1
|
Krevh R, Wang J, Zuniga B, Toor J, Subedi K, Zhou L, Mi QS. TAK1 is essential for MAIT cell development and the differentiation of MAIT1 and MAIT17. Cell Mol Immunol 2023; 20:854-856. [PMID: 36973488 PMCID: PMC10310715 DOI: 10.1038/s41423-023-00999-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Affiliation(s)
- Rachel Krevh
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
| | - Jie Wang
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
| | - Bobby Zuniga
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
| | - Jugmohit Toor
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
| | - Kalpana Subedi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA
| | - Li Zhou
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA.
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA.
- Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA.
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology Research, Department of Dermatology, Henry Ford Health, Detroit, MI, USA.
- Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health, Detroit, MI, USA.
- Department of Internal Medicine, Henry Ford Health, Detroit, MI, USA.
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
2
|
Lee HS, Kim EN, Jeong GS. Oral administration of Helianthus annuus leaf extract ameliorates atopic dermatitis by modulation of T cell activity in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154443. [PMID: 36108372 DOI: 10.1016/j.phymed.2022.154443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 08/08/2022] [Accepted: 09/04/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Atopic dermatitis (AD) is multifactorial disease that is highly involved in the activity of T cells from the skin lesion. Seeds of Helianthus annuus extract have been traditionally used as anti-inflammatory reagent but few studies have been reported on leaf of H. annuus that are discarded uselessly as an immunomodulator. PURPOSE Therefore, here, the regulatory effect of Helianthus annuus extract (HAE) on AD via suppression of T cell activity was investigated. METHODS The efficacy of HAE was evaluated in T cells stimulated with CD3/CD28 antibody and PMA/A23187. And demonstration of the alleviating effect of HAE on AD in the ears of Balb/c female mice stimulated with mite extract and DNCB. RESULTS Pre-treatment with HAE abrogates IL-2 production from activated T cells. It was also found that HAE suppresses the expression of surface molecules in activated T cells. Cell viability results demonstrated that HAE is not associated with cytotoxicity in resting and activated T cells. Besides, we exhibited that regulated phosphorylation of MAPK through TAK1-IKKα-NFκB by pre-treatment with HAE leads to the suppressive effect of HAE on T cell activation. Oral administration of HAE attenuates manifestations of AD including reduced thickness of dermis and epidermis, decreased IgE level in serum, and declined mRNA levels of atopic cytokines on ear tissues. The ameliorative effect of HAE on AD was found to be associated with suppressed activity of T cells from draining lymph nodes. CONCLUSION Therefore, our results provide that HAE alleviates AD symptoms via modulation of T cell activity. In addition, these results suggest the immunomodulatory effect of HAE on T-cell mediated diseases.
Collapse
Affiliation(s)
- Hyun-Su Lee
- Department of Physiology, Daegu Catholic University School of Medicine, Daegu, Republic of Korea
| | - Eun-Nam Kim
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Gil-Saeng Jeong
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea.
| |
Collapse
|
3
|
Zhou M, Ren X, Yan X, Sun Y, Xu T. Rho-GDP-dissociation inhibitor-γ negatively regulates NF-κB signaling by promoting the degradation of TAK1 in miiuy croaker (Miichthys miiuy). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2022; 135:104496. [PMID: 35870543 DOI: 10.1016/j.dci.2022.104496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/03/2022] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Transforming growth factor-beta activated kinase 1 (TAK1) is an adaptor molecular in TLR-mediated NF-κB signaling pathway and plays indispensable roles in innate immunity. As the most typical innate immune pathway, the strict regulation of NF-κB signaling pathway is particularly important. Rho-GDP-dissociation inhibitor-γ (Rho-GDIγ) is a member of the Rho protein family that regulates many important physiological processes. In this study, we demonstrated the mechanism of suppressing TAK1 expression in the teleost and found that Rho-GDIγ negatively regulated the NF-κB signaling pathway mediated by TAK1. We determined that TAK1 could directly interact with Rho-GDIγ. It is interesting that Rho-GDIγ promotes TAK1 degradation through the ubiquitin proteasome pathway. This study brings a new experimental basis to the teleost fish innate immune signaling pathway. Moreover, this discovery may provide new insights into innate immune regulation mechanism in mammals.
Collapse
Affiliation(s)
- Ming Zhou
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaomeng Ren
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Xiaolong Yan
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai, 201306, China.
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, 201306, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266200, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai, 201306, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
4
|
Prospect of ULK1 modulators in targeting regulatory T cells. Bioorg Chem 2022; 129:106141. [PMID: 36137312 DOI: 10.1016/j.bioorg.2022.106141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 08/11/2022] [Accepted: 09/05/2022] [Indexed: 11/20/2022]
Abstract
Regulatory T (Treg) cells play an instrumental role in coordinating immune homeostasis via potent inhibitory effects. Defects in Treg cells lead to autoimmunity, but an overwhelming proportion of Treg cells encourages cancer progression. Hence, targeting Treg cells has emerged as a promising approach for mitigating disease severity. Recent studies have revealed that kinases are a critical component for tuning the fate of Treg cells, but the entire network of Treg-modulating kinases is still unclear. Here, we propose that the autophagy-activating UNC-51-like kinase 1 (ULK1) is a candidate for Treg cell modulation. While accumulating evidence has highlighted the role of autophagy-related kinases in Treg cells, the ULK1-Treg cell axis is yet to be examined. In this review, we predicted the potential role of ULK1 in Treg cell modulation. Furthermore, we summarized current ULK1 activators and inhibitors that can be investigated as Treg-targeting strategies, which might have beneficial outcomes in autoimmunity and cancer.
Collapse
|
5
|
Chuckran CA, Cillo AR, Moskovitz J, Overacre-Delgoffe A, Somasundaram AS, Shan F, Magnon GC, Kunning SR, Abecassis I, Zureikat AH, Luketich J, Pennathur A, Sembrat J, Rojas M, Merrick DT, Taylor SE, Orr B, Modugno F, Buckanovich R, Schoen RE, Kim S, Duvvuri U, Zeh H, Edwards R, Kirkwood JM, Coffman L, Ferris RL, Bruno TC, Vignali DAA. Prevalence of intratumoral regulatory T cells expressing neuropilin-1 is associated with poorer outcomes in patients with cancer. Sci Transl Med 2021; 13:eabf8495. [PMID: 34878821 PMCID: PMC9022491 DOI: 10.1126/scitranslmed.abf8495] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite the success of immune checkpoint blockade therapy, few strategies sufficiently overcome immunosuppression within the tumor microenvironment (TME). Targeting regulatory T cells (Tregs) is challenging, because perturbing intratumoral Treg function must be specific enough to avoid systemic inflammatory side effects. Thus, no Treg-targeted agents have proven both safe and efficacious in patients with cancer. Neuropilin-1 (NRP1) is recognized for its role in supporting intratumoral Treg function while being dispensable for peripheral homeostasis. Nonetheless, little is known about the biology of human NRP1+ Tregs and the signals that regulate NRP1 expression. Here, we report that NRP1 is preferentially expressed on intratumoral Tregs across six distinct cancer types compared to healthy donor peripheral blood [peripheral blood lymphocyte (PBL)] and site-matched, noncancer tissue. Furthermore, NRP1+ Treg prevalence is associated with reduced progression-free survival in head and neck cancer. Human NRP1+ Tregs have broad activation programs and elevated suppressive function. Unlike mouse Tregs, we demonstrate that NRP1 identifies a transient activation state of human Tregs driven by continuous T cell receptor (TCR) signaling through the mitogen-activated protein kinase pathway and interleukin-2 exposure. The prevalence of NRP1+ Tregs in patient PBL correlates with the intratumoral abundance of NRP1+ Tregs and may indicate higher disease burden. These findings support further clinical evaluation of NRP1 as a suitable therapeutic target to enhance antitumor immunity by inhibiting Treg function in the TME.
Collapse
Affiliation(s)
- Christopher A. Chuckran
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, 200 Lothrop St., Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Anthony R. Cillo
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Jessica Moskovitz
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Abigail Overacre-Delgoffe
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Ashwin S. Somasundaram
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Feng Shan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Integrative Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Grant C. Magnon
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Sheryl R. Kunning
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Irina Abecassis
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Amer H. Zureikat
- Department of Surgery, Division of Surgical Oncology, UPMC Hillman Cancer Center and UPMC Pancreatic Cancer Program, Pittsburgh, PA 15213, USA
| | - James Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Arjun Pennathur
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - John Sembrat
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Mauricio Rojas
- Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Daniel T. Merrick
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah E. Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Brian Orr
- Department Obstetrics and Gynecology, Gynecologic Oncology Division, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Francesmary Modugno
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Women’s Cancer Research Center, Magee-Women’s Research Institute and Foundation and Hillman Cancer Center and Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ron Buckanovich
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Robert E. Schoen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Seungwon Kim
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Umamaheswar Duvvuri
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Herbert Zeh
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern, Dallas, TX 75390, USA
| | - Robert Edwards
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - John M. Kirkwood
- Departments of Medicine, Dermatology, and Translational Science, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Lan Coffman
- Division of Hematology/Oncology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Robert L. Ferris
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Tullia C. Bruno
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Dario A. A. Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
6
|
Hou X, Zhu F, Ni Y, Chen T, Du J, Liu X, Han Y, Liu Y, Du W, Li Y, Wang X, Li D, Liang R, Li B, Shi G. USP4 is pathogenic in allergic airway inflammation by inhibiting regulatory T cell response. Life Sci 2021; 281:119720. [PMID: 34144056 DOI: 10.1016/j.lfs.2021.119720] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/05/2021] [Accepted: 06/05/2021] [Indexed: 11/17/2022]
Abstract
AIMS Asthma is characterized by chronic inflammation and airway hyperresponsiveness (AHR). It is controllable, but not curable. Ubiquitin-specific peptidase 4 (USP4) has been verified as a regulator of regulatory T (Treg) cells and Th17 cells in vitro. In this study, we aim to investigate whether USP4 could serve as a therapeutic target for asthma. MAIN METHODS Age-matched USP4 wild-type and knockout mice received an intraperitoneal injection of 100 μg ovalbumin (OVA) mixed in 2 mg aluminum hydroxide in 1 × PBS on days 0, 7 and 14. On days 21 to 27, the mice were challenged with aerosolized 1% OVA in 1 × PBS for 30 min. Tissue histology, ELISA and flow cytometry were applied 24 h after the last OVA challenge. KEY FINDINGS USP4 deficiency protected mice from OVA-induced AHR and decreased the production of several inflammatory cytokines in T cells in vivo. Compared to the lung cells isolated from WT mice, Usp4-/- lung cells decreased secretion of IL-4, IL-13 and IL-17A upon stimulation in vitro. Meanwhile, the percentage of CD4+Foxp3+ Treg cells was elevated, with more CCR6+Foxp3+ Treg cells accumulating in the lungs of OVA-challenged USP4 deficient mice than in their wild-type counterparts. Treatment with the USP4 inhibitor, Vialinin A, reduced inflammatory cell infiltration in the lungs of OVA-challenged mice in vivo. SIGNIFICANCE We found USP4 deficiency contributes to attenuated airway inflammation and AHR in allergen-induced murine asthma, and Vialinin A treatment alleviates asthma pathogenesis and may serve as a promising therapeutic target for asthma.
Collapse
Affiliation(s)
- Xiaoxia Hou
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangming Zhu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Institute Shanghai Key Laboratory of Bio-Energy Crops, School of Life Science, Shanghai University, Shanghai, China
| | - Yingmeng Ni
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiantian Chen
- Department of Pulmonary and Critical Care Medicine, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Du
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinnan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yichao Han
- Department of Thoracic Surgery, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yahui Liu
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Du
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangyang Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxia Wang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dan Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Liang
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Guochao Shi
- Department of Pulmonary and Critical Care Medicine, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Institute of Respiratory Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
7
|
Fulford TS, Grumont R, Wirasinha RC, Ellis D, Barugahare A, Turner SJ, Naeem H, Powell D, Lyons PA, Smith KGC, Scheer S, Zaph C, Klein U, Daley SR, Gerondakis S. c-Rel employs multiple mechanisms to promote the thymic development and peripheral function of regulatory T cells in mice. Eur J Immunol 2021; 51:2006-2026. [PMID: 33960413 DOI: 10.1002/eji.202048900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 03/12/2021] [Accepted: 05/05/2021] [Indexed: 01/03/2023]
Abstract
The NF-κB transcription factor c-Rel is a critical regulator of Treg ontogeny, controlling multiple points of the stepwise developmental pathway. Here, we found that the thymic Treg defect in c-Rel-deficient (cRel-/- ) mice is quantitative, not qualitative, based on analyses of TCR repertoire and TCR signaling strength. However, these parameters are altered in the thymic Treg-precursor population, which is also markedly diminished in cRel-/- mice. Moreover, c-Rel governs the transcriptional programme of both thymic and peripheral Tregs, controlling a core of genes involved with immune signaling, and separately in the periphery, cell cycle progression. Last, the immune suppressive function of peripheral cRel-/- tTregs is diminished in a lymphopenic model of T cell proliferation and is associated with decreased stability of Foxp3 expression. Collectively, we show that c-Rel is a transcriptional regulator that controls multiple aspects of Treg development, differentiation, and function via distinct mechanisms.
Collapse
Affiliation(s)
- Thomas S Fulford
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Raelene Grumont
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Rushika C Wirasinha
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Darcy Ellis
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Adele Barugahare
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia.,Monash Bioinformatics Platform, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - Stephen J Turner
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia.,Department of Microbiology, Monash University, Melbourne, Australia
| | - Haroon Naeem
- Monash Bioinformatics Platform, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - David Powell
- Monash Bioinformatics Platform, School of Biomedical Sciences, Monash University, Melbourne, Australia
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, England, UK.,Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge, England, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, England, UK.,Department of Medicine, University of Cambridge, University of Cambridge School of Clinical Medicine, Cambridge, England, UK
| | - Sebastian Scheer
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Colby Zaph
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Ulf Klein
- Division of Haematology & Immunology, Leeds Institute of Medical Research at St. James's, University of Leeds, Leeds, LS2 7TF
| | - Stephen R Daley
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Steve Gerondakis
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| |
Collapse
|
8
|
Xu YR, Lei CQ. TAK1-TABs Complex: A Central Signalosome in Inflammatory Responses. Front Immunol 2021; 11:608976. [PMID: 33469458 PMCID: PMC7813674 DOI: 10.3389/fimmu.2020.608976] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-β (TGF-β)-activated kinase 1 (TAK1) is a member of the MAPK kinase kinase (MAPKKK) family and has been implicated in the regulation of a wide range of physiological and pathological processes. TAK1 functions through assembling with its binding partners TAK1-binding proteins (TAB1, TAB2, and TAB3) and can be activated by a variety of stimuli such as tumor necrosis factor α (TNFα), interleukin-1β (IL-1β), and toll-like receptor ligands, and they play essential roles in the activation of NF-κB and MAPKs. Numerous studies have demonstrated that post-translational modifications play important roles in properly controlling the activity, stability, and assembly of TAK1-TABs complex according to the indicated cellular environment. This review focuses on the recent advances in TAK1-TABs-mediated signaling and the regulations of TAK1-TABs complex by post-translational modifications.
Collapse
Affiliation(s)
- Yan-Ran Xu
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Cao-Qi Lei
- Hubei Key Laboratory of Cell Homeostasis, Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
9
|
Blanchett S, Boal-Carvalho I, Layzell S, Seddon B. NF-κB and Extrinsic Cell Death Pathways - Entwined Do-or-Die Decisions for T cells. Trends Immunol 2020; 42:76-88. [PMID: 33246882 DOI: 10.1016/j.it.2020.10.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/09/2020] [Accepted: 10/27/2020] [Indexed: 12/23/2022]
Abstract
NF-κB signaling is required at multiple stages of T cell development and function. The NF-κB pathway integrates signals from many receptors and involves diverse adapters and kinases. Recent advances demonstrate that kinases controlling NF-κB activation, such as the IKK complex, serve dual independent functions because they also control cell death checkpoints. Survival functions previously attributed to NF-κB are in fact mediated by these upstream kinases by novel mechanisms. This new understanding has led to a refined view of how NF-κB and cell death signaling are interlinked and how they regulate cell fate. We discuss how NF-κB activation and control of cell death signaling by common upstream triggers cooperate to regulate different aspects of T cell development and function.
Collapse
Affiliation(s)
- Sam Blanchett
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Ines Boal-Carvalho
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Scott Layzell
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK
| | - Benedict Seddon
- Institute of Immunity and Transplantation, Division of Infection and Immunity, University College London, Royal Free Hospital, Rowland Hill Street, London NW3 2PF, UK.
| |
Collapse
|
10
|
Li C, Jiang P, Wei S, Xu X, Wang J. Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects. Mol Cancer 2020; 19:116. [PMID: 32680511 PMCID: PMC7367382 DOI: 10.1186/s12943-020-01234-1] [Citation(s) in RCA: 516] [Impact Index Per Article: 103.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) characterized by the expression of the master transcription factor forkhead box protein p3 (Foxp3) suppress anticancer immunity, thereby hindering protective immunosurveillance of tumours and hampering effective antitumour immune responses in tumour-bearing hosts, constitute a current research hotspot in the field. However, Tregs are also essential for the maintenance of the immune tolerance of the body and share many molecular signalling pathways with conventional T cells, including cytotoxic T cells, the primary mediators of tumour immunity. Hence, the inability to specifically target and neutralize Tregs in the tumour microenvironment without globally compromising self-tolerance poses a significant challenge. Here, we review recent advances in characterizing tumour-infiltrating Tregs with a focus on the functional roles of costimulatory and inhibitory receptors in Tregs, evaluate their potential as clinical targets, and systematically summarize their roles in potential treatment strategies. Also, we propose modalities to integrate our increasing knowledge on Tregs phenotype and function for the rational design of checkpoint inhibitor-based combination therapies. Finally, we propose possible treatment strategies that can be used to develop Treg-targeted therapies.
Collapse
Affiliation(s)
- Chunxiao Li
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| | - Ping Jiang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Shuhua Wei
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaofei Xu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Key Laboratory of Assisted Reproduction, Ministry of Education, Peking University, Beijing, 100191, China
| | - Junjie Wang
- Department of Radiation Oncology, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
11
|
Joetham A, Schedel M, Ning F, Wang M, Takeda K, Gelfand EW. Dichotomous role of TGF-β controls inducible regulatory T-cell fate in allergic airway disease through Smad3 and TGF-β-activated kinase 1. J Allergy Clin Immunol 2020; 145:933-946.e4. [PMID: 31626843 PMCID: PMC11098441 DOI: 10.1016/j.jaci.2019.09.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/13/2019] [Accepted: 09/27/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Inducible CD4+CD25+ regulatory T (iTreg) cells can become pathogenic effector cells, enhancing lung allergic responses. OBJECTIVE We aimed to define the underlying cellular and molecular pathways activated by TGF-β, which determine the suppressor or enhancing activities of iTreg cells. METHODS Sensitized wild-type and CD8-deficient (CD8-/-) mice were challenged with allergen. Isolated CD4+CD25- T cells were activated by using anti-CD3/anti-CD28. To generate suppressor iTreg cells, cells were then differentiated in the presence of TGF-β, whereas IL-17-producing effector T cells were additionally exposed to IL-6. After TGF-β, Smad3 and TGF-β-activated kinase 1 (TAK1) kinase levels were monitored. The consequences of inhibiting either kinase were determined in vitro and after transfer into CD8-/- recipients. Quantitative PCR and chromatin immunoprecipitation were used to monitor gene expression and histone modifications at the retinoic acid-related orphan receptor γt (Rorγt) locus. RESULTS In wild-type mice, iTreg cells suppressed lung allergic responses linked to Smad3-dependent forkhead box P3 (Foxp3) expression and IL-10 production. In the presence of IL-6, iTreg cells converted to TH17 cells, mediating a neutrophil-dependent enhancement of lung allergic responses in CD8-/- mice. Conversion was regulated by TAK1. Inhibition or silencing of TAK1 prevented expression of Rorγt and TH17 differentiation through histone modifications of Rorγt; Foxp3 expression and iTreg cell-mediated suppression remained intact. In the same cell, TGF-β induced coexpression of Smad3 and TAK1 proteins; in the presence of IL-6, expression of Smad3 and Foxp3 but not TAK1 decreased. CONCLUSION TGF-β regulates iTreg cell outcomes through 2 distinct signal transduction pathways: one Smad3 dependent and the other TAK1 dependent. The balance of these pathways has important implications in TH17-mediated autoimmune diseases and neutrophil-dependent asthma.
Collapse
Affiliation(s)
- Anthony Joetham
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Michaela Schedel
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Fangkun Ning
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Meiqin Wang
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Katsuyuki Takeda
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo
| | - Erwin W Gelfand
- Division of Cell Biology, Department of Pediatrics, National Jewish Health, Denver, Colo.
| |
Collapse
|
12
|
Cheng L, Deng N, Yang N, Zhao X, Lin X. Malt1 Protease Is Critical in Maintaining Function of Regulatory T Cells and May Be a Therapeutic Target for Antitumor Immunity. THE JOURNAL OF IMMUNOLOGY 2019; 202:3008-3019. [DOI: 10.4049/jimmunol.1801614] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 03/14/2019] [Indexed: 01/17/2023]
|
13
|
Fleskens V, Minutti CM, Wu X, Wei P, Pals CEGM, McCrae J, Hemmers S, Groenewold V, Vos HJ, Rudensky A, Pan F, Li H, Zaiss DM, Coffer PJ. Nemo-like Kinase Drives Foxp3 Stability and Is Critical for Maintenance of Immune Tolerance by Regulatory T Cells. Cell Rep 2019; 26:3600-3612.e6. [PMID: 30917315 PMCID: PMC6444001 DOI: 10.1016/j.celrep.2019.02.087] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/06/2018] [Accepted: 02/21/2019] [Indexed: 12/22/2022] Open
Abstract
The Foxp3 transcription factor is a crucial determinant of both regulatory T (TREG) cell development and their functional maintenance. Appropriate modulation of tolerogenic immune responses therefore requires the tight regulation of Foxp3 transcriptional output, and this involves both transcriptional and post-translational regulation. Here, we show that during T cell activation, phosphorylation of Foxp3 in TREG cells can be regulated by a TGF-β activated kinase 1 (TAK1)-Nemo-like kinase (NLK) signaling pathway. NLK interacts and phosphorylates Foxp3 in TREG cells, resulting in the stabilization of protein levels by preventing association with the STUB1 E3-ubiquitin protein ligase. Conditional TREG cell NLK-knockout (NLKΔTREG) results in decreased TREG cell-mediated immunosuppression in vivo, and NLK-deficient TREG cell animals develop more severe experimental autoimmune encephalomyelitis. Our data suggest a molecular mechanism, in which stimulation of TCR-mediated signaling can induce a TAK1-NLK pathway to sustain Foxp3 transcriptional activity through the stabilization of protein levels, thereby maintaining TREG cell suppressive function.
Collapse
Affiliation(s)
- Veerle Fleskens
- Center for Molecular Medicine, Division of Pediatrics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Carlos M Minutti
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
| | - Xingmei Wu
- ENT Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Ping Wei
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, 136 Zhongshaner Road, Chongqing 400014, China
| | - Cornelieke E G M Pals
- Center for Molecular Medicine, Division of Pediatrics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - James McCrae
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK
| | - Saskia Hemmers
- Immunology Program, Howard Hughes Medical Institute, and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vincent Groenewold
- Hubrecht Institute, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Harm-Jan Vos
- Proteins at Work, UMC Utrecht, Utrecht, the Netherlands
| | - Alexander Rudensky
- Immunology Program, Howard Hughes Medical Institute, and Ludwig Center, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fan Pan
- Immunology and Hematopoiesis Division, Department of Oncology, Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Huabin Li
- ENT Department, Affiliated Eye and ENT Hospital, Fudan University, Shanghai, China.
| | - Dietmar M Zaiss
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, Edinburgh, UK.
| | - Paul J Coffer
- Center for Molecular Medicine, Division of Pediatrics, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands; Regenerative Medicine Center, University Medical Centre Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
14
|
Prolonged IKKβ Inhibition Improves Ongoing CTL Antitumor Responses by Incapacitating Regulatory T Cells. Cell Rep 2018; 21:578-586. [PMID: 29045828 DOI: 10.1016/j.celrep.2017.09.082] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 09/05/2017] [Accepted: 09/25/2017] [Indexed: 01/28/2023] Open
Abstract
Regulatory T cells (Tregs) prevent autoimmunity but limit antitumor immunity. The canonical NF-κB signaling pathway both activates immunity and promotes thymic Treg development. Here, we report that mature Tregs continue to require NF-κB signaling through IκB-kinase β (IKKβ) after thymic egress. Mice lacking IKKβ in mature Tregs developed scurfy-like immunopathology due to death of peripheral FoxP3+ Tregs. Also, pharmacological IKKβ inhibition reduced Treg numbers in the circulation by ∼50% and downregulated FoxP3 and CD25 expression and STAT5 phosphorylation. In contrast, activated cytotoxic T lymphocytes (CTLs) were resistant to IKKβ inhibition because other pathways, in particular nuclear factor of activated T cells (NFATc1) signaling, sustained their survival and expansion. In a melanoma mouse model, IKKβ inhibition after CTL cross-priming improved the antitumor response and delayed tumor growth. In conclusion, prolonged IKKβ inhibition decimates circulating Tregs and improves CTL responses when commenced after tumor vaccination, indicating that IKKβ represents a druggable checkpoint.
Collapse
|
15
|
TBK1 as a regulator of autoimmunity and antitumor immunity. Cell Mol Immunol 2018; 15:743-745. [PMID: 29503440 DOI: 10.1038/cmi.2017.165] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 12/14/2017] [Indexed: 01/19/2023] Open
|
16
|
Miraghazadeh B, Cook MC. Nuclear Factor-kappaB in Autoimmunity: Man and Mouse. Front Immunol 2018; 9:613. [PMID: 29686669 PMCID: PMC5900062 DOI: 10.3389/fimmu.2018.00613] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
NF-κB (nuclear factor-kappa B) is a transcription complex crucial for host defense mediated by innate and adaptive immunity, where canonical NF-κB signaling, mediated by nuclear translocation of RelA, c-Rel, and p50, is important for immune cell activation, differentiation, and survival. Non-canonical signaling mediated by nuclear translocation of p52 and RelB contributes to lymphocyte maturation and survival and is also crucial for lymphoid organogenesis. We outline NF-κB signaling and regulation, then summarize important molecular contributions of NF-κB to mechanisms of self-tolerance. We relate these mechanisms to autoimmune phenotypes described in what is now a substantial catalog of immune defects conferred by mutations in NF-κB pathways in mouse models. Finally, we describe Mendelian autoimmune syndromes arising from human NF-κB mutations, and speculate on implications for understanding sporadic autoimmune disease.
Collapse
Affiliation(s)
- Bahar Miraghazadeh
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Acton, ACT, Australia
- Translational Research Unit, Canberra Hospital, Acton, ACT, Australia
| | - Matthew C. Cook
- Centre for Personalised Immunology, John Curtin School of Medical Research, Australian National University, Acton, ACT, Australia
- Translational Research Unit, Canberra Hospital, Acton, ACT, Australia
- Department of Immunology, Canberra Hospital, Acton, ACT, Australia
- *Correspondence: Matthew C. Cook,
| |
Collapse
|
17
|
Oka M, Medrano S, Sequeira-Lόpez MLS, Gómez RA. Chronic Stimulation of Renin Cells Leads to Vascular Pathology. Hypertension 2017; 70:119-128. [PMID: 28533331 DOI: 10.1161/hypertensionaha.117.09283] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/09/2017] [Accepted: 04/04/2017] [Indexed: 01/14/2023]
Abstract
Experimental or spontaneous genomic mutations of the renin-angiotensin system or its pharmacological inhibition in early life leads to renal abnormalities, including poorly developed renal medulla, papillary atrophy, hydronephrosis, inability to concentrate the urine, polyuria, polydipsia, renal failure, and anemia. At the core of such complex phenotype is the presence of unique vascular abnormalities: the renal arterioles do not branch or elongate properly and they have disorganized, concentric hypertrophy. This lesion has been puzzling because it is often found in hypertensive individuals whereas mutant or pharmacologically inhibited animals are hypotensive. Remarkably, when renin cells are ablated with diphtheria toxin, the vascular hypertrophy does not occur, suggesting that renin cells per se may contribute to the vascular disease. To test this hypothesis, on a Ren1c-/- background, we generated mutant mice with reporter expression (Ren1c-/-;Ren1c-Cre;R26R.mTmG and Ren1c-/-;Ren1c-Cre;R26R.LacZ) to trace the fate of reninnull cells. To assess whether reninnull cells maintain their renin promoter active, we used Ren1c-/-;Ren1c-YFP mice that transcribe YFP (yellow fluorescent protein) directed by the renin promoter. We also followed the expression of Akr1b7 and miR-330-5p, markers of cells programmed for the renin phenotype. Contrary to what we expected, reninnull cells did not die or disappear. Instead, they survived, increased in number along the renal arterial tree, and maintained an active molecular memory of the myoepitheliod renin phenotype. Furthermore, null cells of the renin lineage occupied the walls of the arteries and arterioles in a chaotic, directionless pattern directly contributing to the concentric arterial hypertrophy.
Collapse
Affiliation(s)
- Masafumi Oka
- From the Department of Pediatrics, University of Virginia, Charlottesville
| | - Silvia Medrano
- From the Department of Pediatrics, University of Virginia, Charlottesville
| | | | - R Ariel Gómez
- From the Department of Pediatrics, University of Virginia, Charlottesville.
| |
Collapse
|
18
|
Li K, Wang M, Hu Y, Xu N, Yu Q, Wang Q. TAK1 knockdown enhances lipopolysaccharide-induced secretion of proinflammatory cytokines in myeloid cells via unleashing MEKK3 activity. Cell Immunol 2016; 310:193-198. [DOI: 10.1016/j.cellimm.2016.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 09/20/2016] [Accepted: 09/20/2016] [Indexed: 02/06/2023]
|
19
|
The NF-κB transcription factor RelA is required for the tolerogenic function of Foxp3(+) regulatory T cells. J Autoimmun 2016; 70:52-62. [PMID: 27068879 DOI: 10.1016/j.jaut.2016.03.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 12/29/2022]
Abstract
The properties of CD4(+) regulatory T cell (Treg) subsets are dictated by distinct patterns of gene expression determined by FOXP3 and different combinations of various transcription factors. Here we show the NF-κB transcription factor RelA is constitutively active in naïve and effector Tregs. The conditional inactivation of Rela in murine FOXP3(+) cells induces a rapid onset, multi-focal autoimmune disease that depends on RelA being expressed in conventional T cells. In addition to promoting Treg lineage stability, RelA determines the size of the effector Treg population, a function influenced by the presence or absence of RelA in conventional T cells. These findings showing that RelA controls Treg stability and promotes the competitive fitness of effector Tregs highlight the importance of RelA activity in peripheral Treg induced tolerance.
Collapse
|
20
|
Shi JH, Sun SC. TCR signaling to NF-κB and mTORC1: Expanding roles of the CARMA1 complex. Mol Immunol 2015; 68:546-57. [PMID: 26260210 PMCID: PMC4679546 DOI: 10.1016/j.molimm.2015.07.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Revised: 06/07/2015] [Accepted: 07/19/2015] [Indexed: 12/25/2022]
Abstract
Naïve T-cell activation requires signals from both the T-cell receptor (TCR) and the costimulatory molecule CD28. A central mediator of the TCR and CD28 signals is the scaffold protein CARMA1, which functions by forming a complex with partner proteins, Bcl10 and MALT1. A well-known function of the CARMA1 signaling complex is to mediate activation of IκB kinase (IKK) and its target transcription factor NF-κB, thereby promoting T-cell activation and survival. Recent evidence suggests that CARMA1 also mediates TCR/CD28-stimulated activation of the IKK-related kinase TBK1, which plays a role in regulating the homeostasis and migration of T cells. Moreover, the CARMA1 complex connects the TCR/CD28 signals to the activation of mTORC1, a metabolic kinase regulating various aspects of T-cell functions. This review will discuss the mechanism underlying the activation of the CARMA1-dependent signaling pathways and their roles in regulating T-cell functions.
Collapse
Affiliation(s)
- Jian-hong Shi
- Central Laboratory, Affiliated Hospital of Hebei University, 212 Yuhua East Road, Baoding 071000, China
| | - Shao-Cong Sun
- Department of Immunology, The University of Texas MD Anderson Cancer Center, 7455 Fannin Street, Box 902, Houston, TX 77030, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, USA.
| |
Collapse
|