1
|
Xu J, Wakai M, Xiong K, Yang Y, Prabakaran A, Wu S, Ahrens D, Molina-Portela MDP, Ni M, Bai Y, Shavlakadze T, Glass DJ. The pro-inflammatory cytokine IL6 suppresses mitochondrial function via the gp130-JAK1/STAT1/3-HIF1α/ERRα axis. Cell Rep 2025; 44:115403. [PMID: 40056415 DOI: 10.1016/j.celrep.2025.115403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/07/2024] [Accepted: 02/14/2025] [Indexed: 03/10/2025] Open
Abstract
Chronic inflammation and a decline in mitochondrial function are hallmarks of aging. Here, we show that the two mechanisms may be linked. We found that interleukin-6 (IL6) suppresses mitochondrial function in settings where PGC1 (both PGC1α and PGC1β) expression is low. This suppression is mediated by the JAK1/STAT1/3 axis, which activates HIF1α through non-canonical mechanisms involving upregulation of HIF1A and ERRα transcription, and subsequent stabilization of the HIF1A protein by ERRα. HIF1α, in turn, inhibits ERRα, which is a master regulator of mitochondrial biogenesis, thus contributing to the inhibition of mitochondrial function. When expressed at higher levels, PGC1 rescues ERRα to boost baseline mitochondrial respiration, including under IL6-treated conditions. Our study suggests that inhibition of the IL6 signaling axis could be a potential treatment for those inflammatory settings where mitochondrial function is compromised.
Collapse
Affiliation(s)
- Jianing Xu
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| | - Matthew Wakai
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Kun Xiong
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yanfeng Yang
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Adithya Prabakaran
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Sophia Wu
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Diana Ahrens
- Research Flow Cytometry Core, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | | | - Min Ni
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Yu Bai
- Molecular Profiling & Data Science, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA
| | - Tea Shavlakadze
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| | - David J Glass
- Aging/Age-Related Diseases, Regeneron Pharmaceuticals, 777 Old Saw Mill River Road, Tarrytown, NY, USA.
| |
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
|
Heidarian Y, Fasteen TD, Mungcal L, Buddika K, Mahmoudzadeh NH, Nemkov T, D'Alessandro A, Tennessen JM. Hypoxia-inducible factor 1α is required to establish the larval glycolytic program in Drosophila melanogaster. Mol Metab 2025; 93:102106. [PMID: 39894213 PMCID: PMC11869853 DOI: 10.1016/j.molmet.2025.102106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025] Open
Abstract
OBJECTIVES The rapid growth that occurs during Drosophila larval development requires a dramatic rewiring of central carbon metabolism to support biosynthesis. Larvae achieve this metabolic state, in part, by coordinately up-regulating the expression of genes involved in carbohydrate metabolism. The resulting metabolic program exhibits hallmark characteristics of aerobic glycolysis and establishes a physiological state that supports growth. To date, the only factor known to activate the larval glycolytic program is the Drosophila Estrogen-Related Receptor (dERR). However, dERR is dynamically regulated during the onset of this metabolic switch, indicating that other factors must be involved. Here we examine the possibility that the Drosophila ortholog of Hypoxia inducible factor 1α (Hif1α) is also required to activate the larval glycolytic program. METHODS CRISPR/Cas9 was used to generate new loss-of-function alleles in the Drosophila gene similar (sima), which encodes the sole fly ortholog of Hif1α. The resulting mutant strains were analyzed using a combination of metabolomics and RNAseq for defects in carbohydrate metabolism. RESULTS Our studies reveal that sima mutants fail to activate aerobic glycolysis and die during larval development with metabolic phenotypes that mimic those displayed by dERR mutants. Moreover, we demonstrate that dERR and Sima/Hif1α protein accumulation is mutually dependent, as loss of either transcription factor results in decreased abundance of the other protein. CONCLUSIONS These findings demonstrate that Sima/HIF1α is required during embryogenesis to coordinately up-regulate carbohydrate metabolism in preparation for larval growth. Notably, our study also reveals that the Sima/HIF1α-dependent gene expression program shares considerable overlap with that observed in dERR mutant, suggesting that Sima/HIF1α and dERR cooperatively regulate embryonic and larval glycolytic gene expression.
Collapse
Affiliation(s)
- Yasaman Heidarian
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Tess D Fasteen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Liam Mungcal
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA; Member, Melvin and Bren Simon Cancer Center, Indianapolis, IN 46202, USA.
| |
Collapse
|
4
|
Ren Y, Mao X, Lin W, Chen Y, Chen R, Sun P. Targeting estrogen-related receptors to mitigate tumor resistance: A comprehensive approach to bridging cellular energy metabolism. Biochim Biophys Acta Rev Cancer 2025; 1880:189256. [PMID: 39743156 DOI: 10.1016/j.bbcan.2024.189256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
The war between humanity and malignant tumors has been ongoing, with continuous advancements in classic chemotherapy and radiotherapy regimens, targeted drugs, endocrine therapy, and immunotherapy. However, tumor cells can develop primary or secondary resistance to these treatment options, making the issue of tumor resistance a major factor affecting patient prognosis and leading to recurrence. Estrogen-related receptors (ERRs) are members of the nuclear receptor superfamily, primarily involved in regulating glucose, lipid, and amino acid metabolism, serving as a central hub for intracellular energy metabolism. ERRs not only mediate insulin resistance but also participate in the mechanisms of drug resistance in various tumors, including breast cancer, osteosarcoma, endometrial cancer, lung cancer, and liver cancer, and even mediate resistance to radiation and immunotherapy. They mainly resist tumor treatment methods through metabolic reprogramming within cells, affecting mitochondrial energy metabolism, regulating metabolites such as cholesterol, glutamine, and lactate, or other signaling pathways, or by influencing the immune microenvironment. ERRs are promising targets for addressing the dilemma of tumor resistance. Currently, electrochemical luminescence biosensors for detecting ERRα in bodily fluids have been developed, making large-scale, low-cost detection of ERRα possible. Additionally, targeted inhibitors of ERRs have shown significant effects in suppressing cancer cell proliferation and reversing tumor resistance. This article reviews the research progress of ERRs in tumor resistance, providing important references for developing more effective anti-tumor treatment strategies.
Collapse
Affiliation(s)
- Yuan Ren
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Xiaodan Mao
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Wenyu Lin
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China
| | - Yi Chen
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| | - Rongfeng Chen
- National Center for Occupational Safety and Health, Beijing, 102308, China; NHC Key Laboratory for Engineering Control of Dust Hazard, Beijing 102308, China
| | - Pengming Sun
- Fujian Clinical Research Center for Gynecological Oncology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou 350001, Fujian, China; Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, China; School of Group Medicine and Public Health, Peking Union Medical College, Beijing 100091, China.
| |
Collapse
|
5
|
Heidarian Y, Fasteen TD, Mungcal L, Buddika K, Mahmoudzadeh NH, Nemkov T, D'Alessandro A, Tennessen JM. Hypoxia-inducible factor 1α is required to establish the larval glycolytic program in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631819. [PMID: 39829828 PMCID: PMC11741260 DOI: 10.1101/2025.01.07.631819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The rapid growth that occurs during Drosophila larval development requires a dramatic rewiring of central carbon metabolism to support biosynthesis. Larvae achieve this metabolic state, in part, by coordinately up-regulating the expression of genes involved in carbohydrate metabolism. The resulting metabolic program exhibits hallmark characteristics of aerobic glycolysis and establishes a physiological state that supports growth. To date, the only factor known to activate the larval glycolytic program is the Drosophila Estrogen-Related Receptor (dERR). However, dERR is dynamically regulated during the onset of this metabolic switch, indicating that other factors must be involved. Here we discover that Sima, the Drosophila ortholog of Hif1α, is also essential for establishing the larval glycolytic program. Using a multi-omics approach, we demonstrate that sima mutants fail to properly activate aerobic glycolysis and die during larval development with metabolic defects that phenocopy dERR mutants. Moreover, we demonstrate that dERR and Sima/Hif1α protein accumulation is mutually dependent, as loss of either transcription factor results in decreased abundance of the other protein. Considering that the mammalian homologs of ERR and Hif1α also cooperatively regulate aerobic glycolysis in cancer cells, our findings establish the fly as a powerful genetic model for studying the interaction between these two key metabolic regulators.
Collapse
Affiliation(s)
- Yasaman Heidarian
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Tess D Fasteen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Liam Mungcal
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | - Kasun Buddika
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
| | | | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Jason M Tennessen
- Department of Biology, Indiana University, Bloomington, IN 47405, USA
- Affiliate Member, Melvin and Bren Simon Cancer Center, Indianapolis, IN, 46202, USA
| |
Collapse
|
6
|
Du Y, Pang M, Chen H, Zhou X, Geng R, Zhang Y, Yang L, Li J, Han Y, Liu J, Zhang R, Bi H, Guo D. Inhibitory effect of Zhujing Pill on myopia progression: Mechanistic insights based on metabonomics and network pharmacology. PLoS One 2024; 19:e0312379. [PMID: 39625993 PMCID: PMC11614212 DOI: 10.1371/journal.pone.0312379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 10/02/2024] [Indexed: 12/06/2024] Open
Abstract
OBJECTIVES This study endeavored to uncover the mechanisms by which Zhujing pill (ZJP) slows myopia progression. METHODS We employed biometric analyses to track diopter and axial length changes in guinea pigs with negative lens-induced myopia (LIM). Through integrating metabonomics and network pharmacology, we aimed to predict the anti-myopic targets and active ingredients of ZJP. Subsequent analysis, including real-time fluorescent quantitative PCR (qPCR) and Western blotting (WB), assessed the expression levels of CHRNA7, LPCAT1, and NOS2 in retinal tissues. KEY FINDINGS Our findings demonstrate that ZJP significantly mitigates diopter increase and axial elongation in LIM guinea pigs. Metabonomic analysis revealed significant changes in 13 serum metabolites, with ZJP reversing the expression of 5 key metabolites. By integrating metabonomics with network pharmacology, we identified core targets of ZJP against myopia and constructed a compound-gene-disease-metabolite network. The expressions of LPCAT1 and CHRNA7 were found to decrease in the LIM group but increase with ZJP treatment, whereas NOS2 expression showed the opposite pattern. CONCLUSIONS This investigation provides the first evidence of ZJP's multifaceted effectiveness in managing myopia, highlighting its impact on multiple components, targets, and pathways, including the novel involvement of LPCAT1 and CHRNA7 in myopia pathogenesis.
Collapse
Affiliation(s)
- Yongle Du
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mengran Pang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haoyu Chen
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangkun Zhou
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruyue Geng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanan Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Linqi Yang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawen Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yufeng Han
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinpeng Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruixue Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, China
- Shandong Academy of Eye Disease Prevention and Therapy, Jinan, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
7
|
Chow ST, Fan J, Zhang X, Wang Y, Li Y, Ng CF, Pei X, Zheng Q, Wang F, Wu D, Chan FL. Nuclear receptor TLX functions to promote cancer stemness and EMT in prostate cancer via its direct transactivation of CD44 and stem cell-regulatory transcription factors. Br J Cancer 2024; 131:1450-1462. [PMID: 39322688 PMCID: PMC11519473 DOI: 10.1038/s41416-024-02843-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: 08/25/2023] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/27/2024] Open
Abstract
BACKGROUND Prostate cancer stem cells (PCSCs) play crucial roles in therapy-resistance and metastasis in castration-resistant prostate cancer (CRPC). Certain functional link between cancer stemness and epithelial-mesenchymal transition (EMT) is involved in CRPC. However, up-stream regulators controlling these two processes in PCSCs are still poorly understood. Recently, we have shown that orphan nuclear receptor TLX can promote tumour initiation and progression in CRPC by repressing androgen receptor and oncogene-induced senescence. METHODS PCSCs were isolated from various prostate cancer cell lines and clinical tumour tissues using multiple methods for various in vitro and in vivo oncogenic growth analyses. Direct targets of TLX involved in stemness and EMT regulation were determined by specific reporter gene assays and ligand-driven modulation of TLX activity. RESULTS PCSCs isolated from various sources exhibited increased expression of TLX. Functional and molecular characterisation showed that TLX could function to promote cancer stemness and EMT in prostate cancer cells via its direct transactivation of CD44, SOX2, POU5F1 and NANOG, which share certain functional crosstalk in these two cellular processes. CONCLUSIONS TLX could act as a key up-stream regulator in transcriptional control of stemness and EMT in PCSCs, which contribute to their tumorigenicity, castration-resistance and metastasis potentials in advanced prostate cancer.
Collapse
Affiliation(s)
- Sin Ting Chow
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China
| | - Jiaqi Fan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China
| | - Xingxing Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China
| | - Youjia Li
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China
| | - Chi-Fai Ng
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China
| | - Xiaojuan Pei
- Department of Pathology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guandong, China
| | - Qingyou Zheng
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong Province, China
| | - Fei Wang
- Department of Urology, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan Province, China
| | - Dinglan Wu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China.
- Department of Urology and The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China.
| | - Franky Leung Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong, China.
| |
Collapse
|
8
|
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
|
9
|
Rosell-Garcia T, Rivas-Muñoz S, Kin K, Romero-Albillo V, Alcaraz S, Fernandez-Tornero C, Rodriguez-Pascual F. Multimerization of HIF enhances transcription of target genes containing the hypoxia ancillary sequence. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2023; 1866:194963. [PMID: 37499936 DOI: 10.1016/j.bbagrm.2023.194963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
Transcriptional activity of the hypoxia inducible factor (HIF) relies on the formation of a heterodimer composed of an oxygen-regulated α-subunit and a stably expressed β-subunit. Heterodimeric HIF activates expression by binding to RCGTG motifs within promoters of hypoxia-activated genes. Some hypoxia targets also possess an adjacent HIF ancillary sequence (HAS) reported to increase transcription but whose function remains obscure. Here, we investigate the contribution of the HAS element to the hypoxia response and its mechanism of action, using the HAS-containing prolyl 4-hydroxylase subunit α1 (P4HA1) as a gene model in NIH/3T3 mouse embryonic fibroblasts and HEK293 human embryonic kidney cells. Our HIF overexpression experiments demonstrate that the HAS motif is essential for full induction by hypoxia and that the presence of the tandem HAS/HIF, as opposed to HIF-only sequences, provides HIF proteins with the capacity to form complexes of stoichiometry beyond the classical heterodimer, likely tetramers, to cooperatively potentiate hypoxia-induced transcription. We also provide evidence of the crucial role played by the Fα helix of the PAS-B domain of the HIF1β subunit to support the interaction between heterodimers. Functional analysis showed that human genes containing the HAS/HIF motifs are better responders to hypoxia, and their promoters are enriched for specific transcription factor binding sites. Gene ontology enrichment revealed a predominance of HAS/HIF in genes primarily related to tissue formation and development. Our findings add an extra level of regulation of the hypoxia/HIF signaling through multimerization of HIF proteins on regulatory elements containing the HAS/HIF motifs.
Collapse
Affiliation(s)
- Tamara Rosell-Garcia
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | - Sergio Rivas-Muñoz
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | - Koryu Kin
- Institut de Biologia Evolutiva (CSIC-Universitat Pompeu Fabra), Barcelona, Spain
| | - Verónica Romero-Albillo
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | - Silvia Alcaraz
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain
| | | | - Fernando Rodriguez-Pascual
- Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas (C.S.I.C.)-Universidad Autónoma de Madrid (U.A.M.), Madrid, Spain.
| |
Collapse
|
10
|
Narkar VA. Exercise and Ischemia-Activated Pathways in Limb Muscle Angiogenesis and Vascular Regeneration. Methodist Debakey Cardiovasc J 2023; 19:58-68. [PMID: 38028974 PMCID: PMC10655757 DOI: 10.14797/mdcvj.1304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Exercise has a profound effect on cardiovascular disease, particularly through vascular remodeling and regeneration. Peripheral artery disease (PAD) is one such cardiovascular condition that benefits from regular exercise or rehabilitative physical therapy in terms of slowing the progression of disease and delaying amputations. Various rodent pre-clinical studies using models of PAD and exercise have shed light on molecular pathways of vascular regeneration. Here, I review key exercise-activated signaling pathways (nuclear receptors, kinases, and hypoxia inducible factors) in the skeletal muscle that drive paracrine regenerative angiogenesis. The rationale for highlighting the skeletal muscle is that it is the largest organ recruited during exercise. During exercise, skeletal muscle releases several myokines, including angiogenic factors and cytokines that drive tissue vascular regeneration via activation of endothelial cells, as well as by recruiting immune and endothelial progenitor cells. Some of these core exercise-activated pathways can be extrapolated to vascular regeneration in other organs. I also highlight future areas of exercise research (including metabolomics, single cell transcriptomics, and extracellular vesicle biology) to advance our understanding of how exercise induces vascular regeneration at the molecular level, and propose the idea of "exercise-mimicking" therapeutics for vascular recovery.
Collapse
Affiliation(s)
- Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, US
| |
Collapse
|
11
|
Su P, Mao X, Ma J, Huang L, Yu L, Tang S, Zhuang M, Lu Z, Osafo KS, Ren Y, Wang X, Lin X, Huang L, Huang X, Braicu EI, Sehouli J, Sun P. ERRα promotes glycolytic metabolism and targets the NLRP3/caspase-1/GSDMD pathway to regulate pyroptosis in endometrial cancer. J Exp Clin Cancer Res 2023; 42:274. [PMID: 37864196 PMCID: PMC10588109 DOI: 10.1186/s13046-023-02834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023] Open
Abstract
BACKGROUND Tumor cells can resist chemotherapy-induced pyroptosis through glycolytic reprogramming. Estrogen-related receptor alpha (ERRα) is a central regulator of cellular energy metabolism associated with poor cancer prognosis. Herein, we refine the oncogenic role of ERRα in the pyroptosis pathway and glycolytic metabolism. METHODS The interaction between ERRα and HIF-1α was verified using co-immunoprecipitation. The transcriptional binding sites of ERRα and NLRP3 were confirmed using dual-luciferase reporter assay and cleavage under targets and tagmentation (CUT&Tag). Flow cytometry, transmission electron microscopy, scanning electron microscopy, cell mito stress test, and extracellular acidification rate analysis were performed to investigate the effects of ERRα on the pyroptosis pathway and glycolytic metabolism. The results of these experiments were further confirmed in endometrial cancer (EC)-derived organoids and nude mice. In addition, the expression of ERRα-related pyroptosis genes was analyzed using The Cancer Genome Atlas and Gene Expression Omnibus database. RESULTS Triggered by a hypoxic microenvironment, highly expressed ERRα could bind to the promoter of NLRP3 and inhibit caspase-1/GSDMD signaling, which reduced inflammasome activation and increased pyroptosis resistance, thereby resulting in the resistance of cancer cells to cisplatin. Moreover, ERRα activated glycolytic rate-limiting enzyme to bridge glycolytic metabolism and pyroptosis in EC. This phenomenon was further confirmed in EC-derived organoids and nude mice. CUT & Tag sequencing and The Cancer Genome Atlas database analysis showed that ERRα participated in glycolysis and programmed cell death, which resulted in EC progression. CONCLUSIONS ERRα inhibits pyroptosis in an NLRP3-dependent manner and induces glycolytic metabolism, resulting in cisplatin resistance in EC cells.
Collapse
Affiliation(s)
- Pingping Su
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaodan Mao
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Jincheng Ma
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Lixiang Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Lirui Yu
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Shuting Tang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Mingzhi Zhuang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, Fujian, China
| | - Zhonglei Lu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, Fujian, China
| | - Kelvin Stefan Osafo
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Yuan Ren
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xinrui Wang
- Medical Research Center, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- NHC Key Laboratory of Technical Evaluation of Fertility Regulation for Non-Human Primate, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
| | - Xite Lin
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Leyi Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaoli Huang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Fujian Medical University, FuzhouFujian, 350005, China
| | - Elena Ioana Braicu
- Department of Gynecology and Obstetrics, Charité Virchow University Hospital, Augustenberger Platz1, 13353, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology and Obstetrics, Charité Virchow University Hospital, Augustenberger Platz1, 13353, Berlin, Germany
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Maternity and Child Health Hospital, Fujian Medical University, Fuzhou, 350001, Fujian, China.
- Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China.
- Fujian Clinical Research Center for Gynecologic Oncology, Fujian Maternity and Child Health Hospital, Fujian Obstetrics and Gynecology Hospital, Fuzhou, 350001, Fujian, China.
- National Key Clinical Specialty Construction Program of China (Gynecology), Fujian Maternity and Child Health Hospital, Fuzhou, 350001, Fujian, China.
| |
Collapse
|
12
|
Sopariwala DH, Hao NTT, Narkar VA. Estrogen-related Receptor Signaling in Skeletal Muscle Fitness. Int J Sports Med 2023; 44:609-617. [PMID: 36787804 PMCID: PMC11168301 DOI: 10.1055/a-2035-8192] [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] [Indexed: 02/16/2023]
Abstract
Skeletal muscle is a highly plastic tissue that can alter its metabolic and contractile features, as well as regenerative potential in response to exercise and other conditions. Multiple signaling factors including metabolites, kinases, receptors, and transcriptional factors have been studied in the regulation of skeletal muscle plasticity. Recently, estrogen-related receptors (ERRs) have emerged as a critical transcriptional hub in control of skeletal muscle homeostasis. ERRα and ERRγ - the two highly expressed ERR sub-types in the muscle respond to various extracellular cues such as exercise, hypoxia, fasting and dietary factors, in turn regulating gene expression in the skeletal muscle. On the other hand, conditions such as diabetes and muscular dystrophy suppress expression of ERRs in the skeletal muscle, likely contributing to disease progression. We highlight key functions of ERRs in the skeletal muscle including the regulation of fiber type, mitochondrial metabolism, vascularization, and regeneration. We also describe how ERRs are regulated in the skeletal muscle, and their interaction with important muscle regulators (e. g. AMPK and PGCs). Finally, we identify critical gaps in our understanding of ERR signaling in the skeletal muscle, and suggest future areas of investigation to advance ERRs as potential targets for function promoting therapeutics in muscle diseases.
Collapse
Affiliation(s)
- Danesh H. Sopariwala
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| | - Nguyen Thi Thu Hao
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| |
Collapse
|
13
|
Chaltel-Lima L, Domínguez F, Domínguez-Ramírez L, Cortes-Hernandez P. The Role of the Estrogen-Related Receptor Alpha (ERRa) in Hypoxia and Its Implications for Cancer Metabolism. Int J Mol Sci 2023; 24:ijms24097983. [PMID: 37175690 PMCID: PMC10178695 DOI: 10.3390/ijms24097983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Under low oxygen conditions (hypoxia), cells activate survival mechanisms including metabolic changes and angiogenesis, which are regulated by HIF-1. The estrogen-related receptor alpha (ERRα) is a transcription factor with important roles in the regulation of cellular metabolism that is overexpressed in hypoxia, suggesting that it plays a role in cell survival in this condition. This review enumerates and analyses the recent evidence that points to the role of ERRα as a regulator of hypoxic genes, both in cooperation with HIF-1 and through HIF-1- independent mechanisms, in invertebrate and vertebrate models and in physiological and pathological scenarios. ERRα's functions during hypoxia include two mechanisms: (1) direct ERRα/HIF-1 interaction, which enhances HIF-1's transcriptional activity; and (2) transcriptional activation by ERRα of genes that are classical HIF-1 targets, such as VEGF or glycolytic enzymes. ERRα is thus gaining recognition for its prominent role in the hypoxia response, both in the presence and absence of HIF-1. In some models, ERRα prepares cells for hypoxia, with important clinical/therapeutic implications.
Collapse
Affiliation(s)
- Leslie Chaltel-Lima
- Segal Cancer Center, Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada
- Division of Experimental Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Fabiola Domínguez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco 74360, Mexico
| | - Lenin Domínguez-Ramírez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco 74360, Mexico
| | - Paulina Cortes-Hernandez
- Centro de Investigación Biomédica de Oriente (CIBIOR), Instituto Mexicano del Seguro Social (IMSS), Atlixco 74360, Mexico
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Estrogens revert neutrophil hyperplasia by inhibiting Hif1α-cMyb pathway in zebrafish myelodysplastic syndromes models. Cell Death Dis 2022; 8:323. [PMID: 35842445 PMCID: PMC9288432 DOI: 10.1038/s41420-022-01121-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/29/2022] [Accepted: 07/05/2022] [Indexed: 11/09/2022]
Abstract
Myelodysplastic syndromes (MDS) are characterized by daunting genetic heterogeneity and a high risk of leukemic transformation, which presents great challenges for clinical treatment. To identify new chemicals for MDS, we screened a panel of FDA-approved drugs and verified the neutrophil hyperplasia inhibiting role of 17β-estradiol (E2, a natural estrogen) in several zebrafish MDS models (pu.1G242D/G242D, irf8Δ57Δ/57 and c-mybhyper). However, the protective mechanism of estrogen in the development of hematological malignancies remains to be explored. Here, analyzing the role of E2 in the development of each hematopoietic lineage, we found that E2 exhibited a specific neutrophil inhibiting function. This neutrophil inhibitory function of E2 is attributed to its down-regulation of c-myb, which leads to accelerated apoptosis and decreased proliferation of neutrophils. We further showed that knockdown of hif1α could mimic the neutrophil inhibiting role of E2, and hif1α overexpression could reverse the protective function of E2. Collectively, our findings highlight the protective role of E2 on MDS by inhibiting hif1α-c-myb pathway, suggesting that E2 is a promising and effective drug for hematopoietic tumors associated with abnormal neutrophil hyperplasia.
Collapse
|
17
|
Su P, Yu L, Mao X, Sun P. Role of HIF-1α/ERRα in Enhancing Cancer Cell Metabolism and Promoting Resistance of Endometrial Cancer Cells to Pyroptosis. Front Oncol 2022; 12:881252. [PMID: 35800058 PMCID: PMC9253301 DOI: 10.3389/fonc.2022.881252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/25/2022] [Indexed: 12/24/2022] Open
Abstract
Oxygen is critical to energy metabolism, and tumors are often characterized by a hypoxic microenvironment. Owing to the high metabolic energy demand of malignant tumor cells, their survival is promoted by metabolic reprogramming in the hypoxic microenvironment, which can confer tumor cell resistance to pyroptosis. Pyroptosis resistance can inhibit anti-tumor immunity and promote the development of malignant tumors. Hypoxia inducible factor-1α (HIF-1α) is a key regulator of metabolic reprogramming in tumor cells, and estrogen-related receptor α (ERRα) plays a key role in regulating cellular energy metabolism. Therefore, the close interaction between HIF-1α and ERRα influences the metabolic and functional changes in cancer cells. In this review, we summarize the reprogramming of tumor metabolism involving HIF-1α/ERRα. We review our understanding of the role of HIF-1α/ERRα in promoting tumor growth adaptation and pyroptosis resistance, emphasize its key role in energy homeostasis, and explore the regulation of HIF-1α/ERRα in preventing and/or treating endometrial carcinoma patients. This review provides a new perspective for the study of the molecular mechanisms of metabolic changes in tumor progression.
Collapse
Affiliation(s)
- Pingping Su
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lirui Yu
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaodan Mao
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecological Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Women and Children’s Critical Diseases Research, Fuzhou, China
- *Correspondence: Pengming Sun,
| |
Collapse
|
18
|
Sakellakis M. Orphan receptors in prostate cancer. Prostate 2022; 82:1016-1024. [PMID: 35538397 DOI: 10.1002/pros.24370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/22/2022] [Accepted: 04/22/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND The identification of new cellular receptors has been increasing rapidly. A receptor is called "orphan" if an endogenous ligand has not been identified yet. METHODS Here we review receptors that contribute to prostate cancer and are considered orphan or partially orphan. This means that the full spectrum of their endogenous ligands remains unknown. RESULTS The orphan receptors are divided into two major families. The first group includes G protein-coupled receptors. Most are orphan olfactory receptors. OR51E1 inhibits cell proliferation and induces senescence in prostate cancer. OR51E2 inhibits prostate cancer growth, but promotes invasiveness and metastasis. GPR158, GPR110, and GPCR-X play significant roles in prostate cancer development and progression. However, GPR160 induces cell cycle arrest and apoptosis. The other major subset of orphan receptors are nuclear receptors. Receptor-related orphan receptor α (RORα) inhibits tumor growth, but RORγ stimulates androgen receptor signaling. PXR contributes to metabolic deactivation of androgens and inhibits cell proliferation. TLX has protumorigenic effects in prostate cancer, while its knockdown triggers cellular senescence and growth arrest. Estrogen-related receptor ERRγ can inhibit tumor growth but ERRα is protumorigenic. Dax1 and short heterodimeric partner are also inhibitory in prostate cancer. CONCLUSION There is a "zoo" of relatively underappreciated orphan receptors that play key roles in prostate cancer.
Collapse
Affiliation(s)
- Minas Sakellakis
- Fourth Oncology Department and Comprehensive Clinical Trials Center, Metropolitan Hospital, Athens, Greece
| |
Collapse
|
19
|
Xie K, Tan K, Naylor MJ. Transcription Factors as Novel Therapeutic Targets and Drivers of Prostate Cancer Progression. Front Oncol 2022; 12:854151. [PMID: 35547880 PMCID: PMC9082354 DOI: 10.3389/fonc.2022.854151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/23/2022] [Indexed: 11/24/2022] Open
Abstract
Prostate cancer is the second most diagnosed cancer among men worldwide. Androgen deprivation therapy, the most common targeted therapeutic option, is circumvented as prostate cancer progresses from androgen dependent to castrate-resistant disease. Whilst the nuclear receptor transcription factor, androgen receptor, drives the growth of prostate tumor during initial stage of the disease, androgen resistance is associated with poorly differentiated prostate cancer. In the recent years, increased research has highlighted the aberrant transcriptional activities of a small number of transcription factors. Along with androgen receptors, dysregulation of these transcription factors contributes to both the poorly differentiated phenotypes of prostate cancer cells and the initiation and progression of prostate carcinoma. As master regulators of cell fate decisions, these transcription factors may provide opportunity for the development of novel therapeutic targets for the management of prostate cancer. Whilst some transcriptional regulators have previously been notoriously difficult to directly target, technological advances offer potential for the indirect therapeutic targeting of these transcription factors and the capacity to reprogram cancer cell phenotype. This mini review will discuss how recent advances in our understanding of transcriptional regulators and material science pave the way to utilize these regulatory molecules as therapeutic targets in prostate cancer.
Collapse
Affiliation(s)
- Kangzhe Xie
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney, NSW, Australia
| | - Keely Tan
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney, NSW, Australia
| | - Matthew J Naylor
- Charles Perkins Centre, School of Medical Sciences, Faculty of Medicine & Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
20
|
Gao W, Wang Y, Yu S, Wang Z, Ma T, Chan AML, Chiu PKF, Ng CF, Wu D, Chan FL. Endothelial nitric oxide synthase (eNOS)-NO signaling axis functions to promote the growth of prostate cancer stem-like cells. Stem Cell Res Ther 2022; 13:188. [PMID: 35526071 PMCID: PMC9080127 DOI: 10.1186/s13287-022-02864-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/24/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Accumulating evidence supports that prostate cancer stem-like cells (PCSCs) play significant roles in therapy resistance and metastasis of prostate cancer. Many studies also show that nitric oxide (NO) synthesized by NO synthases can function to promote tumor progression. However, the exact roles of NOSs and NO signaling in the growth regulation of PCSCs and castration-resistant prostate cancer (CRPC) are still not fully understood. METHODS The regulatory functions of NOS-NO signaling were evaluated in prostate cancer cells, especially in PCSCs enriched by 3D spheroid culture and CD133/CD44 cell sorting. The molecular mechanisms of NOS-NO signaling in PCSCs growth regulation and tumor metastasis were investigated in PCSCs and mice orthotopic prostate tumor model. RESULTS Endothelial NOS (eNOS) exhibited a significant upregulation in high-grade prostate cancer and metastatic CRPC. Xenograft models of CRPC exhibited notable increased eNOS expression and higher intracellular NO levels. PCSCs isolated from various models displayed significant enhanced eNOS-NO signaling. Functional analyses demonstrated that increased eNOS expression could promote in vivo tumorigenicity and metastatic potential of prostate cancer cells. Characterization of eNOS-NO involved downstream pathway which confirmed that enhanced eNOS signaling could promote the growth of PCSCs and antiandrogen-resistant prostate cancer cells via an activated downstream NO-sGC-cGMP-PKG effector signaling pathway. Interestingly, eNOS expression could be co-targeted by nuclear receptor ERRα and transcription factor ERG in prostate cancer cells and PCSCs. CONCLUSIONS Enhanced eNOS-NO signaling could function to promote the growth of PCSCs and also the development of metastatic CRPC. Besides eNOS-NO as potential targets, targeting its upstream regulators (ERRα and ERG) of eNOS-NO signaling could also be the therapeutic strategy for the management of advanced prostate cancer, particularly the aggressive cancer carrying with the TMPRSS2:ERG fusion gene.
Collapse
Affiliation(s)
- Weijie Gao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Shan Yu
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Zhu Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Taiyang Ma
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Andrew Man-Lok Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Peter Ka-Fung Chiu
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Chi-Fai Ng
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dinglan Wu
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Franky Leung Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
21
|
Ranhotra HS. Estrogen-related receptor alpha in select host functions and cancer: new frontiers. Mol Cell Biochem 2022; 477:1349-1359. [PMID: 35138514 DOI: 10.1007/s11010-022-04380-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/27/2022] [Indexed: 01/03/2023]
Abstract
Eukaryotic gene expression is under the tight control of transcription factors, which includes the estrogen-related receptor alpha (ERRα). The endogenous ligand(s) acting as ERRα agonist has not been identified and confirmed. ERRα is a prominent member of the nuclear receptors super-family with major roles in energy metabolism, including immunity, cell growth, proliferation and differentiation and a host of other functions in animals. The actions exerted by ERRα towards gene expression regulation are often in association with other transcriptional factors, receptors and signal mediators. Metabolic regulation by ERRα is known for some time that has tremendous impact on host biology like autophagy, angiogenesis, mitochondrial activity, including lipid metabolism. Cellular metabolism and cancer has intricate relationship. On account of the participation of ERRα in metabolism, it has been implicated in various types of cancer onset and progression. In a number of findings, ERRα has been demonstrated to influence several types of cancers, exhibiting as a negative prognostic marker for many. Such diverse role associated with ERRα is due to its interaction with numerous transcriptional factors and other signalling pathways that culminate in providing optimal gene regulation. These observations points to the crucial regulatory roles of ERRα in health and disease. In this article, some of the new findings on the influence of ERRα in host metabolism and biology including cancer, shall be reviewed that will provide a concise understanding of this receptor.
Collapse
Affiliation(s)
- Harmit S Ranhotra
- Department of Biochemistry, St. Edmund's College, Shillong, 793 003, India.
| |
Collapse
|
22
|
Lappano R, Todd LA, Stanic M, Cai Q, Maggiolini M, Marincola F, Pietrobon V. Multifaceted Interplay between Hormones, Growth Factors and Hypoxia in the Tumor Microenvironment. Cancers (Basel) 2022; 14:539. [PMID: 35158804 PMCID: PMC8833523 DOI: 10.3390/cancers14030539] [Citation(s) in RCA: 14] [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: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 02/07/2023] Open
Abstract
Hormones and growth factors (GFs) are signaling molecules implicated in the regulation of a variety of cellular processes. They play important roles in both healthy and tumor cells, where they function by binding to specific receptors on target cells and activating downstream signaling cascades. The stages of tumor progression are influenced by hormones and GF signaling. Hypoxia, a hallmark of cancer progression, contributes to tumor plasticity and heterogeneity. Most solid tumors contain a hypoxic core due to rapid cellular proliferation that outgrows the blood supply. In these circumstances, hypoxia-inducible factors (HIFs) play a central role in the adaptation of tumor cells to their new environment, dramatically reshaping their transcriptional profile. HIF signaling is modulated by a variety of factors including hormones and GFs, which activate signaling pathways that enhance tumor growth and metastatic potential and impair responses to therapy. In this review, we summarize the role of hormones and GFs during cancer onset and progression with a particular focus on hypoxia and the interplay with HIF proteins. We also discuss how hypoxia influences the efficacy of cancer immunotherapy, considering that a hypoxic environment may act as a determinant of the immune-excluded phenotype and a major hindrance to the success of adoptive cell therapies.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Lauren A. Todd
- Department of Biology, University of Waterloo, Waterloo, ON N2L 3G1, Canada;
| | - Mia Stanic
- Department of Laboratory Medicine & Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Qi Cai
- Kite Pharma Inc., Santa Monica, CA 90404, USA; (Q.C.); (F.M.)
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | | | | |
Collapse
|
23
|
Alderdice R, Pernice M, Cárdenas A, Hughes DJ, Harrison PL, Boulotte N, Chartrand K, Kühl M, Suggett DJ, Voolstra CR. Hypoxia as a physiological cue and pathological stress for coral larvae. Mol Ecol 2021; 31:571-587. [PMID: 34716959 DOI: 10.1111/mec.16259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/15/2021] [Accepted: 10/20/2021] [Indexed: 11/30/2022]
Abstract
Ocean deoxygenation events are intensifying worldwide and can rapidly drive adult corals into a state of metabolic crisis and bleaching-induced mortality, but whether coral larvae are subject to similar stress remains untested. We experimentally exposed apo-symbiotic coral larvae of Acropora selago to deoxygenation stress with subsequent reoxygenation aligned to their night-day light cycle, and followed their gene expression using RNA-Seq. After 12 h of deoxygenation stress (~2 mg O2 /L), coral planulae demonstrated a low expression of HIF-targeted hypoxia response genes concomitant with a significantly high expression of PHD2 (a promoter of HIFα proteasomal degradation), similar to corresponding adult corals. Despite exhibiting a consistent swimming phenotype compared to control samples, the differential gene expression observed in planulae exposed to deoxygenation-reoxygenation suggests a disruption of pathways involved in developmental regulation, mitochondrial activity, lipid metabolism, and O2 -sensitive epigenetic regulators. Importantly, we found that treated larvae exhibited a disruption in the expression of conserved HIF-targeted developmental regulators, for example, Homeobox (HOX) genes, corroborating how changes in external oxygen levels can affect animal development. We discuss how the observed deoxygenation responses may be indicative of a possible acclimation response or alternatively may imply negative latent impacts for coral larval fitness.
Collapse
Affiliation(s)
- Rachel Alderdice
- Faculty of Science, Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia
| | - Mathieu Pernice
- Faculty of Science, Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia
| | - Anny Cárdenas
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - David J Hughes
- Faculty of Science, Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia
| | - Peter L Harrison
- Marine Ecology Research Centre, Southern Cross University, Lismore, NSW, Australia
| | - Nadine Boulotte
- Marine Ecology Research Centre, Southern Cross University, Lismore, NSW, Australia
| | - Katie Chartrand
- Centre of Tropical Water and Aquatic Ecosystem Research, James Cook University, Townsville, Qld, Australia
| | - Michael Kühl
- Faculty of Science, Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia.,Marine Biology Section, Department of Biology, University of Copenhagen, Helsingør, Denmark
| | - David J Suggett
- Faculty of Science, Climate Change Cluster, University of Technology Sydney, Ultimo, NSW, Australia
| | | |
Collapse
|
24
|
Scholtes C, Giguère V. Transcriptional Regulation of ROS Homeostasis by the ERR Subfamily of Nuclear Receptors. Antioxidants (Basel) 2021; 10:antiox10030437. [PMID: 33809291 PMCID: PMC7999130 DOI: 10.3390/antiox10030437] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 01/08/2023] Open
Abstract
Reactive oxygen species (ROS) such as superoxide anion (O2•-) and hydrogen peroxide (H2O2) are generated endogenously by processes such as mitochondrial oxidative phosphorylation, or they may arise from exogenous sources like bacterial invasion. ROS can be beneficial (oxidative eustress) as signaling molecules but also harmful (oxidative distress) to cells when ROS levels become unregulated in response to physiological, pathological or pharmacological insults. Indeed, abnormal ROS levels have been shown to contribute to the etiology of a wide variety of diseases. Transcriptional control of metabolic genes is a crucial mechanism to coordinate ROS homeostasis. Therefore, a better understanding of how ROS metabolism is regulated by specific transcription factors can contribute to uncovering new therapeutic strategies. A large body of work has positioned the estrogen-related receptors (ERRs), transcription factors belonging to the nuclear receptor superfamily, as not only master regulators of cellular energy metabolism but, most recently, of ROS metabolism. Herein, we will review the role played by the ERRs as transcriptional regulators of ROS generation and antioxidant mechanisms and also as ROS sensors. We will assess how the control of ROS homeostasis by the ERRs can be linked to physiology and disease and the possible contribution of manipulating ERR activity in redox medicine.
Collapse
Affiliation(s)
- Charlotte Scholtes
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada;
| | - Vincent Giguère
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montréal, QC H3A 1A3, Canada;
- Department of Biochemistry, McGill University, Montréal, QC H3G 1Y6, Canada
- Correspondence:
| |
Collapse
|
25
|
Zhou J, Wang Y, Wu D, Wang S, Chen Z, Xiang S, Chan FL. Orphan nuclear receptors as regulators of intratumoral androgen biosynthesis in castration-resistant prostate cancer. Oncogene 2021; 40:2625-2634. [PMID: 33750894 PMCID: PMC8049868 DOI: 10.1038/s41388-021-01737-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/18/2021] [Accepted: 02/24/2021] [Indexed: 01/31/2023]
Abstract
Castration-resistant prostate cancer (CRPC) almost invariably occurs after androgen-deprivation therapy (ADT) for the advanced metastatic disease. It is generally believed that among multiple mechanisms and signaling pathways, CRPC is significantly driven by the reactivation of androgen receptor (AR) signaling in ADT-treated patients with castrate levels of androgen, partially at least mediated by the androgen biosynthesis within the tumor, also known as intratumoral or intraprostatic androgen biosynthesis. Steroidogenic enzymes, such as CYP11A1, CYP17A1, HSD3B1, AKR1C3 and SRD5A, are essential to catalyze the conversion of the initial substrate cholesterol into potent androgens that confers the CRPC progression. Accumulating evidences indicate that many steroidogenic enzymes are upregulated in the progression setting; however, little is known about the dysregulation of these enzymes in CRPC. Orphan nuclear receptors (ONRs) are members of the nuclear receptor superfamily, of which endogenous physiological ligands are unknown and which are constitutively active independent of any physiological ligands. Studies have validated that besides AR, ONRs could be the potential therapeutic targets for prostate cancer, particularly the lethal CRPC progression. Early studies reveal that ONRs play crucial roles in the transcriptional regulation of steroidogenic enzyme genes. Notably, we and others show that three distinct ONRs, including liver receptor homolog-1 (LRH-1, NR5A2), steroidogenic factor 1 (SF-1, AD4BP, NR5A1) and estrogen-related receptor α (ERRα, NR3B1), can contribute to the CRPC progression by promotion of the intratumoral androgen synthesis via their direct transcriptional regulation on multiple steroidogenic enzymes. This review presents an overview of the current understanding on the intratumoral androgen biosynthesis in CRPC, with a special focus on the emerging roles of ONRs in this process.
Collapse
Affiliation(s)
- Jianfu Zhou
- grid.411866.c0000 0000 8848 7685Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China ,grid.411866.c0000 0000 8848 7685The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China ,grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuliang Wang
- grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dinglan Wu
- grid.488521.2Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Shusheng Wang
- grid.411866.c0000 0000 8848 7685Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhiqiang Chen
- grid.411866.c0000 0000 8848 7685Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Songtao Xiang
- grid.411866.c0000 0000 8848 7685Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Franky Leung Chan
- grid.10784.3a0000 0004 1937 0482School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Xia L, Sun J, Xie S, Chi C, Zhu Y, Pan J, Dong B, Huang Y, Xia W, Sha J, Xue W. PRKAR2B-HIF-1α loop promotes aerobic glycolysis and tumour growth in prostate cancer. Cell Prolif 2020; 53:e12918. [PMID: 33025691 PMCID: PMC7653268 DOI: 10.1111/cpr.12918] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES Reprogramming of cellular metabolism is profoundly implicated in tumorigenesis and can be exploited to cancer treatment. Cancer cells are known for their propensity to use glucose-dependent glycolytic pathway instead of mitochondrial oxidative phosphorylation for energy generation even in the presence of oxygen, a phenomenon known as Warburg effect. The type II beta regulatory subunit of protein kinase A (PKA), PRKAR2B, is highly expressed in castration-resistant prostate cancer (CRPC) and contributes to tumour growth and metastasis. However, whether PRKAR2B regulates glucose metabolism in prostate cancer remains largely unknown. MATERIALS AND METHODS Loss-of-function and gain-of-function studies were used to investigate the regulatory role of PRKAR2B in aerobic glycolysis. Real-time qPCR, Western blotting, luciferase reporter assay and chromatin immunoprecipitation were employed to determine the underlying mechanisms. RESULTS PRKAR2B was sufficient to enhance the Warburg effect as demonstrated by glucose consumption, lactate production and extracellular acidification rate. Mechanistically, loss-of-function and gain-of-function studies showed that PRKAR2B was critically involved in the tumour growth of prostate cancer. PRKAR2B was able to increase the expression level of hypoxia-inducible factor 1α (HIF-1α), which is a key mediator of the Warburg effect. Moreover, we uncovered that HIF-1α is a key transcription factor responsible for inducing PRKAR2B expression in prostate cancer. Importantly, inhibition of glycolysis by the glycolytic inhibitor 2-deoxy-d-glucose (2-DG) or replacement of glucose in the culture medium with galactose (which has a much lower rate than glucose entry into glycolysis) largely compromised PRKAR2B-mediated tumour-promoting effect. Similar phenomenon was noticed by genetic silencing of HIF-1α. CONCLUSIONS Our study identified that PRKAR2B-HIF-1α loop enhances the Warburg effect to enable growth advantage in prostate cancer.
Collapse
Affiliation(s)
- Lei Xia
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jian Sun
- Department of UrologyAffiliated Hospital of Jiangnan UniversityJiangsuChina
| | - Shaowei Xie
- Department of UltrasoundRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Chenfei Chi
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yinjie Zhu
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jiahua Pan
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Baijun Dong
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Yiran Huang
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Weiliang Xia
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Jianjun Sha
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Wei Xue
- Department of UrologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
27
|
Wang Y, Gao W, Li Y, Chow ST, Xie W, Zhang X, Zhou J, Chan FL. Interplay between orphan nuclear receptors and androgen receptor-dependent or-independent growth signalings in prostate cancer. Mol Aspects Med 2020; 78:100921. [PMID: 33121737 DOI: 10.1016/j.mam.2020.100921] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/08/2020] [Accepted: 10/15/2020] [Indexed: 12/14/2022]
Abstract
It is well-established that both the initial and advanced growth of prostate cancer depends critically on androgens and thus on the activated androgen receptor (AR) -mediated signaling pathway. The unique hormone-dependent feature of prostate cancer forms the biological basis of hormone or androgen-deprivation therapy (ADT) that aims to suppress the AR signaling by androgen depletion or AR antagonists. ADT still remains the mainstay treatment option for locally advanced or metastatic prostate cancer. However, most patients upon ADT will inevitably develop therapy-resistance and progress to relapse in the form of castration-resistant disease (castration-resistant prostate cancer or CRPC) or even a more aggressive androgen-independent subtype (therapy-related neuroendocrine prostate cancer or NEPC). Recent advances show that besides AR, some ligand-independent members of nuclear receptor superfamily-designated as orphan nuclear receptors (ONRs), as their endogenous physiological ligands are either absent or not yet identified to date, also play significant roles in the growth regulation of prostate cancer via multiple AR-dependent or -independent (AR-bypass) pathways or mechanisms. In this review, we summarize the recent progress in the newly elucidated roles of ONRs in prostate cancer, with a focus on their interplay in the AR-dependent pathways (intratumoral androgen biosynthesis and suppression of AR signaling) and AR-independent pathways or cellular processes (hypoxia, oncogene- or tumor suppressor-induced senescence, apoptosis and regulation of prostate cancer stem cells). These ONRs with their newly characterized roles not only can serve as novel biomarkers but also as potential therapeutic targets for management of advanced prostate cancer.
Collapse
Affiliation(s)
- Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Weijie Gao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Youjia Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Sin Ting Chow
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenjuan Xie
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xingxing Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jianfu Zhou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China; Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510370, China
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
28
|
Schoepke E, Billon C, Haynes KM, Avdagic A, Sitaula S, Sanders R, Adeyemi CM, Walker JK, Burris TP. A Selective ERRα/γ Inverse Agonist, SLU-PP-1072, Inhibits the Warburg Effect and Induces Apoptosis in Prostate Cancer Cells. ACS Chem Biol 2020; 15:2338-2345. [PMID: 32897058 DOI: 10.1021/acschembio.0c00670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The estrogen related receptors (ERRs) are a subgroup of nuclear receptors that play a role in regulation of cellular metabolism. Prostate cancer (PCa) cells display altered metabolic signatures, such as the Warburg effect, and the ERRs have been implicated in driving this phenotype. Despite the lack of a known endogenous ligand, synthetic ligands that target the ERRs have been discovered. For example, the ERRα inverse agonist XCT790 modulates metabolic pathways in PCa cells, but it also functions as a mitochondrial uncoupler independent of targeting ERRα. Here, we describe a novel dual ERRα/γ inverse agonist, SLU-PP-1072, derived from the GSK4716 ERRγ agonist scaffold that is distinct from the XCT790 scaffold. SLU-PP-1072 alters PCa cell metabolism and gene expression, resulting in cell cycle dysregulation and increased apoptosis without acute mitochondrial uncoupling activity. Our data suggest that inhibition of ERRα/γ may be beneficial in treatment of PCa, and SLU-PP-1072 provides a unique chemical tool to evaluate the pharmacology of ERRα and ERRγ.
Collapse
Affiliation(s)
- Emmalie Schoepke
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Cyrielle Billon
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Keith M Haynes
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Amer Avdagic
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Sadichha Sitaula
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Ryan Sanders
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Christiana M Adeyemi
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - John K Walker
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Thomas P Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| |
Collapse
|
29
|
Huang H, Li J, Shen J, Lin L, Wu X, Xiang S, Li Y, Xu Y, Zhao Q, Zhao Y, Kaboli PJ, Li M, Li X, Wang W, Wen Q, Xiao Z. Increased ABCC4 Expression Induced by ERRα Leads to Docetaxel Resistance via Efflux of Docetaxel in Prostate Cancer. Front Oncol 2020; 10:1474. [PMID: 33014785 PMCID: PMC7493678 DOI: 10.3389/fonc.2020.01474] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 07/10/2020] [Indexed: 12/11/2022] Open
Abstract
Docetaxel is a major treatment for advanced prostate cancer (PCa); however, its resistance compromises clinical effectiveness. Estrogen receptor-related receptor alpha (ERRα) belongs to an orphan nuclear receptor superfamily and was recently found to be closely involved in cancer. In the present study, we found that ERRα was involved in docetaxel resistance in PCa. Overexpression of ERRα conferred docetaxel resistance in PCa cell lines, and cells with ERRα downregulation were more sensitive to docetaxel. Among the drug resistance-related genes, ABCC4 demonstrated synchronous expression after ERRα manipulation in cells. Moreover, both ERRα and ABCC4 were overexpressed in the docetaxel-resistant cell, which could be reversed by ERRα knockdown. The knockdown of ERRα also reversed the reduced drug accumulation in the docetaxel-resistant cell. We also demonstrated for the first time that ABCC4 was a direct target of ERRα as determined by the CHIP and luciferase assays. Bioinformatics analysis revealed high expression of ERRα and ABCC4 in PCa patients, and a number of potential ERRα/ABCC4 targets were predicted. In conclusion, our study demonstrated a critical role for ERRα in docetaxel resistance by directly targeting ABCC4 and stressed the importance of ERRα as a potential therapeutic target for drug-resistant PCa.
Collapse
Affiliation(s)
- Houbao Huang
- Department of Urology, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, China
| | - Jing Li
- Department of Oncology and Hematology, Hospital (T.C.M) Affiliated to Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Ling Lin
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Shixin Xiang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yawei Li
- Department of Urology, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, China
| | - Yujie Xu
- Department of Urology, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| | - Xiang Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Weiping Wang
- Department of Pharmacy, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, China
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.,South Sichuan Institute of Translational Medicine, Luzhou, China
| |
Collapse
|
30
|
Matos B, Howl J, Jerónimo C, Fardilha M. The disruption of protein-protein interactions as a therapeutic strategy for prostate cancer. Pharmacol Res 2020; 161:105145. [PMID: 32814172 DOI: 10.1016/j.phrs.2020.105145] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/11/2020] [Accepted: 08/11/2020] [Indexed: 12/14/2022]
Abstract
Prostate cancer (PCa) is one of the most common male-specific cancers worldwide, with high morbidity and mortality rates associated with advanced disease stages. The current treatment options of PCa are prostatectomy, hormonal therapy, chemotherapy or radiotherapy, the selection of which is usually dependent upon the stage of the disease. The development of PCa to a castration-resistant phenotype (CRPC) is associated with a more severe prognosis requiring the development of a new and effective therapy. Protein-protein interactions (PPIs) have been recognised as an emerging drug modality and targeting PPIs is a promising therapeutic approach for several diseases, including cancer. The efficacy of several compounds in which target PPIs and consequently impair disease progression were validated in phase I/II clinical trials for different types of cancer. In PCa, various small molecules and peptides proved successful in inhibiting important PPIs, mainly associated with the androgen receptor (AR), Bcl-2 family proteins, and kinases/phosphatases, thus impairing the growth of PCa cells in vitro. Moreover, a majority of these compounds require further validation in vivo and, preferably, in clinical trials. In addition, several other PPIs associated with PCa progression have been identified and now require experimental validation as potential therapeutic loci. In conclusion, we consider the disruption of PPIs to be a promising though challenging therapeutic strategy for PCa. Agents which modulate PPIs might be employed as a monotherapy or as an adjunct to classical chemotherapeutics to overcome drug resistance and improve efficacy. The discovery of new PPIs with important roles in disease progression, and of novel optimized strategies to target them are major challenges for the scientific and pharmacological communities.
Collapse
Affiliation(s)
- Bárbara Matos
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal
| | - John Howl
- Molecular Pharmacology Group, Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton WV1 1LY, UK
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Institute of Oncology of Porto (IPO Porto), Research Center-LAB 3, F Bdg., 1st Floor, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar- University of Porto (ICBAS-UP), Porto, Portugal
| | - Margarida Fardilha
- Laboratory of Signal Transduction, Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, 3810-193, Aveiro, Portugal.
| |
Collapse
|
31
|
Xu Z, Ma T, Zhou J, Gao W, Li Y, Yu S, Wang Y, Chan FL. Nuclear receptor ERRα contributes to castration-resistant growth of prostate cancer via its regulation of intratumoral androgen biosynthesis. Theranostics 2020; 10:4201-4216. [PMID: 32226548 PMCID: PMC7086365 DOI: 10.7150/thno.35589] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 02/17/2020] [Indexed: 12/29/2022] Open
Abstract
Enhanced intratumoral androgen biosynthesis and persistent androgen receptor (AR) signaling are key factors responsible for the relapse growth of castration-resistant prostate cancer (CRPC). Residual intraprostatic androgens can be produced by de novo synthesis of androgens from cholesterol or conversion from adrenal androgens by steroidogenic enzymes expressed in prostate cancer cells via different steroidogenic pathways. However, the dysregulation of androgen biosynthetic enzymes in CRPC still remains poorly understood. This study aims to elucidate the role of the nuclear receptor, estrogen-related receptor alpha (ERRα, ESRRA), in the promotion of androgen biosynthesis in CRPC growth. Methods: ERRα expression in CRPC patients was analyzed using Gene Expression Omnibus (GEO) datasets and validated in established CRPC xenograft model. The roles of ERRα in the promotion of castration-resistant growth were elucidated by overexpression and knockdown studies and the intratumoral androgen levels were measured by UPLC-MS/MS. The effect of suppression of ERRα activity in the potentiation of sensitivity to androgen-deprivation was determined using an ERRα inverse agonist. Results: ERRα exhibited an increased expression in metastatic CRPC and CRPC xenograft model, could act to promote castration-resistant growth via direct transactivation of two key androgen synthesis enzymes CYP11A1 and AKR1C3, and hence enhance intraprostatic production of dihydrotestosterone (DHT) and activation of AR signaling in prostate cancer cells. Notably, inhibition of ERRα activity by an inverse agonist XCT790 could reduce the DHT production and suppress AR signaling in prostate cancer cells. Conclusion: Our study reveals a new role of ERRα in the intratumoral androgen biosynthesis in CRPC via its transcriptional control of steroidogenic enzymes, and also provides a novel insight that targeting ERRα could be a potential androgen-deprivation strategy for the management of CRPC.
Collapse
Affiliation(s)
- Zhenyu Xu
- Precision Medicine Centre, Yijishan Affiliated Hospital of Wannan Medical College, Wuhu, Anhui Province, PR China
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Taiyang Ma
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Jianfu Zhou
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
- Department of Urology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weijie Gao
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Youjia Li
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Shan Yu
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
32
|
Nuclear receptor HNF4α performs a tumor suppressor function in prostate cancer via its induction of p21-driven cellular senescence. Oncogene 2019; 39:1572-1589. [PMID: 31695151 PMCID: PMC7018660 DOI: 10.1038/s41388-019-1080-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 10/17/2019] [Accepted: 10/17/2019] [Indexed: 12/20/2022]
Abstract
Hepatocyte nuclear factor 4α (HNF4α, NR2A1) is a highly conserved member of the nuclear receptor superfamily. Recent advances reveal that it is a key transcriptional regulator of genes, broadly involved in xenobiotic and drug metabolism and also cancers of gastrointestinal tract. However, the exact functional roles of HNF4α in prostate cancer progression are still not fully understood. In this study, we determined the functional significance of HNF4α in prostate cancer. Our results showed that HNF4α exhibited a reduced expression pattern in clinical prostate cancer tissues, prostate cancer cell lines and xenograft model of castration-relapse prostate cancer. Stable HNF4α knockdown not only could promote cell proliferation and suppress doxorubicin (Dox)-induced cellular senescence in prostate cancer cells, but also confer resistance to paclitaxel treatment and enhance colony formation capacity and in vivo tumorigenicity of prostate cancer cells. On the contrary, ectopic overexpression of HNF4α could significantly inhibit the cell proliferation of prostate cancer cells, induce cell-cycle arrest at G2/M phase and trigger the cellular senescence in prostate cancer cells by activation of p21 signal pathway in a p53-independent manner via its direct transactivation of CDKN1A. Together, our results show that HNF4α performs a tumor suppressor function in prostate cancer via a mechanism of p21-driven cellular senescence.
Collapse
|
33
|
Li P, Wang J, Wu D, Ren X, Wu W, Zuo R, Zeng Q, Wang B, He X, Yuan J, Xie N. ERRα is an aggressive factor in lung adenocarcinoma indicating poor prognostic outcomes. Cancer Manag Res 2019; 11:8111-8123. [PMID: 31564971 PMCID: PMC6730612 DOI: 10.2147/cmar.s204732] [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: 02/10/2019] [Accepted: 07/28/2019] [Indexed: 12/12/2022] Open
Abstract
Purpose Lung cancer is one of the most life-threatening cancer worldwide with poor prognosis attributed to the lack of early diagnosis and proper therapy. The estrogen-related receptor alpha (ERRα) is a multifunctional protein not limited to bind ligands and has been reported to be associated with numerous cancers. This study aimed to investigate the potential role of ERRα in lung cancer and to provide a novel perspective for lung cancer early diagnosis, targeted therapy, and prognosis assessment. Methods The correlation between ERRα mRNA expression and survival time of the online clinical data about lung cancer was analyzed by using Kaplan–Meier (KM) plotter. A mouse model of lung adenocarcinoma (LUAD) was constructed to detect the expression level of ERRα in tumor tissues. ERRα-knockdown LUAD cells were generated and the impacts of ERRα on cell proliferation, invasion, and metastasis were further analyzed. Cancerous and paracancerous tissues were collected to semi-quantitative the levels of ERRα in LUAD clinical samples (n=88), combined with clinical information for prognostic analysis. Results The KM plotter analysis suggested that ERRα is correlated with poor prognosis in LUAD (n=720) rather than in lung squamous cell carcinoma (LSCC) (n=524). ERRα is also upregulated in tumor tissues obtained from LUAD model mice. Quantitative analysis suggested an abnormal elevation of ERRα in LUAD cells rather than in LSCC cells. The results demonstrated that downregulation of ERRα impairs proliferation, invasion and migration abilities (P<0.01). The prognostic analysis showed that the overexpressed ERRα in LUAD was positively correlated with low survival rates (HR=1.597). The results indicate that the death risk of ERRα high expression is 1.597 times higher than ERRα low level in LUAD patients. Conclusion In summary, our findings suggest that ERRα is a potential aggressive factor of LUAD which implies poor prognosis.
Collapse
Affiliation(s)
- Ping Li
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China.,Department of Medicine, University of South China, Hengyang 421001, People's Republic of China.,Department of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518035, People's Republic of China
| | - Jian Wang
- Department of Thoracic Surgery, The Shenzhen People's Hospital, Shenzhen 518020, People's Republic of China
| | - Desheng Wu
- Department of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518035, People's Republic of China
| | - Xiaohu Ren
- Department of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518035, People's Republic of China
| | - Wen Wu
- Department of Medicine, University of South China, Hengyang 421001, People's Republic of China.,Department of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518035, People's Republic of China
| | - Ran Zuo
- Department of Medicine, University of South China, Hengyang 421001, People's Republic of China
| | - Qingbo Zeng
- Department of Toxicology, Shenzhen Center for Disease Control and Prevention, Shenzhen 518035, People's Republic of China
| | - Bingyu Wang
- Department of Medicine, University of South China, Hengyang 421001, People's Republic of China
| | - Xi He
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China
| | - Jianhui Yuan
- Department of Medicine, University of South China, Hengyang 421001, People's Republic of China.,Department of Occupational Health, Shenzhen Nanshan District Center for Disease Control and Prevention, Shenzhen 518054, People's Republic of China
| | - Ni Xie
- Biobank, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen 518035, People's Republic of China.,Department of Medicine, University of South China, Hengyang 421001, People's Republic of China
| |
Collapse
|
34
|
Ye X, Guo J, Zhang H, Meng Q, Ma Y, Lin R, Yi X, Lu H, Bai X, Cheng J. The enhanced expression of estrogen-related receptor α in human bladder cancer tissues and the effects of estrogen-related receptor α knockdown on bladder cancer cells. J Cell Biochem 2019; 120:13841-13852. [PMID: 30977157 DOI: 10.1002/jcb.28657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/17/2018] [Accepted: 01/02/2019] [Indexed: 01/15/2023]
Abstract
Estrogen-related receptor α (ERRα) belongs to the superfamily of nuclear orphan receptors. However, the role of ERRα in bladder cancer remains unknown. This study examined the expression of ERRα in bladder cancer tissues and explored the molecular mechanisms of ERRα in bladder cancer progression. The expression of ERRα in bladder cancer tissues from 61 patients was determined by immunohistochemistry. We performed quantitative real-time polymerase chain reaction assay to detect the gene expression levels and carried out Western blot assay to measure protein levels. In vitro functional assays, including colony formation, Cell Counting Kit-8, Transwell invasion, and migration assays, were performed to detect bladder cancer cell growth, proliferation, invasion, and migration, respectively. Flow cytometry was used to determine the cell apoptotic rate of bladder cancer cells. Among the 61 detected bladder cancer tissues, 39 bladder cancer tissues showed positive ERRα immunoreactivity. Higher ERRα immunoreactivity score was significantly associated with TNM stage, tumor grade, distant metastasis, and poor survival in patients with bladder cancer. Univariate and multivariate analyses showed that ERRα immunoreactivity was an independent prognostic factor for overall survival in patients with bladder cancer. ERRα was found to be upregulated in bladder cancer cell lines, and knockdown of ERRα suppressed bladder cancer cell growth, proliferation, invasion, and migration; promoted bladder cancer cell apoptosis; and inhibited the epithelial-mesenchymal transition of bladder cancer cells. On the other hand, bladder cancer cell proliferation, invasion, and migration were significantly enhanced after cells were transfected with an ERRα-overexpressing vector. In vivo tumor growth and metastasis assays showed that ERRα knockdown resulted in remarkable inhibition of tumor growth and tumor metastasis in nude mice. Collectively, our results suggest that the enhanced expression of ERRα may play a key role in the development and progression of bladder cancer and ERRα may serve as an important prognostic factor for bladder cancer.
Collapse
Affiliation(s)
- Xinqing Ye
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jinan Guo
- Department of Urology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen Minimally Invasive Engineering Center, Shenzhen, China
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Hongxiang Zhang
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qinggui Meng
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yun Ma
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Rui Lin
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xianlin Yi
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Haoyuan Lu
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xianzhong Bai
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jiwen Cheng
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
35
|
Zonneveld MI, Keulers TGH, Rouschop KMA. Extracellular Vesicles as Transmitters of Hypoxia Tolerance in Solid Cancers. Cancers (Basel) 2019; 11:cancers11020154. [PMID: 30699970 PMCID: PMC6406242 DOI: 10.3390/cancers11020154] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 02/07/2023] Open
Abstract
Tumour hypoxia is a common feature of solid tumours that contributes to poor prognosis after treatment. This is mainly due to increased resistance of hypoxic cells to radio- and chemotherapy and the association of hypoxic cells with increased metastasis development. It is therefore not surprising that an increased hypoxic tumour fraction is associated with poor patient survival. The extent of hypoxia within a tumour is influenced by the tolerance of individual tumor cells to hypoxia, a feature that differs considerably between tumors. High numbers of hypoxic cells may, therefore, be a direct consequence of enhanced cellular capability inactivation of hypoxia tolerance mechanisms. These include HIF-1α signaling, the unfolded protein response (UPR) and autophagy to prevent hypoxia-induced cell death. Recent evidence shows hypoxia tolerance can be modulated by distant cells that have experienced episodes of hypoxia and is mediated by the systemic release of factors, such as extracellular vesicles (EV). In this review, the evidence for transfer of a hypoxia tolerance phenotype between tumour cells via EV is discussed. In particular, proteins, mRNA and microRNA enriched in EV, derived from hypoxic cells, that impact HIF-1α-, UPR-, angiogenesis- and autophagy signalling cascades are listed.
Collapse
Affiliation(s)
- Marijke I Zonneveld
- Maastricht Radiation Oncology (MaastRO) lab, GROW⁻School for Oncology and Developmental Biology, Maastricht University, 6200 MD Maastricht, The Netherlands.
| | - Tom G H Keulers
- Maastricht Radiation Oncology (MaastRO) lab, GROW⁻School for Oncology and Developmental Biology, Maastricht University, 6200 MD Maastricht, The Netherlands.
| | - Kasper M A Rouschop
- Maastricht Radiation Oncology (MaastRO) lab, GROW⁻School for Oncology and Developmental Biology, Maastricht University, 6200 MD Maastricht, The Netherlands.
| |
Collapse
|
36
|
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
|
37
|
Liu G, Sun P, Dong B, Sehouli J. Key regulator of cellular metabolism, estrogen-related receptor α, a new therapeutic target in endocrine-related gynecological tumor. Cancer Manag Res 2018; 10:6887-6895. [PMID: 30588094 PMCID: PMC6296681 DOI: 10.2147/cmar.s182466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The estrogen-related receptor α (ERRα), is an orphan transcription factor. Recently, many studies have reported its regulatory mechanisms and transcriptional targets after identification. Therefore, it may be eligible to join the rank of other nuclear receptors that control almost all aspects of cell metabolism. Cellular metabolism reprogramming plays a key role in fueling malignant change. The purpose of this review was to demonstrate that the ERRα plays an important role in the association between gynecological endocrine-related tumors and energy metabolism. Furthermore, regulation of ERRα may represent a promising strategy to induce cellular metabolic vulnerability of cancer from different origins. Thus, a comprehensive understanding of current treatment strategies may be achieved.
Collapse
Affiliation(s)
- GuiFen Liu
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - PengMing Sun
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China, .,Department of Gynaecology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - BinHua Dong
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - Jalid Sehouli
- Department of Gynaecologic Oncology and Gynaecology, Charité/Campus Virchow-Klinikum, European Competence Centre for Ovarian Cancer University of Berlin, Berlin 13353, Germany
| |
Collapse
|
38
|
Xu Z, Wang Y, Xiao ZG, Zou C, Zhang X, Wang Z, Wu D, Yu S, Chan FL. Nuclear receptor ERRα and transcription factor ERG form a reciprocal loop in the regulation of TMPRSS2:ERG fusion gene in prostate cancer. Oncogene 2018; 37:6259-6274. [PMID: 30042415 PMCID: PMC6265259 DOI: 10.1038/s41388-018-0409-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 05/17/2018] [Accepted: 06/19/2018] [Indexed: 11/08/2022]
Abstract
The TMPRSS2:ERG (T:E) fusion gene is generally believed to be mainly regulated by the activated androgen receptor (AR) signaling in androgen-dependent prostate cancer. However, its persistent expression in castration-resistant and neuroendocrine prostate cancers implies that other transcription factors might also regulate its expression. Here, we showed that up-regulation of nuclear receptor estrogen-related receptor alpha (ERRα) was closely associated with the oncogenic transcription factor ERG expression in prostate cancer, and their increased coexpression patterns were closely associated with high Gleason scores and metastasis in patients. Both ERRα and ERG exhibited a positive expression correlation in a castration-resistant prostate cancer (CRPC) xenograft model VCaP-CRPC. We showed that ERRα could directly transactivate T:E fusion gene in both AR-positive and -negative prostate cancer cells via both ERR-binding element- and AR-binding element-dependent manners. Ectopic T:E expression under ERRα regulation could promote both in vitro invasion and in vivo metastasis capacities of AR-negative prostatic cells. Intriguingly, ERG expressed by the T:E fusion could also transactivate the ERRα (ESRRA) gene. Hereby, ERRα and ERG can synergistically regulate each other and form a reciprocal regulatory loop to promote the advanced growth of prostate cancer. Inhibition of ERRα activity by ERRα inverse agonist could suppress T:E expression in prostate cancer cells, implicating that targeting ERRα could be a potential therapeutic strategy for treating the aggressive T:E-positive prostate cancer.
Collapse
Affiliation(s)
- Zhenyu Xu
- Department of Pharmacy, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, Anhui, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhan Gang Xiao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Pharmacology, Southwest Medical University, Luzhou, China
| | - Chang Zou
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Clinical Medical Research Center, Shenzhen People's Hospital, Second Clinical Medical College, Jinan University, Shenzhen, China
| | - Xian Zhang
- Department of Pharmacy, Yijishan Affiliated Hospital, Wannan Medical College, Wuhu, Anhui, China
| | - Zhu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dinglan Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shan Yu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
39
|
Gao W, Wu D, Wang Y, Wang Z, Zou C, Dai Y, Ng CF, Teoh JYC, Chan FL. Development of a novel and economical agar-based non-adherent three-dimensional culture method for enrichment of cancer stem-like cells. Stem Cell Res Ther 2018; 9:243. [PMID: 30257704 PMCID: PMC6158801 DOI: 10.1186/s13287-018-0987-x] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/07/2018] [Accepted: 08/17/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Non-adherent or ultra-low attachment three-dimensional (3D) culture, also called sphere formation assay, has been widely used to assess the malignant phenotype and stemness potential of transformed or cancer cells. This method is also popularly used to isolate the cancer stem-like cells (CSCs) or tumor-initiating cells based on their unique anchorage-independent growth or anoikis-resistant capacity. Different non-adhesive coating agents, such as poly-2-hydroxyethyl methacrylate (poly-HEMA) and synthetic hydrogels, have been used in this non-adherent 3D culture. However, preparation of non-adherent culture-ware is labor-intensive and technically demanding, and also costs of commercial non-adherent culture-ware prepared with various coating agents are relatively expensive and the culture-ware cannot be used repeatedly. METHODS In this study, we developed a non-adherent 3D culture method based on agar coating for growing tumor spheres derived from various cancer cell lines and primary prostate cancer tissues under a non-adherent and serum-free condition. The tumor spheres generated by this 3D culture method were analyzed on their expression profiles of CSC-associated markers by reverse transcription quantitative polymerase chain reaction, presence and relative proportion of CSCs by fluorescence-activated cell sorting (CD133+/CD44+ cell sorting) and also a CSC-visualizing reporter system responsive to OCT4 and SOX2 (SORE6), and in vivo tumorigenicity. The repeated use of agar-coated plates for serial passages of tumor spheres was also evaluated. RESULTS Our results validated that the multicellular tumor spheres generated by this culture method were enriched of CSCs, as evidenced by their enhanced expression profiles of CSC markers, presence of CD133+/CD44+ or SORE6+ cells, enhanced self-renewal capacity, and in vivo tumorigenicity, indicating its usefulness in isolation and enrichment of CSCs. The agar-coated plates could be used multiple times in serial passages of tumor spheres. CONCLUSIONS The described agar-based 3D culture method offers several advantages as compared with other methods in isolation of CSCs, including its simplicity and low-cost and repeated use of agar-coated plates for continuous passages of CSC-enriched spheres.
Collapse
Affiliation(s)
- Weijie Gao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Dinglan Wu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China. .,Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center, Shenzhen Hospital, Southern Medical University, Shenzhen, 518110, China.
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Zhu Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518000, China
| | - Yong Dai
- Clinical Medical Research Center, The Second Clinical Medical School of Jinan University, Shenzhen People's Hospital, Shenzhen, 518000, China
| | - Chi-Fai Ng
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeremy Yuen-Chun Teoh
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China.
| |
Collapse
|
40
|
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
|
41
|
Abstract
The eukaryotic nuclear receptors (NRs) super-family of transcriptional factors include the estrogen-related receptors (ERRs) that have diverse roles in control of cellular energy balance, general metabolism, growth and development, immunity etc. Mouse knock-out models of specific ERR isoforms (ERRα, ERRβ and ERRγ) exhibit defects in several phenotypic traits. Newer findings indicate important roles of ERRs in the regulation of brown adipocyte tissue mitochondrial oxidative functions as well as metabolic control in association with hypoxia-inducible factors during cellular hypoxic state. Genes involved in cardiac metabolism is also influenced by ERRα and ERRγ in association with the co-activators PGC-1α and PGC-1β. On the other hand, ERRs have crucial involvement at the interface of metabolism and diseases such as cancer. Recent findings have implicated ERRα in the progression of tumor and malignancy of the breast, prostate, colon, endometrium etc. In this article, new insights into the regulatory role of ERRs in metabolism and cancer shall be reviewed.
Collapse
Affiliation(s)
- Harmit S Ranhotra
- a Department of Biochemistry , St. Edmund's College , Shillong , India
| |
Collapse
|
42
|
Orphan nuclear receptor TLX contributes to androgen insensitivity in castration-resistant prostate cancer via its repression of androgen receptor transcription. Oncogene 2018; 37:3340-3355. [PMID: 29555975 PMCID: PMC6013422 DOI: 10.1038/s41388-018-0198-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 01/11/2018] [Accepted: 02/03/2018] [Indexed: 02/03/2023]
Abstract
The metastatic castration-resistant prostate cancer (CRPC) is a lethal form of prostate cancer, in which the expression of androgen receptor (AR) is highly heterogeneous. Indeed, lower AR expression and attenuated AR signature activity is shown in CRPC tissues, especially in the subset of neuroendocrine prostate cancer (NEPC) and prostate cancer stem-like cells (PCSCs). However, the significance of AR downregulation in androgen insensitivity and de-differentiation of tumor cells in CRPC is poorly understood and much neglected. Our previous study shows that the orphan nuclear receptor TLX (NR2E1), which is upregulated in prostate cancer, plays an oncogenic role in prostate carcinogenesis by suppressing oncogene-induced senescence. In the present study, we further established that TLX exhibited an increased expression in metastatic CRPC. Further analyses showed that overexpression of TLX could confer resistance to androgen deprivation and anti-androgen in androgen-dependent prostate cancer cells in vitro and in vivo, whereas knockdown of endogenous TLX could potentiate the sensitivity to androgen deprivation and anti-androgen in prostate cancer cells. Our study revealed that the TLX-induced resistance to androgen deprivation and anti-androgen was mediated through its direct suppression of AR gene transcription and signaling in both androgen-stimulated and -unstimulated prostate cancer cells. We also characterized that TLX could bind directly to AR promoter and repress AR transcription by recruitment of histone modifiers, including HDAC1, HDAC3, and LSD1. Together, our present study shows, for the first time, that TLX can contribute to androgen insensitivity in CRPC via repression of AR gene transcription and signaling, and also implicates that targeting the druggable TLX may have a potential therapeutic significance in CRPC management, particularly in NEPC and PCSCs.
Collapse
|
43
|
Fujimura T, Takayama K, Takahashi S, Inoue S. Estrogen and Androgen Blockade for Advanced Prostate Cancer in the Era of Precision Medicine. Cancers (Basel) 2018; 10:cancers10020029. [PMID: 29360794 PMCID: PMC5836061 DOI: 10.3390/cancers10020029] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 12/14/2022] Open
Abstract
Androgen deprivation therapy (ADT) has been widely prescribed for patients with advanced prostate cancer (PC) to control key signaling pathways via androgen receptor (AR) and AR-collaborative transcriptional factors; however, PC gradually acquires a lethal phenotype and results in castration-resistant PC (CRPC) during ADT. Therefore, new therapeutic strategies are required in clinical practice. In addition, ARs; estrogen receptors (ERs; ERα and ERβ); and estrogen-related receptors (ERRs; ERRα, ERRβ, and ERRγ) have been reported to be involved in the development or regulation of PC. Recent investigations have revealed the role of associated molecules, such as KLF5, FOXO1, PDGFA, VEGF-A, WNT5A, TGFβ1, and micro-RNA 135a of PC, via ERs and ERRs. Selective ER modulators (SERMs) have been developed. Recently, estrogen and androgen blockade (EAB) using a combination of toremifene and ADT has been demonstrated to improve biochemical recurrence rate in treatment-naïve bone metastatic PC. In the future, the suitability of ADT alone or EAB for individuals may be evaluated by making clinical decisions on the basis of information obtained from RT-PCR, gene-panel, or liquid biopsy to create a “personalized medicine” or “precision medicine”. In this review, we summarize ER and ERR signaling pathways, molecular diagnosis, and SERMs as candidates for advanced PC treatment.
Collapse
Affiliation(s)
- Tetsuya Fujimura
- Department of Urology, National Center for Global Health and Medicine, Tokyo 162-8655, Japan.
| | - Kenichi Takayama
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan.
| | - Satoru Takahashi
- Department of Urology, Nihon University School of Medicine, Tokyo 173-8610, Japan.
| | - Satoshi Inoue
- Department of Functional Biogerontology, Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan.
| |
Collapse
|
44
|
Wang Z, Wu D, Ng CF, Teoh JYC, Yu S, Wang Y, Chan FL. Nuclear receptor profiling in prostatospheroids and castration-resistant prostate cancer. Endocr Relat Cancer 2018; 25:35-50. [PMID: 29042395 DOI: 10.1530/erc-17-0280] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
Nuclear receptors (NRs), which belong to a superfamily of transcription factors and consist of a total of 48 members in humans, govern the expression of genes involved in a board range of developmental, reproductive, metabolic and immunological programs. Given the significant importance of androgen receptor and a few known NRs in the progression of prostate cancer, we surveyed the expression profiles of the entire NR superfamily in three-dimensional cultured prostatospheroids derived from different prostate cancer cell lines and a tumor xenograft model of castration-resistant prostate cancer VCaP-CRPC by quantitative real-time RT-PCR. Our results revealed that prostatospheroids and castration-relapse VCaP-CRPC xenografts, both contained enriched populations of prostate cancer stem/progenitor-like cells (PCSCs), displayed distinct expression patterns of NRs. Intriguingly, most of these differentially expressed NRs were orphan NRs and showed upregulation. Pairwise analysis identified five orphan NRs (including RORβ, TLX, COUP-TFII, NURR1 and LRH-1) that showed common upregulation in both mRNA and protein levels in the prostatospheroids and castration-relapse VCaP-CRPC xenografts, and overexpression of these orphan NRs could increase cancer stem cell marker expressions and enhance spheroid formation capacity in prostate cancer cells, suggesting that these orphan NRs might perform positive roles in the growth regulation of PCSCs and castration-resistant prostate cancer. Together, our NR expression dataset not only revealed the distinct physiologic status and regulatory roles governed by the networks of specific NRs but also some of these identified orphan NRs could be the potential therapeutic targets for PCSCs or castration-resistant prostate cancer.
Collapse
MESH Headings
- Animals
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- COUP Transcription Factor II/genetics
- COUP Transcription Factor II/metabolism
- Humans
- Male
- Mice, SCID
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Orphan Nuclear Receptors
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/pathology
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Receptors, Retinoic Acid/genetics
- Receptors, Retinoic Acid/metabolism
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Zhu Wang
- School of Biomedical SciencesThe Chinese University of Hong Kong, Hong Kong, China
- Department of UrologyPeople's Hospital of Longhua, Shenzhen, China
| | - Dinglan Wu
- School of Biomedical SciencesThe Chinese University of Hong Kong, Hong Kong, China
- The Clinical Innovation & Research CenterShenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chi-Fai Ng
- Department of SurgeryFaculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeremy Yuen-Chun Teoh
- Department of SurgeryFaculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Shan Yu
- School of Biomedical SciencesThe Chinese University of Hong Kong, Hong Kong, China
| | - Yuliang Wang
- School of Biomedical SciencesThe Chinese University of Hong Kong, Hong Kong, China
| | - Franky L Chan
- School of Biomedical SciencesThe Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
45
|
Yu G, Lin J, Liu C, Hou K, Liang M, Shi B. Long non-coding RNA SPRY4-IT1 promotes development of hepatic cellular carcinoma by interacting with ERRα and predicts poor prognosis. Sci Rep 2017; 7:17176. [PMID: 29214989 PMCID: PMC5719451 DOI: 10.1038/s41598-017-16781-9] [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: 02/21/2017] [Accepted: 10/05/2017] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) has become one of the most common leading causes of cancer-related deaths worldwide. This study investigates the role of lncRNA, SPRY4-IT1 in the development of HCC. Quantitative real-time PCR (qRT-PCR) was performed and the results showed that SPRY4-IT1 expression was up-regulated in HCC tissues and high expression of SPRY4-IT1 was associated with poor 5-year overall survival in the HCC patient cohort. Clinicopathological analysis showed that the expression of SPRY4-IT1 was significantly correlated with TNM stage in HCC patients. In vitro CCK-8 assay, colony formation assay, cell invasion and migration assays demonstrated that knock-down of SPRY4-IT1 suppressed cell proliferation, colony formation, cell invasion and migration in HCC cells. Flow cytometric analysis showed that knock-down of SPRY4-IT1 induced cell cycle arrest at G0/G1 phase and induced apoptosis. In addition, knock-down of SPRY4-IT1 also suppressed the mRNA and protein expression of estrogen-related receptor α (ERRα). Similarly, knock-down of ERRα inhibited cell proliferation, colony formation, cell invasion and migration in HCC cells. More importantly, ERRα overexpression antagonized the effects of SPRY4-IT1 knock-down on cell proliferation, colony formation, cell invasion and migration in HCC cells. Taken together, our data highlights the pivotal role of SPRY4-IT1 in the tumorigenesis of HCC.
Collapse
Affiliation(s)
- Guifang Yu
- The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China.
| | - Jieheng Lin
- The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China
| | - Chengcheng Liu
- The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China
| | - Kailian Hou
- The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China
| | - Min Liang
- The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China
| | - Boyun Shi
- The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou, Guangdong, China
| |
Collapse
|
46
|
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
|
47
|
Bondanese VP, Lamboux A, Simon M, Lafont JE, Albalat E, Pichat S, Vanacker JM, Telouk P, Balter V, Oger P, Albarède F. Hypoxia induces copper stable isotope fractionation in hepatocellular carcinoma, in a HIF-independent manner. Metallomics 2017; 8:1177-1184. [PMID: 27500357 DOI: 10.1039/c6mt00102e] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most frequent type of primary liver cancer, with increasing incidence worldwide. The unrestrained proliferation of tumour cells leads to tumour hypoxia which in turn promotes cancer aggressiveness. While changes in the concentration of copper (Cu) have long been observed upon cancerization, we have recently reported that the isotopic composition of copper is also altered in several types of cancer. In particular, we showed that in hepatocellular carcinoma, tumour tissue contains heavier copper compared to the surrounding parenchyma. However, the reasons behind such isotopic signature remained elusive. Here we show that hypoxia causes heavy copper enrichment in several human cell lines. We also demonstrate that this effect of hypoxia is pH, HIF-1 and -2 independent. Our data identify a previously unrecognized cellular process associated with hypoxia, and suggests that in vivo tumour hypoxia determines copper isotope fractionation in HCC and other solid cancers.
Collapse
Affiliation(s)
- Victor P Bondanese
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Aline Lamboux
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Melanie Simon
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Jérôme E Lafont
- Institute for Biology and Chemistry of Proteins, CNRS, UMR 5305 Laboratory of Tissue Biology and Therapeutic Engineering, Université Claude Bernard-Lyon 1 and University of Lyon, France
| | - Emmanuelle Albalat
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Sylvain Pichat
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 69007 Lyon, France
| | - Philippe Telouk
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Vincent Balter
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Philippe Oger
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| | - Francis Albarède
- Univ Lyon, ENS de Lyon, Université Lyon 1, CNRS, UMR 5276 LGL-TPE, F-69342, Lyon, France.
| |
Collapse
|
48
|
Wu Y, Zhang Q, Zhang R. Kaempferol targets estrogen-related receptor α and suppresses the angiogenesis of human retinal endothelial cells under high glucose conditions. Exp Ther Med 2017; 14:5576-5582. [PMID: 29285095 DOI: 10.3892/etm.2017.5261] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 03/17/2017] [Indexed: 12/22/2022] Open
Abstract
Diabetic retinopathy (DR) is the most common complication of diabetes and a major cause of new-onset blindness in the developed world. The present study aimed to examine the effect of kaempferol on high glucose-induced human retinal endothelial cells (HRECs) in vitro. The expression levels of various mRNAs and proteins were measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting, respectively. The target of kaempferol was determined using a luciferase reporter assay. In addition, HREC proliferation, migration and cell sprouting were determined using Cell Counting kit-8, wound scratch and tube formation assays, respectively. RT-qPCR and western blotting results showed that treatment with 30 mM glucose for 12, 24 and 48 h increased the expression level of estrogen-related receptor α (ERRα) mRNA and protein. The luciferase reporter assay demonstrated that kaempferol inhibited ERRα activity in HRECs. Compared with 5 mM normal glucose treatment, high (30 mM) glucose significantly promoted the proliferation, migration and tube formation of HRECs, which was antagonized by 10 and 30 µM kaempferol in a dose-dependent manner. Treatment with 30 mM glucose also increased the expression of vascular endothelial growth factor (VEGF) mRNA and protein, and the expression levels of VEGF mRNA and protein were suppressed by kaempferol (10 and 30 µM). Kaempferol (30 µM) treatment also increased the expression levels of thrombospondin 1 (TSP-1) and a disintegrin and metalloproteinase with thrombospondin motifs 1 (ADAMTS-1) mRNA; however, TSP-1 and ADAMTS-1 levels did not differ between high glucose and normal (5 mM) glucose conditions. The results of this study suggest that kaempferol targets ERRα and suppresses the angiogenesis of HRECs under high glucose conditions. Kaempferol may be a potential drug for use in controlling the progression of DR; however, in vivo studies are required to evaluate its efficacy and safety.
Collapse
Affiliation(s)
- Yan Wu
- Department of Endocrinology, The Second Clinical College of Jinan University, Shenzhen Peoples' Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Qinmei Zhang
- Department of Endocrinology, The Second Clinical College of Jinan University, Shenzhen Peoples' Hospital, Shenzhen, Guangdong 518020, P.R. China
| | - Rui Zhang
- Department of Endocrinology, The Second Clinical College of Jinan University, Shenzhen Peoples' Hospital, Shenzhen, Guangdong 518020, P.R. China
| |
Collapse
|
49
|
Semenza GL. A compendium of proteins that interact with HIF-1α. Exp Cell Res 2017; 356:128-135. [PMID: 28336293 DOI: 10.1016/j.yexcr.2017.03.041] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/18/2017] [Indexed: 12/23/2022]
Abstract
Hypoxia-inducible factor 1 (HIF-1) is the founding member of a family of transcription factors that function as master regulators of oxygen homeostasis. HIF-1 is composed of an O2-regulated HIF-1α subunit and a constitutively expressed HIF-1β subunit. This review provides a compendium of proteins that interact with the HIF-1α subunit, many of which regulate HIF-1 activity in either an O2-dependent or O2-independent manner.
Collapse
Affiliation(s)
- Gregg L Semenza
- Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, MD 21205 USA.
| |
Collapse
|
50
|
Fradet A, Bouchet M, Delliaux C, Gervais M, Kan C, Benetollo C, Pantano F, Vargas G, Bouazza L, Croset M, Bala Y, Leroy X, Rosol TJ, Rieusset J, Bellahcène A, Castronovo V, Aubin JE, Clézardin P, Duterque-Coquillaud M, Bonnelye E. Estrogen related receptor alpha in castration-resistant prostate cancer cells promotes tumor progression in bone. Oncotarget 2016; 7:77071-77086. [PMID: 27776343 PMCID: PMC5363569 DOI: 10.18632/oncotarget.12787] [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: 07/04/2016] [Accepted: 10/13/2016] [Indexed: 12/19/2022] Open
Abstract
Bone metastases are one of the main complications of prostate cancer and they are incurable. We investigated whether and how estrogen receptor-related receptor alpha (ERRα) is involved in bone tumor progression associated with advanced prostate cancer. By meta-analysis, we first found that ERRα expression is correlated with castration-resistant prostate cancer (CRPC), the hallmark of progressive disease. We then analyzed tumor cell progression and the associated signaling pathways in gain-of-function/loss-of-function CRPC models in vivo and in vitro. Increased levels of ERRα in tumor cells led to rapid tumor progression, with both bone destruction and formation, and direct impacts on osteoclasts and osteoblasts. VEGF-A, WNT5A and TGFβ1 were upregulated by ERRα in tumor cells and all of these factors also significantly and positively correlated withERRα expression in CRPC patient specimens. Finally, high levels of ERRα in tumor cells stimulated the pro-metastatic factor periostin expression in the stroma, suggesting that ERRα regulates the tumor stromal cell microenvironment to enhance tumor progression. Taken together, our data demonstrate that ERRα is a regulator of CRPC cell progression in bone. Therefore, inhibiting ERRα may constitute a new therapeutic strategy for prostate cancer skeletal-related events.
Collapse
Affiliation(s)
- Anais Fradet
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | - Mathilde Bouchet
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | - Carine Delliaux
- CNRS-UMR8161, F-59021 Lille, France
- Université-Lille, F-59000 Lille, France
| | - Manon Gervais
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | - Casina Kan
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | - Claire Benetollo
- Université-Lyon1, F-69008 Lyon, France
- InsermU1028-CNRS-UMR5292, Lyon, France
| | | | - Geoffrey Vargas
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | - Lamia Bouazza
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | - Martine Croset
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | - Yohann Bala
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | | | | | | | | | | | - Jane E Aubin
- University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Philippe Clézardin
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| | | | - Edith Bonnelye
- InsermUMR1033, F-69372 Lyon, France
- Université-Lyon1, F-69008 Lyon, France
| |
Collapse
|