1
|
Lazo PA. VRK2 kinase pathogenic pathways in cancer and neurological diseases. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119949. [PMID: 40187568 DOI: 10.1016/j.bbamcr.2025.119949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/07/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The VRK2 ser-thr kinase, belonging to the dark kinome, is implicated in the pathogenesis of cancer progression, neurological and psychiatric diseases. The VRK2 gene codes for two isoforms. The main isoform (VRK2A) is mainly located in the cytoplasm, and anchored to different types of membranes, such as the endoplasmic reticulum, mitochondria and nuclear envelope. The VRK2A isoform interacts with signaling modules assembled on scaffold proteins such as JIP1 or KSR1, forming stable complexes and blocking the activation of regulatory signaling pathways by altering their intracellular localization and the balance among them. VRK2 regulates apoptosis, nuclear membrane organization, immune responses, and Cajal bodies. Wild-type VRK2 is overexpressed in tumors and contributes to cancer development. In cells and tumors with low levels of nuclear VRK1, VRK2 generates by alternative splicing a shorter isoform (VRK2B) that lacks the C-terminal hydrophobic tail and permits its relocation to nuclei. Furthermore, rare VRK2 gene variants are associated with different neurological or psychiatric diseases such as schizophrenia, epilepsy, bipolar disorder, depression, autism, circadian clock alterations and insomnia, but their pathogenic mechanism is unknown. These diseases are a likely consequence of an altered balance among different signaling pathways that are regulated by VRK2.
Collapse
Affiliation(s)
- Pedro A Lazo
- Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Salamanca, 37007 Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Hospital Universitario de Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
2
|
Solomon RN, Pittner NA, McCoy JR, Warwick PA, McBride JW. Cell signaling in Ehrlichia infection and cancer: Parallels in pathogenesis. Front Cell Infect Microbiol 2025; 15:1539847. [PMID: 40028182 PMCID: PMC11868041 DOI: 10.3389/fcimb.2025.1539847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Ehrlichia chaffeensis (E. chaffeensis) has recently emerged as an intracellular bacterial pathogen with sophisticated survival mechanisms that include repurposing evolutionarily conserved eukaryotic cell signaling pathways for immune evasion. E. chaffeensis exploits four major developmental signaling pathways (Wnt, Notch, Hedgehog, and Hippo) using short linear motif (SLiM) ligand mimicry to initiate signaling cascades. Dysregulation of these major signaling pathways leading to unchecked cell survival is implicated in various diseases, most notably cancer. E. chaffeensis exploits Wnt, Notch, Hedgehog and Hippo signaling pathways to inhibit apoptosis and co-opt other cellular functions to promote infection. This review will explore the signaling pathways exploited during Ehrlichia infection and the new discoveries that have illuminated this interesting example of the cell signaling convergence in cellular infection and cancer biology.
Collapse
Affiliation(s)
- Regina N. Solomon
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Nicholas A. Pittner
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jaclyn R. McCoy
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Paityn A. Warwick
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jere W. McBride
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
- Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, TX, United States
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, United States
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
3
|
Gazzeri S, Zubchuk N, Montaudon E, Nemati F, Huot-Marchand S, Berardi G, Pucciarelli A, Dib Y, Nerini D, Oddou C, Pezet M, David-Boudet L, Ardin C, de Fraipont F, Maraver A, Girard N, Decaudin D, Toffart AC, Eymin B. PPP3CB overexpression mediates EGFR TKI resistance in lung tumors via calcineurin/MEK/ERK signaling. Life Sci Alliance 2024; 7:e202402873. [PMID: 39353739 PMCID: PMC11447527 DOI: 10.26508/lsa.202402873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024] Open
Abstract
Despite initial high response rates to first-line EGFR TKI, all non-small-cell lung cancer (NSCLC) with EGFR-activating mutation will ultimately develop resistance to treatment. Identification of resistance mechanisms is critical to adapt treatment and improve patient outcomes. Here, we show that a PPP3CB transcript that encodes full-length catalytic subunit 2B of calcineurin accumulates in EGFR-mutant NSCLC cells with acquired resistance against different EGFR TKIs and in post-progression biopsies of NSCLC patients treated with EGFR TKIs. Neutralization of PPP3CB by siRNA or inactivation of calcineurin by cyclosporin A induces apoptosis in resistant cells treated with EGFR TKIs. Mechanistically, EGFR TKIs increase the cytosolic level of calcium and trigger activation of a calcineurin/MEK/ERK pathway that prevents apoptosis. Combining EGFR, calcineurin, and MEK inhibitors overcomes resistance to EGFR TKI in both in vitro and in vivo models. Our results identify PPP3CB overexpression as a new mechanism of acquired resistance to EGFR TKIs, and provide a promising therapeutic approach for NSCLC patients that progress under TKI treatment.
Collapse
Affiliation(s)
- Sylvie Gazzeri
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Nadiia Zubchuk
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Elodie Montaudon
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Fariba Nemati
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Sarah Huot-Marchand
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Giulia Berardi
- Department of Pneumology and Physiology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Amelie Pucciarelli
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Yassir Dib
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Dylan Nerini
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| | - Christiane Oddou
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team Epigenetics, Immunity, Metabolism, Cell Signaling and Cancer, Institute for Advanced Biosciences, Grenoble, France
| | - Mylène Pezet
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Platform MicroCell, Institute for Advanced Biosciences, Grenoble, France
| | - Laurence David-Boudet
- Department of Cytology and Pathology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Camille Ardin
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
- Department of Pneumology and Physiology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Florence de Fraipont
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
- Medical Unit of Molecular Genetic (Hereditary Diseases and Oncology), Grenoble-Alpes University Hospital, Grenoble, France
| | - Antonio Maraver
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194-ICM-Université de Montpellier, Montpellier, France
| | - Nicolas Girard
- Institut du Thorax Curie-Montsouris, Institut Curie, Paris, France
| | - Didier Decaudin
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Anne-Claire Toffart
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
- Department of Pneumology and Physiology, Grenoble-Alpes University Hospital, Grenoble, France
| | - Beatrice Eymin
- University Grenoble Alpes, Inserm U1209, CNRS UMR 5309, Team RNA Splicing, Cell Signaling and Response to Therapy, Institute for Advanced Biosciences, Grenoble, France
| |
Collapse
|
4
|
Hang Q, Zuo S, Yang Y, Wang Y, Li C, Li W, Guo J, Hou S, Huang H. USP33 is an integrin α6 deubiquitinase and promotes esophageal squamous cell carcinoma cell migration and metastasis. J Cancer Res Clin Oncol 2024; 150:511. [PMID: 39589547 PMCID: PMC11599434 DOI: 10.1007/s00432-024-06041-5] [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: 10/22/2024] [Accepted: 11/16/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE The deubiquitinating enzymes (DUBs) have been linked to cancer initiation and progression. Although ubiquitin-specific protease 33 (USP33) represents a significant factor in regulating various tumor cell behaviors, its specific biological functions and precise mechanisms in esophageal squamous cell carcinoma (ESCC) progression remain unclear. METHODS The expressions of USP33 mRNA in GEO databases, clinical ESCC samples, and USP33 protein were analyzed using bioinformatics, RT-PCR, and immunohistochemistry, respectively. Using Kaplan-Meier survival curves, the log-rank test was used to determine the cumulative survival rate. Western blotting was used to determine indicated protein expression. The cell biological functions were evaluated by cell growth assay, transwell, cell adhesion, and cell spreading assay, respectively. The interaction between USP33 and integrins was detected by immunoprecipitation, and the deubiquitination was performed by deubiquitination assay. The metastatic ability was checked by tail vein injection. RESULTS A significant positive correlation was found between USP33 expression and clinical TNM stage, T classification, and poor prognosis in patients with ESCC. USP33 promoted laminin-dependent adhesion, spreading, and migration of ESCC cells but not their proliferation. Mechanistically, USP33 mediates cell migration through binding, deubiquinating, and stabilizing integrin α6. USP33 knockdown could inhibit ESCC cell migration and metastasis majorly through integrin α6. CONCLUSION This study reveals a novel mechanism of USP33 in promoting laminin-dependent ESCC cell migration and metastasis through integrin α6, suggesting that USP33 may be a promising target for treating ESCC.
Collapse
Affiliation(s)
- Qinglei Hang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Jiangsu Provincial Innovation and Practice Base for Postdoctors, Suining People's Hospital, Affiliated Hospital of Xuzhou Medical University, Suining, Jiangsu Province, 221200, China.
| | - Shiying Zuo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Yawen Yang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
| | - Yuanzhi Wang
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Caimin Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Wenqian Li
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Jingya Guo
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China
| | - Sicong Hou
- Department of Clinical Medicine, Medical College, Key laboratory of Jiangsu province university for Nucleic Acid & Cell Fate Manipulation, Yangzhou University, Yangzhou, Jiangsu Province, 225009, China.
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu Province, 225001, China.
| | - Haifeng Huang
- Department of Laboratory Medicine, The First People's Hospital of Yancheng, Yancheng, Jiangsu Province, 224006, China.
- Department of Laboratory Medicine, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng, Jiangsu Province, 224006, China.
| |
Collapse
|
5
|
Chinbold B, Kwon HM, Park R. TonEBP degradation is essential for microtubule nucleation and regrowth. Biochem Biophys Res Commun 2024; 734:150791. [PMID: 39369538 DOI: 10.1016/j.bbrc.2024.150791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
TonEBP is a transcription factor known for its involvement in diverse physiological processes, including cell cycle, mitosis, migration, and cytoskeletal remodeling. However, the role of TonEBP regarding microtubules, essential structural components of the cytoskeleton, remains unclear. Here, we introduce a novel function for TonEBP as a regulator of microtubule nucleation. Our initial findings reveal that Nocodazole, a well-known microtubule depolymerizing agent, significantly downregulates the protein level of TonEBP. Moreover, microtubule depolymerization induces rapid degradation of TonEBP through the ubiquitin-proteasome pathway. Knockdown of TonEBP results in enhanced microtubule polymerization and regrowth, whereas the presence of TonEBP impairs microtubule nucleation. Collectively, our data suggest that TonEBP negatively regulates microtubule nucleation.
Collapse
Affiliation(s)
- Batchingis Chinbold
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyug Moo Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
6
|
Michl M, Taverna F, Woischke C, Li P, Klauschen F, Kirchner T, Heinemann V, von Bergwelt-Baildon M, Stahler A, Herold TM, Jurinovic V, Engel J, Kumbrink J, Neumann J. Identification of a gene expression signature associated with brain metastasis in colorectal cancer. Clin Transl Oncol 2024; 26:1886-1895. [PMID: 38558282 PMCID: PMC11249597 DOI: 10.1007/s12094-024-03408-5] [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: 01/31/2024] [Accepted: 02/11/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Brain metastasis (BM) in colorectal cancer (CRC) is a rare event with poor prognosis. Apart from (K)RAS status and lung and bone metastasis no biomarkers exist to identify patients at risk. This study aimed to identify a gene expression signature associated with colorectal BM. METHODS Three patient groups were formed: 1. CRC with brain metastasis (BRA), 2. exclusive liver metastasis (HEP) and, 3. non-metastatic disease (M0). RNA was extracted from primary tumors and mRNA expression was measured using a NanoString Panel (770 genes). Expression was confirmed by qPCR in a validation cohort. Statistical analyses including multivariate logistic regression followed by receiver operating characteristic (ROC) analysis were performed. RESULTS EMILIN3, MTA1, SV2B, TMPRSS6, ACVR1C, NFAT5 and SMC3 were differentially expressed in BRA and HEP/M0 groups. In the validation cohort, differential NFAT5, ACVR1C and SMC3 expressions were confirmed. BRA patients showed highest NFAT5 levels compared to HEP/M0 groups (global p = 0.02). High ACVR1C expression was observed more frequently in the BRA group (42.9%) than in HEP (0%) and M0 (7.1%) groups (global p = 0.01). High SMC3 expressions were only detectable in the BRA group (global p = 0.003). Only patients with BM showed a combined high expression of NFAT5, ACVR1C or SMC3 as well as of all three genes. ROC analysis revealed a good prediction of brain metastasis by the three genes (area under the curve (AUC) = 0.78). CONCLUSIONS The NFAT5, ACVR1C and SMC3 gene expression signature is associated with colorectal BM. Future studies should further investigate the importance of this biomarker signature.
Collapse
Affiliation(s)
- Marlies Michl
- Department of Medicine III, University Hospital, Ludwig-Maximilian-University of Munich, Munich, Germany
- Department of Haematology and Oncology, Comprehensive Cancer Center Munich, Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Francesco Taverna
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Christine Woischke
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Pan Li
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Frederick Klauschen
- Department of Haematology and Oncology, Comprehensive Cancer Center Munich, Ludwig-Maximilian-University of Munich, Munich, Germany
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Thomas Kirchner
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Volker Heinemann
- Department of Medicine III, University Hospital, Ludwig-Maximilian-University of Munich, Munich, Germany
- Department of Haematology and Oncology, Comprehensive Cancer Center Munich, Ludwig-Maximilian-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig-Maximilian-University of Munich, Munich, Germany
- Department of Haematology and Oncology, Comprehensive Cancer Center Munich, Ludwig-Maximilian-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Arndt Stahler
- Department of Hematology, Oncology, and Tumorimmunology, Corporate Member of Freie Universitaet Berlin and Humbolt-Universitaet zu Berlin, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Tobias Marcus Herold
- Department of Medicine III, University Hospital, Ludwig-Maximilian-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Vindi Jurinovic
- Institute for Medical Information Processing, Biometry and Epidemiology, Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Jutta Engel
- Munich Cancer Registry (MCR), Ludwig-Maximilian-University of Munich, Munich, Germany
| | - Jörg Kumbrink
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany.
| | - Jens Neumann
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilian-University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Centre (DKFZ), Heidelberg, Germany
| |
Collapse
|
7
|
Chinbold B, Kwon HM, Park R. TonEBP inhibits ciliogenesis by controlling aurora kinase A and regulating centriolar satellite integrity. Cell Commun Signal 2024; 22:348. [PMID: 38961488 PMCID: PMC11221002 DOI: 10.1186/s12964-024-01721-8] [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: 02/19/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Primary cilia on the surface of eukaryotic cells serve as sensory antennas for the reception and transmission in various cell signaling pathways. They are dynamic organelles that rapidly form during differentiation and cell cycle exit. Defects in these organelles cause a group of wide-ranging disorders called ciliopathies. Tonicity-responsive enhancer-binding protein (TonEBP) is a pleiotropic stress protein that mediates various physiological and pathological cellular responses. TonEBP is well-known for its role in adaptation to a hypertonic environment, to which primary cilia have been reported to contribute. Furthermore, TonEBP is involved in a wide variety of other signaling pathways, such as Sonic Hedgehog and WNT signaling, that promote primary ciliogenesis, suggesting a possible regulatory role. However, the functional relationship between TonEBP and primary ciliary formation remains unclear. METHODS TonEBP siRNAs and TonEBP-mCherry plasmids were used to examine their effects on cell ciliation rates, assembly and disassembly processes, and regulators. Serum starvation was used as a condition to induce ciliogenesis. RESULTS We identified a novel pericentriolar localization for TonEBP. The results showed that TonEBP depletion facilitates the formation of primary cilia, whereas its overexpression results in fewer ciliated cells. Moreover, TonEBP controlled the expression and activity of aurora kinase A, a major negative regulator of ciliogenesis. Additionally, TonEBP overexpression inhibited the loss of CP110 from the mother centrioles during the early stages of primary cilia assembly. Finally, TonEBP regulated the localization of PCM1 and AZI1, which are necessary for primary cilia formation. CONCLUSIONS This study proposes a novel role for TonEBP as a pericentriolar protein that regulates the integrity of centriolar satellite components. This regulation has shown to have a negative effect on ciliogenesis. Investigations into cilium assembly and disassembly processes suggest that TonEBP acts upstream of the aurora kinase A - histone deacetylase 6 signaling pathway and affects basal body formation to control ciliogenesis. Taken together, our data proposes previously uncharacterized regulation of primary cilia assembly by TonEBP.
Collapse
Affiliation(s)
- Batchingis Chinbold
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Hyug Moo Kwon
- School of Life Sciences, Ulsan National Institute of Science and Technology, Ulsan, Republic of Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea.
| |
Collapse
|
8
|
Miskin RP, DiPersio CM. Roles for epithelial integrin α3β1 in regulation of the microenvironment during normal and pathological tissue remodeling. Am J Physiol Cell Physiol 2024; 326:C1308-C1319. [PMID: 38497112 PMCID: PMC11371326 DOI: 10.1152/ajpcell.00128.2024] [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: 02/23/2024] [Revised: 03/08/2024] [Accepted: 03/08/2024] [Indexed: 03/19/2024]
Abstract
Integrin receptors for the extracellular matrix activate intracellular signaling pathways that are critical for tissue development, homeostasis, and regeneration/repair, and their loss or dysregulation contributes to many developmental defects and tissue pathologies. This review will focus on tissue remodeling roles for integrin α3β1, a receptor for laminins found in the basement membranes (BMs) that underlie epithelial cell layers. As a paradigm, we will discuss literature that supports a role for α3β1 in promoting ability of epidermal keratinocytes to modify their tissue microenvironment during skin development, wound healing, or tumorigenesis. Preclinical and clinical studies have shown that this role depends largely on ability of α3β1 to govern the keratinocyte's repertoire of secreted proteins, or the "secretome," including 1) matrix proteins and proteases involved in matrix remodeling and 2) paracrine-acting growth factors/cytokines that stimulate other cells with important tissue remodeling functions (e.g., endothelial cells, fibroblasts, inflammatory cells). Moreover, α3β1 signaling controls gene expression that helps epithelial cells carry out these functions, including genes that encode secreted matrix proteins, proteases, growth factors, or cytokines. We will review what is known about α3β1-dependent gene regulation through both transcription and posttranscriptional mRNA stability. Regarding the latter, we will discuss examples of α3β1-dependent alternative splicing (AS) or alternative polyadenylation (APA) that prevents inclusion of cis-acting mRNA sequences that would otherwise target the transcript for degradation via nonsense-mediated decay or destabilizing AU-rich elements (AREs) in the 3'-untranslated region (3'-UTR). Finally, we will discuss prospects and anticipated challenges of exploiting α3β1 as a clinical target for the treatment of cancer or wound healing.
Collapse
Affiliation(s)
| | - C Michael DiPersio
- Department of Surgery, Albany Medical College, Albany, New York, United States
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, New York, United States
| |
Collapse
|
9
|
Dashti NK, Perret R, Balzer B, Naous R, Michal M, Dermawan JK, Antonescu CR. Vascular Neoplasms With NFATC1/C2 Gene Alterations : Expanding the Clinicopathologic and Molecular Characteristics of a Distinct Entity. Am J Surg Pathol 2024; 48:487-496. [PMID: 38189436 PMCID: PMC11591551 DOI: 10.1097/pas.0000000000002175] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Despite significant advances in their molecular pathogenesis, skeletal vascular tumors remain diagnostically challenging due to their aggressive radiologic appearance and significant morphologic overlap. Within the epithelioid category and at the benign end of the spectrum, recurrent FOS/FOSB fusions have defined most epithelioid hemangiomas, distinguishing them from epithelioid hemangioendothelioma and angiosarcoma. More recently, the presence of EWSR1/FUS :: NFATC1/2 fusions emerged as the genetic hallmark of a novel group of unusual vascular proliferations, often displaying epithelioid morphology, with alternating vasoformative and solid growth, variable atypia, reminiscent of composite hemangioendothelioma. In this study, we further our understanding and morphologic spectrum of NFATC -fusion positive vascular neoplasms by describing 9 new cases, including soft tissue locations and novel fusion partners. Combining with the initial cohort of 5 cases, a total of 14 patients were analyzed, showing slight female predilection and an age range of 10 to 66 (mean 42 y). Twelve patients had solitary lesions, while 2 had multifocal polyostotic (pelvic bones) disease. Overall, 12 lesions were intra-osseous and 2 in soft tissue. By targeted RNA Fusion panels or FISH, there were 6 cases of EWSR1::NFATC1 , 4 EWSR1::NFATC2 , 2 FUS::NFATC2 , 1 EWSR1 rearrangement, and 1 with a novel FABP4::NFATC2 fusion. Follow-up was available in 4 patients. One patient experienced 2 local recurrences, 11 and 15 years postdiagnosis, and one patient experienced progressive disease despite multimodality treatment (curettings, embolization, radiation) over 3 years. In summary, our extended investigation confirms that NFATC -related fusions define a distinct group of vascular neoplasms with variable architecture, epithelioid phenotype, and cytologic atypia, commonly located in the bone, occasionally multifocal and with potential for local recurrence and aggressive behavior but no metastatic potential. Molecular analysis is recommended in diagnostically challenging cases with atypical histology to exclude malignancy.
Collapse
Affiliation(s)
- Nooshin K. Dashti
- Department of Pathology and Laboratory Medicine Dartmouth Health, Lebanon, NH, USA
- Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Raul Perret
- Department of Biopathology, Institut Bergonié, Comprehensive Cancer Center, Bordeaux, France
- Bordeaux Institute of Oncology, BRIC, INSERM, Bordeaux University, Bergonié Institute, Bordeaux, France
| | | | - Rana Naous
- University of Pittsburgh Medical Center, Shadyside, Pittsburgh, PA, USA
| | - Michael Michal
- Biopticka Laboratory, Pilsen, Czech Republic
- Department of Pathology, Charles University, Faculty of Medicine in Plzen, Czech Republic
| | - Josephine K. Dermawan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Cristina R. Antonescu
- Department of Pathology and Lab Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
10
|
Ge L, Rui Y, Wang C, Wu Y, Wang H, Wang J. The RNA m 6A reader IGF2BP3 regulates NFAT1/IRF1 axis-mediated anti-tumor activity in gastric cancer. Cell Death Dis 2024; 15:192. [PMID: 38448411 PMCID: PMC10917814 DOI: 10.1038/s41419-024-06566-0] [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: 11/07/2022] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
N6-methyladenosine (m6A) and its associated reader protein insulin like growth factor 2 mRNA binding protein 3 (IGF2BP3) are involved in tumor initiation and progression via regulating RNA metabolism. This study aims to investigate the biological function and clinical significance of IGF2BP3 in gastric cancer (GC). The clinical significance of IGF2BP3 was evaluated using tumor related databases and clinical tissues. The biological role and molecular mechanism of IGF2BP3 in GC progression were investigated by multi-omics analysis including Ribosome sequence (Ribo-seq), RNA sequence (RNA-seq) and m6A sequence (m6A-seq) combined with gain- and loss- of function experiments. IGF2BP3 expression is significantly elevated in GC tissues and associated with poor prognosis of GC patients. Knockdown of IGF2BP3 significantly weakens the migration and clonogenic ability, promotes the apoptosis, inhibits translation, and suppresses in vitro growth and progression of GC cells. Mechanistically, IGF2BP3 regulates the mRNA stability and translation of the nuclear factor of activated T cells 1(NFAT1) in a m6A dependent manner. Then NFAT1 induced by IGF2BP3 acts as a transcription factor (TF) to negatively regulates the promoter activities of interferon regulatory factor 1 (IRF1) to inhibit its expression. Inhibition of IGF2BP3-induced expression of IRF1 activates interferon (IFN) signaling pathway and then exerts its anti-tumor effect. Elevated IGF2BP3 promotes in vivo and in vitro GC progression via regulation of NFAT1/IRF1 pathways. Targeted inhibition of IGF2BP3 might be a potential therapeutic approach for GC treatment.
Collapse
Affiliation(s)
- Lichen Ge
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Yalan Rui
- Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Cheng Wang
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Yingmin Wu
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Department of Physiology, School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550009, China
| | - Hongsheng Wang
- Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Junjun Wang
- Department of Clinical Laboratory, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
11
|
Song HJ, Kim YH, Choi HN, Kim T, Kim SJ, Kang MW, Lee SD. TonEBP/NFAT5 expression is associated with cisplatin resistance and migration in macrophage-induced A549 cells. BMC Mol Cell Biol 2024; 25:6. [PMID: 38438872 PMCID: PMC10913585 DOI: 10.1186/s12860-024-00502-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/26/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Macrophages promote angiogenesis, metastasis, and drug resistance in several cancers. Similarly, TonEBP/NFAT5 induces metastasis in renal carcinoma and colon cancer cells. However, the role of this transcription factor and that of macrophages in lung cancer cells remains unclear. Therefore, this study investigated the effects of macrophages and TonEBP/NFAT5 expression on cisplatin resistance and migration in A549 lung adenocarcinoma cells. RESULTS A549 cells were cultured alone or indirectly co-cultured with THP-1-derived macrophages using a transwell culture chamber. Cisplatin-induced cell death was markedly decreased and migration increased in co-cultured A549 cells. Macrophage-conditioned media (CM) showed a similar effect on drug resistance and migration. Cisplatin-induced apoptosis, DNA fragmentation, and cleaved apoptotic proteins PARP and caspase-3 were markedly reduced in macrophage CM-induced A549 cells. Here, ERK, p38, JNK, and NF-κB activities were increased by macrophage CM. Furthermore, the proteins involved in cisplatin resistance and cancer cell migration were identified using specific inhibitors of each protein. ERK and NF-κB inhibition considerably reduced cisplatin resistance. The increase in macrophage CM-induced migration was partially reduced by treatment with ERK, JNK, and NF-κB inhibitors. TonEBP/NFAT5 expression was increased by macrophages, resulting in increased cisplatin resistance, cell migration, and invasion. Moreover, RNAi-mediated knockdown of TonEBP/NFAT5 reduced cisplatin resistance, migration, and invasion in macrophage CM-induced A549 cells. CONCLUSIONS These findings demonstrate that paracrine factors secreted from macrophages can change A549 cells, resulting in the induction of drug resistance against cisplatin and migration. In addition, the TonEBP/NFAT5 ratio, increased by macrophages, is an important regulator of the malignant transformation of cells.
Collapse
Affiliation(s)
- Hee Ju Song
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Young Hwan Kim
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Han Na Choi
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Taehee Kim
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Soo Jin Kim
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Min Woong Kang
- Department of thoracic surgery, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Sang Do Lee
- Department of Physiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
| |
Collapse
|
12
|
Machado I, Llombart-Bosch A, Charville GW, Navarro S, Domínguez Franjo MP, Bridge JA, Linos K. Sarcomas with EWSR1::Non-ETS Fusion (EWSR1::NFATC2 and EWSR1::PATZ1). Surg Pathol Clin 2024; 17:31-55. [PMID: 38278606 DOI: 10.1016/j.path.2023.07.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
The wide application of increasingly advanced molecular studies in routine clinical practice has allowed a detailed, albeit still incomplete, genetic subclassification of undifferentiated round cell sarcomas. The WHO classification continues to include provisional molecular entities, whose clinicopathologic features are in the early stages of evolution. This review focuses on the clinicopathologic, molecular, and prognostic features of undifferentiated round cell sarcomas with EWSR1/FUS::NFATC2 or EWSR1::PATZ1 fusions. Classic histopathologic findings, uncommon variations, and diagnostic pitfalls are addressed, along with the utility of recently developed immunohistochemical and molecular markers.
Collapse
Affiliation(s)
- Isidro Machado
- Pathology Department, Instituto Valenciano de Oncología, Valencia, Spain; Patologika Laboratory, Hospital Quiron-Salud, Valencia, Spain; Pathology Department, University of Valencia, Valencia, Spain.
| | - Antonio Llombart-Bosch
- Pathology Department, university of Valencia, Spain and Cancer CIBER (CIBERONC), Madrid, Spain
| | | | - Samuel Navarro
- Pathology Department, university of Valencia, Spain and Cancer CIBER (CIBERONC), Madrid, Spain
| | | | - Julia A Bridge
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA; Division of Molecular Pathology, ProPath, Dallas, TX, USA
| | - Konstantinos Linos
- Department of Pathology & Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
13
|
Zhang Y, Xu H, Pi S, Tan H, Huang B, Chen Y. The prognostic and immunological role of FKBP1A in an integrated muti-omics cancers analysis, especially lung cancer. J Cancer Res Clin Oncol 2023; 149:16589-16608. [PMID: 37715833 DOI: 10.1007/s00432-023-05362-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 08/28/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND AND AIM FKBP1A, a gene encoding the FK506-binding protein 1A, has emerged as a significant player in cancer progression and prognosis. This study aimed to comprehensively investigate the multifaceted role of FKBP1A in cancer, focusing on its differential expression patterns, prognostic implications, genetic alterations, and associations with the tumor microenvironment. METHODS AND RESULTS Using large-scale datasets, including GTEx, TCGA, HPA, and cBioPortal, we analyzed FKBP1A expression across normal tissues and various cancer types. Our findings revealed that FKBP1A exhibited aberrant upregulation in most human cancers, making it a potential biomarker for malignancy. Moreover, FKBP1A expression correlated with poor overall survival, disease-specific survival, disease-free interval, and progression-free interval in several cancers, indicating its prognostic significance. Genetic alteration analysis showed that FKBP1A gene amplification was prevalent, particularly in ovarian cancer. Furthermore, FKBP1A expression was associated with tumor mutational burden and microsatellite instability, highlighting its potential involvement in tumor-immune response. Notably, FKBP1A expression positively correlated with stromal and immune cell scores, suggesting its role in shaping the tumor microenvironment. Additionally, according to the functional enrichment analysis, experimental validation in lung adenocarcinoma confirmed the role of FKBP1A through the regulation of EGFR signaling by apoptosis, which is consistent with drug sensitivity analysis to some extent. CONCLUSION In conclusion, FKBP1A exhibits differential expression in cancer, serves as a prognostic indicator, undergoes genetic alterations, and influences the tumor-immune microenvironment. These findings shed light on the multifaceted role of FKBP1A in cancer development and progression, suggesting its potential as a therapeutic target and guidance of clinical drugs selection, and provide valuable insights into patient prognosis for interventions based on pharmaceuticals.
Collapse
Affiliation(s)
- Yi Zhang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Haifeng Xu
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Sainan Pi
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Huiqian Tan
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Bihui Huang
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Youpeng Chen
- Department of Infectious Diseases, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
14
|
Hammad AS, Yu F, Al-Hamaq J, Horgen FD, Machaca K. STIM1 signals through NFAT1 independently of Orai1 and SOCE to regulate breast cancer cell migration. Cell Calcium 2023; 114:102779. [PMID: 37399784 DOI: 10.1016/j.ceca.2023.102779] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/25/2023] [Accepted: 06/27/2023] [Indexed: 07/05/2023]
Abstract
Store-operated calcium entry (SOCE) contributes to several physiological and pathological conditions including transcription, secretion, immunodeficiencies, and cancer. SOCE has been shown to be important for breast cancer cell migration where knockdown of SOCE components (STIM1 or Orai1) decreases cancer metastasis. Here we show unexpectedly that complete knockout of STIM1 (STIM1-KO) using gene editing in metastatic MDA-MB-231 breast cancer cells results in faster migration and enhanced invasion capacity. In contrast, Orai1-KO cells, which have similar levels of SOCE inhibition as STIM1-KO, migrate slower than the parental cell line. This shows that the enhanced migration phenotype of STIM1-KO cells is not due to the loss of Ca2+ entry through SOCE, rather it involves transcriptional remodeling as elucidated by RNA-seq analyses. Interestingly, NFAT1 is significantly downregulated in STIM1-KO cells and overexpression of NFAT1 reversed the enhanced migration of STIM1-KO cells. STIM1 knockout in other breast cancer cells, independent of their metastatic potential, also enhanced cell migration while reducing NFAT1 expression. These data argue that in breast cancer cells STIM1 modulates NFAT1 expression and cell migration independently of its role in SOCE.
Collapse
Affiliation(s)
- Ayat S Hammad
- College of Health and Life Science, Hamad bin Khalifa University, Doha, Qatar; Calcium Signaling Group, Research Department, Weill Cornell Medicine, Doha, Qatar
| | - Fang Yu
- Calcium Signaling Group, Research Department, Weill Cornell Medicine, Doha, Qatar; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States
| | - Jawaher Al-Hamaq
- Calcium Signaling Group, Research Department, Weill Cornell Medicine, Doha, Qatar
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Honolulu, Hawaii, United States
| | - Khaled Machaca
- Calcium Signaling Group, Research Department, Weill Cornell Medicine, Doha, Qatar; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, United States.
| |
Collapse
|
15
|
Zhen H, Yao Y, Yang H. SAFB2 Inhibits the Progression of Breast Cancer by Suppressing the Wnt/β-Catenin Signaling Pathway via NFAT5. Mol Biotechnol 2023; 65:1465-1475. [PMID: 36652182 DOI: 10.1007/s12033-022-00649-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023]
Abstract
Aberrant scaffold attachment factor-B2 (SAFB2) expression is associated with several malignant tumors. In this study, we investigated how SAFB2 worked in the process of breast cancer as well as the underlying mechanism. Quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting analysis were used to investigate the expression of SAFB2 and nuclear factor of activated T cells 5 (NFAT5). Cellular proliferative ability was detected with cell counting kit 8 (CCK8), colony formation and 5-Ethynyl-2'-deoxyuridine (EdU) staining assays. Cell apoptosis was measured via flow cytometry and western blotting analysis. Wound healing, transwell assays, and western blotting analysis were executed to estimate cell migration and invasion. The relationship between SAFB2 and NFAT5 was verified by RNA immunoprecipitation (RIP) assay and NFAT5 mRNA stability was examined with actinomycin (Act) D assay. Western blotting analysis also tested the expression of Wnt/β-catenin signaling-associated proteins. As a result, SAFB2 was downregulated in breast cancer cell lines, while NFAT5 was highly expressed in most breast cancer cell lines. Overexpression of SAFB2 suppressed the proliferation, migration, and invasion while exacerbated the apoptosis of breast cancer cells. SAFB2 interacted with NFAT5 mRNA and declined the stability of NFAT5 mRNA. Overexpression of NFAT5 counteracted anti-proliferative, anti-metastatic and pro-apoptotic effects of SAFB2 in breast cancer cells. Mechanistically, SAFB2 overexpression inhibited the Wnt/β-catenin signaling pathway, while this effect was partially eliminated by NFAT5. Collectively, SAFB2 hindered breast cancer development and inactivated Wnt/β-catenin signaling via regulation of NFAT5, suggesting that SAFB2 might be a promising therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Huifen Zhen
- Department of Breast Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan, 030032, Shanxi Province, China
| | - Yarong Yao
- Department of Breast Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan, 030032, Shanxi Province, China
| | - Haibo Yang
- Department of Breast Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, 99 Longcheng Street, Taiyuan, 030032, Shanxi Province, China.
| |
Collapse
|
16
|
Liu C, Lin J, Yang H, Li N, Tang L, Neumann D, Ding X, Zhu L. NFAT5 Restricts Bovine Herpesvirus 1 Productive Infection in MDBK Cell Cultures. Microbiol Spectr 2023; 11:e0011723. [PMID: 37227295 PMCID: PMC10434061 DOI: 10.1128/spectrum.00117-23] [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: 01/08/2023] [Accepted: 05/04/2023] [Indexed: 05/26/2023] Open
Abstract
Bovine herpesvirus 1 (BoHV-1), an important bovine viral pathogen, causes severe disease in the upper respiratory tract and reproductive system. Tonicity-responsive enhancer-binding protein (TonEBP), also known as nuclear factor of activated T cells 5 (NFAT5), is a pleiotropic stress protein involved in a range of cellular processes. In this study, we showed that the knockdown of NFAT5 by siRNA increased BoHV-1 productive infection and overexpression of NFAT5 via plasmid transfection decreased virus production in bovine kidney (MDBK) cells. Virus productive infection at later stages significantly increased transcription of NFAT5 but not appreciably alter measurable NFAT5 protein levels. Virus infection relocalized NFAT5 protein and decreased the cytosol accumulation. Importantly, we found a subset of NFAT5 resides in mitochondria, and virus infection led to the depletion of mitochondrial NFAT5. In addition to full-length NFAT5, another two isoforms with distinct molecular weights were exclusively detected in the nucleus, where the accumulation was differentially affected following virus infection. In addition, virus infection differentially altered mRNA levels of PGK1, SMIT, and BGT-1, the canonical downstream targets regulated by NFAT5. Taken together, NFAT5 is a potential host factor that restricts BoHV-1 productive infection, and virus infection hijacks NFAT5 signaling transduction by relocalization of NFAT5 molecules in cytoplasm, nucleus, and mitochondria, as well as altered expression of its downstream targets. IMPORTANCE Accumulating studies have revealed that NFAT5 regulates disease development due to infection of numerous viruses, underlying the importance of the host factor in virus pathogenesis. Here, we report that NFAT5 has capacity to restrict BoHV-1 productive infection in vitro. And virus productive infection at later stages may alter NFAT5 signaling pathway as observed by relocalization of NFAT5 protein, reduced accumulation of NFAT5 in cytosol, and differential expression of NFAT5 downstream targets. Importantly, for the first time, we found that a subset of NFAT5 resides in mitochondria, implying that NFAT5 may regulate mitochondrial functions, which will extend our knowledge on NFAT5 biological activities. Moreover, we found two NFAT5 isoforms with distinct molecular weights were exclusively detected in the nucleus, where the accumulation was differentially affected following virus infection, representing a novel regulation mechanism on NFAT5 function in response to BoHV-1infection.
Collapse
Affiliation(s)
- Chang Liu
- College of Life Sciences, Hebei University, Baoding, China
| | - Jiayu Lin
- College of Life Sciences, Hebei University, Baoding, China
| | - Hao Yang
- College of Life Sciences, Hebei University, Baoding, China
| | - Ningxi Li
- College of Life Sciences, Hebei University, Baoding, China
| | - Linke Tang
- College of Life Sciences, Hebei University, Baoding, China
| | - Donna Neumann
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Xiuyan Ding
- College of Life Sciences, Hebei University, Baoding, China
| | - Liqian Zhu
- College of Life Sciences, Hebei University, Baoding, China
- Key Laboratory of Microbial Diversity Research and Application of Hebei Province, College of Life Science, Hebei University, Baoding, China
| |
Collapse
|
17
|
Wang X, Sun Q, Wang W, Liu B, Gu Y, Chen L. Decoding key cell sub-populations and molecular alterations in glioblastoma at recurrence by single-cell analysis. Acta Neuropathol Commun 2023; 11:125. [PMID: 37525259 PMCID: PMC10391841 DOI: 10.1186/s40478-023-01613-x] [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: 04/27/2023] [Accepted: 07/01/2023] [Indexed: 08/02/2023] Open
Abstract
Glioblastoma (GBM) is the most frequent malignant brain tumor, the relapse of which is unavoidable following standard treatment. However, the effective treatment for recurrent GBM is lacking, necessitating the understanding of key mechanisms driving tumor recurrence and the identification of new targets for intervention. Here, we integrated single-cell RNA-sequencing data spanning 36 patient-matched primary and recurrent GBM (pGBM and rGBM) specimens, with 6 longitudinal GBM spatial transcriptomics to explore molecular alterations at recurrence, with each cell type characterized in parallel. Genes involved in extracellular matrix (ECM) organization are preferentially enriched in rGBM cells, and MAFK is highlighted as a potential regulator. Notably, we uncover a unique subpopulation of GBM cells that is much less detected in pGBM and highly expresses ECM and mesenchyme related genes, suggesting it may contribute to the molecular transition of rGBM. Further regulatory network analysis reveals that transcription factors, such as NFATC4 and activator protein 1 members, may function as hub regulators. All non-tumor cells alter their specific sets of genes as well and certain subgroups of myeloid cells appear to be physically associated with the mesenchyme-like GBM subpopulation. Altogether, our study provides new insights into the molecular understanding of GBM relapse and candidate targets for rGBM treatment.
Collapse
Affiliation(s)
- Xin Wang
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
- BGI Research, Hangzhou, 310030 China
| | - Qian Sun
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Weiwen Wang
- China National GeneBank, BGI Research, Shenzhen, 518120 China
| | - Baohui Liu
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| | - Ying Gu
- BGI Research, Hangzhou, 310030 China
- BGI Research, Shenzhen, 518083 China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083 China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049 China
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, 430072, China
| |
Collapse
|
18
|
Li Y, Gao Z, Wang Y, Pang B, Zhang B, Hu R, Wang Y, Liu C, Zhang X, Yang J, Mei M, Wang Y, Zhou X, Li M, Ren Y. Lysine methylation promotes NFAT5 activation and determines temozolomide efficacy in glioblastoma. Nat Commun 2023; 14:4062. [PMID: 37429858 DOI: 10.1038/s41467-023-39845-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 06/30/2023] [Indexed: 07/12/2023] Open
Abstract
Temozolomide (TMZ) therapy offers minimal clinical benefits in patients with glioblastoma multiforme (GBM) with high EGFR activity, underscoring the need for effective combination therapy. Here, we show that tonicity-responsive enhancer binding protein (NFAT5) lysine methylation, is a determinant of TMZ response. Mechanistically, EGFR activation induces phosphorylated EZH2 (Ser21) binding and triggers NFAT5 methylation at K668. Methylation prevents NFAT5 cytoplasm interaction with E3 ligase TRAF6, thus blocks NFAT5 lysosomal degradation and cytosol localization restriction, which was mediated by TRAF6 induced K63-linked ubiquitination, resulting in NFAT5 protein stabilization, nuclear accumulation and activation. Methylated NFAT5 leads to the upregulation of MGMT, a transcriptional target of NFAT5, which is responsible for unfavorable TMZ response. Inhibition of NFAT5 K668 methylation improved TMZ efficacy in orthotopic xenografts and patient-derived xenografts (PDX) models. Notably, NFAT5 K668 methylation levels are elevated in TMZ-refractory specimens and confer poor prognosis. Our findings suggest targeting NFAT5 methylation is a promising therapeutic strategy to improve TMZ response in tumors with EGFR activation.
Collapse
Affiliation(s)
- Yatian Li
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhenyue Gao
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuhong Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Bo Pang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Binbin Zhang
- Department of Neuro-oncology, Tianjin Huanhu Hospital, Tianjin, China
| | - Ruxin Hu
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuqing Wang
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chao Liu
- Department of Maxillofacial and Otorhinolaryngology Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China
| | - Xuebin Zhang
- Department of Pathology, Tianjin Huanhu Hospital, Tianjin, China
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mei Mei
- Department of Cell Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Yongzhi Wang
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngology Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Cancer Institute, National Clinical Research Center of Cancer, Tianjin, China.
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Yu Ren
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
19
|
Merhi M, Ahmad F, Taib N, Inchakalody V, Uddin S, Shablak A, Dermime S. The complex network of transcription factors, immune checkpoint inhibitors and stemness features in colorectal cancer: A recent update. Semin Cancer Biol 2023; 89:1-17. [PMID: 36621515 DOI: 10.1016/j.semcancer.2023.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Cancer immunity is regulated by several mechanisms that include co-stimulatory and/or co-inhibitory molecules known as immune checkpoints expressed by the immune cells. In colorectal cancer (CRC), CTLA-4, LAG3, TIM-3 and PD-1 are the major co-inhibitory checkpoints involved in tumor development and progression. On the other hand, the deregulation of transcription factors and cancer stem cells activity plays a major role in the development of drug resistance and in the spread of metastatic disease in CRC. In this review, we describe how the modulation of such transcription factors affects the response of CRC to therapies. We also focus on the role of cancer stem cells in tumor metastasis and chemoresistance and discuss both preclinical and clinical approaches for targeting stem cells to prevent their tumorigenic effect. Finally, we provide an update on the clinical applications of immune checkpoint inhibitors in CRC and discuss the regulatory effects of transcription factors on the expression of the immune inhibitory checkpoints with specific focus on the PD-1 and PD-L1 molecules.
Collapse
Affiliation(s)
- Maysaloun Merhi
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Fareed Ahmad
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Nassiba Taib
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Varghese Inchakalody
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Alaaeldin Shablak
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
20
|
Ren F, Zhu K, Wang Y, Zhou F, Pang S, Chen L. Proliferation, apoptosis and invasion of human lung cancer cells are associated with NFATc1. Exp Ther Med 2022; 25:49. [PMID: 36588811 PMCID: PMC9780516 DOI: 10.3892/etm.2022.11748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 08/31/2022] [Indexed: 12/12/2022] Open
Abstract
The expression of nuclear factor of activated T cells c1 (NFATc1) is closely associated with the progression of numerous types of cancer. When NFATc1 expression becomes dysregulated in some types of cancer, this alteration can promote malignant transformation and thereby progression of cancer. NFATc1 expression has been demonstrated to be upregulated in lung cancer cells. This suggests that knockdown of NFATc1 in lung cancer cells may be a therapeutic marker for the treatment of cancer. In the present study, the effects of NFATc1 on the proliferation, apoptosis, invasion and migration of NCI-H1299 and A549 lung cancer cell lines were explored. Lentivirus infection was used to establish a cell model of NFATc1 knockdown in A549 and NCI-H1299 lung cancer cells. Reverse transcription-quantitative PCR was subsequently performed to detect NFATc1 expression in these human lung cancer cells. MTT, wound healing, colony formation and Transwell invasion assays, and flow cytometry were then performed to measure the proliferation, invasion, apoptosis and cell cycle of the cells. Finally, western blot analysis was performed to investigate the mechanism underlying the involvement of NFATc1 in these processes. NFATc1 knockdown was found to significantly inhibit the proliferation, clone formation, migration and invasion of the cells. Furthermore, the cell cycle was arrested at the G1 phase and the expression levels of the target proteins located downstream in the signaling pathway, namely CDK4, c-Myc, ERK, p38 and N-cadherin, were decreased. Following NFATc1 knockdown, the percentages of apoptotic cells were increased, and the expression levels of Bax, cleaved caspase-3 and E-cadherin were also increased. Taken together, the results of the present study suggested that NFATc1 serves an oncogenic role in lung cancer. In terms of the underlying mechanism, NFATc1 promoted the proliferation of lung cancer cells by inhibiting the MAPK and epithelial-to-mesenchymal transition signaling pathways, suggesting that NFATc1 may be a novel target for therapeutic intervention for the treatment of lung cancer.
Collapse
Affiliation(s)
- Fenghai Ren
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Kaibin Zhu
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Yanbo Wang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Fucheng Zhou
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Sainan Pang
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China
| | - Lantao Chen
- Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, P.R. China,Correspondence to: Dr Lantao Chen, Department of Thoracic Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
21
|
Human complete NFAT1 deficiency causes a triad of joint contractures, osteochondromas, and B-cell malignancy. Blood 2022; 140:1858-1874. [PMID: 35789258 DOI: 10.1182/blood.2022015674] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 06/20/2022] [Indexed: 11/20/2022] Open
Abstract
The discovery of humans with monogenic disorders has a rich history of generating new insights into biology. Here we report the first human identified with complete deficiency of nuclear factor of activated T cells 1 (NFAT1). NFAT1, encoded by NFATC2, mediates calcium-calcineurin signals that drive cell activation, proliferation, and survival. The patient is homozygous for a damaging germline NFATC2 variant (c.2023_2026delTACC; p.Tyr675Thrfs∗18) and presented with joint contractures, osteochondromas, and recurrent B-cell lymphoma. Absence of NFAT1 protein in chondrocytes caused enrichment in prosurvival and inflammatory genes. Systematic single-cell-omic analyses in PBMCs revealed an environment that promotes lymphomagenesis with accumulation of naïve B cells (enriched for oncogenic signatures MYC and JAK1), exhausted CD4+ T cells, impaired T follicular helper cells, and aberrant CD8+ T cells. This work highlights the pleiotropic role of human NFAT1, will empower the diagnosis of additional patients with NFAT1 deficiency, and further defines the detrimental effects associated with long-term use of calcineurin inhibitors.
Collapse
|
22
|
Coillard L, Guaddachi F, Ralu M, Brabencova E, Garbar C, Bensussan A, Le Bras M, Lehmann-Che J, Jauliac S. The NFAT3/RERG Complex in Luminal Breast Cancers Is Required to Inhibit Cell Invasion and May Be Correlated With an Absence of Axillary Lymph Nodes Colonization. Front Oncol 2022; 12:804868. [PMID: 35847954 PMCID: PMC9280138 DOI: 10.3389/fonc.2022.804868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 05/25/2022] [Indexed: 11/18/2022] Open
Abstract
Luminal breast cancers represent 70% of newly diagnosed breast cancers per annum and have a relatively good prognosis compared with triple-negative breast cancers. Luminal tumors that are responsive to hormonal therapy are particularly associated with a favorable prognosis. Nonetheless, the absolute number of metastatic relapses in luminal cancers is larger than in triple-negative breast cancers. A better understanding of the biology of luminal cancers, control of metastases formation, and identification of predictive markers of their evolution are therefore still necessary. In this context, we previously disclosed the key role of NFAT3 in regulating luminal breast cancer invasion. We have now identified a specific inhibitory region, in the C-terminal part of NFAT3, required for the inhibition of invasion of the human luminal breast cancer cell line T-47D. Indeed, we showed that this 85 amino acid C-terminal region acts as a dominant negative form of NFAT3 and that its overexpression in the T-47D cell line led to increased cell invasion. Mechanistically, we have revealed that this region of NFAT3 interacts with the small Ras GTPase RERG (RAS like estrogen regulated growth inhibitor) and shown that RERG expression is required for NFAT3 to impede T-47D cell invasion. We have validated the association of NFAT3 with RERG in human luminal breast cancer tissues. We have shown an increase of the quantity of the NFAT3/RERG complexes in patients without axillary lymph node colonization and therefore proposed that the detection of this complex may be a non-invasive marker of axillary lymph node colonization.
Collapse
Affiliation(s)
- Lucie Coillard
- Université de Paris, Research Saint Louis Institute (IRSL), Institut National de la Santé et de la Recherche Médicale, Human Immunology Pathophysiology Immunotherapy (INSERM HIPI) U976, Paris, France
| | - Frédéric Guaddachi
- Université de Paris, Research Saint Louis Institute (IRSL), Institut National de la Santé et de la Recherche Médicale, Human Immunology Pathophysiology Immunotherapy (INSERM HIPI) U976, Paris, France
| | - Maëlle Ralu
- Université de Paris, Research Saint Louis Institute (IRSL), Institut National de la Santé et de la Recherche Médicale, Human Immunology Pathophysiology Immunotherapy (INSERM HIPI) U976, Paris, France
| | - Eva Brabencova
- Department of Biopathology, Centre Régional de Lutte Contre le Cancer, Institut Godinot, Reims, France
| | - Christian Garbar
- Department of Biopathology, Centre Régional de Lutte Contre le Cancer, Institut Godinot, Reims, France
| | - Armand Bensussan
- Université de Paris, Research Saint Louis Institute (IRSL), Institut National de la Santé et de la Recherche Médicale, Human Immunology Pathophysiology Immunotherapy (INSERM HIPI) U976, Paris, France
| | - Morgane Le Bras
- Université de Paris, Research Saint Louis Institute (IRSL), Institut National de la Santé et de la Recherche Médicale, Human Immunology Pathophysiology Immunotherapy (INSERM HIPI) U976, Paris, France
| | - Jacqueline Lehmann-Che
- Université de Paris, Research Saint Louis Institute (IRSL), Institut National de la Santé et de la Recherche Médicale, Human Immunology Pathophysiology Immunotherapy (INSERM HIPI) U976, Paris, France
- Molecular Oncology Unit, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Louis, Paris, France
| | - Sébastien Jauliac
- Université de Paris, Research Saint Louis Institute (IRSL), Institut National de la Santé et de la Recherche Médicale, Human Immunology Pathophysiology Immunotherapy (INSERM HIPI) U976, Paris, France
| |
Collapse
|
23
|
Sánchez-Morales A, Biçer A, Panagiotopoulos V, Crecente-Garcia S, Benaiges C, Bayod S, Luís Hernández J, Busqué F, Matsoukas MT, Pérez-Riba M, Alibés R. Design and synthesis of a novel non peptide CN-NFATc signaling inhibitor for tumor suppression in triple negative breast cancer. Eur J Med Chem 2022; 238:114514. [DOI: 10.1016/j.ejmech.2022.114514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 11/04/2022]
|
24
|
Teo T, Kasirzadeh S, Albrecht H, Sykes MJ, Yang Y, Wang S. An Overview of CDK3 in Cancer: Clinical Significance and Pharmacological Implications. Pharmacol Res 2022; 180:106249. [DOI: 10.1016/j.phrs.2022.106249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022]
|
25
|
Pan-cancer analysis of microRNA expression profiles highlights microRNAs enriched in normal body cells as effective suppressors of multiple tumor types: A study based on TCGA database. PLoS One 2022; 17:e0267291. [PMID: 35476804 PMCID: PMC9045663 DOI: 10.1371/journal.pone.0267291] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/05/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are frequently deregulated in various types of cancer. While antisense oligonucleotides are used to block oncomiRs, delivery of tumour-suppressive miRNAs holds great potential as a potent anti-cancer strategy. Here, we aim to determine, and functionally analyse, miRNAs that are lowly expressed in various types of tumour but abundantly expressed in multiple normal tissues. METHODS The miRNA sequencing data of 14 cancer types were downloaded from the TCGA dataset. Significant differences in miRNA expression between tumor and normal samples were calculated using limma package (R programming). An adjusted p value < 0.05 was used to compare normal versus tumor miRNA expression profiles. The predicted gene targets were obtained using TargetScan, miRanda, and miRDB and then subjected to gene ontology analysis using Enrichr. Only GO terms with an adjusted p < 0.05 were considered statistically significant. All data from wet-lab experiments (cell viability assays and flow cytometry) were expressed as means ± SEM, and their differences were analyzed using GraphPad Prism software (Student's t test, p < 0.05). RESULTS By compiling all publicly available miRNA profiling data from The Cancer Genome Atlas (TCGA) Pan-Cancer Project, we reveal a small set of tumour-suppressing miRNAs (which we designate as 'normomiRs') that are highly expressed in 14 types of normal tissues but poorly expressed in corresponding tumour tissues. Interestingly, muscle-enriched miRNAs (e.g. miR-133a/b and miR-206) and miRNAs from DLK1-DIO3 locus (e.g. miR-381 and miR-411) constitute a large fraction of the normomiRs. Moreover, we define that the CCCGU motif is absent in the oncomiRs' seed sequences but present in a fraction of tumour-suppressive miRNAs. Finally, the gain of function of candidate normomiRs across several cancer cell types indicates that miR-206 and miR-381 exert the most potent inhibition on multiple cancer types in vitro. CONCLUSION Our results reveal a pan-cancer set of tumour-suppressing miRNAs and highlight the potential of miRNA-replacement therapies for targeting multiple types of tumour.
Collapse
|
26
|
Rammohan M, Harris E, Bhansali RS, Zhao E, Li LS, Crispino JD. The chromosome 21 kinase DYRK1A: emerging roles in cancer biology and potential as a therapeutic target. Oncogene 2022; 41:2003-2011. [PMID: 35220406 PMCID: PMC8977259 DOI: 10.1038/s41388-022-02245-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/02/2022] [Accepted: 02/11/2022] [Indexed: 11/09/2022]
Abstract
Dual-specificity tyrosine phosphorylation-regulated kinase 1 A (DYRK1A) is a serine/threonine kinase that belongs to the DYRK family of proteins, a subgroup of the evolutionarily conserved CMGC protein kinase superfamily. Due to its localization on chromosome 21, the biological significance of DYRK1A was initially characterized in the pathogenesis of Down syndrome (DS) and related neurodegenerative diseases. However, increasing evidence has demonstrated a prominent role in cancer through its ability to regulate biologic processes including cell cycle progression, DNA damage repair, transcription, ubiquitination, tyrosine kinase activity, and cancer stem cell maintenance. DYRK1A has been identified as both an oncogene and tumor suppressor in different models, underscoring the importance of cellular context in its function. Here, we review mechanistic contributions of DYRK1A to cancer biology and its role as a potential therapeutic target.
Collapse
Affiliation(s)
- Malini Rammohan
- Driskill Graduate Program in Life Sciences, Northwestern University, Chicago, IL, USA
| | - Ethan Harris
- University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rahul S Bhansali
- Department of Medicine, Division of Hematology/Oncology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Emily Zhao
- Weinberg College of Arts and Sciences, Northwestern University, Chicago, IL, USA
| | - Loretta S Li
- Molecular and Translational Cancer Biology Program, Stanley Manne Children's Research Institute, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
- Department of Pediatrics, Division of Hematology, Oncology, and Stem Cell Transplantation, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - John D Crispino
- Division of Experimental Hematology, Department of Hematology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
27
|
Liu W, Ren D, Xiong W, Jin X, Zhu L. A novel FBW7/NFAT1 axis regulates cancer immunity in sunitinib-resistant renal cancer by inducing PD-L1 expression. J Exp Clin Cancer Res 2022; 41:38. [PMID: 35081978 PMCID: PMC8790872 DOI: 10.1186/s13046-022-02253-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/12/2022] [Indexed: 01/11/2023] Open
Abstract
Background Tyrosine kinase inhibitors (TKIs) alone and in combination with immune checkpoint inhibitors (ICIs) have been shown to be beneficial for the survival of metastatic renal cell carcinoma (mRCC) patients, but resistance to targeted therapy and ICIs is common in the clinic. Understanding the underlying mechanism is critical for further prolonging the survival of renal cancer patients. Nuclear factor of activated T cell 1 (NFAT1) is expressed in immune and nonimmune cells, and the dysregulation of NFAT1 contributes to the progression of various type of malignant tumors. However, the specific role of NFAT1 in RCC is elusive. As a regulator of the immune response, we would like to systemically study the role of NFAT1 in RCC. Methods TCGA-KIRC dataset analysis, Western blot analysis and RT-qPCR analysis was used to determine the clinic-pathological characteristic of NFAT1 in RCC. CCK-8 assays, colony formation assays and xenograft assays were performed to examine the biological role of NFAT1 in renal cancer cells. RNA-seq analysis was used to examine the pathways changed after NFAT1 silencing. ChIP-qPCR, coimmunoprecipitation analysis, Western blot analysis and RT-qPCR analysis were applied to explore the mechanism by NAFT1 was regulated in the renal cancer cells. Results In our study, we found that NFAT1 was abnormally overexpressed in RCC and that NFAT1 overexpression was associated with an unfavorable prognosis. Then, we showed that NFAT1 enhanced tumor growth and regulated the immune response by increasing PD-L1 expression in RCC. In addition, we demonstrated that NFAT1 was stabilized in sunitinib-resistant RCC via hyperactivation of the PI3K/AKT/GSK-3β signaling pathway. Furthermore, our study indicated that downregulation of the expression of FBW7, which promotes NFAT1 degradation, was induced by FOXA1 and SETD2 in sunitinib-resistant RCC. Finally, FBW7 was found to contribute to modulating the immune response in RCC. Conclusions Our data reveal a novel role for the FBW7/NFAT1 axis in the RCC response to TKIs and ICIs. NFAT1 and its associated signaling pathway might be therapeutic targets for RCC treatment, especially when combined with ICIs and/or TKIs. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02253-0.
Collapse
|
28
|
Kim W, Kim HJ, Trinh NT, Yeon HR, Kim JH, Choi IA, Kim HA, Jung JY, Lee KE. Association between nuclear factor of activated T cells C2 polymorphisms and treatment response in rheumatoid arthritis patients receiving tumor necrosis factor-alpha inhibitors. Pharmacogenet Genomics 2022; 32:10-15. [PMID: 34320607 DOI: 10.1097/fpc.0000000000000446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Nuclear factor of activated T cells C2 (NFATC2) is known as a member of the transcription family and enhances tumor necrosis factor-alpha (TNF-α) synthesis in human T cells at the gene transcription level. Although NFATC2 has a potential role in rheumatoid arthritis (RA) progression and treatment, no study has investigated the association between NFATC2 gene polymorphisms and response status in RA patients receiving TNF-α inhibitors. This study aimed to examine the effects of polymorphisms in NFATC2, a TNF-α transcription factor, on response to TNF-α inhibitors. METHODS This prospective observational study was performed in two centers. Seven single nucleotide polymorphisms (SNPs) were investigated. Good responders were defined as patients with disease activity score (DAS)28 ≤3.2 after 6 months of treatment. Logistic regression analyses were used to investigate the association between genetic polymorphisms and response to the treatment. To test the model's goodness of fit, a Hosmer-Lemeshow test was performed. RESULTS This study included 98 patients, among whom 46 showed favorable responses to the treatment. Patients with hypertension revealed an approximately three-fold lower response to TNF-α inhibitors compared to those without hypertension (23.5 vs. 76.5%; P = 0.049). After adjusting for covariates, C allele carriers of NFATC2 rs3787186 exhibited approximately three-fold lower rates of treatment response compared to those with TT genotype (P = 0.037). The Hosmer-Lemeshow test showed that the fitness of the multivariable analysis model was satisfactory (χ2 = 9.745; 8 degrees of freedom; P = 0.283). CONCLUSION This study suggested an association between the C allele of rs3787186 and treatment response in RA patients receiving TNF-α inhibitors.
Collapse
Affiliation(s)
- Woorim Kim
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Hyun Jeong Kim
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Nga Thi Trinh
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Ha Rim Yeon
- College of Pharmacy, Chungbuk National University, Yeonje-ri, Osong-eup, Heungdeok-gu, Cheongju-si
| | - Joo Hee Kim
- College of Pharmacy, Ajou University, Worldcup-ro, Yeongtong-gu, Suwon
| | - In Ah Choi
- Division of Rheumatology, Department of Internal Medicine, Chungbuk National University Hospital, 1sunhwan-ro, Seowon-gu, Cheongju
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University School of Medicine, Worldcup-ro, Yeongtong-gu, Suwon, Republic of Korea
| | - Ju-Yang Jung
- Department of Rheumatology, Ajou University School of Medicine, Worldcup-ro, Yeongtong-gu, Suwon, Republic of Korea
| | - Kyung Eun Lee
- Department of Rheumatology, Ajou University School of Medicine, Worldcup-ro, Yeongtong-gu, Suwon, Republic of Korea
| |
Collapse
|
29
|
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: 24] [Impact Index Per Article: 6.0] [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
|
30
|
Shen T, Yue C, Wang X, Wang Z, Wu Y, Zhao C, Chang P, Sun X, Wang W. NFATc1 promotes epithelial-mesenchymal transition and facilitates colorectal cancer metastasis by targeting SNAI1. Exp Cell Res 2021; 408:112854. [PMID: 34597678 DOI: 10.1016/j.yexcr.2021.112854] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 01/16/2023]
Abstract
Metastatic recurrence remains a major cause of colorectal cancer (CRC) mortality. In this study, we investigated the mechanistic role of nuclear factor of activated T cells 1 (NFATc1) in CRC metastasis. First, we explored the potential role of NFATc1 in CRC using bioinformatics and hypothesized that NFATc1 might play different roles at different stages of CRC development. Then, we examined the relative expression of NFATc1 in 25 CRC tissues and adjacent normal tissues, and further analyzed the correlation between NFATc1 expression levels and clinical stages in 120 CRC patients. The role of NFATc1 in CRC metastasis and the molecular mechanisms were investigated in both in vitro and in vivo models. Our results showed that the expression of NFATc1 was increased in metastatic CRC tissues and positively associated with clinical stages (stage I vs. stage II, III or IV) of CRC. Overexpression of NFATc1 promoted CRC cell migration, invasion, and epithelial-mesenchymal transition (EMT). Moreover, SNAI1 was verified as the direct transcriptional target of NFATc1 and interacted with SLUG to promote EMT. Remarkably, our lung and liver metastasis mouse model demonstrated that NFATc1 overexpression accelerated CRC metastasis, and treatment with FK506, a calcineurin-NFAT pathway inhibitor, could suppress CRC metastasis in vivo. Taken together, our findings suggest that NFATc1 could transcriptionally activate SNAI1, which in turn interacts with SLUG to mediate EMT to promote CRC metastasis. Thus, making NFATc1 a promising therapeutic target in the treatment of metastatic CRC.
Collapse
Affiliation(s)
- Tianli Shen
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chenyang Yue
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Xingjie Wang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Zijun Wang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Yunhua Wu
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Chenye Zhao
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Pengkang Chang
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Xuejun Sun
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| | - Wei Wang
- Department of General Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, China.
| |
Collapse
|
31
|
Topalov NE, Mayr D, Scherer C, Chelariu-Raicu A, Beyer S, Hester A, Kraus F, Zheng M, Kaltofen T, Kolben T, Burges A, Mahner S, Trillsch F, Jeschke U, Czogalla B. Actin Beta-Like 2 as a New Mediator of Proliferation and Migration in Epithelial Ovarian Cancer. Front Oncol 2021; 11:713026. [PMID: 34631538 PMCID: PMC8495414 DOI: 10.3389/fonc.2021.713026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/25/2021] [Indexed: 11/16/2022] Open
Abstract
The impact of Actin beta-like 2 (ACTBL2), a novel described actin isoform, on epithelial ovarian cancer (EOC) biology has not been investigated so far. In this study, we analyzed the prognostic and functional significance of ACTBL2 and its regulatory element Nuclear factor of activated T-cells 5 (NFAT5). The expression of ACTBL2 and NFAT5 was examined in tissue microarrays of 156 ovarian cancer patients by immunohistochemistry. Aiming to assess the molecular impact of ACTBL2 on cellular characteristics, functional assays were executed in vitro upon siRNA knockdown of ACTBL2 and NFAT5. ACTBL2 expression was identified as an independent negative prognostic factor for overall survival of EOC patients. EOC cell lines showed a significantly increased mRNA and protein level of ACTBL2 compared to the benign control. In vitro analyses upon siRNA knockdown of ACTBL2 displayed a significantly reduced cellular viability, proliferation and migration. siRNA knockdown of NFAT5 proved a significant molecular interplay by inducing a downregulation of ACTBL2 with a thus resulting concordant alteration in cellular functions, predominantly reflected in a decreased migratory potential of EOC cells. Our results provide significant evidence on the negative prognostic impact of ACTBL2 in EOC, suggesting its crucial importance in ovarian carcinogenesis by modulating cellular motility and proliferation.
Collapse
Affiliation(s)
| | - Doris Mayr
- Institute of Pathology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Clemens Scherer
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany.,5DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, University Hospital, LMU Munich, Munich, Germany
| | - Anca Chelariu-Raicu
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Susanne Beyer
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Anna Hester
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Fabian Kraus
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Mingjun Zheng
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Till Kaltofen
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Thomas Kolben
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Alexander Burges
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany.,Department of Obstetrics and Gynecology, University Hospital Augsburg, Augsburg, Germany
| | - Bastian Czogalla
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
32
|
Pontrelli P, Gigante M, Spadaccino F, Netti GS, Saldarelli M, Balducci L, Gigante M, Battaglia M, Storkus WJ, Castellano G, Stallone G, Gesualdo L, Ranieri E. CD40 Cross-Linking Induces Migration of Renal Tumor Cell through Nuclear Factor of Activated T Cells (NFAT) Activation. Int J Mol Sci 2021; 22:ijms22168871. [PMID: 34445576 PMCID: PMC8396205 DOI: 10.3390/ijms22168871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/11/2022] Open
Abstract
CD40 crosslinking plays an important role in regulating cell migration, adhesion and proliferation in renal cell carcinoma (RCC). CD40/CD40L interaction on RCC cells activates different intracellular pathways but the molecular mechanisms leading to cell scattering are not yet clearly defined. Aim of our study was to investigate the main intracellular pathways activated by CD40 ligation and their specific involvement in RCC cell migration. CD40 ligation increased the phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun NH (2)-terminal kinase (JNK) and p38 MAPK. Furthermore, CD40 crosslinking activated different transcriptional factors on RCC cell lines: AP-1, NFkB and some members of the Nuclear Factor of Activated T cells (NFAT) family. Interestingly, the specific inhibition of NFAT factors by cyclosporine A, completely blocked RCC cell motility induced by CD40 ligation. In tumor tissue, we observed a higher expression of NFAT factors and in particular an increased activation and nuclear migration of NFATc4 on RCC tumor tissues belonging to patients that developed metastases when compared to those who did not. Moreover, CD40-CD40L interaction induced a cytoskeleton reorganization and increased the expression of integrin β1 on RCC cell lines, and this effect was reversed by cyclosporine A and NFAT inhibition. These data suggest that CD40 ligation induces the activation of different intracellular signaling pathways, in particular the NFATs factors, that could represent a potential therapeutic target in the setting of patients with metastatic RCC.
Collapse
Affiliation(s)
- Paola Pontrelli
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Margherita Gigante
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Federica Spadaccino
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Giuseppe Stefano Netti
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Marilisa Saldarelli
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Luigi Balducci
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Maddalena Gigante
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Michele Battaglia
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Walter J. Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA;
| | - Giuseppe Castellano
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Giovanni Stallone
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
| | - Loreto Gesualdo
- Department of Emergency and Organ Transplantation, Divisions of Nephrology and Urology, University of Bari, Piazza G. Cesare 11, 70124 Bari, Italy; (P.P.); (M.B.); (L.G.)
| | - Elena Ranieri
- Department of Medical and Surgical Sciences, Divisions of Clinical Pathology and Nephrology, University of Foggia, Policlinico Riuniti, Viale L. Pinto, 71100 Foggia, Italy; (M.G.); (F.S.); (G.S.N.); (M.S.); (L.B.); (M.G.); (G.C.); (G.S.)
- Correspondence: ; Tel.: +39-0881-732611; Fax: +39-0881-732627
| |
Collapse
|
33
|
Role of Nuclear Factor of Activated T Cells (NFAT) Pathway in Regulating Autophagy and Inflammation in Retinal Pigment Epithelial Cells. Int J Mol Sci 2021; 22:ijms22168684. [PMID: 34445390 PMCID: PMC8395439 DOI: 10.3390/ijms22168684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 01/03/2023] Open
Abstract
Nuclear factor of activated T cells (NFAT) family of transcription factors are substrates of calcineurin and play an important role in integrating Ca2+ signaling with a variety of cellular functions. Of the five NFAT proteins (NFAT1-5), NFAT1-4 are subject to dephosphorylation and activation by calcineurin, a Ca2+-calmodulin-dependent phosphatase. Increased levels of intracellular Ca2+ activates calcineurin, which in turn dephosphorylates and promotes nuclear translocation of NFAT. We investigated the functions of NFAT proteins in the retinal pigment epithelial cells (RPE). Our results show that NFAT-mediated luciferase activity was induced upon treatment with the bacterial endotoxin, lipopolysaccharide (LPS) and treatment with the NFAT peptide inhibitor, MAGPHPVIVITGPHEE (VIVIT) decreased LPS-induced NFAT luciferase activity. LPS-induced activation of NFAT-regulated cytokines (IL-6 and IL-8) is inhibited by treatment of cells with VIVIT. We also investigated the effects of NFAT signaling on the autophagy pathway. Our results show that inhibition of NFAT with VIVIT in cells deprived of nutrients resulted in cytosolic retention of transcription Factor EB (TFEB), decreased expression of TFEB-regulated coordinated Lysosomal Expression and Regulation CLEAR network genes and decreased starvation-induced autophagy flux in the RPE cells. In summary, these studies suggest that the NFAT pathway plays an important role in the regulation of autophagy and inflammation in the RPE.
Collapse
|
34
|
NFAT transcription factors are essential and redundant actors for leukemia initiating potential in T-cell acute lymphoblastic leukemia. PLoS One 2021; 16:e0254184. [PMID: 34234374 PMCID: PMC8263285 DOI: 10.1371/journal.pone.0254184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/21/2021] [Indexed: 11/21/2022] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy with few available targeted therapies. We previously reported that the phosphatase calcineurin (Cn) is required for LIC (leukemia Initiating Capacity) potential of T-ALL pointing to Cn as an interesting therapeutic target. Calcineurin inhibitors have however unwanted side effect. NFAT transcription factors play crucial roles downstream of calcineurin during thymocyte development, T cell differentiation, activation and anergy. Here we elucidate NFAT functional relevance in T-ALL. Using murine T-ALL models in which Nfat genes can be inactivated either singly or in combination, we show that NFATs are required for T-ALL LIC potential and essential to survival, proliferation and migration of T-ALL cells. We also demonstrate that Nfat genes are functionally redundant in T-ALL and identified a node of genes commonly deregulated upon Cn or NFAT inactivation, which may serve as future candidate targets for T-ALL.
Collapse
|
35
|
Dai ZT, Xiang Y, Wang Y, Bao LY, Wang J, Li JP, Zhang HM, Lu Z, Ponnambalam S, Liao XH. Prognostic value of members of NFAT family for pan-cancer and a prediction model based on NFAT2 in bladder cancer. Aging (Albany NY) 2021; 13:13876-13897. [PMID: 33962392 PMCID: PMC8202856 DOI: 10.18632/aging.202982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 03/26/2021] [Indexed: 04/23/2023]
Abstract
Bladder cancer (BLCA) is one of the common malignant tumors of the urinary system. The poor prognosis of BLCA patients is due to the lack of early diagnosis and disease recurrence after treatment. Increasing evidence suggests that gene products of the nuclear factor of activated T-cells (NFAT) family are involved in BLCA progression and subsequent interaction(s) with immune surveillance. In this study, we carried out a pan-cancer analysis of the NFAT family and found that NFAT2 is an independent prognostic factor for BLCA. We then screened for differentially expressed genes (DEGs) and further analyzed such candidate gene loci using gene ontology enrichment to curate the KEGG database. We then used Lasso and multivariate Cox regression to identify 4 gene loci (FER1L4, RNF128, EPHB6, and FN1) which were screened together with NFAT2 to construct a prognostic model based on using Kaplan-Meier analysis to predict the overall survival of BLCA patients. Moreover, the accuracy of our proposed model is supported by deposited datasets in the Gene Expression Omnibus (GEO) database. Finally, a nomogram of this prognosis model for BLCA was established which could help to provide better disease management and treatment.
Collapse
Affiliation(s)
- Zhou-Tong Dai
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| | - Yuan Xiang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
- Department of Medical Laboratory, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei, P.R. China
| | - Yundan Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| | - Le-Yuan Bao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| | - Jun Wang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| | - Jia-Peng Li
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| | - Hui-Min Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| | - Zhongxin Lu
- Department of Medical Laboratory, Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, Hubei, P.R. China
| | - Sreenivasan Ponnambalam
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Xing-Hua Liao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P.R. China
| |
Collapse
|
36
|
Pastor TP, Peixoto BC, Viola JPB. The Transcriptional Co-factor IRF2BP2: A New Player in Tumor Development and Microenvironment. Front Cell Dev Biol 2021; 9:655307. [PMID: 33996817 PMCID: PMC8116537 DOI: 10.3389/fcell.2021.655307] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/06/2021] [Indexed: 12/13/2022] Open
Abstract
Interferon regulatory factor 2-binding protein 2 (IRF2BP2) encodes a member of the IRF2BP family of transcriptional regulators, which includes IRF2BP1, IRF2BP2, and IRF2BPL (EAP1). IRF2BP2 was initially identified as a transcriptional corepressor that was dependent on Interferon regulatory factor-2 (IRF-2). The IRF2BP2 protein is found in different organisms and has been described as ubiquitously expressed in normal and tumor cells and tissues, indicating a possible role for this transcriptional cofactor in different cell signaling pathways. Recent data suggest the involvement of IRF2BP2 in the regulation of several cellular functions, such as the cell cycle, cell death, angiogenesis, inflammation and immune response, thereby contributing to physiological cell homeostasis. However, an imbalance in IRF2BP2 function may be related to the pathophysiology of cancer. Some studies have shown the association of IRF2BP2 expression in hematopoietic and solid tumors through mechanisms based on gene fusion and point mutations in gene coding sequences, and although the biological functions of these types of hybrid and mutant proteins are not yet known, they are thought to be involved in an increase in the likelihood of tumor development. In this review, we address the possible involvement of IRF2BP2 in tumorigenesis through its regulation of important pathways involved in tumor development.
Collapse
Affiliation(s)
- Tatiane P Pastor
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - Barbara C Peixoto
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| | - João P B Viola
- Program of Immunology and Tumor Biology, Brazilian National Cancer Institute (INCA), Rio de Janeiro, Brazil
| |
Collapse
|
37
|
Ma J, Du R, Huang Y, Zhong W, Gui H, Mao C, Song X, Lu J. Expression, Prognosis and Gene Regulation Network of NFAT Transcription Factors in Non-Small Cell Lung Cancer. Pathol Oncol Res 2021; 27:529240. [PMID: 34257525 PMCID: PMC8262184 DOI: 10.3389/pore.2021.529240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 02/26/2021] [Indexed: 12/09/2022]
Abstract
Non-small cell lung cancer (NSCLC) is the leading cause of cancer-related death worldwide. The nuclear factor of activated T cells (NFAT) family is implicated in tumorigenesis and progression in various types of cancer. However, little is known about their expression patterns, distinct prognostic values, and potential regulatory networks in NSCLC. In this study, we comprehensively analyzed the distinct expression and prognostic value of NFATs in NSCLC through various large databases, including the Oncomine, UCSC Xena Browser, UALCAN databases, Kaplan–Meier Plotter, cBioPortal, and Enrichr. In lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC), NFAT1/2/4/5 mRNA expression levels were significantly decreased and NFAT3 mRNA expression level was significantly increased. The cBioPortal database analysis showed that the mRNA dysregulation was one of the single most important factors for NFAT alteration in LUAD and LUSC and that both LUAD and LUSC cases with the alterations in the mRNA expression of NFATs had significantly better overall survival (OS). High expression levels of NFAT1/2/4/5 were significantly associated with better OS in LUAD, whereas high NFAT3 expression led to a worse OS. Overexpression of NFAT1/2 predicted better OS in LUSC, whereas high NFAT5 expression led to a worse OS. The networks for NFATs and the 50 most frequently altered neighbor genes in LUAD and LUSC were also constructed. NFATs and genes significantly associated with NFAT mRNA expression in LUAD and LUSC were significantly enriched in the cGMP-dependent protein kinase and Wnt signaling pathways. These results showed that the NFAT family members displayed varying degrees of abnormal expressions, suggesting that NFATs may be therapeutic targets for patients with NSCLC. Aberrant expression of NFATs was found to be associated with OS in the patients with NSCLC; among NFATs, NFAT3/4 may be new biomarkers for the prognosis of LUAD. However, further studies are required to validate our findings.
Collapse
Affiliation(s)
- Jin Ma
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China
| | - Rao Du
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China
| | - Yan Huang
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China
| | - Wen Zhong
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China
| | - Huan Gui
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China
| | - Chenmei Mao
- Department of Pharmacy, Children's Hospital of Soochow University, Suzhou, China
| | - Xiudao Song
- Clinical Pharmaceutical Laboratory of Traditional Chinese Medicine, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Jun Lu
- Department of Haematology, Children's Hospital of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Karedath T, Al-Dasim FM, Ahmed I, Al-Qurashi A, Raza A, Andrews SS, Ahmed AA, Ali Mohamoud Y, Dermime S, Malek JA. Regulation of Circular RNA CircNFATC3 in Cancer Cells Alters Proliferation, Migration, and Oxidative Phosphorylation. Front Cell Dev Biol 2021; 9:595156. [PMID: 33816459 PMCID: PMC8017239 DOI: 10.3389/fcell.2021.595156] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 02/26/2021] [Indexed: 12/13/2022] Open
Abstract
Circular RNAs were once considered artifacts of transcriptome sequencing but have recently been identified as functionally relevant in different types of cancer. Although there is still no clear main function of circRNAs, several studies have revealed that circRNAs are expressed in various eukaryotic organisms in a regulated manner often independent of their parental linear isoforms demonstrating conservation across species. circNFATC3, an abundant and uncharacterized circular RNA of exon 2 and 3 from NFATC3, was identified in transcriptomic data of solid tumors. Here we show that circNFATC3 gain- and loss-of-function experiments using RNAi-mediated circRNA silencing and circular mini vector-mediated overexpression of circularized constructs in breast and ovarian cancer cell lines affect molecular phenotypes. The knockdown of circNFATC3 induces a reduction in cell proliferation, invasion, migration, and oxidative phosphorylation. Gain-of-function of circNFATC3 in MDA-MB-231 and SK-OV-3 cells show a significant increase in cell proliferation, migration, and respiration. The above results suggest that circNFATC3 is a functionally relevant circular RNA in breast and ovarian cancer.
Collapse
Affiliation(s)
- Thasni Karedath
- Weill Cornell Medicine-Qatar, Doha, Qatar
- Sidra Medicine, Doha, Qatar
| | | | - Ikhlak Ahmed
- Weill Cornell Medicine-Qatar, Doha, Qatar
- Sidra Medicine, Doha, Qatar
| | | | - Afsheen Raza
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | | | | | | | - Said Dermime
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | | |
Collapse
|
39
|
Laplana M, Bieg M, Faltus C, Melnik S, Bogatyrova O, Gu Z, Muley T, Meister M, Dienemann H, Herpel E, Amos CI, Schlesner M, Eils R, Plass C, Risch A. Differentially methylated regions within lung cancer risk loci are enriched in deregulated enhancers. Epigenetics 2021; 17:117-132. [PMID: 33595421 PMCID: PMC8865272 DOI: 10.1080/15592294.2021.1878723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified SNPs linked with lung cancer risk. Our aim was to discover the genes, non-coding RNAs, and regulatory elements within GWAS-identified risk regions that are deregulated in non-small cell lung carcinoma (NSCLC) to identify novel, clinically targetable genes and mechanisms in carcinogenesis. A targeted bisulphite-sequencing approach was used to comprehensively investigate DNA methylation changes occurring within lung cancer risk regions in 17 NSCLC and adjacent normal tissue pairs. We report differences in differentially methylated regions between adenocarcinoma and squamous cell carcinoma. Among the minimal regions found to be differentially methylated in at least 50% of the patients, 7 candidates were replicated in 2 independent cohorts (n = 27 and n = 87) and the potential of 6 as methylation-dependent regulatory elements was confirmed by functional assays. This study contributes to understanding the pathways implicated in lung cancer initiation and progression, and provides new potential targets for cancer treatment.
Collapse
Affiliation(s)
- Marina Laplana
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias Bieg
- Center for Digital Health, Berlin Institute of Health and Charité - Universitätsmedizin Berlin, Berlin, Germany.,Heidelberg Center for Personalized Oncology (DKFZ-HIPO), Heidelberg, Germany
| | - Christian Faltus
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Biosciences, Allergy-Cancer-BioNano Research Centre, University of Salzburg, Salzburg, Austria
| | - Svitlana Melnik
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Olga Bogatyrova
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Zuguang Gu
- Heidelberg Center for Personalized Oncology (DKFZ-HIPO), Heidelberg, Germany.,Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Thomas Muley
- Translational Research Unit, Thoraxklinik-Heidelberg gGmbH, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michael Meister
- Translational Research Unit, Thoraxklinik-Heidelberg gGmbH, University of Heidelberg, Heidelberg, Germany.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Hendrik Dienemann
- Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Thoracic Surgery, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Esther Herpel
- Tissue Bank of the National Center for Tumor Diseases (NCT) and Institute of Pathology, Heidelberg University Hospital, Germany
| | - Christopher I Amos
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Matthias Schlesner
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health and Charité - Universitätsmedizin Berlin, Berlin, Germany.,Heidelberg Center for Personalized Oncology (DKFZ-HIPO), Heidelberg, Germany.,Health Data Science Unit, University Hospital Heidelberg, Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Angela Risch
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Biosciences, Allergy-Cancer-BioNano Research Centre, University of Salzburg, Salzburg, Austria.,Translational Lung Research Center Heidelberg (TLRC), Member of the German Center for Lung Research (DZL), Heidelberg, Germany.,Cancer Cluster Salzburg, Austria
| |
Collapse
|
40
|
Ray S, Tillo D, Durell SR, Khund-Sayeed S, Vinson C. REL Domain of NFATc2 Binding to Five Types of DNA Using Protein Binding Microarrays. ACS OMEGA 2021; 6:4147-4154. [PMID: 33644537 PMCID: PMC7906578 DOI: 10.1021/acsomega.0c04069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/25/2020] [Indexed: 06/12/2023]
Abstract
NFATc2 is a DNA binding protein in the Rel family transcription factors, which binds a CGGAA motif better when both cytosines in the CG dinucleotide are methylated. Using protein binding microarrays (PBMs), we examined the DNA binding of NFATc2 to three additional types of DNA: single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA) with either 5-methylcytosine (5mC, M) or 5-hydroxymethylcytosine (5hmC, H) in one strand and a cytosine in the second strand. ATTTCCAC, the complement of the core GGAA motif, is better bound as ssDNA compared to dsDNA. dsDNA containing the 5-mer CGGAA with either 5mC or 5hmC in one DNA strand is bound stronger than CGGAA. In contrast, the reverse complement TTCCG is bound weaker when it contains 5mC. Analysis of the available NFATc2:dsDNA complexes rationalizes these PBM data.
Collapse
Affiliation(s)
- Sreejana Ray
- Laboratory
of Metabolism, National Cancer Institute,
National Institutes of Health, 37 Convent Drive, Building 37, Room 5000, Bethesda, Maryland 20892, United States
| | - Desiree Tillo
- Laboratory
of Metabolism, National Cancer Institute,
National Institutes of Health, 37 Convent Drive, Building 37, Room 5000, Bethesda, Maryland 20892, United States
| | - Stewart R. Durell
- Laboratory
of Cell Biology, National Cancer Institute,
National Institutes of Health, 37 Convent Drive, Building 37, Room 5000, Bethesda, Maryland 20892, United States
| | - Syed Khund-Sayeed
- Laboratory
of Metabolism, National Cancer Institute,
National Institutes of Health, 37 Convent Drive, Building 37, Room 5000, Bethesda, Maryland 20892, United States
| | - Charles Vinson
- Laboratory
of Metabolism, National Cancer Institute,
National Institutes of Health, 37 Convent Drive, Building 37, Room 5000, Bethesda, Maryland 20892, United States
| |
Collapse
|
41
|
Snuggs JW, Tessier S, Bunning RAB, Shapiro IM, Risbud MV, Le Maitre CL. TonEBP regulates the hyperosmotic expression of aquaporin 1 and 5 in the intervertebral disc. Sci Rep 2021; 11:3164. [PMID: 33542263 PMCID: PMC7862284 DOI: 10.1038/s41598-021-81838-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 12/22/2020] [Indexed: 01/30/2023] Open
Abstract
The central region of the intervertebral disc (IVD) is rich in proteoglycans, leading to a hyperosmotic environment, which fluctuates with daily loading. The cells of the nucleus pulposus (NP cells) have adapted to this environment via the function of tonicity enhancer binding protein (TonEBP), and NP cells have been shown to express several water channels known as aquaporins (AQP). We have previously shown that AQP1 and 5 decrease during IVD degeneration. Here, the regulation of AQP1 and 5 by hyperosmotic conditions and the role of TonEBP in this regulation was investigated. AQP1 and 5 gene expression was upregulated by hyperosmotic conditions mimicking the osmolality of the healthy IVD, which was abrogated by TonEBP knockdown. Furthermore, AQP1 and 5 immunopositivity was significantly reduced in TonEBPΔ/Δ E17.5 mice when compared with wildtype controls, indicating in vivo expression of AQP1 and 5 is controlled at least in part by TonEBP. This hyperosmotic regulation of AQP1 and 5 could help to explain the decreased AQP1 and 5 expression during degeneration, when the osmolality of the NP decreases. Together this data suggests that TonEBP-regulated osmo-adaptation may be disrupted during IVD degeneration when the expression of both AQPs is reduced.
Collapse
Affiliation(s)
- J. W. Snuggs
- grid.5884.10000 0001 0303 540XBiomolecular Sciences Research Centre, Sheffield Hallam University, City Campus, Howard Street, Sheffield, S1 1WB UK
| | - S. Tessier
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA USA
| | - R. A. B. Bunning
- grid.5884.10000 0001 0303 540XBiomolecular Sciences Research Centre, Sheffield Hallam University, City Campus, Howard Street, Sheffield, S1 1WB UK
| | - I. M. Shapiro
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
| | - M. V. Risbud
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
| | - C. L. Le Maitre
- grid.5884.10000 0001 0303 540XBiomolecular Sciences Research Centre, Sheffield Hallam University, City Campus, Howard Street, Sheffield, S1 1WB UK
| |
Collapse
|
42
|
Integrin α3β1 Promotes Invasive and Metastatic Properties of Breast Cancer Cells through Induction of the Brn-2 Transcription Factor. Cancers (Basel) 2021; 13:cancers13030480. [PMID: 33513758 PMCID: PMC7866210 DOI: 10.3390/cancers13030480] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Metastatic triple-negative breast cancer (TNBC) is highly lethal with limited therapy options. Integrin α3β1 is a cell surface receptor that interacts with the extracellular matrix and facilitates communication between tumor cells and their microenvironment. α3β1 is implicated in breast cancer progression and metastasis, so understanding mechanisms by which α3β1 promotes invasion and metastasis will facilitate the development of this integrin as a potential therapeutic target. Here we identify a novel role for α3β1 in promoting the expression of the transcription factor Brain-2 (Brn-2) in triple-negative breast cancer cells. We further report that Brn-2 promotes invasion and metastasis and partially restores invasion to cells in which expression of α3β1 has been suppressed. Bioinformatic analysis of patient datasets revealed a positive correlation between the expression of the genes encoding the integrin α3 subunit and Brn-2. In summary, our work identifies α3β1-mediated induction of Brn-2 as a mechanism that regulates invasive and metastatic properties of breast cancer cells. Abstract In the current study, we demonstrate that integrin α3β1 promotes invasive and metastatic traits of triple-negative breast cancer (TNBC) cells through induction of the transcription factor, Brain-2 (Brn-2). We show that RNAi-mediated suppression of α3β1 in MDA-MB-231 cells caused reduced expression of Brn-2 mRNA and protein and reduced activity of the BRN2 gene promoter. In addition, RNAi-targeting of Brn-2 in MDA-MB-231 cells decreased invasion in vitro and lung colonization in vivo, and exogenous Brn-2 expression partially restored invasion to cells in which α3β1 was suppressed. α3β1 promoted phosphorylation of Akt in MDA-MB-231 cells, and treatment of these cells with a pharmacological Akt inhibitor (MK-2206) reduced both Brn-2 expression and cell invasion, indicating that α3β1-Akt signaling contributes to Brn-2 induction. Analysis of RNAseq data from patients with invasive breast carcinoma revealed that high BRN2 expression correlates with poor survival. Moreover, high BRN2 expression positively correlates with high ITGA3 expression in basal-like breast cancer, which is consistent with our experimental findings that α3β1 induces Brn-2 in TNBC cells. Together, our study demonstrates a pro-invasive/pro-metastatic role for Brn-2 in breast cancer cells and identifies a role for integrin α3β1 in regulating Brn-2 expression, thereby revealing a novel mechanism of integrin-dependent breast cancer cell invasion.
Collapse
|
43
|
Yoshimoto S, Morita H, Matsuda M, Katakura Y, Hirata M, Hashimoto S. NFAT5 promotes oral squamous cell carcinoma progression in a hyperosmotic environment. J Transl Med 2021; 101:38-50. [PMID: 32901097 PMCID: PMC7758185 DOI: 10.1038/s41374-020-00486-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 12/27/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) is highly expressed in several types of cancer cells including oral squamous cell carcinoma (OSCC). EGF/EGFR signaling is recognized as an important molecular target in cancer therapy. However, cancer cells often become tolerant to EGF/EGFR signaling-targeted therapies. In the tumor microenvironment, the tumor incites inflammation and the inflammation-derived cytokines make a considerable impact on cancer development. In addition, hyperosmolarity is also induced, but the role of osmotic stress in cancer development has not been fully understood. This study demonstrates molecular insights into hyperosmolarity effect on OSCC development and shows that NFAT5 transcription factor plays an important functional role in enhancing the oral cancer cell proliferation by inducing the EGFR translocation from the endoplasmic reticulum to the plasma membrane through increase the expression of DPAGT1, an essential enzyme for catalyzing the first committed step of N-linked protein glycosylation. These results suggest that hyperosmolarity-induced intra-nuclear translocation of NFAT5 essential for DPAGT1 activation and EGFR subcellular translocation responsible for OSCC tumor progression.
Collapse
Affiliation(s)
- Shohei Yoshimoto
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Hiromitsu Morita
- The Center for Visiting Dental Service, Department of General Dentistry, Fukuoka Dental College, Fukuoka Dental College, Fukuoka, Japan
| | - Miho Matsuda
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yoshinori Katakura
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Masato Hirata
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Shuichi Hashimoto
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan.
| |
Collapse
|
44
|
Su F, Shi M, Zhang J, Zheng Q, Wang H, Li X, Chen J. MiR-223/NFAT5 signaling suppresses arterial smooth muscle cell proliferation and motility in vitro. Aging (Albany NY) 2020; 12:26188-26198. [PMID: 33373321 PMCID: PMC7803580 DOI: 10.18632/aging.202395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 10/31/2020] [Indexed: 01/11/2023]
Abstract
Aberrant proliferation and migration of vascular smooth muscle cells contributes to cardiovascular diseases (CVDs), including atherosclerosis. MicroRNA-223 (miR-223) protects against atherosclerotic CVDs. We investigated the contribution of miR-223 to platelet-derived growth factor-BB (PDGF-BB)-induced proliferation and migration of human aortic smooth muscle cells (HASMCs). We found that miR-223 was downregulated in PDGF-BB-treated HASMCs in a dose- and time-dependent manner, while nuclear factor of activated T cells 5 (NFAT5) was upregulated. Gain- and loss-of-function studies demonstrated that miR-223 treatment reduced PDGF-BB-induced HASMC proliferation and motility, whereas miR-223 inhibitor enhanced these processes. Moreover, NFAT5 was identified as a direct target of miR-223 in HASMC. The inhibitory effects of miR-223 on HASMC proliferation and migration were partly rescued by NFAT5 restoration. Overall, these findings suggest that miR-223 inhibits the PDGF-BB-induced proliferation and motility of HASMCs by targeting NFAT5 and that miR-223 and NFAT5 may be potential therapeutic targets for atherosclerosis.
Collapse
Affiliation(s)
- Feifei Su
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China.,Department of Cardiology, Air Force General Hospital, PLA, Beijing 100142, China
| | - Miaoqian Shi
- Department of Cardiology, PLA Army General Hospital, Beijing 100700, China
| | - Jian Zhang
- Department of Cardiology, Beijing Chest Hospital Heart Center, Capital Medical University, Beijing 101149, China
| | - Qiangsun Zheng
- Division of Cardiology, Second Affiliated Hospital of Xi’an Jiao Tong University, Xi'an 710004, Shaanxi, China
| | - Haichang Wang
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| | - Xue Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| | - Jianghong Chen
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi’an 710038, Shaanxi, China
| |
Collapse
|
45
|
NFAT5 directs hyperosmotic stress-induced fibrin deposition and macrophage infiltration via PAI-1 in endothelium. Aging (Albany NY) 2020; 13:3661-3679. [PMID: 33410782 PMCID: PMC7906158 DOI: 10.18632/aging.202330] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/31/2020] [Indexed: 12/12/2022]
Abstract
Although stress can significantly promote atherosclerosis, the underlying mechanisms are still not completely understood. Here we successfully unveiled that high salt-induced nuclear factor of activated T cells 5 (NFAT5) control the endothelial-dependent fibrinolytic activity and the inflammatory adhesion-related molecules expression through regulation of plasminogen activator inhibitor-1 (PAI-1). We first observed that high salt diets instigated the expression of NFAT5 and PAI-1 in the endothelium which brought about the fibrin deposition and macrophage infiltration in the atherosclerotic arteries of ApoE-/- mice. Overexpression of NFAT5 increased PAI-1-mediated antifibrinolytic activity and activated inflammatory adhesion-related genes in endothelial cells. Knockdown of NFAT5 by siRNA inhibited the expression of PAI-1, antifibrinolytic and adhesive molecules. Moreover, chromatin immunoprecipitation assay demonstrated that high salt intake significantly promoted the binding of NFAT5 to PAI-1 promoter (TGGAATTATTT) in endothelial cells. Our study identified that NFAT5 has great potential to activate the PAI-1-mediated fibrinolytic dysfunction and inflammatory cell adhesion, thus promoting high salt-induced atherosclerosis disease.
Collapse
|
46
|
Transcription factors in colorectal cancer: molecular mechanism and therapeutic implications. Oncogene 2020; 40:1555-1569. [PMID: 33323976 DOI: 10.1038/s41388-020-01587-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is a major cause of cancer mortality worldwide, however, the molecular mechanisms underlying the pathogenesis of CRC remain largely unclear. Recent studies have revealed crucial roles of transcription factors in CRC development. Transcription factors essential for the regulation of gene expression by interacting with transcription corepressor/enhancer complexes and they orchestrate downstream signal transduction. Deregulation of transcription factors is a frequent occurrence in CRC, and the accompanying drastic changes in gene expression profiles play fundamental roles in multistep process of tumorigenesis, from cellular transformation, disease progression to metastatic disease. Herein, we summarized current and emerging key transcription factors that participate in CRC tumorigenesis, and highlighted their oncogenic or tumor suppressive functions. Moreover, we presented critical transcription factors of CRC, emphasized the major molecular mechanisms underlying their effect on signal cascades associated with tumorigenesis, and summarized of their potential as molecular biomarkers for CRC prognosis therapeutic response, as well as drug targets for CRC treatment. A better understanding of transcription factors involved in the development of CRC will provide new insights into the pathological mechanisms and reveal novel prognostic biomarkers and therapeutic strategies for CRC.
Collapse
|
47
|
Nie L, Wang C, Li N, Feng X, Lee N, Su D, Tang M, Yao F, Chen J. Proteome-wide Analysis Reveals Substrates of E3 Ligase RNF146 Targeted for Degradation. Mol Cell Proteomics 2020; 19:2015-2030. [PMID: 32958691 PMCID: PMC7710139 DOI: 10.1074/mcp.ra120.002290] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Indexed: 12/28/2022] Open
Abstract
Specific E3 ligases target tumor suppressors for degradation. Inhibition of such E3 ligases may be an important approach to cancer treatment. RNF146 is a RING domain and PARylation-dependent E3 ligase that functions as an activator of the β-catenin/Wnt and YAP/Hippo pathways by targeting the degradation of several tumor suppressors. Tankyrases 1 and 2 (TNKS1/2) are the only known poly-ADP-ribosyltransferases that require RNF146 to degrade their substrates. However, systematic identification of RNF146 substrates have not yet been performed. To uncover substrates of RNF146 that are targeted for degradation, we generated RNF146 knockout cells and TNKS1/2-double knockout cells and performed proteome profiling with label-free quantification as well as transcriptome analysis. We identified 160 potential substrates of RNF146, which included many known substrates of RNF146 and TNKS1/2 and 122 potential TNKS-independent substrates of RNF146. In addition, we validated OTU domain-containing protein 5 and Protein mono-ADP-ribosyltransferase PARP10 as TNKS1/2-independent substrates of RNF146 and SARDH as a novel substrate of TNKS1/2 and RNF146. Our study is the first proteome-wide analysis of potential RNF146 substrates. Together, these findings not only demonstrate that proteome profiling can be a useful general approach for the systemic identification of substrates of E3 ligases but also reveal new substrates of RNF146, which provides a resource for further functional studies.
Collapse
Affiliation(s)
- Litong Nie
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chao Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nan Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xu Feng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Namsoo Lee
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dan Su
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mengfan Tang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fan Yao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Junjie Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
48
|
Timucin AC. Structure based peptide design, molecular dynamics and MM-PBSA studies for targeting C terminal dimerization of NFAT5 DNA binding domain. J Mol Graph Model 2020; 103:107804. [PMID: 33248341 DOI: 10.1016/j.jmgm.2020.107804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 11/27/2022]
Abstract
NFAT5 as a transcription factor with an established role in osmotic stress response, has also been revealed to be active under numerous settings, including pathological conditions such as diabetic microvascular complications, chronic arthritis and cancer. Despite these links, current strategies for downregulating NFAT5 activity only relies on indirect modulators, not directly targeting NFAT5, itself. With this study, through using a computational approach, an original peptide was explored to directly target C terminal dimerization of NFAT5 RHR, located in its DNA binding domain. At first, homodimeric NFAT5 RHR bound to its consensus DNA was used for prediction of a preliminary peptide sequence. Possible amino acid replacements for this preliminary peptide were predicted for optimization, which was followed by addition of a cell penetrating peptide sequence. These attempts yielded a small peptide library, which was further investigated for peptide affinities towards C terminal of NFAT5 RHR through molecular docking, 50 ns and 250 ns molecular dynamics simulations, followed by estimation of MM-PBSA based relative binding free energies. Results indicated that after receiving mutations on the preliminary peptide sequence for optimization, a unique peptide could target C terminal dimerization region of NFAT5 RHR through using its cell penetrating peptide sequence. In conclusion, this is the first study presenting computational evidence on identification of a novel peptide capable of directly targeting NFAT5 dimerization. Besides, future implications of these observations were also discussed in terms of methodology and possible applications.
Collapse
Affiliation(s)
- Ahmet Can Timucin
- Department of Chemical Engineering, Faculty of Natural Sciences and Engineering, Üsküdar University, Turkey; Neuropsychopharmacology Application and Research Center (NPARC), Üsküdar University, Turkey.
| |
Collapse
|
49
|
Xin B, Ji KQ, Liu YS, Zhao XD. NFAT Overexpression Correlates with CA72-4 and Poor Prognosis of Ovarian Clear-Cell Carcinoma Subtype. Reprod Sci 2020; 28:745-756. [PMID: 33125687 DOI: 10.1007/s43032-020-00368-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 10/19/2020] [Indexed: 11/29/2022]
Abstract
Current biomarkers did not overcome the limitations of clinical application due to the heterogeneity of ovarian tumors. The role of nuclear factor of activated T cells (NFAT) in the prognosis of different histological subtypes of ovarian cancer remains unclear. NFAT expression was analyzed in 302 ovarian tumors from The Cancer Genome Atlas (TCGA) dataset and was further confirmed by 88 ovarian tumor specimens, including 30 clear-cell carcinoma, 34 serous carcinoma, and 24 papillary serous cystadenocarcinoma. The correlations between NFAT expression, cancer biomarkers, and clinical characteristics in different subtypes of ovarian tumors were analyzed. ALGGEN PROMO, reporter assay, and NFAT overexpression and knockdown were used to identify chondroadherin (CHAD) as the downstream target of NFAT. NFAT was significantly upregulated only in late-stage clear-cell carcinoma, but not in other two subtypes. NFAT levels were correlated with CA72-4 levels and poor overall survival and disease-free survival (P < 0.05), suggesting that NFAT together with CA72-4 were specific prognostic markers for clear-cell carcinoma. Pathological stage and lymph node metastasis were the prognostic factors affecting serous carcinoma (P < 0.05), while CA-125 was the prognostic factor affecting papillary serous cystadenocarcinoma (P < 0.05). PROMO and reporter assay indicated that CHAD was the downstream target of NFAT. In addition, NFAT overexpression and silencing increased and reduced CHAD expression, respectively. NFAT together with CA72-4 were specific tumor markers for risk assessment of unique clear-cell subtype of ovarian tumors. CHAD was identified as the downstream target gene of NAFT and was associated with poor survival of ovarian cancer.
Collapse
Affiliation(s)
- Bing Xin
- Department of Obstetrics and Gynaecology, ShengJing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China.
| | - Kai-Qiang Ji
- Department of ICU, ShengJing Hospital of China Medical University, Shenyang, 110004, China
| | - Yi-Si Liu
- Department of Obstetrics and Gynaecology, ShengJing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, China
| | - Xiao-Dong Zhao
- Department of Pathology, ShengJing Hospital of China Medical University, Shenyang, 110004, China
| |
Collapse
|
50
|
Chen Y, Huang L, Dong Y, Tao C, Zhang R, Shao H, Shen H. Effect of AKT1 (p. E17K) Hotspot Mutation on Malignant Tumorigenesis and Prognosis. Front Cell Dev Biol 2020; 8:573599. [PMID: 33123537 PMCID: PMC7573235 DOI: 10.3389/fcell.2020.573599] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/03/2020] [Indexed: 12/12/2022] Open
Abstract
The substitution of the seventeenth amino acid glutamate by lysine in the homologous structural domain of the Akt1 gene pleckstrin is a somatic cellular mutation found in breast, colorectal, and ovarian cancers, named p. Glu17Lys or E17K. In recent years, a growing number of studies have suggested that this mutation may play a unique role in the development of tumors. In this review article, we describe how AKT1(E17K) mutations stimulate downstream signals that cause cells to emerge transformed; we explore the differential regulation and function of E17K in different physiological and pathological settings; and we also describe the phenomenon that E17K impedes tumor growth by interfering with growth-promoting and chemotherapy-resistant AKT1lowQCC generation, an intriguing finding that mutants may prolong tumor patient survival by activating feedback mechanisms and disrupting transcription. This review is intended to provide a better understanding of the role of AKT1(E17K) in cancer and to inform the development of AKT1(E17K)-based antitumor strategies.
Collapse
Affiliation(s)
- Ying Chen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lan Huang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongjian Dong
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Changli Tao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Rongxin Zhang
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongwei Shao
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Han Shen
- Guangdong Province Key Laboratory for Biotechnology Drug Candidates, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|