characteristic signal transduction pathways involved in Treg biology such as T-cell receptor-, Toll-like receptor-, transforming growth factor-β-, JAK/STAT (Janus kinase/signal transducers and activators of transcription)- and mammalian target of rapamycin signaling, and allowed for the prediction of specific pathway activities on the basis of miR and mRNA transcript levels in a probabilistic manner. These data encourage new concepts for targeted control of Treg cell effector functions.
Collapse
|
21
|
Abstract
The cytokine TGF-β plays an integral role in regulating immune responses. TGF-β has pleiotropic effects on adaptive immunity, especially in the regulation of effector and regulatory CD4(+) T cell responses. Many immune and nonimmune cells can produce TGF-β, but it is always produced as an inactive complex that must be activated to exert functional effects. Thus, activation of latent TGF-β provides a crucial layer of regulation that controls TGF-β function. In this review, we highlight some of the important functional roles for TGF-β in immunity, focusing on its context-specific roles in either dampening or promoting T cell responses. We also describe how activation of TGF-β controls its function in the immune system, with a focus on the key roles for members of the integrin family in this process.
Collapse
Affiliation(s)
- Mark A Travis
- Manchester Collaborative Center for Inflammation Research
| | | |
Collapse
|
22
|
Spiering R, Wagenaar-Hilbers J, Huijgen V, van der Zee R, van Kooten PJS, van Eden W, Broere F. Membrane-Bound Metallothionein 1 of Murine Dendritic Cells Promotes the Expansion of Regulatory T Cells In Vitro. Toxicol Sci 2013; 138:69-75. [DOI: 10.1093/toxsci/kft268] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
23
|
The TGFβ1 pathway is required for NFκB dependent gene expression in mouse keratinocytes. Cytokine 2013; 64:652-9. [PMID: 24075100 DOI: 10.1016/j.cyto.2013.09.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Revised: 06/27/2013] [Accepted: 09/01/2013] [Indexed: 12/21/2022]
Abstract
The transforming growth factor-beta 1 (TGFβ1) and NFκB pathways are important regulators of epidermal homeostasis, inflammatory responses and carcinogenesis. Previous studies have shown extensive crosstalk between these pathways that is cell type and context dependent, but this has not been well-characterized in epidermal keratinocytes. Here we show that in primary mouse keratinocytes, TGFβ1 induces NFκB-luciferase reporter activity that is dependent on both NFκB and Smad3. TGFβ1-induced NFκB-luciferase activity was blocked by the IκB inhibitor parthenolide, the IκB super-repressor, a dominant negative TGFβ1-activated kinase 1 (TAK1) and genetic deletion of NFκB1. Coexpression of NFκB p50 or p65 subunits enhanced NFκB-luciferase activity. Similarly, inhibition of the TGFβ1 type I receptor with SB431542 or genetic deletion of Smad3 blocked TGFβ1 induction of NFκB-luciferase. TGFβ1 rapidly induced IKK phosphorylation but did not cause a detectable decrease in cytoplasmic IκB levels or nuclear translocation of NFκB subunits, although EMSA showed rapid NFκB nuclear binding activity that could be blocked by SB431542 treatment. TNFα, a well characterized NFκB target gene was also induced by TGFβ1 and this was blocked in NFκB+/- and -/- keratinocytes and by the IκB super-repressor. To test the effects of the TGFβ1 pathway on a biologically relevant activator of NFκB, we exposed mice and primary keratinocytes in culture to UVB irradiation. In primary keratinocytes UVB caused a detectable increase in levels of Smad2 phosphorylation that was dependent on ALK5, but no significant increase in SBE-dependent gene expression. Inhibition of TGFβ1 signaling in primary keratinocytes with SB431542 or genetic deletion of Tgfb1 or Smad3 suppressed UVB induction of TNFα message. Similarly, UVB induction of TNFα mRNA was blocked in skin of Tgfb1+/- mice. These studies demonstrate that intact TGFβ1 signaling is required for NFκB-dependent gene expression in mouse keratinocytes and skin and suggest that a convergence of these pathways in the nucleus rather than the cytoplasm may be critical for regulation of inflammatory pathways in skin by TGFβ1.
Collapse
|
24
|
Qiao G, Zhao Y, Li Z, Tang PQ, Langdon WY, Yang T, Zhang J. T cell activation threshold regulated by E3 ubiquitin ligase Cbl-b determines fate of inducible regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:632-9. [PMID: 23749633 DOI: 10.4049/jimmunol.1202068] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
E3 ubiquitin ligase Casitas-B-lineage lymphoma protein-b (Cbl-b) is critical for establishing the threshold for T cell activation and is essential for induction of T cell anergy. Recent studies suggest that Cbl-b is involved in the development of CD4(+)CD25(+) inducible regulatory T cells (iTregs). In this study, we report that the optimal induction of Foxp3 by naive CD4(+)CD25(-) T cells requires suboptimal TCR triggering. In the absence of Cbl-b, the TCR strength for optimal Foxp3 induction is downregulated in vitro. Using TCR-transgenic Rag(-/-) mice in combination with Cbl-b deficiency, we show that in vivo iTreg development is also controlled by Cbl-b via tuning the TCR strength. Furthermore, we show that Akt-2 but not Akt-1 regulates Foxp3 expression downstream of Cbl-b. Therefore, we demonstrate that Cbl-b regulates the fate of iTregs via controlling the threshold for T cell activation.
Collapse
Affiliation(s)
- Guilin Qiao
- Section of Nephrology, Department of Medicine, The University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Evaluation of the immunosuppressive activity of artesunate in vitro and in vivo. Int Immunopharmacol 2013; 16:306-12. [PMID: 23583335 DOI: 10.1016/j.intimp.2013.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 02/19/2013] [Accepted: 03/13/2013] [Indexed: 01/01/2023]
Abstract
Artemisinin and its derivatives have been reported to have immunosuppressive activity in some laboratory studies. However, the detail of mechanism remains to be demonstrated. The objective of this study is to clarify the immunosuppressive activity of artesunate (AST), one kind of artemisinin derivatives, and to find its unexplored mode of action. In vitro, the proliferation of T lymphocytes and its cytotoxicity were measured by WST-1 and MTT assay. In vivo, the immunomodulatory effect of AST was evaluated in a mouse model of delayed type hypersensitivity reaction (DTH), which was based on a T cell-mediated immune response. The data displayed that AST had a relatively high immunosuppressive activity with low toxicity, and could inhibit T lymphocyte proliferation induced by mitogen and alloantigen. Meanwhile, topical administration of AST could suppress DTH response significantly. Moreover, AST could also increase the secretion of TFG-β, coupling with the striking enhance of NF-κB/p65 and Smad2/3 signaling. The promotion of CD4(+)CD25(+) regulatory T cells (Tregs) was shown to be a possible mechanism involved in AST-mediated regulation. Taken together, these observations exhibit the potential of developing AST as a novel safe remedy for the treatment of T cell-mediated immune disorders.
Collapse
|
26
|
Reissig S, Hövelmeyer N, Weigmann B, Nikolaev A, Kalt B, Wunderlich TF, Hahn M, Neurath MF, Waisman A. The tumor suppressor CYLD controls the function of murine regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:4770-6. [PMID: 23066153 DOI: 10.4049/jimmunol.1201993] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
CYLD was originally identified as a tumor suppressor gene mutated in familial cylindromatosis, an autosomal dominant predisposition to multiple benign neoplasms of the skin known as cylindromas. The CYLD protein is a deubiquitinating enzyme that acts as a negative regulator of NF-κB and JNK signaling through its interaction with NEMO and TNFR-associated factor 2. We have previously described a novel mouse strain that expresses solely and excessively a naturally occurring splice variant of CYLD (CYLD(ex7/8)). In this study, we demonstrate that CYLD plays a critical role in Treg development and function. T cells of CYLD(ex7/8) mice had a hyperactive phenotype manifested by increased production of inflammatory cytokines and constitutive activation of the NF-κB pathway. Furthermore, the amount of Foxp3(+) regulatory T cells in these mice was markedly enhanced in thymus and peripheral organs. Importantly, these regulatory T cells displayed decreased expression levels of CD25 and CTLA-4 associated with impaired suppressive capacity. Hence, our data emphasize an essential role of CYLD in maintaining T cell homeostasis as well as normal T regulatory cell function, thereby controlling abnormal T cell responses.
Collapse
Affiliation(s)
- Sonja Reissig
- Institute for Molecular Medicine, Johannes Gutenberg-University of Mainz, 55131 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Immunomodulatory Effect of Nuclear Factor-κB Inhibition by Dehydroxymethylepoxyquinomicin in Combination With Donor-Specific Blood Transfusion. Transplantation 2012; 93:777-86. [DOI: 10.1097/tp.0b013e318248ca5f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
28
|
Zhang C, Zhang X, Chen XH. Hypothesis: Human Umbilical Cord Blood-Derived Stromal Cells Regulate the Foxp3 Expression of Regulatory T Cells Through the TGF-β1/Smad3 Pathway. Cell Biochem Biophys 2011; 62:463-6. [DOI: 10.1007/s12013-011-9328-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
29
|
Molinero LL, Alegre ML. Role of T cell-nuclear factor κB in transplantation. Transplant Rev (Orlando) 2011; 26:189-200. [PMID: 22074783 DOI: 10.1016/j.trre.2011.07.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/17/2011] [Accepted: 07/12/2011] [Indexed: 11/30/2022]
Abstract
Nuclear factor (NF) κB is a pleiotropic transcription factor that is ubiquitously expressed. After transplantation of solid organs, NF-κB in the graft is activated within a few hours as a consequence of ischemia/reperfusion and then again after a few days in intragraft infiltrating cells during the process of acute allograft rejection. In the present article, we review the components of the NF-κB pathway, their mechanisms of activation, and their role in T cell and antigen-presenting cell activation and differentiation and in solid organ allograft rejection. Targeted inhibition of NF-κB in selected cell types may promote graft survival with fewer adverse effects compared with global immunosuppressive therapies.
Collapse
Affiliation(s)
- Luciana L Molinero
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
30
|
Hahn JN, Falck VG, Jirik FR. Smad4 deficiency in T cells leads to the Th17-associated development of premalignant gastroduodenal lesions in mice. J Clin Invest 2011; 121:4030-42. [PMID: 21881210 DOI: 10.1172/jci45114] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2010] [Accepted: 07/06/2011] [Indexed: 12/11/2022] Open
Abstract
While there is evidence that specific T cell populations can promote the growth of established tumors, instances where T cell activity causes neoplasms to arise de novo are infrequent. Here, we employed two conditional mutagenesis systems to delete the TGF-β signaling pathway component Smad4 in T cells and observed the spontaneous development of massive polyps within the gastroduodenal regions of mice. The epithelial lesions contained increased levels of transcripts encoding IL-11, IL-6, TGF-β, IL-1β, and TNF-α, and lamina propria cells isolated from lesions contained abundant IL-17A+CD4+ T cells. Furthermore, we found that Smad4 deficiency attenuated TGF-β-mediated in vitro polarization of FoxP3+CD4+ T cells, but not IL-17A+CD4+ T cells, suggesting that the epithelial lesions may have arisen as a consequence of unchecked Th17 cell activity. Proinflammatory cytokine production likely accounted for the raised levels of IL-11, a cytokine known to promote gastric epithelial cell survival and hyperplasia. Consistent with IL-11 having a pathogenic role in this model, we found evidence of Stat3 activation in the gastric polyps. Thus, our data indicate that a chronic increase in gut Th17 cell activity can be associated with the development of premalignant lesions of the gastroduodenal region.
Collapse
Affiliation(s)
- Jennifer Nancy Hahn
- Department of Biochemistry and Molecular Biology, The McCaig Institute for Bone and Joint Health, University of Calgary, Calgary, Alberta, Canada
| | | | | |
Collapse
|
31
|
The suppressive effect of CD25+Treg cells on Th1 differentiation requires cell–cell contact partially via TGF-β production. Cell Biol Int 2011; 35:705-12. [DOI: 10.1042/cbi20100528] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Cell-intrinsic NF-κB activation is critical for the development of natural regulatory T cells in mice. PLoS One 2011; 6:e20003. [PMID: 21625598 PMCID: PMC3097234 DOI: 10.1371/journal.pone.0020003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2011] [Accepted: 04/08/2011] [Indexed: 12/21/2022] Open
Abstract
Background Naturally occurring CD4+CD25+Foxp3+ regulatory T (Treg) cells develop in the thymus and represent a mature T cell subpopulation critically involved in maintaining peripheral tolerance. The differentiation of Treg cells in the thymus requires T cell receptor (TCR)/CD28 stimulation along with cytokine-promoted Foxp3 induction. TCR-mediated nuclear factor kappa B (NF-κB) activation seems to be involved in differentiation of Treg cells because deletion of components of the NF-κB signaling pathway, as well as of NF-κB transcription factors, leads to markedly decreased Treg cell numbers in thymus and periphery. Methodology/Principal Findings To investigate if Treg cell-intrinsic NF-κB activation is required for thymic development and peripheral homeostasis of Treg cells we used transgenic (Tg) mice with thymocyte-specific expression of a stable IκBα mutant to inhibit NF-κB activation solely within the T cell lineage. Here we show that Treg cell-intrinsic NF-κB activation is important for the generation of cytokine-responsive Foxp3− thymic Treg precursors and their further differentiation into mature Treg cells. Treg cell development could neither be completely rescued by the addition of exogenous Interleukin 2 (IL-2) nor by the presence of wild-type derived cells in adoptive transfer experiments. However, peripheral NF-κB activation appears to be required for IL-2 production by conventional T cells, thereby participating in Treg cell homeostasis. Moreover, pharmacological NF-κB inhibition via the IκB kinase β (IKKβ) inhibitor AS602868 led to markedly diminished thymic and peripheral Treg cell frequencies. Conclusion/Significance Our results indicate that Treg cell-intrinsic NF-κB activation is essential for thymic Treg cell differentiation, and further suggest pharmacological NF-κB inhibition as a potential therapeutic approach for manipulating this process.
Collapse
|
33
|
Barbarulo A, Grazioli P, Campese AF, Bellavia D, Di Mario G, Pelullo M, Ciuffetta A, Colantoni S, Vacca A, Frati L, Gulino A, Felli MP, Screpanti I. Notch3 and canonical NF-kappaB signaling pathways cooperatively regulate Foxp3 transcription. THE JOURNAL OF IMMUNOLOGY 2011; 186:6199-206. [PMID: 21508258 DOI: 10.4049/jimmunol.1002136] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Notch3 overexpression has been previously shown to positively regulate the generation and function of naturally occurring regulatory T cells and the expression of Foxp3, in cooperation with the pTα/pre-TCR pathway. In this study, we show that Notch3 triggers the trans activation of Foxp3 promoter depending on the T cell developmental stage. Moreover, we discovered a novel CSL/NF-κB overlapping binding site within the Foxp3 promoter, and we demonstrate that the activation of NF-κB, mainly represented by p65-dependent canonical pathway, plays a positive role in Notch3-dependent regulation of Foxp3 transcription. Accordingly, the deletion of protein kinase C, which mediates canonical NF-κB activation, markedly reduces regulatory T cell number and per cell Foxp3 expression in transgenic mice with a constitutive activation of Notch3 signaling. Collectively, our data indicate that the cooperation among Notch3, protein kinase C, and p65/NF-κB subunit modulates Foxp3 expression, adding new insights in the understanding of the molecular mechanisms involved in regulatory T cell homeostasis and function.
Collapse
|
34
|
Benayoun BA, Caburet S, Veitia RA. Forkhead transcription factors: key players in health and disease. Trends Genet 2011; 27:224-32. [PMID: 21507500 DOI: 10.1016/j.tig.2011.03.003] [Citation(s) in RCA: 241] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/17/2011] [Accepted: 03/17/2011] [Indexed: 12/16/2022]
Abstract
Forkhead box (FOX) proteins constitute an evolutionarily conserved family of transcription factors with a central role not only during development, but also in the adult organism. Thus, the misregulation and/or mutation of FOX genes often induce human genetic diseases, promote cancer or deregulate ageing. Indeed, germinal FOX gene mutations cause diseases ranging from infertility to language and/or speech disorders and immunological defects. Moreover, because of their central role in signalling pathways and in the regulation of homeostasis, somatic misregulation and/or mutation of FOX genes are associated with cancer. FOX proteins have undergone diversification in terms of their sequence, regulation and function. In addition to dedicated roles, evidence suggests that Forkhead factors have retained some functional redundancy. Thus, combinations of slightly defective alleles might induce disease phenotypes in humans, acting as quantitative trait loci. Uncovering such variants would be a big step towards understanding the functional interdependencies of different FOX members and their implications in complex pathologies.
Collapse
Affiliation(s)
- Bérénice A Benayoun
- CNRS UMR 7592, Institut Jacques Monod, Equipe Génétique et Génomique du Développement Gonadique, 75205 Paris Cedex 13, France
| | | | | |
Collapse
|
35
|
Son JS, Sahoo A, Chae CS, Hwang JS, Park ZY, Im SH. JunB and c-Rel cooperatively enhance Foxp3 expression during induced regulatory T cell differentiation. Biochem Biophys Res Commun 2011; 407:141-7. [PMID: 21371435 DOI: 10.1016/j.bbrc.2011.02.126] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Accepted: 02/25/2011] [Indexed: 10/18/2022]
|
36
|
Molinero LL, Miller ML, Evaristo C, Alegre ML. High TCR stimuli prevent induced regulatory T cell differentiation in a NF-κB-dependent manner. THE JOURNAL OF IMMUNOLOGY 2011; 186:4609-17. [PMID: 21411734 DOI: 10.4049/jimmunol.1002361] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The concentration of Ag or mitogenic stimuli is known to play an important role in controlling the differentiation of naive CD4(+) T cells into different effector phenotypes. In particular, whereas TCR engagement at low Ag doses in the presence of TGF-β and IL-2 can promote differentiation of Foxp3-expressing induced regulatory T cells (iTregs), high levels of Ag have been shown in vitro and in vivo to prevent Foxp3 upregulation. This tight control of iTreg differentiation dictated by Ag dose most likely determines the quality and duration of an immune response. However, the molecular mechanism by which this high-dose inhibition of Foxp3 induction occurs is not well understood. In this study, we demonstrate that when cells are in the presence of CD28 costimulation, TCR-dependent NF-κB signaling is essential for Foxp3 inhibition at high doses of TCR engagement in mouse T cells. Prevention of Foxp3 induction depends on the production of NF-κB-dependent cytokines by the T cells themselves. Moreover, T cells that fail to upregulate Foxp3 under iTreg-differentiating conditions and high TCR stimulation acquire the capacity to make TNF and IFN-γ, as well as IL-17 and IL-9. Thus, NF-κB helps T cells control their differentiation fate in a cell-intrinsic manner and prevents peripheral iTreg development under conditions of high Ag load that may require more vigorous effector T cell responses.
Collapse
Affiliation(s)
- Luciana L Molinero
- Section of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | |
Collapse
|
37
|
Soligo M, Camperio C, Caristi S, Scottà C, Del Porto P, Costanzo A, Mantel PY, Schmidt-Weber CB, Piccolella E. CD28 costimulation regulates FOXP3 in a RelA/NF-κB-dependent mechanism. Eur J Immunol 2011; 41:503-13. [PMID: 21268019 DOI: 10.1002/eji.201040712] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Revised: 10/18/2010] [Accepted: 11/25/2010] [Indexed: 02/04/2023]
Abstract
The molecular mechanisms whereby CD28 alone or associated with TCR can regulate FOXP3 expression are not understood, although the importance of CD28 as a pivotal regulator of CD4(+) CD25(+) FOXP3(+) T cells is well recognized. We previously demonstrated that unique CD28-induced, NF-κB-dependent signals were sufficient to activate FOXP3 transcription in human CD4(+) CD25(-) T cells; however, the exact mechanisms are currently unknown. In this study, we have identified novel κB-binding sites on FOXP3 gene and demonstrated that CD28 signals mediated FOXP3 trans activation by nuclear translocation of RelA/NF-κB and not of c-Rel. The occupancy of FOXP3 κB-binding sites by RelA dimers that correlated with histone acetylation and recruitment of Pol II were required both to initiate FOXP3 transcription and to control the promoter occupancy by NFAT. Interestingly, knockdown of RelA in CD4(+) CD25(-) T cells stimulated through TCR and CD28 significantly affected FOXP3 expression, confirming that also the transcriptional activation of FOXP3 gene by TCR in the presence of CD28-costimulatory signals is RelA-dependent. In conclusion, these data suggest a new mechanism by which FOXP3 is activated and supports the critical role of CD28 in the regulation of peripheral tolerance.
Collapse
Affiliation(s)
- Marzia Soligo
- Department of Biology and Biotechnology C. Darwin, University Sapienza of Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Transcriptional regulation of Foxp3 in regulatory T cells. Immunobiology 2010; 216:678-85. [PMID: 21122941 DOI: 10.1016/j.imbio.2010.11.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 11/05/2010] [Accepted: 11/05/2010] [Indexed: 12/30/2022]
Abstract
Regulatory T (Treg) cells constitute a unique T-cell lineage that plays a pivotal role in the maintenance of the peripheral tolerance. The transcription factor Foxp3 (Forkhead box P3) was identified as a master regulator for the development and function of Treg cells. It is well defined that Foxp3 expression is critical to program CD4+CD25+ Treg cell development and function; however, the molecular mechanisms that are involved in the regulation of the Foxp3 expression remain unclear. Recent studies have showed an indication that this process is influenced by a number of transcription factors. In this review, we summarize the current knowledge of how Foxp3 expression is controlled at molecular level by focusing on these factors.
Collapse
|
39
|
Malhotra N, Robertson E, Kang J. SMAD2 is essential for TGF beta-mediated Th17 cell generation. J Biol Chem 2010; 285:29044-8. [PMID: 20656683 DOI: 10.1074/jbc.c110.156745] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TGFβ is the quintessential cytokine of T cell homeostasis. TGFβ signaling is required for the efficient differentiation and maintenance of CD4(+)FOXP3(+) T cells that inhibit immune responses. Conversely, in conjunction with the inflammatory cytokine IL-6, TGFβ promotes Th17 cell differentiation. The mechanism by which TGFβ signals synergize with IL-6 to generate inflammatory versus immunosuppressive T cell subsets is unclear. TGFβ signaling activates receptor SMADs, SMAD2 and SMAD3, which associate with a variety of nuclear factors to regulate gene transcription. Defining relative contributions of distinct SMAD molecules for CD4 T cell differentiation is critical for mapping the versatile intracellular TGFβ-signaling pathways that tailor TGFβ activities to the state of host interaction with pathogens. We show here that SMAD2 is essential for Th17 cell differentiation and that it acts in part by modulating the expression of IL-6R on T cells. Although mice lacking SMAD2 specifically in T cells do not develop spontaneous lymphoproliferative autoimmunity, Smad2-deficient T cells are impaired in their response to TGFβ in vitro and in vivo, and they are more pathogenic than controls when transferred into lymphopenic mice. These results demonstrate that SMAD2 is uniquely essential for TGFβ signaling in CD4(+) T effector cell differentiation.
Collapse
Affiliation(s)
- Nidhi Malhotra
- Department of Pathology, Graduate Program in Immunology and Virology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | |
Collapse
|
40
|
Eckerstorfer P, Novy M, Burgstaller-Muehlbacher S, Paster W, Schiller HB, Mayer H, Stockinger H. Proximal human FOXP3 promoter transactivated by NF-kappaB and negatively controlled by feedback loop and SP3. Mol Immunol 2010; 47:2094-102. [PMID: 20462637 PMCID: PMC6340486 DOI: 10.1016/j.molimm.2010.04.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 03/31/2010] [Accepted: 04/09/2010] [Indexed: 12/29/2022]
Abstract
Forkhead box protein 3 (Foxp3) is indispensable for the development of CD4(+)CD25(+) regulatory T cells (Tregs). Here we analyzed three prominent evolutionary conserved regions (ECRs) upstream of the transcription start site of the human FOXP3 gene. We show that ECR2 and ECR3 fragments derived from positions -1.3 to -2.0 kb and -5.0 to -6.0 kb, respectively, display basal transcriptional activity. Reporter constructs derived from ECR1, located between -0.6 and +0.23 kb and thus the most proximal ECR in respect of transcription initiation, remained almost inactive. However, ECR1 was transactivated by the NF-kappaB subunit p65 in HEK 293 cells. In Jurkat and primary T cells, in addition to p65, a second stimulus delivered by either T-cell receptor stimulation or addition of PMA was needed. Co-expression of I kappaB alpha inhibited p65-mediated FOXP3 proximal promoter transactivation, and the NF-kappaB inhibitor curcumin reduced Foxp3 neoexpression in IL-2/CD3/CD28/TGF-beta stimulated PBMCs. Moreover, proximal FOXP3 promoter transactivation was inhibited by Foxp3 and the SP transcription factor family member SP3. Thus, the human proximal FOXP3 promoter is controlled by activation through the TCR involving PKC and the NF-kappaB subunit p65 and by inhibition through a negative feedback loop and SP3.
Collapse
Affiliation(s)
- Paul Eckerstorfer
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Michael Novy
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Sebastian Burgstaller-Muehlbacher
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Wolfgang Paster
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Herbert B. Schiller
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Herbert Mayer
- Competence Center for Biomolecular Therapeutics-Research Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| | - Hannes Stockinger
- Molecular Immunology Unit, Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
- Competence Center for Biomolecular Therapeutics-Research Vienna, Lazarettgasse 19, A-1090 Vienna, Austria
| |
Collapse
|
41
|
Lu L, Wang J, Zhang F, Chai Y, Brand D, Wang X, Horwitz DA, Shi W, Zheng SG. Role of SMAD and non-SMAD signals in the development of Th17 and regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:4295-306. [PMID: 20304828 DOI: 10.4049/jimmunol.0903418] [Citation(s) in RCA: 158] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Whereas TGF-beta is essential for the development of peripherally induced Foxp3(+) regulatory T cells (iTreg cells) and Th17 cells, the intracellular signaling mechanism by which TGF-beta regulates development of both cell subsets is less understood. In this study, we report that neither Smad2 nor Smad3 gene deficiency abrogates TGF-beta-dependent iTreg induction by a deacetylase inhibitor trichostatin A in vivo, although the loss of the Smad2 or Smad3 gene partially reduces iTreg induction in vitro. Similarly, SMAD2 and SMAD3 have a redundant role in development of Th17 in vitro and in experimental autoimmune encephalomyelitis. In addition, ERK and/or JNK pathways were shown to be involved in regulating iTreg cells, whereas the p38 pathway predominately modulated Th17 and experimental autoimmune encephalomyelitis induction. Therefore, selective targeting of these intracellular TGF-beta signaling pathways during iTreg and Th17 cell development might lead to the development of therapies in treating autoimmune and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ling Lu
- Division of Rheumatology, Department of Medicine, Saban Research Institute, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Lee AJ, Wu X, Cheng H, Zhou X, Cheng X, Sun SC. CARMA1 regulation of regulatory T cell development involves modulation of interleukin-2 receptor signaling. J Biol Chem 2010; 285:15696-703. [PMID: 20233721 DOI: 10.1074/jbc.m109.095190] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
T cell receptor-stimulated NF-kappaB activation requires CARMA1 and is negatively regulated by the deubiquitinase CYLD. Recent studies suggest that CARMA1 regulates regulatory T cell (Treg) development, although the role of NF-kappaB in this event is incompletely understood. We show that CYLD deficiency causes constitutive NF-kappaB activation in thymocytes, which is associated with enhanced frequency of Treg cells. The NF-kappaB activation in CYLD-deficient thymocytes is independent of CARMA1, because the NF-kappaB activation was also detected in CYLD/CARMA1 double knock-out thymocytes. Interestingly, although loss of CYLD causes NF-kappaB activation in the CARMA1-deficient thymocytes, the CYLD deficiency fails to rescue the defect of CARMA1 knock-out mice in Treg development. Furthermore, inhibition of canonical NF-kappaB by an IkappaBalpha transgene only partially inhibits Treg development. We demonstrate that CARMA1 regulates IL-2 receptor signaling and controls the IL-2-stimulated maturation of Treg precursors to mature Tregs. These results suggest that the role of CARMA1 in Treg regulation involves both NF-kappaB activation and IL-2 receptor signaling.
Collapse
Affiliation(s)
- Andrew Joon Lee
- Department of Immunology, The University of Texas MD Anderson Cancer Center, University of Texas, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
43
|
Nagar M, Jacob-Hirsch J, Vernitsky H, Berkun Y, Ben-Horin S, Amariglio N, Bank I, Kloog Y, Rechavi G, Goldstein I. TNF Activates a NF-κB–Regulated Cellular Program in Human CD45RA– Regulatory T Cells that Modulates Their Suppressive Function. THE JOURNAL OF IMMUNOLOGY 2010; 184:3570-81. [DOI: 10.4049/jimmunol.0902070] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|