1
|
Mackiewicz J, Tomczak J, Lisek M, Sakowicz A, Guo F, Boczek T. NFATc4 Knockout Promotes Neuroprotection and Retinal Ganglion Cell Regeneration After Optic Nerve Injury. Mol Neurobiol 2024; 61:9383-9401. [PMID: 38639863 PMCID: PMC11496353 DOI: 10.1007/s12035-024-04129-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Retinal ganglion cells (RGCs), neurons transmitting visual information via the optic nerve, fail to regenerate their axons after injury. The progressive loss of RGC function underlies the pathophysiology of glaucoma and other optic neuropathies, often leading to irreversible blindness. Therefore, there is an urgent need to identify the regulators of RGC survival and the regenerative program. In this study, we investigated the role of the family of transcription factors known as nuclear factor of activated T cells (NFAT), which are expressed in the retina; however, their role in RGC survival after injury is unknown. Using the optic nerve crush (ONC) model, widely employed to study optic neuropathies and central nervous system axon injury, we found that NFATc4 is specifically but transiently up-regulated in response to mechanical injury. In the injured retina, NFATc4 immunolocalized primarily to the ganglionic cell layer. Utilizing NFATc4-/- and NFATc3-/- mice, we demonstrated that NFATc4, but not NFATc3, knockout increased RGC survival, improved retina function, and delayed axonal degeneration. Microarray screening data, along with decreased immunostaining of cleaved caspase-3, revealed that NFATc4 knockout was protective against ONC-induced degeneration by suppressing pro-apoptotic signaling. Finally, we used lentiviral-mediated NFATc4 delivery to the retina of NFATc4-/- mice and reversed the pro-survival effect of NFATc4 knockout, conclusively linking the enhanced survival of injured RGCs to NFATc4-dependent mechanisms. In summary, this study is the first to demonstrate that NFATc4 knockout may confer transient RGC neuroprotection and decelerate axonal degeneration after injury, providing a potent therapeutic strategy for optic neuropathies.
Collapse
Affiliation(s)
- Joanna Mackiewicz
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Julia Tomczak
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Malwina Lisek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland
| | - Agata Sakowicz
- Department of Medical Biotechnology, Medical University of Lodz, Lodz, Poland
| | - Feng Guo
- Department of Pharmaceutical Toxicology, China Medical University, Shenyang, China.
| | - Tomasz Boczek
- Department of Molecular Neurochemistry, Medical University of Lodz, Lodz, Poland.
| |
Collapse
|
2
|
Jain A, Jung HJ, Aubee J, O’Neil JN, Muhammad LA, Khan S, Thompson K, Fluitt MB, Lee DL, Klinge CM, Khundmiri SJ. Role of NHERF1 in MicroRNA Landscape Changes in Aging Mouse Kidneys. Biomolecules 2024; 14:1048. [PMID: 39334814 PMCID: PMC11430241 DOI: 10.3390/biom14091048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
MicroRNAs (miRNAs) play important roles in the regulation of cellular function and fate via post-transcriptional regulation of gene expression. Although several miRNAs are associated with physiological processes and kidney diseases, not much is known about changes in miRNAs in aging kidneys. We previously demonstrated that sodium hydrogen exchanger 1 (NHERF1) expression regulates cellular responses to cisplatin, age-dependent salt-sensitive hypertension, and sodium-phosphate cotransporter trafficking. However, the mechanisms driving these regulatory effects of NHERF1 on cellular processes are unknown. Here, we hypothesize that dysregulation of miRNA-mediated gene regulatory networks that induce fibrosis and cytokines may depend on NHERF1 expression. To address this hypothesis, we compared miRNA expression in kidneys from both male and female old (12-18-month-old) and young (4-7-month-old) wild-type (WT) and NHERF1 knockout (NHERF1-/-) mice. Our results identified that miRNAs significantly decreased in NHERF1-/- mice included miR-669m, miR-590-3p, miR-153, miR-673-3p, and miR-127. Only miR-702 significantly decreased in aged WT mice, while miR-678 decreased in both WT and NHERF1-/- old versus young mice. miR-153 was shown to downregulate transcription factors NFATc2 and NFATc3 which regulate the transcription of several cytokines. Immunohistochemistry and western blotting revealed a significant increase in nuclear NFATc2 and NFATc3 in old NHERF1-/- mice compared to old WT mice. Our data further show that expression of the cytokines IL-1β, IL-6, IL-17A, MCP1, and TNF-α significantly increased in the old NHERF1-/- mice compared to the WT mice. We conclude that loss of NHERF1 expression induces cytokine expression in the kidney through interactive regulation between miR-153 and NFATc2/NFATc3 expression.
Collapse
Affiliation(s)
- Anish Jain
- Department of Physiology, Howard University College of Medicine, Washington, DC 20059, USA; (A.J.); (J.N.O.); (L.A.M.); (S.K.); (D.L.L.)
| | - Hyun Jun Jung
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Joseph Aubee
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (J.A.); (K.T.)
| | - Jahn N. O’Neil
- Department of Physiology, Howard University College of Medicine, Washington, DC 20059, USA; (A.J.); (J.N.O.); (L.A.M.); (S.K.); (D.L.L.)
| | - Laila A. Muhammad
- Department of Physiology, Howard University College of Medicine, Washington, DC 20059, USA; (A.J.); (J.N.O.); (L.A.M.); (S.K.); (D.L.L.)
| | - Shaza Khan
- Department of Physiology, Howard University College of Medicine, Washington, DC 20059, USA; (A.J.); (J.N.O.); (L.A.M.); (S.K.); (D.L.L.)
| | - Karl Thompson
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (J.A.); (K.T.)
| | - Maurice B. Fluitt
- Department of Medicine, Howard University College of Medicine, Washington, DC 20059, USA;
| | - Dexter L. Lee
- Department of Physiology, Howard University College of Medicine, Washington, DC 20059, USA; (A.J.); (J.N.O.); (L.A.M.); (S.K.); (D.L.L.)
| | - Carolyn M. Klinge
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Syed J. Khundmiri
- Department of Physiology, Howard University College of Medicine, Washington, DC 20059, USA; (A.J.); (J.N.O.); (L.A.M.); (S.K.); (D.L.L.)
| |
Collapse
|
3
|
Lin Y, Song Y, Zhang Y, Shi M, Hou A, Han S. NFAT signaling dysregulation in cancer: Emerging roles in cancer stem cells. Biomed Pharmacother 2023; 165:115167. [PMID: 37454598 DOI: 10.1016/j.biopha.2023.115167] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/08/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
The nuclear factor of activated T cells (NFAT) was first identified as a transcriptional regulator of activated T cells. The NFAT family is involved in the development of tumors. Furthermore, recent evidence reveals that NFAT proteins regulate the development of inflammatory and immune responses. New discoveries have also been made about the mechanisms by which NFAT regulates cancer progression through cancer stem cells (CSC). Here, we discuss the role of the NFAT family in the immune system and various cancer types.
Collapse
Affiliation(s)
- Yibin Lin
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yifu Song
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yaochuan Zhang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Mengwu Shi
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang 110001, China.
| | - Sheng Han
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
4
|
Kaye J. Integrating T Cell Activation Signals to Regulate Gene Expression through Cyclosporin-Sensitive NFAT. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:323-324. [PMID: 37987776 DOI: 10.4049/jimmunol.2300224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
This Pillars of Immunology article is a commentary on three pivotal articles: “Nuclear factor of activated T cells contains Fos and Jun,” an article written by J. Jain, P. G. McCaffrey, V. E. Valge-Archer, and A. Rao, and published in Nature, in 1992, https://www.nature.com/articles/356801a0; “The T-cell transcription factor NFATp is a substrate for calcineurin and interacts with Fos and Jun,” written by J. Jain, P. G. McCaffrey, Z. Miner, T. K. Kerppola, J. N. Lambert, G. L. Verdine, T. Curran, and A. Rao, and published in Nature, in 1993, https://www.nature.com/articles/365352a0; and “Isolation of the cyclosporin-sensitive T cell transcription factor NFATp,” written by P. G. McCaffrey, C. Luo, T. K. Kerppola, J. Jain, T. M. Badalian, A. M. Ho, E. Burgeon, W. S. Lane, J. N. Lambert, T. Curran, et al., and published in Science, in 1993, https://www.science.org/doi/10.1126/science.8235597.
Collapse
Affiliation(s)
- Jonathan Kaye
- Research Division of Immunology, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA
| |
Collapse
|
5
|
Seth A, Yokokura Y, Choi JY, Shyer JA, Vidyarthi A, Craft J. AP-1-independent NFAT signaling maintains follicular T cell function in infection and autoimmunity. J Exp Med 2023; 220:e20211110. [PMID: 36820828 PMCID: PMC9998660 DOI: 10.1084/jem.20211110] [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: 05/20/2021] [Revised: 09/05/2022] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
Coordinated gene expression programs enable development and function of T cell subsets. Follicular helper T (Tfh) cells coordinate humoral immune responses by providing selective and instructive cues to germinal center B cells. Here, we show that AP-1-independent NFAT gene expression, a program associated with hyporesponsive T cell states like anergy or exhaustion, is also a distinguishing feature of Tfh cells. NFAT signaling in Tfh cells, maintained by NFAT2 autoamplification, is required for their survival. ICOS signaling upregulates Bcl6 and induces an AP-1-independent NFAT program in primary T cells. Using lupus-prone mice, we demonstrate that genetic disruption or pharmacologic inhibition of NFAT signaling specifically impacts Tfh cell maintenance and leads to amelioration of autoantibody production and renal injury. Our data provide important conceptual and therapeutic insights into the signaling mechanisms that regulate Tfh cell development and function.
Collapse
Affiliation(s)
- Abhinav Seth
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Yoshiyuki Yokokura
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Jin-Young Choi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Justin A. Shyer
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Aurobind Vidyarthi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Joe Craft
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
6
|
Huang H, Dong J, Jiang J, Yang F, Zheng Y, Wang S, Wang N, Ma J, Hou M, Ding Y, Meng L, Zhuo W, Yang D, Qian W, Chen Q, You G, Qian G, Gu L, Lv H. The role of FOXO4/NFAT2 signaling pathway in dysfunction of human coronary endothelial cells and inflammatory infiltration of vasculitis in Kawasaki disease. Front Immunol 2023; 13:1090056. [PMID: 36700213 PMCID: PMC9869249 DOI: 10.3389/fimmu.2022.1090056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/19/2022] [Indexed: 01/12/2023] Open
Abstract
Aims The Ca+/NFAT (Nuclear factor of activated T cells) signaling pathway activation is implicated in the pathogenesis of Kawasaki disease (KD); however, we lack detailed information regarding the regulatory network involved in the human coronary endothelial cell dysfunction and cardiovascular lesion development. Herein, we aimed to use mouse and endothelial cell models of KD vasculitis in vivo and in vitro to characterize the regulatory network of NFAT pathway in KD. Methods and Results Among the NFAT gene family, NFAT2 showed the strongest transcriptional activity in peripheral blood mononuclear cells (PBMCs) from patients with KD. Then, NFAT2 overexpression and knockdown experiments in Human coronary artery endothelial cells (HCAECs) indicated that NFAT2 overexpression disrupted endothelial cell homeostasis by regulation of adherens junctions, whereas its knockdown protected HCAECs from such dysfunction. Combined analysis using RNA-sequencing and transcription factor (TF) binding site analysis in the NFAT2 promoter region predicted regulation by Forkhead box O4 (FOXO4). Western blotting, chromatin immunoprecipitation, and luciferase assays validated that FOXO4 binds to the promoter and transcriptionally represses NFAT2. Moreover, Foxo4 knockout increased the extent of inflamed vascular tissues in a mouse model of KD vasculitis. Functional experiments showed that inhibition NFAT2 relieved Foxo4 knockout exaggerated vasculitis in vivo. Conclusions Our findings revealed the FOXO4/NFAT2 axis as a vital pathway in the progression of KD that is associated with endothelial cell homeostasis and cardiovascular inflammation development.
Collapse
Affiliation(s)
- Hongbiao Huang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China,Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China,Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jinfeng Dong
- Department of Hematology, the First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Jiaqi Jiang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Fang Yang
- Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Yiming Zheng
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Shuhui Wang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Nana Wang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jin Ma
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Miao Hou
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yueyue Ding
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lijun Meng
- Department of Hematology, Children’s Hospital of Soochow University, Suzhou, China
| | - Wenyu Zhuo
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Daoping Yang
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Weiguo Qian
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qiaobin Chen
- Department of Pediatrics, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Guoping You
- Department of Emergency, Fujian Provincial Hospital, Fujian Provincial Clinical College of Fujian Medical University, Fuzhou, Fujian, China
| | - Guanghui Qian
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lei Gu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany,Cardiopulmonary Institute (CPI), Bad Nauheim, Germany,*Correspondence: Haitao Lv, ; Lei Gu,
| | - Haitao Lv
- Department of Pediatrics, Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China,*Correspondence: Haitao Lv, ; Lei Gu,
| |
Collapse
|
7
|
Role of PARP Inhibitors in Cancer Immunotherapy: Potential Friends to Immune Activating Molecules and Foes to Immune Checkpoints. Cancers (Basel) 2022; 14:cancers14225633. [PMID: 36428727 PMCID: PMC9688455 DOI: 10.3390/cancers14225633] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/04/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
Abstract
Poly (ADP-ribose) polymerase (PARP) inhibitors (PARPi) induce cytotoxic effects as single agents in tumors characterized by defective repair of DNA double-strand breaks deriving from BRCA1/2 mutations or other abnormalities in genes associated with homologous recombination. Preclinical studies have shown that PARPi-induced DNA damage may affect the tumor immune microenvironment and immune-mediated anti-tumor response through several mechanisms. In particular, increased DNA damage has been shown to induce the activation of type I interferon pathway and up-regulation of PD-L1 expression in cancer cells, which can both enhance sensitivity to Immune Checkpoint Inhibitors (ICIs). Despite the recent approval of ICIs for a number of advanced cancer types based on their ability to reinvigorate T-cell-mediated antitumor immune responses, a consistent percentage of treated patients fail to respond, strongly encouraging the identification of combination therapies to overcome resistance. In the present review, we analyzed both established and unexplored mechanisms that may be elicited by PARPi, supporting immune reactivation and their potential synergism with currently used ICIs. This analysis may indicate novel and possibly patient-specific immune features that might represent new pharmacological targets of PARPi, potentially leading to the identification of predictive biomarkers of response to their combination with ICIs.
Collapse
|
8
|
Abstract
Undifferentiated small round cell sarcomas (SRCSs) of bone and soft tissue comprise a heterogeneous group of highly aggressive tumours associated with a poor prognosis, especially in metastatic disease. SRCS entities mainly occur in the third decade of life and can exhibit striking disparities regarding preferentially affected sex and tumour localization. SRCSs comprise new entities defined by specific genetic abnormalities, namely EWSR1-non-ETS fusions, CIC-rearrangements or BCOR genetic alterations, as well as EWSR1-ETS fusions in the prototypic SRCS Ewing sarcoma. These gene fusions mainly encode aberrant oncogenic transcription factors that massively rewire the transcriptome and epigenome of the as yet unknown cell or cells of origin. Additional mutations or copy number variants are rare at diagnosis and, depending on the tumour entity, may involve TP53, CDKN2A and others. Histologically, these lesions consist of small round cells expressing variable levels of CD99 and specific marker proteins, including cyclin B3, ETV4, WT1, NKX3-1 and aggrecan, depending on the entity. Besides locoregional treatment that should follow standard protocols for sarcoma management, (neo)adjuvant treatment is as yet ill-defined but generally follows that of Ewing sarcoma and is associated with adverse effects that might compromise quality of life. Emerging studies on the molecular mechanisms of SRCSs and the development of genetically engineered animal models hold promise for improvements in early detection, disease monitoring, treatment-related toxicity, overall survival and quality of life.
Collapse
|
9
|
A Novel Prognostic Four-Gene Signature of Breast Cancer Identified by Integrated Bioinformatics Analysis. DISEASE MARKERS 2022; 2022:5925982. [PMID: 35265226 PMCID: PMC8898848 DOI: 10.1155/2022/5925982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 11/20/2022]
Abstract
Molecular analysis facilitates the prediction of overall survival (OS) of breast cancer and decision-making of the treatment plan. The current study was designed to identify new prognostic genes for breast cancer and construct an effective prognostic signature with integrated bioinformatics analysis. Differentially expressed genes in breast cancer samples from The Cancer Genome Atlas (TCGA) dataset were filtered by univariate Cox regression analysis. The prognostic model was optimized by the Akaike information criterion and further validated using the TCGA dataset (n = 1014) and Gene Expression Omnibus (GEO) dataset (n = 307). The correlation between the risk score and clinical information was assessed by univariate and multivariate Cox regression analyses. Functional pathways in relation to high-risk and low-risk groups were analyzed using gene set enrichment analysis (GSEA). Four prognostic genes (EXOC6, GPC6, PCK2, and NFATC2) were screened and used to construct a prognostic model, which showed robust performance in classifying the high-risk and low-risk groups. The risk score was significantly related to clinical features and OS. We identified 19 functional pathways significantly associated with the risk score. This study constructed a new prognostic model with a high prediction performance for breast cancer. The four-gene prognostic signature could serve as an effective tool to predict prognosis and assist the management of breast cancer patients.
Collapse
|
10
|
Yassouf MY, Zhang X, Huang Z, Zhai D, Sekiya R, Kawabata T, Li TS. Biphasic effect of mechanical stress on lymphocyte activation. J Cell Physiol 2022; 237:1521-1531. [PMID: 34724217 DOI: 10.1002/jcp.30623] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/19/2021] [Accepted: 10/22/2021] [Indexed: 01/08/2023]
Abstract
Mechanical forces can modulate the immune response, mostly described as promoting the activation of immune cells, but the role and mechanism of pathological levels of mechanical stress in lymphocyte activation have not been focused on before. By an ex vivo experimental approach, we observed that mechanical stressing of murine spleen lymphocytes with 50 mmHg for 3 h induced the nuclear localization of NFAT1, increased C-Jun, and increased the expression of early activation marker CD69 in resting CD8+ cells. Interestingly, 50 mmHg mechanical stressing induced the nuclear localization of NFAT1; but conversely decreased C-Jun and inhibited the expression of CD69 in lymphocytes under lipopolysaccharide or phorbol 12-myristate 13-acetate/ionomycin stimulation. Additionally, we observed similar changes trends when comparing RNA-seq data of hypertensive and normotensive COVID-19 patients. Our results indicate a biphasic effect of mechanical stress on lymphocyte activation, which provides insight into the variety of immune responses in pathologies involving elevated mechanical stress.
Collapse
Affiliation(s)
- Mhd Yousuf Yassouf
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Xu Zhang
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Zisheng Huang
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Da Zhai
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Reiko Sekiya
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
11
|
Ong SL, Lam SW, van den Akker BE, Kroon HM, Briaire-de Bruijn IH, Cleven AH, Savci-Heijink DC, Cleton-Jansen AM, Baumhoer D, Szuhai K, Bovée JV. Expanding the Spectrum of EWSR1-NFATC2-rearranged Benign Tumors: A Common Genomic Abnormality in Vascular Malformation/Hemangioma and Simple Bone Cyst. Am J Surg Pathol 2021; 45:1669-1681. [PMID: 34081036 PMCID: PMC8598111 DOI: 10.1097/pas.0000000000001748] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A simple bone cyst (SBC) is a cystic bone lesion predominantly affecting young males. The cyst is lined by a fibrous membrane and filled with serosanguinous fluid. EWSR1/FUS-NFATC2 rearrangements were recently identified in SBC. We here report exactly the same rearrangement in 3 lesions diagnosed as vascular malformations of 2 elderly patients. In total, through Archer FusionPlex, fluorescence in situ hybridization and/or reverse transcriptase-polymerase chain reaction the EWSR1-NFATC2 rearrangement was identified in 6 of 9 SBC, 3 of 12 benign vascular tumors, and none of 5 aneurysmal bone cyst lacking USP6 fusion. Using fluorescence in situ hybridization, it was apparent that amplification of the fusion, as seen in EWSR1-NFATC2 round cell sarcomas, was absent, and that in the vascular tumors the fusion was present both in the lining cells as well as in the surrounding spindle cells. Of note, not all of the spaces in the vascular malformations were lined by endothelial cells. Aggrecan was positive in all cases but was not specific. NKX2-2 and NKX3-1 staining were negative in all cases. Thus, even though the overlap between the 2 entities is limited to the presence of few thick-walled cysts lacking endothelial lining in the benign vascular malformations, the spectrum of benign tumors containing NFATC2 fusions should be expanded and contains not only SBC in the young, but also vascular malformation/hemangioma in elderly patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Daniel Baumhoer
- Department of Medical Genetics and Pathology, Bone Tumor Reference Center, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Karoly Szuhai
- Cell and Chemical Biology, Leiden University Medical Center, Leiden
| | | |
Collapse
|
12
|
Rao A. Scientific divagations: from signaling and transcription to chromatin changes in T cells. Nat Immunol 2021; 21:1473-1476. [PMID: 33173218 DOI: 10.1038/s41590-020-00823-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Anjana Rao
- Division of Signaling and Gene Expression, La Jolla Institute for Immunology, La Jolla, CA, USA.
| |
Collapse
|
13
|
Sallinger M, Tiffner A, Schmidt T, Bonhenry D, Waldherr L, Frischauf I, Lunz V, Derler I, Schober R, Schindl R. Luminal STIM1 Mutants that Cause Tubular Aggregate Myopathy Promote Autophagic Processes. Int J Mol Sci 2020; 21:E4410. [PMID: 32575830 PMCID: PMC7352373 DOI: 10.3390/ijms21124410] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022] Open
Abstract
Stromal interaction molecule 1 (STIM1) is a ubiquitously expressed Ca2+ sensor protein that induces permeation of Orai Ca2+ channels upon endoplasmic reticulum Ca2+-store depletion. A drop in luminal Ca2+ causes partial unfolding of the N-terminal STIM1 domains and thus initial STIM1 activation. We compared the STIM1 structure upon Ca2+ depletion from our molecular dynamics (MD) simulations with a recent 2D NMR structure. Simulation- and structure-based results showed unfolding of two α-helices in the canonical and in the non-canonical EF-hand. Further, we structurally and functionally evaluated mutations in the non-canonical EF-hand that have been shown to cause tubular aggregate myopathy. We found these mutations to cause full constitutive activation of Ca2+-release-activated Ca2+ currents (ICRAC) and to promote autophagic processes. Specifically, heterologously expressed STIM1 mutations in the non-canonical EF-hand promoted translocation of the autophagy transcription factors microphthalmia-associated transcription factor (MITF) and transcription factor EB (TFEB) into the nucleus. These STIM1 mutations additionally stimulated an enhanced production of autophagosomes. In summary, mutations in STIM1 that cause structural unfolding promoted Ca2+ down-stream activation of autophagic processes.
Collapse
Affiliation(s)
- Matthias Sallinger
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Adéla Tiffner
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Tony Schmidt
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
| | - Daniel Bonhenry
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, CZ-373 33 Nove Hrady, Czech Republic;
| | - Linda Waldherr
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
| | - Irene Frischauf
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Victoria Lunz
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
| | - Romana Schober
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria; (M.S.); (A.T.); (I.F.); (V.L.); (I.D.)
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
| | - Rainer Schindl
- Gottfried Schatz Research Center, Medical University of Graz, A-8010 Graz, Austria; (T.S.); (L.W.)
- BioTechMed-Graz, A-8010 Graz, Austria
| |
Collapse
|
14
|
Cole AJ, Iyengar M, Panesso-Gómez S, O'Hayer P, Chan D, Delgoffe GM, Aird KM, Yoon E, Bai S, Buckanovich RJ. NFATC4 promotes quiescence and chemotherapy resistance in ovarian cancer. JCI Insight 2020; 5:131486. [PMID: 32182216 DOI: 10.1172/jci.insight.131486] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 03/11/2020] [Indexed: 12/30/2022] Open
Abstract
Development of chemotherapy resistance is a major problem in ovarian cancer. One understudied mechanism of chemoresistance is the induction of quiescence, a reversible nonproliferative state. Unfortunately, little is known about regulators of quiescence. Here, we identify the master transcription factor nuclear factor of activated T cells cytoplasmic 4 (NFATC4) as a regulator of quiescence in ovarian cancer. NFATC4 is enriched in ovarian cancer stem-like cells and correlates with decreased proliferation and poor prognosis. Treatment of cancer cells with cisplatin resulted in NFATC4 nuclear translocation and activation of the NFATC4 pathway, while inhibition of the pathway increased chemotherapy response. Induction of NFATC4 activity resulted in a marked decrease in proliferation, G0 cell cycle arrest, and chemotherapy resistance, both in vitro and in vivo. Finally, NFATC4 drove a quiescent phenotype in part via downregulation of MYC. Together, these data identify NFATC4 as a driver of quiescence and a potential new target to combat chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Alexander J Cole
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mangala Iyengar
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Santiago Panesso-Gómez
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Patrick O'Hayer
- Department of Cellular and Molecular Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Daniel Chan
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Greg M Delgoffe
- Tumor Microenvironment Center, UPMC Hillman Cancer Center; and Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine M Aird
- Department of Cellular & Molecular Physiology, Penn State College of Medicine, Hershey, Pennsylvania, USA
| | - Euisik Yoon
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Michigan, USA
| | - Shoumei Bai
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ronald J Buckanovich
- Department of Internal Medicine and Magee-Womens Research Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.,Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Dominant mutants of the calcineurin catalytic subunit (CNA-1) showed developmental defects, increased sensitivity to stress conditions, and CNA-1 interacts with CaM and CRZ-1 in Neurospora crassa. Arch Microbiol 2019; 202:921-934. [DOI: 10.1007/s00203-019-01768-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 10/12/2019] [Accepted: 10/31/2019] [Indexed: 12/20/2022]
|
16
|
Zhou X, Zhang S, Yu F, Zhao G, Geng S, Yu W, Wang XY, Wang B. Tolerogenic vaccine composited with islet-derived multipeptides and cyclosporin A induces pTreg and prevents Type 1 diabetes in murine model. Hum Vaccin Immunother 2019; 16:240-250. [PMID: 31070990 PMCID: PMC7062422 DOI: 10.1080/21645515.2019.1616504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Regulatory T cells (Tregs) play a crucial role in the control of the initiation and progression of type 1 diabetes (T1D). Various immunological interventions including those to ex vivo expansion Tregs transfer, in vivo induction of peripherally derived Treg (pTreg) have been considered as promising approaches for T1D therapy. In this study, we developed a novel tolerogenic vaccine using four autoantigenic peptides of islet-derived with cyclosporine A (CsA) as the pTreg inducer, designated as GAD-IN+CsA. This vaccine immunized into prediabetic NOD mice subcutaneously could induce IL-10 and TGF-β expressing pTregs and lead to suppressing autoreactive T cells responses, resulting in the prevention of T1D in these animals. Furthermore, we demonstrated that CsA with autoantigenic peptides modulates dendritic cells (DCs) to become immature IL-10hiCD40lo DCs. Such modulated DCs could foster naïve CD4+CD25− T cell into Tregs when presenting antigen peptides in vitro. This novel approach offers an alternative strategy to induce pTregs to treat T1D.
Collapse
Affiliation(s)
- Xian Zhou
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shijie Zhang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Fan Yu
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Gan Zhao
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Shuang Geng
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Wencong Yu
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China
| | - Xuan-Yi Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology of the Ministry of Health and the Ministry of Education, Shanghai Basic Medical College, Fudan University, Shanghai, China.,Children Hospital of Fudan University, Shanghai, China
| |
Collapse
|
17
|
Seal S, Polley S, Sau S. A staphylococcal cyclophilin carries a single domain and unfolds via the formation of an intermediate that preserves cyclosporin A binding activity. PLoS One 2019; 14:e0210771. [PMID: 30925148 PMCID: PMC6440624 DOI: 10.1371/journal.pone.0210771] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 03/18/2019] [Indexed: 12/22/2022] Open
Abstract
Cyclophilin (Cyp), a peptidyl-prolyl cis-trans isomerase (PPIase), acts as a virulence factor in many bacteria including Staphylococcus aureus. The enzymatic activity of Cyp is inhibited by cyclosporin A (CsA), an immunosuppressive drug. To precisely determine the unfolding mechanism and the domain structure of Cyp, we have investigated a chimeric S. aureus Cyp (rCyp) using various probes. Our limited proteolysis and the consequent analysis of the proteolytic fragments indicate that rCyp is composed of one domain with a short flexible tail at the C-terminal end. We also show that the urea-induced unfolding of both rCyp and rCyp-CsA is completely reversible and proceeds via the synthesis of at least one stable intermediate. Both the secondary structure and the tertiary structure of each intermediate appears very similar to those of the corresponding native protein. Conversely, the hydrophobic surface areas of the intermediates are comparatively less. Further analyses reveal no loss of CsA binding activity in rCyp intermediate. The thermodynamic stability of rCyp was also significantly increased in the presence of CsA, recommending that this protein could be employed to screen new CsA derivatives in the future.
Collapse
Affiliation(s)
- Soham Seal
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | - Soumitra Polley
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
| | - Subrata Sau
- Department of Biochemistry, Bose Institute, Kolkata, West Bengal, India
- * E-mail:
| |
Collapse
|
18
|
Yamaguchi K. Tacrolimus treatment for infertility related to maternal-fetal immune interactions. Am J Reprod Immunol 2019; 81:e13097. [PMID: 30689243 DOI: 10.1111/aji.13097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/29/2022] Open
Abstract
Many approaches have been used to achieve successful pregnancies in patients with infertility, though existing treatments remain unsatisfactory in patients with infertility caused by abnormal maternal-fetal immunity. However, our understanding of the immunological aspects of infertility has steadily progressed, aided by recent research into organ transplantation and cancer. The results of these recent analyses have led to the development and evaluation of several candidate immunological treatments, but the use of immunological treatments remains a novel approach. The current paper presents the hypothesis that tacrolimus may have potential as a candidate agent for the treatment of maternal-fetal immunity-related infertility.
Collapse
Affiliation(s)
- Koushi Yamaguchi
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
19
|
Oxysterol-binding protein-related protein 4L promotes cell proliferation by sustaining intracellular Ca2+ homeostasis in cervical carcinoma cell lines. Oncotarget 2018; 7:65849-65861. [PMID: 27588468 PMCID: PMC5323197 DOI: 10.18632/oncotarget.11671] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 08/09/2016] [Indexed: 12/30/2022] Open
Abstract
Oxsterol binding protein-related protein 4 (ORP4) is essential for cell proliferation, but the underlying mechanism is unclear. ORP4 is expressed as three variants, ORP4L, ORP4M and ORP4S. Here, we reported that silencing of ORP4L with specific small interfering RNA (siRNA) inhibited the proliferation of human cervical cancer cell lines C33A, HeLa and CaSki, the reverse effect being observed in ORP4L overexpressing cells. For molecular insight, we found that ORP4L maintained intracellular Ca2+ homeostasis. Through this mechanism, ORP4L activated nuclear factor of activated T cells (NFAT) activity and thus promoted expression of a gene cluster which supported cell proliferation. Of note, ORP4L sustained inositol-1,4,5-trisphosphate receptor 1 (IP3R1) expression at both mRNA and protein levels via Ca2+-dependent NFAT3 activation, which offered a mechanic explanation for the role of ORP4L intracellular Ca2+ homeostasis. Furthermore, ORP4L knockdown markedly inhibited tumor growth in a C33A cell xenograft mouse model. To conclude, our results reveal that ORP4L promotes cell proliferation through maintaining intracellular Ca2+ homeostasis.
Collapse
|
20
|
Abstract
Nuclear factor of activated T cells (NFAT) was first described almost three decades ago as a Ca
2+/calcineurin-regulated transcription factor in T cells. Since then, a large body of research uncovered the regulation and physiological function of different NFAT homologues in the immune system and many other tissues. In this review, we will discuss novel roles of NFAT in T cells, focusing mainly on its function in humoral immune responses, immunological tolerance, and the regulation of immune metabolism.
Collapse
Affiliation(s)
- Martin Vaeth
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| | - Stefan Feske
- Department of Pathology, New York University School of Medicine, New York, NY, 10016, USA
| |
Collapse
|
21
|
|
22
|
Ao M, Pan Z, Qian Y, Tang B, Feng Z, Fang H, Wu Z, Chen J, Xue Y, Fang M. Design, synthesis, and biological evaluation of AV6 derivatives as novel dual reactivators of latent HIV-1. RSC Adv 2018; 8:17279-17292. [PMID: 35539279 PMCID: PMC9080425 DOI: 10.1039/c8ra01216d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/24/2018] [Indexed: 01/31/2023] Open
Abstract
The “shock and kill” strategy might be a promising therapeutic approach for HIV/AIDS due to the existence of latent viral reservoirs. A major challenge of the “shock and kill” strategy arises from the general lack of clinically effective latency-reversing agents (LRAs). The 2-methylquinoline derivative, antiviral 6 (AV6) has been reported to induce latent HIV-1 expression and act synergistically with a HDAC inhibitor VA to reverse HIV latency. We report herein the design and identification of AV6 analogues which possess the zinc-binding group of HDAC inhibitors and have dual acting mechanism for the reactivation of HIV-1 from latency. Evaluation of compounds for the reactivation of HIV-1 latency identified two excellent active compounds 12c and 12d. Further bioassays revealed that these two compounds reactivated latent HIV-1 through dual mechanism, the inhibition of HDACs and NFAT-required for early HIV-1 gene expression. Additionally, it was found that 12c and 12d could reactivate HIV-1 transcription by releasing P-TEFb from the inactive complex 7SK snRNP. At last, molecular docking identified their orientation and binding interactions at the active site of HDAC2. This experimental data suggests that 12c and 12d can be served as effective HIV-1 LRAs which can be taken up for further studies. As dual-acting HIV LRAs, compounds 12c and 12d could activate latent HIV-1 via the NFAT-required mechanism and as histone deacetylase (HDAC) inhibitors.![]()
Collapse
|
23
|
|
24
|
Frischauf I, Litviňuková M, Schober R, Zayats V, Svobodová B, Bonhenry D, Lunz V, Cappello S, Tociu L, Reha D, Stallinger A, Hochreiter A, Pammer T, Butorac C, Muik M, Groschner K, Bogeski I, Ettrich RH, Romanin C, Schindl R. Transmembrane helix connectivity in Orai1 controls two gates for calcium-dependent transcription. Sci Signal 2017; 10:eaao0358. [PMID: 29184031 PMCID: PMC6433236 DOI: 10.1126/scisignal.aao0358] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The channel Orai1 requires Ca2+ store depletion in the endoplasmic reticulum and an interaction with the Ca2+ sensor STIM1 to mediate Ca2+ signaling. Alterations in Orai1-mediated Ca2+ influx have been linked to several pathological conditions including immunodeficiency, tubular myopathy, and cancer. We screened large-scale cancer genomics data sets for dysfunctional Orai1 mutants. Five of the identified Orai1 mutations resulted in constitutively active gating and transcriptional activation. Our analysis showed that certain Orai1 mutations were clustered in the transmembrane 2 helix surrounding the pore, which is a trigger site for Orai1 channel gating. Analysis of the constitutively open Orai1 mutant channels revealed two fundamental gates that enabled Ca2+ influx: Arginine side chains were displaced so they no longer blocked the pore, and a chain of water molecules formed in the hydrophobic pore region. Together, these results enabled us to identify a cluster of Orai1 mutations that trigger Ca2+ permeation associated with gene transcription and provide a gating mechanism for Orai1.
Collapse
Affiliation(s)
- Irene Frischauf
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
| | - Monika Litviňuková
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Cardiovascular and Metabolic Sciences, Berlin D-13125, Germany
| | - Romana Schober
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
| | - Vasilina Zayats
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Nové Hrady CZ-373 33, Czech Republic
- Center of New Technologies, University of Warsaw, Warsaw 02-097, Poland
| | - Barbora Svobodová
- Institute for Biophysics, Medical University of Graz, Graz A-8010, Austria
| | - Daniel Bonhenry
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Nové Hrady CZ-373 33, Czech Republic
| | - Victoria Lunz
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
| | - Sabrina Cappello
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg August University Göttingen, Göttingen, Niedersachsen 37073, Germany
- Department of Biophysics, Center for Integrative Physiology and Molecular Medicine, Medical Faculty, Saarland University, Homburg D-66421, Germany
| | - Laura Tociu
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Nové Hrady CZ-373 33, Czech Republic
- University of Chicago, Chicago, IL 60637, USA
| | - David Reha
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Nové Hrady CZ-373 33, Czech Republic
- Faculty of Sciences, University of South Bohemia, Nové Hrady CZ-373 33, Czech Republic
| | - Amrutha Stallinger
- Institute for Molecular Biosciences, Karl-Franzens-University Graz, Graz A-8010, Austria
| | - Anna Hochreiter
- Institute for Experimental and Clinical Cell Therapy, Paracelsus Medical University, Salzburg A-5020, Austria
| | - Teresa Pammer
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
| | - Carmen Butorac
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
| | - Martin Muik
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
| | - Klaus Groschner
- Institute for Biophysics, Medical University of Graz, Graz A-8010, Austria
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg August University Göttingen, Göttingen, Niedersachsen 37073, Germany
| | - Rüdiger H Ettrich
- Center for Nanobiology and Structural Biology, Institute of Microbiology, Academy of Sciences of the Czech Republic, Nové Hrady CZ-373 33, Czech Republic
- Faculty of Sciences, University of South Bohemia, Nové Hrady CZ-373 33, Czech Republic
| | - Christoph Romanin
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, Linz A-4020, Austria
| | - Rainer Schindl
- Institute for Biophysics, Medical University of Graz, Graz A-8010, Austria.
| |
Collapse
|
25
|
Wang Z, Yang H, Si S, Han Z, Tao J, Chen H, Ge Y, Guo M, Wang K, Tan R, Wei JF, Gu M. Polymorphisms of nucleotide factor of activated T cells cytoplasmic 2 and 4 and the risk of acute rejection following kidney transplantation. World J Urol 2017; 36:111-116. [PMID: 29103109 PMCID: PMC5758697 DOI: 10.1007/s00345-017-2117-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/27/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Acute rejection (AR) is a common complication of kidney transplantation. Nuclear factors of activated T cells (NFATs) are transcription factors involved in the activation of T lymphocytes, but their association with AR is unclear. METHODS This retrospective, case-control study included 200 renal transplant recipients who were divided into the AR group (n = 69) and stable group (n = 131). Their blood samples were collected, and DNA was extracted from the whole blood. High-throughput next-generation sequencing was used to identify single nucleotide polymorphisms (SNPs) of the NFATC2 and NFATC4 genes. The correlation of these SNPs with AR was determined by logistic analysis. RESULTS Seventy-one SNPs of the NFATC2 and NFATC4 genes were identified by the sequencing and Hardy-Weinberg equilibrium analyses. After adjusting for age, gender and immunosuppressive protocols, 27 SNPs were correlated with AR, of which the SNP rs2426295 of the NFATC2 gene showed a significant correlation with AR in the HET model (AA vs. AC: OR = 0.43, 95% CI = 0.19-0.98, P = 0.045), but no significant NFATC4 SNPs were identified. CONCLUSIONS Our study shows that the rs2426295 variant of the NFATC2 gene is significantly associated with the occurrence of AR following kidney transplantation. And patients with AA genotypes in rs2426295 are inclined to suffer from AR pathogenesis.
Collapse
Affiliation(s)
- Zijie Wang
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Haiwei Yang
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Shuhui Si
- Research Division of Clinical Pharmacology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Zhijian Han
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Jun Tao
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Hao Chen
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Yuqiu Ge
- School of Public Health, Nanjing Medical University, Nanjing, 211166, People's Republic of China
| | - Miao Guo
- Research Division of Clinical Pharmacology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China
| | - Ji-Fu Wei
- Research Division of Clinical Pharmacology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China.
| | - Min Gu
- Department of Urology, The First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, People's Republic of China.
| |
Collapse
|
26
|
Regulation of the phosphatase PP2B by protein-protein interactions. Biochem Soc Trans 2017; 44:1313-1319. [PMID: 27911714 DOI: 10.1042/bst20160150] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/09/2016] [Accepted: 07/14/2016] [Indexed: 02/06/2023]
Abstract
Protein dephosphorylation is important for regulating cellular signaling in a variety of contexts. Protein phosphatase-2B (PP2B), or calcineurin, is a widely expressed serine/threonine phosphatase that acts on a large cross section of potential protein substrates when activated by increased levels of intracellular calcium in concert with calmodulin. PxIxIT and LxVP targeting motifs are important for maintaining specificity in response to elevated calcium. In the present study, we describe the mechanism of PP2B activation, discuss its targeting by conserved binding motifs and review recent advances in the understanding of an A-kinase anchoring protein 79/PP2B/protein kinase A complex's role in synaptic long-term depression. Finally, we discuss potential for targeting PP2B anchoring motifs for therapeutic benefit.
Collapse
|
27
|
Hogan PG. Calcium-NFAT transcriptional signalling in T cell activation and T cell exhaustion. Cell Calcium 2017; 63:66-69. [PMID: 28153342 PMCID: PMC5739523 DOI: 10.1016/j.ceca.2017.01.014] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 01/26/2017] [Indexed: 02/02/2023]
Abstract
A cornerstone of the adaptive immune response is the T cell receptor (TcR)-calcium-calcineurin signalling pathway leading to T cell activation. The 'nuclear factor of activated T cells' proteins NFAT1, NFAT2, and NFAT4 are transcription factors that promote expression of a panel of genes required for activation. It has become apparent that these same NFAT transcription factors underlie an alternative transcriptional program in T cells that serves to limit the immune response. This duality in NFAT transcriptional functions raises the possibility that NFAT transcriptional complexes could be targeted therapeutically to alter the relative strength of the effector and alternative transcriptional programs, thereby modulating immune responses.
Collapse
Affiliation(s)
- Patrick G Hogan
- La Jolla Institute for Allergy & Immunology, 9420 Athena Circle, La Jolla, CA 92037, United States.
| |
Collapse
|
28
|
Lückemann L, Unteroberdörster M, Kirchhof J, Schedlowski M, Hadamitzky M. Applications and limitations of behaviorally conditioned immunopharmacological responses. Neurobiol Learn Mem 2017; 142:91-98. [PMID: 28216206 DOI: 10.1016/j.nlm.2017.02.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 02/10/2017] [Accepted: 02/15/2017] [Indexed: 12/17/2022]
Abstract
The importance of placebo responses for the treatment of various medical conditions has increasingly been recognized, whereas knowledge and systematic application in clinical settings are still sparse. One possible application for placebo responses in pharmacotherapy is given by learning paradigms, such as behaviorally conditioned immunosuppression, aiming at drug dose reduction while maintaining therapeutic efficacy of drug treatment. In an established learning paradigm of conditioned taste aversion/avoidance (CTA) in both, rats and humans, respectively, a novel-tasting drinking solution (conditioned stimulus, CS) is paired with an injection of the immunosuppressive drug cyclosporine A (CsA) as unconditioned stimulus (US). The conditioned response, evoked by re-presenting the CS alone at a later time, is reflected by avoidance behavior of consuming the solution (conditioned taste aversion; CTA) and a diminished interleukin (IL)-2 and interferon (IFN)-γ cytokine production as well as mRNA expression of rat splenic T cells or human peripheral T lymphocytes, closely mimicking the immunosuppressive effects of CsA. However, due to unreinforced CS-re-exposure conditioned responses progressively decreases over time (extinction), reflecting a considerable challenge for potential clinical applications of this learned immunosuppression. The present article discusses and critically reviews actual approaches, applications but also limitations of learning paradigms in immune pharmacotherapy.
Collapse
Affiliation(s)
- Laura Lückemann
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Meike Unteroberdörster
- Department of Neurosurgery, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Julia Kirchhof
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Manfred Schedlowski
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany
| | - Martin Hadamitzky
- Institute of Medical Psychology and Behavioral Immunobiology, University Hospital Essen, University of Duisburg-Essen, 45122 Essen, Germany.
| |
Collapse
|
29
|
Gabriel CH, Gross F, Karl M, Stephanowitz H, Hennig AF, Weber M, Gryzik S, Bachmann I, Hecklau K, Wienands J, Schuchhardt J, Herzel H, Radbruch A, Krause E, Baumgrass R. Identification of Novel Nuclear Factor of Activated T Cell (NFAT)-associated Proteins in T Cells. J Biol Chem 2016; 291:24172-24187. [PMID: 27637333 DOI: 10.1074/jbc.m116.739326] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 09/13/2016] [Indexed: 12/12/2022] Open
Abstract
Transcription factors of the nuclear factor of activated T cell (NFAT) family are essential for antigen-specific T cell activation and differentiation. Their cooperative DNA binding with other transcription factors, such as AP1 proteins (FOS, JUN, and JUNB), FOXP3, IRFs, and EGR1, dictates the gene regulatory action of NFATs. To identify as yet unknown interaction partners of NFAT, we purified biotin-tagged NFATc1/αA, NFATc1/βC, and NFATc2/C protein complexes and analyzed their components by stable isotope labeling by amino acids in cell culture-based mass spectrometry. We revealed more than 170 NFAT-associated proteins, half of which are involved in transcriptional regulation. Among them are many hitherto unknown interaction partners of NFATc1 and NFATc2 in T cells, such as Raptor, CHEK1, CREB1, RUNX1, SATB1, Ikaros, and Helios. The association of NFATc2 with several other transcription factors is DNA-dependent, indicating cooperative DNA binding. Moreover, our computational analysis discovered that binding motifs for RUNX and CREB1 are found preferentially in the direct vicinity of NFAT-binding motifs and in a distinct orientation to them. Furthermore, we provide evidence that mTOR and CHEK1 kinase activity influence NFAT's transcriptional potency. Finally, our dataset of NFAT-associated proteins provides a good basis to further study NFAT's diverse functions and how these are modulated due to the interplay of multiple interaction partners.
Collapse
Affiliation(s)
- Christian H Gabriel
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin
| | - Fridolin Gross
- the Institute for Theoretical Biology, Charité and Humboldt University Berlin, 10015 Berlin
| | - Martin Karl
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin
| | | | - Anna Floriane Hennig
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin
| | - Melanie Weber
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin
| | - Stefanie Gryzik
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin
| | | | - Katharina Hecklau
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin
| | - Jürgen Wienands
- the Institute of Cellular and Molecular Immunology, Georg-August-University of Göttingen, 37073 Göttingen, Germany
| | | | - Hanspeter Herzel
- the Institute for Theoretical Biology, Charité and Humboldt University Berlin, 10015 Berlin
| | - Andreas Radbruch
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin
| | - Eberhard Krause
- the Leibniz-Institut für Molekulare Pharmakologie, 13125 Berlin
| | - Ria Baumgrass
- From the German Rheumatism Research Center (DRFZ), Leibniz Institute, 10117 Berlin,
| |
Collapse
|
30
|
AKAP150 participates in calcineurin/NFAT activation during the down-regulation of voltage-gated K(+) currents in ventricular myocytes following myocardial infarction. Cell Signal 2015; 28:733-40. [PMID: 26724383 DOI: 10.1016/j.cellsig.2015.12.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 12/22/2015] [Indexed: 12/19/2022]
Abstract
The Ca(2+)-responsive phosphatase calcineurin/protein phosphatase 2B dephosphorylates the transcription factor NFATc3. In the myocardium activation of NFATc3 down-regulates the expression of voltage-gated K(+) (Kv) channels after myocardial infarction (MI). This prolongs action potential duration and increases the probability of arrhythmias. Although recent studies infer that calcineurin is activated by local and transient Ca(2+) signals the molecular mechanism that underlies the process is unclear in ventricular myocytes. Here we test the hypothesis that sequestering of calcineurin to the sarcolemma of ventricular myocytes by the anchoring protein AKAP150 is required for acute activation of NFATc3 and the concomitant down-regulation of Kv channels following MI. Biochemical and cell based measurements resolve that approximately 0.2% of the total calcineurin activity in cardiomyocytes is associated with AKAP150. Electrophysiological analyses establish that formation of this AKAP150-calcineurin signaling dyad is essential for the activation of the phosphatase and the subsequent down-regulation of Kv channel currents following MI. Thus AKAP150-mediated targeting of calcineurin to sarcolemmal micro-domains in ventricular myocytes contributes to the local and acute gene remodeling events that lead to the down-regulation of Kv currents.
Collapse
|
31
|
|
32
|
Placing ion channels into a signaling network of T cells: from maturing thymocytes to healthy T lymphocytes or leukemic T lymphoblasts. BIOMED RESEARCH INTERNATIONAL 2015; 2015:750203. [PMID: 25866806 PMCID: PMC4383400 DOI: 10.1155/2015/750203] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022]
Abstract
T leukemogenesis is a multistep process, where the genetic errors during T cell maturation cause the healthy progenitor to convert into the leukemic precursor that lost its ability to differentiate but possesses high potential for proliferation, self-renewal, and migration. A new misdirecting "leukemogenic" signaling network appears, composed by three types of participants which are encoded by (1) genes implicated in determined stages of T cell development but deregulated by translocations or mutations, (2) genes which normally do not participate in T cell development but are upregulated, and (3) nondifferentially expressed genes which become highly interconnected with genes expressed differentially. It appears that each of three groups may contain genes coding ion channels. In T cells, ion channels are implicated in regulation of cell cycle progression, differentiation, activation, migration, and cell death. In the present review we are going to reveal a relationship between different genetic defects, which drive the T cell neoplasias, with calcium signaling and ion channels. We suggest that changes in regulation of various ion channels in different types of the T leukemias may provide the intracellular ion microenvironment favorable to maintain self-renewal capacity, arrest differentiation, induce proliferation, and enhance motility.
Collapse
|
33
|
Caldwell KL, Wang J. Cell-based articular cartilage repair: the link between development and regeneration. Osteoarthritis Cartilage 2015; 23:351-62. [PMID: 25450846 PMCID: PMC4339504 DOI: 10.1016/j.joca.2014.11.004] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/02/2014] [Accepted: 11/01/2014] [Indexed: 02/02/2023]
Abstract
Clinical efforts to repair damaged articular cartilage (AC) currently face major obstacles due to limited intrinsic repair capacity of the tissue and unsuccessful biological interventions. This highlights a need for better therapeutic strategies. This review summarizes the recent advances in the field of cell-based AC repair. In both animals and humans, AC defects that penetrate into the subchondral bone marrow are mainly filled with fibrocartilaginous tissue through the differentiation of bone marrow mesenchymal stem cells (MSCs), followed by degeneration of repaired cartilage and osteoarthritis (OA). Cell therapy and tissue engineering techniques using culture-expanded chondrocytes, bone marrow MSCs, or pluripotent stem cells with chondroinductive growth factors may generate cartilaginous tissue in AC defects but do not form hyaline cartilage-based articular surface because repair cells often lose chondrogenic activity or result in chondrocyte hypertrophy. The new evidence that AC and synovium develop from the same pool of precursors with similar gene profiles and that synovium-derived chondrocytes have stable chondrogenic activity has promoted use of synovium as a new cell source for AC repair. The recent finding that NFAT1 and NFAT2 transcription factors (TFs) inhibit chondrocyte hypertrophy and maintain metabolic balance in AC is a significant advance in the field of AC repair. The use of synovial MSCs and discovery of upstream transcriptional regulators that help maintain the AC phenotype have opened new avenues to improve the outcome of AC regeneration.
Collapse
Affiliation(s)
| | - Jinxi Wang
- Corresponding Author: Jinxi Wang, Address: University of Kansas Medical Center, Department of Orthopedic Surgery, 3901 Rainbow Blvd., Mail Stop 3017, Kansas City, KS 66160, USA, Phone: +1 913-588-0870, Fax: +1 913-945-7773,
| |
Collapse
|
34
|
Qin JJ, Nag S, Wang W, Zhou J, Zhang WD, Wang H, Zhang R. NFAT as cancer target: mission possible? Biochim Biophys Acta Rev Cancer 2014; 1846:297-311. [PMID: 25072963 DOI: 10.1016/j.bbcan.2014.07.009] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 07/17/2014] [Accepted: 07/22/2014] [Indexed: 12/30/2022]
Abstract
The NFAT signaling pathway regulates various aspects of cellular functions; NFAT acts as a calcium sensor, integrating calcium signaling with other pathways involved in development and growth, immune response, and inflammatory response. The NFAT family of transcription factors regulates diverse cellular functions such as cell survival, proliferation, migration, invasion, and angiogenesis. The NFAT isoforms are constitutively activated and overexpressed in several cancer types wherein they transactivate downstream targets that play important roles in cancer development and progression. Though the NFAT family has been conclusively proved to be pivotal in cancer progression, the different isoforms play distinct roles in different cellular contexts. In this review, our discussion is focused on the mechanisms that drive the activation of various NFAT isoforms in cancer. Additionally, we analyze the potential of NFAT as a valid target for cancer prevention and therapy.
Collapse
Affiliation(s)
- Jiang-Jiang Qin
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Subhasree Nag
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Wei Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Jianwei Zhou
- Department of Molecular Cell Biology and Toxicology, Cancer Center, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu 210029, PR China
| | - Wei-Dong Zhang
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Hui Wang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing 100021, PR China
| | - Ruiwen Zhang
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Biology Center, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA.
| |
Collapse
|
35
|
Mass E, Wachten D, Aschenbrenner AC, Voelzmann A, Hoch M. Murine Creld1 controls cardiac development through activation of calcineurin/NFATc1 signaling. Dev Cell 2014; 28:711-26. [PMID: 24697899 DOI: 10.1016/j.devcel.2014.02.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 01/28/2014] [Accepted: 02/13/2014] [Indexed: 11/30/2022]
Abstract
Calcineurin is a heteromeric Ca(2+)-dependent serine/threonine phosphatase. It dephosphorylates the transcription factor nuclear factor of activated T cells (NFAT) in the cytoplasm, which subsequently undergoes nuclear translocation. NFAT regulates numerous biological processes, including inflammatory T cell responses and cardiac development. Our study identifies the Cysteine-Rich with EGF-Like Domains 1 (Creld1) gene as a regulator of calcineurin/NFATc1 signaling. We show that Creld1 is sufficient to promote NFATc1 dephosphorylation and translocation to the nucleus. Creld1 is contained in a joint protein complex with the regulatory subunit of calcineurin, CnB, thereby controlling calcineurin function. Localization of Creld1 at the endoplasmic reticulum (ER) is important to exert its action on calcineurin. By using Creld1KO mice, we demonstrate that Creld1 is essential for heart development. Creld1 function is required for the VEGF-dependent proliferation of endocardial cells by promoting the expression of NFATc1 target-genes. Collectively, our study identifies Creld1 as an important regulator of calcineurin/NFATc1 signaling.
Collapse
Affiliation(s)
- Elvira Mass
- LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, NRW 53115, Germany
| | - Dagmar Wachten
- Center of Advanced European Studies and Research (caesar), Minerva Research Group, Molecular Physiology, Ludwig-Erhard-Allee 2, 53175 Bonn, NRW 53115, Germany
| | - Anna C Aschenbrenner
- LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, NRW 53115, Germany
| | - André Voelzmann
- LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, NRW 53115, Germany
| | - Michael Hoch
- LIMES-Institute, Program Unit Development, Genetics and Molecular Physiology, Molecular Developmental Biology, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, NRW 53115, Germany.
| |
Collapse
|
36
|
Dahiya S, Liu Y, Nonnemacher MR, Dampier W, Wigdahl B. CCAAT enhancer binding protein and nuclear factor of activated T cells regulate HIV-1 LTR via a novel conserved downstream site in cells of the monocyte-macrophage lineage. PLoS One 2014; 9:e88116. [PMID: 24551078 PMCID: PMC3925103 DOI: 10.1371/journal.pone.0088116] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 01/03/2014] [Indexed: 12/11/2022] Open
Abstract
Transcriptional control of the human immunodeficiency virus type 1 (HIV-1) promoter, the long terminal repeat (LTR), is achieved by interactions with cis-acting elements present both upstream and downstream of the start site. In silico transcription factor binding analysis of the HIV-1 subtype B LTR sequences revealed a potential downstream CCAAT enhancer binding protein (C/EBP) binding site. This binding site (+158 to+172), designated DS3, was found to be conserved in 67% of 3,858 unique subtype B LTR sequences analyzed in terms of nucleotide sequence as well as physical location in the LTR. DS3 was found to be well represented in other subtypes as well. Interestingly, DS3 overlaps with a previously identified region that bind members of the nuclear factor of activated T cells (NFAT) family of proteins. NFATc2 exhibited a higher relative affinity for DS3 as compared with members of the C/EBP family (C/EBP α and β). DS3 was able to compete efficiently with the low-affinity upstream C/EBP binding site I with respect to C/EBP binding, suggesting utilization of both NFAT and C/EBP. Moreover, cyclosporine A treatment, which has been shown to prevent dephosphorylation and nuclear translocation of NFAT isoforms, resulted in enhanced C/EBPα binding. The interactions at DS3 were also validated in an integrated HIV-1 LTR in chronically infected U1 cells. A binding knockout of DS3 demonstrated reduced HIV-1 LTR-directed transcription under both basal and interleukin-6-stimulated conditions only in cells of the monocyte-macrophage lineage cells and not in cells of T-cell origin. Thus, the events at DS3 positively regulate the HIV-1 promoter in cells of the monocyte-macrophage lineage.
Collapse
Affiliation(s)
- Satinder Dahiya
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Yujie Liu
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michael R. Nonnemacher
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Will Dampier
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Brian Wigdahl
- Department of Microbiology and Immunology, and Center for Molecular Virology and Translational Neuroscience, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
37
|
Daniel C, Gerlach K, Väth M, Neurath MF, Weigmann B. Nuclear factor of activated T cells - a transcription factor family as critical regulator in lung and colon cancer. Int J Cancer 2013; 134:1767-75. [PMID: 23775822 DOI: 10.1002/ijc.28329] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 05/21/2013] [Accepted: 05/28/2013] [Indexed: 01/03/2023]
Abstract
Nuclear factor of activated T cells (NFAT) was first identified as a transcription factor which is activated upon T cell stimulation. Subsequent studies uncovered that a whole family of individual NFAT proteins exists with pleiotropic functions not only in immune but also in nonimmune cells. However, dysregulation of NFAT thereby favors malignant growth and cancer. Summarizing the recent advances in understanding how individual NFAT factors regulate the immune system, this review gives new insights into the critical role of NFAT in cancer development with special focus on inflammation-associated colorectal cancer.
Collapse
Affiliation(s)
- Carolin Daniel
- Institute of Diabetes Research, Helmholtz Zentrum Muenchen,German Research Center for Environmental Health (GmbH), Munich, Germany
| | | | | | | | | |
Collapse
|
38
|
Abdel-Salam E, Abdel-Meguid I, Korraa S. Assessment of immune function in Down syndrome patients. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2013. [DOI: 10.1016/j.ejmhg.2013.05.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
39
|
Ulrich JD, Kim MS, Houlihan PR, Shutov LP, Mohapatra DP, Strack S, Usachev YM. Distinct activation properties of the nuclear factor of activated T-cells (NFAT) isoforms NFATc3 and NFATc4 in neurons. J Biol Chem 2012; 287:37594-609. [PMID: 22977251 DOI: 10.1074/jbc.m112.365197] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The Ca(2+)/calcineurin-dependent transcription factor NFAT (nuclear factor of activated T-cells) is implicated in regulating dendritic and axonal development, synaptogenesis, and neuronal survival. Despite the increasing appreciation for the importance of NFAT-dependent transcription in the nervous system, the regulation and function of specific NFAT isoforms in neurons are poorly understood. Here, we compare the activation of NFATc3 and NFATc4 in hippocampal and dorsal root ganglion neurons following electrically evoked elevations of intracellular Ca(2+) concentration ([Ca(2+)](i)). We find that NFATc3 undergoes rapid dephosphorylation and nuclear translocation that are essentially complete within 20 min, although NFATc4 remains phosphorylated and localized to the cytosol, only exhibiting nuclear localization following prolonged (1-3 h) depolarization. Knocking down NFATc3, but not NFATc4, strongly diminished NFAT-mediated transcription induced by mild depolarization in neurons. By analyzing NFATc3/NFATc4 chimeras, we find that the region containing the serine-rich region-1 (SRR1) mildly affects initial NFAT translocation, although the region containing the serine-proline repeats is critical for determining the magnitude of NFAT activation and nuclear localization upon depolarization. Knockdown of glycogen synthase kinase 3β (GSK3β) significantly increased the depolarization-induced nuclear localization of NFATc4. In contrast, inhibition of p38 or mammalian target of rapamycin (mTOR) kinases had no significant effect on nuclear import of NFATc4. Thus, electrically evoked [Ca(2+)](i) elevation in neurons rapidly and strongly activates NFATc3, whereas activation of NFATc4 requires a coincident increase in [Ca(2+)](i) and suppression of GSK3β, with differences in the serine-proline-containing region giving rise to these distinct activation properties of NFATc3 and NFATc4.
Collapse
Affiliation(s)
- Jason D Ulrich
- Department of Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
40
|
Pont JNA, McArdle CA, López Bernal A. Oxytocin-stimulated NFAT transcriptional activation in human myometrial cells. Mol Endocrinol 2012; 26:1743-56. [PMID: 22902539 PMCID: PMC3507519 DOI: 10.1210/me.2012-1057] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oxytocin (OXT) is a peptide hormone that binds the OXT receptor on myometrial cells, initiating an intracellular signaling cascade, resulting in accumulation of intracellular calcium and smooth muscle contraction. In other systems, an elevation of intracellular Ca(2+) stimulates nuclear translocation of the transcription factor, nuclear factor of activated T cells (NFAT), which is transcriptionally active in arterial and ileal smooth muscle. Here we have investigated the role of NFAT in the mechanism of action of OXT. Human myometrial cells expressed all five NFAT isoforms (NFATC1-C4 and -5). Myometrial cells were transduced with a recombinant adenovirus expressing a NFATC1-EFP reporter, and a semi-automated imaging system was used to monitor effects of OXT on reporter localization in live cells. OXT induced a concentration-dependent nuclear translocation of NFATC1-EFP in a reversible manner, which was inhibited by OXT antagonists and calcineurin inhibitors. Pulsatile stimulation with OXT caused intermittent, pulse-frequency-dependent, nuclear translocation of NFATC1-EFP, which was more efficient than sustained stimulation. OXT induced nuclear translocation of endogenous NFAT that was transcriptionally active, because OXT stimulated activity of a NFAT-response element-luciferase reporter and induced calcineurin-NFAT dependent expression of RGS2, RCAN1, and PTGS2 (COX2) mRNA. Furthermore, OXT-dependent transcription was dependent on protein neosynthesis; cycloheximide abolished RGS2 transcription but augmented RCAN1 and COX2 transcriptional readouts. This study identifies a novel signaling mechanism within the myometrium, whereby calcineurin-NFAT signaling mediates OXT-induced transcriptional activity. Furthermore, we show NFATC1-EFP is responsive to pulses of OXT, a mechanism by which myometrial cells could decode OXT pulse frequency.
Collapse
Affiliation(s)
- Jason N A Pont
- Bristol University, School of Clinical Sciences, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, United Kingdom.
| | | | | |
Collapse
|
41
|
Arumugam A, Walsh SB, Xu J, Afaq F, Elmets CA, Athar M. Combined inhibition of p38 and Akt signaling pathways abrogates cyclosporine A-mediated pathogenesis of aggressive skin SCCs. Biochem Biophys Res Commun 2012; 425:177-81. [PMID: 22820192 DOI: 10.1016/j.bbrc.2012.07.062] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Accepted: 07/13/2012] [Indexed: 11/16/2022]
Abstract
Non-melanoma skin cancers (NMSCs) are the most common neoplasm in organ transplant recipients (OTRs). These cancers are more invasive and metastatic as compared to those developed in normal cohorts. Previously, we have shown that immunosuppressive drug, cyclosporine A (CsA) directly alters tumor phenotype of cutaneous squamous cell carcinomas (SCCs) by activating TGF-β and TAK1/TAB1 signaling pathways. Here, we identified novel molecular targets for the therapeutic intervention of these SCCs. We observed that combined blockade of Akt and p38 kinases-dependent signaling pathways in CsA-promoted human epidermoid carcinoma A431 xenograft tumors abrogated their growth by more than 90%. This diminution in tumor growth was accompanied by a significant decrease in proliferation and an increase in apoptosis. The residual tumors following the combined treatment with Akt inhibitor triciribine and p38 inhibitors SB-203580 showed significantly diminished expression of phosphorylated Akt and p38 and these tumors were less invasive and highly differentiated. Diminished tumor invasiveness was associated with the reduced epithelial-mesenchymal transition as ascertained by the enhanced E-cadherin and reduced vimentin and N-cadherin expression. Consistently, these tumors also manifested reduced MMP-2/9. The decreased p-Akt expression was accompanied by a significant reduction in p-mTOR. These data provide first important combinatorial pharmacological approach to block the pathogenesis of CsA-induced highly aggressive cutaneous neoplasm in OTRs.
Collapse
Affiliation(s)
- Aadithya Arumugam
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294-0019, USA
| | | | | | | | | | | |
Collapse
|
42
|
Learned Immunosuppression: Extinction, Renewal, and the Challenge of Reconsolidation. J Neuroimmune Pharmacol 2012; 8:180-8. [DOI: 10.1007/s11481-012-9388-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Accepted: 07/04/2012] [Indexed: 12/17/2022]
|
43
|
Zelante T, Fric J, Wong AYW, Ricciardi-Castagnoli P. Interleukin-2 production by dendritic cells and its immuno-regulatory functions. Front Immunol 2012; 3:161. [PMID: 22719740 PMCID: PMC3376408 DOI: 10.3389/fimmu.2012.00161] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 05/30/2012] [Indexed: 11/26/2022] Open
Abstract
Dendritic cells (DCs) are uniquely potent antigen presenting cells that acquire microbial products and prime adaptive immune responses against pathogens. Furthermore, DCs also play a key role in induction and maintenance of tolerance. Although numerous studies have assessed the diverse functions of DCs, many unanswered questions remain regarding the molecular mechanisms that DCs use to achieve immunoregulation. While not widely regarded as a significant provider of T-cell growth factors, DCs have previously been identified as a potential source of IL-2 cytokine. Recent research indicates that microbes are the most effective stimuli to trigger IL-2 production in DCs by activating the calcineurin/NFAT signaling pathway. Herein we describe recent insights into the production and function of IL-2 cytokine and IL-2 receptor in DCs early after stimulation through pattern recognition receptors. These findings clarify how DCs fine-tune effector and regulatory responses by modulating IL-2 production in both tolerance and immunity.
Collapse
Affiliation(s)
- Teresa Zelante
- Singapore Immunology Network, Agency for Science, Technology and Research Singapore
| | | | | | | |
Collapse
|
44
|
Fric J, Lim CXF, Koh EGL, Hofmann B, Chen J, Tay HS, Isa SABM, Mortellaro A, Ruedl C, Ricciardi-Castagnoli P. Calcineurin/NFAT signalling inhibits myeloid haematopoiesis. EMBO Mol Med 2012; 4:269-82. [PMID: 22311511 PMCID: PMC3376854 DOI: 10.1002/emmm.201100207] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2011] [Revised: 12/13/2011] [Accepted: 12/15/2011] [Indexed: 01/09/2023] Open
Abstract
Nuclear factor of activated T cells (NFAT) comprises a family of transcription factors that regulate T cell development, activation and differentiation. NFAT signalling can also mediate granulocyte and dendritic cell (DC) activation, but it is unknown whether NFAT influences their development from progenitors. Here, we report a novel role for calcineurin/NFAT signalling as a negative regulator of myeloid haematopoiesis. Reconstituting lethally irradiated mice with haematopoietic stem cells expressing an NFAT-inhibitory peptide resulted in enhanced development of the myeloid compartment. Culturing bone marrow cells in media supplemented with Flt3-L in the presence of the calcineurin/NFAT inhibitor Cyclosporin A increased numbers of differentiated DC. Global gene expression analysis of untreated DC and NFAT-inhibited DC revealed differential expression of transcripts that regulate cell cycle and apoptosis. In conclusion, these results provide evidence that calcineurin/NFAT signalling negatively regulates myeloid lineage development. The finding that inhibition of NFAT enhances myeloid development provides a novel insight into understanding how the treatment with drugs targeting calcineurin/NFAT signalling influence the homeostasis of the innate immune system.
Collapse
Affiliation(s)
- Jan Fric
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| | - Clarice X F Lim
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| | - Esther G L Koh
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| | - Benjamin Hofmann
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| | - Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| | - Hock Soon Tay
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| | | | - Alessandra Mortellaro
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological UniversitySingapore, Singapore
| | - Paola Ricciardi-Castagnoli
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR)Singapore, Singapore
| |
Collapse
|
45
|
Choi JM, Sohn JH, Park TY, Park JW, Lee SK. Cell permeable NFAT inhibitory peptide Sim-2-VIVIT inhibits T-cell activation and alleviates allergic airway inflammation and hyper-responsiveness. Immunol Lett 2012; 143:170-6. [PMID: 22342853 DOI: 10.1016/j.imlet.2012.01.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 11/28/2022]
Abstract
Nuclear factor of activated T cells (NFAT) is an important transcription factor for the production of interleukin (IL)-2 upon T-cell receptor (TcR) signaling. Therefore, inhibition of the NFAT-carcineurin pathway is an important target for inflammatory disease inhibition and graft rejection. A novel cell permeable peptide (CPP), Sim-2, has been identified from a human transcription factor, and Sim-2-CPP conjugated to β-galactosidase or EGFP protein was efficiently delivered into cells in vitro and in vivo. A cell permeable form of the NFAT inhibitory peptide VIVIT (Sim-2-VIVIT) was synthesized and showed inhibitory effects on human CD4 or CD8 T-cell activation through NFAT transcriptional activity suppression and IL-2 inhibition. Intranasal administration of the Sim-2-VIVIT peptide in an ovalbumin (OVA)-induced murine asthma model alleviated peribronchial and perivascular infiltration of inflammatory cells in the lung and caused airway remodeling and airway hyper-responsiveness. These results suggest that cell permeable Sim-2-VIVIT peptide has clinical potential as an immunosuppressive agent for inflammatory diseases.
Collapse
Affiliation(s)
- Je-Min Choi
- Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Republic of Korea.
| | | | | | | | | |
Collapse
|
46
|
Walsh SB, Xu J, Xu H, Kurundkar AR, Maheshwari A, Grizzle WE, Timares L, Huang CC, Kopelovich L, Elmets CA, Athar M. Cyclosporine a mediates pathogenesis of aggressive cutaneous squamous cell carcinoma by augmenting epithelial-mesenchymal transition: role of TGFβ signaling pathway. Mol Carcinog 2011; 50:516-27. [PMID: 21308804 PMCID: PMC3110643 DOI: 10.1002/mc.20744] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2010] [Revised: 12/01/2010] [Accepted: 01/04/2011] [Indexed: 01/07/2023]
Abstract
Organ transplant recipients (OTRs) develop multiple aggressive and metastatic non-melanoma skin cancers (NMSCs). Yet, the underlying mechanism remains elusive. Employing a variety of immune-compromised murine models, immunoblotting, immunohistochemical and immunofluorescence techniques, we show that human squamous xenograft tumors in nude mice grow faster and become significantly larger in size following treatment with the immunosuppressive drug, cyclosporine A (CsA). Re-injected tumor cells isolated from CsA-treated xenografts continued to form larger tumors in nude mice than those from vehicle-controls and retained the CsA-signatures of calcineurin signaling inhibition. Similar results were obtained when these tumors were grown in SCID-beige mice or in immuno-competent mice inoculated with syngeinic tumor cells. Consistently, tumors in the CsA group manifested enhanced cellular proliferation and decreased apoptosis. Tumors in CsA-treated animals also showed an augmented epithelial-mesenchymal transition (EMT) characterized by an increased expression of fibronectin, α-SMA, vimentin, N-cadherin, MMP-9/-2, snail and twist with a concomitant decrease in E-cadherin. CsA-treated xenograft tumors manifested increased TGFβ1 expression and TGFβ-dependent signaling characterized by increased nuclear p-Smad 2/3. Our data demonstrate that CsA alters the phenotype of skin SCCs to an invasive and aggressive tumor-type by enhancing expression of proteins regulating EMT acting through the TGFβ1 signaling pathway providing at least one unique mechanism by which multiple aggressive and metastatic NMSCs develop in OTRs.
Collapse
Affiliation(s)
- Stephanie B. Walsh
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Jianmin Xu
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Hui Xu
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Ashish R. Kurundkar
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, 35233
| | - Akhil Maheshwari
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, Alabama, 35233
| | - William E. Grizzle
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Laura Timares
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Conway C. Huang
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Levy Kopelovich
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Craig A. Elmets
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Mohammad Athar
- Department of Dermatology and Skin Diseases Research Center, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| |
Collapse
|
47
|
Opposing roles of FoxP1 and Nfat3 in transcriptional control of cardiomyocyte hypertrophy. Mol Cell Biol 2011; 31:3068-80. [PMID: 21606195 DOI: 10.1128/mcb.00925-10] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cardiac homeostasis is maintained by a balance of growth-promoting and growth-modulating factors. Sustained elevation of calcium signaling can induce cardiac hypertrophy through activation of Nfat family transcription factors. FoxP family transcription factors are known to interact with Nfat proteins and to modulate their transcriptional activities in lymphocytes. We investigated FoxP1 interaction with Nfat3 (Nfatc4) and their effects on transcription of hypertrophy-associated genes in neonatal rat cardiomyocytes. FoxP1-Nfat3 complexes were visualized using bimolecular fluorescence complementation (BiFC) analysis. Calcineurin activation induced FoxP1-Nfat3 BiFC complex formation. Amino acid substitutions in the predicted interaction interface inhibited it. FoxP1 repressed hypertrophy-associated genes (Myh7, Rcan1, Cx43, Anf, and Bnp) and counteracted their activation by constitutively nuclear Nfat3 (cnNfat3). In contrast, FoxP1 activated genes that maintain normal heart functions (Myh6 and p57Kip2) and cnNfat3 counteracted their activation by FoxP1. Amino acid substitutions in FoxP1 or cnNfat3 that inhibited their interaction abrogated the activation of hypertrophy-associated gene transcription by cnNfat3 and the repression of these genes by FoxP1. FoxP1 and Nfat3 co-occupied the promoter regions of hypertrophy-associated genes in neonatal and adult heart tissue. FoxP1 counteracted hypertrophic cardiomyocyte growth, and connexin 43 mislocalization caused by cnNfat3 expression. These data suggest that the opposing transcriptional activities of FoxP1 and Nfat3 maintain cardiomyocyte homeostasis.
Collapse
|
48
|
Abstract
Cyclosporin A and tacrolimus have been used as immunosuppressive agents initially in organ transplantation after their discovery, and are also used for treatment of the autoimmune disease, providing an excellent therapeutic effect. These agents act targeting on intracellular phosphatase calcineurin (CN), and subsequently inhibit activation of nuclear factor of activated T cells (NFAT), a key regulator of stimulation-dependent gene activation. The CN-NFAT system is involved not only in the immunoregulation including activation and development of helper T cells, regulatory T cells and NKT cells, but in a variety of cellular and developmental events other than immune system. CN inhibitors also affect organs outside of immune system leading to adverse effects, including nephrotoxicity and glucose intolerance. We review recent findings in CN-NFAT system, as well as development of potential CN inhibitors.
Collapse
Affiliation(s)
- Yoshiharu Amasaki
- The Center for Rheumatic Diseases/Tonan Hospital, KKR Sapporo Medical Center, Japan
| |
Collapse
|
49
|
Li H, Rao A, Hogan PG. Interaction of calcineurin with substrates and targeting proteins. Trends Cell Biol 2011; 21:91-103. [PMID: 21115349 PMCID: PMC3244350 DOI: 10.1016/j.tcb.2010.09.011] [Citation(s) in RCA: 282] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2010] [Revised: 09/15/2010] [Accepted: 09/27/2010] [Indexed: 12/18/2022]
Abstract
Calcineurin is a calcium activated protein phosphatase with a major role in calcium signaling in diverse cells and organs and clinical importance as the target of the immunosuppressive drugs cyclosporin A and tacrolimus (FK506). Cell biology studies have focused mainly on the role of calcineurin in transcriptional signaling. Calcium entry in response to extracellular stimuli results in calcineurin activation, and signal transmission from the cytosol into the nucleus through dephosphorylation and nuclear translocation of the transcription factor nuclear factor of activated T cells (NFAT). This initiates a cascade of transcriptional events involved in physiological and developmental processes. Molecular analyses of the calcineurin-NFAT interaction have been extended recently to encompass the interaction of calcineurin with other substrates, targeting proteins and regulators of calcineurin activity. These studies have increased our understanding of how this essential calcium activated enzyme orchestrates intracellular events in cooperation with other signaling pathways, and have suggested a link between altered calcineurin signaling and the developmental anomalies of Down syndrome.
Collapse
Affiliation(s)
- Huiming Li
- Immune Disease Institute and Program in Cellular and Molecular Medicine, Children's Hospital, Boston, MA 02115, USA
| | | | | |
Collapse
|
50
|
Nardozzi JD, Lott K, Cingolani G. Phosphorylation meets nuclear import: a review. Cell Commun Signal 2010; 8:32. [PMID: 21182795 PMCID: PMC3022542 DOI: 10.1186/1478-811x-8-32] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/23/2010] [Indexed: 12/18/2022] Open
Abstract
Phosphorylation is the most common and pleiotropic modification in biology, which plays a vital role in regulating and finely tuning a multitude of biological pathways. Transport across the nuclear envelope is also an essential cellular function and is intimately linked to many degeneration processes that lead to disease. It is therefore not surprising that phosphorylation of cargos trafficking between the cytoplasm and nucleus is emerging as an important step to regulate nuclear availability, which directly affects gene expression, cell growth and proliferation. However, the literature on phosphorylation of nucleocytoplasmic trafficking cargos is often confusing. Phosphorylation, and its mirror process dephosphorylation, has been shown to have opposite and often contradictory effects on the ability of cargos to be transported across the nuclear envelope. Without a clear connection between attachment of a phosphate moiety and biological response, it is difficult to fully understand and predict how phosphorylation regulates nucleocytoplasmic trafficking. In this review, we will recapitulate clue findings in the field and provide some general rules on how reversible phosphorylation can affect the nuclear-cytoplasmic localization of substrates. This is only now beginning to emerge as a key regulatory step in biology.
Collapse
Affiliation(s)
- Jonathan D Nardozzi
- Dept, of Biochemistry and Molecular Biology, Thomas Jefferson University, 233 South 10th Street, Philadelphia, PA 19107, USA.
| | | | | |
Collapse
|