1
|
Hundhausen N, Majumder S, Xiao Y, Haeusl SS, Goehler H, Seal R, Chiarolla CM, Rosenwald A, Eyrich M, Cicin-Sain L, Berberich-Siebelt F. NFAT single-deficient murine T cells reduce the risk of aGvHD while controlling cytomegalovirus infection. iScience 2025; 28:111937. [PMID: 40028277 PMCID: PMC11872454 DOI: 10.1016/j.isci.2025.111937] [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: 10/24/2024] [Revised: 12/13/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
NFAT is a family of transcription factors whose activation is inhibited by calcineurin inhibitors (CNIs). In allogeneic hematopoietic stem cell transplantation (allo-HCT), CNIs are employed to prevent and treat graft-versus-host disease (GvHD). Unfortunately, control of cytomegalovirus (CMV), which exacerbates clinical outcomes, is simultaneously lost. Since single NFAT deficiency in T cells ameliorates GvHD in our major mismatch model, we investigated whether protection is maintained during CMV infection. Reassuringly, NFAT-deficient T cells still improved GvHD upon acute CMV infection and after allo-HCT in latently CMV-infected mice, showing reduced proinflammatory and cytotoxic potential. In sharp contrast, CMV-specific NFAT-deficient CD8+ inflated memory T cells expanded more and with higher levels of interferon gamma (IFN-γ) and GzmB expression, effectively controlling CMV. Notably, NFAT-deficient inflated memory T cells could migrate to non-lymphoid tissues and fight CMV. Therefore, CMV infection does not interfere with the protective effect of NFAT inhibition to attenuate GvHD while allowing an anti-CMV response.
Collapse
Affiliation(s)
- Nadine Hundhausen
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Snigdha Majumder
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Yin Xiao
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Sigrun S. Haeusl
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Helen Goehler
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Rishav Seal
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | | | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Matthias Eyrich
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Luka Cicin-Sain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a Joint Venture of Helmholtz Centre for Infection Research and Medical School Hannover, Hannover, Germany
| | | |
Collapse
|
2
|
Liang C, Spoerl S, Xiao Y, Habenicht KM, Haeusl SS, Sandner I, Winkler J, Strieder N, Eder R, Stanewsky H, Alexiou C, Dudziak D, Rosenwald A, Edinger M, Rehli M, Hoffmann P, Winkler TH, Berberich-Siebelt F. Oligoclonal CD4 +CXCR5 + T cells with a cytotoxic phenotype appear in tonsils and blood. Commun Biol 2024; 7:879. [PMID: 39025930 PMCID: PMC11258247 DOI: 10.1038/s42003-024-06563-1] [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: 09/15/2023] [Accepted: 07/05/2024] [Indexed: 07/20/2024] Open
Abstract
In clinical situations, peripheral blood accessible CD3+CD4+CXCR5+ T-follicular helper (TFH) cells may have to serve as a surrogate indicator for dysregulated germinal center responses in tissues. To determine the heterogeneity of TFH cells in peripheral blood versus tonsils, CD3+CD4+CD45RA-CXCR5+ cells of both origins were sorted. Transcriptomes, TCR repertoires and cell-surface protein expression were analysed by single-cell RNA sequencing, flow cytometry and immunohistochemistry. Reassuringly, all blood-circulating CD3+CD4+CXCR5+ T-cell subpopulations also appear in tonsils, there with some supplementary TFH characteristics, while peripheral blood-derived TFH cells display markers of proliferation and migration. Three further subsets of TFH cells, however, with bona fide T-follicular gene expression patterns, are exclusively found in tonsils. One additional, distinct and oligoclonal CD4+CXCR5+ subpopulation presents pronounced cytotoxic properties. Those 'killer TFH (TFK) cells' can be discovered in peripheral blood as well as among tonsillar cells but are located predominantly outside of germinal centers. They appear terminally differentiated and can be distinguished from all other TFH subsets by expression of NKG7 (TIA-1), granzymes, perforin, CCL5, CCR5, EOMES, CRTAM and CX3CR1. All in all, this study provides data for detailed CD4+CXCR5+ T-cell assessment of clinically available blood samples and extrapolation possibilities to their tonsil counterparts.
Collapse
Affiliation(s)
- Chunguang Liang
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Silvia Spoerl
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Yin Xiao
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Katharina M Habenicht
- Division of Genetics, Department Biology, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Sigrun S Haeusl
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Isabel Sandner
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | - Julia Winkler
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
| | | | - Rüdiger Eder
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | | | - Christoph Alexiou
- Department of Otorhinolaryngology, Head & Neck Surgery, Else Kröner-Fresenius-Foundation-Professorship, Section of Experimental Oncology & Nanomedicine (SEON), University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
- Institute of Immunology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Matthias Edinger
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Michael Rehli
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Petra Hoffmann
- Leibniz Institute for Immunotherapy, Regensburg, Germany
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Thomas H Winkler
- Division of Genetics, Department Biology, Nikolaus-Fiebiger-Center of Molecular Medicine, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
3
|
Chaudhry MZ, Borkner L, Kulkarni U, Berberich-Siebelt F, Cicin-Sain L. NFAT signaling is indispensable for persistent memory responses of MCMV-specific CD8+ T cells. PLoS Pathog 2024; 20:e1012025. [PMID: 38346075 PMCID: PMC10890734 DOI: 10.1371/journal.ppat.1012025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 02/23/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Cytomegalovirus (CMV) induces a unique T cell response, where antigen-specific populations do not contract, but rather inflate during viral latency. It has been proposed that subclinical episodes of virus reactivation feed the inflation of CMV-specific memory cells by intermittently engaging T cell receptors (TCRs), but evidence of TCR engagement has remained lacking. Nuclear factor of activated T cells (NFAT) is a family of transcription factors, where NFATc1 and NFATc2 signal downstream of TCR in mature T lymphocytes. We show selective impacts of NFATc1 and/or NFATc2 genetic ablations on the long-term inflation of MCMV-specific CD8+ T cell responses despite largely maintained responses to acute infection. NFATc1 ablation elicited robust phenotypes in isolation, but the strongest effects were observed when both NFAT genes were missing. CMV control was impaired only when both NFATs were deleted in CD8+ T cells used in adoptive immunotherapy of immunodeficient mice. Transcriptome analyses revealed that T cell intrinsic NFAT is not necessary for CD8+ T cell priming, but rather for their maturation towards effector-memory and in particular the effector cells, which dominate the pool of inflationary cells.
Collapse
Affiliation(s)
- M. Zeeshan Chaudhry
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lisa Borkner
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Upasana Kulkarni
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Luka Cicin-Sain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine, a joint venture of Helmholtz Centre for Infection Research and Medical School Hannover, Hannover, Germany
| |
Collapse
|
4
|
Harris R, Karimi M. Dissecting the regulatory network of transcription factors in T cell phenotype/functioning during GVHD and GVT. Front Immunol 2023; 14:1194984. [PMID: 37441063 PMCID: PMC10333690 DOI: 10.3389/fimmu.2023.1194984] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
Transcription factors play a major role in regulation and orchestration of immune responses. The immunological context of the response can alter the regulatory networks required for proper functioning. While these networks have been well-studied in canonical immune contexts like infection, the transcription factor landscape during alloactivation remains unclear. This review addresses how transcription factors contribute to the functioning of mature alloactivated T cells. This review will also examine how these factors form a regulatory network to control alloresponses, with a focus specifically on those factors expressed by and controlling activity of T cells of the various subsets involved in graft-versus-host disease (GVHD) and graft-versus-tumor (GVT) responses.
Collapse
Affiliation(s)
- Rebecca Harris
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| | - Mobin Karimi
- Department of Microbiology and Immunology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
5
|
Gao R, Li A, Li S, Li X, Zhang S, Zhang X, Xu J. Induced regulatory T cells modified by knocking down T-bet in combination with ectopic expression of inhibitory cytokines effectively protect Graft-versus-Host Disease. Am J Transplant 2023:S1600-6135(23)00415-X. [PMID: 37084847 DOI: 10.1016/j.ajt.2023.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 04/23/2023]
Abstract
Induced regulatory T (iTreg) cells play a vital role in immune tolerance and in controlling chronic inflammation. Generated in the periphery, iTreg cells are suitable for responding to alloantigens and preventing transplant rejection. Nevertheless, their clinical application has been impeded by the plasticity and instability attributed to the loss of Foxp3 expression, raising concerns that iTreg may be converted to Teff cells and even exert a pathogenic effect. Herein, second-generation short hairpin RNAs (shRNAs) loaded with three pairs of small interfering RNAs (siRNAs) were utilized to target the transcription factor T-bet. In addition, two immunosuppressive cytokines, namely transforming growth factor beta (TGF-β) and interleukin-10 (IL-10), were constitutively expressed. This novel engineering strategy allowed the generation of stably-induced iTreg cells (SI Treg), which maintained the expression of Foxp3 even in an unfavorable environment and exerted potent immunosuppressive functions in vitro. Furthermore, SI Treg cells demonstrated an effector transcriptional profile. Finally, SI Treg showed a significant protective effect against GVHD-related deaths in a xenotransplantation model. Collectively, these results signify that SI Treg cells hold great promise for future clinical application and offer a rational therapeutic approach for transplant rejection.
Collapse
Affiliation(s)
- Rongrong Gao
- Clinical Center for Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| | - Ang Li
- Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 201508, P. R. China
| | - Sen Li
- Clinical Center for Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| | - Xiangrong Li
- Clinical Center for Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| | - Shuye Zhang
- Clinical Center for Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China
| | - Xiaoyan Zhang
- Clinical Center for Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China; Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 201508, P. R. China.
| | - Jianqing Xu
- Clinical Center for Biotherapy at Zhongshan Hospital & Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, P. R. China; Shanghai Public Health Clinical Center, Shanghai Medical College, Fudan University, Shanghai, 201508, P. R. China.
| |
Collapse
|
6
|
Mohammadpour H, Tsuji T, MacDonald CR, Sarow JL, Rosenheck H, Daneshmandi S, Choi JE, Qiu J, Matsuzaki J, Witkiewicz AK, Attwood K, Blazar BR, Odunsi K, Repasky EA, McCarthy PL. Galectin-3 expression in donor T cells reduces GvHD severity and lethality after allogeneic hematopoietic cell transplantation. Cell Rep 2023; 42:112250. [PMID: 36924493 PMCID: PMC10116561 DOI: 10.1016/j.celrep.2023.112250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 01/05/2023] [Accepted: 02/25/2023] [Indexed: 03/17/2023] Open
Abstract
Abundant donor cytotoxic T cells that attack normal host organs remain a major problem for patients receiving allogeneic hematopoietic cell transplantation (allo-HCT). Despite an increase in our knowledge of the pathobiology of acute graft versus host disease (aGvHD), the mechanisms regulating the proliferation and function of donor T cells remain unclear. Here, we show that activated donor T cells express galectin-3 (Gal-3) after allo-HCT. In both major and minor histocompatibility-mismatched models of murine aGvHD, expression of Gal-3 is associated with decreased T cell activation and suppression of the secretion of effector cytokines, including IFN-γ and GM-CSF. Mechanistically, Gal-3 results in activation of NFAT signaling, which can induce T cell exhaustion. Gal-3 overexpression in human T cells prevents severe disease by suppressing cytotoxic T cells in xenogeneic aGvHD models. Together, these data identify the Gal-3-dependent regulatory pathway in donor T cells as a critical component of inflammation in aGvHD.
Collapse
Affiliation(s)
- Hemn Mohammadpour
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| | - Takemasa Tsuji
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Cameron R MacDonald
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Joseph L Sarow
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Hanna Rosenheck
- Department of Medicine, Transplant and Cellular Therapy Program, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Saeed Daneshmandi
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA; Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Jee Eun Choi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Jingxin Qiu
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Junko Matsuzaki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Agnieszka K Witkiewicz
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Kristopher Attwood
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Kunle Odunsi
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA
| | - Philip L McCarthy
- Center for Immunotherapy, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA.
| |
Collapse
|
7
|
Walton K, Walker K, Riddle M, Koehn BH, Reff J, Sagatys EM, Linden MA, Pidala J, Kim J, Lee MC, Kiluk JV, Hui JYC, Yun SY, Xing Y, Stefanski H, Lawrence HR, Lawrence NJ, Tolar J, Anasetti C, Blazar BR, Sebti SM, Betts BC. Dual JAK2/Aurora kinase A inhibition prevents human skin graft rejection by allo-inactivation and ILC2-mediated tissue repair. Am J Transplant 2022; 22:717-730. [PMID: 34668635 PMCID: PMC8897228 DOI: 10.1111/ajt.16870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 01/25/2023]
Abstract
Prevention of allograft rejection often requires lifelong immune suppression, risking broad impairment of host immunity. Nonselective inhibition of host T cell function increases recipient risk of opportunistic infections and secondary malignancies. Here we demonstrate that AJI-100, a dual inhibitor of JAK2 and Aurora kinase A, ameliorates skin graft rejection by human T cells and provides durable allo-inactivation. AJI-100 significantly reduces the frequency of skin-homing CLA+ donor T cells, limiting allograft invasion and tissue destruction by T effectors. AJI-100 also suppresses pathogenic Th1 and Th17 cells in the spleen yet spares beneficial regulatory T cells. We show dual JAK2/Aurora kinase A blockade enhances human type 2 innate lymphoid cell (ILC2) responses, which are capable of tissue repair. ILC2 differentiation mediated by GATA3 requires STAT5 phosphorylation (pSTAT5) but is opposed by STAT3. Further, we demonstrate that Aurora kinase A activation correlates with low pSTAT5 in ILC2s. Importantly, AJI-100 maintains pSTAT5 levels in ILC2s by blocking Aurora kinase A and reduces interference by STAT3. Therefore, combined JAK2/Aurora kinase A inhibition is an innovative strategy to merge immune suppression with tissue repair after transplantation.
Collapse
Affiliation(s)
- Kelly Walton
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Kirsti Walker
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Megan Riddle
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Brent H. Koehn
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jordan Reff
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA
| | - Elizabeth M. Sagatys
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA,Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Michael A. Linden
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Joseph Pidala
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA,Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Blood and Marrow Transplantation – Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, USA
| | - Marie C Lee
- Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - John V. Kiluk
- Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Breast Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | | | - Sang Y. Yun
- Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA
| | - Yan Xing
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Heather Stefanski
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Harshani R. Lawrence
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA
| | - Nicholas J. Lawrence
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL, USA
| | - Jakub Tolar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Claudio Anasetti
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, USA,Department of Oncologic Sciences, Moffitt Cancer Center, Tampa, FL, USA,Department of Blood and Marrow Transplantation – Cellular Immunotherapy, Moffitt Cancer Center, Tampa, FL, USA
| | - Bruce R. Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Said M. Sebti
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia USA
| | - Brian C. Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
8
|
Koenig A, Vaeth M, Xiao Y, Chiarolla CM, Erapaneedi R, Klein M, Dietz L, Hundhausen N, Majumder S, Schuessler F, Bopp T, Klein-Hessling S, Rosenwald A, Berberich I, Berberich-Siebelt F. NFATc1/αA and Blimp-1 Support the Follicular and Effector Phenotype of Tregs. Front Immunol 2022; 12:791100. [PMID: 35069572 PMCID: PMC8770984 DOI: 10.3389/fimmu.2021.791100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
CD4+CXCR5+Foxp3+ T-follicular regulatory (TFR) cells control the germinal center responses. Like T-follicular helper cells, they express high levels of Nuclear Factor of Activated T-cells c1, predominantly its short isoform NFATc1/αA. Ablation of NFATc1 in Tregs prevents upregulation of CXCR5 and migration of TFR cells into B-cell follicles. By contrast, constitutive active NFATc1/αA defines the surface density of CXCR5, whose level determines how deep a TFR migrates into the GC and how effectively it controls antibody production. As one type of effector Treg, TFR cells express B lymphocyte-induced maturation protein-1 (Blimp-1). Blimp-1 can directly repress Cxcr5 and NFATc1/αA is necessary to overcome this Blimp-1-mediated repression. Interestingly, Blimp-1 even reinforces the recruitment of NFATc1 to Cxcr5 by protein-protein interaction and by those means cooperates with NFATc1 for Cxcr5 transactivation. On the contrary, Blimp-1 is necessary to counterbalance NFATc1/αA and preserve the Treg identity. This is because although NFATc1/αA strengthens the follicular development of Tregs, it bears the inherent risk of causing an ex-Treg phenotype.
Collapse
Affiliation(s)
- Anika Koenig
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Martin Vaeth
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Yin Xiao
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Raghu Erapaneedi
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany
| | - Lena Dietz
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Snigdha Majumder
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Felix Schuessler
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI), University Medical Center, University of Mainz, Mainz, Germany.,University Cancer Center Mainz, University Medical Center, University of Mainz, Mainz, Germany.,German Cancer Consortium (DKTK), Frankfurt/Mainz, Germany
| | - Stefan Klein-Hessling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, Würzburg, Germany.,Comprehensive Cancer Centre Mainfranken, University of Würzburg, Würzburg, Germany
| | - Ingolf Berberich
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
9
|
Campe J, Ullrich E. T Helper Cell Lineage-Defining Transcription Factors: Potent Targets for Specific GVHD Therapy? Front Immunol 2022; 12:806529. [PMID: 35069590 PMCID: PMC8766661 DOI: 10.3389/fimmu.2021.806529] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Allogenic hematopoietic stem cell transplantation (allo-HSCT) represents a potent and potentially curative treatment for many hematopoietic malignancies and hematologic disorders in adults and children. The donor-derived immunity, elicited by the stem cell transplant, can prevent disease relapse but is also responsible for the induction of graft-versus-host disease (GVHD). The pathophysiology of acute GVHD is not completely understood yet. In general, acute GVHD is driven by the inflammatory and cytotoxic effect of alloreactive donor T cells. Since several experimental approaches indicate that CD4 T cells play an important role in initiation and progression of acute GVHD, the contribution of the different CD4 T helper (Th) cell subtypes in the pathomechanism and regulation of the disease is a central point of current research. Th lineages derive from naïve CD4 T cell progenitors and lineage commitment is initiated by the surrounding cytokine milieu and subsequent changes in the transcription factor (TF) profile. Each T cell subtype has its own effector characteristics, immunologic function, and lineage specific cytokine profile, leading to the association with different immune responses and diseases. Acute GVHD is thought to be mainly driven by the Th1/Th17 axis, whereas Treg cells are attributed to attenuate GVHD effects. As the differentiation of each Th subset highly depends on the specific composition of activating and repressing TFs, these present a potent target to alter the Th cell landscape towards a GVHD-ameliorating direction, e.g. by inhibiting Th1 and Th17 differentiation. The finding, that targeting of Th1 and Th17 differentiation appears more effective for GVHD-prevention than a strategy to inhibit Th1 and Th17 cytokines supports this concept. In this review, we shed light on the current advances of potent TF inhibitors to alter Th cell differentiation and consecutively attenuate GVHD. We will focus especially on preclinical studies and outcomes of TF inhibition in murine GVHD models. Finally, we will point out the possible impact of a Th cell subset-specific immune modulation in context of GVHD.
Collapse
Affiliation(s)
- Julia Campe
- Experimental Immunology, Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Evelyn Ullrich
- Experimental Immunology, Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Children's University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany.,Frankfurt Cancer Institute, Goethe University Frankfurt, Frankfurt am Main, Germany.,German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung (DKTK)), Partner Site Frankfurt/Mainz, Frankfurt am Main, Germany
| |
Collapse
|
10
|
Karl F, Hudecek M, Berberich-Siebelt F, Mackensen A, Mougiakakos D. T-Cell Metabolism in Graft Versus Host Disease. Front Immunol 2021; 12:760008. [PMID: 34777373 PMCID: PMC8586445 DOI: 10.3389/fimmu.2021.760008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/11/2021] [Indexed: 01/23/2023] Open
Abstract
Allogeneic-hematopoietic stem cell transplantation (allo-HSCT) represents the only curative treatment option for numerous hematological malignancies. Elimination of malignant cells depends on the T-cells' Graft-versus-Tumor (GvT) effect. However, Graft-versus-Host-Disease (GvHD), often co-occurring with GvT, remains an obstacle for therapeutic efficacy. Hence, approaches, which selectively alleviate GvHD without compromising GvT activity, are needed. As already explored for autoimmune and inflammatory disorders, immuno-metabolic interventions pose a promising option to address this unmet challenge. Being embedded in a complex regulatory framework, immunological and metabolic pathways are closely intertwined, which is demonstrated by metabolic reprograming of T-cells upon activation or differentiation. In this review, current knowledge on the immuno-metabolic signature of GvHD-driving T-cells is summarized and approaches to metabolically interfere are outlined. Furthermore, we address the metabolic impact of standard medications for GvHD treatment and prophylaxis, which, in conjunction with the immuno-metabolic profile of alloreactive T-cells, could allow more targeted interventions in the future.
Collapse
Affiliation(s)
- Franziska Karl
- Department of Medicine 5, Hematology and Clinical Oncology, Friedrich Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | | | - Andreas Mackensen
- Department of Medicine 5, Hematology and Clinical Oncology, Friedrich Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| | - Dimitrios Mougiakakos
- Department of Medicine 5, Hematology and Clinical Oncology, Friedrich Alexander University (FAU) Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Erlangen, Germany
| |
Collapse
|
11
|
Xiao Y, Qureischi M, Dietz L, Vaeth M, Vallabhapurapu SD, Klein-Hessling S, Klein M, Liang C, König A, Serfling E, Mottok A, Bopp T, Rosenwald A, Buttmann M, Berberich I, Beilhack A, Berberich-Siebelt F. Lack of NFATc1 SUMOylation prevents autoimmunity and alloreactivity. J Exp Med 2021; 218:152124. [PMID: 32986812 PMCID: PMC7953626 DOI: 10.1084/jem.20181853] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 04/22/2020] [Accepted: 07/17/2020] [Indexed: 12/16/2022] Open
Abstract
Posttranslational modification with SUMO is known to regulate the activity of transcription factors, but how SUMOylation of individual proteins might influence immunity is largely unexplored. The NFAT transcription factors play an essential role in antigen receptor-mediated gene regulation. SUMOylation of NFATc1 represses IL-2 in vitro, but its role in T cell-mediated immune responses in vivo is unclear. To this end, we generated a novel transgenic mouse in which SUMO modification of NFATc1 is prevented. Avoidance of NFATc1 SUMOylation ameliorated experimental autoimmune encephalomyelitis as well as graft-versus-host disease. Elevated IL-2 production in T cells promoted T reg expansion and suppressed autoreactive or alloreactive immune responses. Mechanistically, increased IL-2 secretion counteracted IL-17 and IFN-γ expression through STAT5 and Blimp-1 induction. Then, Blimp-1 repressed IL-2 itself, as well as the induced, proliferation-associated survival factor Bcl2A1. Collectively, these data demonstrate that prevention of NFATc1 SUMOylation fine-tunes T cell responses toward lasting tolerance. Thus, targeting NFATc1 SUMOylation presents a novel and promising strategy to treat T cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Yin Xiao
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Musga Qureischi
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Department of Medicine II, Center for Interdisciplinary Clinical Research, University Hospital Wuerzburg, Wuerzburg, Germany.,Graduate School of Life Sciences, University of Wuerzburg, Wuerzburg, Germany
| | - Lena Dietz
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Martin Vaeth
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Stefan Klein-Hessling
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Department of Molecular Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Matthias Klein
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany
| | - Chunguang Liang
- Functional Genomics and Systems Biology Group, Department of Bioinformatics, Biocenter, University of Wuerzburg, Wuerzburg, Germany
| | - Anika König
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Edgar Serfling
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Department of Molecular Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Anja Mottok
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Tobias Bopp
- Institute for Immunology, University Medical Center, University of Mainz, Mainz, Germany.,Research Center for Immunotherapy, University Medical Center, University of Mainz, Mainz, Germany.,University Cancer Center Mainz, University Medical Center, University of Mainz, Mainz, Germany.,German Cancer Consortium, University Medical Center, University of Mainz, Mainz, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Centre Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - Mathias Buttmann
- Department of Neurology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ingolf Berberich
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Andreas Beilhack
- Department of Medicine II, Center for Interdisciplinary Clinical Research, University Hospital Wuerzburg, Wuerzburg, Germany
| | | |
Collapse
|
12
|
Kubota H, Masuda T, Noura M, Furuichi K, Matsuo H, Hirata M, Kataoka TR, Hiramatsu H, Yasumi T, Nakahata T, Imai Y, Takita J, Adachi S, Sugiyama H, Kamikubo Y. RUNX inhibitor suppresses graft-versus-host disease through targeting RUNX-NFATC2 axis. EJHAEM 2021; 2:449-458. [PMID: 35844683 PMCID: PMC9175814 DOI: 10.1002/jha2.230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 11/13/2022]
Abstract
Patients with refractory graft-versus-host disease (GVHD) have a dismal prognosis. Therefore, novel therapeutic targets are still needed to be identified. Runt-related transcriptional factor (RUNX) family transcription factors are essential transcription factors that mediate the essential roles in effector T cells. However, whether RUNX targeting can suppress, and GVHD is yet unknown. Here, we showed that RUNX family members have a redundant role in directly transactivating NFATC2 expression in T cells. We also found that our novel RUNX inhibitor, Chb-M', which is the inhibitor that switches off the entire RUNX family by alkylating agent-conjugated pyrrole-imidazole (PI) polyamides, inhibited T-cell receptor mediated T cell proliferation and allogenic T cell response. These were designed to specifically bind to consensus RUNX-binding sequences (TGTGGT). Chb-M' also suppressed the expression of NFATC2 and pro-inflammatory cytokine genes in vitro. Using xenogeneic GVHD model, mice injected by Chb-M' showed almost no sign of GVHD. Especially, the CD4 T cell was decreased and GVHD-associated cytokines including tissue necrosis factor-α and granulocyte-macrophage colony-stimulating factor were reduced in the peripheral blood of Chb-M' injected mice. Taken together, our data demonstrates that RUNX family transcriptionally upregulates NFATC2 in T cells, and RUNX-NFATC2 axis can be a novel therapeutic target against GVHD.
Collapse
Affiliation(s)
- Hirohito Kubota
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Tatsuya Masuda
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Mina Noura
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Kana Furuichi
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Hidemasa Matsuo
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Masahiro Hirata
- Department of Diagnostic PathologyKyoto University HospitalSakyo‐kuKyotoJapan
| | - Tatsuki R. Kataoka
- Department of Diagnostic PathologyKyoto University HospitalSakyo‐kuKyotoJapan
| | - Hidefumi Hiramatsu
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Takahiro Yasumi
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Tatsutoshi Nakahata
- Drug Discovery Technology Development OfficeCenter for iPS cell research and application (CiRA)Kyoto UniversitySakyo‐kuKyotoJapan
| | - Yoichi Imai
- Department of Hematology/OncologyIMSUT HospitalThe Institute of Medical ScienceThe University of TokyoTokyoJapan
| | - Junko Takita
- Department of PediatricsGraduate School of MedicineKyoto UniversitySakyo‐kuKyotoJapan
| | - Souichi Adachi
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| | - Hiroshi Sugiyama
- Department of ChemistryGraduate School of ScienceKyoto UniversitySakyo‐kuKyotoJapan
| | - Yasuhiko Kamikubo
- Department of Human Health SciencesGraduate School of MedicineKyoto, University, Sakyo‐kuKyotoJapan
| |
Collapse
|
13
|
Majumder S, Jugovic I, Saul D, Bell L, Hundhausen N, Seal R, Beilhack A, Rosenwald A, Mougiakakos D, Berberich-Siebelt F. Rapid and Efficient Gene Editing for Direct Transplantation of Naive Murine Cas9 + T Cells. Front Immunol 2021; 12:683631. [PMID: 34367143 PMCID: PMC8335400 DOI: 10.3389/fimmu.2021.683631] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/06/2021] [Indexed: 01/10/2023] Open
Abstract
Gene editing of primary T cells is a difficult task. However, it is important for research and especially for clinical T-cell transfers. CRISPR/Cas9 is the most powerful gene-editing technique. It has to be applied to cells by either retroviral transduction or electroporation of ribonucleoprotein complexes. Only the latter is possible with resting T cells. Here, we make use of Cas9 transgenic mice and demonstrate nucleofection of pre-stimulated and, importantly, of naive CD3+ T cells with guideRNA only. This proved to be rapid and efficient with no need of further selection. In the mixture of Cas9+CD3+ T cells, CD4+ and CD8+ conventional as well as regulatory T cells were targeted concurrently. IL-7 supported survival and naivety in vitro, but T cells were also transplantable immediately after nucleofection and elicited their function like unprocessed T cells. Accordingly, metabolic reprogramming reached normal levels within days. In a major mismatch model of GvHD, not only ablation of NFATc1 and/or NFATc2, but also of the NFAT-target gene IRF4 in naïve primary murine Cas9+CD3+ T cells by gRNA-only nucleofection ameliorated GvHD. However, pre-activated murine T cells could not achieve long-term protection from GvHD upon single NFATc1 or NFATc2 knockout. This emphasizes the necessity of gene-editing and transferring unstimulated human T cells during allogenic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Snigdha Majumder
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Isabelle Jugovic
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Domenica Saul
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, Erlangen, Wuerzburg, Germany
| | - Luisa Bell
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | | | - Rishav Seal
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany
| | - Andreas Beilhack
- Department of Medicine II, Center for Interdisciplinary Clinical Research (IZKF), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Centre Mainfranken, University of Wuerzburg, Wuerzburg, Germany
| | - Dimitrios Mougiakakos
- Department of Internal Medicine 5, Hematology and Oncology, Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, Erlangen, Wuerzburg, Germany.,Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University (FAU) of Erlangen-Nuremberg, Erlangen, Germany
| | | |
Collapse
|
14
|
Carter NM, Pomerantz JL. Calcineurin inhibitors target Lck activation in graft-versus-host disease. J Clin Invest 2021; 131:e149934. [PMID: 34060488 DOI: 10.1172/jci149934] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Calcineurin inhibitors (CNIs) such as cyclosporin A and FK506 are widely administered immunosuppressive drugs. Calcineurin relieves inhibitory phosphorylation from nuclear factor of activated T cells (NFAT) transcription factors downstream of T cell receptor engagement, resulting in their nuclear translocation and the production of cytokines, including IL-2, IFN-γ, and TNF-α. It was previously believed that CNIs downregulate immunity by reducing NFAT activation. However, work from Otsuka et al. in this issue of the JCI revealed a second mechanism by which CNIs suppress T cell function. The authors previously reported that calcineurin removes an inhibitory phosphate from the tyrosine kinase Lck at Ser59 (Lck-S59) and that this dephosphorylation positively regulates T cell activation. In the present work, the authors showed that inhibition of Lck-S59 dephosphorylation was essential for the CNI-mediated suppression of acute graft-versus-host disease (aGVHD). These findings have important implications for future approaches to the management of aGVHD, organ transplant rejection, and autoimmune disease.
Collapse
|
15
|
Mammadli M, Huang W, Harris R, Sultana A, Cheng Y, Tong W, Pu J, Gentile T, Dsouza S, Yang Q, Bah A, August A, Karimi M. Targeting Interleukin-2-Inducible T-Cell Kinase (ITK) Differentiates GVL and GVHD in Allo-HSCT. Front Immunol 2020; 11:593863. [PMID: 33324410 PMCID: PMC7726260 DOI: 10.3389/fimmu.2020.593863] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/29/2020] [Indexed: 01/04/2023] Open
Abstract
Allogeneic hematopoietic stem cell transplantation is a potentially curative procedure for many malignant diseases. Donor T cells prevent disease recurrence via graft-versus-leukemia (GVL) effect. Donor T cells also contribute to graft-versus-host disease (GVHD), a debilitating and potentially fatal complication. Novel treatment strategies are needed which allow preservation of GVL effects without causing GVHD. Using murine models, we show that targeting IL-2-inducible T cell kinase (ITK) in donor T cells reduces GVHD while preserving GVL effects. Both CD8+ and CD4+ donor T cells from Itk-/- mice produce less inflammatory cytokines and show decrease migration to GVHD target organs such as the liver and small intestine, while maintaining GVL efficacy against primary B-cell acute lymphoblastic leukemia (B-ALL). Itk-/- T cells exhibit reduced expression of IRF4 and decreased JAK/STAT signaling activity but upregulating expression of Eomesodermin (Eomes) and preserve cytotoxicity, necessary for GVL effect. Transcriptome analysis indicates that ITK signaling controls chemokine receptor expression during alloactivation, which in turn affects the ability of donor T cells to migrate to GVHD target organs. Our data suggest that inhibiting ITK could be a therapeutic strategy to reduce GVHD while preserving the beneficial GVL effects following allo-HSCT treatment.
Collapse
Affiliation(s)
- Mahinbanu Mammadli
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Weishan Huang
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States.,Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Rebecca Harris
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Aisha Sultana
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Ying Cheng
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Wei Tong
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jeffery Pu
- Department of Hematology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Teresa Gentile
- Department of Hematology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Shanti Dsouza
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Qi Yang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, United States
| | - Alaji Bah
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, United States
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Mobin Karimi
- Department of Microbiology and Immunology, SUNY Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
16
|
Bendickova K, Fric J. Roles of IL-2 in bridging adaptive and innate immunity, and as a tool for cellular immunotherapy. J Leukoc Biol 2020; 108:427-437. [PMID: 32480431 PMCID: PMC7384134 DOI: 10.1002/jlb.5mir0420-055r] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/01/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
IL-2 was initially characterized as a T cell growth factor in the 1970s, and has been studied intensively ever since. Decades of research have revealed multiple and diverse roles for this potent cytokine, indicating a unique linking role between adaptive and innate arms of the immune system. Here, we review the literature showing that IL-2 is expressed in a plethora of cell types across the immune system, where it has indispensable functions in orchestrating cellular interactions and shaping the nature and magnitude of immune responses. Emerging from the basic research that has revealed the molecular mechanisms and the complexity of the biologic actions of IL-2, several immunotherapeutic approaches have now focused on manipulating the levels of this cytokine in patients. These strategies range from inhibition of IL-2 to achieve immunosuppression, to the application of IL-2 as a vaccine adjuvant and in cancer therapies. This review will systematically summarize the major findings in the field and identify key areas requiring further research in order to realize the potential of IL-2 in the treatment of human diseases.
Collapse
Affiliation(s)
- Kamila Bendickova
- International Clinical Research CenterSt. Anne's University Hospital BrnoBrnoCzech Republic
| | - Jan Fric
- International Clinical Research CenterSt. Anne's University Hospital BrnoBrnoCzech Republic
- Institute of Hematology and Blood TransfusionPragueCzech Republic
| |
Collapse
|
17
|
Glycogen Synthase Kinase-3β Facilitates Cytokine Production in 12-O-Tetradecanoylphorbol-13-Acetate/Ionomycin-Activated Human CD4 + T Lymphocytes. Cells 2020; 9:cells9061424. [PMID: 32521784 PMCID: PMC7348852 DOI: 10.3390/cells9061424] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/02/2022] Open
Abstract
Cytokines are the major immune regulators secreted from activated CD4+ T lymphocytes that activate adaptive immunity to eradicate nonself cells, including pathogens, tumors, and allografts. The regulation of glycogen synthase kinase (GSK)-3β, a serine/threonine kinase, controls cytokine production by regulating transcription factors. The artificial in vitro activation of CD4+ T lymphocytes by a combination of 12-O-tetradecanoylphorbol-13-acetate and ionomycin, the so-called T/I model, led to an inducible production of cytokines, such as interferon-γ, tumor necrosis factor-α, and interleukin-2. As demonstrated by the approaches of pharmacological targeting and genetic knockdown of GSK-3β, T/I treatment effectively caused GSK-3β activation followed by GSK-3β-regulated cytokine production. In contrast, pharmacological inhibition of the proline-rich tyrosine kinase 2 and calcineurin signaling pathways blocked cytokine production, probably by deactivating GSK-3β. The blockade of GSK-3β led to the inhibition of the nuclear translocation of T-bet, a vital transcription factor of T lymphocyte cytokines. In a mouse model, treatment with the GSK-3β inhibitor 6-bromoindirubin-3’-oxime significantly inhibited T/I-induced mortality and serum cytokine levels. In summary, targeting GSK-3β effectively inhibits CD4+ T lymphocyte activation and cytokine production.
Collapse
|
18
|
Walton K, Fernandez MR, Sagatys EM, Reff J, Kim J, Lee MC, Kiluk JV, Hui JYC, McKenna D, Hupp M, Forster C, Linden MA, Lawrence NJ, Lawrence HR, Pidala J, Pavletic SZ, Blazar BR, Sebti SM, Cleveland JL, Anasetti C, Betts BC. Metabolic reprogramming augments potency of human pSTAT3-inhibited iTregs to suppress alloreactivity. JCI Insight 2020; 5:136437. [PMID: 32255769 DOI: 10.1172/jci.insight.136437] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 04/01/2020] [Indexed: 12/18/2022] Open
Abstract
Immunosuppressive donor Tregs can prevent graft-versus-host disease (GVHD) or solid-organ allograft rejection. We previously demonstrated that inhibiting STAT3 phosphorylation (pSTAT3) augments FOXP3 expression, stabilizing induced Tregs (iTregs). Here we report that human pSTAT3-inhibited iTregs prevent human skin graft rejection and xenogeneic GVHD yet spare donor antileukemia immunity. pSTAT3-inhibited iTregs express increased levels of skin-homing cutaneous lymphocyte-associated antigen, immunosuppressive GARP and PD-1, and IL-9 that supports tolerizing mast cells. Further, pSTAT3-inhibited iTregs significantly reduced alloreactive conventional T cells, Th1, and Th17 cells implicated in GVHD and tissue rejection and impaired infiltration by pathogenic Th2 cells. Mechanistically, pSTAT3 inhibition of iTregs provoked a shift in metabolism from oxidative phosphorylation (OxPhos) to glycolysis and reduced electron transport chain activity. Strikingly, cotreatment with coenzyme Q10 restored OxPhos in pSTAT3-inhibited iTregs and augmented their suppressive potency. These findings support the rationale for clinically testing the safety and efficacy of metabolically tuned, human pSTAT3-inhibited iTregs to control alloreactive T cells.
Collapse
Affiliation(s)
- Kelly Walton
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, and
| | | | - John V Kiluk
- Department of Breast Oncology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - David McKenna
- Department of Laboratory Medicine and Pathology, and
| | - Meghan Hupp
- Department of Laboratory Medicine and Pathology, and
| | - Colleen Forster
- Bionet Histology Research Laboratory, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | - Joseph Pidala
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Steven Z Pavletic
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Said M Sebti
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia USA
| | | | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, Tampa, Florida, USA
| | - Brian C Betts
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
19
|
Kai W, Lin C, Jin Y, Ping-Lin H, Xun L, Bastian A, Arnulf S, Sha-Sha X, Xu L, Shu C. Urethral meatus stricture BOO stimulates bladder smooth muscle cell proliferation and pyroptosis via IL‑1β and the SGK1‑NFAT2 signaling pathway. Mol Med Rep 2020; 22:219-226. [PMID: 32468047 PMCID: PMC7248470 DOI: 10.3892/mmr.2020.11092] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 03/25/2020] [Indexed: 11/29/2022] Open
Abstract
Bladder outlet obstruction (BOO), which is primarily caused by benign prostatic hyperplasia, is a common chronic disease. However, previous studies have most commonly investigated BOO using the acute obstruction model. In the present study, a chronic obstruction model was established to investigate the different pathological alterations in the bladder between acute and chronic obstruction. Compared with chronic obstruction, acute obstruction led to increased expression of proliferating cell nuclear antigen and interleukin-1β, which are markers of proliferation and inflammation, respectively. Furthermore, increased fibrosis in the bladder at week 2 was observed. Low pressure promoted mice bladder smooth muscle cell (MBSMC) proliferation, and pressure overload inhibited cell proliferation and increased the proportion of dead MBSMCs. Further investigation using serum/glucocorticoid regulated kinase 1 (SGK1) small interfering RNAs indicated that low pressure may promote MBSMC proliferation by upregulating SGK1 and nuclear factor of activated T-cell expression levels. Therefore, the present study suggested that acute obstruction led to faster decompensation of bladder function and chronic bladder obstruction displayed an enhanced ability to progress to BOO.
Collapse
Affiliation(s)
- Wang Kai
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Chen Lin
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Yang Jin
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - He Ping-Lin
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Liu Xun
- Department of Urology, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610041, P.R. China
| | - Amend Bastian
- Department of Urology, University of Tübingen, D‑72070 Tübingen, Baden‑Württemberg, Germany
| | - Stenzl Arnulf
- Department of Urology, University of Tübingen, D‑72070 Tübingen, Baden‑Württemberg, Germany
| | - Xing Sha-Sha
- Central Laboratory, Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Luo Xu
- Department of Urology, Zunyi Medical University, Guiyang, Guizhou 563000, P.R. China
| | - Cui Shu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
20
|
Fu Y, Liao C, Cui K, Liu X, Fang W. Antitumor pharmacotherapy of colorectal cancer in kidney transplant recipients. Ther Adv Med Oncol 2019; 11:1758835919876196. [PMID: 31579127 PMCID: PMC6759705 DOI: 10.1177/1758835919876196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 08/19/2019] [Indexed: 11/15/2022] Open
Abstract
Renal transplantation has become the sole most preferred therapy modality for end-stage renal disease patients. The growing tendency for renal transplants, and prolonged survival of renal recipients, have resulted in a certain number of post-transplant colorectal cancer patients. Antitumor pharmacotherapy in these patients is a dilemma. Substantial impediments such as carcinogenesis of immunosuppressive drugs (ISDs), drug interaction between ISDs and anticancer drugs, and toxicity of anticancer drugs exist. However, experience of antitumor pharmacotherapy in these patients is limited, and the potential risks and benefits have not been reviewed systematically. This review evaluates the potential impediments, summarizes current experience, and provides potential antitumor strategies, including adjuvant, palliative, and subsequent regimens. Moreover, special pharmaceutical care, such as ISDs therapeutic drug monitoring, metabolic enzymes genotype, and drug interaction, are also highlighted.
Collapse
Affiliation(s)
- Yuanyuan Fu
- Department of Pharmacy, First Affiliated Hospital of Nanjing Medical University, China
| | - Chengheng Liao
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Kai Cui
- Department of Pharmacy, Liaocheng Infectious Disease Hospital, Liaocheng, Shandong, China
| | - Xiao Liu
- Department of Pharmacy, Qinghai provincial Peoples Hospital, Xining, China
| | - Wentong Fang
- Department of Pharmacy, First Affiliated Hospital of Nanjing Medical University, No 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| |
Collapse
|
21
|
Kawalkowska JZ, Ogbechi J, Venables PJ, Williams RO. cIAP1/2 inhibition synergizes with TNF inhibition in autoimmunity by down-regulating IL-17A and inducing T regs. SCIENCE ADVANCES 2019; 5:eaaw5422. [PMID: 31049403 PMCID: PMC6494502 DOI: 10.1126/sciadv.aaw5422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/19/2019] [Indexed: 06/09/2023]
Abstract
IL-17 and TNF-α are major effector cytokines in chronic inflammation. TNF-α inhibitors have revolutionized the treatment of rheumatoid arthritis (RA), although not all patients respond, and most relapse after treatment withdrawal. This may be due to a paradoxical exacerbation of TH17 responses by TNF-α inhibition. We examined the therapeutic potential of targeting cellular inhibitors of apoptosis 1 and 2 (cIAP1/2) in inflammation by its influence on human TH subsets and mice with collagen-induced arthritis. Inhibition of cIAP1/2 abrogated CD4+ IL-17A differentiation and IL-17 production. This was a direct effect on T cells, mediated by reducing NFATc1 expression. In mice, cIAP1/2 inhibition, when combined with etanercept, abrogated disease activity, which was associated with an increase in Tregs and was sustained after therapy retraction. We reveal an unexpected role for cIAP1/2 in regulating the balance between TH17 and Tregs and suggest that combined therapeutic inhibition could induce long-term remission in inflammatory diseases.
Collapse
|
22
|
Bendíčková K, Tidu F, De Zuani M, Kohoutková MH, Andrejčinová I, Pompeiano A, Bělášková S, Forte G, Zelante T, Frič J. Calcineurin inhibitors reduce NFAT-dependent expression of antifungal pentraxin-3 by human monocytes. J Leukoc Biol 2019; 107:497-508. [PMID: 30934147 PMCID: PMC7064969 DOI: 10.1002/jlb.4vma0318-138r] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 02/20/2019] [Accepted: 03/22/2019] [Indexed: 01/14/2023] Open
Abstract
Calcineurin (CN) inhibitors are effective clinical immunosuppressants but leave patients vulnerable to potentially fatal fungal infections. This study tested the hypothesis that CN inhibition interferes with antifungal immune defenses mediated by monocytes. We showed that NFAT is expressed by human monocytes, and is activated by exposure to fungal ligands. We confirmed that NFAT translocation potently activated target gene transcription using a human monocytic reporter cell line. Inhibition of CN‐NFAT by cyclosporine A significantly reduced monocyte production of TNF‐α, IL‐10, and MCP‐1 proteins in response to pattern recognition receptor ligands as well as to Aspergillus fumigatus conidia. Moreover, we revealed that human monocytes express the antifungal protein pentraxin‐3 under control of NFAT. In conclusion, clinical CN inhibitors have the potential to interfere with the novel NFAT‐dependent pentraxin‐3 pathway as well as antifungal cytokine production in human monocytes, thereby impeding monocyte‐mediated defenses against fungal infection in immune‐suppressed patients.
Collapse
Affiliation(s)
- Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Federico Tidu
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marco De Zuani
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | | | - Ivana Andrejčinová
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Antonio Pompeiano
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Silvie Bělášková
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Giancarlo Forte
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - Teresa Zelante
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
23
|
Flores C, Fouquet G, Moura IC, Maciel TT, Hermine O. Lessons to Learn From Low-Dose Cyclosporin-A: A New Approach for Unexpected Clinical Applications. Front Immunol 2019; 10:588. [PMID: 30984176 PMCID: PMC6447662 DOI: 10.3389/fimmu.2019.00588] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/05/2019] [Indexed: 01/09/2023] Open
Abstract
Cyclosporin-A has been known and used for a long time, since its "fast track" approval in the early 80's. This molecule has rapidly demonstrated unexpected immunosuppressive properties, transforming the history of organ transplantation. Cyclosporin's key effect relies on modulation on T-lymphocyte activity, which explains its role in the prevention of graft rejection. However, whether cyclosporin-A exerts other effects on immune system remains to be determined. Until recently, cyclosporin-A was mainly used at a high-dose, but given the drug toxicity and despite the fear of losing its immunosuppressive effects, there is nowadays a tendency to decrease its dose. The literature has been reporting data revealing a paradoxical effect of low dosage of cyclosporin-A. These low-doses appear to have immunomodulatory properties, with different effects from high-doses on CD8+ T lymphocyte activation, auto-immune diseases, graft-vs.-host disease and cancer. The aim of this review is to discuss the role of cyclosporin-A, not only as a consecrated immunosuppressive agent, but also as an immunomodulatory drug when administrated at low-dose. The use of low-dose cyclosporin-A may become a new therapeutic strategy, particularly to treat cancer.
Collapse
Affiliation(s)
- Camila Flores
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Guillemette Fouquet
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Ivan Cruz Moura
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Thiago Trovati Maciel
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
| | - Olivier Hermine
- INSERM UMR1163 and CNRS URL 8254, Imagine Institute, Paris, France
- Paris Descartes University-Sorbonne Paris Cité, Paris, France
- Department of Hematology, Necker Children's Hospital, APHP, Paris, France
| |
Collapse
|
24
|
Vaeth M, Wang YH, Eckstein M, Yang J, Silverman GJ, Lacruz RS, Kannan K, Feske S. Tissue resident and follicular Treg cell differentiation is regulated by CRAC channels. Nat Commun 2019; 10:1183. [PMID: 30862784 PMCID: PMC6414608 DOI: 10.1038/s41467-019-08959-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 02/11/2019] [Indexed: 12/30/2022] Open
Abstract
T regulatory (Treg) cells maintain immunological tolerance and organ homeostasis. Activated Treg cells differentiate into effector Treg subsets that acquire tissue-specific functions. Ca2+ influx via Ca2+ release-activated Ca2+ (CRAC) channels formed by STIM and ORAI proteins is required for the thymic development of Treg cells, but its function in mature Treg cells remains unclear. Here we show that deletion of Stim1 and Stim2 genes in mature Treg cells abolishes Ca2+ signaling and prevents their differentiation into follicular Treg and tissue-resident Treg cells. Transcriptional profiling of STIM1/STIM2-deficient Treg cells reveals that Ca2+ signaling regulates transcription factors and signaling pathways that control the identity and effector differentiation of Treg cells. In the absence of STIM1/STIM2 in Treg cells, mice develop a broad spectrum of autoantibodies and fatal multiorgan inflammation. Our findings establish a critical role of CRAC channels in controlling lineage identity and effector functions of Treg cells. Regulatory T (Treg) cells are important for maintaining immune homeostasis. Here the authors show that STIM1 and STIM2, which activate the Ca2+ channel ORAI1, are essential for the differentiation of peripheral Treg cells into tissue-resident and follicular Treg cells and their ability to limit autoimmunity in mice.
Collapse
Affiliation(s)
- Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA.,Institute for Systems Immunology, Julius-Maximilians University of Würzburg, 97078, Würzburg, Germany
| | - Yin-Hu Wang
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Miriam Eckstein
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA.,Institute for Systems Immunology, Julius-Maximilians University of Würzburg, 97078, Würzburg, Germany
| | - Jun Yang
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Gregg J Silverman
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Rodrigo S Lacruz
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Kasthuri Kannan
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA.,Genome Technology Center, New York University School of Medicine, New York, NY, 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
25
|
Chang YJ, Zhao XY, Huang XJ. Strategies for Enhancing and Preserving Anti-leukemia Effects Without Aggravating Graft-Versus-Host Disease. Front Immunol 2018; 9:3041. [PMID: 30619371 PMCID: PMC6308132 DOI: 10.3389/fimmu.2018.03041] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/10/2018] [Indexed: 12/29/2022] Open
Abstract
Allogeneic stem cell transplantation (allo-SCT) is a curable method for the treatment of hematological malignancies. In the past two decades, the establishment of haploidentical transplant modalities make “everyone has a donor” become a reality. However, graft-versus-host disease (GVHD) and relapse remain the major two causes of death either in the human leukocyte antigen (HLA)-matched transplant or haploidentical transplant settings, both of which restrict the improvement of transplant outcomes. Preclinical mice model showed that both donor-derived T cells and natural killer (NK) cells play important role in the pathogenesis of GVHD and the effects of graft-versus-leukemia (GVL). Hence, understanding the immune mechanisms of GVHD and GVL would provide potential strategies for the control of leukemia relapse without aggravating GVHD. The purpose of the current review is to summarize the biology of GVHD and GVL responses in preclinical models and to discuss potential novel therapeutic strategies to reduce the relapse rate after allo-SCT. We will also review the approaches, including optimal donor selection and, conditioning regimens, donor lymphocyte infusion, BCR/ABL-specific CTL, and chimeric antigen receptor-modified T cells, which have been successfully used in the clinic to enhance and preserve anti-leukemia activity, especially GVL effects, without aggravating GVHD or alleviate GVHD.
Collapse
Affiliation(s)
- Ying-Jun Chang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital & Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Beijing, China
| |
Collapse
|
26
|
Betts BC, Locke FL, Sagatys EM, Pidala J, Walton K, Menges M, Reff J, Saha A, Djeu JY, Kiluk JV, Lee MC, Kim J, Kang CW, Tang CH, Frieling J, Lynch CC, List A, Rodriguez PC, Blazar BR, Conejo-Garcia JR, Del Valle JR, Hu CC, Anasetti C. Inhibition of Human Dendritic Cell ER Stress Response Reduces T Cell Alloreactivity Yet Spares Donor Anti-tumor Immunity. Front Immunol 2018; 9:2887. [PMID: 30574153 PMCID: PMC6291501 DOI: 10.3389/fimmu.2018.02887] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 11/26/2018] [Indexed: 11/13/2022] Open
Abstract
Acute graft- vs. -host disease (GVHD) is an important cause of morbidity and death after allogeneic hematopoietic cell transplantation (HCT). We identify a new approach to prevent GVHD that impairs monocyte-derived dendritic cell (moDC) alloactivation of T cells, yet preserves graft- vs.-leukemia (GVL). Exceeding endoplasmic reticulum (ER) capacity results in a spliced form of X-box binding protein-1 (XBP-1s). XBP-1s mediates ER stress and inflammatory responses. We demonstrate that siRNA targeting XBP-1 in moDCs abrogates their stimulation of allogeneic T cells. B-I09, an inositol-requiring enzyme-1α (IRE1α) inhibitor that prevents XBP-1 splicing, reduces human moDC migration, allo-stimulatory potency, and curtails moDC IL-1β, TGFβ, and p40 cytokines, suppressing Th1 and Th17 cell priming. B-I09-treated moDCs reduce responder T cell activation via calcium flux without interfering with regulatory T cell (Treg) function or GVL effects by cytotoxic T lymphocytes (CTL) and NK cells. In a human T cell mediated xenogeneic GVHD model, B-I09 inhibition of XBP-1s reduced target-organ damage and pathogenic Th1 and Th17 cells without impacting donor Tregs or anti-tumor CTL. DC XBP-1s inhibition provides an innovative strategy to prevent GVHD and retain GVL.
Collapse
Affiliation(s)
- Brian C Betts
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States.,Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Elizabeth M Sagatys
- Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL, United States
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Kelly Walton
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States.,Division of Hematology, Oncology, and Transplantation, University of Minnesota, Minneapolis, MN, United States
| | - Meghan Menges
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Jordan Reff
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Asim Saha
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,The Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Julie Y Djeu
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - John V Kiluk
- Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Marie C Lee
- Comprehensive Breast Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, FL, United States
| | - Chang Won Kang
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Chih-Hang Tang
- Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, PA, United States
| | - Jeremy Frieling
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, United States
| | - Conor C Lynch
- Department of Tumor Biology, Moffitt Cancer Center, Tampa, FL, United States
| | - Alan List
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL, United States
| | - Paulo C Rodriguez
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| | - Bruce R Blazar
- Division of Blood and Marrow Transplantation, Department of Pediatrics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN, United States.,The Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
| | | | - Juan R Del Valle
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Chih-Chi Hu
- Department of Translational Tumor Immunology, The Wistar Institute, Philadelphia, PA, United States
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Tampa, FL, United States.,Department of Immunology, Moffitt Cancer Center, Tampa, FL, United States
| |
Collapse
|
27
|
Lee JU, Kim LK, Choi JM. Revisiting the Concept of Targeting NFAT to Control T Cell Immunity and Autoimmune Diseases. Front Immunol 2018; 9:2747. [PMID: 30538703 PMCID: PMC6277705 DOI: 10.3389/fimmu.2018.02747] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 11/08/2018] [Indexed: 01/15/2023] Open
Abstract
The nuclear factor of activated T cells (NFAT) family of transcription factors, which includes NFAT1, NFAT2, and NFAT4, are well-known to play important roles in T cell activation. Most of NFAT proteins are controlled by calcium influx upon T cell receptor and costimulatory signaling results increase of IL-2 and IL-2 receptor. NFAT3 however is not shown to be expressed in T cells and NFAT5 has not much highlighted in T cell functions yet. Recent studies demonstrate that the NFAT family proteins involve in function of lineage-specific transcription factors during differentiation of T helper 1 (Th1), Th2, Th17, regulatory T (Treg), and follicular helper T cells (Tfh). They have been studied to make physical interaction with the other transcription factors like GATA3 or Foxp3 and they also regulate Th cell signature gene expressions by direct binding on promotor region of target genes. From last decades, NFAT functions in T cells have been targeted to develop immune modulatory drugs for controlling T cell immunity in autoimmune diseases like cyclosporine A, FK506, etc. Due to their undesirable side defects, only limited application is available in human diseases. This review focuses on the recent advances in development of NFAT targeting drug as well as our understanding of each NFAT family protein in T cell biology. We also discuss updated detail molecular mechanism of NFAT functions in T cells, which would lead us to suggest an idea for developing specific NFAT inhibitors as a therapeutic drug for autoimmune diseases.
Collapse
Affiliation(s)
- Jae-Ung Lee
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Li-Kyung Kim
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| | - Je-Min Choi
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, South Korea.,Research Institute for Natural Sciences, Hanyang University, Seoul, South Korea
| |
Collapse
|
28
|
Kagoya Y, Nakatsugawa M, Saso K, Guo T, Anczurowski M, Wang CH, Butler MO, Arrowsmith CH, Hirano N. DOT1L inhibition attenuates graft-versus-host disease by allogeneic T cells in adoptive immunotherapy models. Nat Commun 2018; 9:1915. [PMID: 29765028 PMCID: PMC5954061 DOI: 10.1038/s41467-018-04262-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 04/11/2018] [Indexed: 12/24/2022] Open
Abstract
Adoptive T-cell therapy is a promising therapeutic approach for cancer patients. The use of allogeneic T-cell grafts will improve its applicability and versatility provided that inherent allogeneic responses are controlled. T-cell activation is finely regulated by multiple signaling molecules that are transcriptionally controlled by epigenetic mechanisms. Here we report that inhibiting DOT1L, a histone H3-lysine 79 methyltransferase, alleviates allogeneic T-cell responses. DOT1L inhibition reduces miR-181a expression, which in turn increases the ERK phosphatase DUSP6 expression and selectively ameliorates low-avidity T-cell responses through globally suppressing T-cell activation-induced gene expression alterations. The inhibition of DOT1L or DUSP6 overexpression in T cells attenuates the development of graft-versus-host disease, while retaining potent antitumor activity in xenogeneic and allogeneic adoptive immunotherapy models. These results suggest that DOT1L inhibition may enable the safe and effective use of allogeneic antitumor T cells by suppressing unwanted immunological reactions in adoptive immunotherapy.
Collapse
Affiliation(s)
- Yuki Kagoya
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Munehide Nakatsugawa
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Kayoko Saso
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Tingxi Guo
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Mark Anczurowski
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Chung-Hsi Wang
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Marcus O Butler
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada.,Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Medicine, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium and Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G 1L7, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada
| | - Naoto Hirano
- Tumor Immunotherapy Program, Campbell Family Institute for Breast Cancer Research, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G 2M9, Canada. .,Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
29
|
Marangoni F, Zhang R, Mani V, Thelen M, Ali Akbar NJ, Warner RD, Äijö T, Zappulli V, Martinez GJ, Turka LA, Mempel TR. Tumor Tolerance-Promoting Function of Regulatory T Cells Is Optimized by CD28, but Strictly Dependent on Calcineurin. THE JOURNAL OF IMMUNOLOGY 2018; 200:3647-3661. [PMID: 29661826 DOI: 10.4049/jimmunol.1701220] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 03/13/2018] [Indexed: 01/07/2023]
Abstract
Regulatory T cells (Treg) restrain immune responses against malignant tumors, but their global depletion in cancer patients will likely be limited by systemic autoimmune toxicity. Instead, approaches to "tune" their activities may allow for preferential targeting of tumor-reactive Treg. Although Ag recognition regulates Treg function, the roles of individual TCR-dependent signaling pathways in enabling Treg to promote tumor tolerance are not well characterized. In this study, we examined in mouse tumor models the role of calcineurin, a key mediator of TCR signaling, and the role of the costimulatory receptor CD28 in the differentiation of resting central Treg into effector Treg endowed with tumor tropism. We find that calcineurin, although largely dispensable for suppressive activity in vitro, is essential for upregulation of ICOS and CTLA-4 in Treg, as well as for expression of chemokine receptors driving their accumulation in tumors. In contrast, CD28 is not critical, but optimizes the formation of tumor-homing Treg and their fitness in tumor tissue. Accordingly, although deletion of either CnB or CD28 strongly impairs Treg-mediated tumor tolerance, lack of CnB has an even more pronounced impact than lack of CD28. Hence, our studies reveal distinct roles for what has classically been defined as signal 1 and signal 2 of conventional T cell activation in the context of Treg-mediated tumor tolerance.
Collapse
Affiliation(s)
- Francesco Marangoni
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114; .,Harvard Medical School, Boston, MA 02115
| | - Ruan Zhang
- Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02114
| | - Vinidhra Mani
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114.,Harvard Medical School, Boston, MA 02115
| | - Martin Thelen
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Noor J Ali Akbar
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Ross D Warner
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114
| | - Tarmo Äijö
- Center for Computational Biology, Flatiron Institute, New York, NY 10010
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padua, 35020 Legnaro, Padua, Italy; and
| | - Gustavo J Martinez
- Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064
| | - Laurence A Turka
- Harvard Medical School, Boston, MA 02115.,Center for Transplantation Sciences, Massachusetts General Hospital, Boston, MA 02114
| | - Thorsten R Mempel
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA 02114; .,Harvard Medical School, Boston, MA 02115
| |
Collapse
|
30
|
Abstract
Nuclear factor of activated T cells (NFAT) was first described almost three decades ago as a Ca
2+/calcineurin-regulated transcription factor in T cells. Since then, a large body of research uncovered the regulation and physiological function of different NFAT homologues in the immune system and many other tissues. In this review, we will discuss novel roles of NFAT in T cells, focusing mainly on its function in humoral immune responses, immunological tolerance, and the regulation of immune metabolism.
Collapse
Affiliation(s)
- Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
31
|
Mencarelli A, Vacca M, Khameneh HJ, Acerbi E, Tay A, Zolezzi F, Poidinger M, Mortellaro A. Calcineurin B in CD4 + T Cells Prevents Autoimmune Colitis by Negatively Regulating the JAK/STAT Pathway. Front Immunol 2018. [PMID: 29515579 PMCID: PMC5826051 DOI: 10.3389/fimmu.2018.00261] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Calcineurin (Cn) is a protein phosphatase that regulates the activation of the nuclear factor of activated T-cells (NFAT) family of transcription factors, which are key regulators of T-cell development and function. Here, we generated a conditional Cnb1 mouse model in which Cnb1 was specifically deleted in CD4+ T cells (Cnb1CD4 mice) to delineate the role of the Cn–NFAT pathway in immune homeostasis of the intestine. The Cnb1CD4 mice developed severe, spontaneous colitis characterized at the molecular level by an increased T helper-1-cell response but an unaltered regulatory T-cell compartment. Antibiotic treatment ameliorated the intestinal inflammation observed in Cnb1CD4 mice, suggesting that the microbiota contributes to the onset of colitis. CD4+ T cells isolated from Cnb1CD4 mice produced high levels of IFNγ due to increased activation of the JAK2/STAT4 pathway induced by IL-12. Our data highlight that Cn signaling in CD4+ T cells is critical for intestinal immune homeostasis in part by inhibiting IL-12 responsiveness of CD4+ T cells.
Collapse
Affiliation(s)
- Andrea Mencarelli
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Maurizio Vacca
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hanif Javanmard Khameneh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Enzo Acerbi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Alicia Tay
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Francesca Zolezzi
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore
| | - Alessandra Mortellaro
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR), Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
32
|
Betts BC, Veerapathran A, Pidala J, Yang H, Horna P, Walton K, Cubitt CL, Gunawan S, Lawrence HR, Lawrence NJ, Sebti SM, Anasetti C. Targeting Aurora kinase A and JAK2 prevents GVHD while maintaining Treg and antitumor CTL function. Sci Transl Med 2018; 9:9/372/eaai8269. [PMID: 28077684 DOI: 10.1126/scitranslmed.aai8269] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 08/17/2016] [Accepted: 10/10/2016] [Indexed: 12/12/2022]
Abstract
Graft-versus-host disease (GVHD) is a leading cause of nonrelapse mortality after allogeneic hematopoietic cell transplantation. T cell costimulation by CD28 contributes to GVHD, but prevention is incomplete when targeting CD28, downstream mammalian target of rapamycin (mTOR), or Aurora A. Likewise, interleukin-6 (IL-6)-mediated Janus kinase 2 (JAK2) signaling promotes alloreactivity, yet JAK2 inhibition does not eliminate GVHD. We provide evidence that blocking Aurora A and JAK2 in human T cells is synergistic in vitro, prevents xenogeneic GVHD, and maintains antitumor responses by cytotoxic T lymphocytes (CTLs). Aurora A/JAK2 inhibition is immunosuppressive but permits the differentiation of inducible regulatory T cells (iTregs) that are hyperfunctional and CD39 bright and efficiently scavenge adenosine triphosphate (ATP). Increased iTreg potency is primarily a function of Aurora A blockade, whereas JAK2 inhibition suppresses T helper 17 (TH17) differentiation. Inhibiting either Aurora A or JAK2 significantly suppresses TH1 T cells. However, CTL generated in vivo retains tumor-specific killing despite Aurora A/JAK2 blockade. Thus, inhibiting CD28 and IL-6 signal transduction pathways in donor T cells can increase the Treg/Tconv ratio, prevent GVHD, and preserve antitumor CTL.
Collapse
Affiliation(s)
- Brian C Betts
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA. .,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Anandharaman Veerapathran
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Joseph Pidala
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Hua Yang
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Pedro Horna
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Hematopathology and Laboratory Medicine, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Kelly Walton
- Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA
| | | | - Steven Gunawan
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| | - Harshani R Lawrence
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Nicholas J Lawrence
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Said M Sebti
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA.,Department of Drug Discovery, Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Claudio Anasetti
- Department of Blood and Marrow Transplantation, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Immunology, Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncologic Sciences, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
33
|
Vaeth M, Yang J, Yamashita M, Zee I, Eckstein M, Knosp C, Kaufmann U, Karoly Jani P, Lacruz RS, Flockerzi V, Kacskovics I, Prakriya M, Feske S. ORAI2 modulates store-operated calcium entry and T cell-mediated immunity. Nat Commun 2017; 8:14714. [PMID: 28294127 PMCID: PMC5355949 DOI: 10.1038/ncomms14714] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 01/25/2017] [Indexed: 12/11/2022] Open
Abstract
Store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels is critical for lymphocyte function and immune responses. CRAC channels are hexamers of ORAI proteins that form the channel pore, but the contributions of individual ORAI homologues to CRAC channel function are not well understood. Here we show that deletion of Orai1 reduces, whereas deletion of Orai2 increases, SOCE in mouse T cells. These distinct effects are due to the ability of ORAI2 to form heteromeric channels with ORAI1 and to attenuate CRAC channel function. The combined deletion of Orai1 and Orai2 abolishes SOCE and strongly impairs T cell function. In vivo, Orai1/Orai2 double-deficient mice have impaired T cell-dependent antiviral immune responses, and are protected from T cell-mediated autoimmunity and alloimmunity in models of colitis and graft-versus-host disease. Our study demonstrates that ORAI1 and ORAI2 form heteromeric CRAC channels, in which ORAI2 fine-tunes the magnitude of SOCE to modulate immune responses.
Collapse
Affiliation(s)
- Martin Vaeth
- Experimental Pathology Program, Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 316, New York, New York 10016, USA
| | - Jun Yang
- Experimental Pathology Program, Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 316, New York, New York 10016, USA
| | - Megumi Yamashita
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Isabelle Zee
- Experimental Pathology Program, Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 316, New York, New York 10016, USA
| | - Miriam Eckstein
- NYU College of Dentistry, New York University, New York, New York 10010, USA
| | - Camille Knosp
- Experimental Pathology Program, Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 316, New York, New York 10016, USA
| | - Ulrike Kaufmann
- Experimental Pathology Program, Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 316, New York, New York 10016, USA
| | | | - Rodrigo S. Lacruz
- NYU College of Dentistry, New York University, New York, New York 10010, USA
| | - Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, School of Medicine, Saarland University, Homburg 66421, Germany
| | | | - Murali Prakriya
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | - Stefan Feske
- Experimental Pathology Program, Department of Pathology, New York University School of Medicine, 550 First Avenue, Smilow 316, New York, New York 10016, USA
| |
Collapse
|
34
|
Roseotoxin B Improves Allergic Contact Dermatitis through a Unique Anti-Inflammatory Mechanism Involving Excessive Activation of Autophagy in Activated T Lymphocytes. J Invest Dermatol 2016; 136:1636-1646. [DOI: 10.1016/j.jid.2016.04.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 04/14/2016] [Accepted: 04/18/2016] [Indexed: 12/31/2022]
|
35
|
Panda S, Srivastava S, Li Z, Vaeth M, Fuhs SR, Hunter T, Skolnik EY. Identification of PGAM5 as a Mammalian Protein Histidine Phosphatase that Plays a Central Role to Negatively Regulate CD4(+) T Cells. Mol Cell 2016; 63:457-69. [PMID: 27453048 DOI: 10.1016/j.molcel.2016.06.021] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 05/18/2016] [Accepted: 06/14/2016] [Indexed: 12/18/2022]
Abstract
Whereas phosphorylation of serine, threonine, and tyrosine is exceedingly well characterized, the role of histidine phosphorylation in mammalian signaling is largely unexplored. Here we show that phosphoglycerate mutase family 5 (PGAM5) functions as a phosphohistidine phosphatase that specifically associates with and dephosphorylates the catalytic histidine on nucleoside diphosphate kinase B (NDPK-B). By dephosphorylating NDPK-B, PGAM5 negatively regulates CD4(+) T cells by inhibiting NDPK-B-mediated histidine phosphorylation and activation of the K(+) channel KCa3.1, which is required for TCR-stimulated Ca(2+) influx and cytokine production. Using recently developed monoclonal antibodies that specifically recognize phosphorylation of nitrogens at the N1 (1-pHis) or N3 (3-pHis) positions of the imidazole ring, we detect for the first time phosphoisoform-specific regulation of histidine-phosphorylated proteins in vivo, and we link these modifications to TCR signaling. These results represent an important step forward in studying the role of histidine phosphorylation in mammalian biology and disease.
Collapse
Affiliation(s)
- Saswati Panda
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY 10016, USA; The Helen L. and Martin S. Kimmel Center for Biology and Medicine, New York University Langone Medical Center, New York, NY 10016, USA; Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Shekhar Srivastava
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY 10016, USA; The Helen L. and Martin S. Kimmel Center for Biology and Medicine, New York University Langone Medical Center, New York, NY 10016, USA; Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, NY 10016, USA; Division of Nephrology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Zhai Li
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY 10016, USA; The Helen L. and Martin S. Kimmel Center for Biology and Medicine, New York University Langone Medical Center, New York, NY 10016, USA; Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, NY 10016, USA
| | - Martin Vaeth
- Department of Pathology, New York University Langone Medical Center, New York, NY 10016, USA
| | - Stephen R Fuhs
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Edward Y Skolnik
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Medical Center, New York, NY 10016, USA; The Helen L. and Martin S. Kimmel Center for Biology and Medicine, New York University Langone Medical Center, New York, NY 10016, USA; Skirball Institute for Biomolecular Medicine, New York University Langone Medical Center, New York, NY 10016, USA; Division of Nephrology, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
36
|
Shoshan E, Braeuer RR, Kamiya T, Mobley AK, Huang L, Vasquez ME, Velazquez-Torres G, Chakravarti N, Ivan C, Prieto V, Villares GJ, Bar-Eli M. NFAT1 Directly Regulates IL8 and MMP3 to Promote Melanoma Tumor Growth and Metastasis. Cancer Res 2016; 76:3145-55. [PMID: 27013197 PMCID: PMC4891299 DOI: 10.1158/0008-5472.can-15-2511] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 03/21/2016] [Indexed: 12/21/2022]
Abstract
Nuclear factor of activated T cell (NFAT1, NFATC2) is a transcription factor that binds and positively regulates IL2 expression during T-cell activation. NFAT1 has important roles in both innate and adaptive immune responses, but its involvement in cancer is not completely understood. We previously demonstrated that NFAT1 contributes to melanoma growth and metastasis by regulating the autotaxin gene (Enpp2). Here, we report a strong correlation between NFAT1 expression and metastatic potential in melanoma cell lines and tumor specimens. To elucidate the mechanisms underlying NFAT1 overexpression during melanoma progression, we conducted a microarray on a highly metastatic melanoma cell line in which NFAT1 expression was stably silenced. We identified and validated two downstream targets of NFAT1, IL8, and MMP3. Accordingly, NFAT1 depletion in metastatic melanoma cell lines was associated with reduced IL8 and MMP3 expression, whereas NFAT1 overexpression in a weakly metastatic cell line induced expression of these targets. Restoration of NFAT1 expression recovered IL8 and MMP3 expression levels back to baseline, indicating that both are direct targets of NFAT1. Moreover, in vivo studies demonstrated that NFAT1 and MMP3 promoted melanoma tumor growth and lung metastasis. Collectively, our findings assign a new role for NFAT1 in melanoma progression, underscoring the multifaceted functions that immunomodulatory factors may acquire in an unpredictable tumor microenvironment. Cancer Res; 76(11); 3145-55. ©2016 AACR.
Collapse
Affiliation(s)
- Einav Shoshan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Russell R Braeuer
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Takafumi Kamiya
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Aaron K Mobley
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Li Huang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mayra E Vasquez
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Nitin Chakravarti
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cristina Ivan
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Victor Prieto
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Menashe Bar-Eli
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
37
|
Vaeth M, Eckstein M, Shaw PJ, Kozhaya L, Yang J, Berberich-Siebelt F, Clancy R, Unutmaz D, Feske S. Store-Operated Ca(2+) Entry in Follicular T Cells Controls Humoral Immune Responses and Autoimmunity. Immunity 2016; 44:1350-64. [PMID: 27261277 DOI: 10.1016/j.immuni.2016.04.013] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Revised: 02/10/2016] [Accepted: 04/22/2016] [Indexed: 12/22/2022]
Abstract
T follicular helper (Tfh) cells promote affinity maturation of B cells in germinal centers (GCs), whereas T follicular regulatory (Tfr) cells limit the GC reaction. Store-operated Ca(2+) entry (SOCE) through Ca(2+) release-activated Ca(2+) (CRAC) channels mediated by STIM and ORAI proteins is a fundamental signaling pathway in T lymphocytes. Conditional deletion of Stim1 and Stim2 genes in T cells abolished SOCE and strongly reduced antibody-mediated immune responses following viral infection caused by impaired differentiation and function of Tfh cells. Conversely, aging Stim1Stim2-deficient mice developed humoral autoimmunity with spontaneous autoantibody production due to abolished Tfr cell differentiation in the presence of residual Tfh cells. Mechanistically, SOCE controlled Tfr and Tfh cell differentiation through NFAT-mediated IRF4, BATF, and Bcl-6 transcription-factor expression. SOCE had a dual role in controlling the GC reaction by regulating both Tfh and Tfr cell differentiation, thus enabling protective B cell responses and preventing humoral autoimmunity.
Collapse
Affiliation(s)
- Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Miriam Eckstein
- NYU College of Dentistry, New York University, New York, NY 10010, USA
| | - Patrick J Shaw
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | - Lina Kozhaya
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Jun Yang
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA
| | | | - Robert Clancy
- Department of Medicine, New York University School of Medicine, New York, NY 10016, USA
| | - Derya Unutmaz
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|