1
|
Libby AE, Solt CM, Jackman MR, Sherk VD, Foright RM, Johnson GC, Nguyen TT, Breit MJ, Hulett N, Rudolph MC, Roberson PA, Wellberg EA, Jambal P, Scalzo RL, Higgins J, Kumar TR, Wierman ME, Pan Z, Shankar K, Klemm DJ, Moreau KL, Kohrt WM, MacLean PS. Effects of follicle-stimulating hormone on energy balance and tissue metabolic health after loss of ovarian function. Am J Physiol Endocrinol Metab 2024; 326:E626-E639. [PMID: 38536037 PMCID: PMC11208003 DOI: 10.1152/ajpendo.00400.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/29/2024] [Accepted: 03/20/2024] [Indexed: 05/01/2024]
Abstract
Loss of ovarian function imparts increased susceptibility to obesity and metabolic disease. These effects are largely attributed to decreased estradiol (E2), but the role of increased follicle-stimulating hormone (FSH) in modulating energy balance has not been fully investigated. Previous work that blocked FSH binding to its receptor in mice suggested this hormone may play a part in modulating body weight and energy expenditure after ovariectomy (OVX). We used an alternate approach to isolate the individual and combined contributions of FSH and E2 in mediating energy imbalance and changes in tissue-level metabolic health. Female Wistar rats were ovariectomized and given the gonadotropin releasing hormone (GnRH) antagonist degarelix to suppress FSH production. E2 and FSH were then added back individually and in combination for a period of 3 wk. Energy balance, body mass composition, and transcriptomic profiles of individual tissues were obtained. In contrast to previous studies, suppression and replacement of FSH in our paradigm had no effect on body weight, body composition, food intake, or energy expenditure. We did, however, observe organ-specific effects of FSH that produced unique transcriptomic signatures of FSH in retroperitoneal white adipose tissue. These included reductions in biological processes related to lipogenesis and carbohydrate transport. In addition, rats administered FSH had reduced liver triglyceride concentration (P < 0.001), which correlated with FSH-induced changes at the transcriptomic level. Although not appearing to modulate energy balance after loss of ovarian function in rats, FSH may still impart tissue-specific effects in the liver and white adipose tissue that might affect the metabolic health of those organs.NEW & NOTEWORTHY We find no effect of follicle-stimulating hormone (FSH) on energy balance using a novel model in which rats are ovariectomized, subjected to gonadotropin-releasing hormone antagonism, and systematically given back FSH by osmotic pump. However, tissue-specific effects of FSH on adipose tissue and liver were observed in this study. These include unique transcriptomic signatures induced by the hormone and a stark reduction in hepatic triglyceride accumulation.
Collapse
Affiliation(s)
- Andrew E Libby
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Claudia M Solt
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Matthew R Jackman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Vanessa D Sherk
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Center for Scientific Review, National Institutes of Health, Bethesda, Maryland, United States
| | - Rebecca M Foright
- Department of Anatomy and Cell Biology, University of Kansas Medical Campus, Kansas City, Kansas, United States
| | - Ginger C Johnson
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Thi-Tina Nguyen
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Matthew J Breit
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Nicholas Hulett
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Michael C Rudolph
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Campus, Oklahoma City, Oklahoma, United States
| | - Paul A Roberson
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Elizabeth A Wellberg
- Stephenson Cancer Center, University of Oklahoma Health Sciences Campus, Oklahoma City, Oklahoma, United States
| | - Purevsuren Jambal
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Rebecca L Scalzo
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Janine Higgins
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - T Rajendra Kumar
- Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Margaret E Wierman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Zhaoxing Pan
- Section of Endocrinology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kartik Shankar
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Dwight J Klemm
- Cardiovascular Pulmonary Research Laboratory, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Kerrie L Moreau
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Wendy M Kohrt
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Paul S MacLean
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
2
|
Kanwore K, Kanwore K, Guo X, Xia Y, Zhou H, Zhang L, Adzika GK, Joseph AA, Abiola AA, Mu P, Kambey PA, Noah MLN, Gao D. Testosterone upregulates glial cell line-derived neurotrophic factor (GDNF) and promotes neuroinflammation to enhance glioma cell survival and proliferation. Inflamm Regen 2023; 43:49. [PMID: 37833789 PMCID: PMC10571473 DOI: 10.1186/s41232-023-00300-7] [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: 05/10/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Testosterone contributes to male organism development, such as bone density, muscle development, and fat repartition. Estrogen (derived from testosterone) also contributes to female reproductive system development. Here, we investigated the effect of testosterone on glioma cells and brain neuron inflammation essential for cancer development and progression. METHODS The human astrocyte and glioma cell lines were treated with 6 ng/ml exogenous testosterone in vitro. We performed cell counting kit-8, transwell, and wound healing assays to determine the effect of testosterone on glioma cell proliferation, migration, and invasion. The glioma cells were injected into the xenograft and treated with 5 µl concentrated testosterone. Transcriptional suppression of glial cell line-derived neurotrophic factor (GDNF) was performed to evaluate brain neuron inflammation and survival. The tumor tissues were assessed by hematoxylin-eosin staining and immunohistochemistry. RESULTS Testosterone upregulates GDNF to stimulate proliferation, migration, and invasion of glioma cells. Pathologically, the augmentation of GDNF and cyclophilin A contributed to neuroprotection when treated with testosterone. Our investigation showed that testosterone contributes to brain neuron and astrocyte inflammation through the upregulation of nuclear factor erythroid 2-related factor 2 (NRF2), glial fibrillary acid protein (GFAP), and sirtuin 5 (SIRT5), resulting in pro-inflammatory macrophages recruitments into the neural microenvironment. Mechanically, testosterone treatment regulates GDNF translocation from the glioma cells and astrocyte nuclei to the cytoplasm. CONCLUSION Testosterone upregulates GDNF in glioma cells and astrocytes essential for microglial proliferation, migration, and invasion. Testosterone contributes to brain tumor growth via GDNF and inflammation. The contribution of testosterone, macrophages, and astrocytes, in old neuron rescue, survival, and proliferation. During brain neuron inflammation, the organism activates and stimulates the neuron rescue through the enrichment of the old neuron microenvironment with growth factors such as GDNF, BDNF, SOX1/2, and MAPK secreted by the surrounding neurons and glial cells to maintain the damaged neuron by inflammation alive even if the axon is dead. The immune response also contributes to brain cell survival through the secretion of proinflammatory cytokines, resulting in inflammation maintenance. The rescued old neuron interaction with infiltrated macrophages contributes to angiogenesis to supplement the old neuron with more nutrients leading to metabolism activation and surrounding cell uncontrollable cell growth.
Collapse
Affiliation(s)
- Kouminin Kanwore
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Konimpo Kanwore
- Mixed Faculty of Medicine and Pharmacy, University of Lomé, Lomé, Togo
| | - Xiaoxiao Guo
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ying Xia
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Han Zhou
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Lin Zhang
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | | | | | - Ayanlaja Abdulrahman Abiola
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Peipei Mu
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Piniel Alphayo Kambey
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | | | - DianShuai Gao
- Public Experimental Research Center, Department of Neurobiology and Anatomy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
3
|
Leo J, Dondossola E, Basham KJ, Wilson NR, Alhalabi O, Gao J, Kurnit KC, White MG, McQuade JL, Westin SN, Wellberg EA, Frigo DE. Stranger Things: New Roles and Opportunities for Androgen Receptor in Oncology Beyond Prostate Cancer. Endocrinology 2023; 164:bqad071. [PMID: 37154098 PMCID: PMC10413436 DOI: 10.1210/endocr/bqad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
The androgen receptor (AR) is one of the oldest therapeutic targets in oncology and continues to dominate the treatment landscape for advanced prostate cancer, where nearly all treatment regimens include some form of AR modulation. In this regard, AR remains the central driver of prostate cancer cell biology. Emerging preclinical and clinical data implicate key roles for AR in additional cancer types, thereby expanding the importance of this drug target beyond prostate cancer. In this mini-review, new roles for AR in other cancer types are discussed as well as their potential for treatment with AR-targeted agents. Our understanding of these additional functions for AR in oncology expand this receptor's potential as a therapeutic target and will help guide the development of new treatment approaches.
Collapse
Affiliation(s)
- Javier Leo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Eleonora Dondossola
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kaitlin J Basham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Nathaniel R Wilson
- Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Omar Alhalabi
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianjun Gao
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Katherine C Kurnit
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, The University of Chicago, Chicago, IL 60637, USA
| | - Michael G White
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jennifer L McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Elizabeth A Wellberg
- Department of Pathology, Harold Hamm Diabetes Center, and Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Daniel E Frigo
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77204, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
4
|
Zhao H, Wang Q, Hu L, Xing S, Gong H, Liu Z, Qin P, Xu J, Du J, Ai W, Peng S, Li Y. Dynamic Alteration of the Gut Microbiota Associated with Obesity and Intestinal Inflammation in Ovariectomy C57BL/6 Mice. Int J Endocrinol 2022; 2022:6600158. [PMID: 35103060 PMCID: PMC8800624 DOI: 10.1155/2022/6600158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Estrogen is a critical hormone that is mainly produced by the ovary in females. Estrogen deficiency leads to various syndromes and diseases, partly due to gut microbiota alterations. Previous studies have shown that estrogen deficiency affects the gut microbiota at 6-8 weeks after ovariectomy, but the immediate effect of estrogen deficiency on the gut microbiota remains poorly understood. METHODS To investigate the short time and dynamic effects of decreased estrogen levels on the gut microbiota and their potential impact on estrogen deficiency-related diseases, we performed metagenomic sequencing of 260 fecal samples from 50 ovariectomy (OVX) and 15 control C57BL/6 female mice at four time points after surgery. RESULTS We found that seven gut microbiota species, including E. coli, Parabacteroides unclassified, Lachnospiraceae bacterium 8_1_57FAA, Bacteroides uniformis, Veillonella unclassified, Bacteroides xylanisolvens, and Firmicutes bacterium M10_2, were abundant in OVX mice. The abundance of these species increased with time after OVX surgery. The relative abundance of the opportunistic pathogen E. coli and the Crohn's disease-related Veillonella spp. was significantly correlated with mouse weight gain in the OVX group. Butyrate production and the Entner-Doudoroff pathway were significantly enriched in the control mouse group, while the degradation of glutamic acid and aspartic acid was enriched in the OVX mouse group. As the time after OVX surgery increased, the bacterial species and metabolic pathways significantly changed and tended to suggest an inflammatory environment, indicating a subhealthy state of the gut microbiota in the OVX mouse group. CONCLUSIONS Taken together, our results show that the dynamic gut microbiota profile alteration caused by estrogen deficiency is related to obesity and inflammation, which may lead to immune and metabolic disorders. This study provides new clues for the treatment of estrogen deficiency-related diseases.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Clinical Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Qi Wang
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730010, Gansu, China
| | - Liqiu Hu
- Department of Spine Surgery, Shenzhen People's Hospital, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Shaojun Xing
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Hui Gong
- Department of Clinical Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Zhe Liu
- Department of Computer Sciences, City University of Hong Kong, Hong Kong 999077, China
| | - Panpan Qin
- Qingdao-Europe Advanced Institute for Life Sciences, BGI-Shenzhen, Qingdao 266555, China
- Shenzhen Key Laboratory of Human Commensal Microorganisms and Health Research, BGI-Shenzhen, Shenzhen 518083, Guangdong, China
| | - Jie Xu
- Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou 730010, Gansu, China
| | - Jihui Du
- Department of Clinical Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| | - Wen Ai
- Medical Research Center of Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518102, China
| | - Songlin Peng
- Department of Spine Surgery, Shenzhen People's Hospital, Jinan University Second College of Medicine, Shenzhen 518020, China
| | - Yifan Li
- Department of Clinical Laboratory, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen 518052, China
| |
Collapse
|
5
|
Wellberg EA, Corleto KA, Checkley LA, Jindal S, Johnson G, Higgins JA, Obeid S, Anderson SM, Thor AD, Schedin PJ, MacLean PS, Giles ED. Preventing ovariectomy-induced weight gain decreases tumor burden in rodent models of obesity and postmenopausal breast cancer. Breast Cancer Res 2022; 24:42. [PMID: 35725493 PMCID: PMC9208221 DOI: 10.1186/s13058-022-01535-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 06/01/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Obesity and adult weight gain are linked to increased breast cancer risk and poorer clinical outcomes in postmenopausal women, particularly for hormone-dependent tumors. Menopause is a time when significant weight gain occurs in many women, and clinical and preclinical studies have identified menopause (or ovariectomy) as a period of vulnerability for breast cancer development and promotion. METHODS We hypothesized that preventing weight gain after ovariectomy (OVX) may be sufficient to prevent the formation of new tumors and decrease growth of existing mammary tumors. We tested this hypothesis in a rat model of obesity and carcinogen-induced postmenopausal mammary cancer and validated our findings in a murine xenograft model with implanted human tumors. RESULTS In both models, preventing weight gain after OVX significantly decreased obesity-associated tumor development and growth. Importantly, we did not induce weight loss in these animals, but simply prevented weight gain. In both lean and obese rats, preventing weight gain reduced visceral fat accumulation and associated insulin resistance. Similarly, the intervention decreased circulating tumor-promoting growth factors and inflammatory cytokines (i.e., BDNF, TNFα, FGF-2), with greater effects in obese compared to lean rats. In obese rats, preventing weight gain decreased adipocyte size, adipose tissue macrophage infiltration, reduced expression of the tumor-promoting growth factor FGF-1 in mammary adipose, and reduced phosphorylated FGFR indicating reduced FGF signaling in tumors. CONCLUSIONS Together, these findings suggest that the underlying mechanisms associated with the anti-tumor effects of weight maintenance are multi-factorial, and that weight maintenance during the peri-/postmenopausal period may be a viable strategy for reducing obesity-associated breast cancer risk and progression in women.
Collapse
Affiliation(s)
- Elizabeth A. Wellberg
- grid.266902.90000 0001 2179 3618Department of Pathology, Harold Hamm Diabetes Center, and Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Karen A. Corleto
- grid.264756.40000 0004 4687 2082Department of Nutrition, Texas A&M University, College Station, TX USA
| | - L. Allyson Checkley
- grid.430503.10000 0001 0703 675XDivisions of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Sonali Jindal
- grid.5288.70000 0000 9758 5690Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR USA ,grid.5288.70000 0000 9758 5690Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
| | - Ginger Johnson
- grid.430503.10000 0001 0703 675XDivisions of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.430503.10000 0001 0703 675XAnschutz Health and Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Janine A. Higgins
- grid.430503.10000 0001 0703 675XDepartment of Pediatrics, Endocrinology Section, University of Colorado Anschutz Medical Campus, Aurora, CO USA
| | - Sarina Obeid
- grid.264756.40000 0004 4687 2082Department of Nutrition, Texas A&M University, College Station, TX USA
| | - Steven M. Anderson
- grid.430503.10000 0001 0703 675XDepartment of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.499234.10000 0004 0433 9255University of Colorado Cancer Center, Aurora, CO USA
| | - Ann D. Thor
- grid.430503.10000 0001 0703 675XDepartment of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.499234.10000 0004 0433 9255University of Colorado Cancer Center, Aurora, CO USA
| | - Pepper J. Schedin
- grid.5288.70000 0000 9758 5690Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR USA ,grid.5288.70000 0000 9758 5690Knight Cancer Institute, Oregon Health & Science University, Portland, OR USA
| | - Paul S. MacLean
- grid.430503.10000 0001 0703 675XDivisions of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.430503.10000 0001 0703 675XAnschutz Health and Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, CO USA ,grid.499234.10000 0004 0433 9255University of Colorado Cancer Center, Aurora, CO USA
| | - Erin D. Giles
- grid.214458.e0000000086837370School of Kinesiology, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
6
|
Scalzo RL, Foright RM, Hull SE, Knaub LA, Johnson-Murguia S, Kinanee F, Kaplan J, Houck JA, Johnson G, Sharp RR, Gillen AE, Jones KL, Zhang AMY, Johnson JD, MacLean PS, Reusch JEB, Wright-Hobart S, Wellberg EA. Breast Cancer Endocrine Therapy Promotes Weight Gain With Distinct Adipose Tissue Effects in Lean and Obese Female Mice. Endocrinology 2021; 162:bqab174. [PMID: 34410380 PMCID: PMC8455348 DOI: 10.1210/endocr/bqab174] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Indexed: 12/19/2022]
Abstract
Breast cancer survivors treated with tamoxifen and aromatase inhibitors report weight gain and have an elevated risk of type 2 diabetes, especially if they have obesity. These patient experiences are inconsistent with, preclinical studies using high doses of tamoxifen which reported acute weight loss. We investigated the impact of breast cancer endocrine therapies in a preclinical model of obesity and in a small group of breast adipose tissue samples from women taking tamoxifen to understand the clinical findings. Mature female mice were housed at thermoneutrality and fed either a low-fat/low-sucrose (LFLS) or a high-fat/high-sucrose (HFHS) diet. Consistent with the high expression of Esr1 observed in mesenchymal stem cells from adipose tissue, endocrine therapy was associated with adipose accumulation and more preadipocytes compared with estrogen-treated control mice but resulted in fewer adipocyte progenitors only in the context of HFHS. Analysis of subcutaneous adipose stromal cells revealed diet- and treatment-dependent effects of endocrine therapies on various cell types and genes, illustrating the complexity of adipose tissue estrogen receptor signaling. Breast cancer therapies supported adipocyte hypertrophy and associated with hepatic steatosis, hyperinsulinemia, and glucose intolerance, particularly in obese females. Current tamoxifen use associated with larger breast adipocyte diameter only in women with obesity. Our translational studies suggest that endocrine therapies may disrupt adipocyte progenitors and support adipocyte hypertrophy, potentially leading to ectopic lipid deposition that may be linked to a greater type 2 diabetes risk. Monitoring glucose tolerance and potential interventions that target insulin action should be considered for some women receiving life-saving endocrine therapies for breast cancer.
Collapse
Affiliation(s)
- Rebecca L Scalzo
- Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Center for Women’s Health Research; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Rebecca M Foright
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Sara E Hull
- Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Leslie A Knaub
- Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stevi Johnson-Murguia
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Research Center, Oklahoma City, OK 73104, USA
| | - Fotobari Kinanee
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jeffrey Kaplan
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Julie A Houck
- Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ginger Johnson
- Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rachel R Sharp
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Research Center, Oklahoma City, OK 73104, USA
| | - Austin E Gillen
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Research Center, Oklahoma City, OK 73104, USA
| | - Anni M Y Zhang
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - James D Johnson
- Diabetes Research Group, Life Sciences Institute, Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Paul S MacLean
- Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Center for Women’s Health Research; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jane E B Reusch
- Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Center for Women’s Health Research; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Sabrina Wright-Hobart
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Elizabeth A Wellberg
- Center for Women’s Health Research; University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, Harold Hamm Diabetes Research Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
7
|
Obesity and Androgen Receptor Signaling: Associations and Potential Crosstalk in Breast Cancer Cells. Cancers (Basel) 2021; 13:cancers13092218. [PMID: 34066328 PMCID: PMC8125357 DOI: 10.3390/cancers13092218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/18/2021] [Accepted: 04/03/2021] [Indexed: 12/24/2022] Open
Abstract
Obesity is an increasing health challenge and is recognized as a breast cancer risk factor. Although obesity-related breast cancer mechanisms are not fully understood, this association has been linked to impaired hormone secretion by the dysfunctional obese adipose tissue (hyperplasic and hypertrophic adipocytes). Among these hormones, altered production of androgens and adipokines is observed, and both, are independently associated with breast cancer development. In this review, we describe and comment on the relationships reported between these factors and breast cancer, focusing on the biological associations that have helped to unveil the mechanisms by which signaling from androgens and adipokines modifies the behavior of mammary epithelial cells. Furthermore, we discuss the potential crosstalk between the two most abundant adipokines produced by the adipose tissue (adiponectin and leptin) and the androgen receptor, an emerging marker in breast cancer. The identification and understanding of interactions among adipokines and the androgen receptor in cancer cells are necessary to guide the development of new therapeutic approaches in order to prevent and cure obesity and breast cancer.
Collapse
|
8
|
Giles ED, Wellberg EA. Preclinical Models to Study Obesity and Breast Cancer in Females: Considerations, Caveats, and Tools. J Mammary Gland Biol Neoplasia 2020; 25:237-253. [PMID: 33146844 PMCID: PMC8197449 DOI: 10.1007/s10911-020-09463-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/09/2020] [Indexed: 12/28/2022] Open
Abstract
Obesity increases the risk for breast cancer and is associated with poor outcomes for cancer patients. A variety of rodent models have been used to investigate these relationships; however, key differences in experimental approaches, as well as unique aspects of rodent physiology lead to variability in how these valuable models are implemented. We combine expertise in the development and implementation of preclinical models of obesity and breast cancer to disseminate effective practices for studies that integrate these fields. In this review, we share, based on our experience, key considerations for model selection, highlighting important technical nuances and tips for use of preclinical models in studies that integrate obesity with breast cancer risk and progression. We describe relevant mouse and rat paradigms, specifically highlighting differences in breast tumor subtypes, estrogen production, and strategies to manipulate hormone levels. We also outline options for diet composition and housing environments to promote obesity in female rodents. While we have applied our experience to understanding obesity-associated breast cancer, the experimental variables we incorporate have relevance to multiple fields that investigate women's health.
Collapse
Affiliation(s)
- Erin D Giles
- Department of Nutrition, Texas A&M University, College Station, TX, USA.
| | - Elizabeth A Wellberg
- Department of Pathology, University of Oklahoma Health Science Center, Stephenson Cancer Center, Harold Hamm Diabetes Center, Oklahoma City, OK, USA
| |
Collapse
|
9
|
Sakle NS, More SA, Mokale SN. A network pharmacology-based approach to explore potential targets of Caesalpinia pulcherima: an updated prototype in drug discovery. Sci Rep 2020; 10:17217. [PMID: 33057155 PMCID: PMC7560621 DOI: 10.1038/s41598-020-74251-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 08/28/2020] [Indexed: 01/19/2023] Open
Abstract
Caesalpinia pulcherima (CP) is a traditional herb used for the treatment of asthma, bronchitis, cancer, anti-bacterial, anti-fungal and as abortifacient. In the present study, bioactive components and potential targets in the treatment of breast cancer validated through in silico, in vitro and in vivo approach. The results for the analysis were as among 29 components, only four components were found active for further study which proved the use of CP as a multi-target herb for betterment of clinical uses. The results found by PPI states that our network has significant interactions which include the ESR-1, ESR-2, ESRRA, MET, VEGF, FGF, PI3K, PDK-1, MAPK, PLK-1, NEK-2, and GRK. Compound-target network involves 4 active compound and 150 target genes which elucidate the mechanisms of drug action in breast cancer treatment. Furthermore, on the basis of the above results the important proteins were fetched for the docking study which helps in predicting the possible interaction between components and targets. The results of the western blotting showed that CP regulates ER and EGFR expression in MCF-7 cell. In addition to this animal experimentation showed that CP significantly improved immunohistological status in MNU induced carcinoma rats. Network pharmacology approach not only helps us to confirm the study of the chosen target but also gave an idea of compound-target network as well as pathways associated to the CP for treating the complex metabolic condition as breast cancer and they importance for experimental verification.
Collapse
Affiliation(s)
- Nikhil S Sakle
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, 431001, India
| | - Shweta A More
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, 431001, India
| | - Santosh N Mokale
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Aurangabad, Maharashtra, 431001, India.
| |
Collapse
|
10
|
Wellberg EA, Kabos P, Gillen AE, Jacobsen BM, Brechbuhl HM, Johnson SJ, Rudolph MC, Edgerton SM, Thor AD, Anderson SM, Elias A, Zhou XK, Iyengar NM, Morrow M, Falcone DJ, El-Hely O, Dannenberg AJ, Sartorius CA, MacLean PS. FGFR1 underlies obesity-associated progression of estrogen receptor-positive breast cancer after estrogen deprivation. JCI Insight 2018; 3:120594. [PMID: 30046001 PMCID: PMC6124402 DOI: 10.1172/jci.insight.120594] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/19/2018] [Indexed: 01/07/2023] Open
Abstract
Obesity increases breast cancer mortality by promoting resistance to therapy. Here, we identified regulatory pathways in estrogen receptor-positive (ER-positive) tumors that were shared between patients with obesity and those with resistance to neoadjuvant aromatase inhibition. Among these was fibroblast growth factor receptor 1 (FGFR1), a known mediator of endocrine therapy resistance. In a preclinical model with patient-derived ER-positive tumors, diet-induced obesity promoted a similar gene expression signature and sustained the growth of FGFR1-overexpressing tumors after estrogen deprivation. Tumor FGFR1 phosphorylation was elevated with obesity and predicted a shorter disease-free and disease-specific survival for patients treated with tamoxifen. In both human and mouse mammary adipose tissue, FGF1 ligand expression was associated with metabolic dysfunction, weight gain, and adipocyte hypertrophy, implicating the impaired response to a positive energy balance in growth factor production within the tumor niche. In conjunction with these studies, we describe a potentially novel graft-competent model that can be used with patient-derived tissue to elucidate factors specific to extrinsic (host) and intrinsic (tumor) tissue that are critical for obesity-associated tumor promotion. Taken together, we demonstrate that obesity and excess energy establish a tumor environment with features of endocrine therapy resistance and identify a role for ligand-dependent FGFR1 signaling in obesity-associated breast cancer progression.
Collapse
Affiliation(s)
| | - Peter Kabos
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Austin E. Gillen
- University of Colorado School of Medicine, RNA Bioscience Initiative, Aurora, Colorado, USA
| | - Britta M. Jacobsen
- Department of Pathology and
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Heather M. Brechbuhl
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | - Michael C. Rudolph
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | | | | | | | - Anthony Elias
- Division of Medical Oncology, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| | - Xi Kathy Zhou
- Department of Healthcare Policy and Research, Weill Cornell Medical College, New York, New York, USA
| | - Neil M. Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center (MSKCC), New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Monica Morrow
- Department of Surgery, MSKCC, New York, New York, USA
| | - Domenick J. Falcone
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical Center, New York, New York, USA
| | - Omar El-Hely
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | | | | | - Paul S. MacLean
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
11
|
Giles ED, Jindal S, Wellberg EA, Schedin T, Anderson SM, Thor AD, Edwards DP, MacLean PS, Schedin P. Metformin inhibits stromal aromatase expression and tumor progression in a rodent model of postmenopausal breast cancer. Breast Cancer Res 2018; 20:50. [PMID: 29898754 PMCID: PMC6000949 DOI: 10.1186/s13058-018-0974-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 04/30/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity and type II diabetes are linked to increased breast cancer risk in postmenopausal women. Patients treated with the antidiabetic drug metformin for diabetes or metabolic syndrome have reduced breast cancer risk, a greater pathologic complete response to neoadjuvant therapy, and improved breast cancer survival. We hypothesized that metformin may be especially effective when targeted to the menopausal transition, as this is a lifecycle window when weight gain and metabolic syndrome increase, and is also when the risk for obesity-related breast cancer increases. METHODS Here, we used an 1-methyl-1-nitrosourea (MNU)-induced mammary tumor rat model of estrogen receptor (ER)-positive postmenopausal breast cancer to evaluate the long-term effects of metformin administration on metabolic and tumor endpoints. In this model, ovariectomy (OVX) induces rapid weight gain, and an impaired whole-body response to excess calories contributes to increased tumor glucose uptake and increased tumor proliferation. Metformin treatment was initiated in tumor-bearing animals immediately prior to OVX and maintained for the duration of the study. RESULTS Metformin decreased the size of existing mammary tumors and inhibited new tumor formation without changing body weight or adiposity. Decreased lipid accumulation in the livers of metformin-treated animals supports the ability of metformin to improve overall metabolic health. We also found a decrease in the number of aromatase-positive, CD68-positive macrophages within the tumor microenvironment, suggesting that metformin targets the immune microenvironment in addition to improving whole-body metabolism. CONCLUSIONS These findings suggest that peri-menopause/menopause represents a unique window of time during which metformin may be highly effective in women with established, or at high risk for developing, breast cancer.
Collapse
Affiliation(s)
- Erin D Giles
- Department of Nutrition & Food Science, Texas A&M University, 373 Olsen Blvd; 2253 TAMU, College Station, TX, 77843, USA.
| | - Sonali Jindal
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Rd, Mailing Code: L215, Portland, OR, 97239, USA
| | - Elizabeth A Wellberg
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Troy Schedin
- Department of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Steven M Anderson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ann D Thor
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dean P Edwards
- Departments of Molecular & Cellular Biology and Pathology Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Paul S MacLean
- Anschutz Health & Wellness Center, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.,Department of Medicine, Divisions of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.,Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, 3181 S.W. Sam Jackson Park Rd, Mailing Code: L215, Portland, OR, 97239, USA.,Knight Cancer Institute, Oregon Health & Science University, 1130 NW 22nd Ave #100, Portland, OR, 97239, USA
| |
Collapse
|
12
|
Hagen KB, Aas T, Kvaløy JT, Søiland H, Lind R. Diet in women with breast cancer compared to healthy controls – What is the difference? Eur J Oncol Nurs 2018; 32:20-24. [DOI: 10.1016/j.ejon.2017.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 11/05/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
|
13
|
Christenson JL, Trepel JB, Ali HY, Lee S, Eisner JR, Baskin-Bey ES, Elias AD, Richer JK. Harnessing a Different Dependency: How to Identify and Target Androgen Receptor-Positive Versus Quadruple-Negative Breast Cancer. Discov Oncol 2018; 9:82-94. [PMID: 29340907 DOI: 10.1007/s12672-017-0314-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/07/2017] [Indexed: 12/12/2022] Open
Abstract
The androgen receptor (AR) is a promising therapeutic target for a subset of triple-negative breast cancers (TNBCs) in which AR is expressed. However, the mechanistic action of AR and the degree to which primary and metastatic tumors depend on AR, both before and after conventional treatment, remain to be defined. We discuss preclinical and clinical data for AR+ TNBC, the difficulties in monitoring AR protein levels, new methods for determining AR status, the influence of AR on "stemness" in the context of TNBC, the role of combined inhibition of sex steroid production and AR, and the role of AR in regulation of the immune system. Although the exact role of AR in subsets of TNBC is still being characterized, new therapies that target AR and the production of androgens may provide additional options for patients with TNBC for whom chemotherapy is currently the sole treatment option.
Collapse
Affiliation(s)
- Jessica L Christenson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jane B Trepel
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Sunmin Lee
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | | | - Anthony D Elias
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Jennifer K Richer
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|