1
|
Shen Y, Chen YF, Yan DF, Contreras LA, Petroze FN, Srinivasan R, Farrar AN, Trainor GL, Desai AB, Adams DJ. XPO1-Targeting Selective Inhibitors of Transcriptional Activation Suppress Graft-versus-Host Disease. J Med Chem 2025. [PMID: 40366915 DOI: 10.1021/acs.jmedchem.5c00989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Exportin-1 (XPO1) is a mediator of nuclear-to-cytoplasmic protein trafficking. The XPO1-targeting selective inhibitor of nuclear export (SINE) Selinexor is FDA-approved for relapsed hematological malignancies. Recently, we reported a unique class of XPO1 modulators that suppressed T cell activation without impairing nuclear export or cell viability (the selective inhibitors of transcriptional activation, or SITAs), suggesting that XPO1 may be a therapeutic target in T cell-driven diseases. Here, we analyzed structure-activity relationships of two structurally distinct subseries of SITAs. A set of pyridine-containing structures attained high cellular potencies (EC50 1 nM), while a complementary set of pyrrolotriazine-containing molecules balanced cellular potency with desirable physicochemical properties. Lead molecules from both subseries demonstrated in vivo XPO1 engagement, were efficacious in a mouse model of graft versus host disease, and showed superior tolerability to Selinexor. This study provides evidence that optimized XPO1-targeting SITAs can extend XPO1 as a therapeutic target to indications beyond oncology.
Collapse
Affiliation(s)
- Yanqiu Shen
- Department of Genetics and Genome Sciences and Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Yi Fan Chen
- Department of Genetics and Genome Sciences and Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - David F Yan
- Department of Genetics and Genome Sciences and Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Lyannah A Contreras
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Frederick N Petroze
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Ramya Srinivasan
- Department of Genetics and Genome Sciences and Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Amanda N Farrar
- Department of Genetics and Genome Sciences and Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - George L Trainor
- Harrington Discovery Institute at University Hospitals, Cleveland, Ohio 44106, United States
| | - Amar B Desai
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Drew J Adams
- Department of Genetics and Genome Sciences and Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| |
Collapse
|
2
|
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
|
3
|
Schoepfer A, Asikainen S, Biedermann L, Kreienbuehl A, Godat A, Dommann C, Straumann A, Greuter T. Swallowed Topical Tacrolimus Induces Clinical and Histological Remission in a Subset of Patients with Severe Lymphocytic Esophagitis. Inflamm Intest Dis 2025; 10:41-49. [PMID: 39898010 PMCID: PMC11785400 DOI: 10.1159/000542812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 11/14/2024] [Indexed: 02/04/2025] Open
Abstract
Introduction Lymphocytic esophagitis (LyE) represents a chronic inflammatory disease of the esophagus with low response rates to topical steroids. Thus, novel treatment options such as swallowed topical tacrolimus, particularly for refractory cases, are urgently needed. Methods We retrospectively analyzed patients with LyE enrolled in the Swiss eosinophilic esophagitis database that received treatment with a swallowed tacrolimus syrup (1 mg bid). We compared clinical (visual analogue scale [VAS] 0-10), endoscopic (VAS, Endoscopic Reference Score [EREFS]), and histological (peak lymphocyte count) disease activity before versus after treatment. Results Out of 17 LyE patients, we identified a total of 7 patients undergoing tacrolimus treatment (4 males, median age 71.3 years, IQR: 61.3-76.5, median diagnostic delay of 51.0 months, IQR: 24.5-62.0). Six patients had been previously treated with PPI, five with topical and/or systemic steroids. All patients were treated with topical tacrolimus corresponding to 1 mg bid (for a median of 13 weeks, IQR: 11-15). All patients had clinically, and histologically active disease at baseline. Topical tacrolimus treatment resulted in histological remission (<30 lymphocytes/hpf) in 3/7 patients (42.9%), while 4/7 patients achieved symptomatic remission (VAS for dysphagia ≤2, 57.1%). Overall, clinical (VAS 5 vs. 2, p = 0.0625) and endoscopic activity (VAS 5 vs. 2, p = 0.0625, and EREFS 3 vs. 2, p = 0.125) decreased. Measurement of tacrolimus trough levels in 4/7 patients (range 2.1-3.9 μg/L) revealed some degree of systemic absorption. Mild adverse events to the tacrolimus treatment were seen in 2 patients (esophageal candidiasis, hyposensitivity around lips). No impact on kidney function was observed during the treatment period. Conclusion Topical tacrolimus appears to be a potential treatment option for severe LyE, particularly after failure of PPI and/or topical steroids. Further studies are needed, in particular regarding the optimal galenic formulation to avoid systemic absorption.
Collapse
Affiliation(s)
- Alain Schoepfer
- Division of Gastroenterology and Hepatology, CHUV University Hospital Lausanne and University of Lausanne UNIL, Lausanne, Switzerland
| | - Sofia Asikainen
- Division of Gastroenterology and Hepatology, CHUV University Hospital Lausanne and University of Lausanne UNIL, Lausanne, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Andrea Kreienbuehl
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Anne Godat
- Division of Gastroenterology and Hepatology, GZO – Zurich Regional Health Center, Wetzikon, Switzerland
| | - Corina Dommann
- Department of Pathology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Thomas Greuter
- Division of Gastroenterology and Hepatology, CHUV University Hospital Lausanne and University of Lausanne UNIL, Lausanne, Switzerland
- Department of Gastroenterology and Hepatology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
- Division of Gastroenterology and Hepatology, GZO – Zurich Regional Health Center, Wetzikon, Switzerland
| |
Collapse
|
4
|
Azeem M, Helal M, Klein-Hessling S, Serfling E, Goebeler M, Muhammad K, Kerstan A. NFATc1 Fosters Allergic Contact Dermatitis Responses by Enhancing the Induction of IL-17-Producing CD8 Cells. J Invest Dermatol 2024:S0022-202X(24)03036-7. [PMID: 39733935 DOI: 10.1016/j.jid.2024.11.014] [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: 11/13/2023] [Revised: 11/15/2024] [Accepted: 11/18/2024] [Indexed: 12/31/2024]
Abstract
A plethora of data supports a major role of CD4+ and CD8+ T lymphocytes for the initiation, progression, and maintenance of allergic contact dermatitis. However, in-depth understanding of the molecular mechanisms is still limited. NFATc1 plays an essential role in T-cell activation. We therefore investigated its impact on contact hypersensitivity, the mouse model for allergic contact dermatitis. The contact hypersensitivity response to 2,4,6-trinitrochlorobenzene was diminished in Nfatc1fl/flxCd4-cre mice (Nfatc1-/-) compared with that in wild-type mice and associated with a lower percentage of IL-17-producing CD8+ T (Tc17) cells in both inflamed skin and draining lymph nodes. In vitro Tc17 polarization assays revealed that Nfatc1-/- CD8+ T cells have a reduced capacity to polarize into Tc17 cells. Applying single-cell RNA sequencing, we realized that NFATc1 controls the T-cell differentiation fate. In the absence of NFATc1, CD8+ T cells favor the development of IFN-γ-secreting CD8+ T (Tc1) lymphocytes, whereas in its presence, they turn into Tc17 cells. Finally, the adoptive transfer of 2,4,6-trinitrochlorobenzene-sensitized wild-type CD8+ T cells restored the contact hypersensitivity response in naïve Nfatc1-/- mice. Our data demonstrate that NFATc1 contributes to the development of Tc17 cells and might present a promising target to alleviate CD8+ T-cell-mediated allergic responses.
Collapse
Affiliation(s)
- Muhammad Azeem
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany; Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Moutaz Helal
- Mildred Scheel Early Career Centre (MSNZ) for Cancer Research, University Hospital Würzburg, Würzburg, Germany
| | - Stefan Klein-Hessling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Edgar Serfling
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Matthias Goebeler
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany
| | - Khalid Muhammad
- Department of Molecular Pathology, Institute of Pathology, University of Würzburg, Würzburg, Germany; Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates.
| | - Andreas Kerstan
- Department of Dermatology, Venereology and Allergology, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
5
|
Chen YF, Ghazala M, Friedrich RM, Cordova BA, Petroze FN, Srinivasan R, Allan KC, Yan DF, Sax JL, Carr K, Tomchuck SL, Fedorov Y, Huang AY, Desai AB, Adams DJ. Targeting the chromatin binding of exportin-1 disrupts NFAT and T cell activation. Nat Chem Biol 2024; 20:1260-1271. [PMID: 38528120 DOI: 10.1038/s41589-024-01586-5] [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: 06/13/2023] [Accepted: 02/22/2024] [Indexed: 03/27/2024]
Abstract
Exportin-1 (XPO1/CRM1) plays a central role in the nuclear-to-cytoplasmic transport of hundreds of proteins and contributes to other cellular processes, such as centrosome duplication. Small molecules targeting XPO1 induce cytotoxicity, and selinexor was approved by the Food and Drug Administration in 2019 as a cancer chemotherapy for relapsed multiple myeloma. Here, we describe a cell-type-dependent chromatin-binding function for XPO1 that is essential for the chromatin occupancy of NFAT transcription factors and thus the appropriate activation of T cells. Additionally, we establish a class of XPO1-targeting small molecules capable of disrupting the chromatin binding of XPO1 without perturbing nuclear export or inducing cytotoxicity. This work defines a broad transcription regulatory role for XPO1 that is essential for T cell activation as well as a new class of XPO1 modulators to enable therapeutic targeting of XPO1 beyond oncology including in T cell-driven autoimmune disorders.
Collapse
Affiliation(s)
- Yi Fan Chen
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Maryam Ghazala
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ryan M Friedrich
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Brittany A Cordova
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Frederick N Petroze
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Ramya Srinivasan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kevin C Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David F Yan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Joel L Sax
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Kelley Carr
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Suzanne L Tomchuck
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yuriy Fedorov
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alex Y Huang
- Department of Pediatrics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Amar B Desai
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Drew J Adams
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Chemical Biology Program, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
6
|
Wang Y, Mei X, Lin Z, Yang X, Cao J, Zhong J, Wang J, Cheng L, Wang Z. Virus infection pattern imprinted and diversified the differentiation of T-cell memory in transcription and function. Front Immunol 2024; 14:1334597. [PMID: 38264657 PMCID: PMC10803622 DOI: 10.3389/fimmu.2023.1334597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/14/2023] [Indexed: 01/25/2024] Open
Abstract
Introduction Memory T (Tm) cells are a subpopulation of immune cells with great heterogeneity. Part of this diversity came from T cells that were primed with different viruses. Understanding the differences among different viral-specific Tms will help develop new therapeutic strategies for viral infections. Methods In this study, we compared the transcriptome of Tm cells that primed with CMV, EBV and SARS-CoV-2 with single-cell sequencing and studied the similarities and differences in terms of subpopulation composition, activation, metabolism and transcriptional regulation. Results We found that CMV is marked by plentiful cytotoxic Temra cells, while EBV is more abundant in functional Tem cells. More importantly, we found that CD28 and CTLA4 can be used as continuous indicators to interrogate the antiviral ability of T cells. Furthermore, we proposed that REL is a main regulatory factor for CMV-specific T cells producing cytokines and plays an antiviral role. Discussion Our data gives deep insight into molecular characteristics of Tm subsets from different viral infection, which is important to understand T cell immunization. Furthermore, our results provide basic background knowledges for T cell based vaccine development in future.
Collapse
Affiliation(s)
- Yuan Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Xinyue Mei
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhengfang Lin
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xiaoyun Yang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Jinpeng Cao
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| | - Jiaying Zhong
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junxiang Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Li Cheng
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhongfang Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
- Guangzhou National Laboratory, Bioland, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Zaichick S, Caraveo G. Harnessing IGF-1 and IL-2 as biomarkers for calcineurin activity to tailor optimal FK506 dosage in α-synucleinopathies. Front Mol Biosci 2023; 10:1292555. [PMID: 38094080 PMCID: PMC10716490 DOI: 10.3389/fmolb.2023.1292555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/16/2023] [Indexed: 02/01/2024] Open
Abstract
Introduction: Rise in Calcium (Ca2+) and hyperactive Ca2+-dependent phosphatase calcineurin represent two key determinants of a-synuclein (a-syn) pathobiology implicated in Parkinson's Disease (PD) and other neurodegenerative diseases. Calcineurin activity can be inhibited with FK506, a Food and Drug Administration (FDA)-approved compound. Our previous work demonstrated a protective effect of low doses of FK506 against a-syn pathology in various models of a-syn related pathobiology. Methods: Control and a-syn-expressing mice (12-18 months old) were injected with vehicle or two single doses of FK506 administered 4 days apart. Cerebral cortex and serum from these mice were collected and assayed using a meso scale discovery quickplex SQ 120 for cytokines and Enzyme-linked immunosorbent assay for IGF-1. Results: In this study we present evidence that reducing calcineurin activity with FK506 in a-syn transgenic mice increased insulin growth factor (IGF-1), while simultaneously decreasing IL-2 levels in both cerebral cortex and serum. Discussion: The highly conserved Ca2+/calcineurin signaling pathway is known to be affected in a-syn-dependent human disease. FK506, an already approved drug for other uses, exhibits high brain penetrance and a proven safety profile. IL-2 and IGF-1 are produced throughout life and can be measured using standard clinical methods. Our findings provide two potential biomarkers that could guide a clinical trial of FK506 in PD patients, without posing significant logistical or regulatory challenges.
Collapse
Affiliation(s)
| | - Gabriela Caraveo
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| |
Collapse
|
8
|
Shim JA, Lee SM, Jeong JW, Kim H, Son WJ, Park JH, Song P, Im SH, Bae S, Park JH, Jo Y, Hong C. NFAT1 and NFκB regulates expression of the common γ-chain cytokine receptor in activated T cells. Cell Commun Signal 2023; 21:309. [PMID: 37904191 PMCID: PMC10617197 DOI: 10.1186/s12964-023-01326-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 09/18/2023] [Indexed: 11/01/2023] Open
Abstract
INTRODUCTION Cytokines of the common γ chain (γc) family are critical for the development, differentiation, and survival of T lineage cells. Cytokines play key roles in immunodeficiencies, autoimmune diseases, allergies, and cancer. Although γc is considered an assistant receptor to transmit cytokine signals and is an indispensable receptor in the immune system, its regulatory mechanism is not yet well understood. OBJECTIVE This study focused on the molecular mechanisms that γc expression in T cells is regulated under T cell receptor (TCR) stimulation. METHODS The γc expression in TCR-stimulated T cells was determined by flow cytometry, western blot and quantitative RT-PCR. The regulatory mechanism of γc expression in activated T cells was examined by promoter-luciferase assay and chromatin immunoprecipitation assays. NFAT1 and NFκB deficient cells generated using CRISPR-Cas9 and specific inhibitors were used to examine their role in regulation of γc expression. Specific binding motif was confirmed by γc promotor mutant cells generated using CRISPR-Cas9. IL-7TgγcTg mice were used to examine regulatory role of γc in cytokine signaling. RESULTS We found that activated T cells significantly upregulated γc expression, wherein NFAT1 and NFκB were key in transcriptional upregulation via T cell receptor stimulation. Also, we identified the functional binding site of the γc promoter and the synergistic effect of NFAT1 and NFκB in the regulation of γc expression. Increased γc expression inhibited IL-7 signaling and rescued lymphoproliferative disorder in an IL-7Tg animal model, providing novel insights into T cell homeostasis. CONCLUSION Our results indicate functional cooperation between NFAT1 and NFκB in upregulating γc expression in activated T cells. As γc expression also regulates γc cytokine responsiveness, our study suggests that γc expression should be considered as one of the regulators in γc cytokine signaling and the development of T cell immunotherapies. Video Abstract.
Collapse
Affiliation(s)
- Ju A Shim
- Department of Anatomy, Pusan National University School of Medicine, Room 504, 49 Busandaehak-Ro, Yangsan, Gyeongsangnam-Do, 50612, South Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - So Min Lee
- Department of Anatomy, Pusan National University School of Medicine, Room 504, 49 Busandaehak-Ro, Yangsan, Gyeongsangnam-Do, 50612, South Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Jin Woo Jeong
- Department of Anatomy, Pusan National University School of Medicine, Room 504, 49 Busandaehak-Ro, Yangsan, Gyeongsangnam-Do, 50612, South Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Hyori Kim
- Department of Anatomy, Pusan National University School of Medicine, Room 504, 49 Busandaehak-Ro, Yangsan, Gyeongsangnam-Do, 50612, South Korea
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Woo Jae Son
- Department of Chemistry, Hanyang University, Seoul, 04763, Republic of Korea
| | - Jun Hong Park
- Herbal Medicine Resources Research Center, Korea Institute of Oriental Medicine, Naju, 58245, Republic of Korea
- University of Science & Technology (UST), KIOM Campus, Korean Convergence Medicine Major, Daejeon, 34054, Republic of Korea
| | - Parkyong Song
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea
| | - Sangsu Bae
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892, USA
| | - Yuna Jo
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
- Department of Anatomy, Pusan National University School of Medicine, Room 515, 49 Busandaehak-Ro, Yangsan, Gyeongsangnam-Do, 50612, South Korea.
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Room 504, 49 Busandaehak-Ro, Yangsan, Gyeongsangnam-Do, 50612, South Korea.
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
- PNU GRAND Convergence Medical Science Education Research Center, Pusan National University School of Medicine, Yangsan, 50612, Republic of Korea.
| |
Collapse
|
9
|
Gamboa M, Kitamura N, Miura K, Noda S, Kaminuma O. Evolutionary mechanisms underlying the diversification of nuclear factor of activated T cells across vertebrates. Sci Rep 2023; 13:6468. [PMID: 37156933 PMCID: PMC10167247 DOI: 10.1038/s41598-023-33751-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 04/18/2023] [Indexed: 05/10/2023] Open
Abstract
The mechanisms of immunity linked to biological evolution are crucial for understanding animal morphogenesis, organogenesis, and biodiversity. The nuclear factor of activated T cells (NFAT) family consists of five members (NFATc1-c4, 5) with different functions in the immune system. However, the evolutionary dynamics of NFATs in vertebrates has not been explored. Herein, we investigated the origin and mechanisms underlying the diversification of NFATs by comparing the gene, transcript and protein sequences, and chromosome information. We defined an ancestral origin of NFATs during the bilaterian development, dated approximately 650 million years ago, where NFAT5 and NFATc1-c4 were derived independently. The conserved parallel evolution of NFATs in multiple species was probably attributed to their innate nature. Conversely, frequent gene duplications and chromosomal rearrangements in the recently evolved taxa have suggested their roles in the adaptive immune evolution. A significant correlation was observed between the chromosome rearrangements with gene duplications and the structural fixation changes in vertebrate NFATs, suggesting their role in NFAT diversification. Remarkably, a conserved gene structure around NFAT genes with vertebrate evolutionary-related breaking points indicated the inheritance of NFATs with their neighboring genes as a unit. The close relationship between NFAT diversification and vertebrate immune evolution was suggested.
Collapse
Affiliation(s)
- Maribet Gamboa
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8553, Japan.
- Department of Ecology, Faculty of Sciences, Universidad Católica de la Santísima Concepción, 4090541, Concepción, Chile.
| | - Noriko Kitamura
- Neurovirology Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, 156-8506, Japan
| | - Kento Miura
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8553, Japan
| | - Satoko Noda
- Graduate School of Science and Engineering, Ibaraki University, Ibaraki, 310-8512, Japan
| | - Osamu Kaminuma
- Department of Disease Model, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, 734-8553, Japan.
| |
Collapse
|
10
|
Knaryan VH, Sarukhanyan FP. [Ca2+-regulated enzymes calpain and calcineurin in neurodegenerative processes and prospects for neuroprotective pharmacotherapy]. Zh Nevrol Psikhiatr Im S S Korsakova 2023; 123:32-40. [PMID: 37490663 DOI: 10.17116/jnevro202312307132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Calcium (Ca2+) and Ca2+-regulated enzymes calpain and calcineurin are the key molecules of signaling mechanisms in neurons and ensure the normal course of intracellular neurochemical and neurophysiological processes. The imbalance and increase in the intracellular level of Ca2+ correlates with the activation of calpain and calcineurin. Inactivation of endogenous inhibitors and/or absence of exogenous pharmacological inhibitors of these enzymes may induce a cascade of intracellular mechanisms that are detrimental to the structural integrity and functional activity of neurons. The interrelated processes of Ca2+ imbalance, dysregulation of calpain and calcineurin are directly related to the development of intracellular pathophysiological reactions leading to the degeneration and death of selective neuronal populations in neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. The review briefly presents the characteristics of calpain and calcineurin, their interrelated role in the neurodegeneration processes. Data on the efficiency of the exogenous inhibitors (in vivo, in vitro) point out the potential role of pharmacological regulation of calpain and calcineurin for neuroprotection.
Collapse
Affiliation(s)
- V H Knaryan
- Buniatian Institute of Biochemistry NAS RA, Yerevan, Armenia
| | - F P Sarukhanyan
- Buniatian Institute of Biochemistry NAS RA, Yerevan, Armenia
| |
Collapse
|
11
|
Mendonça MM, da Cruz KR, Pinheiro DDS, Moraes GCA, Ferreira PM, Ferreira-Neto ML, da Silva ES, Gonçalves RV, Pedrino GR, Fajemiroye JO, Xavier CH. Dysregulation in erythrocyte dynamics caused by SARS-CoV-2 infection: possible role in shuffling the homeostatic puzzle during COVID-19. Hematol Transfus Cell Ther 2022; 44:235-245. [PMID: 35098037 PMCID: PMC8786672 DOI: 10.1016/j.htct.2022.01.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/04/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction The evolving COVID-19 pandemic became a hallmark in human history, not only by changing lifestyles, but also by enriching scientific knowledge on viral infection and its consequences. Objective Although the management of cardiorespiratory changes is pivotal to a favorable prognosis during severe clinical findings, dysregulation of other systems caused by SARS-CoV-2 infection may imbalance erythrocyte dynamics, such as a bidirectional positive feedback loop pathophysiology. Method and Results Recent evidence shows that SARS-CoV-2 is capable of affecting the genetics and dynamics of erythrocytes and this coexists with a non-homeostatic function of cardiovascular, respiratory and renal systems during COVID-19. In hypothesis, SARS-CoV-2-induced systematical alterations of erythrocytes dynamics would constitute a setpoint for COVID-19-related multiple organ failure syndrome and death. Conclusion The present review covers the most frequent erythrocyte-related non-homeostatic findings during COVID-19 capable of providing mechanistic clues of SARS-CoV-2-induced infection and inspiring therapeutic-oriented scientific evidence.
Collapse
Affiliation(s)
| | - Kellen Rosa da Cruz
- Instituto de Ciências Biológicas da Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
| | | | | | - Patricia Maria Ferreira
- Instituto de Ciências Biológicas da Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
| | - Marcos Luiz Ferreira-Neto
- Instituto de Ciências Biomédicas da Universidade Federal de Uberlândia (ICBIM UFU), Uberlândia, MG, Brazil
| | | | | | | | - James O Fajemiroye
- Instituto de Ciências Biológicas da Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil
| | - Carlos Henrique Xavier
- Instituto de Ciências Biológicas da Universidade Federal de Goiás (UFG), Goiânia, GO, Brazil.
| |
Collapse
|
12
|
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
|
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
|
Kader HA, Azeem M, Jwayed SA, Al-Shehhi A, Tabassum A, Ayoub MA, Hetta HF, Waheed Y, Iratni R, Al-Dhaheri A, Muhammad K. Current Insights into Immunology and Novel Therapeutics of Atopic Dermatitis. Cells 2021; 10:1392. [PMID: 34200009 PMCID: PMC8226506 DOI: 10.3390/cells10061392] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Atopic dermatitis (AD) is one of the most prevalent inflammatory disease among non-fatal skin diseases, affecting up to one fifth of the population in developed countries. AD is characterized by recurrent pruritic and localized eczema with seasonal fluctuations. AD initializes the phenomenon of atopic march, during which infant AD patients are predisposed to progressive secondary allergies such as allergic rhinitis, asthma, and food allergies. The pathophysiology of AD is complex; onset of the disease is caused by several factors, including strong genetic predisposition, disrupted epidermal barrier, and immune dysregulation. AD was initially characterized by defects in the innate immune system and a vigorous skewed adaptive Th2 response to environmental agents; there are compelling evidences that the disorder involves multiple immune pathways. Symptomatic palliative treatment is the only strategy to manage the disease and restore skin integrity. Researchers are trying to more precisely define the contribution of different AD genotypes and elucidate the role of various immune axes. In this review, we have summarized the current knowledge about the roles of innate and adaptive immune responsive cells in AD. In addition, current and novel treatment strategies for the management of AD are comprehensively described, including some ongoing clinical trials and promising therapeutic agents. This information will provide an asset towards identifying personalized targets for better therapeutic outcomes.
Collapse
Affiliation(s)
- Hidaya A. Kader
- Department of Biology, College of Science, UAE University, Al Ain 15551, United Arab Emirates; (H.A.K.); (S.A.J.); (A.A.-S.); (M.A.A.); (R.I.)
| | - Muhammad Azeem
- Department of Pathology, University of Würzburg, 97080 Würzburg, Germany;
| | - Suhib A. Jwayed
- Department of Biology, College of Science, UAE University, Al Ain 15551, United Arab Emirates; (H.A.K.); (S.A.J.); (A.A.-S.); (M.A.A.); (R.I.)
| | - Aaesha Al-Shehhi
- Department of Biology, College of Science, UAE University, Al Ain 15551, United Arab Emirates; (H.A.K.); (S.A.J.); (A.A.-S.); (M.A.A.); (R.I.)
| | - Attia Tabassum
- Department of Dermatology, Mayo Hospital, Lahore 54000, Pakistan;
| | - Mohammed Akli Ayoub
- Department of Biology, College of Science, UAE University, Al Ain 15551, United Arab Emirates; (H.A.K.); (S.A.J.); (A.A.-S.); (M.A.A.); (R.I.)
| | - Helal F. Hetta
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt;
| | - Yasir Waheed
- Foundation University Medical College, Foundation University Islamabad, Islamabad 44000, Pakistan;
| | - Rabah Iratni
- Department of Biology, College of Science, UAE University, Al Ain 15551, United Arab Emirates; (H.A.K.); (S.A.J.); (A.A.-S.); (M.A.A.); (R.I.)
| | - Ahmed Al-Dhaheri
- Department of Dermatology, Tawam Hospital, Al Ain 15551, United Arab Emirates;
| | - Khalid Muhammad
- Department of Biology, College of Science, UAE University, Al Ain 15551, United Arab Emirates; (H.A.K.); (S.A.J.); (A.A.-S.); (M.A.A.); (R.I.)
| |
Collapse
|
15
|
Kitamura N, Kaminuma O. Isoform-Selective NFAT Inhibitor: Potential Usefulness and Development. Int J Mol Sci 2021; 22:2725. [PMID: 33800389 PMCID: PMC7962815 DOI: 10.3390/ijms22052725] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/30/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT), which is the pharmacological target of immunosuppressants cyclosporine and tacrolimus, has been shown to play an important role not only in T cells (immune system), from which their name is derived, but also in many biological events. Therefore, functional and/or structural abnormalities of NFAT are linked to the pathogenesis of diseases in various organs. The NFAT protein family consists of five isoforms, and each isoform performs diverse functions and has unique expression patterns in the target tissues. This diversity has made it difficult to obtain ideal pharmacological output for immunosuppressants that inhibit the activity of almost all NFAT family members, causing serious and wide-ranging side effects. Moreover, it remains unclear whether isoform-selective NFAT regulation can be achieved by targeting the structural differences among NFAT isoforms and whether this strategy can lead to the development of better drugs than the existing ones. This review summarizes the role of the NFAT family members in biological events, including the development of various diseases, as well as the usefulness of and problems associated with NFAT-targeting therapies, including those dependent on current immunosuppressants. Finally, we propose a novel therapeutic strategy based on the molecular mechanisms that enable selective regulation of specific NFAT isoforms.
Collapse
Affiliation(s)
- Noriko Kitamura
- Laboratory of Allergy and Immunology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
| | - Osamu Kaminuma
- Laboratory of Allergy and Immunology, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan;
- Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
| |
Collapse
|
16
|
Jia C, Yao Z, Lin Z, Zhao L, Cai X, Chen S, Deng M, Zhang Q. circNFATC3 sponges miR-548I acts as a ceRNA to protect NFATC3 itself and suppressed hepatocellular carcinoma progression. J Cell Physiol 2020; 236:1252-1269. [PMID: 32667692 DOI: 10.1002/jcp.29931] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 06/15/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022]
Abstract
Circular RNAs (circRNA) have been reported as regulators involved in hepatocellular carcinoma (HCC), but their mechanism of activity remains unknown. This study performed quantitative reverse-transcription polymerase chain reaction to determine if circNFATC3 was downregulated in 46 paired HCC tissues and cell lines. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, apoptotic, and transwell assay proved that circNFATC3 can inhibit hepatoma cell proliferation, apoptosis, and migration/invasion in vitro. Mouse xenograft assay demonstrated that circNFATC3 suppressed tumor size and weight and reduced lung metastasis in vivo, and vice versa. The RNA-seq results showed that NFATC3 itself was the most significantly differentially expressed gene when circNFATC3 was manipulated, and bioinformatics and luciferase reporter assays verified circNFATC3 regulated the expression of NFATC3 by interacting with the hsa-miR-548I. Additionally, it was also indicated that the level of NFATC3 was downregulated in HCC patients also and was significantly correlated with the staging and prognosis of HCC. Moreover, both circNFATC3 and NFATC3 were shown to inhibit the phosphorylation of JNK, c-Jun, AKT, and mTOR signaling pathways. Overall, the circNFATC3 can sponge miR-548I to protect NFATC3 itself, then it regulates hepatoma cell function via the JNK, c-Jun, AKT, and mTOR signaling pathways, and the circNFATC3 can be a tumor-repressor on HCC.
Collapse
Affiliation(s)
- Changchang Jia
- Department of Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhicheng Yao
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zexiao Lin
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liyun Zhao
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiurong Cai
- Department of Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shaohong Chen
- Department of Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Meihai Deng
- Department of Hepatobilliary Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qi Zhang
- Department of Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Ali A, Alzeyoudi SAR, Almutawa SA, Alnajjar AN, Vijayan R. Molecular basis of the therapeutic properties of hemorphins. Pharmacol Res 2020; 158:104855. [PMID: 32438036 DOI: 10.1016/j.phrs.2020.104855] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 04/09/2020] [Accepted: 04/20/2020] [Indexed: 12/26/2022]
Abstract
Hemorphins are endogenous peptides, 4-10 amino acids long, belonging to the family of atypical opioid peptides released during the sequential cleavage of hemoglobin protein. Hemorphins have been shown to exhibit diverse therapeutic effects in both human and animal models. However, the precise cellular and molecular mechanisms involved in such effects remain elusive. In this review, we summarize and propose potential mechanisms based on studies that investigated the biological activity of hemorphins of different lengths on multiple therapeutic targets. Special emphasis is given to molecular events related to renin-angiotensin system (RAS), opioid receptors and insulin-regulated aminopeptidase receptor (IRAP). This review provides a comprehensive coverage of the molecular mechanisms that underpin the therapeutic potential of hemorphins. Furthermore, it highlights the role of various hemorphin residues in pathological conditions, which could be explored further for therapeutic purposes.
Collapse
Affiliation(s)
- Amanat Ali
- Department of Biology, College of Science, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | | | - Shamma Abdulla Almutawa
- Department of Biology, College of Science, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Alya Nasir Alnajjar
- Department of Biology, College of Science, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates
| | - Ranjit Vijayan
- Department of Biology, College of Science, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates.
| |
Collapse
|
18
|
Zeng Q, Zhang A. Assessing potential mechanisms of arsenic-induced skin lesions and cancers: Human and in vitro evidence. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2020; 260:113919. [PMID: 31995775 DOI: 10.1016/j.envpol.2020.113919] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 01/03/2020] [Accepted: 01/04/2020] [Indexed: 05/10/2023]
Abstract
Environmental exposure to arsenic is a major public health challenge worldwide. In detailing the hallmark signs of chronic arsenic exposure, previous studies have shown that epigenetic and immune dysfunction are associated with arsenic-induced skin lesions; however, knowledge regarding interactions between the mechanisms listed above is limited. In this study, a total of 106 skin samples were collected over the past 20 years. Based on the presence or absence of high arsenic exposure, the participants were divided into arsenic exposure (72) and reference (34) groups. Additionally, the arsenic exposure group was further divided into the non-cancer group (31, including skin hyperpigmentation and hyperkeratosis) and the skin cancer group (41, including Bowen's disease, basal cell carcinoma and squamous cell carcinoma) according to a skin histopathological examination. First, the associations among miR-155, NF-AT1 with immunological dysfunction and arsenic-induced skin lesions and carcinogenesis were confirmed using these skin samples. In the arsenic-exposed group, miR-155-5p, keratin 1(Krt1), keratin 10 (Krt10), and keratin 6c (Krt6c) were significantly increased in the skin (p < 0.05), while NF-AT1, interleukin-2 (IL-2), and interferon-γ (IFN-γ) were significantly decreased (p < 0.05). Clear correlations were observed among these factors (p < 0.05). In immortalized human keratinocytes, silencing and overexpression of NF-AT1 could alter the expression and secretion of immunological dysfunction indicators (IL-2 and IFN-γ) that are induced by arsenic exposure (p < 0.05); however, miR-155-5p levels did not change significantly (p > 0.05). The miR-155-5p mimic and inhibitor could regulate the NF-AT1-mediated immunological dysfunction caused by arsenic (p < 0.05). Our study provides some limited evidence that miR-155-5p regulates the NF-AT1-mediated immunological dysfunction that is involved in the pathogenesis and carcinogenesis of arsenic. The second major finding was that Krt1 and Krt10 are markers of hyperkeratosis caused by arsenic, and Krt6c is a potential biomarker that can reflect arsenic carcinogenesis.
Collapse
Affiliation(s)
- Qibing Zeng
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, Guizhou Medical University, Guiyang, 550025, China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Department of Toxicology, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
19
|
Bogaert A, Fernandez E, Gevaert K. N-Terminal Proteoforms in Human Disease. Trends Biochem Sci 2020; 45:308-320. [PMID: 32001092 DOI: 10.1016/j.tibs.2019.12.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/06/2019] [Accepted: 12/31/2019] [Indexed: 12/20/2022]
Abstract
The collection of chemically different protein variants, or proteoforms, by far exceeds the number of protein-coding genes in the human genome. Major contributors are alternative splicing and protein modifications. In this review, we focus on those proteoforms that differ at their N termini with a molecular link to disease. We describe the main underlying mechanisms that give rise to such N-terminal proteoforms, these being splicing, initiation of protein translation, and protein modifications. Given their role in several human diseases, it is becoming increasingly clear that several of these N-terminal proteoforms may have potential as therapeutic interventions and/or for diagnosing and prognosing their associated disease.
Collapse
Affiliation(s)
- Annelies Bogaert
- VIB Center for Medical Biotechnology, VIB, B-9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Esperanza Fernandez
- VIB Center for Medical Biotechnology, VIB, B-9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Kris Gevaert
- VIB Center for Medical Biotechnology, VIB, B-9000 Ghent, Belgium; Department of Biomolecular Medicine, Ghent University, B-9000 Ghent, Belgium.
| |
Collapse
|
20
|
Kitamura N, Shindo M, Ohtsuka J, Nakamura A, Tanokura M, Hiroi T, Kaminuma O. Identification of novel interacting regions involving calcineurin and nuclear factor of activated T cells. FASEB J 2020; 34:3197-3208. [PMID: 31909857 DOI: 10.1096/fj.201902229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 12/19/2019] [Indexed: 11/11/2022]
Abstract
Nuclear factor of activated T cells (NFAT) leads to the transcription of diverse inducible genes involved in many biological processes; therefore, aberrant NFAT expression is responsible for the development and exacerbation of various disorders. Since five isoforms of NFAT (NFATc1-c4, NFAT5) exhibit distinct and overlapping functions, selective control of a part, but not all, of NFAT family members is desirable. By comparing the binding activity of each NFATc1-c4 with its regulatory enzyme, calcineurin (CN), using a quantitative immunoprecipitation assay, we found a new CN-binding region (CNBR) selectively functioning in NFATc1 and NFATc4. This region, termed CNBR3, is located between two preexisting CNBR1 and CNBR2, within the Ca2+ regulatory domain. The nuclear translocation of NFATc1 but not NFATc2 in T cells was suppressed by ectopic expression of CNBR3 and, accordingly, NFATc1-dependent cytokine expression was downregulated. Through competition assays using NFATc1-derived partial peptides and mass spectrometry with photoaffinity technology, we identified 18 amino acids in NFATc1 (Arg258 to Pro275 ) and 13 amino acids in CN catalytic subunit (CNA) (Asn77 to Gly89 ) responsible for CNA/CNBR3 binding in which Cys263 and Asp82 , respectively, played crucial roles. The possible selective regulation of NFAT-mediated biological processes by targeting this new CN/NFAT-binding region is suggested.
Collapse
Affiliation(s)
- Noriko Kitamura
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Mayumi Shindo
- Center for Basic Technology Research, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Jun Ohtsuka
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Akira Nakamura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan.,Department of Life Science, Faculty of Science, Gakushuin University, Tokyo, Japan
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takachika Hiroi
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Osamu Kaminuma
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Disease Model, Research Institute of Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
21
|
Yoshida J, Okawa Y, Oyama T, Shimoda N, Uesugi S, Takagi H, Ito Y, Kimura KI. Inhibition of Calcineurin and Glycogen Synthase Kinase-3β by Ricinoleic Acid Derived from Castor Oil. Lipids 2019; 55:89-99. [PMID: 31867745 DOI: 10.1002/lipd.12208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 11/16/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022]
Abstract
Ricinoleic acid (RA) is the main fatty acid component of castor oil and was found to inhibit Ca2+ -signal transduction pathway-mediated cell cycle regulation in a yeast-based drug screening assay. RA is expected to have antidiabetic, antiallergy, and/or anticancer properties but its target molecule is unknown. To identify a novel pharmacological effect of RA, we investigated its target molecule in the Ca2+ -signal transduction pathway. RA inhibition of calcineurin (CN) was examined in a yeast-based CN inhibitor screening assay using the rsp5A401E mutant and in a phosphatase assay using recombinant human CN. RA showed growth-restoration activity at 5 μg/spot in the CN inhibitor screening assay with the rsp5A401E yeast strain. Furthermore, it directly inhibited CN without immunophilins at Ki = 33.7 μM in a substrate-competitive manner. The effects of RA on CN in mammalian cells were further evaluated by measuring β-hexosaminidase (β-HEX) release in RBL-2H3 cells. RA at 50 μM suppressed the release of β-HEX from RBL-2H3 cells. Moreover, this compound was found to inhibit glycogen synthase kinase-3β (GSK-3β), as determined by a kinase assay using recombinant human GSK-3β. RA inhibited GSK-3β at Ki = 1.43 μM in a peptide substrate-competitive manner. The inhibition of GSK-3β by this molecule was further assessed in mammalian cells by measuring the inhibition of glucose production in H4IIE rat hepatoma cells. RA at 25 μM suppressed glucose production in these cells. These findings indicate that RA and/or castor oil could be a useful functional fatty acid to treat allergy or type 2 diabetes.
Collapse
Affiliation(s)
- Jun Yoshida
- Center for Liberal Arts and Sciences, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba, 028-3694, Japan
| | - Yusuke Okawa
- Graduate School of Agriculture, Iwate University, Morioka, 020-8550, Japan
| | - Takuya Oyama
- Faculty of Agriculture, Iwate University, Morioka, 020-8550, Japan
| | - Nozomu Shimoda
- Graduate School of Agriculture, Iwate University, Morioka, 020-8550, Japan
| | - Shota Uesugi
- The United Graduate School of Agricultural Sciences, Iwate University, Morioka, 020-8550, Japan
| | - Hiroshi Takagi
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Yoshiaki Ito
- Graduate School of Agriculture, Iwate University, Morioka, 020-8550, Japan.,Faculty of Agriculture, Iwate University, Morioka, 020-8550, Japan.,The United Graduate School of Agricultural Sciences, Iwate University, Morioka, 020-8550, Japan
| | - Ken-Ichi Kimura
- Graduate School of Agriculture, Iwate University, Morioka, 020-8550, Japan.,Faculty of Agriculture, Iwate University, Morioka, 020-8550, Japan.,The United Graduate School of Agricultural Sciences, Iwate University, Morioka, 020-8550, Japan
| |
Collapse
|
22
|
Giron F, Pastó A, Tasciotti E, Abraham BP. Nanotechnology in the Treatment of Inflammatory Bowel Disease. Inflamm Bowel Dis 2019; 25:1871-1880. [PMID: 31560054 DOI: 10.1093/ibd/izz205] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Indexed: 12/12/2022]
Abstract
Due to the lack of cure for inflammatory bowel disease (IBD) and failure of current medical therapies in many patients with IBD, a need exists in finding novel ways to treat inflammation with a high benefit and the lowest risk possible. With current medical therapies, adverse events or risks of cancer/lymphoma and infections prevent patients-and sometimes providers-in using effective therapies for treatment. Some patients develop systemic side effects that preclude them from continuing a therapy that may have been efficacious, or in other cases, current medical therapies are not adequate to control disease. Nanotechnology is an emerging field where particles, in the size of nanometers, can be used to deliver medications directly to the area of inflammation thus avoiding drug-associated systemic side effects. When using nanoparticles (NPs), only a small amount of the drug is needed, and it can be delivered directly to the inflamed site without exposure to the rest of the body. Here we review conventional and unconventional therapies applied in the treatment of IBD underlying how the introduction of NPs has improved their safety and efficacy.
Collapse
Affiliation(s)
- Fanny Giron
- Universidad Católica de Honduras, San Pedro Sula, Honduras
| | - Anna Pastó
- Istituto Oncologico Veneto-IRCCS, Padova, Italy
| | - Ennio Tasciotti
- Underwood Center for Digestive Disorders, Houston Methodist Hospital, Houston, Texas.,Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| | - Bincy P Abraham
- Underwood Center for Digestive Disorders, Houston Methodist Hospital, Houston, Texas.,Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
23
|
Abstract
De novo donor-specific antibody (DSA) formation is a major problem in transplantation, and associated with long-term graft decline and loss as well as sensitization, limiting future transplant options. Forming high-affinity, long-lived antibody responses involves a process called the germinal center (GC) reaction, and requires interaction between several cell types, including GC B cells, T follicular helper (Tfh) and T follicular regulatory (Tfr) cells. T follicular regulatory cells are an essential component of the GC reaction, limiting its size and reducing nonspecific or self-reactive responses.An imbalance between helper function and regulatory function can lead to excessive antibody production. High proportions of Tfh cells have been associated with DSA formation in transplantation; therefore, Tfr cells are likely to play an important role in limiting DSA production. Understanding the signals that govern Tfr cell development and the balance between helper and regulatory function within the GC is key to understanding how these cells might be manipulated to reduce the risk of DSA development.This review discusses the development and function of Tfr cells and their relevance to transplantation. In particular how current and future immunosuppressive strategies might allow us to skew the ratio between Tfr and Tfh cells to increase or decrease the risk of de novo DSA formation.
Collapse
|
24
|
Bitar M, Boldt A, Freitag MT, Gruhn B, Köhl U, Sack U. Evaluating STAT5 Phosphorylation as a Mean to Assess T Cell Proliferation. Front Immunol 2019; 10:722. [PMID: 31024554 PMCID: PMC6460883 DOI: 10.3389/fimmu.2019.00722] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/18/2019] [Indexed: 01/26/2023] Open
Abstract
Here we present a simple and sensitive flow cytometric-based assay to assess T cell proliferation. Given the critical role STAT5A phosphorylation in T cell proliferation, we decided to evaluate phosphorylation of STAT5A as an indicator of T cell proliferation. We determined pSTAT5A in T cell treated with either CD3/CD28 or PHA. After stimulation, T cells from adult healthy donors displayed a strong long-lasting phosphorylation of STAT5A, reaching a peak value after 24 h. The median fluorescence intensity (MFI) of pSTAT5A increased from 112 ± 17 to 512 ± 278 (CD3/CD28) (24 h) and to 413 ± 123 (PHA) (24 h), the IL-2 receptor-α (CD25) expression was greatly enhanced and after 72 h T cell proliferation amounted to 52.3 ± 10.3% (CD3/CD28) and to 48.4 ± 9.7% (PHA). Treatment with specific JAK3 and STAT5 inhibitors resulted in a complete blockage of phosphorylation of STAT5A, CD25 expression, and suppression of T cell proliferation. Compared with currently available methods, STAT5A phosphorylation is well-suited to predict T cell proliferation. Moreover, the method presented here is not very time consuming (several hours) and delivers functional information from which conclusions about T cell proliferation can be drawn.
Collapse
Affiliation(s)
- Michael Bitar
- Medical Faculty, Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Andreas Boldt
- Medical Faculty, Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Marie-Theres Freitag
- Medical Faculty, Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Bernd Gruhn
- Department of Pediatrics, Jena University Hospital, Jena, Germany
| | - Ulrike Köhl
- Medical Faculty, Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany.,Hannover Medical School, Institute of Cellular Therapeutics, Hannover, Germany.,Fraunhofer Institute for Immunology and Cell Therapy (IZI), Leipzig, Germany
| | - Ulrich Sack
- Medical Faculty, Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| |
Collapse
|
25
|
Ramirez GA, Coletto LA, Bozzolo EP, Citterio L, Delli Carpini S, Zagato L, Rovere-Querini P, Lanzani C, Manunta P, Manfredi AA, Sciorati C. The TRPC6 intronic polymorphism, associated with the risk of neurological disorders in systemic lupus erythematous, influences immune cell function. J Neuroimmunol 2018; 325:43-53. [PMID: 30384327 DOI: 10.1016/j.jneuroim.2018.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 10/02/2018] [Accepted: 10/21/2018] [Indexed: 12/12/2022]
Abstract
Patients with systemic lupus erythematosus (SLE) carrying a TT genotype for the rs7925662 single nucleotide polymorphism (SNP) in the transient receptor potential canonical channel 6 (TRPC6) gene are more likely to develop neuropsychiatric manifestations (NPSLE). We functionally characterised the effects of TRPC6 on peripheral blood mononuclear cells from 18 patients with SLE and 8 healthy controls with a known genotype. TRPC6 influenced calcium currents, apoptosis rates and cytokine secretion in a disease- and genotype-dependent manner. Cells from TT patients with NPSLE were more dependent on TRPC6 for the generation of calcium currents.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | | | - Enrica P Bozzolo
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy
| | - Lorena Citterio
- Unit of Nephrology, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy
| | - Simona Delli Carpini
- Unit of Nephrology, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy
| | - Laura Zagato
- Unit of Nephrology, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy
| | - Patrizia Rovere-Querini
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Disease, San Raffaele Hospital & Scientific Institute Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Chiara Lanzani
- Unit of Nephrology, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy
| | - Paolo Manunta
- Unit of Nephrology, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy
| | - Angelo A Manfredi
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS San Raffaele Hospital & Scientific Institute, Milan, Italy; Division of Immunology, Transplantation and Infectious Disease, San Raffaele Hospital & Scientific Institute Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Clara Sciorati
- Division of Immunology, Transplantation and Infectious Disease, San Raffaele Hospital & Scientific Institute Milan, Italy.
| |
Collapse
|
26
|
Ibáñez M, Carbonell-Caballero J, Such E, García-Alonso L, Liquori A, López-Pavía M, Llop M, Alonso C, Barragán E, Gómez-Seguí I, Neef A, Hervás D, Montesinos P, Sanz G, Sanz MA, Dopazo J, Cervera J. The modular network structure of the mutational landscape of Acute Myeloid Leukemia. PLoS One 2018; 13:e0202926. [PMID: 30303964 PMCID: PMC6179200 DOI: 10.1371/journal.pone.0202926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023] Open
Abstract
Acute myeloid leukemia (AML) is associated with the sequential accumulation of acquired genetic alterations. Although at diagnosis cytogenetic alterations are frequent in AML, roughly 50% of patients present an apparently normal karyotype (NK), leading to a highly heterogeneous prognosis. Due to this significant heterogeneity, it has been suggested that different molecular mechanisms may trigger the disease with diverse prognostic implications. We performed whole-exome sequencing (WES) of tumor-normal matched samples of de novo AML-NK patients lacking mutations in NPM1, CEBPA or FLT3-ITD to identify new gene mutations with potential prognostic and therapeutic relevance to patients with AML. Novel candidate-genes, together with others previously described, were targeted resequenced in an independent cohort of 100 de novo AML patients classified in the cytogenetic intermediate-risk (IR) category. A mean of 4.89 mutations per sample were detected in 73 genes, 35 of which were mutated in more than one patient. After a network enrichment analysis, we defined a single in silico model and established a set of seed-genes that may trigger leukemogenesis in patients with normal karyotype. The high heterogeneity of gene mutations observed in AML patients suggested that a specific alteration could not be as essential as the interaction of deregulated pathways.
Collapse
Affiliation(s)
- Mariam Ibáñez
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Departamento de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad CEU Cardenal Herrera, Valencia, Spain
| | - José Carbonell-Caballero
- ProCURE, Catalan Institute of Oncology, Bellvitge Institute for Biomedical Research (IDIBELL), L’Hospitalet del Llobregat, Barcelona, Spain
| | - Esperanza Such
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Luz García-Alonso
- European Molecular Biology Laboratory—European Bioinformatics Institute, Wellcome Genome Campus, Cambridge, United Kingdom
| | - Alessandro Liquori
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - María López-Pavía
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Marta Llop
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | - Carmen Alonso
- Hematology Service, Hospital Arnau de Villanoba, Valencia, Spain
| | - Eva Barragán
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Department of Medical Pathology, Hospital Universitario La Fe, Valencia, Spain
| | - Inés Gómez-Seguí
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Alexander Neef
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | - Pau Montesinos
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Guillermo Sanz
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Miguel Angel Sanz
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
| | - Joaquín Dopazo
- Functional Genomics Node, Spanish National Institute of Bioinformatics at CIPF, Valencia, Spain
- Bioinformatics of Rare Diseases (BIER), CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
- Clinical Bioinformatics Area, Fundación Progreso y Salud (FPS), CDCA, Hospital Virgen del Rocio, Sevilla, Spain
- * E-mail: (JC); (JD)
| | - José Cervera
- Hematology Service, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigacion Biomédica en Red de Cáncer (CIBERONC), Instituto Carlos III, Madrid, Spain
- Genetics Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- * E-mail: (JC); (JD)
| |
Collapse
|
27
|
Diener C, Hart M, Alansary D, Poth V, Walch-Rückheim B, Menegatti J, Grässer F, Fehlmann T, Rheinheimer S, Niemeyer BA, Lenhof HP, Keller A, Meese E. Modulation of intracellular calcium signaling by microRNA-34a-5p. Cell Death Dis 2018; 9:1008. [PMID: 30262862 PMCID: PMC6160487 DOI: 10.1038/s41419-018-1050-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 12/21/2022]
Abstract
Adjusting intracellular calcium signaling is an important feature in the regulation of immune cell function and survival. Here we show that miR-34a-5p, a small non-coding RNA that is deregulated in many common diseases, is a regulator of store-operated Ca2+ entry (SOCE) and calcineurin signaling. Upon miR-34a-5p overexpression, we observed both a decreased depletion of ER calcium content and a decreased Ca2+ influx through Ca2+ release-activated Ca2+ channels. Based on an in silico target prediction we identified multiple miR-34a-5p target genes within both pathways that are implicated in the balance between T-cell activation and apoptosis including ITPR2, CAMLG, STIM1, ORAI3, RCAN1, PPP3R1, and NFATC4. Functional analysis revealed a decrease in Ca2+ activated calcineurin pathway activity measured by a reduced IL-2 secretion due to miR-34a-5p overexpression. Impacting SOCE and/or downstream calcineurin/NFAT signaling by miR-34a-5p offers a possible future approach to manipulate immune cells for clinical interventions.
Collapse
Affiliation(s)
- Caroline Diener
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany.
| | - Martin Hart
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany
| | - Dalia Alansary
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Vanessa Poth
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Barbara Walch-Rückheim
- Institute of Virology and Center of Human and Molecular Biology, Saarland University, 66421, Homburg, Germany
| | - Jennifer Menegatti
- Institute of Virology and Center of Human and Molecular Biology, Medical School, Saarland University, 66421, Homburg, Germany
| | - Friedrich Grässer
- Institute of Virology and Center of Human and Molecular Biology, Medical School, Saarland University, 66421, Homburg, Germany
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | | | - Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine, School of Medicine, Saarland University, 66421, Homburg, Germany
| | - Hans-Peter Lenhof
- Center for Bioinformatics, Saarland Informatics Campus, Saarland University, 66123, Saarbrücken, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, 66421, Homburg, Germany
| |
Collapse
|
28
|
Patrussi L, Capitani N, Cattaneo F, Manganaro N, Gamberucci A, Frezzato F, Martini V, Visentin A, Pelicci PG, D'Elios MM, Trentin L, Semenzato G, Baldari CT. p66Shc deficiency enhances CXCR4 and CCR7 recycling in CLL B cells by facilitating their dephosphorylation-dependent release from β-arrestin at early endosomes. Oncogene 2018; 37:1534-1550. [PMID: 29326436 DOI: 10.1038/s41388-017-0066-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/06/2017] [Accepted: 11/13/2017] [Indexed: 12/28/2022]
Abstract
Neoplastic cell traffic abnormalities are central to the pathogenesis of chronic lymphocytic leukemia (CLL). Enhanced CXC chemokine receptor-4 (CXCR4) and chemokine receptor-7 (CCR7) recycling contributes to the elevated surface levels of these receptors on CLL cells. Here we have addressed the role of p66Shc, a member of the Shc family of protein adaptors the expression of which is defective in CLL cells, in CXCR4/CCR7 recycling. p66Shc reconstitution in CLL cells reduced CXCR4/CCR7 recycling, lowering their surface levels and attenuating B-cell chemotaxis, due to their accumulation in Rab5+ endosomes as serine-phosphoproteins bound to β-arrestin. This results from the ability of p66Shc to inhibit Ca2+ and PP2B-dependent CXCR4/CCR7 dephosphorylation and β-arrestin release. We also show that ibrutinib, a Btk inhibitor that promotes leukemic cell mobilization from lymphoid organs, reverses the CXCR4/CCR7 recycling abnormalities in CLL cells by increasing p66Shc expression. These results, identifying p66Shc as a regulator of CXCR4/CCR7 recycling in B cells, underscore the relevance of its deficiency to CLL pathogenesis and provide new clues to the mechanisms underlying the therapeutic effects of ibrutinib.
Collapse
Affiliation(s)
- Laura Patrussi
- Department of Life Sciences, University of Siena, Siena, Italy.
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy.,Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | | | - Noemi Manganaro
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Federica Frezzato
- Venetian Institute of Molecular Medicine, Padua, Italy.,Department of Medicine, Hematology and Clinical Immunology Branch, Padua University School of Medicine, Padua, Italy
| | - Veronica Martini
- Venetian Institute of Molecular Medicine, Padua, Italy.,Department of Medicine, Hematology and Clinical Immunology Branch, Padua University School of Medicine, Padua, Italy
| | - Andrea Visentin
- Venetian Institute of Molecular Medicine, Padua, Italy.,Department of Medicine, Hematology and Clinical Immunology Branch, Padua University School of Medicine, Padua, Italy
| | | | - Mario M D'Elios
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | - Livio Trentin
- Venetian Institute of Molecular Medicine, Padua, Italy.,Department of Medicine, Hematology and Clinical Immunology Branch, Padua University School of Medicine, Padua, Italy
| | - Gianpietro Semenzato
- Venetian Institute of Molecular Medicine, Padua, Italy.,Department of Medicine, Hematology and Clinical Immunology Branch, Padua University School of Medicine, Padua, Italy
| | | |
Collapse
|
29
|
High-Throughput Yeast-Based Reporter Assay to Identify Compounds with Anti-inflammatory Potential. Methods Mol Biol 2018; 1449:441-52. [PMID: 27613055 DOI: 10.1007/978-1-4939-3756-1_29] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
The association between altered proteostasis and inflammatory responses has been increasingly recognized, therefore the identification and characterization of novel compounds with anti-inflammatory potential will certainly have a great impact in the therapeutics of protein-misfolding diseases such as degenerative disorders. Although cell-based screens are powerful approaches to identify potential therapeutic compounds, establishing robust inflammation models amenable to high-throughput screening remains a challenge. To bridge this gap, we have exploited the use of yeasts as a platform to identify lead compounds with anti-inflammatory properties. The yeast cell model described here relies on the high-degree homology between mammalian and yeast Ca(2+)/calcineurin pathways converging into the activation of NFAT and Crz1 orthologous proteins, respectively. It consists of a recombinant yeast strain encoding the lacZ gene under the control of Crz1-recongition elements to facilitate the identification of compounds interfering with Crz1 activation through the easy monitoring of β-galactosidase activity. Here, we describe in detail a protocol optimized for high-throughput screening of compounds with potential anti-inflammatory activity as well as a protocol to validate the positive hits using an alternative β-galactosidase substrate.
Collapse
|
30
|
Kaminuma O, Kitamura N, Nishito Y, Nemoto S, Tatsumi H, Mori A, Hiroi T. Downregulation of NFAT3 Due to Lack of T-Box Transcription Factor TBX5 Is Crucial for Cytokine Expression in T Cells. THE JOURNAL OF IMMUNOLOGY 2017; 200:92-100. [PMID: 29180489 DOI: 10.4049/jimmunol.1602113] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 10/19/2017] [Indexed: 11/19/2022]
Abstract
The NFAT family transcription factors play crucial roles in immunological and other biological activities. NFAT3 is rarely expressed in T cells, and the mechanisms and significance of the specific NFAT3 downregulation in T cells have been unknown. In human CD4+ T cells, overexpression of NFAT1 and NFAT3 enhanced and suppressed IL-2 expression, respectively. NFAT3 downregulation in Jurkat cells using RNA interference technology augmented IL-2 expression, whereas a knockdown of NFAT1, NFAT2, and NFAT4 suppressed it. The promoter/enhancer activity of the NFAT-binding site in the IL-2 gene was upregulated and downregulated by NFAT1 and NFAT3, respectively. A study employing NFAT1/NFAT3 chimeric molecules revealed that the region in NFAT3 responsible for NFAT promoter activity inhibition was located within its N-terminal transactivation domain, Ca2+-regulatory domain, and DNA-binding domain. Downregulation of NFAT3 expression in T cells is mediated by lower chromatin accessibility and enhancer activity in its promoter in comparison with aortic smooth muscle cells expressing endogenous NFAT3. The binding sites of T-box transcription factor TBX5 and NK-2 transcription factor-related locus 5 Nkx2.5, which were expressed at higher levels in aortic smooth muscle cells than in T cells, were located within the -387 to +97 NFAT3 promoter region, exhibiting the maximum enhancer activity. Mutating the binding site of TBX5 but not Nkx2.5 diminished the NFAT3 promoter activity, whereas the overexpression of TBX5 enhanced it. Introduction of TBX5 into CD4+ T cells enhanced the expression of NFAT3 and suppressed that of IL-2. TBX5 deficiency-mediated downregulation of NFAT3 is crucial for the high cytokine-producing activity of T cells.
Collapse
Affiliation(s)
- Osamu Kaminuma
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan; .,Center for Life Science Research, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Noriko Kitamura
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| | - Yasumasa Nishito
- Center for Basic Technology Research, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| | - Soichi Nemoto
- Department of Obstetrics and Gynecology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan; and
| | - Hideki Tatsumi
- Department of Obstetrics and Gynecology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan; and
| | - Akio Mori
- Clinical Research Center for Allergy and Rheumatology, National Hospital Organization, Sagamihara National Hospital, Kanagawa 252-0392, Japan
| | - Takachika Hiroi
- Allergy and Immunology Project, The Tokyo Metropolitan Institute of Medical Science, Tokyo 113-8613, Japan
| |
Collapse
|
31
|
Brignall R, Cauchy P, Bevington SL, Gorman B, Pisco AO, Bagnall J, Boddington C, Rowe W, England H, Rich K, Schmidt L, Dyer NP, Travis MA, Ott S, Jackson DA, Cockerill PN, Paszek P. Integration of Kinase and Calcium Signaling at the Level of Chromatin Underlies Inducible Gene Activation in T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 199:2652-2667. [PMID: 28904128 PMCID: PMC5632840 DOI: 10.4049/jimmunol.1602033] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 08/21/2017] [Indexed: 01/20/2023]
Abstract
TCR signaling pathways cooperate to activate the inducible transcription factors NF-κB, NFAT, and AP-1. In this study, using the calcium ionophore ionomycin and/or PMA on Jurkat T cells, we show that the gene expression program associated with activation of TCR signaling is closely related to specific chromatin landscapes. We find that calcium and kinase signaling cooperate to induce chromatin remodeling at ∼2100 chromatin regions, which demonstrate enriched binding motifs for inducible factors and correlate with target gene expression. We found that these regions typically function as inducible enhancers. Many of these elements contain composite NFAT/AP-1 sites, which typically support cooperative binding, thus further reinforcing the need for cooperation between calcium and kinase signaling in the activation of genes in T cells. In contrast, treatment with PMA or ionomycin alone induces chromatin remodeling at far fewer regions (∼600 and ∼350, respectively), which mostly represent a subset of those induced by costimulation. This suggests that the integration of TCR signaling largely occurs at the level of chromatin, which we propose plays a crucial role in regulating T cell activation.
Collapse
Affiliation(s)
- Ruth Brignall
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Pierre Cauchy
- Institute of Biomedical Research, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Sarah L Bevington
- Institute of Biomedical Research, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Bethany Gorman
- Institute of Biomedical Research, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Angela O Pisco
- Centre for Stem Cells and Regenerative Medicine, King's College London, London SE1 9RT, United Kingdom
| | - James Bagnall
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Christopher Boddington
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - William Rowe
- Department of Chemistry, Loughborough University, Loughborough LE11 3TU, United Kingdom
| | - Hazel England
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Kevin Rich
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9PT, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom; and
| | - Lorraine Schmidt
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Nigel P Dyer
- Warwick Systems Biology Centre, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Mark A Travis
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
- Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester M13 9PT, United Kingdom
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester M13 9PT, United Kingdom; and
| | - Sascha Ott
- Warwick Systems Biology Centre, University of Warwick, Coventry CV4 7AL, United Kingdom
| | - Dean A Jackson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Peter N Cockerill
- Institute of Biomedical Research, College of Medicine and Dentistry, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| | - Pawel Paszek
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
32
|
Estrogen receptor α/HDAC/NFAT axis for delphinidin effects on proliferation and differentiation of T lymphocytes from patients with cardiovascular risks. Sci Rep 2017; 7:9378. [PMID: 28839227 PMCID: PMC5570903 DOI: 10.1038/s41598-017-09933-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 08/01/2017] [Indexed: 12/16/2022] Open
Abstract
Delphinidin, an anthocyanin present in red wine, has been reported to preserve the integrity of endothelium via an estrogen receptor alpha (ERα)-dependent mechanism. However, the effect of delphinidin on the immune response in obesity-related inflammation remains unknown. Given the important role of T lymphocytes in obesity-related inflammation, we investigated the effect of delphinidin on proliferation and differentiation of T lymphocytes from healthy subjects and metabolic syndrome patients. Delphinidin decreased the proliferation stimulated by different agents acting through different mechanisms. This effect of delphinidin was associated with its ability to inhibit Ca2+ signaling via reduced store-operated Ca2+ entry and release, and subsequent decrease of HDAC and NFAT activations. Delphinidin also inhibited ERK1/2 activation. Pharmacological inhibition of ER with fulvestrant, or deletion of ERα, prevented the effect of delphinidin. Further, delphinidin suppressed the differentiation of T cells toward Th1, Th17 and Treg without affecting Th2 subsets. Interestingly, delphinidin inhibited both proliferation and differentiation of T cells taken from patients with cardiovascular risks associated with metabolic syndrome. Together, we propose that delphinidin, by acting on ERα via multiple cellular targets, may represent a new approach against chronic inflammation associated with T lymphocyte activation, proliferation and differentiation, in patients with cardiovascular risk factors.
Collapse
|
33
|
Lawrance IC, Baird A, Lightower D, Radford-Smith G, Andrews JM, Connor S. Efficacy of Rectal Tacrolimus for Induction Therapy in Patients With Resistant Ulcerative Proctitis. Clin Gastroenterol Hepatol 2017; 15:1248-1255. [PMID: 28286194 DOI: 10.1016/j.cgh.2017.02.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/22/2017] [Accepted: 02/24/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Resistant ulcerative proctitis can be extremely difficult to manage. Topically administered tacrolimus, however, may be effective in difficult-to-treat proctitis. This was a randomized, double-blind, placebo-controlled induction trial of rectal tacrolimus in patients with active ulcerative colitis. METHODS Eleven patients received rectal tacrolimus (0.5 mg/mL), and 10 placebo, for 8 weeks. The primary endpoint was clinical response by using the Mayo Clinic score. RESULTS A planned interim analysis after 20 patients had completed the study demonstrated highly significant differences between the groups and the study was closed because of ethical considerations with patients already recruited allowed to complete the study. The primary endpoint was met in 8 of 11 patients receiving rectal tacrolimus and 1 of 10 patients receiving placebo (73% vs 10%; P = .004). Of the secondary endpoints, 5 patients with rectal tacrolimus achieved clinical remission compared with none receiving placebo (45% vs 0%; P = .015). Mucosal healing at Week 8 was achieved in 8 patients receiving rectal tacrolimus compared with 1 (73% vs 10%) receiving placebo (P = .004). The Inflammatory Bowel Disease Questionnaire increased ≥16 points over baseline in 5 of the tacrolimus and 2 (45% vs 20%) of the placebo patients (P = .36). Finally, the average partial Mayo score was numerically lower in the tacrolimus-treated group compared with placebo at Week 2 (4.3 ± 0.74 vs 5.8 ± 0.64; P = .15) and Week 4 (3.7 ± 0.96 vs 5.8 ± 0.6; P = .08) but was significantly lower at Week 8 (3.3 ± 1.2 vs 6.7 ± 0.62; P = .01). There were no safety issues identified with rectal tacrolimus use. CONCLUSIONS Rectal tacrolimus was more effective than placebo for induction of a clinical response, clinical remission, and mucosal healing in resistant ulcerative proctitis (Clinicaltrials.gov registration: NCT01418131).
Collapse
Affiliation(s)
- Ian C Lawrance
- Harry Perkins Institute of Medical Research, School of Medicine and Pharmacology, University of Western Australia, Murdoch, WA, Australia; Centre for Inflammatory Bowel Diseases, Saint John of God Hospital, Subiaco, WA, Australia.
| | - Angela Baird
- Harry Perkins Institute of Medical Research, School of Medicine and Pharmacology, University of Western Australia, Murdoch, WA, Australia
| | - Daniel Lightower
- Centre for Inflammatory Bowel Diseases, Saint John of God Hospital, Subiaco, WA, Australia
| | - Graham Radford-Smith
- IBD Research Group, QIMR Berghofer Medical Research Institute, University of Queensland School of Medicine, Brisbane, Queensland, Australia; Department of Gastroenterology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Jane M Andrews
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, SA, Australia; University of Adelaide, School of Medicine, SA, Australia
| | - Susan Connor
- Department of Gastroenterology and Hepatology, Liverpool Hospital, Sydney, NSW, Australia; South Western Sydney Clinical School, University of NSW Medicine, Sydney, NSW, Australia; Ingham Institute for Applied Medical Research, South Western Sydney Clinical School, UNSW Australia, Sydney, NSW, Australia
| |
Collapse
|
34
|
Regulation of CacyBP/SIP expression by NFAT1 transcription factor. Immunobiology 2017; 222:872-877. [PMID: 28526484 DOI: 10.1016/j.imbio.2017.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 05/09/2017] [Indexed: 01/11/2023]
Abstract
In this work we have shown that NFAT1 transcription factor is involved in the regulation of CacyBP/SIP expression. We have demonstrated, by applying Western blot, RT-PCR and luciferase assay that the level of CacyBP/SIP increases upon NFAT1 overexpression. Moreover, inhibition or stimulation of NFAT transcriptional activity exerts a corresponding effect on the expression of CacyBP/SIP gene. Furthermore, EMSA and chromatin immunoprecipitation (ChIP) assay have shown that NFAT1 binds to its specific binding sites within the CacyBP/SIP gene. In conclusion, our data have shown for the first time the regulation of CacyBP/SIP gene expression by NFAT1. Since NFAT transcription factors are involved in processes related to immune response, these results indicate potential involvement of CacyBP/SIP in the immune system.
Collapse
|
35
|
Identification of Spongionella compounds as cyclosporine A mimics. Pharmacol Res 2016; 107:407-414. [DOI: 10.1016/j.phrs.2016.03.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 03/03/2016] [Accepted: 03/03/2016] [Indexed: 01/29/2023]
|
36
|
Lijo J, Vijay N, Dechamma HJ, Reddy GR. Expression of biologically active bovine interleukin 7 and evaluating the activity in vitro. Vet World 2016; 9:160-5. [PMID: 27051202 PMCID: PMC4819366 DOI: 10.14202/vetworld.2016.160-165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/26/2015] [Accepted: 12/31/2015] [Indexed: 01/08/2023] Open
Abstract
Aim: Interleukin 7 (IL-7) is a ϒc family cytokine involved in the homeostatic proliferation and maintenance of immune cells. In the present study, we report the expression of bovine IL-7 (bIL-7) in Escherichia coli and evaluated for its biological activity. Materials and Methods: The sequence coding for bIL-7 (mature protein) was amplified from primary bovine kidney cell culture and cloned into pET28-a vector and expressed in E. coli (BL 21 DE3). The expressed protein was purified by nickel-nitrilotriacetatechromatography, and the reactivity of the protein was confirmed by western blotting using monoclonal antibodies raised against human IL-7. The biological activity of expressed bIL-7 was evaluated by analyzing its effect on the expression of anuclear factor for activated T-cells c1 (NFATc1), B-cell lymphoma 2 (Bcl2), suppressor of cytokine signaling 3 (SOCS3) molecules in bovine peripheral blood mononuclear cells (PBMCs) by quantitative polymerase chain reaction. Ability of the expressed protein was also analyzed by its effect on phosphorylating signal transducer and activator 3 (STAT3) molecule by immunostaining in human embryonic kidney cells 293 (HEK293) cells. Results: The bIL-7 was able to induce the expression of Bcl2 and NFATc1expression in bovine PBMCs by 7 and 5-folds, respectively, whereas a 2-fold decrease was observed in the case of SOCS3 expression. Immunostaining studies in HEK293 cells using antihuman phospho-STAT3 showed activation and nuclear translocation of STAT3 molecule on bIL-7 treatment. Conclusion: bIL-7 gene was successfully amplified, cloned, and expressed in a prokaryotic expression system. The biological activity study showed that the E. coli expressed bIL-7 protein is biologically active. Considering the role of IL-7 in T-cell homeostasis and memory cell generation, this molecule can be used for enhancing the vaccine response and that has to be proved by further experiments.
Collapse
Affiliation(s)
- J Lijo
- FMD Research Laboratory, Indian Veterinary Research Institute, Hebbal, Bengaluru, Karnataka, India
| | - N Vijay
- FMD Research Laboratory, Indian Veterinary Research Institute, Hebbal, Bengaluru, Karnataka, India
| | - H J Dechamma
- FMD Research Laboratory, Indian Veterinary Research Institute, Hebbal, Bengaluru, Karnataka, India
| | - G R Reddy
- FMD Research Laboratory, Indian Veterinary Research Institute, Hebbal, Bengaluru, Karnataka, India
| |
Collapse
|
37
|
Microarray expression profile analysis of long noncoding RNAs in premature brain injury: A novel point of view. Neuroscience 2016; 319:123-33. [PMID: 26812036 DOI: 10.1016/j.neuroscience.2016.01.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 01/14/2016] [Accepted: 01/14/2016] [Indexed: 02/07/2023]
Abstract
Long noncoding RNAs (lncRNAs) are abundant in the central nervous system and have a key role in brain function as well as many neurological disorders. However, the regulatory function of lncRNAs in the premature brain has not been well studied. This study described the expression profile of lncRNAs in premature mice using microarray technology. 1999 differentially expressed lncRNAs and 955 differentially expressed mRNAs were identified. Gene Ontology (GO) and pathway analysis showed that these lncRNAs were involved in multiple biological processes, including the nervous system development and inflammatory response. Additionally, the lncRNA-mRNA-network and TF-gene-lncRNA-network were constructed to identify core regulatory lncRNAs and transcription factors. The sex-determining region of Y chromosome (SRY) gene may be a key transcription factor that regulates premature brain development and injury. This study for the first time represents an expression profile of differentially expressed lncRNAs in the premature brain and may provide a novel point of view into the mechanisms of premature brain injury.
Collapse
|
38
|
Ugajin T, Nishida K, Yamasaki S, Suzuki J, Mita M, Kubo M, Yokozeki H, Hirano T. Zinc-binding metallothioneins are key modulators of IL-4 production by basophils. Mol Immunol 2015; 66:180-8. [DOI: 10.1016/j.molimm.2015.03.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/21/2022]
|
39
|
Dietz L, Frommer F, Vogel AL, Vaeth M, Serfling E, Waisman A, Buttmann M, Berberich-Siebelt F. NFAT1 deficit and NFAT2 deficit attenuate EAE via different mechanisms. Eur J Immunol 2015; 45:1377-89. [DOI: 10.1002/eji.201444638] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Revised: 09/30/2014] [Accepted: 01/27/2015] [Indexed: 11/09/2022]
Affiliation(s)
- Lena Dietz
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Friederike Frommer
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
- Institute for Molecular Medicine; University Medical Center of the Johannes Gutenberg; University of Mainz; Mainz Germany
| | - Anna-Lena Vogel
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Martin Vaeth
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Edgar Serfling
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
| | - Ari Waisman
- Institute for Molecular Medicine; University Medical Center of the Johannes Gutenberg; University of Mainz; Mainz Germany
| | - Mathias Buttmann
- Department of Neurology; University of Wuerzburg; Wuerzburg Germany
| | - Friederike Berberich-Siebelt
- Institute of Pathology; University of Wuerzburg; Wuerzburg Germany
- Comprehensive Cancer Center Mainfranken; University of Wuerzburg; Wuerzburg Germany
| |
Collapse
|
40
|
Selective NFAT targeting in T cells ameliorates GvHD while maintaining antitumor activity. Proc Natl Acad Sci U S A 2015; 112:1125-30. [PMID: 25583478 DOI: 10.1073/pnas.1409290112] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Graft-versus-host disease (GvHD) is a life-threatening immunological complication after allogenic hematopoietic stem cell transplantation (allo-HCT). The intrinsic graft-versus-leukemia (GvL) effect, however, is the desirable curative benefit. Patients with acute GvHD are treated with cyclosporine A (CsA) or tacrolimus (FK506), which not only often causes severe adverse effects, but also interferes with the anticipated GvL. Both drugs inhibit calcineurin, thus at first suppressing activation of the nuclear factor of activated T cells (NFAT). Therefore, we explored the specific contribution of individual NFAT factors in donor T cells in animal models of GvHD and GvL. Ablation of NFAT1, NFAT2, or a combination of both resulted in ameliorated GvHD, due to reduced proliferation, target tissue homing, and impaired effector function of allogenic donor T cells. In contrast, the frequency of Foxp3(+) regulatory T (Treg) cells was increased and NFAT-deficient Tregs were fully protective in GvHD. CD8(+) T-cell recall response and, importantly, the beneficial antitumor activity were largely preserved in NFAT-deficient effector T cells. Thus, specific inhibition of NFAT opens an avenue for an advanced therapy of GvHD maintaining protective GvL.
Collapse
|
41
|
Muhammad K, Alrefai H, Marienfeld R, Pham DAT, Murti K, Patra AK, Avots A, Bukur V, Sahin U, Kondo E, Klein-Hessling S, Serfling E. NF-κB factors control the induction of NFATc1 in B lymphocytes. Eur J Immunol 2014; 44:3392-402. [PMID: 25179582 DOI: 10.1002/eji.201444756] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/16/2014] [Accepted: 08/29/2014] [Indexed: 01/06/2023]
Abstract
In peripheral lymphocytes, the transcription factors (TFs) NF-κB, NFAT, and AP-1 are the prime targets of signals that emerge from immune receptors. Upon activation, these TFs induce gene networks that orchestrate the growth, expansion, and effector function of peripheral lymphocytes. NFAT and NF-κB factors share several properties, such as a similar mode of induction and architecture in their DNA-binding domain, and there is a subgroup of κB-like DNA promoter motifs that are bound by both types of TFs. However, unlike NFAT and AP-1 factors that interact and collaborate in binding to DNA, NFAT, and NF-κB seem neither to interact nor to collaborate. We show here that NF-κB1/p50 and c-Rel, the most prominent NF-κB proteins in BCR-induced splenic B cells, control the induction of NFATc1/αA, a prominent short NFATc1 isoform. In part, this is mediated through two composite κB/NFAT-binding sites in the inducible Nfatc1 P1 promoter that directs the induction of NFATc1/αA by BCR signals. In concert with coreceptor signals that induce NF-κB factors, BCR signaling induces a persistent generation of NFATc1/αA. These data suggest a tight connection between NFATc1 and NF-κB induction in B lymphocytes contributing to the effector function of peripheral B cells.
Collapse
Affiliation(s)
- Khalid Muhammad
- Department of Molecular Pathology, Institute of Pathology and Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Inhibition of interleukin-2 gene expression by human herpesvirus 6B U54 tegument protein. J Virol 2014; 88:12452-63. [PMID: 25122797 DOI: 10.1128/jvi.02030-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human herpesvirus 6B (HHV-6B) is a ubiquitous pathogen causing lifelong infections in approximately 95% of humans worldwide. To persist within its host, HHV-6B has developed several immune evasion mechanisms, such as latency, during which minimal proteins are expressed, and the ability to disturb innate and adaptive immune responses. The primary cellular targets of HHV-6B are CD4(+) T cells. Previous studies by Flamand et al. (L. Flamand, J. Gosselin, I. Stefanescu, D. Ablashi, and J. Menezes, Blood 85:1263-1271, 1995) reported on the capacity of HHV-6A as well as UV-irradiated HHV-6A to inhibit interleukin-2 (IL-2) synthesis in CD4(+) lymphocytes, suggesting that viral structural components could be responsible for this effect. In the present study, we identified the HHV-6B U54 tegument protein (U54) as being capable of inhibiting IL-2 expression. U54 binds the calcineurin (CaN) phosphatase enzyme, causing improper dephosphorylation and nuclear translocation of NFAT (nuclear factor of activated T cells) proteins, resulting in suboptimal IL-2 gene transcription. The U54 GISIT motif (amino acids 293 to 297), analogous to the NFAT PXIXIT motif, contributed to the inhibition of NFAT activation. IMPORTANCE Human herpesvirus 6A (HHV-6A) and HHV-6B are associated with an increasing number of pathologies. These viruses have developed strategies to avoid the immune response allowing them to persist in the host. Several studies have illustrated mechanisms by which HHV-6A and HHV-6B are able to disrupt host defenses (reviewed in L. Dagna, J. C. Pritchett, and P. Lusso, Future Virol. 8:273-287, 2013, doi:10.2217/fvl.13.7). Previous work informed us that HHV-6A is able to suppress synthesis of interleukin-2 (IL-2), a key immune growth factor essential for adequate T lymphocyte proliferation and expansion. We obtained evidence that HHV-6B also inhibits IL-2 gene expression and identified the mechanisms by which it does so. Our work led us to the identification of U54, a virion-associated tegument protein, as being responsible for suppression of IL-2. Consequently, we have identified HHV-6B U54 protein as playing a role in immune evasion. These results further contribute to our understanding of HHV-6 interactions with its human host and the efforts deployed to ensure its long-term persistence.
Collapse
|
43
|
Inhibition of breast cancer cell proliferation through disturbance of the calcineurin/NFAT pathway by human herpesvirus 6B U54 tegument protein. J Virol 2014; 88:12910-4. [PMID: 25122795 DOI: 10.1128/jvi.02107-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Nuclear factor of activated T cell (NFAT) proteins are key regulators involved in multiple physiological mechanisms, such as immune response and cell growth. The capacity of selective calcineurin/NFAT inhibitors to decrease NFAT-dependent cancer cell progression, particularly in breast cancer, has already been demonstrated. In this study, we report a role for the human herpesvirus 6B (HHV-6B) U54 tegument protein in inhibiting MCF-7 breast cancer cell proliferation by inhibiting NFAT activation.
Collapse
|
44
|
Hollerer I, Grund K, Hentze MW, Kulozik AE. mRNA 3'end processing: A tale of the tail reaches the clinic. EMBO Mol Med 2014; 6:16-26. [PMID: 24408965 PMCID: PMC3936486 DOI: 10.1002/emmm.201303300] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Recent advances reveal mRNA 3′end processing as a highly regulated process that fine-tunes posttranscriptional gene expression. This process can affect the site and/or the efficiency of 3′end processing, controlling the quality and the quantity of substrate mRNAs. The regulation of 3′end processing plays a central role in fundamental physiology such as blood coagulation and innate immunity. In addition, errors in mRNA 3′end processing have been associated with a broad spectrum of human diseases, including cancer. We summarize and discuss the paradigmatic shift in the understanding of 3′end processing as a mechanism of posttranscriptional gene regulation that has reached clinical medicine.
Collapse
Affiliation(s)
- Ina Hollerer
- Department of Pediatric Oncology, Hematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | | | | | | |
Collapse
|
45
|
Gal M, Li S, Luna RE, Takeuchi K, Wagner G. The LxVP and PxIxIT NFAT motifs bind jointly to overlapping epitopes on calcineurin's catalytic domain distant to the regulatory domain. Structure 2014; 22:1016-27. [PMID: 24954618 DOI: 10.1016/j.str.2014.05.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Revised: 04/22/2014] [Accepted: 05/04/2014] [Indexed: 11/28/2022]
Abstract
The serine/threonine phosphatase calcineurin (Cn) targets the nuclear factors of activated T cells (NFATs) that activate cytokine genes. Calcium influx activates Cn to dephosphorylate multiple serine residues within the ∼200 residue NFAT regulatory domain, which triggers joint nuclear translocation of NFAT and Cn. The dephosphorylation process relies on the interaction between Cn and the conserved motifs PxIxIT and LxVP, which are located N- and C-terminal to the phosphorylation sites in NFAT's regulatory domain. Here, we show that an NFATc1-derived 15-residue peptide segment containing the conserved LxVP motif binds to an epitope on Cn's catalytic domain (CnA), which overlaps with the previously established PxIxIT binding site on CnA and is distant to the regulatory domain (CnB). Both NFAT motifs partially compete for binding but do not fully displace each other on the CnA epitope, revealing that both segments bind simultaneously to the same epitope on the catalytic domain.
Collapse
Affiliation(s)
- Maayan Gal
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Shuai Li
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Rafael E Luna
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Koh Takeuchi
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA
| | - Gerhard Wagner
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
46
|
Shyu YC, Lee TL, Chen X, Hsu PH, Wen SC, Liaw YW, Lu CH, Hsu PY, Lu MJ, Hwang J, Tsai MD, Hwang MJ, Chen JR, Shen CKJ. Tight regulation of a timed nuclear import wave of EKLF by PKCθ and FOE during Pro-E to Baso-E transition. Dev Cell 2014; 28:409-22. [PMID: 24576425 DOI: 10.1016/j.devcel.2014.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2013] [Revised: 11/24/2013] [Accepted: 01/13/2014] [Indexed: 11/28/2022]
Abstract
Erythropoiesis is a highly regulated process during which BFU-E are differentiated into RBCs through CFU-E, Pro-E, PolyCh-E, OrthoCh-E, and reticulocyte stages. Uniquely, most erythroid-specific genes are activated during the Pro-E to Baso-E transition. We show that a wave of nuclear import of the erythroid-specific transcription factor EKLF occurs during the Pro-E to Baso-E transition. We further demonstrate that this wave results from a series of finely tuned events, including timed activation of PKCθ, phosphorylation of EKLF at S68 by P-PKCθ(S676), and sumoylation of EKLF at K74. The latter EKLF modifications modulate its interactions with a cytoplasmic ankyrin-repeat-protein FOE and importinβ1, respectively. The role of FOE in the control of EKLF nuclear import is further supported by analysis of the subcellular distribution patterns of EKLF in FOE-knockout mice. This study reveals the regulatory mechanisms of the nuclear import of EKLF, which may also be utilized in the nuclear import of other factors.
Collapse
Affiliation(s)
- Yu-Chiau Shyu
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Beitou, Taipei 112, Taiwan, ROC; Department of Education and Research, Taipei City Hospital, Da'an, Taipei 103, Taiwan, ROC; Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC.
| | - Tung-Liang Lee
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - Xin Chen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - Pang-Hung Hsu
- The Genomics Research Center, Academia Sinica, Nankang, Taipei 115, Taiwan, ROC
| | - Shau-Ching Wen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - Yi-Wei Liaw
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - Chi-Huan Lu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - Po-Yen Hsu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - Mu-Jie Lu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - JauLang Hwang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC
| | - Ming-Daw Tsai
- Institute of Biological Chemistry, Academia Sinica, Nankang, Taipei 115, Taiwan, ROC
| | - Ming-Jing Hwang
- Institute of Biomedical Sciences, Academia Sinica, Nankang, Taipei 115, Taiwan, ROC
| | - Jim-Ray Chen
- Department of Pathology, Keelung Chang Gung Memorial Hospital, Anle, Keelung 204, Taiwan, ROC; College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 259, Taiwan, ROC
| | - Che-Kun James Shen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei, Taiwan 115, ROC.
| |
Collapse
|
47
|
Vaeth M, Müller G, Stauss D, Dietz L, Klein-Hessling S, Serfling E, Lipp M, Berberich I, Berberich-Siebelt F. Follicular regulatory T cells control humoral autoimmunity via NFAT2-regulated CXCR5 expression. ACTA ACUST UNITED AC 2014; 211:545-61. [PMID: 24590764 PMCID: PMC3949566 DOI: 10.1084/jem.20130604] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
T cell–specific NFAT2 deletion results in reduced CXCR5+ follicular regulatory T cells, leading to uncontrolled germinal center responses and humoral autoimmunity. Maturation of high-affinity B lymphocytes is precisely controlled during the germinal center reaction. This is dependent on CD4+CXCR5+ follicular helper T cells (TFH) and inhibited by CD4+CXCR5+Foxp3+ follicular regulatory T cells (TFR). Because NFAT2 was found to be highly expressed and activated in follicular T cells, we addressed its function herein. Unexpectedly, ablation of NFAT2 in T cells caused an augmented GC reaction upon immunization. Consistently, however, TFR cells were clearly reduced in the follicular T cell population due to impaired homing to B cell follicles. This was TFR-intrinsic because only in these cells NFAT2 was essential to up-regulate CXCR5. The physiological relevance for humoral (auto-)immunity was corroborated by exacerbated lupuslike disease in the presence of NFAT2-deficient TFR cells.
Collapse
Affiliation(s)
- Martin Vaeth
- Department of Molecular Pathology, Institute of Pathology and 4 Comprehensive Cancer Center Mainfranken, Julius-Maximilians-University of Wuerzburg, 97080 Wuerzburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Yan HQ, Shin SS, Ma X, Li Y, Dixon CE. Differential effect of traumatic brain injury on the nuclear factor of activated T Cells C3 and C4 isoforms in the rat hippocampus. Brain Res 2013; 1548:63-72. [PMID: 24389074 DOI: 10.1016/j.brainres.2013.12.028] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 12/20/2013] [Accepted: 12/23/2013] [Indexed: 10/25/2022]
Abstract
The interaction between the phosphatase calcineurin and transcription factor nuclear factor of activated T cells (NFAT) plays an important role numerous signaling and the regulatory events. Although NFAT is mostly known for its transcription function in the immune system, NFAT also has essential functions even in the central nervous system (CNS). The effects of traumatic brain injury (TBI) on NFAT are currently unknown. To determine if there is an alteration in NFAT after TBI, we examined NFATc3 and c4 levels at 6 h, 1 day, 1 week, 2 weeks and 4 weeks post injury. Rats were anesthetized and surgically prepared for controlled cortical impact (CCI) injury or sham surgery. Semi-quantitative measurements of NFATc3 and c4 in the hippocampal homogenates from injured and sham rats sacrificed at the appropriate time after injury were assessed using Western blot analysis. After TBI insult, in the hippocampus ipsilateral to the injury, NFATc3 expression levels were decreased both in the cytoplasmic and nuclear fractions. However, NFATc4 expression levels were increased in the cytoplasmic fraction but decreased in the nuclear fraction. Double labeling (with NeuN and GFAP) immunohistochemistry revealed that NFATc3 was expressed in subset of astrocytes and NFATc4 was expressed primarily in neurons. These differential responses in NFATc3 and c4 expression after TBI insult may indicate long-term changes in hippocampal excitability and may contribute to behavioral deficits. Further study is warranted to illustrate the role of NFATc3 and c4 in the setting of TBI.
Collapse
Affiliation(s)
- Hong Q Yan
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Samuel S Shin
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Xiecheng Ma
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - Youming Li
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260
| | - C Edward Dixon
- Department of Neurological Surgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania 15260.,Veterans Affairs Pittsburgh Healthcare System Pittsburgh, PA 15240
| |
Collapse
|
49
|
Li W, Kong LB, Li JT, Guo ZY, Xue Q, Yang T, Meng YL, Jin BQ, Wen WH, Yang AG. MiR-568 inhibits the activation and function of CD4⁺ T cells and Treg cells by targeting NFAT5. Int Immunol 2013; 26:269-81. [PMID: 24355664 DOI: 10.1093/intimm/dxt065] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
CD4(+) T cells play critical roles in orchestrating adaptive immune responses. Their activation and proliferation are critical steps that occur before they execute their biological functions. Despite the important role of this process, the underlying molecular events are not fully understood. MicroRNAs (miRNAs) have been shown to play important roles in lymphocyte development and function. However, the miRNAs that regulate T-cell differentiation, activation and proliferation are still largely unknown. In our previous study, using a miRNA array, we found that several miRNAs (including miR-202, 33b, 181c, 568 and 576) are differentially expressed between resting and activated CD4(+) T cells. In this study, we focused on the function of miR-568 during CD4(+) T-cell activation. We showed that the expression level of miR-568 decreased during the activation of T cells, including Jurkat cells and human peripheral blood CD4(+) T cells. When Jurkat or human peripheral blood CD4(+) T cells were transfected with miR-568 mimics, cell activation was significantly inhibited, as shown by the inhibited expression of activation markers such as CD25, CD69 and CD154; decreased IL-2 production; and inhibited cell proliferation. Using software predictions and confirmatory experiments, we demonstrated that nuclear factor of activated T cells 5 (NFAT5) is a target of miR-568. Treg cells are an important CD4(+) T-cell subpopulation, so we also evaluated the function of miR-568 in Treg-cell activation and differentiation. We showed that the miR-568 level decreased, while the NFAT5 protein level increased during CD4(+)CD25(+) Treg-cell activation, and the transfection of miR-568 mimics inhibited the NFAT5 expression, inhibited the production of both TGF-β and IL-10 and also inhibited the proliferation of Treg cells. Our further study showed that over-expression of miR-568 can inhibit Treg-cell differentiation and can inhibit the suppressive effect of these cells on effector cells. In addition, inhibition of NFAT5 by siRNA-mediated knockdown can inhibit the activation and differentiation of Treg cells. These findings reveal that miR-568 can inhibit the activation and function of both CD4(+) T cells and Treg cells by targeting NFAT5. Since miR-568 plays an important role in both CD4(+) T cells and Treg cells, these findings may provide leads for the development of novel treatments for human inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Wei Li
- State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an 710032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kim HH, Park SB, Lee S, Kwon TK, Shin TY, Park PH, Lee SH, Kim SH. Inhibitory effect of putranjivain A on allergic inflammation through suppression of mast cell activation. Toxicol Appl Pharmacol 2013; 274:455-61. [PMID: 24361550 DOI: 10.1016/j.taap.2013.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 12/09/2013] [Accepted: 12/10/2013] [Indexed: 12/19/2022]
Abstract
A great number of people are suffering from allergic inflammatory disease such as asthma, atopic dermatitis, and sinusitis. Therefore discovery of drugs for the treatment of these diseases is an important subject in human health. Putranjivain A (PJA), member of ellagitannin, is known to possess beneficial effects including anti-cancer and anti-viral activities. The aim of the present study was to elucidate whether PJA modulates the allergic inflammatory reaction and to study its possible mechanisms of action using mast cell-based in vitro and in vivo models. The study was performed in anaphylaxis mouse model and cultured mast cells. PJA inhibited the expression of pro-inflammatory cytokines in immunoglobulin E-stimulated mast cells. PJA reduced this expression by inhibiting nuclear factor (NF)-κB and nuclear factor of activated T cell. The oral administration of PJA reduced systemic and cutaneous anaphylaxis, the release of serum histamine, and the expression of the histamine H1 receptor. In addition, PJA attenuated the activation of mast cells. PJA inhibited the release of histamine from various types of mast cells by the suppression of intracellular calcium. The inhibitory activity of PJA on the allergic reaction was similar to that of disodium cromoglycate, a known anti-allergic drug. These results suggest that PJA can facilitate the prevention or treatment of allergic inflammatory diseases mediated by mast cells.
Collapse
Affiliation(s)
- Hui-Hun Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Seung-Bin Park
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Soyoung Lee
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 704-701, Republic of Korea
| | - Tae-Yong Shin
- College of Pharmacy, Woosuk University, Jeonju 565-701, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Youngnam University, Kyungsan 712-749, Republic of Korea
| | - Seung-Ho Lee
- College of Pharmacy, Youngnam University, Kyungsan 712-749, Republic of Korea
| | - Sang-Hyun Kim
- CMRI, Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea.
| |
Collapse
|