1
|
He Y, Shen M, Wang X, Yin A, Liu B, Zhu J, Zhang Z. Suppression of Interferon Response and Antiviral Strategies of Bunyaviruses. Trop Med Infect Dis 2024; 9:205. [PMID: 39330894 PMCID: PMC11435552 DOI: 10.3390/tropicalmed9090205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
The order Bunyavirales belongs to the class of Ellioviricetes and is classified into fourteen families. Some species of the order Bunyavirales pose potential threats to human health. The continuously increasing research reveals that various viruses within this order achieve immune evasion in the host through suppressing interferon (IFN) response. As the types and nodes of the interferon response pathway are continually updated or enriched, the IFN suppression mechanisms and target points of different virus species within this order are also constantly enriched and exhibit variations. For instance, Puumala virus (PUUV) and Tula virus (TULV) can inhibit IFN response through their functional NSs inhibiting downstream factor IRF3 activity. Nevertheless, the IFN suppression mechanisms of Dabie bandavirus (DBV) and Guertu virus (GTV) are mostly mediated by viral inclusion bodies (IBs) or filamentous structures (FSs). Currently, there are no effective drugs against several viruses belonging to this order that pose significant threats to society and human health. While the discovery, development, and application of antiviral drugs constitute a lengthy process, our focus on key targets in the IFN response suppression process of the virus leads to potential antiviral strategies, which provide references for both basic research and practical applications.
Collapse
Affiliation(s)
- Yingying He
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Department of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Min Shen
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Department of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiaohe Wang
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Department of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Anqi Yin
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
- Department of Clinical Medicine, Anhui Medical University, Hefei 230032, China
| | - Bingyan Liu
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jie Zhu
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Zhenhua Zhang
- Institute of Clinical Virology, Department of Infectious Diseases, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| |
Collapse
|
2
|
Targeting Estrogens and Various Estrogen-Related Receptors against Non-Small Cell Lung Cancers: A Perspective. Cancers (Basel) 2021; 14:cancers14010080. [PMID: 35008242 PMCID: PMC8750572 DOI: 10.3390/cancers14010080] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Non-small cell lung cancers (NSCLCs) account for ~85% of lung cancer cases worldwide. Mammalian lungs are exposed to both endogenous and exogenous estrogens. The expression of estrogen receptors (ERs) in lung cancer cells has evoked the necessity to evaluate the role of estrogens in the disease progression. Estrogens, specifically 17β-estradiol, promote maturation of several tissue types including lungs. Recent epidemiologic data indicate that women have a higher risk of lung adenocarcinoma, a type of NSCLC, when compared to men, independent of smoking status. Besides ERs, pulmonary tissues both in healthy physiology and in NSCLCs also express G-protein-coupled ERs (GPERs), epidermal growth factor receptor (EGFRs), estrogen-related receptors (ERRs) and orphan nuclear receptors. Premenopausal females between the ages of 15 and 50 years synthesize a large contingent of estrogens and are at a greater risk of developing NSCLCs. Estrogen-ER/GPER/EGFR/ERR-mediated activation of various cell signaling molecules regulates NSCLC cell proliferation, survival and apoptosis. This article sheds light on the most recent achievements in the elucidation of sequential biochemical events in estrogen-activated cell signaling pathways involved in NSCLC severity with insight into the mechanism of regulation by ERs/GPERs/EGFRs/ERRs. It further discusses the success of anti-estrogen therapies against NSCLCs.
Collapse
|
3
|
Cholesterol-Induced Metabolic Reprogramming in Breast Cancer Cells Is Mediated via the ERRα Pathway. Cancers (Basel) 2021; 13:cancers13112605. [PMID: 34073320 PMCID: PMC8198778 DOI: 10.3390/cancers13112605] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/08/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary There is increasing evidence that obesity and high circulating cholesterol levels are associated with an increased risk of recurrence and a higher mortality rate in breast cancer patients via altering the metabolic programming in breast cancer cells. However, the underlying molecular mechanism by which high cholesterol levels reprogram the metabolic pathways in breast cancer cells is not well-understood. We have previously demonstrated that cholesterol acts as an endogenous agonist of estrogen-related receptor α (ERRα), a strong regulator of cellular metabolism. The aim of the current study is to demonstrate whether cholesterol/obesity mediates its pathogenic effect in breast cancer cells via altering metabolic pathways in an ERRα-dependent manner. The findings of this study provide mechanistic insights into the link between cholesterol/obesity and metabolic reprogramming in breast cancer patients and reveal the metabolic vulnerabilities in such breast cancer patients that could be therapeutically targeted. Abstract The molecular mechanism underlying the metabolic reprogramming associated with obesity and high blood cholesterol levels is poorly understood. We previously reported that cholesterol is an endogenous ligand of the estrogen-related receptor alpha (ERRα). Using functional assays, metabolomics, and genomics, here we show that exogenous cholesterol alters the metabolic pathways in estrogen receptor-positive (ER+) and triple-negative breast cancer (TNBC) cells, and that this involves increased oxidative phosphorylation (OXPHOS) and TCA cycle intermediate levels. In addition, cholesterol augments aerobic glycolysis in TNBC cells although it remains unaltered in ER+ cells. Interestingly, cholesterol does not alter the metabolite levels of glutaminolysis, one-carbon metabolism, or the pentose phosphate pathway, but increases the NADPH levels and cellular proliferation, in both cell types. Importantly, we show that the above cholesterol-induced modulations of the metabolic pathways in breast cancer cells are mediated via ERRα. Furthermore, analysis of the ERRα metabolic gene signature of basal-like breast tumours of overweight/obese versus lean patients, using the GEO database, shows that obesity may modulate ERRα gene signature in a manner consistent with our in vitro findings with exogenous cholesterol. Given the close link between high cholesterol levels and obesity, our findings provide a mechanistic explanation for the association between cholesterol/obesity and metabolic reprogramming in breast cancer patients.
Collapse
|
4
|
Huang X, Ruan G, Sun P. Estrogen-related receptor alpha copy number variation is associated with ovarian cancer histological grade. J Obstet Gynaecol Res 2021; 47:1878-1883. [PMID: 33751740 DOI: 10.1111/jog.14741] [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: 09/12/2020] [Revised: 01/27/2021] [Accepted: 02/20/2021] [Indexed: 11/27/2022]
Abstract
AIM Copy number variations (CNVs) are related to the genetic and phenotypic diversity of cancers and identifying genetic alterations could improve treatment strategies. Here, we used The Cancer Genome Atlas (TCGA) to explore associations between estrogen-related receptor alpha (ESRRA) CNVs and histological grade in patients with ovarian cancer (OC). METHODS Gene expression data and clinical information of 620 OC patients were obtained from The Cancer Genome Atlas)TCGA and associations between ESRRA CNVs and clinical characteristics were evaluated. Multivariate logistic regression analyses to obtain odds ratios (ORs) using a 95% confidence interval (CI) were performed, adjusting for race, age, histological grade, and tumor size. RESULTS ESRRA CNVs were associated with histological grade (OR 0.6235 [95% CI, 0.3593-0.8877]; p < 0.05) and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) CNVs (OR -0.6298 [95% CI, -0.9011 to -0.3585]; p < 0.05). In multivariate analyses, ESRRA CNVs remained significantly associated with histological grade (OR 0.6492 [95% CI, 0.3549-0.9435]; p < 0.05) and PPARGC1A CNVs (OR -0.6236 [95% CI, -0.9269 to 0.3203]; p < 0.05). CONCLUSION There was a significant association between ESRRA CNVs in patients with OC and histological grade of the cancer.
Collapse
Affiliation(s)
- Xiqi Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Guanyu Ruan
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
5
|
Theodoris CV, Zhou P, Liu L, Zhang Y, Nishino T, Huang Y, Kostina A, Ranade SS, Gifford CA, Uspenskiy V, Malashicheva A, Ding S, Srivastava D. Network-based screen in iPSC-derived cells reveals therapeutic candidate for heart valve disease. Science 2021; 371:eabd0724. [PMID: 33303684 PMCID: PMC7880903 DOI: 10.1126/science.abd0724] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022]
Abstract
Mapping the gene-regulatory networks dysregulated in human disease would allow the design of network-correcting therapies that treat the core disease mechanism. However, small molecules are traditionally screened for their effects on one to several outputs at most, biasing discovery and limiting the likelihood of true disease-modifying drug candidates. Here, we developed a machine-learning approach to identify small molecules that broadly correct gene networks dysregulated in a human induced pluripotent stem cell (iPSC) disease model of a common form of heart disease involving the aortic valve (AV). Gene network correction by the most efficacious therapeutic candidate, XCT790, generalized to patient-derived primary AV cells and was sufficient to prevent and treat AV disease in vivo in a mouse model. This strategy, made feasible by human iPSC technology, network analysis, and machine learning, may represent an effective path for drug discovery.
Collapse
Affiliation(s)
- Christina V Theodoris
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
- Program in Developmental and Stem Cell Biology (DSCB), University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Ping Zhou
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
| | - Lei Liu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
| | - Yu Zhang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
| | - Tomohiro Nishino
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
| | - Yu Huang
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
| | - Aleksandra Kostina
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
| | - Sanjeev S Ranade
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
| | - Casey A Gifford
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
| | | | - Anna Malashicheva
- Institute of Cytology, Russian Academy of Sciences, Saint Petersburg, Russia
- Almazov Federal Medical Research Centre, Saint Petersburg, Russia
- Saint Petersburg State University, Saint Petersburg, Russia
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, UCSF, San Francisco, CA, USA
| | - Deepak Srivastava
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, USA.
- Roddenberry Stem Cell Center, Gladstone Institutes, San Francisco, CA, USA
- Department of Pediatrics, Department of Biochemistry and Biophysics, UCSF, San Francisco, CA, USA
| |
Collapse
|
6
|
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: 2.5] [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
|
7
|
Carrasco-Carballo A, Guadalupe Hernández-Linares M, Cárdenas-García M, Sandoval-Ramírez J. Synthesis and biological in vitro evaluation of the effect of hydroxyimino steroidal derivatives on breast cancer cells. Steroids 2021; 166:108787. [PMID: 33383064 DOI: 10.1016/j.steroids.2020.108787] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/15/2020] [Accepted: 12/21/2020] [Indexed: 01/29/2023]
Abstract
Breast cancer is the most common cause of cancer death in women, according to Global Cancer Observatory. This fact forces scientists to continue in the search for effective treatments against this aggressive type of cancer. Breast cancer frequently metastasizes to other organs, most often the bones, lungs, and liver. Breast cancer is normally associated with estrogen and progestogen levels and can be hormone or non-hormone dependent. In current experiments herein reported, some hydroxyimino spirostan derivatives showed great potential against MCF-7 breast cancer, a Luminal-A cancer. On the other hand, a set of synthesized 6-hydroxyimino-22-oxocholestane compounds had excellent activity against the MDA-MB-231 breast cancer cell line. The synthesis of hydroxyamino derivatives from spirostan and 22-oxocholestane compounds was improved. The hydroxyimino compounds enhanced the bioactivity when compared with their parent carbonyl skeletons.
Collapse
Affiliation(s)
- Alan Carrasco-Carballo
- Facultad de Ciencias Químicas, Laboratorio de Elucidación y Síntesis en Química Orgánica, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Mexico
| | - María Guadalupe Hernández-Linares
- Herbario y Jardín Botánico Universitario, Benemérita Universidad Autónoma de Puebla, Ciudad Universitaria, 72570 Puebla, Mexico; Centro de Química, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Mexico
| | - Maura Cárdenas-García
- Facultad de Medicina, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Mexico
| | - Jesús Sandoval-Ramírez
- Facultad de Ciencias Químicas, Laboratorio de Elucidación y Síntesis en Química Orgánica, Benemérita Universidad Autónoma de Puebla, 72570 Puebla, Mexico.
| |
Collapse
|
8
|
Huang X, Ruan G, Liu G, Gao Y, Sun P. Immunohistochemical Analysis of PGC-1α and ERRα Expression Reveals Their Clinical Significance in Human Ovarian Cancer. Onco Targets Ther 2020; 13:13055-13062. [PMID: 33376354 PMCID: PMC7764629 DOI: 10.2147/ott.s288332] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/03/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) and estrogen-related receptor alpha (ERRα) play a vital role in various human cancers. The purpose of this study was to investigate whether the PGC-1α/ERRα axis could serve as an effective prognostic marker in ovarian cancer (OC). Patients and Methods We investigated the expression of both PGC-1α and ERRα in 42 ovarian cancer and 31 noncancerous ovarian samples by immunohistochemistry (IHC). The relationship between the expression of PGC-1α and ERRα in OC and the clinical characteristics of patients was evaluated. In addition, data from the Human Protein Atlas (HPA) database were collected to validate the prognostic significance of PGC-1α and ERRα mRNA expression in OC. Results PGC-1α and ERRα showed notably higher expression in OC tissues than in noncancerous tissues (P=0.0059, P=0.002). Moreover, in patients with OC, high ERRα and PGC-1α/ERRα expression significantly correlated with tumor differentiation (P=0.027; P=0.04), lymph node status (P=0.023; P=0.021), CA125 (P=0.036; P=0.021), and HE4 (P=0.021; P=0.05), while high PGC-1α expression was only significantly associated with tumor differentiation (P=0.029). The combined analysis of high PGC-1α and ERRα expression revealed a tendency towards poor cancer-specific survival (P=0.1276). Conclusion PGC-1α and ERRα are overexpressed in OC and might be significant prognostic factors for this cancer.
Collapse
Affiliation(s)
- Xiqi Huang
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Guanyu Ruan
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Guifen Liu
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Yuqin Gao
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China.,Key Laboratory of Women and Children's Critical Diseases Research, Fujian Provincial Maternity and Children's Health Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| |
Collapse
|
9
|
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: 17] [Impact Index Per Article: 3.4] [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
|
10
|
Shahoei SH, Nelson ER. Nuclear receptors, cholesterol homeostasis and the immune system. J Steroid Biochem Mol Biol 2019; 191:105364. [PMID: 31002862 PMCID: PMC6589364 DOI: 10.1016/j.jsbmb.2019.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 04/09/2019] [Accepted: 04/10/2019] [Indexed: 12/30/2022]
Abstract
Cholesterol is essential for maintaining membrane fluidity in eukaryotes. Additionally, the synthetic cascade of cholesterol results in precursor molecules important for cellular function such as lipid raft formation and protein prenylation. As such, cholesterol homeostasis is tightly regulated. Interestingly, it is now known that some cholesterol precursors and many metabolites serve as active signaling molecules, binding to different classes of receptors including the nuclear receptors. Furthermore, many cholesterol metabolites or their nuclear receptors have been implicated in the regulation of the immune system in normal physiology and disease. Therefore, in this focused review, cholesterol homeostasis and nuclear receptors involved in this regulation will be discussed, with particular emphasis on how these cascades influence the immune system.
Collapse
Affiliation(s)
- Sayyed Hamed Shahoei
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States; University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, United States; Carl R. Woese Institute for Genomic Biology, Anticancer Discovery from Pets to People Theme, University of Illinois at Urbana Champaign, Urbana, IL, United States.
| |
Collapse
|
11
|
Therapeutic Efficiency of an External Chinese Herbal Formula of Mammary Precancerous Lesions by BATMAN-TCM Online Bioinformatics Analysis Tool and Experimental Validation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:2795010. [PMID: 30906412 PMCID: PMC6398062 DOI: 10.1155/2019/2795010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 12/09/2018] [Accepted: 12/31/2018] [Indexed: 01/08/2023]
Abstract
Ruyan Neixiao Cream (RYNXC), a patented Chinese herbal formula, was reported to have the effect of treating mammary precancerous disease. In this study, we predicted the potential targets, pathways, and diseases of the ingredients contained in each herbal of RYNXC and constructed an ingredients-targets-diseases network. Then, we analyzed molecular mechanisms of this Chinese herbal formula by MCF-10AT cells and model rats of breast precancerous lesions. BATMAN-TCM prediction showed that ESR1, PGR, PTGS2, EGFR, and Src were mRNA targets of RYNXC. Our results suggested that RYNXC transdermal fluid downregulated ESR1, PGR, PTGS2, EGFR, and Src expression at gene and protein level in MCF-10AT cells. In the rat breast precancerous lesions model, high and low dose RYNXC could also significantly reduce genes and proteins expression of ESR1, PGR, PTGS2, EGFR, and Src. Taken together these data indicate that RYNXC targets multiple molecules responsible for breast precancerous lesion and is an effective Chinese herbal formula. So RYNXC may be a promising external drug for breast precancerous lesions.
Collapse
|
12
|
Lynch C, Zhao J, Sakamuru S, Zhang L, Huang R, Witt KL, Merrick BA, Teng CT, Xia M. Identification of Compounds That Inhibit Estrogen-Related Receptor Alpha Signaling Using High-Throughput Screening Assays. Molecules 2019; 24:E841. [PMID: 30818834 PMCID: PMC6429183 DOI: 10.3390/molecules24050841] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/19/2019] [Accepted: 02/23/2019] [Indexed: 12/20/2022] Open
Abstract
The nuclear receptor, estrogen-related receptor alpha (ERRα; NR3B1), plays a pivotal role in energy homeostasis. Its expression fluctuates with the demands of energy production in various tissues. When paired with the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), the PGC/ERR pathway regulates a host of genes that participate in metabolic signaling networks and in mitochondrial oxidative respiration. Unregulated overexpression of ERRα is found in many cancer cells, implicating a role in cancer progression and other metabolism-related diseases. Using high throughput screening assays, we screened the Tox21 10K compound library in stably transfected HEK293 cells containing either the ERRα-reporter or the reporter plus PGC-1α expression plasmid. We identified two groups of antagonists that were potent inhibitors of ERRα activity and/or the PGC/ERR pathway: nine antineoplastic agents and thirteen pesticides. Results were confirmed using gene expression studies. These findings suggest a novel mechanism of action on bioenergetics for five of the nine antineoplastic drugs. Nine of the thirteen pesticides, which have not been investigated previously for ERRα disrupting activity, were classified as such. In conclusion, we demonstrated that high-throughput screening assays can be used to reveal new biological properties of therapeutic and environmental chemicals, broadening our understanding of their modes of action.
Collapse
Affiliation(s)
- Caitlin Lynch
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Jinghua Zhao
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Srilatha Sakamuru
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Li Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| | - Kristine L Witt
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA.
| | - B Alex Merrick
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA.
| | - Christina T Teng
- Division of the National Toxicology Program, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, NC 27709, USA.
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health (NIH), Bethesda, MD 20814, USA.
| |
Collapse
|
13
|
Liu G, Sun P, Dong B, Sehouli J. Key regulator of cellular metabolism, estrogen-related receptor α, a new therapeutic target in endocrine-related gynecological tumor. Cancer Manag Res 2018; 10:6887-6895. [PMID: 30588094 PMCID: PMC6296681 DOI: 10.2147/cmar.s182466] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The estrogen-related receptor α (ERRα), is an orphan transcription factor. Recently, many studies have reported its regulatory mechanisms and transcriptional targets after identification. Therefore, it may be eligible to join the rank of other nuclear receptors that control almost all aspects of cell metabolism. Cellular metabolism reprogramming plays a key role in fueling malignant change. The purpose of this review was to demonstrate that the ERRα plays an important role in the association between gynecological endocrine-related tumors and energy metabolism. Furthermore, regulation of ERRα may represent a promising strategy to induce cellular metabolic vulnerability of cancer from different origins. Thus, a comprehensive understanding of current treatment strategies may be achieved.
Collapse
Affiliation(s)
- GuiFen Liu
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - PengMing Sun
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China, .,Department of Gynaecology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - BinHua Dong
- Laboratory of Gynaecologic Oncology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, 350001 Fuzhou, Fujian, People's Republic of China,
| | - Jalid Sehouli
- Department of Gynaecologic Oncology and Gynaecology, Charité/Campus Virchow-Klinikum, European Competence Centre for Ovarian Cancer University of Berlin, Berlin 13353, Germany
| |
Collapse
|
14
|
Calahorra J, Martínez-Lara E, De Dios C, Siles E. Hypoxia modulates the antioxidant effect of hydroxytyrosol in MCF-7 breast cancer cells. PLoS One 2018; 13:e0203892. [PMID: 30235254 PMCID: PMC6147459 DOI: 10.1371/journal.pone.0203892] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/29/2018] [Indexed: 12/30/2022] Open
Abstract
Although cancer is multifactorial, a strong correlation between this pathology and increased oxidative stress has long been stablished. Hypoxia, inherent to solid tumors, increases reactive oxygen species and should be taken into account when analyzing the response of tumor cells to antioxidants. The Mediterranean diet has been related to a lower incidence of cancer, and particularly of breast cancer. Given that hydroxytyrosol (HT) is largely responsible for the antioxidant properties of olive oil, we have performed a comprehensive and comparative study of its effect on the oxidative stress response of the human breast cancer cell line MCF-7 in hypoxia and normoxia. Our results demonstrate that the antioxidant action of HT is particularly effective in a hypoxic environment. Moreover, we have observed that this polyphenol modulates the transcription and translation of members of the PGC-1α/ERRα and PGC-1α/Nrf2 pathways. However, while the transcriptional effects of HT are similar in normoxic and hypoxic conditions, its translational action is less prominent and partially attenuated in hypoxia, and therefore cannot completely explain the antioxidant effect of HT. Consequently, our results underscore that the hypoxic environment of tumor cells should be considered when analyzing the effect of bioactive compounds. Besides, this study also points to the importance of assessing the regulatory role of HT at both mRNA and protein level to get a complete picture of its effects.
Collapse
Affiliation(s)
- Jesús Calahorra
- Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, Jaén, Spain
| | - Esther Martínez-Lara
- Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, Jaén, Spain
| | - Cristina De Dios
- Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, Jaén, Spain
| | - Eva Siles
- Department of Experimental Biology, University of Jaén, Campus Las Lagunillas s/n, Jaén, Spain
| |
Collapse
|
15
|
Lynch C, Zhao J, Huang R, Kanaya N, Bernal L, Hsieh JH, Auerbach SS, Witt KL, Merrick BA, Chen S, Teng CT, Xia M. Identification of Estrogen-Related Receptor α Agonists in the Tox21 Compound Library. Endocrinology 2018; 159:744-753. [PMID: 29216352 PMCID: PMC5774247 DOI: 10.1210/en.2017-00658] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/28/2017] [Indexed: 12/17/2022]
Abstract
The estrogen-related receptor α (ERRα) is an orphan nuclear receptor (NR) that plays a role in energy homeostasis and controls mitochondrial oxidative respiration. Increased expression of ERRα in certain ovarian, breast, and colon cancers has a negative prognosis, indicating an important role for ERRα in cancer progression. An interaction between ERRα and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) has also recently been shown to regulate an enzyme in the β-oxidation of free fatty acids, thereby suggesting that ERRα plays an important role in obesity and type 2 diabetes. Therefore, it would be prudent to identify compounds that can act as activators of ERRα. In this study, we screened ∼10,000 (8311 unique) compounds, known as the Tox21 10K collection, to identify agonists of ERRα. We performed this screen using two stably transfected HEK 293 cell lines, one with the ERRα-reporter alone and the other with both ERRα-reporter and PGC-1α expression vectors. After the primary screening, we identified more than five agonist clusters based on compound structural similarity analysis (e.g., statins). By examining the activities of the confirmed ERRα modulators in other Tox21 NR assays, eliminating those with promiscuous NR activity, and performing follow-up assays (e.g., small interfering RNA knockdown), we identified compounds that might act as endocrine disrupters through effects on ERRα signaling. To our knowledge, this study is the first comprehensive analysis in discovering potential endocrine disrupters that affect the ERRα signaling pathway.
Collapse
Affiliation(s)
- Caitlin Lynch
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| | - Jinghua Zhao
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| | - Noriko Kanaya
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | - Lauren Bernal
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | - Jui-Hua Hsieh
- Kelly Government Solutions, Durham, North Carolina 27560
| | - Scott S. Auerbach
- Division of the National Toxicology Program, Biomolecular Screening Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Kristine L. Witt
- Division of the National Toxicology Program, Biomolecular Screening Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - B. Alex Merrick
- Division of the National Toxicology Program, Biomolecular Screening Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Shiuan Chen
- Department of Cancer Biology, Beckman Research Institute of the City of Hope, Duarte, California 91010
| | - Christina T. Teng
- Division of the National Toxicology Program, Biomolecular Screening Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709
| | - Menghang Xia
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
16
|
Casaburi I, Chimento A, De Luca A, Nocito M, Sculco S, Avena P, Trotta F, Rago V, Sirianni R, Pezzi V. Cholesterol as an Endogenous ERRα Agonist: A New Perspective to Cancer Treatment. Front Endocrinol (Lausanne) 2018; 9:525. [PMID: 30254608 PMCID: PMC6141749 DOI: 10.3389/fendo.2018.00525] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/21/2018] [Indexed: 01/01/2023] Open
Abstract
The estrogen-related receptors (ERRs) are important members of nuclear receptors which contain three isoforms (α, β, and γ). ERRα is the best-characterized isoform expressed mainly in high-energy demanding tissues where it preferentially works in association with the peroxisome proliferator-activated receptor-γ co-activator 1α (PGC-1α) and PGC-1β. ERRα together with its cofactors modulates cellular metabolism, supports the growth of rapidly dividing cells, directs metabolic programs required for cell differentiation and maintains cellular energy homeostasis in differentiated cells. In cancer cells, the functional association between ERRα and PGC-1s is further influenced by oncogenic signals and induces metabolic programs favoring cell growth and proliferation as well as tumor progression. Recently, cholesterol has been identified as a natural ERRα ligand using a combined biochemical strategy. This new finding highlighted some important physiological aspects related to the use of cholesterol-lowering drugs such as statins and bisphosphonates. Even more meaningful is the link between increased cholesterol levels and certain cancer phenotypes characterized by an overexpressed ERRα such as mammary, prostatic, and colorectal cancers, where the metabolic adaptation affects many cancer processes. Moreover, high-energy demanding cancer-related processes are strictly related to the cross-talk between tumor cells and some key players of tumor microenvironment, such as tumor-associated macrophage that fuels cancer progression. Some evidence suggests that high cholesterol content and ERRα activity favor the inflammatory environment by the production of different cytokines. In this review, starting from the most recent observations on the physiological role of the new signaling activated by the natural ligand of ERRα, we propose a new hypothesis on the suitability to control cholesterol levels as a chance in modulating ERRα activity in those tumors in which its expression and activity are increased.
Collapse
|
17
|
Abstract
Mitochondria are essential organelles for many aspects of cellular homeostasis, including energy harvesting through oxidative phosphorylation. Alterations of mitochondrial function not only impact on cellular metabolism but also critically influence whole-body metabolism, health, and life span. Diseases defined by mitochondrial dysfunction have expanded from rare monogenic disorders in a strict sense to now also include many common polygenic diseases, including metabolic, cardiovascular, neurodegenerative, and neuromuscular diseases. This has led to an intensive search for new therapeutic and preventive strategies aimed at invigorating mitochondrial function by exploiting key components of mitochondrial biogenesis, redox metabolism, dynamics, mitophagy, and the mitochondrial unfolded protein response. As such, new findings linking mitochondrial function to the progression or outcome of this ever-increasing list of diseases has stimulated the discovery and development of the first true mitochondrial drugs, which are now entering the clinic and are discussed in this review.
Collapse
Affiliation(s)
- Vincenzo Sorrentino
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland;
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, Ottawa K1H 8M5, Canada;
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
18
|
Ning Y, Chen H, Du Y, Ling H, Zhang L, Chen L, Qi H, Shi X, Li Q. A novel compound LingH2-10 inhibits the growth of triple negative breast cancer cells in vitro and in vivo as a selective inverse agonist of estrogen-related receptor α. Biomed Pharmacother 2017; 93:913-922. [PMID: 28715872 DOI: 10.1016/j.biopha.2017.07.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 02/07/2023] Open
Abstract
Unlike other breast cancer subtypes, targeted therapies for triple negative breast cancer (TNBC) have yet to progress past clinical trial stage to approval. Accumulating evidences demonstrated that expression of estrogen-related receptor alpha (ERRα) indicated worse prognosis and correlated with poor outcome in breast cancers including TNBC. Therefore, ERRα modulators/regulators may be potential in the therapeutic treatment of breast cancers. In the current study, we presented a novel compound LingH2-10 that bound to ERRα, as identified using a time-resolved fluorescence resonance energy transfer assay (TR-FRET) with the IC50 value of 0.64±0.12μM. Further, functional activity was determined by transient transfection luciferase reporter assay in order to validate the utility of the binding affinity in a cellular context. LingH2-10 showed selective inhibition on ERRα transcriptional activity with the IC50 value of 0.58±0.09μM in cell-based luciferase reporter assay. Moreover, representative in vitro results showed that LingH2-10 suppressed the proliferation of various human cancer cells, and inhibited the migration of triple negative breast cancer cell MDA-MB-231. In addition, our results demonstrated that well known ERRα target genes such as PDK4, Osteopontin and pS2, were all significantly down modulated by LingH2-10. In vivo experiments showed that LingH2-10 (30mg/kg, every other day) observably suppressed the growth of MDA-MB-231 xenograft tumors by 42.02% compared to untreated xenograft tumors. Taken together, all these data suggested that LingH2-10, as a selective inverse agonist of ERRα, was a lead compound of anti-cancer agents for treating TNBC patients.
Collapse
Affiliation(s)
- Yang Ning
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China
| | - Haifei Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Yongli Du
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China.
| | - Hao Ling
- School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, 3501 Daxue Road, Jinan 250353, China
| | - Liudi Zhang
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Lu Chen
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Huijie Qi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China
| | - Xiaojin Shi
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China; Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qunyi Li
- Clinical Pharmacy Laboratory, Huashan Hospital North, Fudan University, Shanghai 201907, China; Clinical Pharmacy Laboratory, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
19
|
Seneviratne APB, Turan Z, Hermant A, Lecine P, Smith WO, Borg JP, Jaulin F, Kreitzer G. Modulation of estrogen related receptor alpha activity by the kinesin KIF17. Oncotarget 2017; 8:50359-50375. [PMID: 28881568 PMCID: PMC5584137 DOI: 10.18632/oncotarget.18104] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 05/12/2017] [Indexed: 12/26/2022] Open
Abstract
Estrogen-related receptor alpha (ERR1) is an orphan nuclear receptor that can bind transcriptional co-activators constitutively. ERR1 expression correlates with poor patient outcomes in breast cancer, heightening interest in this nuclear receptor as a therapeutic target. Because ERR1 has no known regulatory ligand, a major challenge in targeting its activity is to find cellular or synthetic modulators of its function. We identified an interaction between ERR1 and KIF17, a kinesin-2 family microtubule motor, in a yeast-2-hybrid screen. We confirmed the interaction using in vitro biochemical assays and determined that binding is mediated by the ERR1 ligand-binding/AF2 domain and the KIF17 C-terminal tail. Expression of KIF17 tail domain in either ER-negative or ER-positive breast cancer epithelial cells attenuated nuclear accumulation of newly synthesized ERR1 and inhibited ERR1 transcriptional activity. Conversely, ERR1 transcriptional activity was elevated significantly in KIF17 knock-out cells. Sequence analysis of the KIF17 tail domain revealed it contains a nuclear receptor box with a conserved LXXLL motif found in transcriptional co-activators. Expression of a 12 amino-acid peptide containing this motif was sufficient to inhibit ERR1 transcriptional activity and cell invasion, while deletion of this region from the KIF17 tail resulted in increased ERR1 activity. Together, these data suggest KIF17 modifies ERR1 function by two possible, non-exclusive mechanisms: (i) by regulating nuclear-cytoplasmic distribution or (ii) by competing with transcriptional co-activators for binding to ERR1. Thus targeting the ERR1-KIF17 interaction has potential as a novel strategy for treating breast cancer.
Collapse
Affiliation(s)
- Am Pramodh Bandara Seneviratne
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA.,Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,The City University of New York School of Medicine, New York, NY, USA
| | - Zeynep Turan
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,California Institute of Technology, Pasadena, CA, USA
| | - Aurelie Hermant
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Institut Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell Signalling and Cancer, Equipe labellisée Ligue Contre le Cancer, Marseille, France
| | - Patrick Lecine
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Institut Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell Signalling and Cancer, Equipe labellisée Ligue Contre le Cancer, Marseille, France.,BIOASTER, Tony Garnier, Lyon, France
| | - William O Smith
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA
| | - Jean-Paul Borg
- Centre de Recherche en Cancérologie de Marseille, Aix Marseille Univ UM105, Institut Paoli-Calmettes, UMR7258 CNRS, U1068 INSERM, Cell Polarity, Cell Signalling and Cancer, Equipe labellisée Ligue Contre le Cancer, Marseille, France
| | - Fanny Jaulin
- Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,Gustave Roussy Institute, Villejuif, France
| | - Geri Kreitzer
- Department of Molecular, Cellular & Biomedical Sciences, The City University of New York School of Medicine, New York, NY, USA.,Department of Cell and Developmental Biology, Weill Medical College, Cornell University, New York, NY, USA.,The City University of New York School of Medicine, New York, NY, USA
| |
Collapse
|
20
|
Schumacher FR, Schubert S, Hannus M, Sönnichsen B, Ittrich C, Kreideweiss S, Kurz T, Rippmann JF. RNAi Screen for NRF2 Inducers Identifies Targets That Rescue Primary Lung Epithelial Cells from Cigarette Smoke Induced Radical Stress. PLoS One 2016; 11:e0166352. [PMID: 27832175 PMCID: PMC5104413 DOI: 10.1371/journal.pone.0166352] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 10/27/2016] [Indexed: 12/30/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a highly prevalent condition characterized by inflammation and progressive obstruction of the airways. At present, there is no treatment that suppresses the chronic inflammation of the disease, and COPD patients often succumb to the condition. Excessive oxidative stress caused by smoke inhalation is a major driving force of the disease. The transcription factor NRF2 is a critical player in the battle against oxidative stress and its function is impaired in COPD. Increasing NRF2 activity may therefore be a viable therapeutic option for COPD treatment. We show that down regulation of KEAP1, a NRF2 inhibitor, protects primary human lung epithelial cells from cigarette-smoke-extract (CSE) induced cell death in an established in vitro model of radical stress. To identify new potential drug targets with a similar effect, we performed a siRNA screen of the ‘druggable’ genome using a NRF2 transcriptional reporter cell line. This screen identified multiple genes that when down regulated increased NRF2 transcriptional activity and provided a survival benefit in the in vitro model. Our results suggest that inhibiting components of the ubiquitin-proteasome system will have the strongest effects on NRF2 transcriptional activity by increasing NRF2 levels. We also find that down regulation of the small GTPase Rab28 or the Estrogen Receptor ESRRA provide a survival benefit. Rab28 knockdown increased NRF2 protein levels, indicating that Rab28 may regulate NRF2 proteolysis. Conversely ESRRA down regulation increased NRF2 transcriptional activity without affecting NRF2 levels, suggesting a proteasome-independent mechanism.
Collapse
Affiliation(s)
- Frances-Rose Schumacher
- MRC Protein Phosphorylation and Ubiquitylation Unit, The Sir James Black Center, University of Dundee, Dow Street, Dundee, DD1 5EH, United Kingdom
| | | | | | | | - Carina Ittrich
- Boehringer Ingelheim Pharma GmbH & Co. KG, Research Germany, 8400, Biberach a. d. Riss, Germany
| | - Stefan Kreideweiss
- Boehringer Ingelheim Pharma GmbH & Co. KG, Research Germany, 8400, Biberach a. d. Riss, Germany
| | - Thimo Kurz
- MRC Protein Phosphorylation and Ubiquitylation Unit, The Sir James Black Center, University of Dundee, Dow Street, Dundee, DD1 5EH, United Kingdom
- * E-mail:
| | - Jörg F. Rippmann
- Boehringer Ingelheim Pharma GmbH & Co. KG, Research Germany, 8400, Biberach a. d. Riss, Germany
| |
Collapse
|
21
|
Carnesecchi J, Vanacker JM. Estrogen-Related Receptors and the control of bone cell fate. Mol Cell Endocrinol 2016; 432:37-43. [PMID: 26206717 DOI: 10.1016/j.mce.2015.07.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 06/23/2015] [Accepted: 07/17/2015] [Indexed: 11/17/2022]
Abstract
Bone loss is naturally occurring in aging males and females and exacerbated in the latter after menopause, altogether leading to cumulative skeleton fragility and increased fracture risk. Two types of therapeutic strategies can be envisioned to counteract age- or menopause-associated bone loss, aiming at either reducing bone resorption exerted by osteoclasts or, alternatively, promoting bone formation by osteoblasts. We here summarize data suggesting that inhibition of the Estrogen-Related Receptors α and/or γ could promote bone formation and compensate for bone loss induced by ageing or estrogen-deficiency.
Collapse
Affiliation(s)
- Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon I, CNRS UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
22
|
Tan X, Gao J, Shi Z, Tai S, Chan LL, Yang Y, Peng DQ, Liao DF, Jiang ZS, Chang YZ, Gui Y, Zheng XL. MG132 Induces Expression of Monocyte Chemotactic Protein-Induced Protein 1 in Vascular Smooth Muscle Cells. J Cell Physiol 2016; 232:122-8. [PMID: 27035356 DOI: 10.1002/jcp.25396] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/29/2016] [Indexed: 11/07/2022]
Abstract
Monocyte chemoattractant protein-1 (MCP-1) has been reported to induce the expression of monocyte chemotactic protein-induced protein 1 (MCPIP1), which undergoes ubiquitination degradation. Therefore, we predict that in vascular smooth muscle (VSMCs), MCPIP1 may be induced by MCP-1 and undergo degradation, which can be inhibited by the proteasome inhibitor, MG132. Our results showed that treatment of human VSMCs with MCP-1 did not increase the expression of MCPIP1. Treatment with MG132, however, elevated MCPIP1 protein levels through stimulation of the gene transcription, but not through increasing protein stability. MCPIP1 expression induced by MG132 was inhibited by α-amanitin inhibition of gene transcription or cycloheximide inhibition of protein synthesis. Our further studies showed that MCPIP1 expression induced by MG132 was inhibited by the inhibitors of AKT and p38 kinase, suggesting a role of the AKT-p38 pathway in MG132 effects. We also found that treatment with MG132 induces apoptosis, but overexpression of MCPIP1 inhibited bromodeoxyuridine (BrdU) incorporation of human VSMCs without induction of significant apoptosis. In summary, MCPIP1 expression is induced by MG132 likely through activation of the AKT-p38 pathway. MCPIP1 inhibits SMC proliferation without induction of apoptosis. J. Cell. Physiol. 232: 122-128, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Xi Tan
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (Incubation), Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Jie Gao
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Zhan Shi
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Shi Tai
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Leona Loretta Chan
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yang Yang
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Dao-Quan Peng
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Duan-Fang Liao
- Division of Stem Cell Regulation and Application, State Key Laboratory of Chinese Medicine Powder and Medicine Innovation in Hunan (Incubation), Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhi-Sheng Jiang
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Institute of Cardiovascular Disease and Key Lab for Arteriosclerogy of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Ying-Zi Chang
- Department of Pharmacology, A. T. Still University, Kirksville College of Osteopathic Medicine, Kirksville, Missouri
| | - Yu Gui
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, Smooth Muscle Research Group, Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
23
|
The Major Prognostic Features of Nuclear Receptor NR5A2 in Infiltrating Ductal Breast Carcinomas. Int J Genomics 2015; 2015:403576. [PMID: 26366408 PMCID: PMC4561099 DOI: 10.1155/2015/403576] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 07/22/2015] [Indexed: 01/08/2023] Open
Abstract
Background. Gene expression profiles of 181 breast cancer samples were analyzed to identify prognostic features of nuclear receptors NR5A1 and NR5A2 based upon their associated transcriptional networks. Methods. A supervised network analysis approach was used to build the NR5A-mediated transcriptional regulatory network. Other bioinformatic tools and statistical methods were utilized to confirm and extend results from the network analysis methodology. Results. NR5A2 expression is a negative factor in breast cancer prognosis in both ER(-) and ER(-)/ER(+) mixed cohorts. The clinical and cohort significance of NR5A2-mediated transcriptional activities indicates that it may have a significant role in attenuating grade development and cancer related signal transduction pathways. NR5A2 signature that conditions poor prognosis was identified based upon results from 15 distinct probes. Alternatively, the expression of NR5A1 predicts favorable prognosis when concurrent NR5A2 expression is low. A favorable signature of eight transcription factors mediated by NR5A1 was also identified. Conclusions. Correlation of poor prognosis and NR5A2 activity is identified by NR5A2-mediated 15-gene signature. NR5A2 may be a potential drug target for treating a subset of breast cancer tumors across breast cancer subtypes, especially ER(-) breast tumors. The favorable prognostic feature of NR5A1 is predicted by NR5A1-mediated 8-gene signature.
Collapse
|
24
|
Constitutive activities of estrogen-related receptors: Transcriptional regulation of metabolism by the ERR pathways in health and disease. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1912-27. [PMID: 26115970 DOI: 10.1016/j.bbadis.2015.06.016] [Citation(s) in RCA: 142] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022]
Abstract
The estrogen-related receptors (ERRs) comprise a small group of orphan nuclear receptor transcription factors. The ERRα and ERRγ isoforms play a central role in the regulation of metabolic genes and cellular energy metabolism. Although less is known about ERRβ, recent studies have revealed the importance of this isoform in the maintenance of embryonic stem cell pluripotency. Thus, ERRs are essential to many biological processes. The development of several ERR knockout and overexpression models and the application of advanced functional genomics have allowed rapid advancement of our understanding of the physiology regulated by ERR pathways. Moreover, it has enabled us to begin to delineate the distinct programs regulated by ERRα and ERRγ that have overlapping effects on metabolism and growth. The current review primarily focuses on the physiologic roles of ERR isoforms related to their metabolic regulation; therefore, the ERRα and ERRγ are discussed in the greatest detail. We emphasize findings from gain- and loss-of-function models developed to characterize ERR control of skeletal muscle, heart and musculoskeletal physiology. These models have revealed that coordinating metabolic capacity with energy demand is essential for seemingly disparate processes such as muscle differentiation and hypertrophy, innate immune function, thermogenesis, and bone remodeling. Furthermore, the models have revealed that ERRα- and ERRγ-deficiency in mice accelerates progression of pathologic processes and implicates ERRs as etiologic factors in disease. We highlight the human diseases in which ERRs and their downstream metabolic pathways are perturbed, including heart failure and diabetes. While no natural ligand has been identified for any of the ERR isoforms, the potential for using synthetic small molecules to modulate their activity has been demonstrated. Based on our current understanding of their transcriptional mechanisms and physiologic relevance, the ERRs have emerged as potential therapeutic targets for treatment of osteoporosis, muscle atrophy, insulin resistance and heart failure in humans.
Collapse
|
25
|
Dai X, Chen A, Bai Z. Integrative investigation on breast cancer in ER, PR and HER2-defined subgroups using mRNA and miRNA expression profiling. Sci Rep 2014; 4:6566. [PMID: 25338681 PMCID: PMC4206873 DOI: 10.1038/srep06566] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 09/15/2014] [Indexed: 12/20/2022] Open
Abstract
Exploring the molecular difference among breast cancer subtypes is of crucial importance in understanding its heterogeneity and seeking its effective clinical treatment. For this, several layers of information including immunohistochemical markers and a variety of high-throughput genomics approaches have been intensively used. Here we have explored the intrinsic differences among breast cancer subgroups defined by immunohistochemical expression (IHC) of hormone receptors ER and PR as well as human epidermal growth factor receptor 2 (HER2) using the mRNA and miRNA expression profiles of 115 tumors. A core basal group was further defined by epidermal growth factor receptor and cytokeratin 5/6 IHC expression and compared to triple negative group. A set of differentially expressed genes including 1015 mRNAs and 69 miRNAs was found to distinguish tumor subtypes whose generality was demonstrated using two independent data sets. The network was explored for each subtype and biomass synthesis signaling was found to play an important role in the core basal subgroup. This study contributes to elucidating the intrinsic relations among breast cancer subgroups defined by ER, PR and HER2 expression via integrating mRNA and miRNA expression. The results can avail functional studies of breast cancer with translational potential for clinical use.
Collapse
Affiliation(s)
- Xiaofeng Dai
- 1] School of Biotechnology, JiangNan University, Wuxi 214122, China [2] National Engineering Laboratory for Cereal Fermention Technology, Jiangnan University, Wuxi 214122, China
| | - Ana Chen
- 1] School of Biotechnology, JiangNan University, Wuxi 214122, China [2] School of Biochemical Engineering, Anhui Polytechnic University, Wuhu 241000, China
| | - Zhonghu Bai
- 1] School of Biotechnology, JiangNan University, Wuxi 214122, China [2] National Engineering Laboratory for Cereal Fermention Technology, Jiangnan University, Wuxi 214122, China
| |
Collapse
|
26
|
May FE. Novel drugs that target the estrogen-related receptor alpha: their therapeutic potential in breast cancer. Cancer Manag Res 2014; 6:225-52. [PMID: 24904222 PMCID: PMC4041375 DOI: 10.2147/cmar.s35024] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
THE INCIDENCE OF BREAST CANCER CONTINUES TO RISE 1.7 million women were diagnosed with and 521,000 women died from breast cancer in 2012. This review considers first current treatment options: surgery; radiotherapy; and systemic endocrine, anti-biological, and cytotoxic therapies. Clinical management includes prevention, early detection by screening, treatment with curative intent, management of chronic disease, and palliative control of advanced breast cancer. Next, the potential of novel drugs that target DNA repair, growth factor dependence, intracellular and intercellular signal transduction, and cell cycle are considered. Estrogen-related receptor alpha has attracted attention as a therapeutic target in triple-negative breast cancers with de novo resistance to, and in breast cancers with acquired resistance to, endocrine therapies such as antiestrogens and aromatase inhibitors. Estrogen-related receptor alpha is an orphan receptor and transcription factor. Its activity is regulated by coregulator proteins and posttranslational modification. It is an energy sensor that controls adaptation to energy demand and may facilitate glycolytic metabolism and mitochondrial oxidative respiration in breast cancer cells. Estrogen-related receptor alpha increases breast cancer cell migration, proliferation, and tumor development. It is expressed at high levels in estrogen receptor-negative tumors, and is proposed to activate estrogen-responsive genes in endocrine-resistant tumors. The structures and functions of the ligand-binding domains of estrogen receptor alpha and estrogen-related receptor alpha, their ability to bind estrogens, phytoestrogens, and synthetic ligands, and the effects of ligand agonists, antagonists, and inverse agonists on biological activity, are evaluated. Synthetic ligands of estrogen-related receptor alpha have activity in preclinical models of metabolic disorders, diabetes, osteoporosis, and oncology. The clinical settings in which these novel drugs might have utility in the management of advanced breast cancer, and biomarkers for stratification of patients likely to benefit, are discussed. Finally, the potential side effects of the novel drugs on metabolism, osteoporosis, osteo-metastasis, and cachexia are considered.
Collapse
Affiliation(s)
- Felicity Eb May
- Northern Institute for Cancer Research and Department of Pathology, Faculty of Medical Sciences, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| |
Collapse
|
27
|
Teng CT, Beames B, Alex Merrick B, Martin N, Romeo C, Jetten AM. Development of a stable cell line with an intact PGC-1α/ERRα axis for screening environmental chemicals. Biochem Biophys Res Commun 2014; 444:177-81. [PMID: 24457025 PMCID: PMC3967403 DOI: 10.1016/j.bbrc.2014.01.033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 01/10/2014] [Indexed: 01/24/2023]
Abstract
The estrogen-related receptor α (ERRα) and the peroxisome proliferator-activated receptor γ (PPARγ) coactivator 1α (PGC-1α) play critical roles in the control of several physiological functions, including the regulation of genes involved in energy homeostasis. However, little is known about the ability of environmental chemicals to disrupt or modulate this important bioenergetics pathway in humans. The goal of this study was to develop a cell-based assay system with an intact PGC-1α/ERRα axis that could be used as a screening assay for detecting such chemicals. To this end, we successfully generated several stable cell lines expressing PGC-1α and showed that the reporter driven by the native ERRα hormone response unit (AAB-Luc) is active in these cell lines and that the activation is PGC-1α-dependent. Furthermore, we show that this activation can be blocked by the ERRα selective inverse agonist, XCT790. In addition, we find that genistein and bisphenol A further stimulate the reporter activity, while kaempferol has minimal effect. These cell lines will be useful for identifying environmental chemicals that modulate this important pathway.
Collapse
Affiliation(s)
- Christina T Teng
- DNTP, BioMolecular Screening Branch, Division, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States.
| | - Burton Beames
- DNTP, BioMolecular Screening Branch, Division, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| | - B Alex Merrick
- DNTP, BioMolecular Screening Branch, Division, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| | - Negin Martin
- DIR, Viral Core Lab, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| | - Charles Romeo
- DIR, Viral Core Lab, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| | - Anton M Jetten
- DIR Laboratory of Respiratory Biology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States
| |
Collapse
|
28
|
Abstract
Mitochondrial dysfunction is not only a hallmark of rare inherited mitochondrial disorders but also implicated in age-related diseases, including those that affect the metabolic and nervous system, such as type 2 diabetes and Parkinson's disease. Numerous pathways maintain and/or restore proper mitochondrial function, including mitochondrial biogenesis, mitochondrial dynamics, mitophagy and the mitochondrial unfolded protein response. New and powerful phenotypic assays in cell-based models as well as multicellular organisms have been developed to explore these different aspects of mitochondrial function. Modulating mitochondrial function has therefore emerged as an attractive therapeutic strategy for several diseases, which has spurred active drug discovery efforts in this area.
Collapse
|
29
|
Kammerer M, Gutzwiller S, Stauffer D, Delhon I, Seltenmeyer Y, Fournier B. Estrogen Receptor α (ERα) and Estrogen Related Receptor α (ERRα) are both transcriptional regulators of the Runx2-I isoform. Mol Cell Endocrinol 2013; 369:150-60. [PMID: 23403054 DOI: 10.1016/j.mce.2013.01.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 01/09/2013] [Accepted: 01/29/2013] [Indexed: 01/18/2023]
Abstract
Runx2 is a master regulator of bone development and has also been described as an oncogene. Estrogen Receptor α (ERα) and Estrogen Related Receptor α (ERRα), both implicated in bone metabolism and breast cancer, have been shown to share common transcriptional targets. Here, we show that ERα is a positive regulator of Runx2-I transcription. Moreover, ERRα can act as a transcriptional activator of Runx2-I in presence of peroxisome proliferator activated receptor gamma coactivator-1 alpha (PGC-1α). In contrast, ERRα behaves as a negative regulator of Runx2-I transcription in presence of PGC-1β. ERα and ERRα cross-talk via a common estrogen receptor response element on the Runx2-I promoter. In addition, estrogen regulates PGC-1β that in turn is able to modulate both ERα and ERRα transcriptional activity.
Collapse
Affiliation(s)
- Martial Kammerer
- Novartis Institutes for BioMedical Research, Musculoskeletal Disease Unit, CH-4002 Basel, Switzerland
| | | | | | | | | | | |
Collapse
|
30
|
Gallet M, Saïdi S, Haÿ E, Photsavang J, Marty C, Sailland J, Carnesecchi J, Tribollet V, Barenton B, Forcet C, Birling MC, Sorg T, Chassande O, Cohen-Solal M, Vanacker JM. Repression of osteoblast maturation by ERRα accounts for bone loss induced by estrogen deficiency. PLoS One 2013; 8:e54837. [PMID: 23359549 PMCID: PMC3554601 DOI: 10.1371/journal.pone.0054837] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 12/17/2012] [Indexed: 01/09/2023] Open
Abstract
ERRα is an orphan member of the nuclear receptor family, the complete inactivation of which confers resistance to bone loss induced by ageing and estrogen withdrawal to female mice in correlation with increased bone formation in vivo. Furthermore ERRα negatively regulates the commitment of mesenchymal cells to the osteoblast lineage ex vivo as well as later steps of osteoblast maturation. We searched to determine whether the activities of ERRα on osteoblast maturation are responsible for one or both types of in vivo induced bone loss. To this end we have generated conditional knock out mice in which the receptor is normally present during early osteoblast differentiation but inactivated upon osteoblast maturation. Bone ageing in these animals was similar to that observed for control animals. In contrast conditional ERRαKO mice were completely resistant to bone loss induced by ovariectomy. We conclude that the late (maturation), but not early (commitment), negative effects of ERRα on the osteoblast lineage contribute to the reduced bone mineral density observed upon estrogen deficiency.
Collapse
Affiliation(s)
- Marlène Gallet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre national de la recherche scientifique UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Soraya Saïdi
- Institut National de la Santé et de la Recherche Médicale U606, Hôpital Lariboisière, Paris, France
| | - Eric Haÿ
- Institut National de la Santé et de la Recherche Médicale U606, Hôpital Lariboisière, Paris, France
| | - Johann Photsavang
- Institut National de la Santé et de la Recherche Médicale U1026, Bordeaux, France
| | - Caroline Marty
- Institut National de la Santé et de la Recherche Médicale U606, Hôpital Lariboisière, Paris, France
| | - Juliette Sailland
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre national de la recherche scientifique UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Julie Carnesecchi
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre national de la recherche scientifique UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Violaine Tribollet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre national de la recherche scientifique UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Bruno Barenton
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre national de la recherche scientifique UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Christelle Forcet
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre national de la recherche scientifique UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Tania Sorg
- Institut Clinique de la Souris, Illkirch-Graffenstaden, France
| | - Olivier Chassande
- Institut National de la Santé et de la Recherche Médicale U1026, Bordeaux, France
| | - Martine Cohen-Solal
- Institut National de la Santé et de la Recherche Médicale U606, Hôpital Lariboisière, Paris, France
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Centre national de la recherche scientifique UMR5242, Ecole Normale Supérieure de Lyon, Lyon, France
- * E-mail:
| |
Collapse
|
31
|
Chang CY, McDonnell DP. Molecular pathways: the metabolic regulator estrogen-related receptor α as a therapeutic target in cancer. Clin Cancer Res 2012; 18:6089-95. [PMID: 23019305 PMCID: PMC3500439 DOI: 10.1158/1078-0432.ccr-11-3221] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The estrogen-related receptor α (ERRα) is an orphan member of the nuclear receptor superfamily of transcription factors whose activity is regulated by the expression level and/or activity of its obligate coregulators, peroxisome proliferator-activated receptor γ coactivator-1 α and β (PGC-1α or PGC-1β). Under normal physiologic conditions, and in responding to different environmental stimuli, the ERRα/PGC-1 complex is involved in regulating metabolic homeostasis under conditions of high energy demand in brown adipocytes, proliferating T cells, and muscle. Interestingly, increased expression and activity of the ERRα/PGC-1 axis has also been shown to correlate with unfavorable clinical outcomes in both breast and ovarian tumors. The observation that ERRα activity is manifest in all breast tumor subtypes with particularly high activity being evident in ERα-negative, HER2-positive, and triple-negative breast cancers has raised significant interest in targeting this receptor for the treatment of those breast cancers for which therapeutic options are limited.
Collapse
Affiliation(s)
- Ching-yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina 27710, USA
| | | |
Collapse
|
32
|
Bianco S, Sailland J, Vanacker JM. ERRs and cancers: effects on metabolism and on proliferation and migration capacities. J Steroid Biochem Mol Biol 2012; 130:180-5. [PMID: 21414406 DOI: 10.1016/j.jsbmb.2011.03.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 01/20/2011] [Accepted: 03/07/2011] [Indexed: 01/06/2023]
Abstract
ERRs are orphan members of the nuclear receptor superfamily which, at least for ERRα and ERRγ display important roles in the control of various metabolic processes. On other hand, correlations have been found between the expression of ERRα and γ and diverse parameters of tumor progression in human cancers. Whereas it is tempting to speculate that ERR receptors act in tumors through the regulation of metabolism, recent data have suggested that they also may directly regulate tumor proliferation and progression independently of their effects on metabolism. The two aspects of tumoral functions of ERR receptors are the purpose of the present review.
Collapse
Affiliation(s)
- Stéphanie Bianco
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364 Lyon cedex 07, France
| | | | | |
Collapse
|
33
|
Zhao Y, Li Y, Lou G, Zhao L, Xu Z, Zhang Y, He F. MiR-137 targets estrogen-related receptor alpha and impairs the proliferative and migratory capacity of breast cancer cells. PLoS One 2012; 7:e39102. [PMID: 22723937 PMCID: PMC3377602 DOI: 10.1371/journal.pone.0039102] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2011] [Accepted: 05/16/2012] [Indexed: 12/05/2022] Open
Abstract
ERRα is an orphan nuclear receptor emerging as a novel biomarker of breast cancer. Over-expression of ERRα in breast tumor is considered as a prognostic factor of poor clinical outcome. The mechanisms underlying the dysexpression of this nuclear receptor, however, are poorly understood. MicroRNAs (miRNAs) regulate gene expression at the post-transcriptional level and play important roles in tumor initiation and progression. In the present study, we have identified that the expression of ERRα is regulated by miR-137, a potential tumor suppressor microRNA. The bioinformatics search revealed two putative and highly conserved target-sites for miR-137 located within the ERRα 3'UTR at nt 480-486 and nt 596-602 respectively. Luciferase-reporter assay demonstrated that the two predicted target sites were authentically functional. They mediated the repression of reporter gene expression induced by miR-137 in an additive manner. Moreover, ectopic expression of miR-137 down-regulated ERRα expression at both protein level and mRNA level, and the miR-137 induced ERRα-knockdown contributed to the impaired proliferative and migratory capacity of breast cancer cells. Furthermore, transfection with miR-137 mimics suppressed at least two downstream target genes of ERRα-CCNE1 and WNT11, which are important effectors of ERRα implicated in tumor proliferation and migration. Taken together, our results establish a role of miR-137 in negatively regulating ERRα expression and breast cancer cell proliferation and migration. They suggest that manipulating the expression level of ERRα by microRNAs has the potential to influence breast cancer progression.
Collapse
Affiliation(s)
- Yuanyin Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yuping Li
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Guiyu Lou
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Li Zhao
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Yan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| | - Fengtian He
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Third Military Medical University, Chongqing, China
| |
Collapse
|
34
|
Liu LYD, Chang LY, Kuo WH, Hwa HL, Shyu MK, Chang KJ, Hsieh FJ. In Silico Prediction for Regulation of Transcription Factors onTheir Shared Target Genes Indicates Relevant Clinical Implications in a Breast Cancer Population. Cancer Inform 2012; 11:113-37. [PMID: 22553415 PMCID: PMC3337786 DOI: 10.4137/cin.s8470] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Aberrant transcriptional activities have been documented in breast cancers. Studies often find some transcription factors to be inappropriately regulated and enriched in certain pathological states. The promoter regions of most target genes have binding sites for their transcription factors. An ample of evidence supports their combinatorial effect on their shared target gene expressions. Here, we used a new statistic method, bivariate CID, to predict combinatorial interaction activity between ERα and a transcription factor (E2F1or GATA3 or ERRα) in regulating target gene expression via four regulatory mechanisms. We identified gene sets in three signal transduction pathways perturbed in breast tumors: cell cycle, VEGF, and PDGFRB. Bivariate network analysis revealed several target genes previously implicated in tumor angiogenesis are among the predicted shared targets, including VEGFA, PDGFRB. In summary, our analysis suggests the importance for the multivariate space of an inferred ERα transcriptional regulatory network in breast cancer diagnostic and therapeutic development.
Collapse
Affiliation(s)
- Li-Yu D Liu
- Department of Agronomy, Biometry Division, National Taiwan University, Taipei, Taiwan
| | - Li-Yun Chang
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Hung Kuo
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsiao-Lin Hwa
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ming-Kwang Shyu
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - King-Jen Chang
- Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fon-Jou Hsieh
- Department of Obstetrics and Gynecology, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
35
|
Genome-wide discovery of chromosomal copy number variants in human amniotic cell using array-based comparative genomic hybridization. Mol Cell Toxicol 2011. [DOI: 10.1007/s13273-011-0037-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
36
|
Deblois G, Giguère V. Functional and physiological genomics of estrogen-related receptors (ERRs) in health and disease. Biochim Biophys Acta Mol Basis Dis 2011; 1812:1032-40. [DOI: 10.1016/j.bbadis.2010.12.009] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 12/09/2010] [Accepted: 12/10/2010] [Indexed: 12/11/2022]
|
37
|
Yao X, Hao H, Li Y, Li S. Modularity-based credible prediction of disease genes and detection of disease subtypes on the phenotype-gene heterogeneous network. BMC SYSTEMS BIOLOGY 2011; 5:79. [PMID: 21599985 PMCID: PMC3130676 DOI: 10.1186/1752-0509-5-79] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 05/20/2011] [Indexed: 12/05/2022]
Abstract
Background Protein-protein interaction networks and phenotype similarity information have been synthesized together to discover novel disease-causing genes. Genetic or phenotypic similarities are manifested as certain modularity properties in a phenotype-gene heterogeneous network consisting of the phenotype-phenotype similarity network, protein-protein interaction network and gene-disease association network. However, the quantitative analysis of modularity in the heterogeneous network and its influence on disease-gene discovery are still unaddressed. Furthermore, the genetic correspondence of the disease subtypes can be identified by marking the genes and phenotypes in the phenotype-gene network. We present a novel network inference method to measure the network modularity, and in particular to suggest the subtypes of diseases based on the heterogeneous network. Results Based on a measure which is introduced to evaluate the closeness between two nodes in the phenotype-gene heterogeneous network, we developed a Hitting-Time-based method, CIPHER-HIT, for assessing the modularity of disease gene predictions and credibly prioritizing disease-causing genes, and then identifying the genetic modules corresponding to potential subtypes of the queried phenotype. The CIPHER-HIT is free to rely on any preset parameters. We found that when taking into account the modularity levels, the CIPHER-HIT method can significantly improve the performance of disease gene predictions, which demonstrates modularity is one of the key features for credible inference of disease genes on the phenotype-gene heterogeneous network. By applying the CIPHER-HIT to the subtype analysis of Breast cancer, we found that the prioritized genes can be divided into two sub-modules, one contains the members of the Fanconi anemia gene family, and the other contains a reported protein complex MRE11/RAD50/NBN. Conclusions The phenotype-gene heterogeneous network contains abundant information for not only disease genes discovery but also disease subtypes detection. The CIPHER-HIT method presented here is effective for network inference, particularly on credible prediction of disease genes and the subtype analysis of diseases, for example Breast cancer. This method provides a promising way to analyze heterogeneous biological networks, both globally and locally.
Collapse
Affiliation(s)
- Xin Yao
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, Tsinghua National Laboratory for Information Science and Technology, Tsinghua University, Beijing 100084, China
| | | | | | | |
Collapse
|
38
|
Duellman SJ, Calaoagan JM, Sato BG, Fine R, Klebansky B, Chao WR, Hobbs P, Collins N, Sambucetti L, Laderoute KR. A novel steroidal inhibitor of estrogen-related receptor alpha (ERR alpha). Biochem Pharmacol 2010; 80:819-26. [PMID: 20513360 PMCID: PMC2918268 DOI: 10.1016/j.bcp.2010.05.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 05/20/2010] [Accepted: 05/21/2010] [Indexed: 01/15/2023]
Abstract
The orphan nuclear receptor estrogen-related receptor alpha (ERRalpha) has been implicated in the development of various human malignancies, including breast, prostate, ovary, and colon cancer. ERRalpha, bound to a co-activator protein (e.g., peroxisome proliferator receptor gamma co-activator-1alpha, PGC-1alpha), regulates cellular energy metabolism by activating transcription of genes involved in various metabolic processes, such as mitochondrial genesis, oxidative phosphorylation, and fatty acid oxidation. Accumulating evidence suggests that ERRalpha is a novel target for solid tumor therapy, conceivably through effects on the regulation of tumor cell energy metabolism associated with energy stress within solid tumor microenvironments. This report describes a novel steroidal antiestrogen (SR16388) that binds selectively to ERRalpha, but not to ERRbeta or ERRgamma, as determined using a time-resolved fluorescence resonance energy transfer assay. SR16388 potently inhibits ERRalpha's transcriptional activity in reporter gene assays, and prevents endogenous PGC-1alpha and ERRalpha from being recruited to the promoters or enhancers of target genes. Representative in vivo results show that SR16388 inhibited the growth of human prostate tumor xenografts in nude mice as a single agent at 30mg/kg given once daily and 100mg/kg given once weekly. In a combination study, SR16388 (10mg/kg, once daily) and paclitaxel (7.5mg/kg, twice weekly) inhibited the growth of prostate tumor xenografts in nude mice by 61% compared to untreated xenograft tumors. SR16388 also inhibited the proliferation of diverse human tumor cell lines after a 24-h exposure to the compound. SR16388 thus has utility both as an experimental antitumor agent and as a chemical probe of ERRalpha biology.
Collapse
Affiliation(s)
- Sarah J Duellman
- Biosciences Division, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, United States.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Riggins RB, Mazzotta MM, Maniya OZ, Clarke R. Orphan nuclear receptors in breast cancer pathogenesis and therapeutic response. Endocr Relat Cancer 2010; 17:R213-31. [PMID: 20576803 PMCID: PMC3518023 DOI: 10.1677/erc-10-0058] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nuclear receptors comprise a large family of highly conserved transcription factors that regulate many key processes in normal and neoplastic tissues. Most nuclear receptors share a common, highly conserved domain structure that includes a carboxy-terminal ligand-binding domain. However, a subgroup of this gene family is known as the orphan nuclear receptors because to date there are no known natural ligands that regulate their activity. Many of the 25 nuclear receptors classified as orphan play critical roles in embryonic development, metabolism, and the regulation of circadian rhythm. Here, we review the emerging role(s) of orphan nuclear receptors in breast cancer, with a particular focus on two of the estrogen-related receptors (ERRalpha and ERRgamma) and several others implicated in clinical outcome and response or resistance to cytotoxic or endocrine therapies, including the chicken ovalbumin upstream promoter transcription factors, nerve growth factor-induced B, DAX-1, liver receptor homolog-1, and retinoic acid-related orphan receptor alpha. We also propose that a clearer understanding of the function of orphan nuclear receptors in mammary gland development and normal mammary tissues could significantly improve our ability to diagnose, treat, and prevent breast cancer.
Collapse
Affiliation(s)
- Rebecca B. Riggins
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Mary M. Mazzotta
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Omar Z. Maniya
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| | - Robert Clarke
- Lombardi Comprehensive Cancer Center and Department of Oncology, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
- Department of Physiology and Biophysics, Georgetown University School of Medicine, 3970 Reservoir Road NW, Washington, DC 20057, USA
| |
Collapse
|