1
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Gupta P, Sakthivel S, Jiemin N, Arul K, Tikno K, Park SH, Wu Y, Wang L, Bay BH, Ho L, Giguere V, Ghosh S, McDonnell DP, Yen PM, Singh BK. ESRRA (estrogen related receptor, alpha) induces ribosomal protein RPLP1-mediated adaptive hepatic translation during prolonged starvation. Autophagy 2025; 21:1283-1297. [PMID: 39936615 PMCID: PMC12087656 DOI: 10.1080/15548627.2025.2465183] [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: 01/22/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/13/2025] Open
Abstract
Protein translation is an energy-intensive ribosome-driven process that is reduced during nutrient scarcity to conserve cellular resources. During prolonged starvation, cells selectively translate specific proteins to enhance their survival (adaptive translation); however, this process is poorly understood. Accordingly, we analyzed protein translation and mRNA transcription by multiple methods in vitro and in vivo to investigate adaptive hepatic translation during starvation. While acute starvation suppressed protein translation in general, proteomic analysis showed that prolonged starvation selectively induced translation of lysosome and autolysosome proteins. Significantly, the expression of the orphan nuclear receptor, ESRRA (estrogen related receptor, alpha) increased during prolonged starvation and served as a master regulator of this adaptive translation by transcriptionally stimulating Rplp1 (ribosomal protein lateral stalk subunit P1) gene expression. Overexpression or siRNA knockdown of Esrra in vitro or in vivo led to parallel changes in Rplp1 gene expression, lysosome and macroautophagy/autophagy protein translation, and autophagy activity. Remarkably, we have found that ESRRA had dual functions by not only regulating transcription but also controlling adaptive translation via the ESRRA-RPLP1-lysosome-autophagy pathway during prolonged starvation.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Karine Gauthier
- Département de Biologie, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, Lyon, Cedex, France
| | - Reddemma Sandireddy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Priyanka Gupta
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Suganya Sakthivel
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Nah Jiemin
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Kabilesh Arul
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Keziah Tikno
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore, Singapore
| | - Lena Ho
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, Montreal, Québec, Canada
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- Pennington Biomedical Research Center, Laboratory of Bioinformatics and Computational Biology, Baton Rouge, LA, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
- Duke Molecular Physiology Institute and Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Brijesh K. Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore, Singapore
| |
Collapse
|
2
|
Ma T, Xie W, Xu Z, Gao W, Zhou J, Wang Y, Chan FL. Estrogen-related receptor alpha (ERRα) controls the stemness and cellular energetics of prostate cancer cells via its direct regulation of citrate metabolism and zinc transportation. Cell Death Dis 2025; 16:154. [PMID: 40044646 PMCID: PMC11882781 DOI: 10.1038/s41419-025-07460-z] [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: 09/17/2024] [Revised: 01/24/2025] [Accepted: 02/18/2025] [Indexed: 03/09/2025]
Abstract
Compared to most tumors that are more glycolytic, primary prostate cancer is less glycolytic but more dependent on TCA cycle coupled with OXPHOS for its energy demand. This unique metabolic energetic feature is attributed to activation of mitochondrial m-aconitase in TCA caused by decreased cellular Zn level. Evidence suggests that a small subpopulation of cancer cells within prostate tumors, designated as prostate cancer stem cells (PCSCs), play significant roles in advanced prostate cancer progression. However, their cellular energetics status is still poorly understood. Nuclear receptor ERRα (ESRRA) is a key regulator of energy metabolism. Previous studies characterize that ERRα exhibits an upregulation in prostate cancer and can perform multiple oncogenic functions. Here, we demonstrate a novel role of ERRα in the control of stemness and energetics metabolism in PCSCs via a mechanism of combined transrepression of Zn transporter ZIP1 in reducing intracellular Zn uptake and transactivation of ACO2 (m-aconitase) in completion of TCA cycle. Results also showed that restoration of Zn accumulation by treatment with a Zn ionophore Clioquinol could significantly suppress both in vitro growth of PCSCs and also their in vivo tumorigenicity, implicating that enhanced cellular Zn uptake could be a potential therapeutic approach for targeting PCSCs in advanced prostate cancer.
Collapse
Affiliation(s)
- Taiyang Ma
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Wenjuan Xie
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Zhenyu Xu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Weijie Gao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Jianfu Zhou
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuliang Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Franky Leung Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
3
|
Vanacker JM, Forcet C. ERRα: unraveling its role as a key player in cell migration. Oncogene 2024; 43:379-387. [PMID: 38129506 DOI: 10.1038/s41388-023-02899-w] [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: 07/28/2023] [Revised: 10/31/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023]
Abstract
Cell migration is essential throughout the life of multicellular organisms, and largely depends on the spatial and temporal regulation of cytoskeletal dynamics, cell adhesion and signal transduction. Interestingly, Estrogen-related receptor alpha (ERRα) has been identified as a major regulator of cell migration in both physiological and pathological conditions. ERRα is an orphan member of the nuclear hormone receptor superfamily of transcription factors and displays many biological functions. ERRα is a global regulator of energy metabolism, and it is also highly involved in bone homeostasis, development, differentiation, immunity and cancer progression. Importantly, in some instances, the regulation of these biological processes relies on the ability to orchestrate cell movements. Therefore, this review describes how ERRα-mediated cell migration contributes not only to tissue homeostasis but also to tumorigenesis and metastasis, and highlights the molecular and cellular mechanisms by which ERRα finely controls the cell migratory potential.
Collapse
Affiliation(s)
- Jean-Marc Vanacker
- Centre de Recherche en Cancérologie de Lyon, CNRS UMR5286, Inserm U1052, Université de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, Lyon, France.
| |
Collapse
|
4
|
Tripathi M, Gauthier K, Sandireddy R, Zhou J, Gupta P, Sakthivel S, Jiemin N, Arul K, Tikno K, Park SH, Wang L, Ho L, Giguere V, Ghosh S, McDonnell DP, Yen PM, Singh BK. Estrogen receptor-related receptor (Esrra) induces ribosomal protein Rplp1-mediated adaptive hepatic translation during prolonged starvation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.09.574937. [PMID: 38260502 PMCID: PMC10802477 DOI: 10.1101/2024.01.09.574937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Protein translation is an energy-intensive ribosome-driven process that is reduced during nutrient scarcity to conserve cellular resources. During prolonged starvation, cells selectively translate specific proteins to enhance their survival (adaptive translation); however, this process is poorly understood. Accordingly, we analyzed protein translation and mRNA transcription by multiple methods in vitro and in vivo to investigate adaptive hepatic translation during starvation. While acute starvation suppressed protein translation in general, proteomic analysis showed that prolonged starvation selectively induced translation of lysosome and autolysosome proteins. Significantly, the expression of the orphan nuclear receptor, estrogen-related receptor alpha (Esrra) increased during prolonged starvation and served as a master regulator of this adaptive translation by transcriptionally stimulating 60S acidic ribosomal protein P1 (Rplp1) gene expression. Overexpression or siRNA knockdown of Esrra expression in vitro or in vivo led to parallel changes in Rplp1 gene expression, lysosome/autophagy protein translation, and autophagy. Remarkably, we have found that Esrra had dual functions by not only regulating transcription but also controling adaptive translation via the Esrra/Rplp1/lysosome/autophagy pathway during prolonged starvation.
Collapse
Affiliation(s)
- Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d’Italie, 69364 Lyon Cedex 07, France
| | - Reddemma Sandireddy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Priyanka Gupta
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Suganya Sakthivel
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Nah Jiemin
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Kabilesh Arul
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Keziah Tikno
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sung-Hee Park
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Lijin Wang
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Lena Ho
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Vincent Giguere
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Sujoy Ghosh
- Centre for Computational Biology, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Paul M. Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Duke Molecular Physiology Institute and Dept. of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Brijesh K. Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| |
Collapse
|
5
|
Wang W, Gao W, Gong P, Song W, Bu X, Hou J, Zhang L, Zhao B. Neuronal-specific TNFAIP1 ablation attenuates postoperative cognitive dysfunction via targeting SNAP25 for K48-linked ubiquitination. Cell Commun Signal 2023; 21:356. [PMID: 38102610 PMCID: PMC10722859 DOI: 10.1186/s12964-023-01390-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/08/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Synaptosomal-associated protein 25 (SNAP25) exerts protective effects against postoperative cognitive dysfunction (POCD) by promoting PTEN-induced kinase 1 (PINK1)/Parkin-mediated mitophagy and repressing caspase-3/gasdermin E (GSDME)-mediated pyroptosis. However, the regulatory mechanisms of SNAP25 protein remain unclear. METHODS We employed recombinant adeno-associated virus 9 (AAV9)-hSyn to knockdown tumor necrosis factor α-induced protein 1 (TNFAIP1) or SNAP25 and investigate the role of TNFAIP1 in POCD. Cognitive performance, hippocampal injury, mitophagy, and pyroptosis were assessed. Co-immunoprecipitation (co-IP) and ubiquitination assays were conducted to elucidate the mechanisms by which TNFAIP1 stabilizes SNAP25. RESULTS Our results demonstrated that the ubiquitin ligase TNFAIP1 was upregulated in the hippocampus of mice following isoflurane (Iso) anesthesia and laparotomy. The N-terminal region (residues 1-96) of TNFAIP1 formed a conjugate with SNAP25, leading to lysine (K) 48-linked polyubiquitination of SNAP25 at K69. Silencing TNFAIP1 enhanced SH-SY5Y cell viability and conferred antioxidant, pro-mitophagy, and anti-pyroptosis properties in response to Iso and lipopolysaccharide (LPS) challenges. Conversely, TNFAIP1 overexpression reduced HT22 cell viability, increased reactive oxygen species (ROS) accumulation, impaired PINK1/Parkin-dependent mitophagy, and induced caspase-3/GSDME-dependent pyroptosis by suppressing SNAP25 expression. Neuron-specific knockdown of TNFAIP1 ameliorated POCD, restored mitophagy, and reduced pyroptosis, which was reversed by SNAP25 depletion. CONCLUSIONS In summary, our findings demonstrated that inhibiting TNFAIP1-mediated degradation of SNAP25 might be a promising therapeutic approach for mitigating postoperative cognitive decline. Video Abstract.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuhan, 430060, China
| | - Wenwei Gao
- Department of Critical Care Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ping Gong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuhan, 430060, China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, Department of Anesthesiology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Wenqin Song
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuhan, 430060, China
| | - Xueshan Bu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuhan, 430060, China
| | - Jiabao Hou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuhan, 430060, China
| | - Lei Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuhan, 430060, China.
| | - Bo Zhao
- Department of Anesthesiology, Renmin Hospital of Wuhan University, No.238 Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
6
|
Qiu F, He S, Zhang Z, Dai S, Wang J, Liu N, Li Z, Hu X, Xiang S, Wei C. MiR-93 alleviates DEHP plasticizer-induced neurotoxicity by negatively regulating TNFAIP1 and inhibiting ubiquitin-mediated degradation of CK2β. Food Chem Toxicol 2023:113888. [PMID: 37302538 DOI: 10.1016/j.fct.2023.113888] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/28/2023] [Accepted: 06/08/2023] [Indexed: 06/13/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is a plasticizer that is widely used in various products, such as plastic packaging in food industries. As an environmental endocrine disruptor, it induces adverse effects on brain development and function. However, the molecular mechanisms by which DEHP induces learning and memory impairment remain poorly understood. Herein, we found that DEHP impaired learning and memory in pubertal C57BL/6 mice, decreased the number of neurons, downregulated miR-93 and the β subunit of casein kinase 2 (CK2β), upregulated tumor necrosis factor-induced protein 1 (TNFAIP1), and inhibited Akt/CREB pathway in mouse hippocampi. Coimmunoprecipitation and western blotting assays revealed that TNFAIP1 interacted with CK2β and promoted its degradation by ubiquitination. Bioinformatics analysis showed a miR-93 binding site in the 3'-untranslated region of Tnfaip1. A dual-luciferase reporter assay revealed that miR-93 targeted TNFAIP1 and negatively regulated its expression. MiR-93 overexpression prevented DEHP-induced neurotoxicity by downregulating TNFAIP1 and then activating CK2/Akt/CREB pathway. These data indicate that DEHP upregulates TNFAIP1 expression by downregulating miR-93, thus promoting ubiquitin-mediated degradation of CK2β, subsequently inhibiting Akt/CREB pathway, and finally inducing learning and memory impairment. Therefore, miR-93 can relieve DEHP-induced neurotoxicity and may be used as a potential molecular target for prevention and treatment of related neurological disorders.
Collapse
Affiliation(s)
- Feng Qiu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Simei He
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Zilong Zhang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Siyu Dai
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Jin Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Ning Liu
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Zhiwei Li
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Xiang Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China
| | - Chenxi Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, School of Life Sciences, Hunan Normal University, Changsha, 410081, Hunan, China.
| |
Collapse
|
7
|
Cerutti C, Shi JR, Vanacker JM. Multifaceted Transcriptional Network of Estrogen-Related Receptor Alpha in Health and Disease. Int J Mol Sci 2023; 24:ijms24054265. [PMID: 36901694 PMCID: PMC10002233 DOI: 10.3390/ijms24054265] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023] Open
Abstract
Estrogen-related receptors (ERRα, β and γ in mammals) are orphan members of the nuclear receptor superfamily acting as transcription factors. ERRs are expressed in several cell types and they display various functions in normal and pathological contexts. Amongst others, they are notably involved in bone homeostasis, energy metabolism and cancer progression. In contrast to other nuclear receptors, the activities of the ERRs are apparently not controlled by a natural ligand but they rely on other means such as the availability of transcriptional co-regulators. Here we focus on ERRα and review the variety of co-regulators that have been identified by various means for this receptor and their reported target genes. ERRα cooperates with distinct co-regulators to control the expression of distinct sets of target genes. This exemplifies the combinatorial specificity of transcriptional regulation that induces discrete cellular phenotypes depending on the selected coregulator. We finally propose an integrated view of the ERRα transcriptional network.
Collapse
|
8
|
Tribollet V, Cerutti C, Géloën A, Berger E, De Mets R, Balland M, Courchet J, Vanacker JM, Forcet C. ERRα coordinates actin and focal adhesion dynamics. Cancer Gene Ther 2022; 29:1429-1438. [PMID: 35379907 DOI: 10.1038/s41417-022-00461-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/15/2022] [Accepted: 03/18/2022] [Indexed: 11/09/2022]
Abstract
Cell migration depends on the dynamic organisation of the actin cytoskeleton and assembly and disassembly of focal adhesions (FAs). However, the precise mechanisms coordinating these processes remain poorly understood. We previously identified the oestrogen-related receptor α (ERRα) as a major regulator of cell migration. Here, we show that loss of ERRα leads to abnormal accumulation of actin filaments that is associated with an increased level of inactive form of the actin-depolymerising factor cofilin. We further show that ERRα depletion decreases cell adhesion and results in defective FA formation and turnover. Interestingly, specific inhibition of the RhoA-ROCK-LIMK-cofilin pathway rescues the actin polymerisation defects resulting from ERRα silencing, but not cell adhesion. Instead, we found that MAP4K4 is a direct target of ERRα and down-regulation of its activity rescues cell adhesion and FA formation in the ERRα-depleted cells. Altogether, our results highlight a crucial role of ERRα in coordinating the dynamic of actin network and FAs through the independent regulation of the RhoA and MAP4K4 pathways.
Collapse
Affiliation(s)
- Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Alain Géloën
- Université de Lyon, UMR Ecologie Microbienne (LEM), CNRS 5557, INRAE 1418, Université Claude Bernard Lyon 1, VetAgro Sup, Research Team "Bacterial Opportunistic Pathogens and Environment" (BPOE), 69622, Villeurbanne, cedex, France
| | - Emmanuelle Berger
- Université de Lyon, UMR Ecologie Microbienne (LEM), CNRS 5557, INRAE 1418, Université Claude Bernard Lyon 1, VetAgro Sup, Research Team "Bacterial Opportunistic Pathogens and Environment" (BPOE), 69622, Villeurbanne, cedex, France
| | - Richard De Mets
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
| | - Martial Balland
- Laboratoire Interdisciplinaire de Physique, Grenoble Alpes University, 38402, Saint Martin d'Hères, France
| | - Julien Courchet
- Université de Lyon, Université Claude Bernard Lyon 1, CNRS, INSERM, Physiopathologie et Génétique du Neurone et du Muscle, UMR5261, U1315, Institut NeuroMyoGène, 69008, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 69007, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Claude Bernard Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 69007, Lyon, France.
| |
Collapse
|
9
|
Cerutti C, Zhang L, Tribollet V, Shi JR, Brillet R, Gillet B, Hughes S, Forcet C, Shi TL, Vanacker JM. Computational identification of new potential transcriptional partners of ERRα in breast cancer cells: specific partners for specific targets. Sci Rep 2022; 12:3826. [PMID: 35264626 PMCID: PMC8907200 DOI: 10.1038/s41598-022-07744-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/17/2022] [Indexed: 12/26/2022] Open
Abstract
Estrogen related receptors are orphan members of the nuclear receptor superfamily acting as transcription factors (TFs). In contrast to classical nuclear receptors, the activities of the ERRs are not controlled by a natural ligand. Regulation of their activities thus relies on availability of transcriptional co-regulators. In this paper, we focus on ERRα, whose involvement in cancer progression has been broadly demonstrated. We propose a new approach to identify potential co-activators, starting from previously identified ERRα-activated genes in a breast cancer (BC) cell line. Considering mRNA gene expression from two sets of human BC cells as major endpoint, we used sparse partial least squares modeling to uncover new transcriptional regulators associated with ERRα. Among them, DDX21, MYBBP1A, NFKB1, and SETD7 are functionally relevant in MDA-MB-231 cells, specifically activating the expression of subsets of ERRα-activated genes. We studied SET7 in more details and showed its co-localization with ERRα and its ERRα-dependent transcriptional and phenotypic effects. Our results thus demonstrate the ability of a modeling approach to identify new transcriptional partners from gene expression. Finally, experimental results show that ERRα cooperates with distinct co-regulators to control the expression of distinct sets of target genes, thus reinforcing the combinatorial specificity of transcription.
Collapse
Affiliation(s)
- Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
| | - Ling Zhang
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
| | - Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
| | - Jing-Ru Shi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Riwan Brillet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
| | - Benjamin Gillet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
| | - Sandrine Hughes
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
| | - Tie-Liu Shi
- The Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France.
| |
Collapse
|
10
|
Wu J, Shen G, Liu D, Xu H, Jiao M, Zhang Y, Lin Y, Zhao P. The Response of the Estrogen-Related Receptor to 20-Hydroxyecdysone in Bombyx mori: Insight Into the Function of Estrogen-Related Receptor in Insect 20-Hydroxyecdysone Signaling Pathway. Front Physiol 2022; 12:785637. [PMID: 35115955 PMCID: PMC8804299 DOI: 10.3389/fphys.2021.785637] [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: 09/29/2021] [Accepted: 12/29/2021] [Indexed: 01/03/2023] Open
Abstract
Estrogen-related receptor (ERR) is an orphan nuclear receptor that was first discovered in animals, and play an important role in metabolism, development, and reproduction. Despite extensive research on the function of ERR, its transcriptional regulation mechanism remains unclear. In this study, we obtained the upstream region of Bombyx mori ERR (BmERR) and confirmed the promoter activity of this region. Interestingly, we found that 10 and 50 nM 20-hydroxyecdysone (20E) up-regulated the transcriptional activity of BmERR promoter. In addition, eight putative ecdysone response elements (EcREs) were predicted in the upstream sequence of BmERR. Based on their positions, the upstream sequence of BmERR was truncated into different fragments. Finally, an EcRE-like sequence (5′-AGTGCAGTAAACTGT-3′) was identified. Electrophoretic mobility shift assay (EMSA) and cell transfection experiments confirmed that this motif specifically binds to the complex formed between ecdysone receptor (BmEcR) and the ultraspiracle (BmUSP), a key complex in the 20E signaling pathway. Interference of BmERR or BmEcR mRNA in the embryonic cells of Bombyx mori significantly affected the expression of BmEcR and BmUSP. Overall, these results suggested that an EcRE element was identified from BmERR, and this will help understanding the detailed regulatory mechanism of ERR in insects.
Collapse
Affiliation(s)
- Jinxin Wu
- Biological Science Research Center, Southwest University, Chongqing, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Guanwang Shen
- Biological Science Research Center, Southwest University, Chongqing, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Die Liu
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
| | - Haoran Xu
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing, China
| | - Mengyao Jiao
- Biological Science Research Center, Southwest University, Chongqing, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Yungui Zhang
- Biological Science Research Center, Southwest University, Chongqing, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Ying Lin
- Biological Science Research Center, Southwest University, Chongqing, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- *Correspondence: Ying Lin,
| | - Ping Zhao
- Biological Science Research Center, Southwest University, Chongqing, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
- Ping Zhao,
| |
Collapse
|
11
|
STK25 and MST3 Have Overlapping Roles to Regulate Rho GTPases during Cortical Development. J Neurosci 2021; 41:8887-8903. [PMID: 34518307 DOI: 10.1523/jneurosci.0523-21.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 08/13/2021] [Accepted: 09/06/2021] [Indexed: 11/21/2022] Open
Abstract
Precise control of neuronal migration is required for the laminar organization of the neocortex and critical for brain function. We previously reported that the acute disruption of the Stk25 gene (Stk25 conditional knock-out; cKO) during mouse embryogenesis causes anomalous neuronal migration in the neocortex, but paradoxically the Stk25 cKO did not have a cortical phenotype, suggesting some forms of compensation exist. In this study, we report that MST3, another member of the GCKIII subgroup of the Ste20-like kinase family, compensates for loss of Stk25 and vice versa with sex independent manner. MST3 overexpression rescued neuronal migration deficit and abnormal axonogenesis in Stk25 cKO brains. Mechanistically, STK25 leads to Rac1 activation and reduced RhoA levels in the developing brain, both of which are required to fully restore neuronal migration in the Stk25 cKO brain. Abnormal migration phenotypes are also rescued by overexpression of Bacurd1and Cul3, which target RhoA for degradation, and activate Rac1. This study reveals that MST3 upregulation is capable of rescuing acute Stk25 deficiency and resolves details of signaling downstream STK25 required for corticogenesis both common to and distinct from MST3 signaling.SIGNIFICANCE STATEMENT Proper neuronal migration during cortical development is required for normal neuronal function. Here, we show that STK25 and MST3 kinases regulate neuronal migration and polarization in a mutually compensatory manner. Furthermore, STK25 balances Rac1 activity and RhoA level through forming complexes with α-PIX and β-PIX, GTPase regulatory enzymes, and Cullin3-Bacurd1/Kctd13, a pair of RhoA ubiquitination molecules in a kinase activity-independent manner. Our findings demonstrate the importance of overlapping and unique roles of STK25 and MST3 to regulate Rho GTPase activities in cortical development.
Collapse
|
12
|
González-Alvarez ME, McGuire BC, Keating AF. Obesity alters the ovarian proteomic response to zearalenone exposure†. Biol Reprod 2021; 105:278-289. [PMID: 33855340 PMCID: PMC8256104 DOI: 10.1093/biolre/ioab069] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/10/2021] [Accepted: 04/07/2021] [Indexed: 12/21/2022] Open
Abstract
Zearalenone (ZEN), a nonsteroidal estrogenic mycotoxin, is detrimental to female reproduction. Altered chemical biotransformation, depleted primordial follicles and a blunted genotoxicant response have been discovered in obese female ovaries, thus, this study investigated the hypothesis that obesity would enhance ovarian sensitivity to ZEN exposure. Seven-week-old female wild-type nonagouti KK.Cg-a/a mice (lean) and agouti lethal yellow KK.Cg-Ay/J mice (obese) received food and water ad libitum, and either saline or ZEN (40 μg/kg) per os for 15 days. Body and organ weights, and estrous cyclicity were recorded, and ovaries collected posteuthanasia for protein analysis. Body and liver weights were increased (P < 0.05) in the obese mice, but obesity did not affect (P > 0.05) heart, kidney, spleen, uterus, or ovary weight and there was no impact (P > 0.05) of ZEN exposure on body or organ weight in lean or obese mice. Obese mice had shorter proestrus (P < 0.05) and a tendency (P = 0.055) for longer metestrus/diestrus. ZEN exposure in obese mice increased estrus but shortened metestrus/diestrus length. Neither obesity nor ZEN exposure impacted (P > 0.05) circulating progesterone, or ovarian abundance of EPHX1, GSTP1, CYP2E1, ATM, BRCA1, DNMT1, HDAC1, H4K16ac, or H3K9me3. Lean mice exposed to ZEN had a minor increase in γH2AX abundance (P < 0.05). In lean and obese mice, LC-MS/MS identified alterations to proteins involved in chemical metabolism, DNA repair and reproduction. These data identify ZEN-induced adverse ovarian modes of action and suggest that obesity is additive to ZEN-induced ovotoxicity.
Collapse
Affiliation(s)
- M Estefanía González-Alvarez
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames IA, USA
| | - Bailey C McGuire
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames IA, USA
| | - Aileen F Keating
- Department of Animal Science and Interdepartmental Toxicology Graduate Program, Iowa State University, Ames IA, USA
| |
Collapse
|
13
|
Podieh F, Hordijk PL. Regulation of Rho GTPases in the Vasculature by Cullin3-Based E3 Ligase Complexes. Front Cell Dev Biol 2021; 9:680901. [PMID: 34136490 PMCID: PMC8201781 DOI: 10.3389/fcell.2021.680901] [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: 03/15/2021] [Accepted: 05/04/2021] [Indexed: 02/02/2023] Open
Abstract
Cullin3-based ubiquitin E3 ligases induce ubiquitination of substrates leading to their proteasomal or lysosomal degradation. BTB proteins serve as adaptors by binding to Cullin3 and recruiting substrate proteins, which enables specific recognition of a broad spectrum of targets. Hence, Cullin3 and its adaptors are involved in myriad cellular processes and organ functions. Cullin3-based ubiquitin E3 ligase complexes target small GTPases of the Rho subfamily, which are key regulators of cytoskeletal dynamics and cell adhesion. In this mini review, we discuss recent insights in Cullin3-mediated regulation of Rho GTPases and their impact on cellular function and disease. Intriguingly, upstream regulators of Rho GTPases are targeted by Cullin3 complexes as well. Thus, Rho GTPase signaling is regulated by Cullin3 on multiple levels. In addition, we address current knowledge of Cullin3 in regulating vascular function, focusing on its prominent role in endothelial barrier function, angiogenesis and the regulation of blood pressure.
Collapse
Affiliation(s)
- Fabienne Podieh
- Department of Physiology, Amsterdam UMC, Amsterdam, Netherlands
| | - Peter L Hordijk
- Department of Physiology, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
14
|
Kim SY, Song HK, Lee SK, Kim SG, Woo HG, Yang J, Noh HJ, Kim YS, Moon A. Sex-Biased Molecular Signature for Overall Survival of Liver Cancer Patients. Biomol Ther (Seoul) 2020; 28:491-502. [PMID: 33077700 PMCID: PMC7585639 DOI: 10.4062/biomolther.2020.157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/18/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
Sex/gender disparity has been shown in the incidence and prognosis of many types of diseases, probably due to differences in genes, physiological conditions such as hormones, and lifestyle between the sexes. The mortality and survival rates of many cancers, especially liver cancer, differ between men and women. Due to the pronounced sex/gender disparity, considering sex/gender may be necessary for the diagnosis and treatment of liver cancer. By analyzing research articles through a PubMed literature search, the present review identified 12 genes which showed practical relevance to cancer and sex disparities. Among the 12 sex-specific genes, 7 genes (BAP1, CTNNB1, FOXA1, GSTO1, GSTP1, IL6, and SRPK1) showed sex-biased function in liver cancer. Here we summarized previous findings of cancer molecular signature including our own analysis, and showed that sex-biased molecular signature CTNNB1High, IL6High, RHOAHigh and GLIPR1Low may serve as a female-specific index for prediction and evaluation of OS in liver cancer patients. This review suggests a potential implication of sex-biased molecular signature in liver cancer, providing a useful information on diagnosis and prediction of disease progression based on gender.
Collapse
Affiliation(s)
- Sun Young Kim
- Department of Chemistry, College of Natural Sciences, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Hye Kyung Song
- Department of Chemistry, College of Natural Sciences, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Suk Kyeong Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06649, Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University_Seoul, Goyang 10326, Republic of Korea
| | - Hyun Goo Woo
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea.,Department of Biomedical Science, Graduate School, Ajou University, Suwon 16499, Republic of Korea
| | - Jieun Yang
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea.,Department of Biomedical Science, Graduate School, Ajou University, Suwon 16499, Republic of Korea
| | - Hyun-Jin Noh
- Department of Biomedical Science, Graduate School, Ajou University, Suwon 16499, Republic of Korea.,Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - You-Sun Kim
- Department of Biomedical Science, Graduate School, Ajou University, Suwon 16499, Republic of Korea.,Department of Biochemistry, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Aree Moon
- Duksung Innovative Drug Center, College of Pharmacy, Duksung Women's University, Seoul 01369, Republic of Korea
| |
Collapse
|
15
|
Xiao Y, Huang S, Qiu F, Ding X, Sun Y, Wei C, Hu X, Wei K, Long S, Xie L, Xun Y, Chen W, Zhang Z, Liu N, Xiang S. Tumor necrosis factor α-induced protein 1 as a novel tumor suppressor through selective downregulation of CSNK2B blocks nuclear factor-κB activation in hepatocellular carcinoma. EBioMedicine 2020; 51:102603. [PMID: 31901862 PMCID: PMC6950786 DOI: 10.1016/j.ebiom.2019.102603] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/10/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Background Tumor necrosis factor α-induced protein 1 (TNFAIP1) is frequently downregulated in cancer cell lines and promotes cancer cell apoptosis. However, its role, clinical significance and molecular mechanisms in hepatocellular carcinoma (HCC) are unknown. Methods The expression of TNFAIP1 in HCC tumor tissues and cell lines was measured by Western blot and immunohistochemistry. The effects of TNFAIP1 on HCC proliferation, apoptosis, metastasis, angiogenesis and tumor formation were evaluated by Cell Counting Kit-8 (CCK8), Terminal deoxynucleotidyl transferase dUTP Nick-End Labeling (TUNEL), transwell, tube formation assay in vitro and nude mice experiments in vivo. The interaction between TNFAIP1 and CSNK2B was validated by liquid chromatography-tandem mass spectrometry (LC-MS/MS), Co-immunoprecipitation and Western blot. The mechanism of how TNFAIP1 regulated nuclear factor-kappaB (NF-κB) pathway was analyzed by dual-luciferase reporter, immunofluorescence, quantitative Real-time polymerase chain reaction (RT-qPCR) and Western blot. Findings The TNFAIP1 expression is significantly decreased in HCC tissues and cell lines, and negatively correlated with the increased HCC histological grade. Overexpression of TNFAIP1 inhibits HCC cell proliferation, metastasis, angiogenesis and promotes cancer cell apoptosis both in vitro and in vivo, whereas the knockdown of TNFAIP1 in HCC cell displays opposite effects. Mechanistically, TNFAIP1 interacts with CSNK2B and promotes its ubiquitin-mediated degradation with Cul3, causing attenuation of CSNK2B-dependent NF-κB trans-activation in HCC cell. Moreover, the enforced expression of CSNK2B counteracts the inhibitory effects of TNFAIP1 on HCC cell proliferation, migration, and angiogenesis in vitro and in vivo. Interpretation Our results support that TNFAIP1 can act as a tumor suppressor of HCC by modulating TNFAIP1/CSNK2B/NF-κB pathway, implying that TNFAIP1 may represent a potential marker and a promising therapeutic target for HCC.
Collapse
Affiliation(s)
- Ye Xiao
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China; Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Shulan Huang
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Feng Qiu
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xiaofeng Ding
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Yi Sun
- Department of Pathology, Second Xiangya Hospital of Central South University, Changsha, 410011, China
| | - Chenxi Wei
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Xiang Hu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Ke Wei
- Medical school, Hunan University of Traditional Chinese Medicine, Changsha, 410208, China
| | - Shengwen Long
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Lina Xie
- Department of Stomatology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yu Xun
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Wen Chen
- Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China
| | - Zhijian Zhang
- Department of Pathology, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Ning Liu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China.
| | - Shuanglin Xiang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, China; Key Laboratory of Protein Chemistry and Development Biology of State Education Ministry of China, College of Life Science, Hunan Normal University, Changsha, 410081, China.
| |
Collapse
|
16
|
Klebe D, Tibrewal M, Sharma DR, Vanaparthy R, Krishna S, Varghese M, Cheng B, Mouton PR, Velíšková J, Dobrenis K, Hof PR, Ballabh P. Reduced Hippocampal Dendrite Branching, Spine Density and Neurocognitive Function in Premature Rabbits, and Reversal with Estrogen or TrkB Agonist Treatment. Cereb Cortex 2019; 29:4932-4947. [PMID: 30877788 PMCID: PMC6918929 DOI: 10.1093/cercor/bhz033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 01/16/2019] [Accepted: 02/08/2019] [Indexed: 01/02/2023] Open
Abstract
Preterm-born children suffer from neurological and behavioral disorders. Herein, we hypothesized that premature birth and non-maternal care of preterm newborns might disrupt neurobehavioral function, hippocampal dendritic arborization, and dendritic spine density. Additionally, we assessed whether 17β-estradiol (E2) replacement or the TrkB receptor agonist, 7,8-dihydroxyflavone (DHF), would reverse compromised dendritic development and cognitive function in preterm newborns. These hypotheses were tested by comparing preterm (E28.5) rabbit kits cared and gavage-fed by laboratory personnel and term-kits reared and breast-fed by their mother doe at an equivalent postconceptional age. Neurobehavioral tests showed that both premature-birth and formula-feeding with non-maternal care led to increased anxiety behavior, poor social interaction, and lack of novelty preference compared with term-kits. Dendritic branching and number of total or mushroom dendritic spines were reduced in the CA1 field of preterm-kits compared with term controls. While CDC42 and Rac1/2/3 expression levels were lower, RhoA-activity was higher in preterm-kits compared with term controls. Both E2 and DHF treatment reversed prematurity-induced reduction in spine density, reduced total RhoA-GTPase levels, and enhanced cognitive function. Hence, prematurity and non-maternal care result in cognitive deficits, and reduced dendritic arbors and spines in CA1. E2 replacement or DHF treatment might reverse changes in dendritic spines and improve neurodevelopment in premature infants.
Collapse
Affiliation(s)
- Damon Klebe
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Mahima Tibrewal
- Department of Pediatrics, New York Medical College, Valhalla NY, USA
| | - Deep R Sharma
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Rachna Vanaparthy
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Sunil Krishna
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Merina Varghese
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Bokun Cheng
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Peter R Mouton
- Department of Pathology and Cell Biology, College of Medicine, University of South Florida, Tampa FL, USA
| | - Jana Velíšková
- Departments of Cell Biology & Anatomy, Neurology, and Obstetrics & Gynecology, New York Medical College, Valhalla NY, USA
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| | - Patrick R Hof
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Praveen Ballabh
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx NY, USA
| |
Collapse
|
17
|
Zhou Y, Jia Q, Meng X, Chen D, Zhu B. ERRα Regulates OTUB1 Expression to Promote Colorectal Cancer Cell Migration. J Cancer 2019; 10:5812-5819. [PMID: 31737118 PMCID: PMC6843886 DOI: 10.7150/jca.30720] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023] Open
Abstract
Ovarian tumor domain-containing ubiquitin aldehyde binding protein 1 (OTUB1) is overexpressed in many cancers and plays an important role in tumor progression and metastasis. However, the molecular mechanisms underlying OTUB1 overexpression are not clear. In this study, we found that estrogen-related receptor alpha (ERRα, also called NR3B1) binds to OTUB1 promoter and regulates its expression in colorectal cancer. Furthermore, ERRα promoted the migration of CRC cells by inducing vimentin expression via OTUB1. Our data show that OTUB1 is a novel target of ERRα and indicate that ERRα-OTUB1 signaling may play a significant role in CRC metastasis.
Collapse
Affiliation(s)
- Yi Zhou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qingzhu Jia
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoqing Meng
- Department of Hematology, the Third Affiliated Hospital of Chongqing Medical University (Gener Hospital), Chongqing, China
| | - Diangang Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
18
|
Bros M, Haas K, Moll L, Grabbe S. RhoA as a Key Regulator of Innate and Adaptive Immunity. Cells 2019; 8:cells8070733. [PMID: 31319592 PMCID: PMC6678964 DOI: 10.3390/cells8070733] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/13/2022] Open
Abstract
RhoA is a ubiquitously expressed cytoplasmic protein that belongs to the family of small GTPases. RhoA acts as a molecular switch that is activated in response to binding of chemokines, cytokines, and growth factors, and via mDia and the ROCK signaling cascade regulates the activation of cytoskeletal proteins, and other factors. This review aims to summarize our current knowledge on the role of RhoA as a general key regulator of immune cell differentiation and function. The contribution of RhoA for the primary functions of innate immune cell types, namely neutrophils, macrophages, and conventional dendritic cells (DC) to (i) get activated by pathogen-derived and endogenous danger signals, (ii) migrate to sites of infection and inflammation, and (iii) internalize pathogens has been fairly established. In activated DC, which constitute the most potent antigen-presenting cells of the immune system, RhoA is also important for the presentation of pathogen-derived antigen and the formation of an immunological synapse between DC and antigen-specific T cells as a prerequisite to induce adaptive T cell responses. In T cells and B cells as the effector cells of the adaptive immune system Rho signaling is pivotal for activation and migration. More recently, mutations of Rho and Rho-modulating factors have been identified to predispose for autoimmune diseases and as causative for hematopoietic malignancies.
Collapse
Affiliation(s)
- Matthias Bros
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany.
| | - Katharina Haas
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Lorna Moll
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stephan Grabbe
- University Medical Center Mainz, Department of Dermatology, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
19
|
Liao J, Ng SH, Luk AC, Suen HC, Qian Y, Lee AWT, Tu J, Fung JCL, Tang NLS, Feng B, Chan WY, Fouchet P, Hobbs RM, Lee TL. Revealing cellular and molecular transitions in neonatal germ cell differentiation using single cell RNA sequencing. Development 2019; 146:dev174953. [PMID: 30824552 DOI: 10.1242/dev.174953] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 02/17/2019] [Indexed: 12/22/2022]
Abstract
Neonatal germ cell development provides the foundation of spermatogenesis. However, a systematic understanding of this process is still limited. To resolve cellular and molecular heterogeneity in this process, we profiled single cell transcriptomes of undifferentiated germ cells from neonatal mouse testes and employed unbiased clustering and pseudotime ordering analysis to assign cells to distinct cell states in the developmental continuum. We defined the unique transcriptional programs underlying migratory capacity, resting cellular states and apoptosis regulation in transitional gonocytes. We also identified a subpopulation of primitive spermatogonia marked by CD87 (plasminogen activator, urokinase receptor), which exhibited a higher level of self-renewal gene expression and migration potential. We further revealed a differentiation-primed state within the undifferentiated compartment, in which elevated Oct4 expression correlates with lower expression of self-renewal pathway factors, higher Rarg expression, and enhanced retinoic acid responsiveness. Lastly, a knockdown experiment revealed the role of Oct4 in the regulation of gene expression related to the MAPK pathway and cell adhesion, which may contribute to stem cell differentiation. Our study thus provides novel insights into cellular and molecular regulation during early germ cell development.
Collapse
Affiliation(s)
- Jinyue Liao
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Shuk Han Ng
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Alfred Chun Luk
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Hoi Ching Suen
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Yan Qian
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Annie Wing Tung Lee
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Jiajie Tu
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Jacqueline Chak Lam Fung
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
| | - Nelson Leung Sang Tang
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Bo Feng
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Wai Yee Chan
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Joint CUHK-UoS (University of Southampton) Joint Laboratories for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- CUHK-BGI Innovation Institute of Trans-omics Hong Kong, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Pierre Fouchet
- CEA DRF IBFJ IRCM, Laboratoire des Cellules Souches Germinales, 92265 Fontenay-aux-Roses, France
- Université Paris Diderot, Sorbonne Paris Cité, INSERM, UMR 967, 92265 Fontenay-aux-Roses, France
- Université Paris Sud, INSERM, UMR 967, 92265 Fontenay-aux-Roses, France
| | - Robin M Hobbs
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Melbourne, Victoria 3800, Australia
| | - Tin Lap Lee
- Developmental and Regenerative Biology Program, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- The Chinese University of Hong Kong - Shandong University (CUHK-SDU) Joint Laboratory on Reproductive Genetics, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Joint CUHK-UoS (University of Southampton) Joint Laboratories for Stem Cells and Regenerative Medicine, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- CUHK-BGI Innovation Institute of Trans-omics Hong Kong, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| |
Collapse
|
20
|
Zhang L, Zhu Y, Cheng H, Zhang J, Zhu Y, Chen H, Chen L, Qi H, Ren G, Tang J, Zhong M, Hua W, Shi X, Li Q. The Increased Expression of Estrogen-Related Receptor α Correlates with Wnt5a and Poor Prognosis in Patients with Glioma. Mol Cancer Ther 2019; 18:173-184. [PMID: 30322948 DOI: 10.1158/1535-7163.mct-17-0782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 01/10/2018] [Accepted: 10/10/2018] [Indexed: 11/16/2022]
Abstract
Malignant glioma is an often fatal type of cancer. Elevated expression of the orphan nuclear receptor estrogen-related receptor alpha (ERRα) is an unfavorable factor for malignant progression and poor prognosis in several cancers, although the mechanism by which this receptor affects the pathophysiology of cancers remains obscure. However, few studies have been conducted in regard to the role of ERRα in glioma. In the current study, we found that elevated expression of ERRα was observed in 107 glioma cases by means of IHC. Clinically, high expression of ERRα was associated with later stages of disease progression and clinical outcome of patients with glioma. ERRα had the ability to promote cell proliferation and migration in glioma cell lines. Moreover, in a xenograft model, we also found that silencing ERRα had an inhibitory effect on the growth of glioma. Further investigation confirmed that ERRα was involved in the carcinogenesis of glioma via the regulation of the Wnt5a signal pathway in vitro and in vivo Our study was first to show the overexpression of ERRα in glioma tissues and a direct correlation between ERRα expression and clinical prognosis of glioma. Together, these data reveal that ERRα has prognostic significance in glioma, and targeting ERRα may provide a reliable therapeutic strategy for the treatment for human glioma.
Collapse
Affiliation(s)
- Liudi Zhang
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai, China
| | - Yingfeng Zhu
- Department of Pathology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Haixia Cheng
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinsen Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuqian Zhu
- Department of Neurosurgery, Huashan Hospital North, Fudan University, Shanghai, China
| | - Haifei Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai, China
| | - Lu Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai, China
| | - Huijie Qi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai, China
| | - Guoqiang Ren
- Department of Pathology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Jianmin Tang
- Department of Pathology, Huashan Hospital North, Fudan University, Shanghai, China
| | - Mingkang Zhong
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai, China
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
| | - Xiaojin Shi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai, China.
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai, China
| | - Qunyi Li
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai, China.
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
21
|
Zhang L, Carnesecchi J, Cerutti C, Tribollet V, Périan S, Forcet C, Wong J, Vanacker JM. LSD1-ERRα complex requires NRF1 to positively regulate transcription and cell invasion. Sci Rep 2018; 8:10041. [PMID: 29968728 PMCID: PMC6030097 DOI: 10.1038/s41598-018-27676-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/05/2018] [Indexed: 12/20/2022] Open
Abstract
Lysine-specific demethylase 1 (LSD1) exerts dual effects on histone H3, promoting transcriptional repression via Lys4 (H3K4) demethylation or transcriptional activation through Lys9 (H3K9) demethylation. These activities are often exerted at transcriptional start sites (TSSs) and depend on the type of enhancer-bound transcription factor (TFs) with which LSD1 interacts. In particular, the Estrogen-Receptor Related α (ERRα) TF interacts with LSD1 and switches its activities toward H3K9 demethylation, resulting in transcriptional activation of a set of common target genes. However, how are the LSD1-TF and, in particular LSD1-ERRα, complexes determined to act at TSSs is not understood. Here we show that promoter-bound nuclear respiratory factor 1 (NRF1), but not ERRα, is essential to LSD1 recruitment at the TSSs of positive LSD1-ERRα targets. In contrast to ERRα, NRF1 does not impact on the nature of LSD1 enzymatic activity. We propose a three factor model, in which the LSD1 histone modifier requires a TSS tethering factor (NRF1) as well as an activity inducer (ERRα) to transcriptionally activate common targets. The relevance of this common network is illustrated by functional data, showing that all three factors are required for cell invasion in an MMP1 (Matrix MetalloProtease 1)-dependent manner, the expression of which is regulated by NRF1/LSD1/ERRα-mediated H3K9me2 demethylation.
Collapse
Affiliation(s)
- Ling Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
- Department of Developmental Biology, Centre for Organismal Studies (COS), Heidelberg, University of Heidelberg, D-69120, Heidelberg, Germany
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Séverine Périan
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, F-69007, Lyon, France.
| |
Collapse
|
22
|
Denley MCS, Gatford NJF, Sellers KJ, Srivastava DP. Estradiol and the Development of the Cerebral Cortex: An Unexpected Role? Front Neurosci 2018; 12:245. [PMID: 29887794 PMCID: PMC5981095 DOI: 10.3389/fnins.2018.00245] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 03/28/2018] [Indexed: 12/16/2022] Open
Abstract
The cerebral cortex undergoes rapid folding in an "inside-outside" manner during embryonic development resulting in the establishment of six discrete cortical layers. This unique cytoarchitecture occurs via the coordinated processes of neurogenesis and cell migration. In addition, these processes are fine-tuned by a number of extracellular cues, which exert their effects by regulating intracellular signaling pathways. Interestingly, multiple brain regions have been shown to develop in a sexually dimorphic manner. In many cases, estrogens have been demonstrated to play an integral role in mediating these sexual dimorphisms in both males and females. Indeed, 17β-estradiol, the main biologically active estrogen, plays a critical organizational role during early brain development and has been shown to be pivotal in the sexually dimorphic development and regulation of the neural circuitry underlying sex-typical and socio-aggressive behaviors in males and females. However, whether and how estrogens, and 17β-estradiol in particular, regulate the development of the cerebral cortex is less well understood. In this review, we outline the evidence that estrogens are not only present but are engaged and regulate molecular machinery required for the fine-tuning of processes central to the cortex. We discuss how estrogens are thought to regulate the function of key molecular players and signaling pathways involved in corticogenesis, and where possible, highlight if these processes are sexually dimorphic. Collectively, we hope this review highlights the need to consider how estrogens may influence the development of brain regions directly involved in the sex-typical and socio-aggressive behaviors as well as development of sexually dimorphic regions such as the cerebral cortex.
Collapse
Affiliation(s)
- Matthew C. S. Denley
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Nicholas J. F. Gatford
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Katherine J. Sellers
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Deepak P. Srivastava
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| |
Collapse
|
23
|
Long non-coding RNA MT1DP shunts the cellular defense to cytotoxicity through crosstalk with MT1H and RhoC in cadmium stress. Cell Discov 2018; 4:5. [PMID: 29507753 PMCID: PMC5824791 DOI: 10.1038/s41421-017-0005-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/12/2017] [Accepted: 12/12/2017] [Indexed: 12/18/2022] Open
Abstract
Metallothioneins (MTs) are known to protect cells against oxidative stress, especially providing protection against cadmium (Cd) toxicity in hepatocytes. There are various gene variants and pseudogenes for MTs; however, there is little understanding on the functions of those non-coding MT members that are known to be expressed as long non-coding RNAs (lncRNAs) nowadays. Different from most protein-coding MT members, MT1DP was here found that remarkably induced to provoke cytotoxicity in hepatocytes in response to Cd treatment. MT1DP exerted such a pro-apoptotic function in Cd-treated hepatocytes through interacting with two partners: RhoC and MT1H. On one hand, MT1DP interacted with RhoC protein to increase the latter’s stability by preventing lysosome-dependent protein degradation. Therefore, upon Cd stress, MT1DP/RhoC complex was quickly reinforced to activate RhoC-CCN1/2-AKT signaling and potentiate Ca2+ influx, leading to enhanced Cd uptake and elevated Cd toxicity. On the other hand, MT1H, a protein-coding member of the MT family with little known function, was found to quickly respond to Cd exposure along with MT1DP. Mechanistically, MT1H and MT1DP were uncovered to mutually protect each other through a reciprocal ceRNA mechanism, building up a positive feedback loop to enforce MT1DP-conducted signaling upon Cd exposure. Moreover, MT1DP was found to contribute much more to the activation of RhoC-CCN1/2-AKT signaling than MT1H. Considered together, we here unveiled a mystery whether a pseudogene within the MT family, MT1DP, has actual biological functions in regulating Cd-induced cellular defense. Our findings unearthed an important role of pseudogene MT1DP in calibrating the cellular machinery to switch the cellular defense to cytotoxicity through crosslinking an interplay between its two partners, namely MT1H and RhoC, under cadmium stress.
Collapse
|
24
|
Cullin 3-Based Ubiquitin Ligases as Master Regulators of Mammalian Cell Differentiation. Trends Biochem Sci 2017; 43:95-107. [PMID: 29249570 DOI: 10.1016/j.tibs.2017.11.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/23/2017] [Accepted: 11/25/2017] [Indexed: 01/09/2023]
Abstract
Specificity of the ubiquitin proteasome system is controlled by ubiquitin E3 ligases, including their major representatives, the multisubunit cullin-RING ubiquitin (Ub) ligases (CRLs). More than 200 different CRLs are divided into seven families according to their cullin scaffolding proteins (CUL1-7) around which they are assembled. Research over two decades has revealed that different CRL families are specialized to fulfill specific cellular functions. Whereas many CUL1-based CRLs (CRL1s) ubiquitylate cell cycle regulators, CRL4 complexes often associate with chromatin to control DNA metabolism. Based on studies about differentiation programs of mesenchymal stem cells (MSCs), including myogenesis, neurogenesis, chondrogenesis, osteogenesis and adipogenesis, we propose here that CRL3 complexes evolved to fulfill a pivotal role in mammalian cell differentiation.
Collapse
|
25
|
Carnesecchi J, Cerutti C, Vanacker JM, Forcet C. ERRα protein is stabilized by LSD1 in a demethylation-independent manner. PLoS One 2017; 12:e0188871. [PMID: 29190800 PMCID: PMC5708767 DOI: 10.1371/journal.pone.0188871] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022] Open
Abstract
The LSD1 histone demethylase is highly expressed in breast tumors where it constitutes a factor of poor prognosis and promotes traits of cancer aggressiveness such as cell invasiveness. Recent work has shown that the Estrogen-Related Receptor α (ERRα) induces LSD1 to demethylate the Lys 9 of histone H3. This results in the transcriptional activation of a number of common target genes, several of which being involved in cellular invasion. High expression of ERRα protein is also a factor of poor prognosis in breast tumors. Here we show that, independently of its demethylase activities, LSD1 protects ERRα from ubiquitination, resulting in overexpression of the latter protein. Our data also suggests that the elevation of LSD1 mRNA and protein in breast cancer (as compared to normal tissue) may be a key event to increase ERRα protein, independently of its corresponding mRNA.
Collapse
Affiliation(s)
- Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
26
|
Functional analysis of Cullin 3 E3 ligases in tumorigenesis. Biochim Biophys Acta Rev Cancer 2017; 1869:11-28. [PMID: 29128526 DOI: 10.1016/j.bbcan.2017.11.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 11/06/2017] [Accepted: 11/06/2017] [Indexed: 12/14/2022]
Abstract
Cullin 3-RING ligases (CRL3) play pivotal roles in the regulation of various physiological and pathological processes, including neoplastic events. The substrate adaptors of CRL3 typically contain a BTB domain that mediates the interaction between Cullin 3 and target substrates to promote their ubiquitination and subsequent degradation. The biological implications of CRL3 adaptor proteins have been well described where they have been found to play a role as either an oncogene, tumor suppressor, or can mediate either of these effects in a context-dependent manner. Among the extensively studied CRL3-based E3 ligases, the role of the adaptor protein SPOP (speckle type BTB/POZ protein) in tumorigenesis appears to be tissue or cellular context dependent. Specifically, SPOP acts as a tumor suppressor via destabilizing downstream oncoproteins in many malignancies, especially in prostate cancer. However, SPOP has largely an oncogenic role in kidney cancer. Keap1, another well-characterized CRL3 adaptor protein, likely serves as a tumor suppressor within diverse malignancies, mainly due to its specific turnover of its downstream oncogenic substrate, NRF2 (nuclear factor erythroid 2-related factor 2). In accordance with the physiological role the various CRL3 adaptors exhibit, several pharmacological agents have been developed to disrupt its E3 ligase activity, therefore blocking its potential oncogenic activity to mitigate tumorigenesis.
Collapse
|
27
|
He X, Ma S, Tian Y, Wei C, Zhu Y, Li F, Zhang P, Wang P, Zhang Y, Zhong H. ERRα negatively regulates type I interferon induction by inhibiting TBK1-IRF3 interaction. PLoS Pathog 2017; 13:e1006347. [PMID: 28591144 PMCID: PMC5476288 DOI: 10.1371/journal.ppat.1006347] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 06/19/2017] [Accepted: 04/11/2017] [Indexed: 12/21/2022] Open
Abstract
Estrogen-related receptor α (ERRα) is a member of the nuclear receptor superfamily controlling energy homeostasis; however, its precise role in regulating antiviral innate immunity remains to be clarified. Here, we showed that ERRα deficiency conferred resistance to viral infection both in vivo and in vitro. Mechanistically, ERRα inhibited the production of type-I interferon (IFN-I) and the expression of multiple interferon-stimulated genes (ISGs). Furthermore, we found that viral infection induced TBK1-dependent ERRα stabilization, which in turn associated with TBK1 and IRF3 to impede the formation of TBK1-IRF3, IRF3 phosphorylation, IRF3 dimerization, and the DNA binding affinity of IRF3. The effect of ERRα on IFN-I production was independent of its transcriptional activity and PCG-1α. Notably, ERRα chemical inhibitor XCT790 has broad antiviral potency. This work not only identifies ERRα as a critical negative regulator of antiviral signaling, but also provides a potential target for future antiviral therapy.
Collapse
Affiliation(s)
- Xiang He
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Shengli Ma
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Yinyin Tian
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
- Institute of Healthy Science, Anhui University, Hefei, Anhui, P.R. China
| | - Congwen Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Yongjie Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
- Institute of Healthy Science, Anhui University, Hefei, Anhui, P.R. China
| | - Feng Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
- Institute of Healthy Science, Anhui University, Hefei, Anhui, P.R. China
| | - Pingping Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Penghao Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Yanhong Zhang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| | - Hui Zhong
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, P.R. China
| |
Collapse
|
28
|
Seneviratne APB, Turan Z, Hermant A, Lecine P, Smith WO, Borg JP, Jaulin F, Kreitzer G. Modulation of estrogen related receptor alpha activity by the kinesin KIF17. Oncotarget 2017; 8:50359-50375. [PMID: 28881568 PMCID: PMC5584137 DOI: 10.18632/oncotarget.18104] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 05/12/2017] [Indexed: 12/26/2022] Open
Abstract
Estrogen-related receptor alpha (ERR1) is an orphan nuclear receptor that can bind transcriptional co-activators constitutively. ERR1 expression correlates with poor patient outcomes in breast cancer, heightening interest in this nuclear receptor as a therapeutic target. Because ERR1 has no known regulatory ligand, a major challenge in targeting its activity is to find cellular or synthetic modulators of its function. We identified an interaction between ERR1 and KIF17, a kinesin-2 family microtubule motor, in a yeast-2-hybrid screen. We confirmed the interaction using in vitro biochemical assays and determined that binding is mediated by the ERR1 ligand-binding/AF2 domain and the KIF17 C-terminal tail. Expression of KIF17 tail domain in either ER-negative or ER-positive breast cancer epithelial cells attenuated nuclear accumulation of newly synthesized ERR1 and inhibited ERR1 transcriptional activity. Conversely, ERR1 transcriptional activity was elevated significantly in KIF17 knock-out cells. Sequence analysis of the KIF17 tail domain revealed it contains a nuclear receptor box with a conserved LXXLL motif found in transcriptional co-activators. Expression of a 12 amino-acid peptide containing this motif was sufficient to inhibit ERR1 transcriptional activity and cell invasion, while deletion of this region from the KIF17 tail resulted in increased ERR1 activity. Together, these data suggest KIF17 modifies ERR1 function by two possible, non-exclusive mechanisms: (i) by regulating nuclear-cytoplasmic distribution or (ii) by competing with transcriptional co-activators for binding to ERR1. Thus targeting the ERR1-KIF17 interaction has potential as a novel strategy for treating breast cancer.
Collapse
Affiliation(s)
- Am Pramodh Bandara Seneviratne
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA.,Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,The City University of New York School of Medicine, New York, NY, USA
| | - Zeynep Turan
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,California Institute of Technology, Pasadena, CA, USA
| | - Aurelie Hermant
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Institut Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell Signalling and Cancer, Equipe labellisée Ligue Contre le Cancer, Marseille, France
| | - Patrick Lecine
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Institut Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell Signalling and Cancer, Equipe labellisée Ligue Contre le Cancer, Marseille, France.,BIOASTER, Tony Garnier, Lyon, France
| | - William O Smith
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA
| | - Jean-Paul Borg
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Institut Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell Signalling and Cancer, Equipe labellisée Ligue Contre le Cancer, Marseille, France
| | - Fanny Jaulin
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,Gustave Roussy Institute, Villejuif, France
| | - Geri Kreitzer
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA.,Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,The City University of New York School of Medicine, New York, NY, USA
| |
Collapse
|
29
|
Carnesecchi J, Forcet C, Zhang L, Tribollet V, Barenton B, Boudra R, Cerutti C, Billas IML, Sérandour AA, Carroll JS, Beaudoin C, Vanacker JM. ERRα induces H3K9 demethylation by LSD1 to promote cell invasion. Proc Natl Acad Sci U S A 2017; 114:3909-3914. [PMID: 28348226 PMCID: PMC5393192 DOI: 10.1073/pnas.1614664114] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lysine Specific Demethylase 1 (LSD1) removes mono- and dimethyl groups from lysine 4 of histone H3 (H3K4) or H3K9, resulting in repressive or activating (respectively) transcriptional histone marks. The mechanisms that control the balance between these two antagonist activities are not understood. We here show that LSD1 and the orphan nuclear receptor estrogen-related receptor α (ERRα) display commonly activated genes. Transcriptional activation by LSD1 and ERRα involves H3K9 demethylation at the transcriptional start site (TSS). Strikingly, ERRα is sufficient to induce LSD1 to demethylate H3K9 in vitro. The relevance of this mechanism is highlighted by functional data. LSD1 and ERRα coregulate several target genes involved in cell migration, including the MMP1 matrix metallo-protease, also activated through H3K9 demethylation at the TSS. Depletion of LSD1 or ERRα reduces the cellular capacity to invade the extracellular matrix, a phenomenon that is rescued by MMP1 reexpression. Altogether our results identify a regulatory network involving a direct switch in the biochemical activities of a histone demethylase, leading to increased cell invasion.
Collapse
Affiliation(s)
- Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Ling Zhang
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Bruno Barenton
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Rafik Boudra
- Genetics, Reproduction and Development, Université Blaise Pascal Clermont-Ferrand, CNRS UMR 6293, Inserm U1103, Centre de Recherche en Nutrition Humaine, F-63171 Aubière, France
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France
| | - Isabelle M L Billas
- Department of Integrative Structural Biology, Institute of Genetics and Molecular and Cellular Biology, CNRS UMR7104, Inserm U964, Université de Strasbourg, F-67404 Illkirch, France
| | - Aurélien A Sérandour
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, United Kingdom
| | - Claude Beaudoin
- Genetics, Reproduction and Development, Université Blaise Pascal Clermont-Ferrand, CNRS UMR 6293, Inserm U1103, Centre de Recherche en Nutrition Humaine, F-63171 Aubière, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, F-69007 Lyon, France;
| |
Collapse
|
30
|
Doan TB, Graham JD, Clarke CL. Emerging functional roles of nuclear receptors in breast cancer. J Mol Endocrinol 2017; 58:R169-R190. [PMID: 28087820 DOI: 10.1530/jme-16-0082] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 01/12/2017] [Indexed: 12/13/2022]
Abstract
Nuclear receptors (NRs) have been targets of intensive drug development for decades due to their roles as key regulators of multiple developmental, physiological and disease processes. In breast cancer, expression of the estrogen and progesterone receptor remains clinically important in predicting prognosis and determining therapeutic strategies. More recently, there is growing evidence supporting the involvement of multiple nuclear receptors other than the estrogen and progesterone receptors, in the regulation of various processes important to the initiation and progression of breast cancer. We review new insights into the mechanisms of action of NRs made possible by recent advances in genomic technologies and focus on the emerging functional roles of NRs in breast cancer biology, including their involvement in circadian regulation, metabolic reprogramming and breast cancer migration and metastasis.
Collapse
Affiliation(s)
- Tram B Doan
- Westmead Institute for Medical ResearchSydney Medical School - Westmead, University of Sydney, Sydney, New South Wales, Australia
| | - J Dinny Graham
- Westmead Institute for Medical ResearchSydney Medical School - Westmead, University of Sydney, Sydney, New South Wales, Australia
| | - Christine L Clarke
- Westmead Institute for Medical ResearchSydney Medical School - Westmead, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
31
|
Li Q, Zhu L, Zhang L, Chen H, Zhu Y, Du Y, Zhong W, Zhong M, Shi X. Inhibition of estrogen related receptor α attenuates vascular smooth muscle cell proliferation and migration by regulating RhoA/p27 Kip1 and β-Catenin/Wnt4 signaling pathway. Eur J Pharmacol 2017; 799:188-195. [PMID: 28213288 DOI: 10.1016/j.ejphar.2017.02.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 02/08/2017] [Accepted: 02/13/2017] [Indexed: 12/28/2022]
Abstract
RhoA/p27Kip1 and β-Catenin/Wnt4 signaling processes play central roles in proliferation and migration in vascular smooth muscle cells (VSMCs). ERRα, a member of orphan nuclear receptors, is a potent prognostic factor in breast, ovarian, colon and other types of tumors. However, biological significance of ERRα in VSMCs as well as the molecular mechanisms remains largely unknown. Therefore, the present study was designed to investigate whether ERRα is involved in the proliferation and migration of VSMCs in vitro and neointimal formation in vivo. The specific ERRα inverse agonist XCT790 (or ERRα shRNA) resulted in a significant inhibition of proliferation and phenotypic switch in cultured rat aortic SMCs (RASMCs). Furthermore, cycle progression, cell cycle protein transcription as well as hyperphosphorylation of the retinoblastoma protein (Rb) in RASMCs were prevented by downregulation of ERRα. Transwell assay demonstrated that migratory capacity of RASMCs was also inhibited the treatment of XCT790 (or ERRα shRNA). At the molecular levels, RhoA/p27Kip1 and β-Catenin/Wnt4 signaling pathways are involved in ERRα-mediated RASMCs growth and migration. Finally, inhibition of ERRα significantly attenuated neointimal formation in rat artery after balloon injury. These results help to further understand vascular remodeling and suggest that ERRα might be a potential target for the treatment of vascular proliferative diseases.
Collapse
Affiliation(s)
- Qunyi Li
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China.
| | - Lei Zhu
- Department of General Surgery, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Liudi Zhang
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Haifei Chen
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Yingfeng Zhu
- Department of Pathology, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Yongli Du
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan 250353, China
| | - Wanxian Zhong
- Department of Pharmacy, Jinshan Branch of the Sixth People's Hospital, Affiliated with Shanghai Jiaotong University, Shanghai 201500, China
| | - Mingkang Zhong
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China; Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Xiaojin Shi
- Department of Pharmacy, Huashan Hospital North, Fudan University, Shanghai 201907, China.
| |
Collapse
|
32
|
Zhang C, Wang HJ, Bao QC, Wang L, Guo TK, Chen WL, Xu LL, Zhou HS, Bian JL, Yang YR, Sun HP, Xu XL, You QD. NRF2 promotes breast cancer cell proliferation and metastasis by increasing RhoA/ROCK pathway signal transduction. Oncotarget 2016; 7:73593-73606. [PMID: 27713154 PMCID: PMC5342001 DOI: 10.18632/oncotarget.12435] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 09/26/2016] [Indexed: 12/26/2022] Open
Abstract
Nuclear factor erythroid 2-related factor (NRF2) is an important transcription factor in oxidative stress regulation. Overexpression of NRF2 is associated with human breast carcinogenesis, and increased NRF2 mRNA levels predict poor patient outcome for breast cancer. However, the mechanisms linking gain of NRF2 expression and poor prognosis in breast cancer are still unclear. Here, we provide evidence that NRF2 deletion inhibits proliferation and metastasis of breast cancer cells by down-regulating RhoA. Restoration of RhoA in MCF7 and MDA-MB-231 cells induced NRF2 knockdown-suppressed cell growth and metastasis in vitro, and NRF2 silencing suppressed stress fiber and focal adhesion formation leading to decreased cell migration and invasion. Mechanistic studies showed that NRF2 binds to the promoter region of estrogen-related receptor α (ERR1) and may function as a silencer. This may enhance RhoA protein stability and lead to RhoA overexpression in breast cancer cell. Our findings indicate that NRF2 silencing-mediated reduction of RhoA expression contributes, at least in part, to the poor outcome of breast cancer patients with high NRF2 expression.
Collapse
Affiliation(s)
- Chao Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Hui-Jie Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi-Chao Bao
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Lei Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Tian-Kun Guo
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Wei-Lin Chen
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Li-Li Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Hai-Shan Zhou
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Jin-Lei Bian
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Ying-Rui Yang
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Hao-Peng Sun
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiao-Li Xu
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qi-Dong You
- Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
33
|
Han L, Liu B, Jiang L, Liu J, Han S. MicroRNA-497 downregulation contributes to cell proliferation, migration, and invasion of estrogen receptor alpha negative breast cancer by targeting estrogen-related receptor alpha. Tumour Biol 2016; 37:13205-13214. [PMID: 27456360 DOI: 10.1007/s13277-016-5200-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023] Open
Abstract
Metastasis has become the main challenge for treatment of estrogen receptor alpha (ERα) negative breast cancer. Here, we found a negative correlation between miR-497 and estrogen-related receptor alpha (ERRα), a nuclear receptor overexpressed in ERα negative breast cancer. Targeted inhibition of ERRα by si-RNA increased miR-497 expression while overexpression of ERRα inhibited miR-497 expression. Further investigation showed that miR-497 targeted ERRα by binding to the 3'UTR region of ERRα. Luciferase assay and ChIP assay confirmed that ERα directly regulated the transcription of miR-497, suggesting that loss of ERα lowered miR-497 level in ERα negative breast cancer. Further, overexpression of miR-497 not only inhibited ERRα expression but also reduced MIF level and MMP9 activity, which led to significant decreases in cell proliferation, migration, and invasion of ERα negative breast cancer. Taken together, our findings suggested that, in ERα negative breast cancer, the low level of ERα reduced miR-497 expression, which promoted ERRα expression that enhanced cell proliferation, migration, and invasion by increasing MIF expression and MMP9 activity.
Collapse
Affiliation(s)
- Li Han
- Department of Internal Medicine Oncology, Shandong Tumor Hospital and Institute, No.440 Ji Yan Road, Jinan, 250117, China.
| | - Bo Liu
- Department of Internal Medicine Oncology, Shandong Tumor Hospital and Institute, No.440 Ji Yan Road, Jinan, 250117, China
| | - Lixi Jiang
- Department of Internal Medicine Oncology, Shandong Tumor Hospital and Institute, No.440 Ji Yan Road, Jinan, 250117, China
| | - Junyan Liu
- Department of Internal Medicine Oncology, Shandong Tumor Hospital and Institute, No.440 Ji Yan Road, Jinan, 250117, China
| | - Shumei Han
- Department of Internal Medicine Oncology, Shandong Tumor Hospital and Institute, No.440 Ji Yan Road, Jinan, 250117, China
| |
Collapse
|
34
|
Zhang L, Wong J, Vanacker JM. The estrogen-related receptors (ERRs): potential targets against bone loss. Cell Mol Life Sci 2016; 73:3781-7. [PMID: 27514376 PMCID: PMC11108346 DOI: 10.1007/s00018-016-2328-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 08/04/2016] [Indexed: 01/20/2023]
Abstract
Bone loss and the resulting skeletal fragility is induced by several pathological or natural conditions, the most prominent of which being aging as well as the decreased levels of circulating estrogens in post-menopause females. To date, most treatments against bone loss aim at preventing excess bone resorption. We here summarize data indicating that the estrogen-related receptors (ERRs) α and γ prevent bone formation. Inhibiting these receptors may thus constitute an anabolic approach by increasing bone formation.
Collapse
Affiliation(s)
- Ling Zhang
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR5242, Université de Lyon, Université Lyon I, Ecole Normale Supérieure de Lyon, Lyon, France
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiemin Wong
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, CNRS UMR5242, Université de Lyon, Université Lyon I, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
35
|
Griveau A, Devailly G, Eberst L, Navaratnam N, Le Calvé B, Ferrand M, Faull P, Augert A, Dante R, Vanacker JM, Vindrieux D, Bernard D. The PLA2R1-JAK2 pathway upregulates ERRα and its mitochondrial program to exert tumor-suppressive action. Oncogene 2016; 35:5033-42. [PMID: 27041564 DOI: 10.1038/onc.2016.43] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 01/06/2016] [Accepted: 02/08/2016] [Indexed: 12/20/2022]
Abstract
Little is known about the biological role of the phospholipase A2 receptor (PLA2R1) transmembrane protein. In recent years, PLA2R1 has been shown to have an important role in regulating tumor-suppressive responses via JAK2 activation, but the underlying mechanisms are largely undeciphered. In this study, we observed that PLA2R1 increases the mitochondrial content, judged by increased levels of numerous mitochondrial proteins, of the mitochondrial structural component cardiolipin, of the mitochondrial DNA content, and of the mitochondrial DNA replication and transcription factor TFAM. This effect of PLA2R1 relies on a transcriptional program controlled by the estrogen-related receptor alpha1 (ERRα) mitochondrial master regulator. Expression of ERRα and of its nucleus-encoded mitochondrial targets is upregulated upon PLA2R1 ectopic expression, and this effect is mediated by JAK2. Conversely, downregulation of PLA2R1 decreases the level of ERRα and of its nucleus-encoded mitochondrial targets. Finally, blocking the ERRα-controlled mitochondrial program largely inhibits the PLA2R1-induced tumor-suppressive response. Together, our data document ERRα and its mitochondrial program as downstream effectors of the PLA2R1-JAK2 pathway leading to oncosuppression.
Collapse
Affiliation(s)
- A Griveau
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - G Devailly
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - L Eberst
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - N Navaratnam
- Cellular Stress Group, MRC Clinical Sciences Centre, Faculty of Medicine, Imperial College, Hammersmith Campus, London, UK
| | - B Le Calvé
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - M Ferrand
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - P Faull
- Biological Mass Spectrometry and Proteomics Laboratory, MRC Clinical Sciences Centre, Imperial College London, London, UK
| | - A Augert
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - R Dante
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - J M Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - D Vindrieux
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| | - D Bernard
- Inserm U1052, Centre de Recherche en Cancérologie de Lyon, Lyon, France
- CNRS UMR 5286, Lyon, France
- Centre Léon Bérard, Lyon, France
- Université de Lyon, Lyon, France
| |
Collapse
|
36
|
Carnesecchi J, Vanacker JM. Estrogen-Related Receptors and the control of bone cell fate. Mol Cell Endocrinol 2016; 432:37-43. [PMID: 26206717 DOI: 10.1016/j.mce.2015.07.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/23/2015] [Accepted: 07/17/2015] [Indexed: 11/17/2022]
Abstract
Bone loss is naturally occurring in aging males and females and exacerbated in the latter after menopause, altogether leading to cumulative skeleton fragility and increased fracture risk. Two types of therapeutic strategies can be envisioned to counteract age- or menopause-associated bone loss, aiming at either reducing bone resorption exerted by osteoclasts or, alternatively, promoting bone formation by osteoblasts. We here summarize data suggesting that inhibition of the Estrogen-Related Receptors α and/or γ could promote bone formation and compensate for bone loss induced by ageing or estrogen-deficiency.
Collapse
Affiliation(s)
- Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
37
|
Wu D, Cheung A, Wang Y, Yu S, Chan FL. The emerging roles of orphan nuclear receptors in prostate cancer. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1866:23-36. [PMID: 27264242 DOI: 10.1016/j.bbcan.2016.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 05/31/2016] [Accepted: 06/01/2016] [Indexed: 12/25/2022]
Abstract
Orphan nuclear receptors are members of the nuclear receptor (NR) superfamily and are so named because their endogenous physiological ligands are either unknown or may not exist. Because of their important regulatory roles in many key physiological processes, dysregulation of signalings controlled by these receptors is associated with many diseases including cancer. Over years, studies of orphan NRs have become an area of great interest because their specific physiological and pathological roles have not been well-defined, and some of them are promising drug targets for diseases. The recently identified synthetic small molecule ligands, acting as agonists or antagonists, to these orphan NRs not only help to understand better their functional roles but also highlight that the signalings mediated by these ligand-independent NRs in diseases could be therapeutically intervened. This review is a summary of the recent advances in elucidating the emerging functional roles of orphan NRs in cancers, especially prostate cancer. In particular, some orphan NRs, RORγ, TR2, TR4, COUP-IFII, ERRα, DAX1 and SHP, exhibit crosstalk or interference with androgen receptor (AR) signaling in either normal or malignant prostatic cells, highlighting their involvement in prostate cancer progression as androgen and AR signaling pathway play critical roles in this process. We also propose that a better understanding of the mechanism of actions of these orphan NRs in prostate gland or prostate cancer could help to evaluate their potential value as therapeutic targets for prostate cancer.
Collapse
Affiliation(s)
- Dinglan Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Alyson Cheung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Shan Yu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| | - Franky L Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
38
|
Nass N, Dittmer A, Hellwig V, Lange T, Beyer JM, Leyh B, Ignatov A, Weiβenborn C, Kirkegaard T, Lykkesfeldt AE, Kalinski T, Dittmer J. Expression of transmembrane protein 26 (TMEM26) in breast cancer and its association with drug response. Oncotarget 2016; 7:38408-38426. [PMID: 27224909 PMCID: PMC5122400 DOI: 10.18632/oncotarget.9493] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/29/2016] [Indexed: 12/18/2022] Open
Abstract
We have previously shown that stromal cells desensitize breast cancer cells to the anti-estrogen fulvestrant and, along with it, downregulate the expression of TMEM26 (transmembrane protein 26). In an effort to study the function and regulation of TMEM26 in breast cancer cells, we found that breast cancer cells express non-glycosylated and N-glycosylated isoforms of the TMEM26 protein and demonstrate that N-glycosylation is important for its retention at the plasma membrane. Fulvestrant induced significant changes in expression and in the N-glycosylation status of TMEM26. In primary breast cancer, TMEM26 protein expression was higher in ERα (estrogen receptor α)/PR (progesterone receptor)-positive cancers. These data suggest that ERα is a major regulator of TMEM26. Significant changes in TMEM26 expression and N-glycosylation were also found, when MCF-7 and T47D cells acquired fulvestrant resistance. Furthermore, patients who received aromatase inhibitor treatment tend to have a higher risk of recurrence when tumoral TMEM26 protein expression is low. In addition, TMEM26 negatively regulates the expression of integrin β1, an important factor involved in endocrine resistance. Data obtained by spheroid formation assays confirmed that TMEM26 and integrin β1 can have opposite effects in breast cancer cells. These data are consistent with the hypothesis that, in ERα-positive breast cancer, TMEM26 may function as a tumor suppressor by impeding the acquisition of endocrine resistance. In contrast, in ERα-negative breast cancer, particularly triple-negative cancer, high TMEM26 expression was found to be associated with a higher risk of recurrence. This implies that TMEM26 has different functions in ERα-positive and -negative breast cancer.
Collapse
Affiliation(s)
- Norbert Nass
- Otto-von-Guericke-Universität Magdeburg, Institut für Pathologie, Magdeburg, Germany
| | - Angela Dittmer
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Vicky Hellwig
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Theresia Lange
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Johanna Mirjam Beyer
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Benjamin Leyh
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| | - Atanas Ignatov
- Otto-von-Guericke-Universität Magdeburg, Universitätsfrauenklinik, Magdeburg, Germany
| | - Christine Weiβenborn
- Otto-von-Guericke-Universität Magdeburg, Universitätsfrauenklinik, Magdeburg, Germany
| | - Tove Kirkegaard
- Breast Cancer Group, Cell Death and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark.,Present address: Department of Surgery, Koege Hospital, Koege, Denmark
| | - Anne E Lykkesfeldt
- Breast Cancer Group, Cell Death and Metabolism, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Thomas Kalinski
- Otto-von-Guericke-Universität Magdeburg, Institut für Pathologie, Magdeburg, Germany
| | - Jürgen Dittmer
- Klinik für Gynäkologie, Martin-Luther-Universität Halle-Wittenberg, Halle/Saale, Germany
| |
Collapse
|
39
|
Li Q, Wang Y, Zhang L, Chen L, Du Y, Ye T, Shi X. Naringenin exerts anti-angiogenic effects in human endothelial cells: Involvement of ERRα/VEGF/KDR signaling pathway. Fitoterapia 2016; 111:78-86. [PMID: 27105956 DOI: 10.1016/j.fitote.2016.04.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 04/16/2016] [Accepted: 04/18/2016] [Indexed: 12/20/2022]
Abstract
Naringenin (Nar), most abundant in oranges and tomatoes, are known for the hypocholesterolemic, anti-estrogenic, hypolipidemic, anti-hypertensive, and anti-inflammatory activities. Here, the present study was designed to investigate the in vitro and in vivo anti-angiogenesis of Nar. Inhibition of angiogenesis was determined in vitro by using proliferation, apoptosis, migration, and tube-formation assays in Nar-treated human endothelial cell. Finally, CAM assays were used to assess inhibitory effect of Nar on physiological angiogenesis in vivo. The data suggest that Nar should be a direct ERRα inhibitor capable of inhibiting angiogenesis in vitro and in vivo, including endothelial cell proliferation, survival, migration and capillary-like structures formation of HUVECs, as well as reduced neovascularization of the CAM. Furthermore, the effects exerted by Nar are cell cycle related and mediated by VEGF/KDR signaling pathway along with downregulation of certain proangiogenic inflammatory cytokines. Our data thus provide potential molecular mechanisms through which Nar manifests it as a promising anti-angiogenic and anti-cancer agent.
Collapse
Affiliation(s)
- Qunyi Li
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China; Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China.
| | - Yi Wang
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Liudi Zhang
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Lu Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Yongli Du
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan 250353, China
| | - Ting Ye
- General Surgery Unit, Huashan Hospital, Fudan University, 12 Wu Lu Mu Qi M Road, Shanghai 200040, China.
| | - Xiaojin Shi
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China; Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| |
Collapse
|
40
|
Tribollet V, Barenton B, Kroiss A, Vincent S, Zhang L, Forcet C, Cerutti C, Périan S, Allioli N, Samarut J, Vanacker JM. miR-135a Inhibits the Invasion of Cancer Cells via Suppression of ERRα. PLoS One 2016; 11:e0156445. [PMID: 27227989 PMCID: PMC4881992 DOI: 10.1371/journal.pone.0156445] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/13/2016] [Indexed: 01/04/2023] Open
Abstract
MicroRNA-135a (miR-135a) down-modulates parameters of cancer progression and its expression is decreased in metastatic breast cancers (as compared to non-metastatic tumors) as well as in prostate tumors relative to normal tissue. These expression and activity patterns are opposite to those of the Estrogen-Related Receptor α (ERRα), an orphan member of the nuclear receptor family. Indeed high expression of ERRα correlates with poor prognosis in breast and prostate cancers, and the receptor promotes various traits of cancer aggressiveness including cell invasion. Here we show that miR-135a down-regulates the expression of ERRα through specific sequences of its 3'UTR. As a consequence miR-135a also reduces the expression of downstream targets of ERRα. miR-135a also decreases cell invasive potential in an ERRα-dependent manner. Our results suggest that the decreased expression of miR-135a in metastatic tumors leads to elevated ERRα expression, resulting in increased cell invasion capacities.
Collapse
Affiliation(s)
- Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Bruno Barenton
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Auriane Kroiss
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Séverine Vincent
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Ling Zhang
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Catherine Cerutti
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Séverine Périan
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Nathalie Allioli
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
- Institut des Sciences Pharmaceutiques et Biologiques, Faculté de Pharmacie, Université de Lyon, Lyon, France
| | - Jacques Samarut
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
- Faculté de Médecine Lyon-Sud, Université de Lyon, Lyon, France
- UMOMT, Centre Hospitalier Lyon-Sud, Hospices Civils de Lyon, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| |
Collapse
|
41
|
Wertz MH, Winden K, Neveu P, Ng SY, Ercan E, Sahin M. Cell-type-specific miR-431 dysregulation in a motor neuron model of spinal muscular atrophy. Hum Mol Genet 2016; 25:2168-2181. [PMID: 27005422 DOI: 10.1093/hmg/ddw084] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 03/11/2016] [Indexed: 12/17/2022] Open
Abstract
Spinal muscular atrophy (SMA) is an autosomal-recessive pediatric neurodegenerative disease characterized by selective loss of spinal motor neurons. It is caused by mutation in the survival of motor neuron 1, SMN1, gene and leads to loss of function of the full-length SMN protein. microRNAs (miRNAs) are small RNAs that are involved in post-transcriptional regulation of gene expression. Prior studies have implicated miRNAs in the pathogenesis of motor neuron disease. We hypothesized that motor neuron-specific miRNA expression changes are involved in their selective vulnerability in SMA. Therefore, we sought to determine the effect of SMN loss on miRNAs and their target mRNAs in spinal motor neurons. We used microarray and RNAseq to profile both miRNA and mRNA expression in primary spinal motor neuron cultures after acute SMN knockdown. By integrating the miRNA:mRNA profiles, a number of dysregulated miRNAs were identified with enrichment in differentially expressed putative mRNA targets. miR-431 expression was highly increased, and a number of its putative mRNA targets were significantly downregulated in motor neurons after SMN loss. Further, we found that miR-431 regulates motor neuron neurite length by targeting several molecules previously identified to play a role in motor neuron axon outgrowth, including chondrolectin. Together, our findings indicate that cell-type-specific dysregulation of miR-431 plays a role in the SMA motor neuron phenotype.
Collapse
Affiliation(s)
- Mary H Wertz
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kellen Winden
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pierre Neveu
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, 69117 Heidelberg, Germany
| | - Shi-Yan Ng
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA Neurotherapeutics Laboratory, Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
| | - Ebru Ercan
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mustafa Sahin
- Department of Neurology, The F. M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
42
|
Wu YM, Chen ZJ, Jiang GM, Zhang KS, Liu Q, Liang SW, Zhou Y, Huang HB, Du J, Wang HS. Inverse agonist of estrogen-related receptor α suppresses the growth of triple negative breast cancer cells through ROS generation and interaction with multiple cell signaling pathways. Oncotarget 2016; 7:12568-81. [PMID: 26871469 PMCID: PMC4914305 DOI: 10.18632/oncotarget.7276] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/23/2016] [Indexed: 12/19/2022] Open
Abstract
There is an urgent clinical need for targeted therapy approaches for triple-negative breast cancer (TNBC) patients. Increasing evidences suggested that the expression of estrogen-related receptor alpha (ERRα) was correlate with unfavorable clinical outcomes of breast cancer patients. We here show that inhibition of ERRα by its inverse agonist XCT-790 can suppress the proliferation, decrease G2/M phases, and induce mitochondrial-related apoptosis of TNBC cells. XCT-790 elevates the proteins related to endoplasmic reticulum (ER) stress such as ATF4/6, XBT-1 and CHOP. It also increases the expression of growth inhibition related proteins such as p53 and p21. Further, XCT-790 can increase the generation of reactive oxygen species (ROS) in TNBC cells mainly through inhibition of SOD1/2. While ROS scavenger NAC abolishes XCT-790 induced ER-stress and growth arrest. XCT-790 treatment can rapidly activate the signal molecules including ERK1/2, p38-MAPK, JNK, Akt, p65, and IκBα, while NAC attenuates effects of XCT-790 induced phosphorylation of ERK1/2, p38-MAPK and Akt. Further, the inhibitors of ERK1/2, JNK, Akt, and NF-κB attenuate XCT-790 induced ROS generation. These data suggest that AKT/ROS and ERK/ROS positive feedback loops, NF-κB/ROS, and ROS/p38-MAPK, are activated in XCT-790 treated TNBC cells. In vivo experiments show that XCT-790 significantly suppresses the growth of MDA-MB-231 xenograft tumors, which is associated with up regulation of p53, p21, ER-stress related proteins while down regulation of bcl-2. The present discovery makes XCT-790 a promising candidate drug and lays the foundation for future development of ERRα-based therapies for TNBC patients.
Collapse
Affiliation(s)
- Ying-Min Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Zhuo-Jia Chen
- Department of Pharmacy, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Guan-Min Jiang
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Kun-Shui Zhang
- Department of Pharmacy, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Qiao Liu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Shu-Wei Liang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Yan Zhou
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hong-Bin Huang
- Department of Pharmacy, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| |
Collapse
|
43
|
Gladwyn-Ng I, Huang L, Ngo L, Li SS, Qu Z, Vanyai HK, Cullen HD, Davis JM, Heng JIT. Bacurd1/Kctd13 and Bacurd2/Tnfaip1 are interacting partners to Rnd proteins which influence the long-term positioning and dendritic maturation of cerebral cortical neurons. Neural Dev 2016; 11:7. [PMID: 26969432 PMCID: PMC4788816 DOI: 10.1186/s13064-016-0062-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 02/28/2016] [Indexed: 11/10/2022] Open
Abstract
Background The development of neural circuits within the embryonic cerebral cortex relies on the timely production of neurons, their positioning within the embryonic cerebral cortex as well as their terminal differentiation and dendritic spine connectivity. The RhoA GTPases Rnd2 and Rnd3 are important for neurogenesis and cell migration within the embryonic cortex (Nat Commun 4:1635, 2013), and we recently identified the BTB/POZ domain-containing Adaptor for Cul3-mediated RhoA Degradation family member Bacurd2 (also known as Tnfaip1) as an interacting partner to Rnd2 for the migration of embryonic mouse cortical neurons (Neural Dev 10:9, 2015). Findings We have extended this work and report that Bacurd1/Kctd13 and Bacurd2/Tnfaip1 are interacting partners to Rnd2 and Rnd3 in vitro. Given that these genes are expressed during cortical development, we performed a series of in utero electroporation studies in mice and found that disruptions to Bacurd1/Kctd13 or Bacurd2/Tnfaip1 expression impair the long-term positioning of E14.5-born cortical neurons within the postnatal (P17) mouse cerebral cortex. We also find that forced expression of Bacurd1/Kctd13 and Bacurd2/Tnfaip1 alters the branching and dendritic spine properties of layer II/III projection neurons. Conclusions We identify Bacurd1/Kctd13 and Bacurd2/Tnfaip1 as interacting partners to Rnd proteins which influence the development of cortical neurons. Their neurodevelopmental functions are likely to be relevant to human brain development and disease. Electronic supplementary material The online version of this article (doi:10.1186/s13064-016-0062-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ivan Gladwyn-Ng
- EMBL-Australia, The Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia.,The Harry Perkins Institute of Medical Research, 6 Verdun St, Crawley, WA, 6009, Australia.,The Centre for Medical Research, The University of Western Australia, Crawley Avenue, Crawley, WA, 6009, Australia
| | - Lieven Huang
- EMBL-Australia, The Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Linh Ngo
- EMBL-Australia, The Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia.,The Harry Perkins Institute of Medical Research, 6 Verdun St, Crawley, WA, 6009, Australia.,The Centre for Medical Research, The University of Western Australia, Crawley Avenue, Crawley, WA, 6009, Australia
| | - Shan Shan Li
- EMBL-Australia, The Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Zhengdong Qu
- EMBL-Australia, The Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Hannah Kate Vanyai
- The Harry Perkins Institute of Medical Research, 6 Verdun St, Crawley, WA, 6009, Australia.,The Centre for Medical Research, The University of Western Australia, Crawley Avenue, Crawley, WA, 6009, Australia
| | - Hayley Daniella Cullen
- The Harry Perkins Institute of Medical Research, 6 Verdun St, Crawley, WA, 6009, Australia.,The Centre for Medical Research, The University of Western Australia, Crawley Avenue, Crawley, WA, 6009, Australia
| | - John Michael Davis
- EMBL-Australia, The Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia
| | - Julian Ik-Tsen Heng
- EMBL-Australia, The Australian Regenerative Medicine Institute, Monash University, Wellington Road, Clayton, VIC, 3800, Australia. .,The Harry Perkins Institute of Medical Research, 6 Verdun St, Crawley, WA, 6009, Australia. .,The Centre for Medical Research, The University of Western Australia, Crawley Avenue, Crawley, WA, 6009, Australia.
| |
Collapse
|
44
|
Tam IS, Giguère V. There and back again: The journey of the estrogen-related receptors in the cancer realm. J Steroid Biochem Mol Biol 2016; 157:13-9. [PMID: 26151739 DOI: 10.1016/j.jsbmb.2015.06.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/09/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022]
Abstract
The identification of two genes encoding polypeptides with structural features common with the estrogen receptor more than a quarter century ago, referred to as the estrogen-related receptors (ERRs), subsequently led to the discovery of several previously unrecognized hormone responsive systems through the application of reverse endocrinology. Paradoxically, the natural ligand(s) associated with members of the ERR subfamily remains to be identified. While initial studies on the mode of action and physiological functions of the ERRs focused on interaction with estrogen signalling in breast cancer, subsequent work showed that the ERRs are ubiquitous master regulators of cellular energy metabolism. This review aims to demonstrate that the ERRs occupy a central node at the interface of cancer and metabolism, and that modulation of their activity may represent a worthwhile strategy to induce metabolic vulnerability in tumors of various origins and thus achieve a more comprehensive response to current therapies.
Collapse
Affiliation(s)
- Ingrid S Tam
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montréal, QC H3A 1A3, Canada
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montréal, QC H3A 1A3, Canada; Departments of Biochemistry, Medicine and Oncology, McGill University, Montréal, PQ H3G 1Y6, Canada.
| |
Collapse
|
45
|
Chen ZJ, Wei W, Jiang GM, Liu H, Wei WD, Yang X, Wu YM, Liu H, Wong CKC, Du J, Wang HS. Activation of GPER suppresses epithelial mesenchymal transition of triple negative breast cancer cells via NF-κB signals. Mol Oncol 2016; 10:775-88. [PMID: 26842883 DOI: 10.1016/j.molonc.2016.01.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/09/2016] [Accepted: 01/09/2016] [Indexed: 12/13/2022] Open
Abstract
The targeted therapy for triple-negative breast cancer (TNBC) is a great challenge due to our poor understanding on its molecular etiology. In the present study, our clinical data showed that the expression of G-protein coupled estrogen receptor (GPER) is negatively associated with lymph node metastasis, high-grade tumor and fibronectin (FN) expression while positively associated with the favorable outcome in 135 TNBC patients. In our experimental studies, both the in vitro migration and invasion of TNBC cells were inhibited by GPER specific agonist G-1, through the suppression of the epithelial mesenchymal transition (EMT). The G-1 treatment also reduced the phosphorylation, nuclear localization, and transcriptional activities of NF-κB. While over expression of NF-κB attenuated the action of G-1 in suppressing EMT. Our data further illustrated that the phosphorylation of GSK-3β by PI3K/Akt and ERK1/2 mediated, at least partially, the inhibitory effect of G-1 on NF-κB activities. It was further confirmed in a study of MDA-MB-231 tumor xenografts in nude mice. The data showed that G-1 inhibited the in vivo growth and invasive potential of TNBC via suppression of EMT. Our present study demonstrated that an activation of GPER pathway elicits tumor suppressive actions on TNBC, and supports the use of G-1 therapeutics for TNBC metastasis.
Collapse
Affiliation(s)
- Zhuo-Jia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Wei Wei
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Guan-Min Jiang
- Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, China
| | - Hao Liu
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou 510095, China
| | - Wei-Dong Wei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Xiangling Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Clinical Laboratory, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying-Min Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Huanliang Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Department of Clinical Laboratory, Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China; Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Chris K C Wong
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
46
|
Zhang LD, Chen L, Zhang M, Qi HJ, Chen L, Chen HF, Zhong MK, Shi XJ, Li QY. Downregulation of ERRα inhibits angiogenesis in human umbilical vein endothelial cells through regulating VEGF production and PI3K/Akt/STAT3 signaling pathway. Eur J Pharmacol 2015; 769:167-76. [PMID: 26586335 DOI: 10.1016/j.ejphar.2015.11.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 11/12/2015] [Indexed: 12/20/2022]
Abstract
The human estrogen related receptor α (ERRα) is a pivotal regulator involved in energy homeostasis and mitochondrial biogenesis. It has been demonstrated that activation of ERRα in various breast cancer cells results in a significant increase of vascular endothelial growth factor (VEGF) mRNA and protein secretion. However, little is known about the relationship between ERRα and angiogenesis. Thus, the present study is aimed to investigate the effects and mechanism of ERRα suppression on the angiogenesis in human umbilical vein endothelial cells (HUVECs). Here we show that ERRα suppression powerfully inhibits proliferation, migration and capillary-like structures formation of HUVECs. Importantly, we demonstrate that these inhibitory effects are associated with the significantly reduced expression and production of VEGF. Results from further experiments using western blot and luciferase reporter assay exhibit that ERRα suppression inhibits hypoxia-inducible factor 1α (HIF-1α) expression, and phosphorylation of protein kinase B (Akt) and signal transducer and activator of transcription (STAT3) which up-regulated VEGF expression. In summary, we show that ERRα suppression inhibits angiogenesis in HUVECs and deserves further studies for application of rationale therapeutic target for patient with diseases related with aberrant angiogenesis.
Collapse
Affiliation(s)
- Liu-Di Zhang
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 12 Wu Lu Mu Qi M Road, Shanghai 200040, China; Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| | - Li Chen
- Pharmacy Department, Xuhui District Central Hospital, 966 Huai Hai M Road, Shanghai 200031, China
| | - Meng Zhang
- Brunswick Laboratories (China), 5 Xing Han Road, Suzhou Industrial Park 215021, China
| | - Hui-Jie Qi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| | - Lu Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| | - Hai-Fei Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| | - Ming-Kang Zhong
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 12 Wu Lu Mu Qi M Road, Shanghai 200040, China; Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China
| | - Xiao-Jin Shi
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 12 Wu Lu Mu Qi M Road, Shanghai 200040, China; Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China.
| | - Qun-Yi Li
- Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, 12 Wu Lu Mu Qi M Road, Shanghai 200040, China; Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, 108 Luxiang Road, Shanghai 201907, China.
| |
Collapse
|
47
|
Chen Y, Lian P, Liu Y, Xu K. Pachymic acid inhibits tumorigenesis in gallbladder carcinoma cells. Int J Clin Exp Med 2015; 8:17781-17788. [PMID: 26770369 PMCID: PMC4694269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/08/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Gallbladder cancer, with high aggressivity and extremely poor prognosis, is the most common malignancy of the bile duct. Thus, seeking targets gallbladder tumor cells is an attractive goal towards improving clinical treatment. MATERIAL AND METHODS In this study, we investigated the effects of pachymic acid (PA) on the tumorigenesis of human gallbladder cancer cells. RESULTS We found that PA significantly reduced cell growth in a dose- and time-dependent fashion. Meanwhile, cell cycle arrest at G0 phase was induced by PA. PA also significantly inhibited cancer cell migration, invasion in a dose-dependent manner. Interestingly, we demonstrated that cancer cell adhesion ability was suppressed dose-dependently, which may contribute to the inhibition of cell invasion. Finally, we showed that PA inhibited AKT and ERK signaling pathways. And oncoproteins, such as PCNA, ICAM-1 and RhoA which are involved intumorigenesis, were also downregulated by PA. CONCLUSION Our study reveals that PA is able to inhibit gallbladder cancer tumorigenesis involving affection of AKT and ERK signaling pathways. Together, these results encourage further studies of PA as a promising candidate for gallbladder cancer therapy.
Collapse
Affiliation(s)
- Yueguang Chen
- Department of General Surgery, Qilu Hospital, Shandong University Jinan 250012, P. R. China
| | - Peilong Lian
- Department of General Surgery, Qilu Hospital, Shandong University Jinan 250012, P. R. China
| | - Yanfeng Liu
- Department of General Surgery, Qilu Hospital, Shandong University Jinan 250012, P. R. China
| | - Kesen Xu
- Department of General Surgery, Qilu Hospital, Shandong University Jinan 250012, P. R. China
| |
Collapse
|
48
|
Wu YM, Chen ZJ, Liu H, Wei WD, Lu LL, Yang XL, Liang WT, Liu T, Liu HL, Du J, Wang HS. Inhibition of ERRα suppresses epithelial mesenchymal transition of triple negative breast cancer cells by directly targeting fibronectin. Oncotarget 2015; 6:25588-601. [PMID: 26160845 PMCID: PMC4694852 DOI: 10.18632/oncotarget.4436] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/16/2015] [Indexed: 12/03/2022] Open
Abstract
Triple-negative breast cancer (TNBC) patients have poor prognosis due to the aggressive metastatic behaviors. Our study reveals that expression of estrogen related receptor α (ERRα) is significantly (p < 0.01) positively associated with high grade tumors and lymph node metastasis, while negatively correlated with overall survival (OS), in 138 TNBC patients. Targeted inhibition of ERRα by its inverse agonist XCT-790 or si-RNA obviously inhibits in vitro motility of TNBC cells. While over expression of ERRα triggers the invasion and migration of TNBC cells. Further, si-ERRα and XCT-790 inhibit the epithelial mesenchymal transition (EMT) of TNBC cells with increasing the expression of E-cadherin and decreasing fibronectin (FN) and vimentin. While XCT-790 has no effect on the expression of EMT related transcription factors such as Snail or Slug. Further, inhibitors of MAPK, PI3K/Akt, NF-κB signal molecules, which are activated by XCT-790, can not attenuate the suppression effects of XCT-790 on EMT. Alternatively, luciferase reporter gene assays and ChIP analysis indicate that ERRα can directly bind with FN promoter at ERR response element-3 (ERRE-1), ERRE-3, and ERRE-4, while XCT-790 reduces this bond. In vivo data show that ERRα expression is significantly (p < 0.05) correlated with FN in clinical TNBC patients. In MDA-MB-231 tumor xenograft models, XCT-790 decreases the expression of FN, inhibits the growth and lung metastasis, and suppresses the EMT. Our results demonstrate that ERRα functions as a metastasis stimulator and its targeted inhibition may be a new therapeutic strategy for TNBC treatment.
Collapse
Affiliation(s)
- Ying-Min Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zhuo-Jia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Hao Liu
- Cancer Research Institute and Cancer Hospital, Guangzhou Medical University, Guangzhou 510095, China
| | - Wei-Dong Wei
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Lin-Lin Lu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Xiang-Ling Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei-Ting Liang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Tao Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Huan-Liang Liu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, Guangdong Institute of Gastroenterology and The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
- Institute of Human Virology and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, Guangzhou 510080, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hong-Sheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|