1
|
Chen W, Song YS, Lee HS, Lin CW, Lee J, Kang YE, Kim SK, Kim SY, Park YJ, Park JI. Estrogen-related receptor alpha promotes thyroid tumor cell survival via a tumor subtype-specific regulation of target gene networks. Oncogene 2024; 43:2431-2446. [PMID: 38937602 DOI: 10.1038/s41388-024-03078-1] [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: 10/18/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024]
Abstract
Mortalin (encoded by HSPA9) is a mitochondrial chaperone often overexpressed in cancer through as-yet-unknown mechanisms. By searching different RNA-sequencing datasets, we found that ESRRA is a transcription factor highly correlated with HSPA9 in thyroid cancer, especially in follicular, but not C cell-originated, tumors. Consistent with this correlation, ESRRA depletion decreased mortalin expression only in follicular thyroid tumor cells. Further, ESRRA expression and activity were relatively high in thyroid tumors with oncocytic characteristics, wherein ESRRA and mortalin exhibited relatively high functional overlap. Mechanistically, ESRRA directly regulated HSPA9 transcription through a novel ESRRA-responsive element located upstream of the HSPA9 promoter. Physiologically, ESRRA depletion suppressed thyroid tumor cell survival via caspase-dependent apoptosis, which ectopic mortalin expression substantially abrogated. ESRRA depletion also effectively suppressed tumor growth and mortalin expression in the xenografts of oncocytic or ESRRA-overexpressing human thyroid tumor cells in mice. Notably, our Bioinformatics analyses of patient data revealed two ESRRA target gene clusters that contrast oncocytic-like and anaplastic features of follicular thyroid tumors. These findings suggest that ESRRA is a tumor-specific regulator of mortalin expression, the ESRRA-mortalin axis has higher significance in tumors with oncocytic characteristics, and ESRRA target gene networks can refine molecular classification of thyroid cancer.
Collapse
Affiliation(s)
- Wenjing Chen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Young Shin Song
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Han Sai Lee
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chien-Wei Lin
- Division of Biostatistics, Institute for Health and Equity, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Junguee Lee
- Department of Pathology, Konyang University School of Medicine, Daejeon, Republic of Korea
| | - Yea Eun Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chungnam National University Hospital & College of Medicine, Daejeon, Republic of Korea
| | - Seon-Kyu Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seon-Young Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Young Joo Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea
| | - Jong-In Park
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
2
|
Zhang X, Li H, Wang Y, Zhao H, Wang Z, Chan FL. Nuclear receptor NURR1 functions to promote stemness and epithelial-mesenchymal transition in prostate cancer via its targeting of Wnt/β-catenin signaling pathway. Cell Death Dis 2024; 15:234. [PMID: 38531859 DOI: 10.1038/s41419-024-06621-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 03/28/2024]
Abstract
Dysregulated activation of Wnt/β-catenin signaling pathway is a frequent or common event during advanced progression of multiple cancers. With this signaling activation, it enhances their tumorigenic growth and facilitates metastasis and therapy resistance. Advances show that this signaling pathway can play dual regulatory roles in the control of cellular processes epithelial-mesenchymal transition (EMT) and cancer stemness in cancer progression. Aberrant activation of Wnt/β-catenin signaling pathway is shown to be common in prostate cancer and also castration-resistant prostate cancer (CRPC). However, the transcriptional regulators of this pathway in prostate cancer are still not well characterized. NURR1 (NR4A2) is an orphan nuclear receptor and plays an important role in the development of dopaminergic neurons. Previously, we have shown that NURR1 exhibits an upregulation in isolated prostate cancer stem-like cells (PCSCs) and a xenograft model of CRPC. In this study, we further confirmed that NURR1 exhibited an upregulation in prostate cancer and also enhanced expression in prostate cancer cell lines. Functional and molecular analyses showed that NURR1 could act to promote both in vitro (cancer stemness and EMT) and also in vivo oncogenic growth of prostate cancer cells (metastasis and castration resistance) via its direct transactivation of CTNNB1 (β-catenin) and activation of β-catenin to mediate the activation of Wnt/β-catenin signaling pathway. Moreover, we also demonstrated that NURR1 activity in prostate cancer cells could be modulated by small molecules, implicating that NURR1 could be a potential therapeutic target for advanced prostate cancer management.
Collapse
Affiliation(s)
- Xingxing Zhang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Haolong Li
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
- Department of Urology, The People's Hospital of Longhua, Shenzhen, 518109, Guangdong, China
| | - Yuliang Wang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hui Zhao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhu Wang
- Department of Urology, The People's Hospital of Longhua, Shenzhen, 518109, Guangdong, China.
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Alqualo NO, Campos-Fernandez E, Picolo BU, Ferreira EL, Henriques LM, Lorenti S, Moreira DC, Simião MPS, Oliveira LBT, Alonso-Goulart V. Molecular biomarkers in prostate cancer tumorigenesis and clinical relevance. Crit Rev Oncol Hematol 2024; 194:104232. [PMID: 38101717 DOI: 10.1016/j.critrevonc.2023.104232] [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: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023] Open
Abstract
Prostate cancer (PCa) is the second most frequent type of cancer in men and assessing circulating tumor cells (CTCs) by liquid biopsy is a promising tool to help in cancer early detection, staging, risk of recurrence evaluation, treatment prediction and monitoring. Blood-based liquid biopsy approaches enable the enrichment, detection and characterization of CTCs by biomarker analysis. Hence, comprehending the molecular markers, their role on each stage of cancer development and progression is essential to provide information that can help in future implementation of these biomarkers in clinical assistance. In this review, we studied the molecular markers most associated with PCa CTCs to better understand their function on tumorigenesis and metastatic cascade, the methodologies utilized to analyze these biomarkers and their clinical significance, in order to summarize the available information to guide researchers in their investigations, new hypothesis formulation and target choice for the development of new diagnostic and treatment tools.
Collapse
Affiliation(s)
- Nathalia Oliveira Alqualo
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Esther Campos-Fernandez
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Bianca Uliana Picolo
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Emanuelle Lorrayne Ferreira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Laila Machado Henriques
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Sabrina Lorenti
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Danilo Caixeta Moreira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Maria Paula Silva Simião
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Luciana Beatriz Tiago Oliveira
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil
| | - Vivian Alonso-Goulart
- Laboratory of Nanobiotechnology, Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Universidade Federal de Uberlândia, Uberlandia, MG 38400-902, Brazil.
| |
Collapse
|
4
|
Chang X, Ismail NI, Rahman A, Xu D, Chan RWY, Ong SG, Ong SB. Long COVID-19 and the Heart: Is Cardiac Mitochondria the Missing Link? Antioxid Redox Signal 2023; 38:599-618. [PMID: 36053670 PMCID: PMC10025846 DOI: 10.1089/ars.2022.0126] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 08/27/2022] [Indexed: 12/30/2022]
Abstract
Significance: Although corona virus disease 2019 (COVID-19) has now gradually been categorized as an endemic, the long-term effect of COVID-19 in causing multiorgan disorders, including a perturbed cardiovascular system, is beginning to gain attention. Nonetheless, the underlying mechanism triggering post-COVID-19 cardiovascular dysfunction remains enigmatic. Are cardiac mitochondria the key to mediating cardiac dysfunction post-severe acute respiratory syndrome coronavirus 2 (post-SARS-CoV-2) infection? Recent Advances: Cardiovascular complications post-SARS-CoV-2 infection include myocarditis, myocardial injury, microvascular injury, pericarditis, acute coronary syndrome, and arrhythmias (fast or slow). Different types of myocardial damage or reduced heart function can occur after a lung infection or lung injury. Myocardial/coronary injury or decreased cardiac function is directly associated with increased mortality after hospital discharge in patients with COVID-19. The incidence of adverse cardiovascular events increases even in recovered COVID-19 patients. Disrupted cardiac mitochondria postinfection have been postulated to lead to cardiovascular dysfunction in the COVID-19 patients. Further studies are crucial to unravel the association between SARS-CoV-2 infection, mitochondrial dysfunction, and ensuing cardiovascular disorders (CVD). Critical Issues: The relationship between COVID-19 and myocardial injury or cardiovascular dysfunction has not been elucidated. In particular, the role of the cardiac mitochondria in this association remains to be determined. Future Directions: Elucidating the cause of cardiac mitochondrial dysfunction post-SARS-CoV-2 infection may allow a deeper understanding of long COVID-19 and resulting CVD, thus providing a potential therapeutic target. Antioxid. Redox Signal. 38, 599-618.
Collapse
Affiliation(s)
- Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Nur Izzah Ismail
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Attaur Rahman
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Dachun Xu
- Department of Cardiology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Cardiology, Qidong People's Hospital, Qidong, China
| | - Renee Wan Yi Chan
- Department of Paediatrics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Laboratory for Paediatric Respiratory Research, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), Hong Kong Children's Hospital (HKCH), Hong Kong SAR, China
- Department of Paediatrics, Chinese University of Hong Kong-University Medical Center Utrecht Joint Research Laboratory of Respiratory Virus and Immunobiology, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
| | - Sang-Ging Ong
- Department of Pharmacology & Regenerative Medicine, The University of Illinois College of Medicine, Chicago, Illinois, USA
- Division of Cardiology, Department of Medicine, The University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Sang-Bing Ong
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, The Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence (HK HOPE), Hong Kong Children's Hospital (HKCH), Hong Kong SAR, China
- Kunming Institute of Zoology—The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
5
|
Maurya SK, Baghel MS, Gaurav, Chaudhary V, Kaushik A, Gautam A. Putative role of mitochondria in SARS-CoV-2 mediated brain dysfunctions: a prospect. Biotechnol Genet Eng Rev 2022:1-26. [PMID: 35934991 DOI: 10.1080/02648725.2022.2108998] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/26/2022] [Indexed: 12/13/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the COVID-19 pandemic. Though the virus primarily damages the respiratory and cardiovascular systems after binding to the host angiotensin-converting enzyme 2 (ACE2) receptors, it has the potential to affect all major organ systems, including the human nervous system. There are multiple clinical reports of anosmia, dizziness, headache, nausea, ageusia, encephalitis, demyelination, neuropathy, memory loss, and neurological complications in SARS-CoV-2 infected individuals. Though the molecular mechanism of these brain dysfunctions during SARS-CoV-2 infection is elusive, the mitochondria seem to be an integral part of this pathogenesis. Emerging research findings suggest that the dysfunctional mitochondria and associated altered bioenergetics in the infected host cells lead to altered energy metabolism in the brain of Covid-19 patients. The interactome between viral proteins and mitochondrial proteins during Covid-19 pathogenesis also provides evidence for the involvement of mitochondria in SARS-CoV-2-induced brain dysfunctions. The present review discusses the possible role of mitochondria in disturbing the SARS-CoV-2 mediated brain functions, with the potential to use this information to prevent and treat these impairments.
Collapse
Affiliation(s)
| | - Meghraj S Baghel
- Department of Pathology, School of Medicine Johns Hopkins University, Baltimore, MD, USA
| | - Gaurav
- Department of Botany, Ramjas College, University of Delhi, Delhi, India
| | - Vishal Chaudhary
- Research Cell and Department of Physics, Bhagini Nivedita College, University of Delhi, New Delhi, India
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health System Engineering, Department ofEnvironmental Engineering, Florida Polytechnic University, Lakeland, FL, USA
| | - Akash Gautam
- Centre for Neural and Cognitive Sciences, University of Hyderabad, Hyderabad, India
| |
Collapse
|
6
|
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
|
7
|
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: 0.5] [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
|
8
|
PGC1 alpha coactivates ERG fusion to drive antioxidant target genes under metabolic stress. Commun Biol 2022; 5:416. [PMID: 35508713 PMCID: PMC9068611 DOI: 10.1038/s42003-022-03385-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 04/20/2022] [Indexed: 12/02/2022] Open
Abstract
The presence of ERG gene fusion; from developing prostatic intraepithelial neoplasia (PIN) lesions to hormone resistant high grade prostate cancer (PCa) dictates disease progression, altered androgen metabolism, proliferation and metastasis1–3. ERG driven transcriptional landscape may provide pro-tumorigenic cues in overcoming various strains like hypoxia, nutrient deprivation, inflammation and oxidative stress. However, insights on the androgen independent regulation and function of ERG during stress are limited. Here, we identify PGC1α as a coactivator of ERG fusion under various metabolic stress. Deacetylase SIRT1 is necessary for PGC1α-ERG interaction and function. We reveal that ERG drives the expression of antioxidant genes; SOD1 and TXN, benefitting PCa growth. We observe increased expression of these antioxidant genes in patients with high ERG expression correlates with poor survival. Inhibition of PGC1α-ERG axis driven transcriptional program results in apoptosis and reduction in PCa xenografts. Here we report a function of ERG under metabolic stress which warrants further studies as a therapeutic target for ERG fusion positive PCa. PGC1α acts as a co-activator of the ERG transcription factor during metabolic stress resulting in antioxidant functionsand inhibition of the PGC1α-ERG driven transcriptional program reduces prostate cancer growth by inducing ROS mediated apoptosis.
Collapse
|
9
|
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: 2.0] [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
|
10
|
Villa C, Rivellini E, Lavitrano M, Combi R. Can SARS-CoV-2 Infection Exacerbate Alzheimer's Disease? An Overview of Shared Risk Factors and Pathogenetic Mechanisms. J Pers Med 2022; 12:29. [PMID: 35055344 PMCID: PMC8780286 DOI: 10.3390/jpm12010029] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/11/2022] Open
Abstract
The current coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus (SARS-CoV)-2, is affecting every aspect of global society, including public healthcare systems, medical care access, and the economy. Although the respiratory tract is primarily affected by SARS-CoV-2, emerging evidence suggests that the virus may also reach the central nervous system (CNS), leading to several neurological issues. In particular, people with a diagnosis of Alzheimer's disease (AD) are a vulnerable group at high risk of contracting COVID-19, and develop more severe forms and worse outcomes, including death. Therefore, understanding shared links between COVID-19 and AD could aid the development of therapeutic strategies against both. Herein, we reviewed common risk factors and potential pathogenetic mechanisms that might contribute to the acceleration of neurodegenerative processes in AD patients infected by SARS-CoV-2.
Collapse
Affiliation(s)
- Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Eleonora Rivellini
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Romina Combi
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
11
|
Di Girolamo FG, Fiotti N, Sisto UG, Nunnari A, Colla S, Mearelli F, Vinci P, Schincariol P, Biolo G. Skeletal Muscle in Hypoxia and Inflammation: Insights on the COVID-19 Pandemic. Front Nutr 2022; 9:865402. [PMID: 35529457 PMCID: PMC9072827 DOI: 10.3389/fnut.2022.865402] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/09/2022] [Indexed: 12/23/2022] Open
Abstract
SARS-CoV-2 infection is often associated with severe inflammation, oxidative stress, hypoxia and impaired physical activity. These factors all together contribute to muscle wasting and fatigue. In addition, there is evidence of a direct SARS-CoV-2 viral infiltration into skeletal muscle. Aging is often characterized by sarcopenia or sarcopenic obesity These conditions are risk factors for severe acute COVID-19 and long-COVID-19 syndrome. From these observations we may predict a strong association between COVID-19 and decreased muscle mass and functions. While the relationship between physical inactivity, chronic inflammation, oxidative stress and muscle dysfunction is well-known, the effects on muscle mass of COVID-19-related hypoxemia are inadequately investigated. The aim of this review is to highlight metabolic, immunity-related and redox biomarkers potentially affected by reduced oxygen availability and/or muscle fatigue in order to shed light on the negative impact of COVID-19 on muscle mass and function. Possible countermeasures are also reviewed.
Collapse
Affiliation(s)
- Filippo G. Di Girolamo
- Department of Medical Surgical ad Health Science, Clinica Medica, Cattinara Hospital, University of Trieste, Trieste, Italy
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
- *Correspondence: Filippo G. Di Girolamo
| | - Nicola Fiotti
- Department of Medical Surgical ad Health Science, Clinica Medica, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Ugo G. Sisto
- Department of Medical Surgical ad Health Science, Clinica Medica, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Alessio Nunnari
- Department of Medical Surgical ad Health Science, Clinica Medica, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Stefano Colla
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Filippo Mearelli
- Department of Medical Surgical ad Health Science, Clinica Medica, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Pierandrea Vinci
- Department of Medical Surgical ad Health Science, Clinica Medica, Cattinara Hospital, University of Trieste, Trieste, Italy
| | - Paolo Schincariol
- SC Assistenza Farmaceutica, Cattinara Hospital, Azienda Sanitaria Universitaria Giuliano Isontina, Trieste, Italy
| | - Gianni Biolo
- Department of Medical Surgical ad Health Science, Clinica Medica, Cattinara Hospital, University of Trieste, Trieste, Italy
| |
Collapse
|
12
|
Fu Z, Liang X, Shi L, Tang L, Chen D, Liu A, Shao C. SYT8 promotes pancreatic cancer progression via the TNNI2/ERRα/SIRT1 signaling pathway. Cell Death Dis 2021; 7:390. [PMID: 34907162 PMCID: PMC8671424 DOI: 10.1038/s41420-021-00779-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/08/2021] [Accepted: 11/24/2021] [Indexed: 12/26/2022]
Abstract
Pancreatic cancer is a highly lethal malignancy due to failures of early detection and high metastasis in patients. While certain genetic mutations in tumors are associated with severity, the molecular mechanisms responsible for cancer progression are still poorly understood. Synaptotagmin-8 (SYT8) is a membrane protein that regulates hormone secretion and neurotransmission, and its expression is positively regulated by the promoter of the insulin gene in pancreatic islet cells. In this study, we identified a previously unknown role of SYT8 in altering tumor characteristics in pancreatic cancer. SYT8 levels were upregulated in patient tumors and contributed towards increased cell proliferation, migration, and invasion in vitro and in vivo. Increased SYT8 expression also promoted tumor metastasis in an in vivo tumor metastasis model. Furthermore, we showed that SYT8-mediated increase in tumorigenicity was regulated by SIRT1, a protein deacetylase previously known to alter cell metabolism in pancreatic lesions. SIRT1 expression was altered by orphan nuclear receptor ERRα and troponin-1 (TNNI2), resulting in cell proliferation and migration in an SYT8-dependent manner. Together, we identified SYT8 to be a central regulator of tumor progression involving signaling via the SIRT1, ERRα, and TNNI2 axis. This knowledge may provide the basis for the development of therapeutic strategies to restrict tumor metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Zhiping Fu
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xing Liang
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ligang Shi
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Liang Tang
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Danlei Chen
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Anan Liu
- grid.73113.370000 0004 0369 1660Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chenghao Shao
- Department of Pancreatic-Biliary Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
13
|
Prostate cancer and SARS-CoV-2: possible intersections through the TMPRSS2 pathway. Eur J Cancer Prev 2021; 30:481-483. [PMID: 34596106 DOI: 10.1097/cej.0000000000000696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Kaundal RK, Kalvala AK, Kumar A. Neurological Implications of COVID-19: Role of Redox Imbalance and Mitochondrial Dysfunction. Mol Neurobiol 2021; 58:4575-4587. [PMID: 34110602 PMCID: PMC8190166 DOI: 10.1007/s12035-021-02412-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 or COVID-19 has been declared as a pandemic disease by the World Health Organization (WHO). Globally, this disease affected 159 million of the population and reported ~ 3.3 million deaths to the current date (May 2021). There is no definitive treatment strategy that has been identified, although this disease has prevailed in its current form for the past 18 months. The main challenges in the (SARS-CoV)-2 infections are in identifying the heterogeneity in viral strains and the plausible mechanisms of viral infection to human tissues. In parallel to the investigations into the patho-mechanism of SARS-CoV-2 infection, understanding the fundamental processes underlying the clinical manifestations of COVID-19 is very crucial for designing effective therapies. Since neurological symptoms are very apparent in COVID-19 infected patients, here, we tried to emphasize the involvement of redox imbalance and subsequent mitochondrial dysfunction in the progression of the COVID-19 infection. It has been articulated that mitochondrial dysfunction is very apparent and also interlinked to neurological symptoms in COVID-19 infection. Overall, this article provides an in-depth overview of redox imbalance and mitochondrial dysfunction involvement in aggravating COVID-19 infection and its probable contribution to the neurological manifestation of the disease.
Collapse
Affiliation(s)
- Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
- Icahn School of Medicine At Mount Sinai, 1470 Madison Ave, New York, NY, USA
| | - Anil K Kalvala
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, North America, USA
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, India.
| |
Collapse
|
15
|
Rahbar Saadat Y, Hosseiniyan Khatibi SM, Zununi Vahed S, Ardalan M. Host Serine Proteases: A Potential Targeted Therapy for COVID-19 and Influenza. Front Mol Biosci 2021; 8:725528. [PMID: 34527703 PMCID: PMC8435734 DOI: 10.3389/fmolb.2021.725528] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/11/2021] [Indexed: 12/23/2022] Open
Abstract
The ongoing pandemic illustrates limited therapeutic options for controlling SARS-CoV-2 infections, calling a need for additional therapeutic targets. The viral spike S glycoprotein binds to the human receptor angiotensin-converting enzyme 2 (ACE2) and then is activated by the host proteases. Based on the accessibility of the cellular proteases needed for SARS-S activation, SARS-CoV-2 entrance and activation can be mediated by endosomal (such as cathepsin L) and non-endosomal pathways. Evidence indicates that in the non-endosomal pathway, the viral S protein is cleaved by the furin enzyme in infected host cells. To help the virus enter efficiently, the S protein is further activated by the serine protease 2 (TMPRSS2), provided that the S has been cleaved by furin previously. In this review, important roles for host proteases within host cells will be outlined in SARS-CoV-2 infection and antiviral therapeutic strategies will be highlighted. Although there are at least five highly effective vaccines at this time, the appearance of the new viral mutations demands the development of therapeutic agents. Targeted inhibition of host proteases can be used as a therapeutic approach for viral infection.
Collapse
|
16
|
Gupta VK, Murthy MK, Patil S. Can Host Cell Proteins Like ACE2, ADAM17, TMPRSS2, Androgen Receptor be the Efficient Targets in SARS-CoV-2 Infection? Curr Drug Targets 2021; 22:1149-1157. [PMID: 33243116 DOI: 10.2174/1389450121999201125201112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 11/22/2022]
Abstract
A novel betacoronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV- -2), which caused a large disease outbreak in Wuhan, China in December 2019, is currently spreading across the world. Along with binding of the virus spike with the host cell receptor, fusion of the viral envelope with host cell membranes is a critical step in establishing successful infection of SARS-CoV-2. In this entry process, a diversity of host cell proteases and androgen receptor play a very important role directly or indirectly. These features of SARS-CoV-2 entry contribute to its rapid spread and severe symptoms, high fatality rates among infected patients. This review is based on the latest published literature including review articles, research articles, hypothetical manuscript, preprint articles and official documents. The literature search was made from various published papers on physiological aspects relevant to SARS-CoV and SARS-CoV-2. In this report, we focus on the role of host cell proteases (ACE2, ADAM17, TMPRSS2) and androgen receptor (AR) in SARS-CoV-2 infection. The hypotheses put forth by us are based on the role played by the proteases ACE2, ADAM17, TMPRSS2 and AR in SARS-CoV-2 infection, which were deduced based on various studies. We have also summarized how these host proteins increase the pathology and the infective ability of SARS-CoV-2 and we posit that their inhibition may be a therapeutic option for preventing SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Vivek K Gupta
- Department of Biochemistry, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra-282004, India
| | - Madhan K Murthy
- Department of Immunology, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra-282004, India
| | - Shripad Patil
- Department of Immunology, ICMR-National JALMA Institute for Leprosy and Other Mycobacterial Diseases, Tajganj, Agra-282004, India
| |
Collapse
|
17
|
Moravvej H, Pourani MR, Baghani M, Abdollahimajd F. Androgenetic alopecia and COVID-19: A review of the hypothetical role of androgens. Dermatol Ther 2021; 34:e15004. [PMID: 34033224 PMCID: PMC8209856 DOI: 10.1111/dth.15004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/22/2021] [Indexed: 12/20/2022]
Abstract
The coronavirus disease 2019 (COVID‐19) has become the most emerging health issue globally. A prompt investigation regarding disease management and treatment is crucial for decreasing the burden of the disease. Many explorations and hypotheses have been posed, but the definite treatment has not been determined for COVID‐19. Recent studies described a substantial prevalence of COVID‐19 and also a higher rate of morbidity and mortality in men afflicted with severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection. The gender‐related discordance in COVID‐19 infection may be due to hormonal differences, socioeconomic factors, genetic susceptibility, gender‐related comorbidities, and habits like alcohol consumption. On the other hand, several studies proposed that androgens could improve the immune system and have a protective role in COVID‐19, and decreased levels of androgens might be associated with unsatisfactory outcomes. In the field of dermatology, androgenetic alopecia (AGA) is correlated with a hyperandrogenic state and may be related to COVID‐19 severity. Furthermore, recent research has assessed the plausible association of AGA and COVID‐19. In this review, we investigate all evidence on AGA and its relationship with COVID‐19, including the possible role of androgens in COVID‐19 severity and outcomes as well as candidate androgen‐related drugs for the treatment of COVID‐19.
Collapse
Affiliation(s)
- Hamideh Moravvej
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Pourani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Clinical Research Development Unit of Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Moein Baghani
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fahimeh Abdollahimajd
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Clinical Research Development Unit of Shohada-e Tajrish Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Yang Z, Gimple RC, Zhou N, Zhao L, Gustafsson JÅ, Zhou S. Targeting Nuclear Receptors for Cancer Therapy: Premises, Promises, and Challenges. Trends Cancer 2021; 7:541-556. [PMID: 33341430 DOI: 10.1016/j.trecan.2020.11.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/12/2022]
Abstract
Nuclear receptors are a family of transcription factors localized in cell nuclei, sensing specific ligands and fine-tuning a variety of cell physiological events. They have been intensively investigated in cancer biology. With their excellent properties of druggability and actionability, nuclear receptors have demonstrated much promise as novel therapeutic targets for different cancer types. Accumulating evidence has highlighted the essential roles of certain nuclear receptors in tumor immunology, suggesting the possibility for them to serve as cancer immunotherapeutic targets. Here, we summarize the roles of nuclear receptors in cancer biology and tumor immunology, and underscore the current advances of clinical trials for nuclear receptor-based cancer therapeutics.
Collapse
Affiliation(s)
- Zhengnan Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, CA, USA; Department of Pathology, Case Western Reserve University, Cleveland, OH, USA
| | - Nianxin Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Linjie Zhao
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, CA, USA.
| | - Jan-Åke Gustafsson
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX, USA; Center for Medical Innovation, Department of Biosciences and Nutrition at Novum, Karolinska Institute, Stockholm, Sweden.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China.
| |
Collapse
|
19
|
Peng Q, Wong CYP, Cheuk IWY, Teoh JYC, Chiu PKF, Ng CF. The Emerging Clinical Role of Spermine in Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094382. [PMID: 33922247 PMCID: PMC8122740 DOI: 10.3390/ijms22094382] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 01/31/2023] Open
Abstract
Spermine, a member of polyamines, exists in all organisms and is essential for normal cell growth and function. It is highly expressed in the prostate compared with other organs and is detectable in urine, tissue, expressed prostatic secretions, and erythrocyte. A significant reduction of spermine level was observed in prostate cancer (PCa) tissue compared with benign prostate tissue, and the level of urinary spermine was also significantly lower in men with PCa. Decreased spermine level may be used as an indicator of malignant phenotype transformation from normal to malignant tissue in prostate. Studies targeting polyamines and key rate-limiting enzymes associated with spermine metabolism as a tool for PCa therapy and chemoprevention have been conducted with various polyamine biosynthesis inhibitors and polyamine analogues. The mechanism between spermine and PCa development are possibly related to the regulation of polyamine metabolism, cancer-driving pathways, oxidative stress, anticancer immunosurveillance, and apoptosis regulation. Although the specific mechanism of spermine in PCa development is still unclear, ongoing research in spermine metabolism and its association with PCa pathophysiology opens up new opportunities in the diagnostic and therapeutic roles of spermine in PCa management.
Collapse
Affiliation(s)
| | | | | | | | | | - Chi-Fai Ng
- Correspondence: (P.K.-F.C.); (C.-F.N.); Tel.: +85-235-052-625 (C.-F.N.)
| |
Collapse
|
20
|
Abbasi AZ, Kiyani DA, Hamid SM, Saalim M, Fahim A, Jalal N. Spiking dependence of SARS-CoV-2 pathogenicity on TMPRSS2. J Med Virol 2021; 93:4205-4218. [PMID: 33638460 PMCID: PMC8014076 DOI: 10.1002/jmv.26911] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/15/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022]
Abstract
Epidemiological data shows a discrepancy in COVID‐19 susceptibility and outcomes with some regions being more heavily affected than others. However, the factors that determine host susceptibility and pathogenicity remain elusive. An increasing number of publications highlight the role of Transmembrane Serine Protease 2 (TMPRSS2) in the susceptibility of the host cell to SARS‐CoV‐2. Cleavage of viral spike protein via the host cell's TMPRSS2 enzyme activity mediates viral entry into the host cell. The enzyme synthesis is regulated by the TMPRSS2 gene, which has also been implicated in the entry mechanisms of previously reported Coronavirus infections. In this review, we have investigated the pathogenicity of SARS‐CoV‐2 and disease susceptibility dependence on the TMPRSS2 gene as expressed in various population groups. We further discuss how the differential expression of this gene in various ethnic groups can affect the SARS‐CoV‐2 infection and Coronavirus disease (COVID)‐19 outcomes. Moreover, promising new TMPRSS2 protease blockers and inhibitors are discussed for COVID‐19 treatment. 1. Entry of SARS‐CoV‐2 into a host cell depends on host protease TMPRSS2. 2. TMPRSS2 gene has localized expression throughout the human body but highly expressed in cells of the respiratory tract (primary target of SARS‐CoV‐2 in humans), gastrointestinal tract, kidneys and prostate. 3. Differences in expression of TMPRSS2 gene in the respiratory among different population groups can be a basis for discrepancy observed in COVID‐19 susceptibility and disease outcomes. 4. Drugs based on the inhibition or blockage of TMPRSS2 protease are undergoing clinical trials as a therapeutic option.
Collapse
Affiliation(s)
| | | | | | - Muhammad Saalim
- PsiMega2 (Pvt.) Ltd., Islamabad, Pakistan.,School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Ammad Fahim
- National University of Medical Sciences, Rawalpindi, Punjab, Pakistan
| | - Nasir Jalal
- PsiMega2 (Pvt.) Ltd., Islamabad, Pakistan.,Nanjing University of Information Science and Technology, Nanjing, Jiangsu Province, China
| |
Collapse
|
21
|
Mukherjee TK, Malik P, Hoidal JR. The emerging role of estrogen related receptorα in complications of non-small cell lung cancers. Oncol Lett 2021; 21:258. [PMID: 33664821 PMCID: PMC7882887 DOI: 10.3892/ol.2021.12519] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/18/2020] [Indexed: 12/20/2022] Open
Abstract
Approximately 85% of lung cancer cases are recognized as non-small cell lung cancer (NSCLC) with a perilous (13–17%) 5-year survival in Europe and the USA. Although tobacco smoking has consistently emerged as the leading cause of NSCLC complications, its consequences are distinctly manifest with respect to sex bias, due to differential gene and sex hormone expression. Estrogen related receptor α (ERRα), a member of the nuclear orphan receptor superfamily is normally expressed in the lungs, and activates various nuclear genes without binding to the ligands, such as estrogens. In NSCLC ERRα expression is significantly higher compared with healthy individuals. It is well established ERα and ERβ‚ have 93% and 60% identity in the DNA and ligand binding domains, respectively. ERα and ERRα have 69% (70% with ERRα-1) and 34% (35% with ERRα-1) identity, respectively; ERRα and ERRβ‚ have 92 and 61% identity, respectively. However, whether there is distinctive ERRα interaction with mammalian estrogens or concurrent involvement in non-ER signalling pathway activation is not known. Relevant to NSCLC, ERRα promotes proliferation, invasion and migration by silencing the tumor suppressor proteins p53 and pRB, and accelerates G2-M transition during cell division. Epithelial to mesenchymal transition (EMT) and activation of Slug (an EMT associated transcription factor) are the prominent mechanisms by which ERRα activates NSCLC metastasis. Based on these observations, the present article focuses on the feasibility of antiERRα therapy alone and in combination with antiER as a therapeutic strategy for NSCLC complications.
Collapse
Affiliation(s)
- Tapan K Mukherjee
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT 84132, USA.,Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA.,George E. Wahlen Department of Veterans Affairs Medical Centre, Salt Lake City, UT 84132, USA
| | - Parth Malik
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar, Gujarat 382030, India
| | - John R Hoidal
- Division of Respiratory, Critical Care and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT 84132, USA.,Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA.,George E. Wahlen Department of Veterans Affairs Medical Centre, Salt Lake City, UT 84132, USA
| |
Collapse
|
22
|
Ganji R, Reddy PH. Impact of COVID-19 on Mitochondrial-Based Immunity in Aging and Age-Related Diseases. Front Aging Neurosci 2021; 12:614650. [PMID: 33510633 PMCID: PMC7835331 DOI: 10.3389/fnagi.2020.614650] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) has become a deadly pandemic with surging mortality rates and no cure. COVID-19 is caused by the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) with a range of clinical symptoms, including cough, fever, chills, headache, shortness of breath, difficulty breathing, muscle pain, and a loss of smell or taste. Aged individuals with compromised immunity are highly susceptible to COVID-19 and the likelihood of mortality increases with age and the presence of comorbidities such as hypertension, diabetes mellitus, cardiovascular disease, or chronic obstructive pulmonary disease. Emerging evidence suggests that COVID-19 highjacks mitochondria of immune cells, replicates within mitochondrial structures, and impairs mitochondrial dynamics leading to cell death. Mitochondria are the powerhouses of the cell and are largely involved in maintaining cell immunity, homeostasis, and cell survival/death. Increasing evidence suggests that mitochondria from COVID-19 infected cells are highly vulnerable, and vulnerability increases with age. The purpose of our article is to summarize the role of various age-related comorbidities such as diabetes, obesity, and neurological diseases in increasing mortality rates amongst the elderly with COVID-19. Our article also highlights the interaction between coronavirus and mitochondrial dynamics in immune cells. We also highlight the current treatments, lifestyles, and safety measures that can help protect against COVID-19. Further research is urgently needed to understand the molecular mechanisms between the mitochondrial virus and disease progression in COVID-19 patients.
Collapse
Affiliation(s)
- Riya Ganji
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Departments of Neuroscience and Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
23
|
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: 5.3] [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
|
24
|
Wang S, Huo X. Comprehensive Analysis of ESRRA in Endometrial Cancer. Technol Cancer Res Treat 2021; 20:1533033821992083. [PMID: 33525981 PMCID: PMC7871350 DOI: 10.1177/1533033821992083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/30/2020] [Accepted: 01/14/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Estrogen-related receptor alpha (ESRRA) was reported to play an important role in multiple biological processes of neoplastic diseases. The roles of ESRRA in endometrial cancer have not been fully investigated yet. METHODS Expression data and clinicopathological data of patients with uteri corpus endometrial carcinoma (UCEC) were obtained from The Cancer Genome Atlas (TCGA). Comprehensive bioinformatics analysis was performed, including receiver operating characteristics (ROC) curve analysis, Kaplan-Meier survival analysis, gene ontology (GO) enrichment analysis, and Gene Set Enrichment Analysis (GSEA). Immunohistochemistry was used to detect the protein expression level of ESRRA and CCK-8 assay was performed to evaluate the effect of ESRRA on the proliferation ability. RESULTS A total of 552 UCEC tissues and 35 normal tissues were obtained from the TCGA database. The mRNA and protein expression level of ESRRA was highly elevated in UCEC compared with normal tissues, and was closely associated with poor prognosis. ROC analysis indicated a very high diagnostic value of ESRRA for patients with UCEC. GO and GSEA functional analysis showed that ESRRA might be mainly involved in cellular metabolism processes, in turn, tumorigenesis and progression of UCEC. Knockdown of ESRRA inhibited the proliferation of UCEC cells in vitro. Further immune cell infiltration demonstrated that ESRRA enhanced the infiltration level of neutrophil cell and reduced that of T cell (CD4+ naïve), NK cell, and cancer associated fibroblast (CAF). The alteration of immune microenvironment will greatly help in developing immune checkpoint therapy for UCEC. CONCLUSIONS Our study comprehensively analyzed the expression level, clinical value, and possible mechanisms of action of ESRRA in UCEC. These findings showed that ESRRA might be a potential diagnostic and therapeutic target.
Collapse
Affiliation(s)
- Shufang Wang
- Department of Obstetrics and Gynecology, Maternal and Child Health
Care Hospital of Qinhuangdao, Qinhuangdao, China
| | - Xinlong Huo
- Department of Oncology, the First Hospital of Qinhuangdao City,
Qinhuangdao, China
| |
Collapse
|
25
|
Moradi F, Enjezab B, Ghadiri-Anari A. The role of androgens in COVID-19. Diabetes Metab Syndr 2020; 14:2003-2006. [PMID: 33091758 PMCID: PMC7557269 DOI: 10.1016/j.dsx.2020.10.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/11/2020] [Accepted: 10/13/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND AND AIM The coronavirus disease 2019 (COVID-19) pandemic is a global health emergency. According to the findings, male patients with COVID-19 infection are at an increased risk for severe complications than females. The causes of this issue are unknown and are most probably multifactorial. Sexual hormones affect the immune system, so estrogen strengthens the immune system, and testosterone suppresses it. Due to the reports of the high prevalence of androgenic alopecia in hospitalized patients with COVID-19 and a higher risk of respiratory disease and increased use of allergy/asthma medications among patients with polycystic ovary syndrome (PCOS) as a hyperandrogenism condition compared with non-PCOS women, this review aimed to evaluate androgens role in COVID-19. METHODS 42 related articles from 2008 to 2020 were reviewed with the keywords of androgens, hormonal factors, and hair loss in combination with COVID-19 in medical research databases. RESULTS The evidence of transmembrane protease, serine 2 (TMPRSS2) expression in lung tissue, which is an androgen-regulated gene and expressed mainly in the adult prostate may interpret the increased susceptibility of the male gender to severe COVID-19 complications. Moreover, angiotensin-converting enzyme 2 (ACE-2) acts as a functional receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and male hormones are effective in the ACE-2 passageway and simplify SARS-CoV-2 entry into host cells. CONCLUSION Further studies on the severity of symptoms in patients with COVID-19 in other hyperandrogenism conditions compared to the control group are recommended.
Collapse
Affiliation(s)
| | - Behnaz Enjezab
- Research Center for Nursing and Midwifery Care, Department of Midwifery, Faculty of Nursing and Midwifery, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| | - Akram Ghadiri-Anari
- Department of Internal Medicine, Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
26
|
Crevet L, Vanacker JM. Regulation of the expression of the estrogen related receptors (ERRs). Cell Mol Life Sci 2020; 77:4573-4579. [PMID: 32448995 PMCID: PMC11104921 DOI: 10.1007/s00018-020-03549-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 10/23/2019] [Accepted: 05/13/2020] [Indexed: 10/24/2022]
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 tissues and cells and they display various physiological and pathological functions, controlling, amongst others and depending on the receptor, bone homeostasis, energy metabolism, embryonic stem cell pluripotency, and cancer progression. In contrast to classical nuclear receptors, the activities of the ERRs are not controlled by a natural ligand. Regulation of their activities thus rely on other means such as post-translational modification or availability of transcriptional co-regulators. In addition, regulation of their mere expression under given physiological or pathological conditions is a particularly important level of control. Here we discuss the mechanisms involved in the regulation of ERRs expression and the reported means to impact on it using pharmacological approaches.
Collapse
Affiliation(s)
- Lucile Crevet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France
- Laboratory of Stem Cells and Cancer, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS UMR5242, Ecole Normale Supérieure de Lyon, 32-34 Avenue Tony Garnier, 69007, Lyon, France.
| |
Collapse
|
27
|
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: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
28
|
SARS-CoV-2 and cancer: Are they really partners in crime? Cancer Treat Rev 2020; 89:102068. [PMID: 32731090 PMCID: PMC7351667 DOI: 10.1016/j.ctrv.2020.102068] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
The outbreak of the SARS-CoV-2 pandemic has overwhelmed health care systems in many countries. The clinical presentation of the SARS-CoV-2 varies between a subclinical or flu-like syndrome to that of severe pneumonia with multi-organ failure and death. Initial reports have suggested that cancer patients may have a higher susceptibility to get infected by the SARS-CoV-2 virus but current evidence remains poor as it is biased by important confounders. Patients with ongoing or recent cancer treatment for advanced active disease, metastatic solid tumors and hematological malignancies are at higher risk of developing severe COVID-19 respiratory disease that requires hospitalization and have a poorer disease outcome compared to individuals without cancer. However it is not clear whether these are independent risk factors, or mainly driven by male gender, age, obesity, performance status, uncontrolled diabetes, cardiovascular disease and various other medical conditions. These often have a greater influence on the probability to die due to SARS-CoV-2 then cancer. Delayed diagnosis and suboptimal cancer management due to the pandemic results in disease upstaging and has considerable impact cancer on specific death rates. Surgery during the peak of the pandemic seems to increase mortality, but there is no convincing evidence that adjuvant systemic cancer therapy and radiotherapy are contraindicated, implicating that cancer treatment can be provided safely after individual risk/benefit assessment and some adaptive measures. Underlying immunosuppression, elevated cytokine levels, altered expression of the angiotensin converting enzyme (ACE-2) and TMPRSS2, and a prothrombotic status may fuel the effects of a SARS-CoV-2 in some cancer patients, but have the potential to be used as biomarkers for severe disease and therapeutic targets. The rapidly expanding literature on COVID-19 should be interpreted with care as it is often hampered by methodological and statistical flaws.
Collapse
|
29
|
De Toni L, Garolla A, Di Nisio A, Rocca MS, Foresta C. Caution in the management of SARS-CoV-2 infection in males. Andrology 2020; 9:27-29. [PMID: 32452644 PMCID: PMC7283787 DOI: 10.1111/andr.12829] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 01/08/2023]
Abstract
The coronavirus 2 (SARS‐CoV‐2) pandemic carries clinical, economic, and social burdens that are currently being disclosed. The key steps of virus life cycle have been recently clarified, highlighting the role of host type 2 angiotensin‐converting enzyme (ACE2) and TMPRSS2 serine protease in virus‐cell binding and entry, respectively. Importantly, major concerns derive from the androgen‐dependent tissue‐expression of both TMPRSS2 and ACE2, suggesting a differential clinical course of the infection between genders. In agreement with this model, available epidemiological data show that the disease in males has an higher risk to display an heavier pattern and associates with both an increased access to critical care unit and higher mortality rate. In this opinion article, available evidence linking the androgen activity with the gender differences observed in SARS‐CoV‐2 infection are discussed, hypothesizing possible therapeutic approaches in male based on the disruption of androgen signaling. On these bases, gender‐specific recommendations for the management of male patients affected by SARS‐CoV‐2 infection are warmly suggested, in order to improve the clinical course of the disease.
Collapse
Affiliation(s)
- Luca De Toni
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Andrea Garolla
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Andrea Di Nisio
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Maria Santa Rocca
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| | - Carlo Foresta
- Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padova, Padova, Italy
| |
Collapse
|
30
|
Effects of estrogen receptor signaling on prostate cancer carcinogenesis. Transl Res 2020; 222:56-66. [PMID: 32413498 DOI: 10.1016/j.trsl.2020.04.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/11/2020] [Accepted: 04/07/2020] [Indexed: 12/25/2022]
Abstract
Management of advanced prostate cancer remains complex, with substantial changes in treatment options emerging in recent years having implications for treatment selection and sequencing. Recognition of the importance of androgen signaling has led to life-prolonging treatments, as well as "liquid biopsy" techniques to guide these treatments in some settings. Therapies that target estrogen receptor signaling are efficacious but infrequently used options for treatment of castration-resistant prostate cancer. It is possible that nuances of estrogen receptor (ER) signaling, or selective modulation of ER signaling, might favorably influence outcomes in castration-resistant prostate cancer. Expression of ERs and their variants has been investigated in other cancers such as breast. Constitutively activating gene alterations can potentially lead to ER activation and subsequently promote cancer progression. The identification of these aberrations may help identify cancer phenotypes that are susceptible or resistant to therapies involved in ER signaling. This review outlines the current literature regarding ER signaling in prostate cancer, and provides background for exploration of potentially useful ER signaling biomarkers in advanced prostate cancer.
Collapse
|
31
|
Singh KK, Chaubey G, Chen JY, Suravajhala P. Decoding SARS-CoV-2 hijacking of host mitochondria in COVID-19 pathogenesis. Am J Physiol Cell Physiol 2020; 319:C258-C267. [PMID: 32510973 PMCID: PMC7381712 DOI: 10.1152/ajpcell.00224.2020] [Citation(s) in RCA: 223] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 12/21/2022]
Abstract
Because of the ongoing pandemic around the world, the mechanisms underlying the SARS-CoV-2-induced COVID-19 are subject to intense investigation. Based on available data for the SARS-CoV-1 virus, we suggest how CoV-2 localization of RNA transcripts in mitochondria hijacks the host cell's mitochondrial function to viral advantage. Besides viral RNA transcripts, RNA also localizes to mitochondria. SARS-CoV-2 may manipulate mitochondrial function indirectly, first by ACE2 regulation of mitochondrial function, and once it enters the host cell, open-reading frames (ORFs) such as ORF-9b can directly manipulate mitochondrial function to evade host cell immunity and facilitate virus replication and COVID-19 disease. Manipulations of host mitochondria by viral ORFs can release mitochondrial DNA (mtDNA) in the cytoplasm and activate mtDNA-induced inflammasome and suppress innate and adaptive immunity. We argue that a decline in ACE2 function in aged individuals, coupled with the age-associated decline in mitochondrial functions resulting in chronic metabolic disorders like diabetes or cancer, may make the host more vulnerable to infection and health complications to mortality. These observations suggest that distinct localization of viral RNA and proteins in mitochondria must play essential roles in SARS-CoV-2 pathogenesis. Understanding the mechanisms underlying virus communication with host mitochondria may provide critical insights into COVID-19 pathologies. An investigation into the SARS-CoV-2 hijacking of mitochondria should lead to novel approaches to prevent and treat COVID-19.
Collapse
Affiliation(s)
- Keshav K Singh
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, Kaul Genetics Building, Birmingham, Alabama
| | - Gyaneshwer Chaubey
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Jake Y Chen
- Department of Genetics, Computer Science, and Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Prashanth Suravajhala
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research Statue Circle, Jaipur, Rajasthan, India
| |
Collapse
|
32
|
Singh KK, Chaubey G, Chen JY, Suravajhala P. Decoding SARS-CoV-2 hijacking of host mitochondria in COVID-19 pathogenesis. Am J Physiol Cell Physiol 2020. [PMID: 32510973 DOI: 10.1152/ajpcell.00224.202048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Because of the ongoing pandemic around the world, the mechanisms underlying the SARS-CoV-2-induced COVID-19 are subject to intense investigation. Based on available data for the SARS-CoV-1 virus, we suggest how CoV-2 localization of RNA transcripts in mitochondria hijacks the host cell's mitochondrial function to viral advantage. Besides viral RNA transcripts, RNA also localizes to mitochondria. SARS-CoV-2 may manipulate mitochondrial function indirectly, first by ACE2 regulation of mitochondrial function, and once it enters the host cell, open-reading frames (ORFs) such as ORF-9b can directly manipulate mitochondrial function to evade host cell immunity and facilitate virus replication and COVID-19 disease. Manipulations of host mitochondria by viral ORFs can release mitochondrial DNA (mtDNA) in the cytoplasm and activate mtDNA-induced inflammasome and suppress innate and adaptive immunity. We argue that a decline in ACE2 function in aged individuals, coupled with the age-associated decline in mitochondrial functions resulting in chronic metabolic disorders like diabetes or cancer, may make the host more vulnerable to infection and health complications to mortality. These observations suggest that distinct localization of viral RNA and proteins in mitochondria must play essential roles in SARS-CoV-2 pathogenesis. Understanding the mechanisms underlying virus communication with host mitochondria may provide critical insights into COVID-19 pathologies. An investigation into the SARS-CoV-2 hijacking of mitochondria should lead to novel approaches to prevent and treat COVID-19.
Collapse
Affiliation(s)
- Keshav K Singh
- Department of Genetics, School of Medicine, University of Alabama at Birmingham, Kaul Genetics Building, Birmingham, Alabama
| | - Gyaneshwer Chaubey
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Jake Y Chen
- Department of Genetics, Computer Science, and Biomedical Engineering, University of Alabama at Birmingham, Birmingham, Alabama
| | - Prashanth Suravajhala
- Department of Biotechnology and Bioinformatics, Birla Institute of Scientific Research Statue Circle, Jaipur, Rajasthan, India
| |
Collapse
|
33
|
Montopoli M, Zumerle S, Vettor R, Rugge M, Zorzi M, Catapano CV, Carbone GM, Cavalli A, Pagano F, Ragazzi E, Prayer-Galetti T, Alimonti A. Androgen-deprivation therapies for prostate cancer and risk of infection by SARS-CoV-2: a population-based study (N = 4532). Ann Oncol 2020; 31:1040-1045. [PMID: 32387456 PMCID: PMC7202813 DOI: 10.1016/j.annonc.2020.04.479] [Citation(s) in RCA: 381] [Impact Index Per Article: 95.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 04/23/2020] [Accepted: 04/29/2020] [Indexed: 02/08/2023] Open
Abstract
Background Cell entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) depends on binding of the viral spike (S) proteins to angiotensin-converting enzyme 2 and on S protein priming by TMPRSS2. Inhibition of TMPRSS2 may work to block or decrease the severity of SARS-CoV-2 infections. Intriguingly, TMPRSS2 is an androgen-regulated gene that is up-regulated in prostate cancer where it supports tumor progression and is involved in a frequent genetic translocation with the ERG gene. First- or second-generation androgen-deprivation therapies (ADTs) decrease the levels of TMPRSS2. Here we put forward the hypothesis that ADTs may protect patients affected by prostate cancer from SARS-CoV-2 infections. Materials and methods We extracted data regarding 9280 patients (4532 males) with laboratory-confirmed SARS-CoV-2 infection from 68 hospitals in Veneto, one of the Italian regions that was most affected by the coronavirus disease 2019 (COVID-19) pandemic. The parameters used for each COVID-19-positive patient were sex, hospitalization, admission to intensive care unit, death, tumor diagnosis, prostate cancer diagnosis, and ADT. Results There were evaluable 9280 SARS-CoV-2-positive patients in Veneto on 1 April 2020. Overall, males developed more severe complications, were more frequently hospitalized, and had a worse clinical outcome than females. Considering only the Veneto male population (2.4 million men), 0.2% and 0.3% of non-cancer and cancer patients, respectively, tested positive for SARS-CoV-2. Comparing the total number of SARS-CoV-2-positive cases, prostate cancer patients receiving ADT had a significantly lower risk of SARS-CoV-2 infection compared with patients who did not receive ADT (OR 4.05; 95% CI 1.55–10.59). A greater difference was found comparing prostate cancer patients receiving ADT with patients with any other type of cancer (OR 4.86; 95% CI 1.88–12.56). Conclusion Our data suggest that cancer patients have an increased risk of SARS-CoV-2 infections compared with non-cancer patients. However, prostate cancer patients receiving ADT appear to be partially protected from SARS-CoV-2 infections. SARS-CoV-2-infected men have a worse clinical outcome than women. Cancer patients have an increased risk of SARS-CoV-2 infection. Prostate cancer patients receiving androgen-deprivation therapies appear to be partially protected from the infection.
Collapse
Affiliation(s)
- M Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Padova, Italy; VIMM - Veneto Institute of Molecular Medicine, Fondazione per la Ricerca Biomedica Avanzata, Padova, Italy
| | - S Zumerle
- VIMM - Veneto Institute of Molecular Medicine, Fondazione per la Ricerca Biomedica Avanzata, Padova, Italy; Department of Medicine, Università degli Studi di Padova, Padova, Italy
| | - R Vettor
- Department of Medicine, Università degli Studi di Padova, Padova, Italy
| | - M Rugge
- Department of Medicine, Università degli Studi di Padova, Padova, Italy; Veneto Tumour Registry - Azienda Zero, Padova, Italy
| | - M Zorzi
- Veneto Tumour Registry - Azienda Zero, Padova, Italy
| | - C V Catapano
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - G M Carbone
- Institute of Oncology Research, Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - A Cavalli
- Institute for Research in Biomedicine, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - F Pagano
- VIMM - Veneto Institute of Molecular Medicine, Fondazione per la Ricerca Biomedica Avanzata, Padova, Italy
| | - E Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, Università degli Studi di Padova, Padova, Italy
| | - T Prayer-Galetti
- Department of Oncological and Gastroenterological Sciences - Urology Unit, Azienda Ospedaliera di Padova, Padova, Italy
| | - A Alimonti
- VIMM - Veneto Institute of Molecular Medicine, Fondazione per la Ricerca Biomedica Avanzata, Padova, Italy; Department of Medicine, Università degli Studi di Padova, Padova, Italy; Institute of Oncology Research, Oncology Institute of Southern Switzerland, Università della Svizzera Italiana, Bellinzona, Switzerland; Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland.
| |
Collapse
|
34
|
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: 2.8] [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
|
35
|
Chen H, Xing Y, Xie J, Xie J, Xing D, Tang J, Yang F, Yi Z, Qiu WW. Synthesis and biological evaluation of 3-nitro-4-chromanone derivatives as potential antiproliferative agents for castration-resistant prostate cancer. RSC Adv 2019; 9:33794-33799. [PMID: 35528914 PMCID: PMC9073653 DOI: 10.1039/c9ra06420f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 10/10/2019] [Indexed: 01/16/2023] Open
Abstract
A series of novel 3-nitro-4-chromanones were synthesized and their in vitro cytotoxicity was evaluated on castration-resistant prostate cancer cell (CRPC) lines using the sulforhodamine B (SRB) assay. The amide derivatives showed more potent antitumor activity than their corresponding ester derivatives. Most of the tested compounds showed less toxicity towards human fibroblasts (HAF) compared with the tumor cell lines. The optimal compound 36 possessed much more potent antiproliferative activity than the positive compound cisplatin. The colony formation, cell cycle distribution, apoptosis, transwell migration and wound healing assays of 36 were performed on CRPC cell lines.
Collapse
Affiliation(s)
- Huiqing Chen
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200062 China
| | - Yajing Xing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Jia Xie
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Jiuqing Xie
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Dong Xing
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200062 China
| | - Jie Tang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200062 China
| | - Fan Yang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200062 China
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University Shanghai 200241 China
| | - Wen-Wei Qiu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University Shanghai 200062 China
| |
Collapse
|
36
|
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: 2.0] [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
|
37
|
Marín‐Aguilera M, Reig Ò, Milà‐Guasch M, Font A, Domènech M, Rodríguez‐Vida A, Carles J, Suárez C, Alba AG, Jiménez N, Victoria I, Sala‐González N, Ribal MJ, López S, Etxaniz O, Anguera G, Maroto P, Fernández PL, Prat A, Mellado B. The influence of treatment sequence in the prognostic value of
TMPRSS2‐ERG
as biomarker of taxane resistance in castration‐resistant prostate cancer. Int J Cancer 2019; 145:1970-1981. [DOI: 10.1002/ijc.32238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/22/2019] [Accepted: 02/07/2019] [Indexed: 11/06/2022]
Affiliation(s)
- Mercedes Marín‐Aguilera
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Òscar Reig
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Maria Milà‐Guasch
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Albert Font
- Department of Medical OncologyInstitut Català d'Oncologia Badalona Spain
| | | | | | - Joan Carles
- Department of Medical OncologyVall d'Hebron Institute of Oncology. Vall d'Hebron University Hospital Barcelona Spain
| | - Cristina Suárez
- Department of Medical OncologyVall d'Hebron Institute of Oncology. Vall d'Hebron University Hospital Barcelona Spain
| | - Aránzazu González Alba
- Department of Medical OncologyHospital Universitario Puerta de Hierro Majadahonda Madrid Spain
| | - Natalia Jiménez
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Iván Victoria
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | | | - Maria José Ribal
- Department of UrologyHospital Clinic Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
| | - Sandra López
- Department of Medical OncologyHospital Clínic Barcelona Spain
| | - Olatz Etxaniz
- Department of Medical OncologyInstitut Català d'Oncologia Badalona Spain
| | - Geòrgia Anguera
- Department of Medical OncologyHospital de la Santa Cruz y San Pablo Barcelona Spain
| | - Pablo Maroto
- Department of Medical OncologyHospital de la Santa Cruz y San Pablo Barcelona Spain
| | - Pedro Luis Fernández
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
- Department of PathologyHospital Clínic Barcelona Spain
| | - Aleix Prat
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
| | - Begoña Mellado
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryInstitut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) Barcelona Spain
- Translational Genomics and Targeted Therapeutics in Solid Tumors LaboratoryFundació Clínic per a la Recerca Biomèdica Barcelona Spain
- Department of Medical OncologyHospital Clínic Barcelona Spain
- Faculty of MedicineUniversity of Barcelona Barcelona Spain
| |
Collapse
|