1
|
Lu CH, Ma J, Lin MC, Wu CJ, Kuo CY, Chiang-Ni C, Kuo ML. Streptococcal pyrogenic exotoxin a induces regulatory T cells via TNF-α-TNFR2 signaling. Med Microbiol Immunol 2025; 214:25. [PMID: 40402252 DOI: 10.1007/s00430-025-00835-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 05/06/2025] [Indexed: 05/23/2025]
Abstract
Bacterial superantigens are potent immune activators that trigger T cell proliferation and intensive release of cytokines, leading to toxic shock syndrome. Also, they impair host immune responses, increasing bacterial carriage and transmission. Several studies proposed that superantigens can induce regulatory T (Treg) cells, which may suppress immune responses against bacterial infection. However, the mechanism of Treg cell induction by superantigens is still elusive. We here demonstrated that streptococcal pyrogenic exotoxin A (SPEA) promoted human CD4+CD25+Foxp3+ T cell induction in a dose- and time-dependent manner and the induction required antigen-presenting cells (APCs). SPEA-induced CD4+CD25+ T cells could suppress allogeneic T cell proliferation and IL-2 secretion. Flow cytometric analyses demonstrated high expression of TNFR2 on SPEA-induced CD4+CD25+Foxp3+ T cells. Blocking the interaction between TNF-⍺ and TNFR2 reduced SPEA-induced CD25+Foxp3+ Treg cells. Our present study suggests a mechanism that the TNF-⍺ and TNFR2 axis is required for the induction of human CD4+CD25+Foxp3+ Treg cells by SPEA, which implicates a potential strategy to enhance the clearance of Group A streptococcus infection through reducing Treg cell induction by the inhibition of TNFR2 signaling.
Collapse
Affiliation(s)
- Chun-Hao Lu
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jason Ma
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
| | - Ming-Chieh Lin
- Institute of Molecular Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Cheng-Jang Wu
- Division of Biological Sciences, University of California, La Jolla, San Diego, CA, USA
| | - Chieh-Ying Kuo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
| | - Chuan Chiang-Ni
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan
- Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Ming-Ling Kuo
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, Taiwan.
- Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
- Department of Pediatrics, New Taipei Municipal TuCheng Hospital, New Taipei, Taiwan.
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
2
|
Sureka N, Zaheer S. Regulatory T Cells in Tumor Microenvironment: Therapeutic Approaches and Clinical Implications. Cell Biol Int 2025. [PMID: 40365758 DOI: 10.1002/cbin.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Regulatory T cells (Tregs), previously referred to as suppressor T cells, represent a distinct subset of CD4+ T cells that are uniquely specialized for immune suppression. They are characterized by the constitutive expression of the transcription factor FoxP3 in their nuclei, along with CD25 (the IL-2 receptor α-chain) and CTLA-4 on their cell surface. Tregs not only restrict natural killer cell-mediated cytotoxicity but also inhibit the proliferation of CD4+ and CD8+ T-cells and suppress interferon-γ secretion by immune cells, ultimately impairing an effective antitumor immune response. Treg cells are widely recognized as a significant barrier to the effectiveness of tumor immunotherapy in clinical settings. Extensive research has consistently shown that Treg cells play a pivotal role in facilitating tumor initiation and progression. Conversely, the depletion of Treg cells has been linked to a marked delay in tumor growth and development.
Collapse
Affiliation(s)
- Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
3
|
Lee KY, Mei Y, Liu H, Schwarz H. CD137-expressing regulatory T cells in cancer and autoimmune diseases. Mol Ther 2025; 33:51-70. [PMID: 39668561 PMCID: PMC11764688 DOI: 10.1016/j.ymthe.2024.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/25/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining immune homeostasis, with critical roles in preventing aberrant immune responses that occur in autoimmune diseases and chronic inflammation. Conversely, the abundance of Tregs in cancer is associated with impaired anti-tumor immunity, and tumor immune evasion. Recent work demonstrates that CD137, a well-known costimulatory molecule for T cells, is highly expressed on Tregs in pathological conditions, while its expression is minimal or negligible on peripheral Tregs. The expression of CD137 marks Tregs with potent immunosuppressive phenotype that foster cancer progression and are protective against certain autoimmune diseases. Hence CD137 has emerged as a marker for Tregs. However, several important questions still remain regarding the expression and function of CD137 in Tregs. Here, we provide an overview of our current knowledge of Treg mechanisms of action, with a focus on the role of CD137 in modulating Treg activity. We also explore the implications of CD137+ Tregs in both cancer and autoimmune diseases, emphasizing the significance of targeting these cells for therapeutic intervention in these conditions.
Collapse
Affiliation(s)
- Kang Yi Lee
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Yu Mei
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore
| | - Haiyan Liu
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore.
| | - Herbert Schwarz
- NUS Immunology Programme, Life Sciences Institute, Department of Microbiology and Immunology, National University of Singapore, Singapore 117545, Singapore; NUSMED Immunology Translational Research Programme, National University of Singapore, Singapore 117456, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore.
| |
Collapse
|
4
|
Cammarata I, Pinna V, Pacella I, Rotella I, Soresina A, Badolato R, Plebani A, Pignata C, Cirillo E, Zicari AM, Violi F, Carnevale R, Loffredo L, Piconese S. In adult X-CGD patients, regulatory T cells are expanded while activated T cells display a NOX2-independent ROS increase. Immunol Lett 2024; 266:106839. [PMID: 38309375 DOI: 10.1016/j.imlet.2024.106839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 01/19/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
The X-linked chronic granulomatous disease (X-CGD), a rare genetic disease characterised by recurrent infections, is caused by mutations of NOX2. Significant proportions of X-CGD patients display signs of immune dysregulation. Regulatory T cells (Tregs) are CD4+T lymphocytes that expand in active inflammation and prevent autoimmune disorders. Here we asked whether X-CGD is associated to Treg dysfunctions in adult patients. To this aim, the frequency of Tregs was analysed through intracellular flow cytometry in a cohort of adult X-CGD patients, carriers and controls. We found that Tregs were significantly expanded and activated in blood of adult X-CGD patients, and this was associated with activation of conventional CD4+T cells (Tconvs). T cell activation was characterised by accumulation of intracellular ROS, not derived from NOX2 but likely produced by cellular metabolism. The higher TNF production by Tconvs in X-CGD patients might contribute to the expansion of Tregs through the TNFR2 receptor. In summary, our data indicate that Tregs expand in adult X-CGD in response to immune activation, and that the increase of NOX2-independent ROS content is a feature of activated T cells.
Collapse
Affiliation(s)
- Ilenia Cammarata
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Valeria Pinna
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ilenia Pacella
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Ivano Rotella
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Annarosa Soresina
- Department of Clinical and Experimental Sciences, Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Raffaele Badolato
- Department of Clinical and Experimental Sciences, Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Alessandro Plebani
- Department of Clinical and Experimental Sciences, Paediatrics Clinic and Institute for Molecular Medicine A. Nocivelli, ASST-Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Claudio Pignata
- Department of Translational Medical Sciences - Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Emilia Cirillo
- Department of Translational Medical Sciences - Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Anna Maria Zicari
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, Rome, Italy
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy; Mediterranea Cardiocentro-Napoli, Naples, Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy; IRCCS Neuromed, Località Camerelle, Pozzilli, Italy
| | - Lorenzo Loffredo
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Silvia Piconese
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy; Neuroimmunology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy; Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
5
|
Kosinsky RL, Gonzalez MM, Saul D, Barros LL, Sagstetter MR, Fedyshyn Y, Nair A, Sun Z, Hamdan FH, Gibbons HR, Perez Pachon ME, Druliner BR, Johnsen SA, Faubion WA. The FOXP3 + Pro-Inflammatory T Cell: A Potential Therapeutic Target in Crohn's Disease. Gastroenterology 2024; 166:631-644.e17. [PMID: 38211712 PMCID: PMC10960691 DOI: 10.1053/j.gastro.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/27/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
BACKGROUND & AIMS The incidence of Crohn's disease (CD) continues to increase worldwide. The contribution of CD4+ cell populations remains to be elucidated. We aimed to provide an in-depth transcriptional assessment of CD4+ T cells driving chronic inflammation in CD. METHODS We performed single-cell RNA-sequencing in CD4+ T cells isolated from ileal biopsies of patients with CD compared with healthy individuals. Cells underwent clustering analysis, followed by analysis of gene signaling networks. We overlapped our differentially expressed genes with publicly available microarray data sets and performed functional in vitro studies, including an in vitro suppression assay and organoid systems, to model gene expression changes observed in CD regulatory T (Treg) cells and to test predicted therapeutics. RESULTS We identified 5 distinct FOXP3+ regulatory Treg subpopulations. Tregs isolated from healthy controls represent the origin of pseudotemporal development into inflammation-associated subtypes. These proinflammatory Tregs displayed a unique responsiveness to tumor necrosis factor-α signaling with impaired suppressive activity in vitro and an elevated cytokine response in an organoid coculture system. As predicted in silico, the histone deacetylase inhibitor vorinostat normalized gene expression patterns, rescuing the suppressive function of FOXP3+ cells in vitro. CONCLUSIONS We identified a novel, proinflammatory FOXP3+ T cell subpopulation in patients with CD and developed a pipeline to specifically target these cells using the US Food and Drug Administration-approved drug vorinostat.
Collapse
Affiliation(s)
- Robyn Laura Kosinsky
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Michelle M Gonzalez
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Dominik Saul
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota; Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tübingen, BG Trauma Center, Tübingen, Germany
| | - Luísa Leite Barros
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota; Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Mary R Sagstetter
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Asha Nair
- Division of Computational Biology, Mayo Clinic, Rochester, Minnesota
| | - Zhifu Sun
- Division of Computational Biology, Mayo Clinic, Rochester, Minnesota
| | - Feda H Hamdan
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Hunter R Gibbons
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - Brooke R Druliner
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | | | - William A Faubion
- Division of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, Arizona.
| |
Collapse
|
6
|
Wang M, Chen S, He X, Yuan Y, Wei X. Targeting inflammation as cancer therapy. J Hematol Oncol 2024; 17:13. [PMID: 38520006 PMCID: PMC10960486 DOI: 10.1186/s13045-024-01528-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/25/2024] Open
Abstract
Inflammation has accompanied human beings since the emergence of wounds and infections. In the past decades, numerous efforts have been undertaken to explore the potential role of inflammation in cancer, from tumor development, invasion, and metastasis to the resistance of tumors to treatment. Inflammation-targeted agents not only demonstrate the potential to suppress cancer development, but also to improve the efficacy of other therapeutic modalities. In this review, we describe the highly dynamic and complex inflammatory tumor microenvironment, with discussion on key inflammation mediators in cancer including inflammatory cells, inflammatory cytokines, and their downstream intracellular pathways. In addition, we especially address the role of inflammation in cancer development and highlight the action mechanisms of inflammation-targeted therapies in antitumor response. Finally, we summarize the results from both preclinical and clinical studies up to date to illustrate the translation potential of inflammation-targeted therapies.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Siyuan Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No.17, Block3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
7
|
Mensink M, Verleng LJ, Schrama E, Janssen GM, Tjokrodirijo RT, van Veelen PA, Jiang Q, Pascutti MF, van der Hoorn ML, Eikmans M, de Kivit S, Borst J. Tregs from human blood differentiate into nonlymphoid tissue-resident effector cells upon TNFR2 costimulation. JCI Insight 2024; 9:e172942. [PMID: 38341270 PMCID: PMC10972588 DOI: 10.1172/jci.insight.172942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
Tregs can facilitate transplant tolerance and attenuate autoimmune and inflammatory diseases. Therefore, it is clinically relevant to stimulate Treg expansion and function in vivo and to create therapeutic Treg products in vitro. We report that TNF receptor 2 (TNFR2) is a unique costimulus for naive, thymus-derived Tregs (tTregs) from human blood that promotes their differentiation into nonlymphoid tissue-resident (NLT-resident) effector Tregs, without Th-like polarization. In contrast, CD28 costimulation maintains a lymphoid tissue-resident (LT-resident) Treg phenotype. We base this conclusion on transcriptome and proteome analysis of TNFR2- and CD28-costimulated CD4+ tTregs and conventional T cells (Tconvs), followed by bioinformatic comparison with published transcriptomic Treg signatures from NLT and LT in health and disease, including autoimmunity and cancer. These analyses illuminate that TNFR2 costimulation promoted tTreg capacity for survival, migration, immunosuppression, and tissue regeneration. Functional studies confirmed improved migratory ability of TNFR2-costimulated tTregs. Flow cytometry validated the presence of the TNFR2-driven tTreg signature in effector/memory Tregs from the human placenta, as opposed to blood. Thus, TNFR2 can be exploited as a driver of NLT-resident tTreg differentiation for adoptive cell therapy or antibody-based immunomodulation in human disease.
Collapse
|
8
|
Ciudad M, Ouandji S, Lamarthée B, Cladière C, Ghesquière T, Nivet M, Thébault M, Boidot R, Soudry-Faure A, Chevrier S, Richard C, Maillet T, Maurier F, Greigert H, Genet C, Ramon A, Trad M, Predan V, Saas P, Samson M, Bonnotte B, Audia S. Regulatory T-cell dysfunctions are associated with increase in tumor necrosis factor α in autoimmune hemolytic anemia and participate in Th17 polarization. Haematologica 2024; 109:444-457. [PMID: 37534543 PMCID: PMC10828774 DOI: 10.3324/haematol.2023.282859] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023] Open
Abstract
Warm autoimmune hemolytic anemia (wAIHA) is a rare acquired autoimmune disease mediated by antibodies targeting red blood cells. The involvement of CD4 T-helper cells has been scarcely explored, with most findings extrapolated from animal models. Here, we performed quantification of both effector T lymphocytes (Teff) and regulatory T cells (Treg), associated with functional and transcriptomic analyses of Treg in human wAIHA. We observed a shift of Teff toward a Th17 polarization concordant with an increase in serum interleukin-17 concentration that correlates with red blood cell destruction parameters, namely lactate dehydrogenase and bilirubin levels. A decrease in circulating Treg, notably effector Treg, associated with a functional deficiency, as represented by their decrease capability to inhibit Teff proliferation, were also observed. Treg deficiency was associated with a reduced expression of Foxp3, the master transcription factor known to maintain the Treg phenotype stability and suppressive functions. Transcriptomic profiling of Treg revealed activation of the tumor necrosis facto (TNF)-α pathway, which was linked to increased serum TNF-α concentrations that were twice as high as in controls. Treg transcriptomic profiling also suggested that post-translational mechanisms possibly accounted for Foxp3 downregulation and Treg dysfunctions. Since TNF-α participates in the rupture of immune tolerance during wAIHA, its inhibition could be of interest. To this end, the effects of fostamatinib, a SYK inhibitor, were investigated in vitro, and we showed that besides the inhibition of erythrocyte phagocytosis by monocytes, fostamatinib is also able to dampen TNF-α production, thus appearing as a promising multitargeting therapy in wAIHA (clinicaltrials gov. Identifier: NCT02158195).
Collapse
Affiliation(s)
- Marion Ciudad
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Sethi Ouandji
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | | | - Claudie Cladière
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Thibault Ghesquière
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Martin Nivet
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Marine Thébault
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Romain Boidot
- Unit of Molecular Biology, Georges-François Leclerc Cancer Center - F-21000 Dijon
| | - Agnès Soudry-Faure
- Department of Clinical Research and Innovation (DRCI), Clinical Research Unit-Methodological Support Network (USMR), Dijon Bourgogne University Hospital, Dijon
| | - Sandy Chevrier
- Unit of Molecular Biology, Georges-François Leclerc Cancer Center - F-21000 Dijon
| | - Corentin Richard
- Unit of Molecular Biology, Georges-François Leclerc Cancer Center - F-21000 Dijon
| | - Thibault Maillet
- Department of Internal Medicine - Centre Hospitalier de Mâcon, Groupe Hospitalier Bourgogne Méridionale - F-71000 Macon
| | - François Maurier
- Department of Internal Medicine, Groupe Hospitalier UNEOS - F-57000 Metz
| | - Hélène Greigert
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Coraline Genet
- Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - André Ramon
- Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Malika Trad
- Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Valérie Predan
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon
| | - Philippe Saas
- Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Maxime Samson
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Bernard Bonnotte
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon
| | - Sylvain Audia
- Department of Internal Medicine and Clinical Immunology, Referral Center for adult autoimmune cytopenia (CeReCAI) - Dijon University Hospital - F-21000 Dijon, France; Université de Bourgogne, INSERM, UMR1098, RIGHT -F-21000 Dijon.
| |
Collapse
|
9
|
Nowag B, Schäfer D, Hengl T, Corduff N, Goldie K. Biostimulating fillers and induction of inflammatory pathways: A preclinical investigation of macrophage response to calcium hydroxylapatite and poly-L lactic acid. J Cosmet Dermatol 2024; 23:99-106. [PMID: 37593832 DOI: 10.1111/jocd.15928] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/02/2023] [Accepted: 06/29/2023] [Indexed: 08/19/2023]
Abstract
INTRODUCTION Initial macrophage response to biostimulatory substances is key in determining the subsequent behavior of fibroblasts and the organization of newly synthesized collagen. Though histological studies suggest that calcium hydroxylapatite (CaHA) filler initiates a regenerative healing response with collagen and elastin deposition similar to natural, healthy tissue rather than an inflammatory response with fibrosis, the relative activity of macrophages stimulated by CaHA, as well as how this activity compares to that induced by other biostimulatory fillers, has not been explored. The aim of the study is to characterize the in vitro macrophage response to two biostimulory fillers, CaHA and PLLA (poly-L lactic acid), and to evaluate their inflammatory potential. METHODS Primary human macrophages were incubated with two dilutions (1:50 and 1:100) of commercially available CaHA or PLLA. After 24 h incubation, an inflammation array was used to screen for the expression of 40 cytokines, released by macrophages. ELISA was used to confirm array results. RESULTS Four cytokines were significantly upregulated in M1 macrophages incubated with PLLA compared to both unstimulated controls and CaHA: CCL1 (p < 0.001), TNFRII (p < 0.01), MIP-1α (p < 0.05), and IL-8 (p < 0.001). In M2 macrophages, MIP-1α (p < 0.01) and MIP-1β (p < 0.01) were significantly upregulated by PLLA compared to CaHA and unstimulated controls. CONCLUSION Together, these findings indicate that the CaHA mode of action is a non-inflammatory response while PLLA initiates expression of several cytokines known to play a role in inflammation. Our study supports the concept that these two "biostimulatory" fillers follow distinct pathways and should be considered individually with regard to mechanism of action.
Collapse
Affiliation(s)
| | | | - Thomas Hengl
- R&D, Merz Aesthetics GmbH, Frankfurt am Main, Germany
| | | | | |
Collapse
|
10
|
Vaikunthanathan T, Landmann E, Correa DM, Romano M, Trevelin SC, Peng Q, Crespo E, Corrado M, Lozano JJ, Pearce EL, Perpinan E, Zoccarato A, Siew L, Edwards-Hicks J, Khan R, Luu NT, Thursz MR, Newsome PN, Martinez-Llordella M, Shah N, Lechler RI, Shah AM, Sanchez-Fueyo A, Lombardi G, Safinia N. Dysregulated anti-oxidant signalling and compromised mitochondrial integrity negatively influence regulatory T cell function and viability in liver disease. EBioMedicine 2023; 95:104778. [PMID: 37657135 PMCID: PMC10480539 DOI: 10.1016/j.ebiom.2023.104778] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Dysregulated inflammatory responses and oxidative stress are key pathogenic drivers of chronic inflammatory diseases such as liver cirrhosis (LC). Regulatory T cells (Tregs) are essential to prevent excessive immune activation and maintain tissue homeostasis. While inflammatory cues are well known to modulate the function and stability of Tregs, the extent to which Tregs are influenced by oxidative stress has not been fully explored. METHODS The phenotypic and functional properties of CD4+CD25+CD127lo/- Tregs isolated from patients with LC were compared to healthy controls (HC). Treg redox state was investigated by characterizing intracellular reactive oxygen species (ROS), NADPH oxidase-2 (Nox2) activity, mitochondrial function, morphology, and nuclear factor-erythroid 2-related factor (Nrf2) antioxidant signalling. The relevance of Nrf2 and its downstream target, Heme-oxygenase-1 (HO-1), in Treg function, stability, and survival, was further assessed using mouse models and CRISPR/Cas9-mediated HO-1 knock-out. FINDINGS Circulating Tregs from LC patients displayed a reduced suppressive function, correlating with liver disease severity, associated with phenotypic abnormalities and increased apoptosis. Mechanistically, this was linked to a dysregulated Nrf2 signalling with resultant lower levels of HO-1, enhanced Nox2 activation, and impaired mitochondrial respiration and integrity. The functional deficit in LC Tregs could be partially recapitulated by culturing control Tregs in patient sera. INTERPRETATION Our findings reveal that Tregs rely on functional redox homeostasis for their function, stability, and survival. Targeting Treg specific anti-oxidant pathways may have therapeutic potential to reverse the Treg impairment in conditions of oxidative damage such as advanced liver disease. FUNDING This study was funded by the Wellcome Trust (211113/A/18/Z).
Collapse
Affiliation(s)
- Trishan Vaikunthanathan
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Emmanuelle Landmann
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Diana Marin Correa
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Marco Romano
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Silvia Cellone Trevelin
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Qi Peng
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Elena Crespo
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Mauro Corrado
- Bloomberg-Kimmel Institute for Cancer Immunotherapy and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Juan-José Lozano
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Joseph Stelzmannstrasse 26, 50931, Cologne, Germany.
| | - Erika L Pearce
- Centro de Investigación Biomédica en Red Enfermedades Hepáticas y Digestivas (CIBEREHD), Calle Rossello 153 Bajos, O8036, Barcelona, Spain.
| | - Elena Perpinan
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Anna Zoccarato
- Department of Immunometabolism, Max Planck Institute of Immunobiology & Epigenetics, Stübeweg 51, 79108, Freiburg, Germany.
| | - Leonard Siew
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Joy Edwards-Hicks
- Centre for Liver and Gastroenterology Research and Birmingham National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom.
| | - Reenam Khan
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, Liver Unit, 10th Floor QEQM Building, St Mary's Hospital, W2 1NY, London, United Kingdom.
| | - Nguyet-Thin Luu
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, Liver Unit, 10th Floor QEQM Building, St Mary's Hospital, W2 1NY, London, United Kingdom.
| | - Mark R Thursz
- Institute of Liver Sciences, King's College Hospital NHS Foundation Trust, London, SE5 9NU, United Kingdom.
| | - Philip N Newsome
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Imperial College London, Liver Unit, 10th Floor QEQM Building, St Mary's Hospital, W2 1NY, London, United Kingdom.
| | - Marc Martinez-Llordella
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Naina Shah
- James Black Centre, Department of Cardiovascular sciences, British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, SE5 9NU, United Kingdom.
| | - Robert I Lechler
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Ajay M Shah
- Department of Immunometabolism, Max Planck Institute of Immunobiology & Epigenetics, Stübeweg 51, 79108, Freiburg, Germany.
| | - Alberto Sanchez-Fueyo
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, 5th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London, SE1 9RT, United Kingdom.
| | - Niloufar Safinia
- Department of Inflammation Biology, Institute of Liver Studies, School of Immunology and Microbial Sciences, James Black Centre, King's College London, London, SE5 9NU, United Kingdom.
| |
Collapse
|
11
|
Venken K, Jarlborg M, Decruy T, Mortier C, Vlieghe C, Gilis E, De Craemer AS, Coudenys J, Cambré I, Fleury D, Klimowicz A, Van den Bosch F, Hoorens A, Lobaton T, de Roock S, Sparwasser T, Nabozny G, Jacques P, Elewaut D. Distinct immune modulatory roles of regulatory T cells in gut versus joint inflammation in TNF-driven spondyloarthritis. Ann Rheum Dis 2023; 82:1076-1090. [PMID: 37197892 DOI: 10.1136/ard-2022-223757] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 04/19/2023] [Indexed: 05/19/2023]
Abstract
OBJECTIVES Gut and joint inflammation commonly co-occur in spondyloarthritis (SpA) which strongly restricts therapeutic modalities. The immunobiology underlying differences between gut and joint immune regulation, however, is poorly understood. We therefore assessed the immunoregulatory role of CD4+FOXP3+ regulatory T (Treg) cells in a model of Crohn's-like ileitis and concomitant arthritis. METHODS RNA-sequencing and flow cytometry was performed on inflamed gut and joint samples and tissue-derived Tregs from tumour necrosis factor (TNF)∆ARE mice. In situ hybridisation of TNF and its receptors (TNFR) was applied to human SpA gut biopsies. Soluble TNFR (sTNFR) levels were measured in serum of mice and patients with SpA and controls. Treg function was explored by in vitro cocultures and in vivo by conditional Treg depletion. RESULTS Chronic TNF exposure induced several TNF superfamily (TNFSF) members (4-1BBL, TWEAK and TRAIL) in synovium and ileum in a site-specific manner. Elevated TNFR2 messenger RNA levels were noted in TNF∆ARE/+ mice leading to increased sTNFR2 release. Likewise, sTNFR2 levels were higher in patients with SpA with gut inflammation and distinct from inflammatory and healthy controls. Tregs accumulated at both gut and joints of TNF∆ARE mice, yet their TNFR2 expression and suppressive function was significantly lower in synovium versus ileum. In line herewith, synovial and intestinal Tregs displayed a distinct transcriptional profile with tissue-restricted TNFSF receptor and p38MAPK gene expression. CONCLUSIONS These data point to profound differences in immune-regulation between Crohn's ileitis and peripheral arthritis. Whereas Tregs control ileitis they fail to dampen joint inflammation. Synovial resident Tregs are particularly maladapted to chronic TNF exposure.
Collapse
Affiliation(s)
- Koen Venken
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Matthias Jarlborg
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Tine Decruy
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Céline Mortier
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Carolien Vlieghe
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Elisabeth Gilis
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Ann-Sophie De Craemer
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Julie Coudenys
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Isabelle Cambré
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Devan Fleury
- Immunology and Respiratory Department, Boehringer Ingelheim Corp Pharmaceutical Research and Development Centre Ridgefield, Ridgefield, Connecticut, USA
| | - Alexander Klimowicz
- Immunology and Respiratory Department, Boehringer Ingelheim Corp Pharmaceutical Research and Development Centre Ridgefield, Ridgefield, Connecticut, USA
| | - Filip Van den Bosch
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Anne Hoorens
- Department of Pathology, University Hospital Ghent, Gent, Belgium
| | - Triana Lobaton
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Gastroenterology unit), Ghent University, Ghent, Belgium
- Department of Gastroenterology, Ghent University Hospital, Ghent, Belgium
| | - Sytze de Roock
- Department of Pediatric Immunology, Center for Molecular and Cellular Intervention CMCI, Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tim Sparwasser
- University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Gerald Nabozny
- Immunology and Respiratory Department, Boehringer Ingelheim Corp Pharmaceutical Research and Development Centre Ridgefield, Ridgefield, Connecticut, USA
| | - Peggy Jacques
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| | - Dirk Elewaut
- Molecular Immunology and Inflammation Unit, VIB Center for Inflammation Research, Ghent University, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Department of Internal Medicine and Pediatrics (Rheumatology unit), Ghent University, Ghent, Belgium
| |
Collapse
|
12
|
Wayman JA, Thomas A, Bejjani A, Katko A, Almanan M, Godarova A, Korinfskaya S, Cazares TA, Yukawa M, Kottyan LC, Barski A, Chougnet CA, Hildeman DA, Miraldi ER. An atlas of gene regulatory networks for memory CD4 + T cells in youth and old age. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531590. [PMID: 36945549 PMCID: PMC10028906 DOI: 10.1101/2023.03.07.531590] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Aging profoundly affects immune-system function, promoting susceptibility to pathogens, cancers and chronic inflammation. We previously identified a population of IL-10-producing, T follicular helper-like cells (" Tfh10 "), linked to suppressed vaccine responses in aged mice. Here, we integrate single-cell ( sc )RNA-seq, scATAC-seq and genome-scale modeling to characterize Tfh10 - and the full CD4 + memory T cell ( CD4 + TM ) compartment - in young and old mice. We identified 13 CD4 + TM populations, which we validated through cross-comparison to prior scRNA-seq studies. We built gene regulatory networks ( GRNs ) that predict transcription-factor control of gene expression in each T-cell population and how these circuits change with age. Through integration with pan-cell aging atlases, we identified intercellular-signaling networks driving age-dependent changes in CD4 + TM. Our atlas of finely resolved CD4 + TM subsets, GRNs and cell-cell communication networks is a comprehensive resource of predicted regulatory mechanisms operative in memory T cells, presenting new opportunities to improve immune responses in the elderly.
Collapse
|
13
|
The Immunosuppressive Effect of TNFR2 Expression in the Colorectal Cancer Microenvironment. Biomedicines 2023; 11:biomedicines11010173. [PMID: 36672682 PMCID: PMC9856189 DOI: 10.3390/biomedicines11010173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/12/2023] Open
Abstract
Colorectal cancer (CRC) represents one of the most common causes of death among cancers worldwide. Its incidence has been increasing among the young population. Many risk factors contribute to the development and progression of CRC and about 70% of them are sporadic. The CRC microenvironment is highly heterogeneous and represents a very complex immunosuppressive platform. Many cytokines and their receptors are vital participants in this immunosuppressive microenvironment. Tumor necrosis factors (TNFs) and TNF receptor 2 (TNFR2) are critical players in the development of CRC. TNFR2 was observed to have increased the immunosuppressive activity of CRC cells via regulatory T cells (T regs) and myeloid-derived suppressor cells (MDSC) in the CRC microenvironment. However, the exact mechanism of TNFR2 in regulating the CRC prognosis remains elusive. Here, we discuss the role of TNFR2 in immune escape mechanism of CRC in the immunosuppressive cells, including Tregs and MDSCs, and the complex signaling pathways that facilitate the development of CRC. It is suggested that extensive studies on TNFR2 downstream signaling must be done, since TNFR2 has a high potential to be developed into a therapeutic agent and cancer biomarker in the future.
Collapse
|
14
|
Tauber PA, Kratzer B, Schatzlmaier P, Smole U, Köhler C, Rausch L, Kranich J, Trapin D, Neunkirchner A, Zabel M, Jutz S, Steinberger P, Gadermaier G, Brocker T, Stockinger H, Derdak S, Pickl WF. The small molecule inhibitor BX-795 uncouples IL-2 production from inhibition of Th2 inflammation and induces CD4 + T cells resembling iTreg. Front Immunol 2023; 14:1094694. [PMID: 37090735 PMCID: PMC10117943 DOI: 10.3389/fimmu.2023.1094694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/06/2023] [Indexed: 04/25/2023] Open
Abstract
Background Treg cells have been shown to be an important part of immune-homeostasis and IL-2 which is produced upon T cell receptor (TCR)-dependent activation of T lymphocytes has been demonstrated to critically participate in Treg development. Objective To evaluate small molecule inhibitors (SMI) for the identification of novel IL-2/Treg enhancing compounds. Materials and methods We used TCR-dependent and allergen-specific cytokine secretion of human and mouse T cells, next generation messenger ribonucleic acid sequencing (RNA-Seq) and two different models of allergic airway inflammation to examine lead SMI-compounds. Results We show here that the reported 3-phosphoinositide dependent kinase-1 (PDK1) SMI BX-795 increased IL-2 in culture supernatants of Jurkat E6-1 T cells, human peripheral blood mononuclear cells (hPBMC) and allergen-specific mouse T cells upon TCR-dependent and allergen-specific stimulation while concomitantly inhibiting Th2 cytokine secretion. RNA-Seq revealed that the presence of BX-795 during allergen-specific activation of T cells induces a bona fide Treg cell type highly similar to iTreg but lacking Foxp3 expression. When applied in mugwort pollen and house dust mite extract-based models of airway inflammation, BX-795 significantly inhibited Th2 inflammation including expression of Th2 signature transcription factors and cytokines and influx into the lungs of type 2-associated inflammatory cells such as eosinophils. Conclusions BX-795 potently uncouples IL-2 production from Th2 inflammation and induces Th-IL-2 cells, which highly resemble induced (i)Tregs. Thus, BX-795 may be a useful new compound for the treatment of allergic diseases.
Collapse
Affiliation(s)
- Peter A. Tauber
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Kratzer
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Philipp Schatzlmaier
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Ursula Smole
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Cordula Köhler
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Lisa Rausch
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Jan Kranich
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Doris Trapin
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alina Neunkirchner
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Maja Zabel
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sabrina Jutz
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Peter Steinberger
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | | | - Thomas Brocker
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, Ludwig Maximilian University (LMU) Munich, Munich, Germany
| | - Hannes Stockinger
- Institute of Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sophia Derdak
- Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Winfried F. Pickl
- Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
- Karl Landsteiner University of Healthcare, Krems, Austria
- *Correspondence: Winfried F. Pickl,
| |
Collapse
|
15
|
Sun Y, Sun J, Xiao M, Lai W, Li L, Fan C, Pei H. DNA origami-based artificial antigen-presenting cells for adoptive T cell therapy. SCIENCE ADVANCES 2022; 8:eadd1106. [PMID: 36459554 PMCID: PMC10936057 DOI: 10.1126/sciadv.add1106] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 10/17/2022] [Indexed: 06/17/2023]
Abstract
Nanosized artificial antigen-presenting cells (aAPCs) with efficient signal presentation hold great promise for in vivo adoptive cell therapy. Here, we used DNA origami nanostructures as two-dimensional scaffolds to regulate the spatial presentation of activating ligands at nanoscale to construct high-effective aAPCs. The DNA origami-based aAPC comprises costimulatory ligands anti-CD28 antibody anchored at three vertices and T cell receptor (TCR) ligands peptide-major histocompatibility complex (pMHC) anchored at three edges with varying density. The DNA origami scaffold enables quantitative analysis of ligand-receptor interactions in T cell activation at the single-particle, single-molecule resolution. The pMHC-TCR-binding dwell time is increased from 9.9 to 12.1 s with increasing pMHC density, driving functional T cell responses. In addition, both in vitro and in vivo assays demonstrate that the optimized DNA origami-based aAPCs show effective tumor growth inhibiting capability in adoptive immunotherapy. These results provide important insights into the rational design of molecular vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Yueyang Sun
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Jiajia Sun
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Mingshu Xiao
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Wei Lai
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Li Li
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, and Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hao Pei
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| |
Collapse
|
16
|
Chen FY, Geng CA, Chou CK, Zheng JB, Yang Y, Wang YF, Li TZ, Li P, Chen JJ, Chen X. Distepharinamide, a novel dimeric proaporphine alkaloid from Diploclisia glaucescens, inhibits the differentiation and proliferative expansion of CD4 +Foxp3 + regulatory T cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 107:154482. [PMID: 36202057 DOI: 10.1016/j.phymed.2022.154482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 09/17/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND CD4+Foxp3+ regulatory T cells (Tregs) represent the primary cellular mechanism of tumor immune evasion. Elimination of Treg activity by the pharmacological agent may enhance anti-tumor immune responses. However, Treg-eliminating agents, especially those with small molecules, are rarely reported. PURPOSE To identify small molecule inhibitors of Treg cells from natural products. METHODS Compounds from Diploclisia glaucescens were isolated by column chromatography, and structures were identified by spectroscopic evidence and quantum calculations. The tet-On system for Foxp3-GFP expression in Jurkat T cells was generated to screen Treg inhibitors based on Foxp3 expression. The effect of the compound on TNF-induced proliferative expansion of naturally occurring Tregs (nTregs) and TGF-β-induced generation of Tregs (iTregs) from naive CD4+ Tcells was further examined. RESULTS A novel dimeric proaporphine alkaloid, designated as distepharinamide (DSA) with a symmetric structure isolated from the stems of D. glaucescens, restrained the doxycycline (Doxy)-induced Foxp3-tGFP expression, decreased the half-life of Foxp3 mRNA as well as reduced the mRNA levels of chemokine receptors (CCR4, CCR8 and CCR10) in Jurkat T cells with inducible Foxp3-tGFP expression. In lymphocytes or purified Tregs from wild-type C57BL/6 mice or from C57BL/6-Tg(Foxp3-DTR/EGFP)23.2Spar/Mmjax mice, DSA markedly inhibited TNF-induced proliferative expansion of Tregs present in the unfractionated CD4+ T cells, accompanied by the down-regulation of TNFR2, CD25 and CTLA4 expression on Tregs. Furthermore, DSA potently inhibited TGF-β-induced differentiation of Foxp3-expressing iTregs. Importantly, the expression of Foxp3 mRNA by both nTregs and iTregs was decreased by DSA treatment. Nevertheless, DSA at the same concentrations did not inhibit the proliferation of conventional CD4+ and CD8+ T cells stimulated by anti-CD3/CD28 antibodies. CONCLUSION DSA, a novel dimeric proaporphine alkaloid, potently inhibited the expansion of nTregs and generation of iTregs. Therefore, DSA or its analogs may merit further investigation as novel immunotherapeutic agents.
Collapse
Affiliation(s)
- Feng-Yang Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau 999078, China; School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou 310053, China
| | - Chang-An Geng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Chon-Kit Chou
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau 999078, China
| | - Jing-Bin Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau 999078, China
| | - Yang Yang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau 999078, China
| | - Yi-Fei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau 999078, China
| | - Tian-Ze Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China
| | - Ping Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau 999078, China
| | - Ji-Jun Chen
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Science, University of Macau, Macau 999078, China.
| |
Collapse
|
17
|
Skartsis N, Ferreira LMR, Tang Q. The dichotomous outcomes of TNFα signaling in CD4 + T cells. Front Immunol 2022; 13:1042622. [PMID: 36466853 PMCID: PMC9708889 DOI: 10.3389/fimmu.2022.1042622] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/03/2022] [Indexed: 09/26/2023] Open
Abstract
TNFa blocking agents were the first-in-class biologic drugs used for the treatment of autoimmune disease. Paradoxically, however, exacerbation of autoimmunity was observed in some patients. TNFa is a pleiotropic cytokine that has both proinflammatory and regulatory effects on CD4+ T cells and can influence the adaptive immune response against autoantigens. Here, we critically appraise the literature and discuss the intricacies of TNFa signaling that may explain the controversial findings of previous studies. The pleiotropism of TNFa is based in part on the existence of two biologically active forms of TNFa, soluble and membrane-bound, with different affinities for two distinct TNF receptors, TNFR1 and TNFR2, leading to activation of diverse downstream molecular pathways involved in cell fate decisions and immune function. Distinct membrane expression patterns of TNF receptors by CD4+ T cell subsets and their preferential binding of distinct forms of TNFα produced by a diverse pool of cellular sources during different stages of an immune response are important determinants of the differential outcomes of TNFa-TNF receptor signaling. Targeted manipulation of TNFa-TNF receptor signaling on select CD4+ T cell subsets may offer specific therapeutic interventions to dampen inflammation while fortifying immune regulation for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Nikolaos Skartsis
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, United States
- Mayo Clinic William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Leonardo M. R. Ferreira
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, United States
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Qizhi Tang
- Department of Surgery, University of California, San Francisco, San Francisco, CA, United States
- Diabetes Center, University of California, San Francisco, San Francisco, CA, United States
- Gladstone University of California San Francisco (UCSF) Institute of Genome Immunology, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
18
|
Advancing Biologic Therapy for Refractory Autoimmune Hepatitis. Dig Dis Sci 2022; 67:4979-5005. [PMID: 35147819 DOI: 10.1007/s10620-021-07378-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/27/2021] [Indexed: 01/05/2023]
Abstract
Biologic agents may satisfy an unmet clinical need for treatment of refractory autoimmune hepatitis. The goals of this review are to present the types and results of biologic therapy for refractory autoimmune hepatitis, indicate opportunities to improve and expand biologic treatment, and encourage comparative clinical trials. English abstracts were identified in PubMed by multiple search terms. Full-length articles were selected for review, and secondary and tertiary bibliographies were developed. Rituximab (monoclonal antibodies against CD20 on B cells), infliximab (monoclonal antibodies against tumor necrosis factor-alpha), low-dose recombinant interleukin 2 (regulatory T cell promoter), and belimumab (monoclonal antibodies against B cell activating factor) have induced laboratory improvement in small cohorts with refractory autoimmune hepatitis. Ianalumab (monoclonal antibodies against the receptor for B cell activating factor) is in clinical trial. These agents target critical pathogenic pathways, but they may also have serious side effects. Blockade of the B cell activating factor or its receptors may disrupt pivotal B and T cell responses, and recombinant interleukin 2 complexed with certain interleukin 2 antibodies may selectively expand the regulatory T cell population. A proliferation-inducing ligand that enhances T cell proliferation and survival is an unevaluated, potentially pivotal, therapeutic target. Fully human antibodies, expanded target options, improved targeting precision, more effective delivery systems, and biosimilar agents promise to improve efficacy, safety, and accessibility. In conclusion, biologic agents target key pathogenic pathways in autoimmune hepatitis, and early experiences in refractory disease encourage clarification of the preferred target, rigorous clinical trial, and comparative evaluations.
Collapse
|
19
|
Bednar KJ, Lee JH, Ort T. Tregs in Autoimmunity: Insights Into Intrinsic Brake Mechanism Driving Pathogenesis and Immune Homeostasis. Front Immunol 2022; 13:932485. [PMID: 35844555 PMCID: PMC9280893 DOI: 10.3389/fimmu.2022.932485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/16/2022] [Indexed: 11/16/2022] Open
Abstract
CD4+CD25highFoxp3+ regulatory T-cells (Tregs) are functionally characterized for their ability to suppress the activation of multiple immune cell types and are indispensable for maintaining immune homeostasis and tolerance. Disruption of this intrinsic brake system assessed by loss of suppressive capacity, cell numbers, and Foxp3 expression, leads to uncontrolled immune responses and tissue damage. The conversion of Tregs to a pathogenic pro-inflammatory phenotype is widely observed in immune mediated diseases. However, the molecular mechanisms that underpin the control of Treg stability and suppressive capacity are incompletely understood. This review summarizes the concepts of Treg cell stability and Treg cell plasticity highlighting underlying mechanisms including translational and epigenetic regulators that may enable translation to new therapeutic strategies. Our enhanced understanding of molecular mechanism controlling Tregs will have important implications into immune homeostasis and therapeutic potential for the treatment of immune-mediated diseases.
Collapse
|
20
|
Vargas JG, Wagner J, Shaikh H, Lang I, Medler J, Anany M, Steinfatt T, Mosca JP, Haack S, Dahlhoff J, Büttner-Herold M, Graf C, Viera EA, Einsele H, Wajant H, Beilhack A. A TNFR2-Specific TNF Fusion Protein With Improved In Vivo Activity. Front Immunol 2022; 13:888274. [PMID: 35769484 PMCID: PMC9234581 DOI: 10.3389/fimmu.2022.888274] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/10/2022] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor (TNF) receptor-2 (TNFR2) has attracted considerable interest as a target for immunotherapy. Indeed, using oligomeric fusion proteins of single chain-encoded TNFR2-specific TNF mutants (scTNF80), expansion of regulatory T cells and therapeutic activity could be demonstrated in various autoinflammatory diseases, including graft-versus-host disease (GvHD), experimental autoimmune encephalomyelitis (EAE) and collagen-induced arthritis (CIA). With the aim to improve the in vivo availability of TNFR2-specific TNF fusion proteins, we used here the neonatal Fc receptor (FcRn)-interacting IgG1 molecule as an oligomerizing building block and generated a new TNFR2 agonist with improved serum retention and superior in vivo activity. Methods Single-chain encoded murine TNF80 trimers (sc(mu)TNF80) were fused to the C-terminus of an in mice irrelevant IgG1 molecule carrying the N297A mutation which avoids/minimizes interaction with Fcγ-receptors (FcγRs). The fusion protein obtained (irrIgG1(N297A)-sc(mu)TNF80), termed NewSTAR2 (New selective TNF-based agonist of TNF receptor 2), was analyzed with respect to activity, productivity, serum retention and in vitro and in vivo activity. STAR2 (TNC-sc(mu)TNF80 or selective TNF-based agonist of TNF receptor 2), a well-established highly active nonameric TNFR2-specific variant, served as benchmark. NewSTAR2 was assessed in various in vitro and in vivo systems. Results STAR2 (TNC-sc(mu)TNF80) and NewSTAR2 (irrIgG1(N297A)-sc(mu)TNF80) revealed comparable in vitro activity. The novel domain architecture of NewSTAR2 significantly improved serum retention compared to STAR2, which correlated with efficient binding to FcRn. A single injection of NewSTAR2 enhanced regulatory T cell (Treg) suppressive activity and increased Treg numbers by > 300% in vivo 5 days after treatment. Treg numbers remained as high as 200% for about 10 days. Furthermore, a single in vivo treatment with NewSTAR2 upregulated the adenosine-regulating ectoenzyme CD39 and other activation markers on Tregs. TNFR2-stimulated Tregs proved to be more suppressive than unstimulated Tregs, reducing conventional T cell (Tcon) proliferation and expression of activation markers in vitro. Finally, singular preemptive NewSTAR2 administration five days before allogeneic hematopoietic cell transplantation (allo-HCT) protected mice from acute GvHD. Conclusions NewSTAR2 represents a next generation ligand-based TNFR2 agonist, which is efficiently produced, exhibits improved pharmacokinetic properties and high serum retention with superior in vivo activity exerting powerful protective effects against acute GvHD.
Collapse
Affiliation(s)
- Juan Gamboa Vargas
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Jennifer Wagner
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Haroon Shaikh
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Isabell Lang
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Juliane Medler
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Mohamed Anany
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Department of Microbial Biotechnology, Institute of Biotechnology, Giza, Egypt
| | - Tim Steinfatt
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Josefina Peña Mosca
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Stephanie Haack
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Julia Dahlhoff
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Carolin Graf
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Estibaliz Arellano Viera
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research Laboratory, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| |
Collapse
|
21
|
Bossini-Castillo L, Glinos DA, Kunowska N, Golda G, Lamikanra AA, Spitzer M, Soskic B, Cano-Gamez E, Smyth DJ, Cattermole C, Alasoo K, Mann A, Kundu K, Lorenc A, Soranzo N, Dunham I, Roberts DJ, Trynka G. Immune disease variants modulate gene expression in regulatory CD4 + T cells. CELL GENOMICS 2022; 2:None. [PMID: 35591976 PMCID: PMC9010307 DOI: 10.1016/j.xgen.2022.100117] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 11/02/2021] [Accepted: 03/15/2022] [Indexed: 12/30/2022]
Abstract
Identifying cellular functions dysregulated by disease-associated variants could implicate novel pathways for drug targeting or modulation in cell therapies. However, follow-up studies can be challenging if disease-relevant cell types are difficult to sample. Variants associated with immune diseases point toward the role of CD4+ regulatory T cells (Treg cells). We mapped genetic regulation (quantitative trait loci [QTL]) of gene expression and chromatin activity in Treg cells, and we identified 133 colocalizing loci with immune disease variants. Colocalizations of immune disease genome-wide association study (GWAS) variants with expression QTLs (eQTLs) controlling the expression of CD28 and STAT5A, involved in Treg cell activation and interleukin-2 (IL-2) signaling, support the contribution of Treg cells to the pathobiology of immune diseases. Finally, we identified seven known drug targets suitable for drug repurposing and suggested 63 targets with drug tractability evidence among the GWAS signals that colocalized with Treg cell QTLs. Our study is the first in-depth characterization of immune disease variant effects on Treg cell gene expression modulation and dysregulation of Treg cell function.
Collapse
Affiliation(s)
| | - Dafni A. Glinos
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- New York Genome Center, New York, NY, USA
| | - Natalia Kunowska
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Gosia Golda
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Abigail A. Lamikanra
- NHS Blood and Transplant, Oxford, UK
- BRC Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Michaela Spitzer
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Blagoje Soskic
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Eddie Cano-Gamez
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - Deborah J. Smyth
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | | | - Kaur Alasoo
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Alice Mann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Kousik Kundu
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Anna Lorenc
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Nicole Soranzo
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Ian Dunham
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| | - David J. Roberts
- NHS Blood and Transplant, Oxford, UK
- BRC Haematology Theme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Gosia Trynka
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Cambridge, UK
| |
Collapse
|
22
|
Okuzono Y, Muraki Y, Sato S. TNFR2 pathways are fully active in cancer regulatory T cells. Biosci Biotechnol Biochem 2022; 86:351-361. [PMID: 35015831 DOI: 10.1093/bbb/zbab226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/28/2021] [Indexed: 12/11/2022]
Abstract
Tumor necrosis factor receptor 2 (TNFR2), a membrane-bound tumor necrosis factor receptor expressed by regulatory T cells (Tregs), participates in Treg proliferation. Although a specific TNFR2 pathway has been reported, the signaling mechanism has not been completely elucidated. This study sought to clarify TNFR2 signaling in human Tregs using amplicon sequencing and single-cell RNA sequencing to assess Tregs treated with a TNFR2 agonist antibody. Pathway enrichment analysis based on differentially expressed genes highlighted tumor necrosis factor α signaling via nuclear factor kappa B, interleukin-2 signal transducer and activator of transcription 5 signaling, interferon-γ response, and cell proliferation-related pathways in Tregs after TNFR2 activation. TNFR2-high Treg-focused analysis found that these pathways were fully activated in cancer Tregs, showing high TNFR2 expression. Collectively, these findings suggest that TNFR2 orchestrates multiple pathways in cancer Tregs, which could help cancer cells escape immune surveillance, making TNFR2 signaling a potential anticancer therapy target.
Collapse
Affiliation(s)
- Yuumi Okuzono
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Yo Muraki
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Shuji Sato
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| |
Collapse
|
23
|
Skartsis N, Peng Y, Ferreira LMR, Nguyen V, Ronin E, Muller YD, Vincenti F, Tang Q. IL-6 and TNFα Drive Extensive Proliferation of Human Tregs Without Compromising Their Lineage Stability or Function. Front Immunol 2022; 12:783282. [PMID: 35003100 PMCID: PMC8732758 DOI: 10.3389/fimmu.2021.783282] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 11/29/2021] [Indexed: 01/09/2023] Open
Abstract
Treg therapies are being tested in clinical trials in transplantation and autoimmune diseases, however, the impact of inflammation on Tregs remains controversial. We challenged human Tregs ex-vivo with pro-inflammatory cytokines IL-6 and TNFα and observed greatly enhanced proliferation stimulated by anti-CD3 and anti-CD28 (aCD3/28) beads or CD28 superagonist (CD28SA). The cytokine-exposed Tregs maintained high expression of FOXP3 and HELIOS, demethylated FOXP3 enhancer, and low IFNγ, IL-4, and IL-17 secretion. Blocking TNF receptor using etanercept or deletion of TNF receptor 2 using CRISPR/Cas9 blunted Treg proliferation and attenuated FOXP3 and HELIOS expression. These results prompted us to consider using CD28SA together with IL-6 and TNFα without aCD3/28 beads (beadless) as an alternative protocol for therapeutic Treg manufacturing. Metabolomics profiling revealed more active glycolysis and oxidative phosphorylation, increased energy production, and higher antioxidant potential during beadless Treg expansion. Finally, beadless expanded Tregs maintained suppressive functions in vitro and in vivo. These results demonstrate that human Tregs positively respond to proinflammatory cytokines with enhanced proliferation without compromising their lineage identity or function. This property can be harnessed for therapeutic Treg manufacturing.
Collapse
Affiliation(s)
- Nikolaos Skartsis
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States.,Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Yani Peng
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Leonardo M R Ferreira
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Vinh Nguyen
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Emilie Ronin
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Yannick D Muller
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States
| | - Flavio Vincenti
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States.,Division of Nephrology, Department of Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Qizhi Tang
- Department of Surgery, University of California San Francisco, San Francisco, CA, United States.,Diabetes Center, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
24
|
Hu Y, Xu C, Ren J, Zeng Y, Cao F, Fang H, Jintao G, Zhou Y, Li Q. Exposure to Tobacco Smoking Induces a subset of Activated Tumor-resident Tregs in Non-Small Cell Lung Cancer. Transl Oncol 2022; 15:101261. [PMID: 34768099 PMCID: PMC8591366 DOI: 10.1016/j.tranon.2021.101261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 11/09/2022] Open
Abstract
Tobacco smoking is the major cause of non-small-cell-lung cancer (NSCLC). However, it is barely known how smoking impact the tumor immune environment (TIME) of lung cancer. We integrated single-cell RNA-seq and bulk RNA-seq data from several studies to systematically study the impact of smoking on T cells in treatment naïve NSCLC patients. We defined a set of smoking-induced differentially expressed genes (SIDEGs) in different cells in TIME.. Specifically, we defined a smoking-related tumor-specific Treg subset, ADAM12+ CTLA4+ Tregs according to the trajectory analysis and highly express genes in cell adhesion pathways and lipid metabolism. Using independent datasets from treatment naïve patients, we found that the fraction of ADAM12+ CTLA4+ Tregs are significantly increased in patients with smoking history. Moreover, the fraction of ADAM12+ CTLA4+ Tregs are positively correlated with the fraction of exhausted T cells. Additionally, we reconstructed the spatial organization of the tumor immune microenvironment and found that ADAM12+ CTLA4+ Tregs more actively communicate with LAYN+CD8+ exhausted T cells compared with ADAM12-CTLA4+ Tregs. Our data demonstrate that smoking induced a unique subset of tumor-specific activated Tregs which interact with exhausted T cells in the TIME. Our findings not only explained how smoking impact the TIME but also provide new targets and biomarkers for precision immunotherapy of lung cancer.
Collapse
Affiliation(s)
- Yudi Hu
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China
| | - Chaoqun Xu
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China
| | - Jun Ren
- School of Informatics, Xiamen University, Xiamen, 361105, China
| | - Yuanyuan Zeng
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China
| | - Fengyang Cao
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China
| | - Hongkun Fang
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China
| | - Guo Jintao
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China
| | - Ying Zhou
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China.
| | - Qiyuan Li
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, 361102, China; Department of hematology, School of Medicine, Xiamen University, Xiamen, 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
25
|
Ahmad S, Hatmal MM, Lambuk L, Al-Hatamleh MAI, Alshaer W, Mohamud R. The role of TNFR2 + Tregs in COVID-19: An overview and a potential therapeutic strategy. Life Sci 2021; 286:120063. [PMID: 34673116 PMCID: PMC8523334 DOI: 10.1016/j.lfs.2021.120063] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 02/08/2023]
Abstract
COVID-19 is a multi-faceted disease ranging from asymptomatic to severely ill condition that primarily affects the lungs and could advance to other organs as well. It's causing factor, SARS-CoV-2 is recognized to develop robust cell-mediated immunity that responsible to either control or exaggerate the infection. As an important cell subset that control immune responses and are significantly dysregulated in COVID-19, Tregs is proposed to be considered for COVID-19 management. Among its hallmark, TNFR2 is recently recognized to play important role in the function and survival of Tregs. This review gathers available TNFR2 agonists to directly target Tregs as a potential approach to overcome immune dysregulation that affect the severity in COVID-19. Furthermore, this review performs a rigid body docking of TNF-TNFR2 interaction and such interaction with TNFR2 agonist to predict the optimal targeting approach.
Collapse
Affiliation(s)
- Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Ma'mon M Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan
| | - Lidawani Lambuk
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Mohammad A I Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Kelantan, Malaysia.
| |
Collapse
|
26
|
Lou Y, Wang J, Peng P, Wang S, Liu P, Xu LX. Downregulated TNF-α Levels after Cryo-Thermal Therapy Drive Tregs Fragility to Promote Long-Term Antitumor Immunity. Int J Mol Sci 2021; 22:ijms22189951. [PMID: 34576115 PMCID: PMC8468796 DOI: 10.3390/ijms22189951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/04/2021] [Accepted: 09/09/2021] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy has emerged as a therapeutic pillar in tumor treatment, but only a minority of patients get benefit. Overcoming the limitations of immunosuppressive environment is effective for immunotherapy. Moreover, host T cell activation and longevity within tumor are required for the long-term efficacy. In our previous study, a novel cryo-thermal therapy was developed to improve long-term survival in B16F10 melanoma and s.q. 4T1 breast cancer mouse models. We determined that cryo-thermal therapy induced Th1-dominant CD4+ T cell differentiation and the downregulation of Tregs in B16F10 model, contributing to tumor-specific and long-lasting immune protection. However, whether cryo-thermal therapy can affect the differentiation and function of T cells in a s.q. 4T1 model remains unknown. In this study, we also found that cryo-thermal therapy induced Th1-dominant differentiation of CD4+ T cells and the downregulation of effector Tregs. In particular, cryo-thermal therapy drove the fragility of Tregs and impaired their function. Furthermore, we discovered the downregulated level of serum tumor necrosis factor-α at the late stage after cryo-thermal therapy which played an important role in driving Treg fragility. Our findings revealed that cryo-thermal therapy could reprogram the suppressive environment and induce strong and durable antitumor immunity, which facilitate the development of combination strategies in immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Ping Liu
- Correspondence: (P.L.); (L.X.X.)
| | | |
Collapse
|
27
|
Insights into the biology and therapeutic implications of TNF and regulatory T cells. Nat Rev Rheumatol 2021; 17:487-504. [PMID: 34226727 DOI: 10.1038/s41584-021-00639-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2021] [Indexed: 02/06/2023]
Abstract
Treatments that block tumour necrosis factor (TNF) have major beneficial effects in several autoimmune and rheumatic diseases, including rheumatoid arthritis. However, some patients do not respond to TNF inhibitor treatment and rare occurrences of paradoxical disease exacerbation have been reported. These limitations on the clinical efficacy of TNF inhibitors can be explained by the differences between TNF receptor 1 (TNFR1) and TNFR2 signalling and by the diverse effects of TNF on multiple immune cells, including FOXP3+ regulatory T cells. This basic knowledge sheds light on the consequences of TNF inhibitor therapies on regulatory T cells in treated patients and on the limitations of such treatment in the control of diseases with an autoimmune component. Accordingly, the next generation of drugs targeting TNF is likely to be based on agents that selectively block the binding of TNF to TNFR1 and on TNFR2 agonists. These approaches could improve the treatment of rheumatic diseases in the future.
Collapse
|
28
|
Ghods A, Mehdipour F, Shariat M, Talei AR, Ghaderi A. Regulatory T cells express Tumor Necrosis Factor Receptor 2 with the highest intensity among CD4 + T cells in the draining lymph nodes of breast cancer. Mol Immunol 2021; 137:52-56. [PMID: 34214829 DOI: 10.1016/j.molimm.2021.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/27/2021] [Accepted: 06/20/2021] [Indexed: 12/16/2022]
Abstract
Tumor Necrosis Factor Receptor 2 (TNFR2) is one of the receptors of TNF-α, which is expressed on various cell types. TNFR2 signaling has a balancing role between regulatory and effector functions of T cells. Herein, we investigated the expression of TNFR2 on regulatory T cells (Tregs) and non-Tregs in breast tumor-draining lymph nodes. Mononuclear cells were isolated from 16 axillary lymph nodes, and the expressions of TNFR2, Foxp3 and CD25 were assessed in CD4+ T cells by flow cytometry. Our results showed that the majority of TNFR2+CD4+ T cells were Foxp3-CD25-. However, the percentage of TNFR2+ cells was significantly higher in Foxp3+CD25+CD4+ Tregs compared to Foxp3-CD25-CD4+, Foxp3+CD25-CD4+, and Foxp3-CD25+CD4+ T cell subsets. Among these subsets, Foxp3+CD25+TNFR2+CD4+ T cells were found to have the highest intensity of TNFR2 expression. The intensity of Foxp3 expression in Foxp3+CD25+TNFR2+CD4+ Treg cells was significantly higher than in their TNFR2- counterpart. Collectively, we showed that most of TNFR2+CD4+ T lymphocytes were Foxp3-CD25-, while the majority of Foxp3+CD25+CD4+ Tregs were TNFR2+, and they expressed TNFR2 with the highest intensity. This report highlights the importance of TNFR2 expression on Tregs and paves the way for further investigation of the effects of TNF-α on the suppressive activity of Tregs in the tumor microenvironment.
Collapse
Affiliation(s)
- Atri Ghods
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Mehdipour
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahmoud Shariat
- Department of Pathology, Shiraz Central Hospital, Shiraz, Iran
| | - Abdol-Rasoul Talei
- Breast Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
29
|
Mercogliano MF, Bruni S, Mauro F, Elizalde PV, Schillaci R. Harnessing Tumor Necrosis Factor Alpha to Achieve Effective Cancer Immunotherapy. Cancers (Basel) 2021; 13:cancers13030564. [PMID: 33540543 PMCID: PMC7985780 DOI: 10.3390/cancers13030564] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/17/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor necrosis factor alpha (TNFα) is a pleiotropic cytokine known to have contradictory roles in oncoimmunology. Indeed, TNFα has a central role in the onset of the immune response, inducing both activation and the effector function of macrophages, dendritic cells, natural killer (NK) cells, and B and T lymphocytes. Within the tumor microenvironment, however, TNFα is one of the main mediators of cancer-related inflammation. It is involved in the recruitment and differentiation of immune suppressor cells, leading to evasion of tumor immune surveillance. These characteristics turn TNFα into an attractive target to overcome therapy resistance and tackle cancer. This review focuses on the diverse molecular mechanisms that place TNFα as a source of resistance to immunotherapy such as monoclonal antibodies against cancer cells or immune checkpoints and adoptive cell therapy. We also expose the benefits of TNFα blocking strategies in combination with immunotherapy to improve the antitumor effect and prevent or treat adverse immune-related effects.
Collapse
Affiliation(s)
- María Florencia Mercogliano
- Laboratorio de Biofisicoquímica de Proteínas, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales-Consejo Nacional de Investigaciones Científicas y Técnicas (IQUIBICEN-CONICET), Buenos Aires 1428, Argentina;
| | - Sofía Bruni
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Florencia Mauro
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Patricia Virginia Elizalde
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
| | - Roxana Schillaci
- Laboratory of Molecular Mechanisms of Carcinogenesis, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires 1428, Argentina; (S.B.); (F.M.); (P.V.E.)
- Correspondence: ; Tel.: +54-11-4783-2869; Fax: +54-11-4786-2564
| |
Collapse
|
30
|
Wienke J, Brouwers L, van der Burg LM, Mokry M, Scholman RC, Nikkels PG, van Rijn BB, van Wijk F. Human Tregs at the materno-fetal interface show site-specific adaptation reminiscent of tumor Tregs. JCI Insight 2020; 5:137926. [PMID: 32809975 PMCID: PMC7526557 DOI: 10.1172/jci.insight.137926] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
Tregs are crucial for maintaining maternal immunotolerance against the semiallogeneic fetus. We investigated the elusive transcriptional profile and functional adaptation of human uterine Tregs (uTregs) during pregnancy. Uterine biopsies, from placental bed (materno-fetal interface) and incision site (control) and blood were obtained from women with uncomplicated pregnancies undergoing cesarean section. Tregs and CD4+ non-Tregs were isolated for transcriptomic profiling by Cel-Seq2. Results were validated on protein and single cell levels by flow cytometry. Placental bed uTregs showed elevated expression of Treg signature markers, including FOXP3, CTLA-4, and TIGIT. Their transcriptional profile was indicative of late-stage effector Treg differentiation and chronic activation, with increased expression of immune checkpoints GITR, TNFR2, OX-40, and 4-1BB; genes associated with suppressive capacity (HAVCR2, IL10, LAYN, and PDCD1); and transcription factors MAF, PRDM1, BATF, and VDR. uTregs mirrored non-Treg Th1 polarization and tissue residency. The particular transcriptional signature of placental bed uTregs overlapped strongly with that of tumor-infiltrating Tregs and was remarkably pronounced at the placental bed compared with uterine control site. In conclusion, human uTregs acquire a differentiated effector Treg profile similar to tumor-infiltrating Tregs, specifically at the materno-fetal interface. This introduces the concept of site-specific transcriptional adaptation of Tregs within 1 organ. Human regulatory T cells at the maternal-fetal interface show uterine site-specific functional adaptation with late-stage effector differentiation, chronic activation, Th1 polarization, and tumor-infiltrating, Treg-like features.
Collapse
Affiliation(s)
| | | | | | - Michal Mokry
- Regenerative Medicine Utrecht.,Laboratory of Clinical Chemistry and Hematology, and
| | | | - Peter Gj Nikkels
- Department of Pathology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht University, Netherlands
| | - Bas B van Rijn
- Wilhelmina Children's Hospital Birth Center.,Obstetrics and Fetal Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, Netherlands
| | | |
Collapse
|
31
|
Nejatbakhsh Samimi L, Farhadi E, Tahmasebi MN, Jamshidi A, Sharafat Vaziri A, Mahmoudi M. NF-κB signaling in rheumatoid arthritis with focus on fibroblast-like synoviocytes. AUTOIMMUNITY HIGHLIGHTS 2020. [PMCID: PMC7414649 DOI: 10.1186/s13317-020-00135-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The nuclear factor-κB (NF-κB) signaling pathway regulates multiple processes in innate and adaptive immune cells. This pathway is involved in inflammation through the regulation of cytokines, chemokines, and adhesion molecules expression. The NF-κB transcription factor also participates in the survival, proliferation, and differentiation of cells. Therefore, deregulated NF-κB activation contributes to the pathogenesis of inflammatory diseases. Rheumatoid arthritis (RA) is classified as a heterogeneous and complex autoimmune inflammatory disease. Although different immune and non-immune cells contribute to the RA pathogenesis, fibroblast-like synoviocytes (FLSs) play a crucial role in disease progression. These cells are altered during the disease and produce inflammatory mediators, including inflammatory cytokines and matrix metalloproteinases, which result in joint and cartilage erosion. Among different cell signaling pathways, it seems that deregulated NF-κB activation is associated with the inflammatory picture of RA. NF-κB activation can also promote the proliferation of RA-FLSs as well as the inhibition of FLS apoptosis that results in hyperplasia in RA synovium. In this review, the role of NF-κB transcription factor in immune and non-immune cells (especially FLSs) that are involved in RA pathogenesis are discussed.
Collapse
|
32
|
Motwani K, Peters LD, Vliegen WH, El-sayed AG, Seay HR, Lopez MC, Baker HV, Posgai AL, Brusko MA, Perry DJ, Bacher R, Larkin J, Haller MJ, Brusko TM. Human Regulatory T Cells From Umbilical Cord Blood Display Increased Repertoire Diversity and Lineage Stability Relative to Adult Peripheral Blood. Front Immunol 2020; 11:611. [PMID: 32351504 PMCID: PMC7174770 DOI: 10.3389/fimmu.2020.00611] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 03/17/2020] [Indexed: 12/22/2022] Open
Abstract
The human T lymphocyte compartment is highly dynamic over the course of a lifetime. Of the many changes, perhaps most notable is the transition from a predominantly naïve T cell state at birth to the acquisition of antigen-experienced memory and effector subsets following environmental exposures. These phenotypic changes, including the induction of T cell exhaustion and senescence, have the potential to negatively impact efficacy of adoptive T cell therapies (ACT). When considering ACT with CD4+CD25+CD127-/lo regulatory T cells (Tregs) for the induction of immune tolerance, we previously reported ex vivo expanded umbilical cord blood (CB) Tregs remained more naïve, suppressed responder T cells equivalently, and exhibited a more diverse T cell receptor (TCR) repertoire compared to expanded adult peripheral blood (APB) Tregs. Herein, we hypothesized that upon further characterization, we would observe increased lineage heterogeneity and phenotypic diversity in APB Tregs that might negatively impact lineage stability, engraftment capacity, and the potential for Tregs to home to sites of tissue inflammation following ACT. We compared the phenotypic profiles of human Tregs isolated from CB versus the more traditional source, APB. We conducted analysis of fresh and ex vivo expanded Treg subsets at both the single cell (scRNA-seq and flow cytometry) and bulk (microarray and cytokine profiling) levels. Single cell transcriptional profiles of pre-expansion APB Tregs highlighted a cluster of cells that showed increased expression of genes associated with effector and pro-inflammatory phenotypes (CCL5, GZMK, CXCR3, LYAR, and NKG7) with low expression of Treg markers (FOXP3 and IKZF2). CB Tregs were more diverse in TCR repertoire and homogenous in phenotype, and contained fewer effector-like cells in contrast with APB Tregs. Interestingly, expression of canonical Treg markers, such as FOXP3, TIGIT, and IKZF2, were increased in CB CD4+CD127+ conventional T cells (Tconv) compared to APB Tconv, post-expansion, implying perinatal T cells may adopt a default regulatory program. Collectively, these data identify surface markers (namely CXCR3) that could be depleted to improve purity and stability of APB Tregs, and support the use of expanded CB Tregs as a potentially optimal ACT modality for the treatment of autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Keshav Motwani
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Leeana D. Peters
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Willem H. Vliegen
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Ahmed Gomaa El-sayed
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Howard R. Seay
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - M. Cecilia Lopez
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Henry V. Baker
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Amanda L. Posgai
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Maigan A. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Daniel J. Perry
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Rhonda Bacher
- Department of Biostatistics, University of Florida, Gainesville, FL, United States
| | - Joseph Larkin
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Michael J. Haller
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Todd M. Brusko
- Department of Pathology, Immunology and Laboratory Medicine, Diabetes Institute, College of Medicine, University of Florida, Gainesville, FL, United States
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
33
|
Raffin C, Vo LT, Bluestone JA. T reg cell-based therapies: challenges and perspectives. Nat Rev Immunol 2020; 20:158-172. [PMID: 31811270 PMCID: PMC7814338 DOI: 10.1038/s41577-019-0232-6] [Citation(s) in RCA: 473] [Impact Index Per Article: 94.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2019] [Indexed: 12/25/2022]
Abstract
Cellular therapies using regulatory T (Treg) cells are currently undergoing clinical trials for the treatment of autoimmune diseases, transplant rejection and graft-versus-host disease. In this Review, we discuss the biology of Treg cells and describe new efforts in Treg cell engineering to enhance specificity, stability, functional activity and delivery. Finally, we envision that the success of Treg cell therapy in autoimmunity and transplantation will encourage the clinical use of adoptive Treg cell therapy for non-immune diseases, such as neurological disorders and tissue repair.
Collapse
Affiliation(s)
- Caroline Raffin
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Linda T Vo
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey A Bluestone
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
34
|
Lubrano di Ricco M, Ronin E, Collares D, Divoux J, Grégoire S, Wajant H, Gomes T, Grinberg-Bleyer Y, Baud V, Marodon G, Salomon BL. Tumor necrosis factor receptor family costimulation increases regulatory T-cell activation and function via NF-κB. Eur J Immunol 2020; 50:972-985. [PMID: 32012260 PMCID: PMC7383872 DOI: 10.1002/eji.201948393] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 12/10/2019] [Accepted: 01/30/2020] [Indexed: 01/06/2023]
Abstract
Several drugs targeting members of the TNF superfamily or TNF receptor superfamily (TNFRSF) are widely used in medicine or are currently being tested in therapeutic trials. However, their mechanism of action remains poorly understood. Here, we explored the effects of TNFRSF co-stimulation on murine Foxp3+ regulatory T cell (Treg) biology, as they are pivotal modulators of immune responses. We show that engagement of TNFR2, 4-1BB, GITR, and DR3, but not OX40, increases Treg proliferation and survival. Triggering these TNFRSF in Tregs induces similar changes in gene expression patterns, suggesting that they engage common signal transduction pathways. Among them, we identified a major role of canonical NF-κB. Importantly, TNFRSF co-stimulation improves the ability of Tregs to suppress colitis. Our data demonstrate that stimulation of discrete TNFRSF members enhances Treg activation and function through a shared mechanism. Consequently, therapeutic effects of drugs targeting TNFRSF or their ligands may be mediated by their effect on Tregs.
Collapse
Affiliation(s)
- Martina Lubrano di Ricco
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Emilie Ronin
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Davi Collares
- Université Paris Descartes, Sorbonne Paris Cité, Laboratoire NF-κB, Différenciation et Cancer, Paris, France
| | - Jordane Divoux
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sylvie Grégoire
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Harald Wajant
- Division Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Tomás Gomes
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Yenkel Grinberg-Bleyer
- Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052, CNRS 5286, Université Claude Bernard Lyon 1, Labex DEVweCAN, Centre Léon Bérard, Lyon, France
| | - Véronique Baud
- Université Paris Descartes, Sorbonne Paris Cité, Laboratoire NF-κB, Différenciation et Cancer, Paris, France
| | - Gilles Marodon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Benoît L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| |
Collapse
|
35
|
Padutsch T, Sendetski M, Huber C, Peters N, Pfizenmaier K, Bethea JR, Kontermann RE, Fischer R. Superior Treg-Expanding Properties of a Novel Dual-Acting Cytokine Fusion Protein. Front Pharmacol 2019; 10:1490. [PMID: 31920671 PMCID: PMC6930692 DOI: 10.3389/fphar.2019.01490] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/19/2019] [Indexed: 11/13/2022] Open
Abstract
Autoimmune diseases are caused by uncontrolled endogenous immune responses against healthy cells. They may develop due to an impaired function of regulatory T cells (Tregs), which normally suppress self-specific effector immune cells. Interleukin 2 (IL-2) and tumor necrosis factor (TNF) have been identified as key players that promote expansion, function, and stability of Tregs. In vivo, both low-dose IL-2 therapy and TNF receptor 2 (TNFR2) agonism were shown to expand Tregs and alleviate autoimmunity. We here designed a novel dimeric dual-acting fusion cytokine, where mouse IL-2 is genetically linked to a TNFR2-selective single-chain TNF mutein (IL2-EHD2-sc-mTNFR2). IL2-EHD2-sc-mTNFR2 showed high affinity to TNFR2 and efficiently activated IL-2 and TNFR2-selective signaling pathways. Further, IL2-EHD2-sc-mTNFR2 promoted superior Treg expansion, with both the IL-2 and the TNFR2 agonist (sc-mTNFR2) component necessary for this biological response. Ultimately, we propose that IL2-EHD2-sc-mTNFR2 is a dual-acting cytokine that efficiently promotes Treg expansion and might have a superior therapeutic window than conventional IL-2-based drugs.
Collapse
Affiliation(s)
- Tanja Padutsch
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Maksim Sendetski
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Carina Huber
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Nathalie Peters
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - John R Bethea
- Department of Biology, Drexel University, Philadelphia, PA, United States
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Roman Fischer
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Department of Biology, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
36
|
Kumar P, Lele SS, Ragothaman VK, Raghunathan D, Epstein AL, Chiba S, Prabhakar BS. OX40L-JAG1-Induced Expansion of Lineage-Stable Regulatory T Cells Involves Noncanonical NF-κB Signaling. THE JOURNAL OF IMMUNOLOGY 2019; 203:3225-3236. [PMID: 31704879 DOI: 10.4049/jimmunol.1900530] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/07/2019] [Indexed: 12/14/2022]
Abstract
Foxp3+T regulatory cells (Tregs) control autoimmune response by suppressing proliferation and effector functions of self-reactive Foxp3-CD4+/CD8+ T cells and thereby maintain the critical balance between self-tolerance and autoimmunity. Earlier, we had shown that OX40L-JAG1 cosignaling mediated through their cognate receptors OX40 and Notch3 preferentially expressed on murine Tregs can selectively induce their proliferation in the absence of TCR stimulation. However, the differential molecular mechanisms regulating TCR-independent versus TCR-dependent Treg proliferation and lineage stability of the expanded Tregs remained unknown. In this study, we show that OX40L-JAG1 treatment induced TCR-independent proliferation of Tregs in the thymus and periphery. The use of Src kinase inhibitor permitted us to demonstrate selective inhibition of TCR-dependent T cell proliferation with little to no effect on OX40L-JAG1-induced TCR-independent Treg expansion in vitro, which was critically dependent on noncanonical NF-κB signaling. OX40L-JAG1-expanded Tregs showed sustained lineage stability as indicated by stable demethylation marks in Treg signature genes such as Foxp3, Il2ra, Ctla4, Ikzf2, and Ikzf4. Furthermore, OX40L-JAG1 treatment significantly increased CTLA4+ and TIGIT+ Tregs and alleviated experimental autoimmune thyroiditis in mice. Relevance of our findings to humans became apparent when human OX40L and JAG1 induced TCR-independent selective expansion of human Tregs in thymocyte cultures and increased human Tregs in the liver tissue of humanized NSG mice. Our findings suggest that OX40L-JAG1-induced TCR-independent Treg proliferation is a conserved mechanism that can be used to expand lineage-stable Tregs to treat autoimmune diseases.
Collapse
Affiliation(s)
- Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Swarali Surendra Lele
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Vandhana K Ragothaman
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Divya Raghunathan
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612
| | - Alan L Epstein
- Department of Pathology, Keck School of Medicine at University of Southern California, Los Angeles, CA 900933
| | - Shigeru Chiba
- Department of Clinical and Experimental Hematology, Institute of Clinical Medicine, University of Tsukuba, Tsukuba 305-8575, Japan; and
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, Chicago, IL 60612; .,Department of Ophthalmology, University of Illinois College of Medicine, Chicago, IL 60612
| |
Collapse
|
37
|
Bansal SS, Ismahil MA, Goel M, Zhou G, Rokosh G, Hamid T, Prabhu SD. Dysfunctional and Proinflammatory Regulatory T-Lymphocytes Are Essential for Adverse Cardiac Remodeling in Ischemic Cardiomyopathy. Circulation 2019; 139:206-221. [PMID: 30586716 DOI: 10.1161/circulationaha.118.036065] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Heart failure (HF) is a state of inappropriately sustained inflammation, suggesting the loss of normal immunosuppressive mechanisms. Regulatory T-lymphocytes (Tregs) are considered key suppressors of immune responses; however, their role in HF is unknown. We hypothesized that Tregs are dysfunctional in ischemic cardiomyopathy and HF, and they promote immune activation and left ventricular (LV) remodeling. METHODS Adult male wild-type C57BL/6 mice, Foxp3-diphtheria toxin receptor transgenic mice, and tumor necrosis factor (TNF) α receptor-1 (TNFR1)-/- mice underwent nonreperfused myocardial infarction to induce HF or sham operation. LV remodeling was assessed by echocardiography as well as histological and molecular phenotyping. Alterations in Treg profile and function were examined by flow cytometry, immunostaining, and in vitro cell assays. RESULTS Compared with wild-type sham mice, CD4+Foxp3+ Tregs in wild-type HF mice robustly expanded in the heart, circulation, spleen, and lymph nodes in a phasic manner after myocardial infarction, beyond the early phase of wound healing, and exhibited proinflammatory T helper 1-type features with interferon-γ, TNFα, and TNFR1 expression, loss of immunomodulatory capacity, heightened proliferation, and potentiated antiangiogenic and profibrotic properties. Selective Treg ablation in Foxp3-diphtheria toxin receptor mice with ischemic cardiomyopathy reversed LV remodeling and dysfunction, alleviating hypertrophy and fibrosis, while suppressing circulating CD4+ T cells and systemic inflammation and enhancing tissue neovascularization. Tregs reconstituted after ablation exhibited restoration of immunosuppressive capacity and normalized TNFR1 expression. Treg dysfunction was also tightly coupled to Treg-endothelial cell contact- and TNFR1-dependent inhibition of angiogenesis and the mobilization and tissue infiltration of CD34+Flk1+ circulating angiogenic cells in a C-C chemokine ligand 5/C-C chemokine receptor 5-dependent manner. Anti-CD25-mediated Treg depletion in wild-type mice imparted similar benefits on LV remodeling, circulating angiogenic cells, and tissue neovascularization. CONCLUSIONS Proinflammatory and antiangiogenic Tregs play an essential pathogenetic role in chronic ischemic HF to promote immune activation and pathological LV remodeling. The restoration of normal Treg function may be a viable approach to therapeutic immunomodulation in this disease.
Collapse
Affiliation(s)
- Shyam S Bansal
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.).,The current affiliation for S.S.B. is Department of Physiology and Cell Biology, The Dorothy M Davis Heart & Lung Research Institute, Ohio State University Wexner Medical Center, Columbus
| | - Mohamed Ameen Ismahil
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Mehak Goel
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Guihua Zhou
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Gregg Rokosh
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Tariq Hamid
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.)
| | - Sumanth D Prabhu
- Division of Cardiovascular Disease and Comprehensive Cardiovascular Center, University of Alabama at Birmingham (S.S.B., M.A.I., M.G., G.Z., G.R., T.H., S.D.P.).,Medical Service, Birmingham VAMC, AL (S.D.P.)
| |
Collapse
|
38
|
Expression of costimulatory and inhibitory receptors in FoxP3 + regulatory T cells within the tumor microenvironment: Implications for combination immunotherapy approaches. Adv Cancer Res 2019; 144:193-261. [PMID: 31349899 DOI: 10.1016/bs.acr.2019.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The unprecedented success of immune checkpoint inhibitors has given rise to a rapidly growing number of immuno-oncology agents undergoing preclinical and clinical development and an exponential increase in possible combinations. Defining a clear rationale for combinations by identifying synergies between immunomodulatory pathways has therefore become a high priority. Immunosuppressive regulatory T cells (Tregs) within the tumor microenvironment (TME) represent a major roadblock to endogenous and therapeutic tumor immunity. However, Tregs are also essential for the maintenance of immunological self-tolerance, and share many molecular pathways with conventional T cells including cytotoxic T cells, the primary mediators of tumor immunity. Hence the inability to specifically target and neutralize Tregs within the TME of cancer patients without globally compromising self-tolerance poses a significant challenge. Here we review recent advances in the characterization of tumor-infiltrating Tregs with a focus on costimulatory and inhibitory receptors. We discuss receptor expression patterns, their functional role in Treg biology and mechanistic insights gained from targeting these receptors in preclinical models to evaluate their potential as clinical targets. We further outline a framework of parameters that could be used to refine the assessment of Tregs in cancer patients and increase their value as predictive biomarkers. Finally, we propose modalities to integrate our increasing knowledge on Treg phenotype and function for the rational design of checkpoint inhibitor-based combination therapies. Such combinations have great potential for synergy, as they could concomitantly enhance cytotoxic T cells and inhibit Tregs within the TME, thereby increasing the efficacy of current cancer immunotherapies.
Collapse
|
39
|
Mehdipour F, Razmkhah M, Faghih Z, Bagheri M, Talei AR, Ghaderi A. The significance of cytokine-producing B cells in breast tumor-draining lymph nodes. Cell Oncol (Dordr) 2019; 42:381-395. [DOI: 10.1007/s13402-019-00433-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2019] [Indexed: 01/17/2023] Open
|
40
|
Ghods A, Ghaderi A, Shariat M, Talei AR, Mehdipour F. TNFR2 but not TNFR1 is the main TNFR expressed by B and T lymphocytes in breast cancer draining lymph nodes. Immunol Lett 2019; 209:36-44. [DOI: 10.1016/j.imlet.2019.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/06/2019] [Accepted: 03/20/2019] [Indexed: 01/02/2023]
|
41
|
Carter CJ. Autism genes and the leukocyte transcriptome in autistic toddlers relate to pathogen interactomes, infection and the immune system. A role for excess neurotrophic sAPPα and reduced antimicrobial Aβ. Neurochem Int 2019; 126:36-58. [PMID: 30862493 DOI: 10.1016/j.neuint.2019.03.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/22/2019] [Accepted: 03/06/2019] [Indexed: 12/20/2022]
Abstract
Prenatal and early childhood infections have been implicated in autism. Many autism susceptibility genes (206 Autworks genes) are localised in the immune system and are related to immune/infection pathways. They are enriched in the host/pathogen interactomes of 18 separate microbes (bacteria/viruses and fungi) and to the genes regulated by bacterial toxins, mycotoxins and Toll-like receptor ligands. This enrichment was also observed for misregulated genes from a microarray study of leukocytes from autistic toddlers. The upregulated genes from this leukocyte study also matched the expression profiles in response to numerous infectious agents from the Broad Institute molecular signatures database. They also matched genes related to sudden infant death syndrome and autism comorbid conditions (autoimmune disease, systemic lupus erythematosus, diabetes, epilepsy and cardiomyopathy) as well as to estrogen and thyrotropin responses and to those upregulated by different types of stressors including oxidative stress, hypoxia, endoplasmic reticulum stress, ultraviolet radiation or 2,4-dinitrofluorobenzene, a hapten used to develop allergic skin reactions in animal models. The oxidative/integrated stress response is also upregulated in the autism brain and may contribute to myelination problems. There was also a marked similarity between the expression signatures of autism and Alzheimer's disease, and 44 shared autism/Alzheimer's disease genes are almost exclusively expressed in the blood-brain barrier. However, in contrast to Alzheimer's disease, levels of the antimicrobial peptide beta-amyloid are decreased and the levels of the neurotrophic/myelinotrophic soluble APP alpha are increased in autism, together with an increased activity of α-secretase. sAPPα induces an increase in glutamatergic and a decrease in GABA-ergic synapses creating and excitatory/inhibitory imbalance that has also been observed in autism. A literature survey showed that multiple autism genes converge on APP processing and that many are able to increase sAPPalpha at the expense of beta-amyloid production. A genetically programmed tilt of this axis towards an overproduction of neurotrophic/gliotrophic sAPPalpha and underproduction of antimicrobial beta-amyloid may explain the brain overgrowth and myelination dysfunction, as well as the involvement of pathogens in autism.
Collapse
Affiliation(s)
- C J Carter
- PolygenicPathways, 41C Marina, Saint Leonard's on Sea, TN38 0BU, East Sussex, UK.
| |
Collapse
|
42
|
Cammarata I, Martire C, Citro A, Raimondo D, Fruci D, Melaiu O, D'Oria V, Carone C, Peruzzi G, Cerboni C, Santoni A, Sidney J, Sette A, Paroli M, Caccavale R, Milanetti E, Riminucci M, Timperi E, Piconese S, Manzo A, Montecucco C, Scrivo R, Valesini G, Cariani E, Barnaba V. Counter-regulation of regulatory T cells by autoreactive CD8 + T cells in rheumatoid arthritis. J Autoimmun 2019; 99:81-97. [PMID: 30777378 DOI: 10.1016/j.jaut.2019.02.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 02/05/2019] [Accepted: 02/05/2019] [Indexed: 12/21/2022]
Abstract
The mechanisms whereby autoreactive T cells escape peripheral tolerance establishing thus autoimmune diseases in humans remain an unresolved question. Here, we demonstrate that autoreactive polyfunctional CD8+ T cells recognizing self-antigens (i.e., vimentin, actin cytoplasmic 1, or non-muscle myosin heavy chain 9 epitopes) with high avidity, counter-regulate Tregs by killing them, in a consistent percentage of rheumatoid arthritis (RA) patients. Indeed, these CD8+ T cells express a phenotype and gene profile of effector (eff) cells and, upon antigen-specific activation, kill Tregs indirectly in an NKG2D-dependent bystander fashion in vitro. This data provides a mechanistic basis for the finding showing that AE-specific (CD107a+) CD8+ T killer cells correlate, directly with the disease activity score, and inversely with the percentage of activated Tregs, in both steady state and follow-up studies in vivo. In addition, multiplex immunofluorescence imaging analyses of inflamed synovial tissues in vivo show that a remarkable number of CD8+ T cells express granzyme-B and selectively contact FOXP3+ Tregs, some of which are in an apoptotic state, validating hence the possibility that CD8+ Teff cells can counteract neighboring Tregs within inflamed tissues, by killing them. Alternatively, the disease activity score of a different subset of patients is correlated with the expansion of a peculiar subpopulation of autoreactive low avidity, partially-activated (pa)CD8+ T cells that, despite they conserve the conventional naïve (N) phenotype, produce high levels of tumor necrosis factor (TNF)-α and exhibit a gene expression signature of a progressive activation state. Tregs directly correlate with the expansion of this autoreactive (low avidity) paCD8+ TN cell subset in vivo, and efficiently control their differentiation rather their proliferation in vitro. Interestingly, autoreactive high avidity CD8+ Teff cells or low avidity paCD8+ TN cells are significantly expanded in RA patients who would become non-responders or patients who would become responders to TNF-α inhibitor therapy, respectively. These data provide evidence of a previously undescribed role of such mechanisms in the progression and therapy of RA.
Collapse
Affiliation(s)
- Ilenia Cammarata
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Carmela Martire
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Alessandra Citro
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Domenico Raimondo
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, 00161, Rome, Italy
| | - Doriana Fruci
- Dipartimento di Ematologia/Oncologia, Ospedale Pediatrico Bambino Gesù, IRCCS, 00165 Rome, Italy
| | - Ombretta Melaiu
- Dipartimento di Ematologia/Oncologia, Ospedale Pediatrico Bambino Gesù, IRCCS, 00165 Rome, Italy; Dipartimento di Biologia, Università di Pisa, 56126, Pisa, Italy
| | - Valentina D'Oria
- Core Facility Research Laboratories, Ospedale Pediatrico Bambino Gesù, IRCCS, 00165, Rome, Italy
| | - Chiara Carone
- Ospedale Civile S. Agostino-Estense, 41126, Modena, Italy
| | - Giovanna Peruzzi
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy
| | - Cristina Cerboni
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, 00161, Rome, Italy; Istituto Pasteur - Fondazione Cenci Bolognetti, 00185, Rome, Italy
| | - Angela Santoni
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, 00161, Rome, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy; Istituto Pasteur - Fondazione Cenci Bolognetti, 00185, Rome, Italy
| | - John Sidney
- La Jolla Institute for Allergy and Immunology, San Diego, CA, 92121, USA
| | - Alessandro Sette
- La Jolla Institute for Allergy and Immunology, San Diego, CA, 92121, USA
| | - Marino Paroli
- Dipartimento di Scienze e Biotecnologie Medico-Chirurgiche, Sapienza Università di Roma, Polo Pontino, 04100, Latina, Italy
| | - Rosalba Caccavale
- Dipartimento di Scienze e Biotecnologie Medico-Chirurgiche, Sapienza Università di Roma, Polo Pontino, 04100, Latina, Italy
| | - Edoardo Milanetti
- Dipartimento di Fisica, Sapienza Università di Roma, 00185, Rome, Italy
| | - Mara Riminucci
- Dipartimento di Medicina Molecolare, Sapienza Università di Roma, 00161, Rome, Italy
| | - Eleonora Timperi
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Silvia Piconese
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy; Istituto Pasteur - Fondazione Cenci Bolognetti, 00185, Rome, Italy
| | - Antonio Manzo
- Dipartimento di Medicina Interna e Terapia Medica, Fondazione IRCCS Policlinico "San Matteo", Università di Pavia, 27100, Pavia, Italy
| | - Carlomaurizio Montecucco
- Dipartimento di Medicina Interna e Terapia Medica, Fondazione IRCCS Policlinico "San Matteo", Università di Pavia, 27100, Pavia, Italy
| | - Rossana Scrivo
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | - Guido Valesini
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy
| | | | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, 00161, Rome, Italy; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, 00161, Rome, Italy; Istituto Pasteur - Fondazione Cenci Bolognetti, 00185, Rome, Italy.
| |
Collapse
|
43
|
Jung MK, Lee JS, Kwak JE, Shin EC. Tumor Necrosis Factor and Regulatory T Cells. Yonsei Med J 2019; 60:126-131. [PMID: 30666833 PMCID: PMC6342721 DOI: 10.3349/ymj.2019.60.2.126] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/17/2018] [Accepted: 11/17/2018] [Indexed: 12/13/2022] Open
Abstract
CD4⁺CD25⁺FoxP3⁺ regulatory T (Treg) cells play major roles in the maintenance of immune homeostasis. In this review, we comprehensively describe the relationship between tumor necrosis factor (TNF) and Treg cells, focusing on the effects of TNF on Treg cells and on TNF-producing Treg cells. Contradictory results have been reported for the effect of TNF on the suppressive activity of Treg cells. In patients with rheumatoid arthritis, TNF has been shown to reduce the suppressive activity of Treg cells. Meanwhile, however, TNF has also been reported to maintain the suppressive activity of Treg cells via a TNFR2-mediated mechanism. In addition, Treg cells have been found to acquire the ability to produce TNF under inflammatory conditions, such as acute viral hepatitis. These TNF-producing Treg cells exhibit T helper 17-like features and hold significance in various human diseases.
Collapse
Affiliation(s)
- Min Kyung Jung
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Jeong Seok Lee
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Jeong Eun Kwak
- BioMedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Eui Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
- BioMedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute of Science and Technology, Daejeon, Korea.
| |
Collapse
|
44
|
Differential roles of TNFα-TNFR1 and TNFα-TNFR2 in the differentiation and function of CD4 +Foxp3 + induced Treg cells in vitro and in vivo periphery in autoimmune diseases. Cell Death Dis 2019; 10:27. [PMID: 30631042 PMCID: PMC6328545 DOI: 10.1038/s41419-018-1266-6] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022]
Abstract
Tumor Necrosis Factor (TNF) α is a multifunctional cytokine with pro-inflammatory and anti-inflammatory characteristics. Increasing evidence suggests that thymus-derived, natural regulatory T cells (nTreg) express a remarkably high level of TNF Receptor 2 (TNFR2) and TNFα modulates the number or function of nTreg via TNFR2 in autoimmune diseases. Nonetheless, Treg cells consist of at least nTreg and iTreg that are induced in the periphery or in vitro and two subsets may have different biological characteristics. However, the role of TNF-TNFR signaling in development and function of these iTreg cells is less clear. In this study, we systemically studied the effect of TNFα and its receptor signals on iTreg differentiation, proliferation, and function in vitro and in vivo. We further investigated the expression and requirement of TNFR1 or TNFR2 expression on iTreg by utilizing TNFR1-/- and TNFR2-/- mice. We found that exogenous TNFα facilitated iTreg differentiation and function in vitro. TNFR2 deficiency hampered iTreg differentiation, proliferation, and function, while TNFR1 deficiency decreased the differentiation of inflammatory T cells such as Th1 and Th17 cells but maintained the regulatory capabilities of iTreg both in vitro and in vivo. Using colitis model, we also revealed TNFR2 but not TNFR1 deficiency compromised the iTreg functionality. Interestingly, inflammation affects TNFR expression on nTreg but not iTreg subset. Our results demonstrate that exogenous TNFα may enhance the differentiation and function of iTreg via TNFR2 signaling. The expression of TNFR2 on Treg might be downregulated in some autoimmune diseases, accompanied by an increased level of TNFR1. Thus, TNFR2 agonists or TNFR1-specific antagonists hold a potential promise for clinical application in treating patients with autoimmune diseases.
Collapse
|
45
|
Dostert C, Grusdat M, Letellier E, Brenner D. The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond. Physiol Rev 2019; 99:115-160. [DOI: 10.1152/physrev.00045.2017] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The tumor necrosis factor (TNF) and TNF receptor (TNFR) superfamilies (TNFSF/TNFRSF) include 19 ligands and 29 receptors that play important roles in the modulation of cellular functions. The communication pathways mediated by TNFSF/TNFRSF are essential for numerous developmental, homeostatic, and stimulus-responsive processes in vivo. TNFSF/TNFRSF members regulate cellular differentiation, survival, and programmed death, but their most critical functions pertain to the immune system. Both innate and adaptive immune cells are controlled by TNFSF/TNFRSF members in a manner that is crucial for the coordination of various mechanisms driving either co-stimulation or co-inhibition of the immune response. Dysregulation of these same signaling pathways has been implicated in inflammatory and autoimmune diseases, highlighting the importance of their tight regulation. Investigation of the control of TNFSF/TNFRSF activities has led to the development of therapeutics with the potential to reduce chronic inflammation or promote anti-tumor immunity. The study of TNFSF/TNFRSF proteins has exploded over the last 30 yr, but there remains a need to better understand the fundamental mechanisms underlying the molecular pathways they mediate to design more effective anti-inflammatory and anti-cancer therapies.
Collapse
Affiliation(s)
- Catherine Dostert
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Melanie Grusdat
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Elisabeth Letellier
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
46
|
Ahmad S, Azid NA, Boer JC, Lim J, Chen X, Plebanski M, Mohamud R. The Key Role of TNF-TNFR2 Interactions in the Modulation of Allergic Inflammation: A Review. Front Immunol 2018; 9:2572. [PMID: 30473698 PMCID: PMC6238659 DOI: 10.3389/fimmu.2018.02572] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/18/2018] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF) is a pleiotropic cytokine, which is thought to play a major role in the pathogenesis of inflammatory diseases, including allergy. TNF is produced at the early stage of allergen sensitization, and then continues to promote the inflammation cascade in the effector phase of allergic reactions. Consequently, anti-TNF treatment has been proposed as a potential therapeutic option. However, recent studies reveal anti-intuitive effects of TNF in the activation and proliferative expansion of immunosuppressive Tregs, tolerogenic DCs and MDSCs. This immunosuppressive effect of TNF is mediated by TNFR2, which is preferentially expressed by immunosuppressive cells. These findings redefine the role of TNF in allergic reaction, and suggest that targeting TNF-TNFR2 interaction itself may represent a novel strategy in the treatment of allergy.
Collapse
Affiliation(s)
- Suhana Ahmad
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Nor Azrini Azid
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Jennifer C Boer
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - JitKang Lim
- School of Chemical Engineering, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, China
| | | | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kelantan, Malaysia.,Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kelantan, Malaysia
| |
Collapse
|
47
|
Kumar P, Saini S, Khan S, Surendra Lele S, Prabhakar BS. Restoring self-tolerance in autoimmune diseases by enhancing regulatory T-cells. Cell Immunol 2018; 339:41-49. [PMID: 30482489 DOI: 10.1016/j.cellimm.2018.09.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/14/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022]
Abstract
Self-tolerance, the state of unresponsiveness to self-tissues/antigens, is maintained through central and peripheral tolerance mechanisms, and a breach of these mechanisms leads to autoimmune diseases. Foxp3 + T-regulatory cells (Tregs) play an essential role in suppressing autoimmune response directed against self-antigens and thereby regulate self-tolerance. Natural Tregs are differentiated in the thymus on the basis of their higher TCR-affinity to self-antigens and migrate to the periphery where they maintain peripheral tolerance. In addition, extra-thymic differentiation of induced Tregs can occur in the periphery which can control abrupt immune responses under inflammatory conditions. A defect in Treg cell numbers and/or function is found to be associated with the development of autoimmune disease in several experimental models and human autoimmune diseases. Moreover, augmentation of Tregs has been shown to be beneficial in treating autoimmunity in preclinical models, and Treg based cellular therapy has shown initial promise in clinical trials. However, emerging studies have identified an unstable subpopulation of Tregs which expresses pro-inflammatory cytokines under both homeostatic and autoimmune conditions, as well as in ex vivo cultures. In addition, clinical translation of Treg cellular therapy is impeded by limitations such as lack of easier methods for selective expansion of Tregs and higher cost associated with GMP-facilities required for cell sorting, ex vivo expansion and infusion of ex vivo expanded Tregs. Here, we discuss the recent advances in molecular mechanisms regulating Treg differentiation, Foxp3 expression and lineage stability, the role of Tregs in the prevention of various autoimmune diseases, and critically review their clinical utility for treating human autoimmune diseases.
Collapse
Affiliation(s)
- Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Shikha Saini
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Saad Khan
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Swarali Surendra Lele
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA.
| |
Collapse
|
48
|
Colombo M, Mirandola L, Chiriva-Internati M, Basile A, Locati M, Lesma E, Chiaramonte R, Platonova N. Cancer Cells Exploit Notch Signaling to Redefine a Supportive Cytokine Milieu. Front Immunol 2018; 9:1823. [PMID: 30154786 PMCID: PMC6102368 DOI: 10.3389/fimmu.2018.01823] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 07/24/2018] [Indexed: 12/19/2022] Open
Abstract
Notch signaling is a well-known key player in the communication between adjacent cells during organ development, when it controls several processes involved in cell differentiation. Notch-mediated communication may occur through the interaction of Notch receptors with ligands on adjacent cells or by a paracrine/endocrine fashion, through soluble molecules that can mediate the communication between cells at distant sites. Dysregulation of Notch pathway causes a number of disorders, including cancer. Notch hyperactivation may be caused by mutations of Notch-related genes, dysregulated upstream pathways, or microenvironment signals. Cancer cells may exploit this aberrant signaling to "educate" the surrounding microenvironment cells toward a pro-tumoral behavior. This may occur because of key cytokines secreted by tumor cells or it may involve the microenvironment through the activation of Notch signaling in stromal cells, an event mediated by a direct cell-to-cell contact and resulting in the increased secretion of several pro-tumorigenic cytokines. Up to now, review articles were mainly focused on Notch contribution in a specific tumor context or immune cell populations. Here, we provide a comprehensive overview on the outcomes of Notch-mediated pathological interactions in different tumor settings and on the molecular and cellular mediators involved in this process. We describe how Notch dysregulation in cancer may alter the cytokine network and its outcomes on tumor progression and antitumor immune response.
Collapse
Affiliation(s)
- Michela Colombo
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Maurizio Chiriva-Internati
- Kiromic Biopharma Inc., Houston, TX, United States.,Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrea Basile
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Massimo Locati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Milano, Italy.,Humanitas Clinical and Research Center, Rozzano, Italy
| | - Elena Lesma
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| | | | - Natalia Platonova
- Department of Health Sciences, Università degli Studi di Milano, Milano, Italy
| |
Collapse
|
49
|
Wang J, Ferreira R, Lu W, Farrow S, Downes K, Jermutus L, Minter R, Al-Lamki RS, Pober JS, Bradley JR. TNFR2 ligation in human T regulatory cells enhances IL2-induced cell proliferation through the non-canonical NF-κB pathway. Sci Rep 2018; 8:12079. [PMID: 30104686 PMCID: PMC6089958 DOI: 10.1038/s41598-018-30621-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/01/2018] [Indexed: 01/11/2023] Open
Abstract
Human T regulatory cells (T regs) express high levels of TNF receptor 2 (TNFR2). Ligation of TNFR2 with TNF, which can recognise both TNFR1 and TNFR2, or with a TNFR2-selective binding molecule, DARPin 18 (D18) activates canonical NF-κB signalling, assessed by IκBα degradation, and the magnitude of the response correlates with the level of TNFR2 expression. RNA-seq analysis of TNF- or D18-treated human T regs revealed that TNFR2 ligation induces transcription of NFKB2 and RELB, encoding proteins that form the non-canonical NF-κB transcription factor. In combination with IL2, D18 treatment is specific for T regs in (1) stabilising NF-κB-inducing kinase protein, the activator of non-canonical NF-κB signalling, (2) inducing translocation of RelB from cytosol to nucleus, (3) increasing cell cycle entry, and (4) increasing cell numbers. However, the regulatory function of the expanded T regs is unaltered. Inhibition of RelB nuclear translocation blocks the proliferative response. We conclude that ligation of TNFR2 by D18 enhances IL2-induced T regs proliferation and expansion in cell number through the non-canonical NF-κB pathway.
Collapse
Affiliation(s)
- Jun Wang
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom.
| | - Ricardo Ferreira
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Wanhua Lu
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Samatha Farrow
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Lutz Jermutus
- MedImmune Ltd., Granta Park, Cambridge, CB21 6GH, UK
| | - Ralph Minter
- MedImmune Ltd., Granta Park, Cambridge, CB21 6GH, UK
| | - Rafia S Al-Lamki
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Jordan S Pober
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - John R Bradley
- Department of Medicine, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
50
|
He T, Liu S, Chen S, Ye J, Wu X, Bian Z, Chen X. The p38 MAPK Inhibitor SB203580 Abrogates Tumor Necrosis Factor-Induced Proliferative Expansion of Mouse CD4 +Foxp3 + Regulatory T Cells. Front Immunol 2018; 9:1556. [PMID: 30038619 PMCID: PMC6046375 DOI: 10.3389/fimmu.2018.01556] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 06/25/2018] [Indexed: 12/27/2022] Open
Abstract
There is now compelling evidence that tumor necrosis factor (TNF) preferentially activates and expands CD4+Foxp3+ regulatory T cells (Tregs) through TNF receptor type II (TNFR2). However, it remains unclear which signaling transduction pathway(s) of TNFR2 is required for the stimulation of Tregs. Previously, it was shown that the interaction of TNF–TNFR2 resulted in the activation of a number of signaling pathways, including p38 MAPK, NF-κB, in T cells. We thus examined the role of p38 MAPK and NF-κB in TNF-mediated activation of Tregs, by using specific small molecule inhibitors. The results show that treatment with specific p38 MAPK inhibitor SB203580, rather than NF-κB inhibitors (Sulfasalazine and Bay 11-7082), abrogated TNF-induced expansion of Tregs in vitro. Furthermore, upregulation of TNFR2 and Foxp3 expression in Tregs by TNF was also markedly inhibited by SB203580. The proliferative expansion and the upregulation of TNFR2 expression on Tregs in LPS-treated mice were mediated by TNF–TNFR2 interaction, as shown by our previous study. The expansion of Tregs in LPS-treated mice were also markedly inhibited by in vivo treatment with SB203580. Taken together, our data clearly indicate that the activation of p38 MAPK is attributable to TNF/TNFR2-mediated activation and proliferative expansion of Tregs. Our results also suggest that targeting of p38 MAPK by pharmacological agent may represent a novel strategy to up- or downregulation of Treg activity for therapeutic purposes.
Collapse
Affiliation(s)
- Tianzhen He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Shuoyang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Shaokui Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Jingyi Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Xueqiang Wu
- Department of Oncology, Beijing Aerospace General Hospital, Beijing, China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xin Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|