1
|
Strømland PP, Bertelsen BE, Viste K, Chatziioannou AC, Bellerba F, Robinot N, Trolat A, Flågeng MH, Scalbert A, Keski-Rahkonen P, Sears DD, Bonanni B, Gandini S, Johansson H, Mellgren G. Effects of metformin on transcriptomic and metabolomic profiles in breast cancer survivors enrolled in the randomized placebo-controlled MetBreCS trial. Sci Rep 2025; 15:16897. [PMID: 40374694 DOI: 10.1038/s41598-025-01705-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 05/07/2025] [Indexed: 05/17/2025] Open
Abstract
Metformin reduces the incidence of breast cancer in patients with obesity and type 2 diabetes. However, our knowledge of the effects of metformin on breast cancer recurrence is limited. Within the randomized double-blind placebo-controlled phase II trial MetBreCS, we examined changes in breast tissue from breast cancer survivors with BMI > 25 kg/m2 after treatment with metformin. To identify metformin-regulated signaling pathways, we integrated the transcriptomic, metabolomic and steroid hormone profiles using bivariate and functional analyses. We identified MS4A1, HBA2, MT-RNR1, MT-RNR2, EGFL6 and FDCSP expression to be differentially expressed in breast tissues from metformin-treated postmenopausal women. The integration of transcriptomic and metabolomic profiles revealed down-regulation of immune response genes associated with reduced levels of arginine and citrulline in the metformin-treated group. The integration of transcriptomic and steroid hormone profiles showed an enrichment of steroid hormone biosynthesis and metabolism pathways with highly negatively correlated CYP11A1 and CYP1B1 expression in breast tissue from postmenopausal metformin-treated women. Our results indicate that postmenopausal breast cancer survivors treated with metformin have specific changes in breast tissue gene expression that may prevent the development of new tumors.Trial registration: MetBreCs trial is registered at European Union Clinical Trials Register (EudraCT Protocol # 2015-001001-14) on 07/10/2015.
Collapse
Affiliation(s)
- Pouda Panahandeh Strømland
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bjørn-Erik Bertelsen
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Kristin Viste
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | | | - Federica Bellerba
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Nivonirina Robinot
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Amarine Trolat
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Marianne Hauglid Flågeng
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Augustin Scalbert
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Pekka Keski-Rahkonen
- International Agency for Research on Cancer, Nutrition and Metabolism Branch, Lyon, France
| | - Dorothy D Sears
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Moores Cancer Center, University of California San Diego, La Jolla, San Diego, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Bernardo Bonanni
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Sara Gandini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Harriet Johansson
- Division of Cancer Prevention and Genetics, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway.
- Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
2
|
Mariana SM, Brenda RP, Heriberto PG, Cristina L, David B, Guadalupe ÁL. GPER1 activation by estrogenic compounds in the inflammatory profile of breast cancer cells. J Steroid Biochem Mol Biol 2025; 245:106639. [PMID: 39571822 DOI: 10.1016/j.jsbmb.2024.106639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Breast cancer (BC) is the most frequent female neoplasm worldwide. Its establishment and development have been related to inflammatory cytokine expression. Steroid hormones such as estradiol (E2) can regulate proinflammatory cytokine secretion through interaction with its nuclear receptors. However, little is known regarding the activation of its membrane estrogen receptor (GPER1) and the inflammatory cytokine environment in BC. We have studied the synthesis and biological effects of molecules analogs to E2 for hormone replacement therapy (HRT), such as pentolame. Nevertheless, its interaction with GPER1 and the modulation of inflammatory cytokines in different BC types has been barely studied and deserves deeper investigation. In this research, the role of GPER1 in the proliferation and modulation of inflammatory cytokines involved in carcinogenesis and metastatic processes in different BC cell lines was assessed by binding to various compounds. To achieve this goal, the presence of GPER1 was identified in different BC cell lines. Subsequently, cell proliferation after exposure to E2, pentolame and GPER1 agonist, G1, was subsequently determined alone or in combination with the GPER1 antagonist, G15. Finally, the pro-inflammatory cytokine secretion derived from the supernatants of BC cells exposed to the previous treatments was also assessed. Interestingly, GPER1 activation or inhibition has significant effects on the cytokine regulation associated with invasion in BC. Notably, pentolame did not induce cell proliferation or increase the proinflammatory cytokine expression compared to E2 in BC cell lines. In addition, pentolame did not induce the presence of the cell adhesion molecule PECAM-1. In contrast, E2 treatment weakly induced the expression of PECAM-1 in MCF-7 and HCC1937 cells, and G1 treatment showed this effect only in MCF-7 cells. The results suggest that GPER1 might be a significant inflammatory modulator with angiogenic-related effects in BC cells. In addition, pentolame might represent an HRT alternative in patients with BC predisposition.
Collapse
Affiliation(s)
- Segovia-Mendoza Mariana
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| | - Reyes-Plata Brenda
- Facultad de Estudios Superiores Zaragoza. Universidad Nacional Autónoma de México,Ciudad de México, Mexico
| | - Prado-Garcia Heriberto
- Laboratorio de Onco-Inmunobiología, Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias, "Ismael Cosio Villegas" Calzada de Tlalpan 4502, Col. Sección XVI, Ciudad de México 14080, Mexico
| | - Lemini Cristina
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Barrera David
- Departamento de Biología de la Reproducción "Dr. Carlos Gual Castro", Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Av. Vasco de Quiroga No. 15, Col. Belisario Domínguez, Sección XVI, Ciudad de México 14080, Mexico
| | - Ángeles-López Guadalupe
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico.
| |
Collapse
|
3
|
Sheva K, Roy Chowdhury S, Kravchenko-Balasha N, Meirovitz A. Molecular Changes in Breast Cancer Induced by Radiation Therapy. Int J Radiat Oncol Biol Phys 2024; 120:465-481. [PMID: 38508467 DOI: 10.1016/j.ijrobp.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024]
Abstract
PURPOSE Breast cancer treatments are based on prognostic clinicopathologic features that form the basis for therapeutic guidelines. Although the utilization of these guidelines has decreased breast cancer-associated mortality rates over the past three decades, they are not adequate for individualized therapy. Radiation therapy (RT) is the backbone of breast cancer treatment. Although a highly successful therapeutic modality clinically, from a biological perspective, preclinical studies have shown RT to have the potential to alter tumor cell phenotype, immunogenicity, and the surrounding microenvironment, potentially changing the behavior of cancer cells and resulting in a significant variation in RT response. This review presents the recent advances in revealing the complex molecular changes induced by RT in the treatment of breast cancer and highlights the complexities of translating this information into clinically relevant tools for improved prognostic insights and the revelation of novel approaches for optimizing RT. METHODS AND MATERIALS Current literature was reviewed with a focus on recent advances made in the elucidation of tumor-associated radiation-induced molecular changes across molecular, genetic, and proteomic bases. This review was structured with the aim of providing an up-to-date overview over the very broad and complex subject matter of radiation-induced molecular changes and radioresistance, familiarizing the reader with the broader issue at hand. RESULTS The subject of radiation-induced molecular changes in breast cancer has been broached from various physiological focal points including that of the immune system, immunogenicity and the abscopal effect, tumor hypoxia, breast cancer classification and subtyping, molecular heterogeneity, and molecular plasticity. It is becoming increasingly apparent that breast cancer clinical subtyping alone does not adequately account for variation in RT response or radioresistance. Multiple components of the tumor microenvironment and immune system, delivered RT dose and fractionation schedules, radiation-induced bystander effects, and intrinsic tumor physiology and heterogeneity all contribute to the resultant RT outcome. CONCLUSIONS Despite recent advances and improvements in anticancer therapies, tumor resistance remains a significant challenge. As new analytical techniques and technologies continue to provide crucial insight into the complex molecular mechanisms of breast cancer and its treatment responses, it is becoming more evident that personalized anticancer treatment regimens may be vital in overcoming radioresistance.
Collapse
Affiliation(s)
- Kim Sheva
- The Legacy Heritage Oncology Center & Dr Larry Norton Institute, Soroka University Medical Center, Ben Gurion University of the Negev, Faculty of Medicine, Be'er Sheva, Israel.
| | - Sangita Roy Chowdhury
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nataly Kravchenko-Balasha
- The Institute of Biomedical and Oral Research, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Amichay Meirovitz
- The Legacy Heritage Oncology Center & Dr Larry Norton Institute, Soroka University Medical Center, Ben Gurion University of the Negev, Faculty of Medicine, Be'er Sheva, Israel.
| |
Collapse
|
4
|
Anazco D, Acosta A, Cathcart-Rake EJ, D'Andre SD, Hurtado MD. Weight-centric prevention of cancer. OBESITY PILLARS 2024; 10:100106. [PMID: 38495815 PMCID: PMC10943063 DOI: 10.1016/j.obpill.2024.100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Background The link between excess adiposity and carcinogenesis has been well established for multiple malignancies, and cancer is one of the main contributors to obesity-related mortality. The potential role of different weight-loss interventions on cancer risk modification has been assessed, however, its clinical implications remain to be determined. In this clinical review, we present the data assessing the effect of weight loss interventions on cancer risk. Methods In this clinical review, we conducted a comprehensive search of relevant literature using MEDLINE, Embase, Web of Science, and Google Scholar databases for relevant studies from inception to January 20, 2024. In this clinical review, we present systematic reviews and meta-analysis, randomized clinical trials, and prospective and retrospective observational studies that address the effect of different treatment modalities for obesity in cancer risk. In addition, we incorporate the opinions from experts in the field of obesity medicine and oncology regarding the potential of weight loss as a preventative intervention for cancer. Results Intentional weight loss achieved through different modalities has been associated with a reduced cancer incidence. To date, the effect of weight loss on the postmenopausal women population has been more widely studied, with multiple reports indicating a protective effect of weight loss on hormone-dependent malignancies. The effect of bariatric interventions as a protective intervention for cancer has been studied extensively, showing a significant reduction in cancer incidence and mortality, however, data for the effect of bariatric surgery on certain specific types of cancer is conflicting or limited. Conclusion Medical nutrition therapy, exercise, antiobesity medication, and bariatric interventions, might lead to a reduction in cancer risk through weight loss-dependent and independent factors. Further evidence is needed to better determine which population might benefit the most, and the amount of weight loss required to provide a clinically significant preventative effect.
Collapse
Affiliation(s)
- Diego Anazco
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Maria D. Hurtado
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
5
|
Ruan GT, Deng L, Xie HL, Shi JY, Liu XY, Zheng X, Chen Y, Lin SQ, Zhang HY, Liu CA, Ge YZ, Song MM, Hu CL, Zhang XW, Yang M, Hu W, Cong MH, Zhu LC, Wang KH, Shi HP. Systemic inflammation and insulin resistance-related indicator predicts poor outcome in patients with cancer cachexia. Cancer Metab 2024; 12:3. [PMID: 38273418 PMCID: PMC10809764 DOI: 10.1186/s40170-024-00332-8] [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: 07/12/2023] [Accepted: 01/12/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The C-reactive protein (CRP)-triglyceride-glucose (TyG) index (CTI), which is a measure representing the level of inflammation and insulin resistance (IR), is related to poor cancer prognosis; however, the CTI has not been validated in patients with cancer cachexia. Thus, this study aimed to explore the potential clinical value of the CTI in patients with cancer cachexia. METHODS In this study, our prospective multicenter cohort included 1411 patients with cancer cachexia (mean age 59.45 ± 11.38, 63.3% male), which was a combined analysis of multiple cancer types. We randomly selected 30% of the patients for the internal test cohort (mean age 58.90 ± 11.22% 61.4% male). Additionally, we included 307 patients with cancer cachexia in the external validation cohort (mean age 61.16 ± 11, 58.5% male). Receiver operating characteristic (ROC) and calibration curves were performed to investigate the prognostic value of CTI. The prognostic value of the CTI was also investigated performing univariate and multivariate survival analyses. RESULTS The survival curve indicated that the CTI showed a significant prognostic value in the total, internal, and external validation cohorts. Prognostic ROC curves and calibration curves revealed that the CTI showed good consistency in predicting the survival of patients with cancer cachexia. Multivariate survival analysis showed that an elevated CTI increased the risk of death by 22% (total cohort, 95% confidence interval [CI] = 1.13-1.33), 34% (internal test cohort, 95%CI = 1.11-1.62), and 35% (external validation cohort, 95%CI = 1.14-1.59) for each increase in the standard deviation of CTI. High CTI reliably predicted shorter survival (total cohort, hazard ratio [HR] = 1.45, 95%CI = 1.22-1.71; internal test cohort, HR = 1.62, 95%CI = 1.12-2.36; external validation cohort, HR = 1.61, 95%CI = 1.15-2.26). High CTI significantly predicted shorter survival in different tumor subgroups, such as esophageal [HR = 2.11, 95%CI = 1.05-4.21] and colorectal cancer [HR = 2.29, 95%CI = 1.42-3.71]. The mediating effects analysis found that the mediating proportions of PGSGA, ECOG PS, and EORTC QLQ-C30 on the direct effects of CTI were 21.72%, 19.63%, and 11.61%, respectively We found that there was a significant positive correlation between the CTI and 90-day [HR = 2.48, 95%CI = 1.52-4.14] and 180-day mortality [HR = 1.77,95%CI = 1.24-2.55] in patients with cancer cachexia. CONCLUSION The CTI can predict the short- and long-term survival of patients with cancer cachexia and provide a useful prognostic tool for clinical practice.
Collapse
Affiliation(s)
- Guo-Tian Ruan
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Li Deng
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Hai-Lun Xie
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Jin-Yu Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Xiao-Yue Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Xin Zheng
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Yue Chen
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Shi-Qi Lin
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - He-Yang Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Chen-An Liu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Yi-Zhong Ge
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Meng-Meng Song
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Chun-Lei Hu
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Xiao-Wei Zhang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Ming Yang
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China
| | - Wen Hu
- Clinical Nutrition Department, Sichuan University West China Hospital, Chengdu, 610041, Sichuan, China
| | - Ming-Hua Cong
- Comprehensive Oncology Department, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100038, China
| | - Li-Chen Zhu
- Department of Immunology, School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, China
| | - Kun-Hua Wang
- Yunnan University, Kunming, 650091, China
- General Surgery Clinical Medical Center of Yunnan Province, Kunming, 650032, China
| | - Han-Ping Shi
- Department of Gastrointestinal Surgery/Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China.
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, 10 Tie Yi Road, Beijing, 100053, China.
- Key Laboratory of Cancer FSMP for State Market Regulation, 10 Tie Yi Road, Beijing, 100038, China.
- Laboratory for Clinical Medicine, Capital Medical University, 10 Tie Yi Road, Beijing, 100038, China.
| |
Collapse
|
6
|
Nguyen HL, Geukens T, Maetens M, Aparicio S, Bassez A, Borg A, Brock J, Broeks A, Caldas C, Cardoso F, De Schepper M, Delorenzi M, Drukker CA, Glas AM, Green AR, Isnaldi E, Eyfjörð J, Khout H, Knappskog S, Krishnamurthy S, Lakhani SR, Langerod A, Martens JWM, McCart Reed AE, Murphy L, Naulaerts S, Nik-Zainal S, Nevelsteen I, Neven P, Piccart M, Poncet C, Punie K, Purdie C, Rakha EA, Richardson A, Rutgers E, Vincent-Salomon A, Simpson PT, Schmidt MK, Sotiriou C, Span PN, Tan KTB, Thompson A, Tommasi S, Van Baelen K, Van de Vijver M, Van Laere S, Van't Veer L, Viale G, Viari A, Vos H, Witteveen AT, Wildiers H, Floris G, Garg AD, Smeets A, Lambrechts D, Biganzoli E, Richard F, Desmedt C. Obesity-associated changes in molecular biology of primary breast cancer. Nat Commun 2023; 14:4418. [PMID: 37479706 PMCID: PMC10361985 DOI: 10.1038/s41467-023-39996-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 07/06/2023] [Indexed: 07/23/2023] Open
Abstract
Obesity is associated with an increased risk of developing breast cancer (BC) and worse prognosis in BC patients, yet its impact on BC biology remains understudied in humans. This study investigates how the biology of untreated primary BC differs according to patients' body mass index (BMI) using data from >2,000 patients. We identify several genomic alterations that are differentially prevalent in overweight or obese patients compared to lean patients. We report evidence supporting an ageing accelerating effect of obesity at the genetic level. We show that BMI-associated differences in bulk transcriptomic profile are subtle, while single cell profiling allows detection of more pronounced changes in different cell compartments. These analyses further reveal an elevated and unresolved inflammation of the BC tumor microenvironment associated with obesity, with distinct characteristics contingent on the estrogen receptor status. Collectively, our analyses imply that obesity is associated with an inflammaging-like phenotype. We conclude that patient adiposity may play a significant role in the heterogeneity of BC and should be considered for BC treatment tailoring.
Collapse
Affiliation(s)
- Ha-Linh Nguyen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Tatjana Geukens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marion Maetens
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Samuel Aparicio
- Department of Molecular Oncology, BC Cancer Research Centre, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Ayse Bassez
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Ake Borg
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
- Lund University Cancer Center Lund, Lund, Sweden
- CREATE Health Strategic Centre for Translational Cancer Research, Lund University, Lund, Sweden
- Department of Clinical Sciences, SCIBLU Genomics, Lund University, Lund, Sweden
| | - Jane Brock
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Annegien Broeks
- Departments of Core Facility, Molecular Pathology and Biobanking, Antoni van Leeuwenhoek, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute and Department of Oncology, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Fatima Cardoso
- Breast Unit, Champalimaud Clinical Center/Champalimaud Foundation, Lisbon, Portugal
| | - Maxim De Schepper
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Mauro Delorenzi
- Department of Oncology, University of Lausanne, Epalinges, Switzerland
- SIB Swiss Institute of Bioinformatics, Bioinformatics Core Facility, Lausanne, Switzerland
| | - Caroline A Drukker
- Department of Surgical Oncology, Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | | | - Andrew R Green
- Nottingham Breast Cancer Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Edoardo Isnaldi
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jórunn Eyfjörð
- BioMedical Center, School of Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Hazem Khout
- Department of Breast Surgery, Glenfield Hospital, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Stian Knappskog
- Department of Clinical Science, Faculty of Medicine, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Savitri Krishnamurthy
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sunil R Lakhani
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
- Pathology Queensland, The Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Anita Langerod
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Ullernchausseen, Oslo, Norway
| | - John W M Martens
- Department of Medical Oncology and Cancer Genomics Netherlands, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Amy E McCart Reed
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Leigh Murphy
- University of Manitoba and Cancer Care Manitoba Research Institute, Winnipeg, MB, Canada
| | - Stefan Naulaerts
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Serena Nik-Zainal
- Department of Medical Genetics, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- MRC Cancer Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Ines Nevelsteen
- Department of Surgical Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Patrick Neven
- Department of Gynecological Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Martine Piccart
- Institut Jules Bordet and Université Libre de Bruxelles, Brussels, Belgium
| | - Coralie Poncet
- European Organisation for Research and Treatment of Cancer (EORTC) Headquarters, Brussels, Belgium
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Unit, Leuven Cancer Institute and University Hospitals Leuven, Leuven, Belgium
| | - Colin Purdie
- Department of Pathology, University of Dundee, NHS Tayside, Dundee, UK
| | - Emad A Rakha
- Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Histopathology, Nottingham University Hospital NHS Trust, City Hospital Campus, Nottingham, UK
| | | | - Emiel Rutgers
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Anne Vincent-Salomon
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL Research University, Paris, France
| | - Peter T Simpson
- UQ Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, the Netherlands
| | - Christos Sotiriou
- Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Paul N Span
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Kiat Tee Benita Tan
- Department of General Surgery, Sengkang General Hospital, Singapore, Singapore
- Department of Breast Surgery, Singapore General Hospital, Singapore, Singapore
- Department of Breast Surgery, National Cancer Centre, Singapore, Singapore
| | - Alastair Thompson
- Department of Surgery, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumouri "Giovanni Paolo II", Bari, Italy
| | - Karen Van Baelen
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Marc Van de Vijver
- Department of Pathology, Amsterdam University Medical Centers, Cancer Center Amsterdam, University of Amsterdam, Amsterdam, the Netherlands
| | - Steven Van Laere
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, Antwerp, Belgium
| | - Laura Van't Veer
- Department of Laboratory Medicine, UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Giuseppe Viale
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Alain Viari
- Synergie Lyon Cancer, Plateforme de Bio-informatique 'Gilles Thomas', Lyon, France
| | - Hanne Vos
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | | | - Hans Wildiers
- Department of General Medical Oncology and Multidisciplinary Breast Unit, Leuven Cancer Institute and University Hospitals Leuven, Leuven, Belgium
| | - Giuseppe Floris
- Department of Pathology, University Hospitals Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Ann Smeets
- Department of Surgical Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Elia Biganzoli
- Unit of Medical Statistics, Biometry and Epidemiology, Department of Biomedical and Clinical Sciences (DIBIC) "L. Sacco" & DSRC, LITA Vialba campus, Università degli Studi di Milano, Milan, Italy
| | - François Richard
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Christine Desmedt
- Laboratory for Translational Breast Cancer Research, Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
7
|
Silva C, Andrade N, Guimarães JT, Cardoso E, Meireles C, Pinto V, Paiva J, Martel F. The pro-proliferative effect of insulin in human breast epithelial DMBA-transformed and non-transformed cell lines is PI3K-, mTOR- and GLUT1-dependent. Cell Biochem Funct 2022; 40:127-137. [PMID: 35014047 DOI: 10.1002/cbf.3681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/03/2021] [Indexed: 11/06/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is linked to an increased risk of breast cancer. We aimed to investigate how T2DM-associated characteristics (high levels of glucose, insulin, leptin, inflammatory mediators and oxidative stress) influence breast cancer carcinogenesis, in DMBA-treated (MCF-12ADMBA ) and non-treated breast epithelial (MCF-12A) cell lines. Insulin (50 nM) promotes cell proliferation, 3 H-DG uptake and lactic acid production in both cell lines. The stimulatory effects of insulin upon cell proliferation and 3 H-DG uptake were hampered by rapamycin, LY294001 and BAY-876, in both cell lines. In conclusion, hyperinsulinemia, one important characteristic of T2DM, contributes to the initiation of breast cancer by a PI3K- and mTOR-dependent mechanism involving increased GLUT1-mediated glucose uptake. SIGNIFICANCE: The pro-proliferative effect of insulin in human breast epithelial DMBA-transformed and non-transformed cell lines is PI3K-, mTOR- and GLUT1-dependent.
Collapse
Affiliation(s)
- Cláudia Silva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Nelson Andrade
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal.,REQUIMTE/LAQV, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - João Tiago Guimarães
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Clinical Pathology, São João Hospital Centre, Porto, Portugal.,Institute of Public Health, University of Porto, Porto, Portugal
| | - Emília Cardoso
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Catarina Meireles
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| | - Vanessa Pinto
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,iLoF, Intelligent Lab on Fiber, Limited, Oxford, UK
| | - Joana Paiva
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,iLoF, Intelligent Lab on Fiber, Limited, Oxford, UK.,Instituto de Ciências Biomédicas Abel Salazar, University of Porto, Porto, Portugal
| | - Fátima Martel
- Unit of Biochemistry, Department of Biomedicine, Faculty of Medicine, University of Porto, Porto, Portugal.,Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
| |
Collapse
|
8
|
Sohouli MH, Hadizadeh M, Omrani M, Baniasadi M, Sanati V, Zarrati M. Adherence to Lifelines Diet Score (LLDS) Is Associated with a Reduced Risk of Breast Cancer (BrCa): A Case-Control Study. Int J Clin Pract 2022; 2022:7726126. [PMID: 35685489 PMCID: PMC9159231 DOI: 10.1155/2022/7726126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/20/2021] [Accepted: 12/14/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Previous evidence suggests a link between diet quality and breast cancer (BrCa); however, the link between the Lifelines Diet Score (LLDS)-a fully food-based score that uses the 2015 Dutch Dietary Guidelines-and risk of BrCa has not yet been evaluated. Therefore, the aim of this study was to observe the relationship between adherence to an LLDS and risk of BrCa in Iranian adults. METHODS In the hospital-based case-control study, 253 patients with BrCa and 267 non-BrCa controls were enrolled. Individual's food consumption was recorded to calculate LLDS using a semiquantitative food frequency questionnaire. In adjusted models, the association between the inflammatory potential of the diet and the risk of BrCa was estimated by using binary logistic regression. RESULTS Compared with control individuals, BrCa patients significantly had higher waist circumference (WC), first pregnancy age, abortion history, and number of children. In addition, the mean intake of vitamin D supplements and anti-inflammatory drugs in the case group was significantly lower than the control group. Furthermore, after adjusted potential confounders, individuals in the highest vs. lowest quartiles of LLDS showed statistically significant lower risk of BrCa in overall population (OR: 0.21; 95% CI: 0.11-0.43; P trend <0.001), premenopausal (OR: 0.26; 95% CI: 0.10-0.68; P trend = 0.003), and post-menopausal women (OR: 0.20; 95% CI: 0.06-0.60; P trend = 0.015). CONCLUSION Findings of this study reflected that higher LLDS decreased risk of BrCa, but need further investigation in later studies.
Collapse
Affiliation(s)
- Mohammad Hassan Sohouli
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadizadeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Omrani
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mansoureh Baniasadi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Sanati
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Zarrati
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
Calcineurin regulates the stability and activity of estrogen receptor α. Proc Natl Acad Sci U S A 2021; 118:2114258118. [PMID: 34711683 DOI: 10.1073/pnas.2114258118] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 08/27/2021] [Indexed: 12/14/2022] Open
Abstract
Estrogen receptor α (ER-α) mediates estrogen-dependent cancer progression and is expressed in most breast cancer cells. However, the molecular mechanisms underlying the regulation of the cellular abundance and activity of ER-α remain unclear. We here show that the protein phosphatase calcineurin regulates both ER-α stability and activity in human breast cancer cells. Calcineurin depletion or inhibition down-regulated the abundance of ER-α by promoting its polyubiquitination and degradation. Calcineurin inhibition also promoted the binding of ER-α to the E3 ubiquitin ligase E6AP, and calcineurin mediated the dephosphorylation of ER-α at Ser294 in vitro. Moreover, the ER-α (S294A) mutant was more stable and activated the expression of ER-α target genes to a greater extent compared with the wild-type protein, whereas the extents of its interaction with E6AP and polyubiquitination were attenuated. These results suggest that the phosphorylation of ER-α at Ser294 promotes its binding to E6AP and consequent degradation. Calcineurin was also found to be required for the phosphorylation of ER-α at Ser118 by mechanistic target of rapamycin complex 1 and the consequent activation of ER-α in response to β-estradiol treatment. Our study thus indicates that calcineurin controls both the stability and activity of ER-α by regulating its phosphorylation at Ser294 and Ser118 Finally, the expression of the calcineurin A-α gene (PPP3CA) was associated with poor prognosis in ER-α-positive breast cancer patients treated with tamoxifen or other endocrine therapeutic agents. Calcineurin is thus a promising target for the development of therapies for ER-α-positive breast cancer.
Collapse
|
10
|
Dittmer J. Nuclear Mechanisms Involved in Endocrine Resistance. Front Oncol 2021; 11:736597. [PMID: 34604071 PMCID: PMC8480308 DOI: 10.3389/fonc.2021.736597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/26/2021] [Indexed: 12/27/2022] Open
Abstract
Endocrine therapy is a standard treatment offered to patients with ERα (estrogen receptor α)-positive breast cancer. In endocrine therapy, ERα is either directly targeted by anti-estrogens or indirectly by aromatase inhibitors which cause estrogen deficiency. Resistance to these drugs (endocrine resistance) compromises the efficiency of this treatment and requires additional measures. Endocrine resistance is often caused by deregulation of the PI3K/AKT/mTOR pathway and/or cyclin-dependent kinase 4 and 6 activities allowing inhibitors of these factors to be used clinically to counteract endocrine resistance. The nuclear mechanisms involved in endocrine resistance are beginning to emerge. Exploring these mechanisms may reveal additional druggable targets, which could help to further improve patients' outcome in an endocrine resistance setting. This review intends to summarize our current knowledge on the nuclear mechanisms linked to endocrine resistance.
Collapse
Affiliation(s)
- Jürgen Dittmer
- Clinic for Gynecology, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
11
|
Ortega MA, Fraile-Martínez O, Saez MA, Álvarez-Mon MA, Gómez-Lahoz AM, Bravo C, Luis JADL, Sainz F, Coca S, Asúnsolo Á, Monserrat J, Guijarro LG, Álvarez-Mon M, Bujan J, García-Honduvilla N. Abnormal proinflammatory and stressor environmental with increased the regulatory cellular IGF-1/PAPP-A/STC and Wnt-1/β-Catenin canonical pathway in placenta of women with Chronic venous Disease during Pregnancy. Int J Med Sci 2021; 18:2814-2827. [PMID: 34220309 PMCID: PMC8241785 DOI: 10.7150/ijms.58992] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022] Open
Abstract
Lower limbs venous insufficiency refers to a wide variety of venous disorders grouped by the term of chronic venous disease (CVD). Hemodynamic and hormonal changes related to pregnancy period, may promote the development of CVD affecting approximately 1 in 3 women. It has been shown that the presence of this condition is associated with damage and placental suffering. Thus, taking IGF-1/PAPP-A/STC-2, inflammatory cytokines production, PI3K/Akt and Wnt/ β-catenin pathways as a part of the alterations that occurs in the placenta due to CVD, the aim of this study will be to examine the main components of these pathways. Genic and protein expression of PAPP-A, STC-2, IGF-1, IRS-4 Wnt-1, β-catenin, c-myc, Cyclin D1, IL-4/IL-6 and PI3K/Akt/mTOR pathway will be analysed through RT-qPCR and immunohistochemical techniques in women with CVD (n=62) and pregnant women without this condition (HC) (n=52). PAPP-A, IGF-1, IL-4, IL-6, IRS-4, PI3K, Akt, mTOR, Wnt-1, β-catenin, c-myc and Cyclin D1 expression were found to be increased in women with CVD, whereas STC-2 were decreased in this group, compared to non-affected women. Our study has demonstrated that IGF-1/PAPP-A/STC-2 axis, PI3K/Akt and Wnt/β-catenin pathways, along with c-myc, Cyclin D1 and inflammatory cytokines are altered in placenta women with CVD. These results extent the knowledge that CVD is associated to a placenta damage with abnormal tissue environment and cellular regulation.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, 28806 Alcalá de Henares, Spain.,University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Miguel A Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, 28801 Alcalá de Henares, Madrid, Spain
| | - Miguel A Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Ana M Gómez-Lahoz
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Coral Bravo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,Service of Gynecology and Obstetrics, Central University Hospital of Defense-UAH, Madrid, Spain
| | - Juan A De León Luis
- Department of Public and Maternal and Child Health, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain.,Department of Obstetrics and Gynecology, University Hospital Gregorio Marañón, Madrid 28009, Spain.,Health Research Institute Gregorio Marañón, 28009 Madrid, Spain
| | - Felipe Sainz
- Angiology and Vascular Surgery Unit, Central University Hospital of Defense-UAH, Madrid, Spain.,Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Santiago Coca
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain
| | - Luis G Guijarro
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,Unit of Biochemistry and Molecular Biology (CIBEREHD), Department of System Biology, University of Alcalá, 28801 Alcalá de Henares, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain.,Immune System Diseases-Rheumatology, Oncology Service an Internal Medicine, University Hospital Príncipe de Asturias, (CIBEREHD), 28806 Alcalá de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcalá de Henares, Spain.,Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain.,University Center for the Defense of Madrid (CUD-ACD), 28047 Madrid, Spain
| |
Collapse
|
12
|
Insulin-like growth factor-I rescue of primary keratinocytes from pre- and post-ultraviolet B radiation effects. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 209:111951. [DOI: 10.1016/j.jphotobiol.2020.111951] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 01/13/2023]
|
13
|
Xu S, Yu S, Dong D, Lee LTO. G Protein-Coupled Estrogen Receptor: A Potential Therapeutic Target in Cancer. Front Endocrinol (Lausanne) 2019; 10:725. [PMID: 31708873 PMCID: PMC6823181 DOI: 10.3389/fendo.2019.00725] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 10/09/2019] [Indexed: 12/13/2022] Open
Abstract
The G protein-coupled estrogen receptor (GPER) is a seven-transmembrane-domain receptor that mediates non-genomic estrogen related signaling. After ligand activation, GPER triggers multiple downstream pathways that exert diverse biological effects on the regulation of cell growth, migration and programmed cell death in a variety of tissues. A significant correlation between GPER and the progression of multiple cancers has likewise been reported. Therefore, a better understanding of the role GPER plays in cancer biology may lead to the identification of novel therapeutic targets, especially among estrogen-related cancers. Here, we review cell signaling and detail the functions of GPER in malignancies.
Collapse
Affiliation(s)
- Shen Xu
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shan Yu
- Faculty of Health Sciences, Centre of Reproduction Development and Aging, University of Macau, Macau, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China
| | - Daming Dong
- Department of Orthopedics, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Leo Tsz On Lee
- Faculty of Health Sciences, Centre of Reproduction Development and Aging, University of Macau, Macau, China
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, China
| |
Collapse
|
14
|
Muñoz E, Domingo J, De Castro G, Lorenzo I, García-Velasco JA, Bellver J, Pellicer A, Garrido N. Ovarian stimulation for oocyte vitrification does not modify disease-free survival and overall survival rates in patients with early breast cancer. Reprod Biomed Online 2019; 39:860-867. [PMID: 31564650 DOI: 10.1016/j.rbmo.2019.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/03/2019] [Accepted: 07/05/2019] [Indexed: 11/28/2022]
Abstract
RESEARCH QUESTION Does ovarian stimulation for oocyte vitrification affect disease-free survival and overall survival rates in women with early breast cancer? DESIGN This cohort study included 259 patients with early breast cancer; 148 patients underwent ovarian stimulation, whereas 111 patients did not. Patients were treated between January 2008 and December 2016. To calculate the disease-free survival time and overall survival rate, the time of definitive surgery was defined as the starting point. The follow-up was conducted up to 5 years. RESULTS Exposed and non-exposed groups were comparable in tumour, node and metastases classification, Nottingham grade, hormonal receptor status, tumour molecular phenotype, histology and pathology stage. The exposed group was younger than the non-exposed. Recurrences occurred in 9/148 women (6.1%) in the exposed group and 15/111 women (13.5%) in the non-exposed group, with no significant difference. The mean disease-free survival time was 63.9 months (95% confidence interval [CI]: 61.5-66.4) in the exposed group and 60.6 months (95% CI: 56.9-64.2) in the non-exposed, with no significant difference (log-rank [Mantel-Cox] test). Overall survival rates were comparable; 2/148 (1.4%) and 4/111 (3.6%) patients died, in exposed and non-exposed groups, respectively, during the period analysed. Mean overall survival times were 67.2 months (95% CI: 66.2-68.2) in the exposed group and 65.9 months (95% CI: 64.0-67.9) in the unexposed, with no significant difference (log-rank [Mantel-Cox] test). CONCLUSIONS This study suggests that ovarian stimulation in patients with early-stage breast cancer is safe in the long term.
Collapse
Affiliation(s)
- Elkin Muñoz
- IVIRMA Vigo, Plaza Francisco Fernández del Riego, 7 36203, Vigo Pontevedra, Spain.
| | | | | | | | | | - Jose Bellver
- Instituto Universitario IVI Valencia, Universidad de Valencia; IVI Foundation, Valencia
| | - Antonio Pellicer
- IVI Foundation, Valencia; Instituto de Investigación Sanitaria La Fe, Valencia, Spain; IVIRMA Rome
| | - Nicolás Garrido
- IVI Foundation, Valencia; Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
15
|
Insulin, estradiol levels and body mass index in pre- and post-menopausal women with breast cancer. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2019. [DOI: 10.1016/j.jrras.2015.07.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
16
|
Engin AB, Engin A, Gonul II. The effect of adipocyte-macrophage crosstalk in obesity-related breast cancer. J Mol Endocrinol 2019; 62:R201-R222. [PMID: 30620711 DOI: 10.1530/jme-18-0252] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 01/07/2019] [Indexed: 12/11/2022]
Abstract
Adipose tissue is the primary source of many pro-inflammatory cytokines in obesity. Macrophage numbers and pro-inflammatory gene expression are positively associated with adipocyte size. Free fatty acid and tumor necrosis factor-α involve in a vicious cycle between adipocytes and macrophages aggravating inflammatory changes. Thereby, M1 macrophages form a characteristic 'crown-like structure (CLS)' around necrotic adipocytes in obese adipose tissue. In obese women, CLSs of breast adipose tissue are responsible for both increase in local aromatase activity and aggressive behavior of breast cancer cells. Interlinked molecular mechanisms between adipocyte-macrophage-breast cancer cells in obesity involve seven consecutive processes: Excessive release of adipocyte- and macrophage-derived inflammatory cytokines, TSC1-TSC2 complex-mTOR crosstalk, insulin resistance, endoplasmic reticulum (ER) stress and excessive oxidative stress generation, uncoupled respiration and hypoxia, SIRT1 controversy, the increased levels of aromatase activity and estrogen production. Considering elevated risks of estrogen receptor (E2R)-positive postmenopausal breast cancer growth in obesity, adipocyte-macrophage crosstalk is important in the aforementioned issues. Increased mTORC1 signaling in obesity ensures the strong activation of oncogenic signaling in E2Rα-positive breast cancer cells. Since insulin and insulin-like growth factors have been identified as tumor promoters, hyperinsulinemia is an independent risk factor for poor prognosis in breast cancer despite peripheral insulin resistance. The unpredictable effects of adipocyte-derived leptin-estrogen-macrophage axis, and sirtuin 1 (SIRT1)-adipose-resident macrophage axis in obese postmenopausal patients with breast cancer are unresolved mechanistic gaps in the molecular links between the tumor growth and adipocytokines.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Department of Toxicology, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Atilla Engin
- Department of General Surgery, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Ipek Isik Gonul
- Department of Pathology, Faculty of Medicine, Gazi University, Ankara, Turkey
| |
Collapse
|
17
|
Efficacy and safety of the combination of metformin, everolimus and exemestane in overweight and obese postmenopausal patients with metastatic, hormone receptor-positive, HER2-negative breast cancer: a phase II study. Invest New Drugs 2019; 37:345-351. [PMID: 30610588 DOI: 10.1007/s10637-018-0700-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023]
Abstract
Background Increased adiposity is thought to result in worse clinical outcomes in patients with breast cancer through increased estrogen production, hyperinsulinemia, insulin resistance, and activation of the phosphatidylinositol-3-kinase/AKT/mammalian target of rapamycin (mTOR) pathway. Thus, we hypothesized that the addition of metformin to everolimus and exemestane, could lead to better outcomes in overweight and obese patients with metastatic, hormone receptor-positive, HER2-negative breast cancer. We conducted a phase II trial to evaluate the efficacy and safety of the combination of metformin, everolimus and exemestane in overweight and obese postmenopausal women with metastatic, hormone receptor-positive, HER2-negative breast cancer. Methods Twenty-two patients with a body mass index ≥25 kg/m2 were treated with metformin 1000 mg twice daily, everolimus 10 mg daily and exemestane 25 mg daily. Median progression-free (PFS) and overall survival (OS) were estimated using the Kaplan-Meier method. Results Median PFS and OS were 6.3 months (95% confidence interval [CI]: 3.8-11.3 months) and 28.8 months (95% CI: 17.5-59.7 months), respectively. Five patients had a partial response and 7 had stable disease for ≥24 weeks yielding a clinical benefit rate of 54.5%. Compared with overweight patients, obese patients had an improved PFS on univariable (p = 0.015) but not multivariable analysis (p = 0.215). Thirty-two percent of patients experienced a grade 3 treatment-related adverse event (TRAE). There were no grade 4 TRAEs and 7 patients experienced a grade 3 TRAE. Conclusion The combination of metformin, everolimus and exemestane was safe and had moderate clinical benefit in overweight and obese with patients metastatic, hormone receptor-positive, HER2-negative breast cancer.
Collapse
|
18
|
Insulin-like growth factor type I selectively binds to G-quadruplex structures. Biochim Biophys Acta Gen Subj 2018; 1863:31-38. [PMID: 30278241 DOI: 10.1016/j.bbagen.2018.09.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 11/20/2022]
Abstract
BACKGROUND G-quadruplex has been viewed as a promising therapeutic target in oncology due to its potentially important roles in physiological and pathological processes. Emerging evidence suggests that the biological functions of G-quadruplexes are closely related to the binding of some proteins. Insulin-like growth factor type I (IGF-1), as a significant modulator of cell growth and development, may serve as a quadruplex-binding protein. METHODS The binding affinity and selectivity of IGF-1 to different DNA motifs in solution were measured by using fluorescence spectroscopy, Surface Plasmon Resonance (SPR), and force-induced remnant magnetization (FIRM). The effects of IGF-1 on the formation and stability of G-quadruplex structures were evaluated by circular dichroism (CD) and melting fluorescence resonance energy transfer (FRET) spectroscopy. The influence of quadruplex-specific ligands on the binding of G-quadruplexes with IGF-1 was determined by FIRM. RESULTS IGF-1 shows a binding specificity for G-quadruplex structures, especially the G-quadruplex structure with a parallel topology. The quadruplex-specific ligands TMPyP4 and PDS (Pyridostatin) can inhibit the interaction between G-quadruplexes and proteins. CONCLUSIONS IGF-1 is demonstrated to selectively bind with G-quadruplex structures. The use of quadruplex-interactive ligands could modulate the binding of IGF-1 to G-quadruplexes. GENERAL SIGNIFICANCE This study provides us with a new perspective to understand the possible physiological relationship between IGF-1 and G-quadruplexes and also conveys a strategy to regulate the interaction between G-quadruplex DNA and proteins.
Collapse
|
19
|
Rong C, Meinert ÉFRC, Hess J. Estrogen Receptor Signaling in Radiotherapy: From Molecular Mechanisms to Clinical Studies. Int J Mol Sci 2018; 19:ijms19030713. [PMID: 29498642 PMCID: PMC5877574 DOI: 10.3390/ijms19030713] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Revised: 02/02/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022] Open
Abstract
Numerous studies have established a proof of concept that abnormal expression and function of estrogen receptors (ER) are crucial processes in initiation and development of hormone-related cancers and also affect the efficacy of anti-cancer therapy. Radiotherapy has been applied as one of the most common and potent therapeutic strategies, which is synergistic with surgical excision, chemotherapy and targeted therapy for treating malignant tumors. However, the impact of ionizing radiation on ER expression and ER-related signaling in cancer tissue, as well as the interaction between endocrine and irradiation therapy remains largely elusive. This review will discuss recent findings on ER and ER-related signaling, which are relevant for cancer radiotherapy. In addition, we will summarize pre-clinical and clinical studies that evaluate the consequences of anti-estrogen and irradiation therapy in cancer, including emerging studies on head and neck cancer, which might improve the understanding and development of novel therapeutic strategies for estrogen-related cancers.
Collapse
Affiliation(s)
- Chao Rong
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
| | - Étienne Fasolt Richard Corvin Meinert
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
- Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Jochen Hess
- Section Experimental and Translational Head and Neck Oncology, Department of Otolaryngology, Head and Neck Surgery, University Hospital Heidelberg, 69120 Heidelberg, Germany.
- Research Group Molecular Mechanisms of Head and Neck Tumors, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
20
|
Wei ML, Duan P, Wang ZM, Ding M, Tu P. High glucose and high insulin conditions promote MCF‑7 cell proliferation and invasion by upregulating IRS1 and activating the Ras/Raf/ERK pathway. Mol Med Rep 2017; 16:6690-6696. [PMID: 28901503 PMCID: PMC5865785 DOI: 10.3892/mmr.2017.7420] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 04/04/2017] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus is associated with an increased risk of breast cancer, but the molecular mechanism underlying this association remains unclear. The aim of the present study was to investigate the effect of high glucose and high insulin conditions on MCF‑7 breast cancer cells and to elucidate the molecular mechanisms underlying these effects. High glucose and high insulin conditions resulted in increased viability, proliferation, and invasion in MCF‑7 cells compared with normal glucose and low insulin conditions. Reverse transcription‑quantitative polymerase chain reaction and western blot analyses revealed that insulin receptor substrate 1 (IRS1) was significantly upregulated following high glucose and high insulin treatment compared with normal glucose and low insulin conditions. Furthermore, high glucose and high insulin treatment increased the Ras family of proto‑oncogenes (Ras) and RAF1 proto‑oncogene (Raf‑1) protein expression, and activated the phosphorylation of extracellular signal‑regulated kinase (ERK) 1/2. These findings suggest that high glucose and high insulin conditions promoted the proliferation and invasion of MCF‑7 cells by upregulating IRS1 and activating the Ras/Raf/ERK pathway.
Collapse
Affiliation(s)
- Mei-Lin Wei
- Department of Endocrinology and Metabolism, The Third Hospital of Nanchang, Nanchang Key Laboratory of Diabetes, Nanchang, Jiangxi 330009, P.R. China
| | - Peng Duan
- Department of Endocrinology and Metabolism, The Third Hospital of Nanchang, Nanchang Key Laboratory of Diabetes, Nanchang, Jiangxi 330009, P.R. China
| | - Zhi-Ming Wang
- Department of Endocrinology and Metabolism, The Third Hospital of Nanchang, Nanchang Key Laboratory of Diabetes, Nanchang, Jiangxi 330009, P.R. China
| | - Miao Ding
- Department of Endocrinology and Metabolism, The Third Hospital of Nanchang, Nanchang Key Laboratory of Diabetes, Nanchang, Jiangxi 330009, P.R. China
| | - Ping Tu
- Department of Endocrinology and Metabolism, The Third Hospital of Nanchang, Nanchang Key Laboratory of Diabetes, Nanchang, Jiangxi 330009, P.R. China,Correspondence to: Professor Ping Tu, Department of Endocrinology and Metabolism, The Third Hospital of Nanchang, Nanchang Key Laboratory of Diabetes, 2 South Xiangshan Road, Nanchang, Jiangxi 330009, P.R. China, E-mail:
| |
Collapse
|
21
|
Kabat GC, Kim MY, Lee JS, Ho GY, Going SB, Beebe-Dimmer J, Manson JE, Chlebowski RT, Rohan TE. Metabolic Obesity Phenotypes and Risk of Breast Cancer in Postmenopausal Women. Cancer Epidemiol Biomarkers Prev 2017; 26:1730-1735. [PMID: 28939589 DOI: 10.1158/1055-9965.epi-17-0495] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/09/2017] [Accepted: 09/18/2017] [Indexed: 12/27/2022] Open
Abstract
Background: Obesity and the metabolic syndrome (MetS) have both been linked to increased risk of postmenopausal breast cancer; however, their relative contributions are poorly understood.Methods: We examined the association of metabolic phenotypes of obesity defined by presence of the MetS (yes and no) and body mass index (BMI; normal, overweight, obese) with risk of postmenopausal breast cancer in a prospective analysis of a cohort of postmenopausal women (n ∼ 21,000) with baseline measurements of blood glucose, triglycerides, HDL-cholesterol, blood pressure, waist circumference, and BMI. Women were classified into 6 metabolic obesity phenotypes according to their BMI (18.5-<25.0, 25.0-<30.0, ≥30.0 kg/m2) and presence of the MetS (≥3 of the following: waist circumference ≥88 cm, triglycerides ≥150 mg/dL, HDL-C <50 mg/dL, glucose ≥100 mg/dL, and systolic/diastolic blood pressure ≥130/85 mmHg or treatment for hypertension). HRs for incident breast cancer and 95% confidence intervals (95% CI) were estimated using Cox proportional hazards models.Results: Over 15 years of follow-up, 1,176 cases of invasive breast cancer were diagnosed. Obesity, regardless of metabolic health, was associated with increased risk of breast cancer. Being obese and metabolically unhealthy was associated with the highest risk: HR, 1.62; 95% CI, 1.33-1.96. These associations were stronger in women who had never used hormone therapy.Conclusions: Our findings suggest that both obesity and metabolic dysregulation are associated with breast cancer risk.Impact: Beyond BMI, metabolic health should be considered a clinically relevant and modifiable risk factor for breast cancer. Cancer Epidemiol Biomarkers Prev; 26(12); 1730-5. ©2017 AACR.
Collapse
Affiliation(s)
- Geoffrey C Kabat
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York.
| | - Mimi Y Kim
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Jennifer S Lee
- Departments of Medicine and Health Research and Policy, Stanford University Medical Center, Stanford, California.,Palo Alto VA Health Care System, Stanford, California
| | - Gloria Y Ho
- Department of Occupational Medicine, Epidemiology and Prevention, Feinstein Institute for Medical Research, Hofstra Northwell School of Medicine, Great Neck, New York
| | - Scott B Going
- Department of Nutrition Sciences, University of Arizona, Tucson, Arizona
| | - Jennifer Beebe-Dimmer
- Department of Oncology, Wayne State University School of Medicine, Detroit, Michigan
| | - JoAnn E Manson
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Thomas E Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
22
|
Tuna BG, Atalay PB, Altunbek M, Kalkan BM, Dogan S. Effects of Chronic and Intermittent Calorie Restriction on Adropin Levels in Breast Cancer. Nutr Cancer 2017; 69:1003-1010. [DOI: 10.1080/01635581.2017.1359314] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Bilge G. Tuna
- Department of Biophysics, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Pinar B. Atalay
- Department of Medical Biology, School of Medicine, Maltepe University, Istanbul, Turkey
| | - Mine Altunbek
- Department of Genetics and Engineering, Faculty of Engineering, Yeditepe University, Istanbul, Turkey
| | - Batuhan M. Kalkan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkey
| | - Soner Dogan
- Department of Medical Biology, School of Medicine, Yeditepe University, Istanbul, Turkey
| |
Collapse
|
23
|
Unar-Munguía M, Torres-Mejía G, Colchero MA, González de Cosío T. Breastfeeding Mode and Risk of Breast Cancer: A Dose-Response Meta-Analysis. J Hum Lact 2017; 33:422-434. [PMID: 28196329 DOI: 10.1177/0890334416683676] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Breastfeeding reduces women's risk of breast cancer. Since exclusive breastfeeding has a stronger hormonal effect, it could theoretically result in a greater reduction in breast cancer risk than any breastfeeding mode. No meta-analysis has examined breast cancer risk by breastfeeding mode. Research aim: The authors conducted a meta-analysis for breast cancer risk in parous women who breastfed exclusively or in any mode versus parous women who formula fed their infants, and they estimated the summary dose-response association by the accumulated duration of any breastfeeding mode. METHODS A systematic review of studies published between 2005 and 2015 analyzing breastfeeding and breast cancer risk in women was conducted in PubMed and EBSCOhost. A meta-analysis ( n = 65 studies) with fixed effects (or random effects, if heterogeneity existed) was carried out stratified by breastfeeding mode and menopausal and parity status. A summary dose-response association was estimated using the generalized least-squares method. RESULTS The summary relative risk (SRR) for breast cancer in parous women who breastfed exclusively was 0.72, 95% confidence interval (CI) [0.58, 0.90], versus parous women who had never breastfed. For parous women who breastfed in any mode, the SRR was lower in both premenopausal women (0.86, 95% CI [0.80, 0.93]) and postmenopausal women (0.89, 95% CI [0.83, 0.95]). There was no heterogeneity or publication bias. There is weak evidence of a difference between exclusive and any breastfeeding mode ( p = .08). The summary dose-response curve was nonlinear ( p < .001). CONCLUSION Exclusive breastfeeding among parous women reduces the risk of breast cancer compared with parous women who do not breastfeed exclusively.
Collapse
Affiliation(s)
- Mishel Unar-Munguía
- 1 Center for Research on Health and Nutrition, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - Gabriela Torres-Mejía
- 2 Center for Research on Population Health, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | - M Arantxa Colchero
- 3 Center for Health Systems Research, National Institute of Public Health, Cuernavaca, Morelos, Mexico
| | | |
Collapse
|
24
|
Differential Effects of Hormones on Cellular Metabolism in Keratoconus In Vitro. Sci Rep 2017; 7:42896. [PMID: 28211546 PMCID: PMC5314412 DOI: 10.1038/srep42896] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/18/2017] [Indexed: 12/28/2022] Open
Abstract
Keratoconus (KC) is a corneal thinning disease with an onset commonly immediately post-puberty and stabilization by 40 to 50 years of age. The role of hormones in regulating corneal tissue structure in homeostatic and pathological conditions is unknown. Our group recently linked altered hormone levels to KC. Our current study sought to investigate and delineate the effects of exogenous hormones, such as androgen, luteotropin, and estrogen, on corneal stroma bioenergetics. We utilized our established 3D in vitro model to characterize the effects of DHEA, prolactin, 17β-estradiol on insulin-growth factor-1 and -2 (IGF-1, -2) signaling and metabolic function in primary corneal fibroblasts from healthy controls (HCFs) and KC patients (HKCs). Our data showed that exogenous DHEA significantly downregulated IGF-1 and its receptor in both HCFs and HKCs with HKCs showing consistently lower basal pentose phosphate flux. Prolactin caused no significant change in IGF-1 levels and an increase in IGF-2 in HKCs correlating with an increase in ATP and NADH levels. 17β-estradiol led to a significant upregulation in pentose phosphate flux and glycolytic intermediates in HCFs. Our results identified hormone-specific responses regulated in HKCs compared to HCFs revealing a novel role for hormones on bioenergetics in KC.
Collapse
|
25
|
Bak MJ, Das Gupta S, Wahler J, Lee HJ, Li X, Lee MJ, Yang CS, Suh N. Inhibitory Effects of γ- and δ-Tocopherols on Estrogen-Stimulated Breast Cancer In Vitro and In Vivo. Cancer Prev Res (Phila) 2017; 10:188-197. [PMID: 28096236 DOI: 10.1158/1940-6207.capr-16-0223] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/24/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022]
Abstract
Estrogens have been implicated as complete carcinogens for breast and other tissues through mechanisms involving increased cell proliferation, oxidative stress, and DNA damage. Because of their potent antioxidant activity and other effects, tocopherols have been shown to exert antitumor activities in various cancers. However, limited information is available on the effect of different forms of tocopherols in estrogen-mediated breast cancer. To address this, we examined the effects of α-, γ-, and δ-tocopherols as well as a natural γ-tocopherol-rich mixture of tocopherols, γ-TmT, on estrogen-stimulated MCF-7 cells in vitro and in vivo For the in vivo studies, MCF-7 cells were injected into the mammary fat pad of immunodeficient mice previously implanted with estrogen pellets. Mice were then administered diets containing 0.2% α-, γ-, δ-tocopherol, or γ-TmT for 5 weeks. Treatment with α-, γ-, δ-tocopherols, and γ-TmT reduced tumor volumes by 29% (P < 0.05), 45% (P < 0.05), 41% (P < 0.05), and 58% (P < 0.01), as well as tumor weights by 20%, 37% (P < 0.05), 39% (P < 0.05), and 52% (P < 0.05), respectively. γ- and δ-tocopherols and γ-TmT inhibited the expression of cell proliferation-related genes such as cyclin D1 and c-Myc, and estrogen-related genes such as TFF/pS2, cathepsin D, and progesterone receptor in estrogen-stimulated MCF-7 cells in vitro Further, γ- and δ-tocopherols decreased the levels of estrogen-induced oxidative stress and nitrosative stress markers, 8-hydroxy-2'-deoxyguanosine and nitrotyrosine, as well as the DNA damage marker, γ-H2AX. Our results suggest that γ- and δ-tocopherols and the γ-tocopherol-rich mixture are effective natural agents for the prevention and treatment of estrogen-mediated breast cancer. Cancer Prev Res; 10(3); 188-97. ©2017 AACR.
Collapse
Affiliation(s)
- Min Ji Bak
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Soumyasri Das Gupta
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Joseph Wahler
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Hong Jin Lee
- Department of Food Science and Technology, Chung-Ang University, Anseong, South Korea
| | - Xiaowei Li
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Mao-Jung Lee
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Chung S Yang
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Nanjoo Suh
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey.
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
26
|
Kharel S, Lee WL, Lee XY, Loo SCJ. Osmogen-Mediated One-Step Technique of Fabricating Hollow Microparticles for Encapsulation and Delivery of Bioactive Molecules. Macromol Biosci 2016; 17. [DOI: 10.1002/mabi.201600328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/07/2016] [Indexed: 01/28/2023]
Affiliation(s)
- Sharad Kharel
- School of Materials Science and Engineering; Nanyang Technological University; 50 Nanyang Avenue Singapore 639798
| | - Wei Li Lee
- School of Materials Science and Engineering; Nanyang Technological University; 50 Nanyang Avenue Singapore 639798
| | - Xuan Yi Lee
- School of Materials Science and Engineering; Nanyang Technological University; 50 Nanyang Avenue Singapore 639798
| | - Say Chye Joachim Loo
- School of Materials Science and Engineering; Nanyang Technological University; 50 Nanyang Avenue Singapore 639798
- Singapore Centre on Environmental Life Sciences Engineering (SCELSE); Nanyang Technological University; Singapore 637551
| |
Collapse
|
27
|
The Anti-Cancer Effect of Polyphenols against Breast Cancer and Cancer Stem Cells: Molecular Mechanisms. Nutrients 2016; 8:nu8090581. [PMID: 27657126 PMCID: PMC5037565 DOI: 10.3390/nu8090581] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 08/25/2016] [Accepted: 09/09/2016] [Indexed: 02/07/2023] Open
Abstract
The high incidence of breast cancer in developed and developing countries, and its correlation to cancer-related deaths, has prompted concerned scientists to discover novel alternatives to deal with this challenge. In this review, we will provide a brief overview of polyphenol structures and classifications, as well as on the carcinogenic process. The biology of breast cancer cells will also be discussed. The molecular mechanisms involved in the anti-cancer activities of numerous polyphenols, against a wide range of breast cancer cells, in vitro and in vivo, will be explained in detail. The interplay between autophagy and apoptosis in the anti-cancer activity of polyphenols will also be highlighted. In addition, the potential of polyphenols to target cancer stem cells (CSCs) via various mechanisms will be explained. Recently, the use of natural products as chemotherapeutics and chemopreventive drugs to overcome the side effects and resistance that arise from using chemical-based agents has garnered the attention of the scientific community. Polyphenol research is considered a promising field in the treatment and prevention of breast cancer.
Collapse
|
28
|
Zelenko Z, Gallagher EJ, Antoniou IM, Sachdev D, Nayak A, Yee D, LeRoith D. EMT reversal in human cancer cells after IR knockdown in hyperinsulinemic mice. Endocr Relat Cancer 2016; 23:747-58. [PMID: 27435064 PMCID: PMC4990486 DOI: 10.1530/erc-16-0142] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 07/19/2016] [Indexed: 12/26/2022]
Abstract
Type 2 diabetes (T2D) is associated with increased cancer risk and cancer-related mortality. Data herein show that we generated an immunodeficient hyperinsulinemic mouse by crossing the Rag1(-/-) mice, which have no mature B or T lymphocytes, with the MKR mouse model of T2D to generate the Rag1(-/-) (Rag/WT) and Rag1(-/-)/MKR(+/+) (Rag/MKR) mice. The female Rag/MKR mice are insulin resistant and have significantly higher nonfasting plasma insulin levels compared with the Rag/WT controls. Therefore, we used these Rag/MKR mice to investigate the role of endogenous hyperinsulinemia on human cancer progression. In this study, we show that hyperinsulinemia in the Rag/MKR mice increases the expression of mesenchymal transcription factors, TWIST1 and ZEB1, and increases the expression of the angiogenesis marker, vascular endothelial growth factor A (VEGFA). We also show that silencing the insulin receptor (IR) in the human LCC6 cancer cells leads to decreased tumor growth and metastases, suppression of mesenchymal markers vimentin, SLUG, TWIST1 and ZEB1, suppression of angiogenesis markers, VEGFA and VEGFD, and re-expression of the epithelial marker, E-cadherin. The data in this paper demonstrate that IR knockdown in primary tumors partially reverses the growth-promoting effects of hyperinsulinemia as well as highlighting the importance of the insulin receptor signaling pathway in cancer progression, and more specifically in epithelial-mesenchymal transition.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/pathology
- Disease Models, Animal
- Epithelial-Mesenchymal Transition
- Female
- Gene Silencing
- Humans
- Hyperinsulinism/genetics
- Hyperinsulinism/metabolism
- Hyperinsulinism/pathology
- Male
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Mice, Transgenic
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Receptor, Insulin/genetics
- Receptor, Insulin/metabolism
- Signal Transduction
- Snail Family Transcription Factors/genetics
- Snail Family Transcription Factors/metabolism
- Twist-Related Protein 1/genetics
- Twist-Related Protein 1/metabolism
- Vascular Endothelial Growth Factor A
- Vimentin/metabolism
- Zinc Finger E-box-Binding Homeobox 1/genetics
- Zinc Finger E-box-Binding Homeobox 1/metabolism
Collapse
Affiliation(s)
- Zara Zelenko
- Division of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Emily Jane Gallagher
- Division of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Irini Markella Antoniou
- Division of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Deepali Sachdev
- Department of Medicine and Masonic Cancer CenterUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Anupma Nayak
- Department of Pathology and Laboratory MedicineThe Mount Sinai Hospital and Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Douglas Yee
- Department of Medicine and Masonic Cancer CenterUniversity of Minnesota, Minneapolis, Minnesota, USA
| | - Derek LeRoith
- Division of EndocrinologyDiabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
29
|
Smith LC, Clark JC, Bisesi JH, Ferguson PL, Sabo-Attwood T. Differential recruitment of co-regulatory proteins to the human estrogen receptor 1 in response to xenoestrogens. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2016; 19:159-173. [PMID: 27156127 DOI: 10.1016/j.cbd.2016.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 04/11/2016] [Accepted: 04/17/2016] [Indexed: 01/24/2023]
Abstract
The diverse biological effects of xenoestrogens may be explained by their ability to differentially recruit co-regulatory proteins to the estrogen receptor (ER). We employed high-throughput receptor affinity binding and co-regulatory protein recruitment screening assays based on fluorescence polarization and time resolved florescence resonance energy transfer (TR-FRET), respectively, to assess xenoestrogen-specific binding and co-regulatory protein recruitment to the ER. Then we used a functional proteomic assay based on co-immunoprecipitation of ER-bound proteins to isolate and identify intact co-regulatory proteins recruited to a ligand-activated ER. Through these approaches, we revealed differential binding affinity of bisphenol-A (BPA) and genistein (GEN) to the human ERα (ESR1) and ligand-dependent recruitment of SRC-1 and SRC-3 peptides. Recruitment profiles were variable for each ligand and in some cases were distinct compared to 17β-estradiol (E2). For example, E2 and GEN recruited both SRC-1 and -3 peptides whereas BPA recruited only SRC-1 peptides. Results of the functional proteomic assay showed differential recruitment between ligands where E2 recruited the greatest number of proteins followed by BPA then GEN. A number of proteins share previously identified relationships with ESR1 as determined by STRING analysis. Although there was limited overlap in proteins identified between treatments, all ligands recruited proteins involved in cell growth as determined by subnetwork enrichment analysis (p<0.05). A comparative, in silico analysis revealed that fewer interactions exist between zebrafish (Danio rerio) esr1 and zebrafish orthologs of proteins identified in our functional proteomic analysis. Taken together these results identify recruitment of known and previously unknown co-regulatory proteins to ESR1 and highlight new methods to assay recruitment of low abundant and intact, endogenous co-regulatory proteins to ESR1 or other nuclear receptors, in both human and aquatic species.
Collapse
Affiliation(s)
- L Cody Smith
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | - Jessica C Clark
- Department of Civil and Environmental Engineering and Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Joseph H Bisesi
- Department of Environmental and Global Health and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA
| | - P Lee Ferguson
- Department of Civil and Environmental Engineering and Nicholas School of the Environment, Duke University, Durham, NC 27708, USA
| | - Tara Sabo-Attwood
- Department of Environmental and Global Health and Center for Environmental and Human Toxicology, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
30
|
Simone V, D'Avenia M, Argentiero A, Felici C, Rizzo FM, De Pergola G, Silvestris F. Obesity and Breast Cancer: Molecular Interconnections and Potential Clinical Applications. Oncologist 2016; 21:404-17. [PMID: 26865587 DOI: 10.1634/theoncologist.2015-0351] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/23/2015] [Indexed: 01/22/2023] Open
Abstract
UNLABELLED Obesity is an important risk factor for breast cancer (BC) in postmenopausal women; interlinked molecular mechanisms might be involved in the pathogenesis. Increased levels of estrogens due to aromatization of the adipose tissue, inflammatory cytokines such as tumor necrosis factor-α, interleukin-6, and prostaglandin E2, insulin resistance and hyperactivation of insulin-like growth factors pathways, adipokines, and oxidative stress are all abnormally regulated in obese women and contribute to cancerogenesis. These molecular factors interfere with intracellular signaling in the mitogen-activated protein kinase and phosphatydilinositol-3-phosphate/mammalian target of rapamycin (mTOR) pathways, which regulate the progression of the cell cycle, apoptosis, and protein synthesis. In this context, structural defects of typical genes related to both BC and obesity, such as leptin, leptin receptor, serum paraoxonase/arylesterase 1, the fat mass and obesity-associated gene and melanocortin receptor 4, have been associated with a high or low risk of BC development. The early detection of these gene alterations might be useful as risk predictors in obese women, and targeting these pathways involved in the BC pathogenesis in obese women is a potential therapeutic tool. In particular, mTOR pathway deregulation concurs in both obesity and BC, and inhibition of this might disrupt the molecular interlinks in a similar manner to that of metformin, which exerts definite anticancer activity and is currently used as an antidiabetic drug with a weight-reducing property. The identification of both genetic and pharmacological implications on the prevention and management of BC is the ultimate aim of these studies. IMPLICATIONS FOR PRACTICE Obese women are at risk of breast cancer, but clinicians lack concrete tools for the prevention or early diagnosis of this risk. The present study, starting from the biology and the molecular defects characterizing both obesity and breast cancer, analyzed the potential molecules and genetic defects whose early identification could delineate a risk profile. Three steps are proposed that are potentially achievable in the clinical assessment of obese women, namely the evaluation of altered levels of serum molecules, the identification of genetic polymorphisms, and the study of the transcriptomic profile of premalignant lesions. Finally, the therapeutic implications of this molecular assessment were evaluated.
Collapse
Affiliation(s)
- Valeria Simone
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Morena D'Avenia
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Antonella Argentiero
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Claudia Felici
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Francesca Maria Rizzo
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Giovanni De Pergola
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| | - Franco Silvestris
- Department of Biomedical Sciences and Human Oncology, University of Bari "A. Moro," Bari, Italy
| |
Collapse
|
31
|
Wairagu PM, Phan ANH, Kim MK, Han J, Kim HW, Choi JW, Kim KW, Cha SK, Park KH, Jeong Y. Insulin priming effect on estradiol-induced breast cancer metabolism and growth. Cancer Biol Ther 2016; 16:484-92. [PMID: 25701261 DOI: 10.1080/15384047.2015.1016660] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Diabetes is a risk factor for breast cancer development and is associated with poor prognosis for breast cancer patients. However, the molecular and biochemical mechanisms underlying the association between diabetes and breast cancer have not been fully elucidated. Here, we investigated estradiol response in MCF-7 breast cancer cells with or without chronic exposure to insulin. We found that insulin priming is necessary and specific for estradiol-induced cancer cell growth, and induces anaplerotic shunting of glucose into macromolecule biosynthesis in the estradiol treated cells. Treatment with ERK or Akt specific inhibitors, U0126 or LY294002, respectively, suppressed estradiol-induced growth. Interestingly, molecular analysis revealed that estradiol treatment markedly increases expression of cyclin A and B, and decreases p21 and p27 in the insulin-primed cells. In addition, estradiol treatment activated metabolic genes in pentose phosphate (PPP) and serine biosynthesis pathways in the insulin-primed cells while insulin priming decreased metabolic gene expression associated with glucose catabolism in the breast cancer cells. Finally, we found that anti-diabetic drug metformin and AMPK ligand AICAR, but not thiazolidinediones (TZDs), specifically suppress the estradiol-induced cellular growth in the insulin-primed cells. These findings suggest that estrogen receptor (ER) activation under chronic hyperinsulinemic condition increases breast cancer growth through the modulation of cell cycle and apoptotic factors and nutrient metabolism, and further provide a mechanistic evidence for the clinical benefit of metformin use for ER-positive breast cancer patients with diabetes.
Collapse
Key Words
- AR, androgen receptor
- CKI, cyclin dependent-kinase inhibitor
- DHT, dihydrotestosterone
- ER, estrogen receptor
- G6PD, glucose-6-phosphodehydrogenase
- GLUT1, glucose transporter 1
- HER2, human epidermal growth factor receptor 2
- IGF-1, insulin-like growth factor 1
- LDHA, lactate dehydrogenase A
- MCF-7
- PDK1, pyruvate dehydrogenase kinase 1
- PFK, phosphofructokinase
- PFKFB3, 6-phosphofructo-2-kinase/fructose-2, 6-bisphosphate 3
- PGD, 6-phosphogluconate dehydrogenase
- PHGDH, phosphoglycerate dehydrogenase
- PKM, pyruvate kinase M
- PPAR γ, peroxisome proliferator-activated receptor gamma
- PPP, pentose phosphate pathway
- PR, progesterone receptor
- PSPH, phosphoserine phosphatase
- RPE, ribulose-5-phosphate-3-epimerase
- RPIA, ribulose-5-phosphate isomerase A
- SHMT, serine hydroxymethyltransferase
- TALDO1, transaldolase 1
- TKT, transketolase
- TZDs, thiazolidinediones
- breast cancer
- diabetes
- estradiol
- estrogen receptor
- iInsulin priming
- metformin
Collapse
Affiliation(s)
- Peninah M Wairagu
- a Department of Biochemistry ; Wonju College of Medicine ; Yonsei University ; Wonju , Gangwon-do , Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mocanu MM, Nagy P, Szöllősi J. Chemoprevention of Breast Cancer by Dietary Polyphenols. Molecules 2015; 20:22578-620. [PMID: 26694341 PMCID: PMC6332464 DOI: 10.3390/molecules201219864] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 02/07/2023] Open
Abstract
The review will discuss in detail the effects of polyphenols on breast cancer, including both the advantages and disadvantages of the applications of these natural compounds. First, we focus on the characterization of the main classes of polyphenols and then on in vitro and in vivo experiments carried out in breast cancer models. Since the therapeutic effects of the administration of a single type of polyphenol might be limited because of the reduced bioavailability of these drugs, investigations on combination of several polyphenols or polyphenols with conventional therapy will also be discussed. In addition, we present recent data focusing on clinical trials with polyphenols and new approaches with nanoparticles in breast cancer. Besides the clinical and translational findings this review systematically summarizes our current knowledge about the molecular mechanisms of anti-cancer effects of polyphenols, which are related to apoptosis, cell cycle regulation, plasma membrane receptors, signaling pathways and epigenetic mechanisms. At the same time the effects of polyphenols on primary tumor, metastasis and angiogenesis in breast cancer are discussed. The increasing enthusiasm regarding the combination of polyphenols and conventional therapy in breast cancer might lead to additional efforts to motivate further research in this field.
Collapse
Affiliation(s)
- Maria-Magdalena Mocanu
- Department of Biophysics, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Péter Nagy
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| | - János Szöllősi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
- MTA-DE Cell Biology and Signaling Research Group, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary.
| |
Collapse
|
33
|
Fernandez TL, Van Lonkhuyzen DR, Dawson RA, Kimlin MG, Upton Z. Insulin-like growth factor-I and UVB photoprotection in human keratinocytes. Exp Dermatol 2015; 24:235-8. [PMID: 25607472 DOI: 10.1111/exd.12637] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2015] [Indexed: 11/26/2022]
Abstract
Ultraviolet radiation (UVR), in particular the UVB spectrum, is a risk factor for skin cancer development. The generation and accumulation of UVB-induced genetic mutations are fundamental premalignant events. Keratinocyte interactions between other cutaneous cell populations and the surrounding microenvironment determine cell fate and acute photoresponses. In this study, the importance of the insulin-like growth factor (IGF) system, in particular the insulin-like growth factor-I (IGF-I), on influencing key processes in the keratinocyte acute photoresponse was investigated. Exogenous IGF-I and other growth factors present in dermal fibroblast-conditioned media (CM) were found to significantly enhance keratinocyte survival following UVB irradiation in vitro. This pretreatment was also shown to cause a shift in the expression levels of various DNA damage response proteins. Consequently, this was associated with accelerated rates of UVB-induced cyclobutane pyrimidine dimer removal in these samples. Finally, activation of the IGF system influenced cell cycle progression in UVB-irradiated keratinocytes. Taken together, these results highlight the importance of the IGF signalling network in initiating the repair of potentially mutagenic DNA damage in human keratinocytes. The dysregulation of these processes may therefore have significant implications in the aetiology of skin cancers and other cutaneous diseases.
Collapse
Affiliation(s)
- Tara Lyn Fernandez
- Tissue Repair and Regeneration Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Qld, Australia
| | | | | | | | | |
Collapse
|
34
|
Shao Y, Cheng S, Hou J, Zuo Y, Zheng W, Xia M, Mu N. Insulin is an important risk factor of endometrial cancer among premenopausal women: a case-control study in China. Tumour Biol 2015; 37:4721-6. [PMID: 26511973 DOI: 10.1007/s13277-015-4229-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/12/2015] [Indexed: 02/06/2023] Open
Abstract
The aim of the article was to evaluate the important role played by insulin in the development of endometrial cancer (EC) among Chinese premenopausal women. In this study, 128 endometrial cancer patients and 294 controls who were all premenopausal were included. Baseline characteristics data were collected and serum insulin, C-peptide, sex hormone-binding globulin, C-reaction protein, interleukin-6, and tumor necrosis factor-α levels were measured. Paired t test, χ(2) test, Spearman correlation coefficients, and univariate and multivariate logistic regression models were used in data analysis. Furthermore, insulin levels were categorized into quartiles, and likelihood ratio was calculated for the four categories. Blood insulin levels of the patients were significantly higher than those of the controls (P < 0.001). Factor analysis identified insulin (OR = 2.46; 95 % confidence interval (CI) = 1.55-3.91; P < 0.001) as the independent risk factor of EC. When insulin levels were categorized into quartiles, we found that insulin was positively associated with endometrial cancer risk [HR comparing extreme quartiles (HR q4-q1) = 4.44; 95 % CI = 2.59-7.62; P trend = 0.025]. After adjustment for body mass index (BMI) or waist-hip ratio (WHR), this association was attenuated, but still significant. In conclusion, insulin plays an important role in the carcinogenesis of EC among premenopausal women. Treatment targeting down-regulation of blood insulin levels seems effective in the prevention of this malignancy.
Collapse
Affiliation(s)
- Yanli Shao
- Department of Gynecology and Obstetrics, First People's Hospital of Laishan District, Yantai, Shandong Province, People's Republic of China, 264003
| | - Shijie Cheng
- Department of Gynecology and Obstetrics, Heze Municipal Hospital, Heze, Shandong Province, People's Republic of China, 274031
| | - Jianqing Hou
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital, NO 20, Yuhuangding East Road, Zhifu District, Yantai, Shandong Province, People's Republic of China, 264000
| | - Ying Zuo
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital, NO 20, Yuhuangding East Road, Zhifu District, Yantai, Shandong Province, People's Republic of China, 264000
| | - Wei Zheng
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital, NO 20, Yuhuangding East Road, Zhifu District, Yantai, Shandong Province, People's Republic of China, 264000
| | - Min Xia
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital, NO 20, Yuhuangding East Road, Zhifu District, Yantai, Shandong Province, People's Republic of China, 264000.
| | - Nan Mu
- Department of Gynecology and Obstetrics, Yantai Yuhuangding Hospital, NO 20, Yuhuangding East Road, Zhifu District, Yantai, Shandong Province, People's Republic of China, 264000.
| |
Collapse
|
35
|
Gong Y, He H, Liu H, Zhang C, Zhao W, Shao RG. Phosphorylation of myofibrillogenesis regulator-1 activates the MAPK signaling pathway and induces proliferation and migration in human breast cancer MCF7 cells. FEBS Lett 2014; 588:2903-10. [PMID: 25066297 DOI: 10.1016/j.febslet.2014.07.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 07/05/2014] [Accepted: 07/15/2014] [Indexed: 12/24/2022]
Abstract
Myofibrillogenesis regulator-1 (MR-1) has been characterized as a tumor promoter in many cancers. However, its mechanism of action has not been fully elucidated. Here, we report that MR-1 is overexpressed in human breast cancer cells and participates in tumor promotion in human breast cancer MCF7 cells by activating the ERK1/2 signaling pathway. MR-1 interacts with MEK1/2 and ERK1, and its N-terminal sequence plays a major role in promoting the MEK/ERK cascade. Furthermore, six phosphorylation sites of MR-1 were identified, and phosphorylation at S46 was shown to be critical for the activation of MEK/ERK. Therefore, our findings suggest that MR-1 functions as a tumor promoter in MCF7 cells by activating the MEK/ERK signaling.
Collapse
Affiliation(s)
- Yuyan Gong
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing 100050, China
| | - Hongwei He
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing 100050, China
| | - Hong Liu
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing 100050, China
| | - Caixia Zhang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing 100050, China
| | - Wuli Zhao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing 100050, China
| | - Rong-Guang Shao
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, 1 Tiantan Xili, Beijing 100050, China.
| |
Collapse
|
36
|
Wang T, Ning G, Bloomgarden Z. Diabetes and cancer relationships. J Diabetes 2013; 5:378-90. [PMID: 23574745 DOI: 10.1111/1753-0407.12057] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 03/10/2013] [Accepted: 03/24/2013] [Indexed: 12/29/2022] Open
Abstract
Diabetes and cancer are both heterogeneous and multifactorial diseases with tremendous impact on health worldwide. Epidemiologic evidence suggests that certain malignancies may be associated with diabetes, as well as with diabetes risk factors and, perhaps, with certain diabetes treatments. Numerous biological mechanisms could account for these relationships. Insulin-like growth factor (IGF)-1, IGF-2, IGF-1 receptors, insulin, and the insulin receptor play roles in the development and progression of cancers. Although evidence from randomized controlled trials does not support or refute associations of diabetes and its treatments with either increased or reduced risk of cancer incidence or prognosis, consideration of malignancy incidence rates and the magnitude of the trials that would be required to address these issues explains why such studies may not be readily undertaken.
Collapse
Affiliation(s)
- Tiange Wang
- Key Laboratory for Endocrine and Metabolic Diseases of Ministry of Health, Shanghai, China; Shanghai Clinical Center for Endocrine and Metabolic Diseases, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; E-Institute of Shanghai Universities, Shanghai, China
| | | | | |
Collapse
|
37
|
Hawsawi Y, El-Gendy R, Twelves C, Speirs V, Beattie J. Insulin-like growth factor - oestradiol crosstalk and mammary gland tumourigenesis. Biochim Biophys Acta Rev Cancer 2013; 1836:345-53. [PMID: 24189571 DOI: 10.1016/j.bbcan.2013.10.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2013] [Revised: 10/15/2013] [Accepted: 10/24/2013] [Indexed: 12/22/2022]
Abstract
Development and differentiation of the mammary gland are dependent on the appropriate temporal expression of both systemically acting hormones and locally produced growth factors. A large body of evidence suggests that molecular crosstalk between these hormonal and growth factor axes is crucial for appropriate cell and tissue function. Two of the most important trophic factors involved in this process are the oestrogen (E) and insulin-like growth factor (IGF) molecular axes. The reciprocal crosstalk that exists between these pathways occurs at transcriptional/post-transcriptional and translational/post-translational levels regulate the expression and activity of genes involved in this process. In a clinical context an important consequence of such crosstalk in the mammary gland is the role which it may play in the aetiology, maintenance and development of breast tumours. Although oestradiol (E2) acting through oestrogen receptors α and β (ERα/β) is important for normal mammary gland function it can also provide a mitogenic drive to ER+ breast tumours. Therefore over several years anti-oestrogen therapeutic regimens in the form of selective oestrogen receptor modulators (SERMs - e.g. tamoxifen), aromatase inhibitors (AI e.g. anastrozole) or selective oestrogen receptor down regulators (SERDs - e.g. fulvestrant) have been used in an adjuvant setting to control tumour growth. Although initial response is usually encouraging, large cohorts of patients eventually develop resistance to these treatments leading to tumour recurrence and poor prognosis. There are potentially many routes by which breast cancer (BC) cells could escape anti-oestrogen based therapeutic strategies and one of the most studied is the possible growth factor mediated activation of ER(s). Because of this, growth factor modulation of ER activity has been an intensively studied route of molecular crosstalk in the mammary gland. The insulin-like growth factors (IGF-1 and -2) are amongst the most potent mitogens for mammary epithelial cells and there is accumulating evidence that they interact with the E2 axis to regulate mitogenesis, apoptosis, adhesion, migration and differentiation of mammary epithelial cells. Such interactions are bi-directional and E2 has been shown to regulate the expression and activity of IGF axis genes with the general effect of sensitising breast epithelial cells to the actions of IGFs and insulin. In this short review we discuss the evidence for the involvement of crosstalk between the insulin-like growth factor (IGF) and oestrogen axes in the mammary gland and comment on the relevance of such studies in the aetiology and treatment of BC.
Collapse
|
38
|
Woo HD, Park KS, Shin A, Ro J, Kim J. Glycemic Index and Glycemic Load Dietary Patterns and the Associated Risk of Breast Cancer: A Case-control Study. Asian Pac J Cancer Prev 2013; 14:5193-8. [DOI: 10.7314/apjcp.2013.14.9.5193] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
39
|
Delort L, Lequeux C, Dubois V, Dubouloz A, Billard H, Mojallal A, Damour O, Vasson MP, Caldefie-Chézet F. Reciprocal interactions between breast tumor and its adipose microenvironment based on a 3D adipose equivalent model. PLoS One 2013; 8:e66284. [PMID: 23750285 PMCID: PMC3672201 DOI: 10.1371/journal.pone.0066284] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 05/09/2013] [Indexed: 11/25/2022] Open
Abstract
Breast cancer has become the most common cancer among women in industrialized countries. Obesity is well established as a risk factor, in particular owing to the attendant secretion of the entities called adipokines; there is growing evidence for a role of cells and factors present in the mammary tumor microenvironment such as fibroblasts, preadipocytes, adipocytes and their secretions. To study how the microenvironment influences breast cancer growth, we developed a novel tridimensional adipose model epithelialized with normal human keratinocytes or with breast cancer cell lines. These mimicked a breast tumor in contact with an adipose microenvironment and allowed monitoring of the interactions between the cells. Leptin and adiponectin, two major adipokines, and their respective receptors, ObRt and AdipoR1, were expressed in the model, but not the second adiponectin receptor, AdipoR2. The differentiation of preadipocytes into adipocytes was greater when they were in contact with the breast cancer cell lines. The contact of breast cancer cell lines with the microenvironment completely modified their transcriptional programs by increasing the expression of genes involved in cell proliferation (cyclinD1, MAPK), angiogenesis (MMP9, VEGF) and hormonal pathways (ESR1, IL6). This tridimensional adipose model provides new insights into the interactions between breast cancer cells and their adipose microenvironment, and provides a tool to develop new drugs for the treatment of both cancer and obesity.
Collapse
Affiliation(s)
- Laetitia Delort
- Clermont Université, Université d'Auvergne, UFR Pharmacie, Laboratoire SVFp, Clermont-Ferrand, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhou YH, Liao SJ, Li D, Luo J, Wei JJ, Yan B, Sun R, Shu Y, Wang Q, Zhang GM, Feng ZH. TLR4 ligand/H₂O₂ enhances TGF-β1 signaling to induce metastatic potential of non-invasive breast cancer cells by activating non-Smad pathways. PLoS One 2013; 8:e65906. [PMID: 23734265 PMCID: PMC3667026 DOI: 10.1371/journal.pone.0065906] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Accepted: 04/29/2013] [Indexed: 12/17/2022] Open
Abstract
TGF-β1 has the potential to activate multiple signaling pathways required for inducing metastatic potential of tumor cells. However, TGF-β1 was inefficient in inducing metastatic potential of many non-invasive human tumor cells. Here we report that the enhancement of TGF-β1 signaling is required for inducing metastatic potential of non-invasive breast cancer cells. TGF-β1 alone could not efficiently induce the sustained activation of Smad and non-Smad pathways in non-invasive breast cancer cells. TLR4 ligand (LPS) and H2O2 cooperated with TGF-β1 to enhance the sustained activation of non-Smad pathways, including p38MAPK, ERK, JNK, PI3K, and NF-κB. The activation of MAPK and PI3K pathways resulted in a positive feed-back effect on TGF-β1 signaling by down-regulating Nm23-H1 expression and up-regulating the expression of TβRI and TβRII, favoring further activation of multiple signaling pathways. Moreover, the enhanced TGF-β1 signaling induced higher expression of SNAI2, which also promoted TβRII expression. Therefore, the sustained activation levels of both Smad and non-Smad pathways were gradually increased after prolonged stimulation with TGF-β1/H2O2/LPS. Consistent with the activation pattern of signaling pathways, the invasive capacity and anoikis-resistance of non-invasive breast cancer cells were gradually increased after prolonged stimulation with TGF-β1/H2O2/LPS. The metastatic potential induced by TGF-β1/H2O2/LPS was sufficient for tumor cells to extravasate and form metastatic foci in an experimental metastasis model in nude mice. The findings in this study suggested that the enhanced signaling is required for inducing higher metastatic capacity of tumor cells, and that targeting one of stimuli or signaling pathways might be potential approach in comprehensive strategy for tumor therapy.
Collapse
Affiliation(s)
- Yuan-Hong Zhou
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Sheng-Jun Liao
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Dong Li
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Jing Luo
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Jing-Jing Wei
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Bin Yan
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Rui Sun
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Yu Shu
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Qi Wang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
| | - Gui-Mei Zhang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
- * E-mail: (GMZ); (ZHF)
| | - Zuo-Hua Feng
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, The People's Republic of China
- * E-mail: (GMZ); (ZHF)
| |
Collapse
|
41
|
Alegre MM, Knowles MH, Robison RA, O'Neill KL. Mechanics behind Breast Cancer Prevention - Focus on Obesity, Exercise and Dietary Fat. Asian Pac J Cancer Prev 2013; 14:2207-12. [DOI: 10.7314/apjcp.2013.14.4.2207] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
42
|
Bruchim I, Werner H. Targeting IGF-1 signaling pathways in gynecologic malignancies. Expert Opin Ther Targets 2013; 17:307-20. [PMID: 23294364 DOI: 10.1517/14728222.2013.749863] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION The signaling pathways of the insulin-like growth factors (IGF) have been implicated in the etiology of a number of epithelial neoplasms including prostate, breast, colon and more recently, gynecologic cancers. The insulin-like growth factor-1 receptor (IGF-1R) is expressed in most transformed cells, where it displays potent anti-apoptotic, cell-survival and potentially, transforming activities. IGF-1R expression and activation are typical hallmarks associated with tumor initiation and progression. Multiple approaches have been used to abrogate IGF-1R signaling for targeted cancer therapy including antibodies and small molecule tyrosine kinase inhibitors. These novel IGF-1R targeting agents have produced significant experimental and clinical results in many cancers and generated considerable optimism in the field of cancer therapy. AREAS COVERED The authors will review important research advances regarding the role of the IGF axis in cancer, particularly preclinical and clinical studies in cervical, uterine and ovarian cancers. The significance of tumor expression and circulating levels of the IGF pathway as well as targeting therapies of the IGF axis in the gynecologic cancers will be discussed. EXPERT OPINION Accumulating data confirm that the IGF-1R pathway has an important role in gynecologic cancers and in vivo and in vitro studies have shown a significant impact of IGF-1R targeted therapies in these malignancies, mainly ovarian and endometrial cancers. Currently, ongoing preclinical and clinical trials are evaluating the efficacy of IGF-1R targeting. A better understanding of the complex mechanisms underlying the regulation of the IGF system will improve the ability to develop effective treatment modalities for these malignancies.
Collapse
Affiliation(s)
- Ilan Bruchim
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
43
|
Lu J, Zhao H, Xu J, Zhang L, Yan L, Shen Z. Elevated cyclin D1 expression is governed by plasma IGF-1 through Ras/Raf/MEK/ERK pathway in rumen epithelium of goats supplying a high metabolizable energy diet. J Anim Physiol Anim Nutr (Berl) 2012; 97:1170-8. [DOI: 10.1111/jpn.12026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Accepted: 11/21/2012] [Indexed: 12/15/2022]
Affiliation(s)
- J. Lu
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
- Jiangsu Animal Husbandry & Veterinary College; Nanjing China
| | - H. Zhao
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - J. Xu
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - L. Zhang
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - L. Yan
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| | - Z. Shen
- Lab of Animal Physiology and Biochemistry; College of Veterinary Medicine; Nanjing Agricultural University; Nanjing China
| |
Collapse
|
44
|
Gaben AM, Sabbah M, Redeuilh G, Bedin M, Mester J. Ligand-free estrogen receptor activity complements IGF1R to induce the proliferation of the MCF-7 breast cancer cells. BMC Cancer 2012; 12:291. [PMID: 22799881 PMCID: PMC3476977 DOI: 10.1186/1471-2407-12-291] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 06/26/2012] [Indexed: 11/13/2022] Open
Abstract
Background Ligand-dependent activation of the estrogen receptor (ER) as well as of the insulin-like growth factor type 1 (IGF1R) induces the proliferation of luminal breast cancer cells. These two pathways cooperate and are interdependent. We addressed the question of the mechanisms of crosstalk between the ER and IGF1R. Methods We evaluated the mitogenic effects of estradiol (E2; agonist ligand of ER) and of insulin (a ligand of IGF1R) in the MCF-7 cells by flow cytometry and by analyzing the cell levels of cell cycle-related proteins (immunoblotting) and mRNA (RT-QPCR). To verify the requirement for the kinase activity of Akt (a downstream target of IGF1R) in the mitogenic action of estradiol, we used shRNA strategy and shRNA-resistant expression vectors. Results The activation of the ER by E2 is unable to induce the cell cycle progression when the phosphatidyl inositol-3 kinase (PI3K)/Akt signaling is blocked by a chemical inhibitor (LY 294002) or by shRNA targeting Akt1 and Akt2. shRNA-resistant Akt wild-type constructs efficiently complemented the mitogenic signaling activity of E2 whereas constructs with inactivated kinase function did not. In growth factor-starved cells, the residual PI3K/Akt activity is sufficient to complement the mitogenic action of E2. Conversely, when ER function is blocked by the antiestrogen ICI 182780, IGF1R signaling is intact but does not lead to efficient reinitiation of the cell cycle in quiescent, growth factor-starved MCF-7 cells. The basal transcription-promoting activity of ligand-free ER in growth factor-starved cells is sufficient to complement the mitogenic action of the IGF1R-dependent signaling. Conclusions The basal ER activity in the absence of ligand is sufficient to allow efficient mitogenic action of IGF1R agonists and needs to be blocked to prevent the cell cycle progression.
Collapse
Affiliation(s)
- Anne-Marie Gaben
- Inserm U938, Centre de Recherche Saint-Antoine, Hôpital Saint-Antoine, Bâtiment Kourilsky, Paris cedex, France.
| | | | | | | | | |
Collapse
|
45
|
Liu D, Lin Y, Kang T, Huang B, Xu W, Garcia-Barrio M, Olatinwo M, Matthews R, Chen YE, Thompson WE. Mitochondrial dysfunction and adipogenic reduction by prohibitin silencing in 3T3-L1 cells. PLoS One 2012; 7:e34315. [PMID: 22479600 PMCID: PMC3316679 DOI: 10.1371/journal.pone.0034315] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 02/28/2012] [Indexed: 11/18/2022] Open
Abstract
Increase in mitochondrial biogenesis has been shown to accompany brown and white adipose cell differentiation. Prohibitins (PHBs), comprised of two evolutionarily conserved proteins, prohibitin-1 (PHB1) and prohibitin-2 (PHB2), are present in a high molecular-weight complex in the inner membrane of mitochondria. However, little is known about the effect of mitochondrial PHBs in adipogenesis. In the present study, we demonstrate that the levels of both PHB1 and PHB2 are significantly increased during adipogenesis of 3T3-L1 preadipocytes, especially in mitochondria. Knockdown of PHB1 or PHB2 by oligonucleotide siRNA significantly reduced the expression of adipogenic markers, the accumulation of lipids and the phosphorylation of extracellular signal-regulated kinases. In addition, fragmentation of mitochondrial reticulum, loss of mitochondrial cristae, reduction of mitochondrial content, impairment of mitochondrial complex I activity and excessive production of ROS were observed upon PHB-silencing in 3T3-L1 cells. Our results suggest that PHBs are critical mediators in promoting 3T3-L1 adipocyte differentiation and may be the potential targets for obesity therapies.
Collapse
Affiliation(s)
- Dong Liu
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- Department of Physiology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (DL); (WET)
| | - Yiming Lin
- Department of Endocrinology, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Ting Kang
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- Division of Cardiology, the First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Bo Huang
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- Department of Clinical Laboratory Sciences, the Second Affiliated Hospital, Nanchang University, Nanchang, China
| | - Wei Xu
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Minerva Garcia-Barrio
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Moshood Olatinwo
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Roland Matthews
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
| | - Y. Eugene Chen
- Cardiovascular Center, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Winston E. Thompson
- Department of Obstetrics and Gynecology, Morehouse School of Medicine, Atlanta, Georgia, United States of America
- * E-mail: (DL); (WET)
| |
Collapse
|
46
|
Bartella V, De Marco P, Malaguarnera R, Belfiore A, Maggiolini M. New advances on the functional cross-talk between insulin-like growth factor-I and estrogen signaling in cancer. Cell Signal 2012; 24:1515-21. [PMID: 22481093 DOI: 10.1016/j.cellsig.2012.03.012] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2012] [Accepted: 03/20/2012] [Indexed: 01/07/2023]
Abstract
There is increasing awareness that estrogens may affect cell functions through the integration with a network of signaling pathways. The IGF system is a phylogenetically highly conserved axis that includes the insulin receptor (IR) and the insulin-like growth factor I receptor (IGF-IR) pathways, which are of crucial importance in the regulation of metabolism and cell growth in relationship to nutrient availability. Numerous studies nowadays document that estrogens cooperate with IGF system at multiple levels both in physiology and in disease. Several studies have focused on this bidirectional cross-talk in central nervous system, in mammary gland development and in cancer. Notably, cancer cells show frequent deregulation of the IGF system with overexpression of IR and/or IGF-IR and their ligands as well as frequent upregulation of the classical estrogen receptor (ER)α and the novel ER named GPER. Recent studies have, therefore, unraveled further mechanisms of cross-talk involving membrane initiated estrogen actions and the IGF system in cancer, that converge in the stimulation of pro-tumoral effects. These studies offer hope for new strategies aimed at the treatment of estrogen related cancers in order to prevent an estrogen-independent and more aggressive tumor progression.
Collapse
Affiliation(s)
- Viviana Bartella
- Department of Pharmaco-Biology, University of Calabria, 87030 Rende, Italy
| | | | | | | | | |
Collapse
|
47
|
Wang Y, Zhu Y, Zhang L, Tian W, Hua S, Zhao J, Zhang H, Xue F. Insulin promotes proliferation, survival, and invasion in endometrial carcinoma by activating the MEK/ERK pathway. Cancer Lett 2012; 322:223-31. [PMID: 22459351 DOI: 10.1016/j.canlet.2012.03.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Revised: 03/21/2012] [Accepted: 03/21/2012] [Indexed: 11/24/2022]
Abstract
The involvement of insulin in endometrial carcinoma (EC) was investigated using radioimmunoassay, Western blot, immunoprecipitation, MTT, and Annexin V-FITC/PI assays in tissue samples and cultured cells. Serum levels of insulin, p-p52Shc, p-p46Shc, Shc·Grb2 complexes, p-MEK, p-ERK, and cyclin D1 were elevated in patients with EC. Expression of key proteins in the MEK/ERK pathway, including p-p52Shc, Shc·Grb2 complexes, p-MEK, p-ERK, and cyclin D1, was significantly higher in patients with advanced FIGO stage, high grade, and lymph-node metastasis and correlated positively with serum insulin concentration. Insulin promotes Ishikawa 3-H-12 cell proliferation, survival, and invasion, and these effects induced by insulin were significantly blocked by MEK inhibitor PD98059. Insulin thus promotes EC cell proliferation, survival, and invasion via the MEK/ERK pathway.
Collapse
Affiliation(s)
- Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Insulin-like growth factor-I regulates GPER expression and function in cancer cells. Oncogene 2012; 32:678-88. [PMID: 22430216 DOI: 10.1038/onc.2012.97] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Functional cross talk between insulin-like growth factor-I (IGF-I) system and estrogen signaling has been largely reported, although the underlying molecular mechanisms remain to be fully elucidated. As GPR30/GPER mediates rapid cell responses to estrogens, we evaluated the potential of IGF-I to regulate GPER expression and function in estrogen receptor (ER)α-positive breast (MCF-7) and endometrial (Ishikawa) cancer cells. We found that IGF-I transactivates the GPER promoter sequence and upregulates GPER mRNA and protein levels in both cells types. Similar data were found, at least in part, in carcinoma-associated fibroblasts. The upregulation of GPER expression by IGF-I involved the IGF-IR/PKCδ/ERK/c-fos/AP1 transduction pathway and required ERα, as ascertained by specific pharmacological inhibitors and gene-silencing. In both MCF-7 and Ishikawa cancer cells, the IGF-I-dependent cell migration required GPER and its main target gene CTGF, whereas the IGF-I-induced proliferation required both GPER and cyclin D1. Our data demonstrate that the IGF-I system regulates GPER expression and function, triggering the activation of a signaling network that leads to the migration and proliferation of cancer cells.
Collapse
|
49
|
Wang Y, Hua S, Tian W, Zhang L, Zhao J, Zhang H, Zhang W, Xue F. Mitogenic and anti-apoptotic effects of insulin in endometrial cancer are phosphatidylinositol 3-kinase/Akt dependent. Gynecol Oncol 2012; 125:734-41. [PMID: 22426488 DOI: 10.1016/j.ygyno.2012.03.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2011] [Revised: 03/07/2012] [Accepted: 03/08/2012] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To determine serum insulin levels, expression and phosphorylation of InsR, IRS-1 and Akt in endometrial cancer (EC) tissues, and to explore the correlation between them. To investigate if insulin-induced mitogenic and anti-apoptotic effects are PI3K-dependent in EC cells. METHODS Serum insulin levels were measured by radioimmunoassay. We performed RT-PCR and western blotting to evaluate the expression and activation of key proteins of PI3K/Akt pathway in 63 EC tissues. The proliferation and apoptosis rates were determined with MTT, BrdU and annexin V/PI assays. RESULTS Serum insulin levels and InsR, IRS-1 and Akt expression and phosphorylation were significantly elevated in patients with EC compared to those without EC. Additionally, levels of p-InsR, p-IRS-1, and p-Akt were significantly higher in patients with high-grade, advanced stage, deep myometrial invasion, and lymph-node metastasis. The expression and activation of InsR, IRS-1, and p-Akt were positively related with the levels of serum insulin. The insulin-induced mitogenic and anti-apoptotic effects in EC cells were blocked when cells were pre-incubated with LY294002. Ishikawa 3-H-12 cells showed increased p-Akt levels after treatment with insulin at 10(-8)M for 15min. The insulin-induced Akt activation was inhibited by LY294002 in a dose-dependent manner. CONCLUSION Insulin played an essential role in EC tumorigenesis. Activation of InsR, IRS-1, and Akt was associated with features of aggressive EC. Insulin was a mitogenic and anti-apoptotic agent for EC cells, and these effects were dependent on PI3K/Akt pathway. Decreasing insulin level and blocking the InsR-IRS-PI3K-Akt pathway could be viable preventive and therapeutic strategies for EC.
Collapse
Affiliation(s)
- Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Ferguson RD, Novosyadlyy R, Fierz Y, Alikhani N, Sun H, Yakar S, LeRoith D. Hyperinsulinemia enhances c-Myc-mediated mammary tumor development and advances metastatic progression to the lung in a mouse model of type 2 diabetes. Breast Cancer Res 2012; 14:R8. [PMID: 22226054 PMCID: PMC3496123 DOI: 10.1186/bcr3089] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 10/31/2011] [Accepted: 01/07/2012] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Hyperinsulinemia, which is common in early type 2 diabetes (T2D) as a result of the chronically insulin-resistant state, has now been identified as a specific factor which can worsen breast cancer prognosis. In breast cancer, a high rate of mortality persists due to the emergence of pulmonary metastases. METHODS Using a hyperinsulinemic mouse model (MKR+/+) and the metastatic, c-Myc-transformed mammary carcinoma cell line Mvt1, we investigated how high systemic insulin levels would affect the progression of orthotopically inoculated primary mammary tumors to lung metastases. RESULTS We found that orthotopically injected Mvt1 cells gave rise to larger mammary tumors and to a significantly higher mean number of pulmonary macrometastases in hyperinsulinemic mice over a period of six weeks (hyperinsulinemic, 19.4 ± 2.7 vs. control, 4.0 ± 1.3). When Mvt1-mediated mammary tumors were allowed to develop and metastasize for approximately two weeks and were then surgically removed, hyperinsulinemic mice demonstrated a significantly higher number of lung metastases after a four-week period (hyperinsulinemic, 25.1 ± 4.6 vs. control, 7.4 ± 0.42). Similarly, when Mvt1 cells were injected intravenously, hyperinsulinemic mice demonstrated a significantly higher metastatic burden in the lung than controls after a three-week period (hyperinsulinemic, 6.0 ± 1.63 vs. control, 1.5 ± 0.68). Analysis of Mvt1 cells both in vitro and in vivo revealed a significant up-regulation of the transcription factor c-Myc under hyperinsulinemic conditions, suggesting that hyperinsulinemia may promote c-Myc signaling in breast cancer. Furthermore, insulin-lowering therapy using the beta-adrenergic receptor agonist CL-316243 reduced metastatic burden in hyperinsulinemic mice to control levels. CONCLUSIONS Hyperinsulinemia in a mouse model promotes breast cancer metastasis to the lung. Therapies to reduce insulin levels in hyperinsulinemic patients suffering from breast cancer could lessen the likelihood of metastatic progression.
Collapse
MESH Headings
- Animals
- Blood Glucose
- Cell Line, Tumor
- Cell Proliferation
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Dioxoles/pharmacology
- Dioxoles/therapeutic use
- Female
- Hyperinsulinism/complications
- Hyperinsulinism/drug therapy
- Hyperinsulinism/metabolism
- Hyperinsulinism/pathology
- Hypoglycemic Agents/pharmacology
- Hypoglycemic Agents/therapeutic use
- Insulin/blood
- Lung Neoplasms/metabolism
- Lung Neoplasms/prevention & control
- Lung Neoplasms/secondary
- Mammary Neoplasms, Experimental/etiology
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/pathology
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Transgenic
- Neoplasm Transplantation
- Phosphorylation
- Proto-Oncogene Proteins c-akt/metabolism
- Proto-Oncogene Proteins c-myc/metabolism
- Receptor, IGF Type 1/metabolism
- Receptor, Insulin/metabolism
- Tumor Burden
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Rosalyn D Ferguson
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Ruslan Novosyadlyy
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Yvonne Fierz
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Nyosha Alikhani
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Hui Sun
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Shoshana Yakar
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Diseases, The Samuel Bronfman Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029, USA
| |
Collapse
|