Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
1.
Am J Physiol Endocrinol Metab ; 326(5): E626-E639, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38536037

RESUMEN

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.


Asunto(s)
Metabolismo Energético , Estradiol , Hormona Folículo Estimulante , Ovariectomía , Ratas Wistar , Animales , Femenino , Metabolismo Energético/efectos de los fármacos , Ratas , Hormona Folículo Estimulante/metabolismo , Estradiol/farmacología , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ovario/efectos de los fármacos , Ovario/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Hígado/metabolismo , Hígado/efectos de los fármacos , Transcriptoma/efectos de los fármacos
2.
Nat Rev Endocrinol ; 20(1): 16-26, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37783846

RESUMEN

Breast cancer is the most common invasive malignancy in the world, with millions of survivors living today. Type 2 diabetes mellitus (T2DM) is also a globally prevalent disease that is a widely studied risk factor for breast cancer. Most breast tumours express the oestrogen receptor and are treated with systemic therapies designed to disrupt oestrogen-dependent signalling. Since the advent of targeted endocrine therapy six decades ago, the mortality from breast cancer has steadily declined; however, during the past decade, an elevated risk of T2DM after breast cancer treatment has been reported, particularly for those who received endocrine therapy. In this Review, we highlight key events in the history of endocrine therapies, beginning with the development of tamoxifen. We also summarize the sequence of reported adverse metabolic effects, which include dyslipidaemia, hepatic steatosis and impaired glucose tolerance. We discuss the limitations of determining a causal role for breast cancer treatments in T2DM development from epidemiological data and describe informative preclinical studies that suggest complex mechanisms through which endocrine therapy might drive T2DM risk and progression. We also reinforce the life-saving benefits of endocrine therapy and highlight the need for better predictive biomarkers of T2DM risk and preventive strategies for the growing population of breast cancer survivors.


Asunto(s)
Neoplasias de la Mama , Supervivientes de Cáncer , Diabetes Mellitus Tipo 2 , Humanos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/epidemiología , Neoplasias de la Mama/metabolismo , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/epidemiología , Tamoxifeno/efectos adversos , Estrógenos/metabolismo , Estrógenos/uso terapéutico
3.
Front Endocrinol (Lausanne) ; 14: 1217875, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37800138

RESUMEN

Obesity and type 2 diabetes are chronic metabolic diseases that impact tens to hundreds of millions of adults, especially in developed countries. Each condition is associated with an elevated risk of breast cancer and with a poor prognosis after treatment. The mechanisms connecting poor metabolic health to breast cancer are numerous and include hyperinsulinemia, inflammation, excess nutrient availability, and adipose tissue dysfunction. Here, we focus on adipose tissue, highlighting important roles for both adipocytes and fibroblasts in breast cancer progression. One potentially important mediator of adipose tissue effects on breast cancer is the fibroblast growth factor receptor (FGFR) signaling network. Among the many roles of FGFR signaling, we postulate that key mechanisms driving aggressive breast cancer include epithelial-to-mesenchymal transition and cellular metabolic reprogramming. We also pose existing questions that may help better understand breast cancer biology in people with obesity, type 2 diabetes, and poor metabolic health.


Asunto(s)
Neoplasias de la Mama , Diabetes Mellitus Tipo 2 , Humanos , Femenino , Neoplasias de la Mama/metabolismo , Diabetes Mellitus Tipo 2/complicaciones , Tejido Adiposo/metabolismo , Adipocitos/metabolismo , Obesidad/metabolismo
4.
Breast Cancer Res ; 25(1): 99, 2023 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-37608351

RESUMEN

BACKGROUND: Obesity increases breast cancer risk and breast cancer-specific mortality, particularly for people with estrogen receptor (ER)-positive tumors. Body mass index (BMI) is used to define obesity, but it may not be the best predictor of breast cancer risk or prognosis on an individual level. Adult weight gain is an independent indicator of breast cancer risk. Our previous work described a murine model of obesity, ER-positive breast cancer, and weight gain and identified fibroblast growth factor receptor (FGFR) as a potential driver of tumor progression. During adipose tissue expansion, the FGF1 ligand is produced by hypertrophic adipocytes as a stimulus to stromal preadipocytes that proliferate and differentiate to provide additional lipid storage capacity. In breast adipose tissue, FGF1 production may stimulate cancer cell proliferation and tumor progression. METHODS: We explored the effects of FGF1 on ER-positive endocrine-sensitive and resistant breast cancer and compared that to the effects of the canonical ER ligand, estradiol. We used untargeted proteomics, specific immunoblot assays, gene expression profiling, and functional metabolic assessments of breast cancer cells. The results were validated in tumors from obese mice and breast cancer datasets from women with obesity. RESULTS: FGF1 stimulated ER phosphorylation independently of estradiol in cells that grow in obese female mice after estrogen deprivation treatment. Phospho- and total proteomic, genomic, and functional analyses of endocrine-sensitive and resistant breast cancer cells show that FGF1 promoted a cellular phenotype characterized by glycolytic metabolism. In endocrine-sensitive but not endocrine-resistant breast cancer cells, mitochondrial metabolism was also regulated by FGF1. Comparison of gene expression profiles indicated that tumors from women with obesity shared hallmarks with endocrine-resistant breast cancer cells. CONCLUSIONS: Collectively, our data suggest that one mechanism by which obesity and weight gain promote breast cancer progression is through estrogen-independent ER activation and cancer cell metabolic reprogramming, partly driven by FGF/FGFR. The first-line treatment for many patients with ER-positive breast cancer is inhibition of estrogen synthesis using aromatase inhibitors. In women with obesity who are experiencing weight gain, locally produced FGF1 may activate ER to promote cancer cell metabolic reprogramming and tumor progression independently of estrogen.


Asunto(s)
Neoplasias de la Mama , Factor 1 de Crecimiento de Fibroblastos , Receptores de Estrógenos , Animales , Femenino , Ratones , Estradiol , Estrógenos , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Ligandos , Obesidad/complicaciones , Proteómica , Receptores de Estrógenos/genética , Aumento de Peso , Neoplasias de la Mama/metabolismo
5.
Biochem Pharmacol ; 216: 115763, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37625554

RESUMEN

Anti-hormone therapies are not efficacious for reducing the incidence of triple negative breast cancer (TNBC), which lacks both estrogen and progesterone receptors. While the etiology of this aggressive breast cancer subtype is unclear, visceral obesity is a strong risk factor for both pre- and post-menopausal cases. The mechanism by which excessive deposition of visceral adipose tissue (VAT) promotes the malignant transformation of hormone receptor-negative mammary epithelial cells is currently unknown. We developed a novel in vitro system of malignant transformation in which non-tumorigenic human breast epithelial cells (MCF-10A) grow in soft agar when cultured with factors released from VAT. These cells, which acquire the capacity for 3D growth, show elevated aryl hydrocarbon receptor (AhR) protein and AhR target genes, suggesting that AhR activity may drive malignant transformation by VAT. AhR is a ligand-dependent transcription factor that generates biological responses to exogenous carcinogens and to the endogenous tryptophan pathway metabolite, kynurenine. The serum kynurenine to tryptophan ratio has been shown to be elevated in patients with obesity. Herein, we demonstrate that AhR inhibitors or knockdown of AhR in MCF-10A cells prevents VAT-induced malignant transformation. Specifically, VAT-induced transformation is inhibited by Kyn-101, an inhibitor for the endogenous ligand binding site of AhR. Mass spectrometry analysis demonstrates that adipocytes metabolize tryptophan and release kynurenine, which is taken up by MCF-10A cells and activates the AhR to induce CYP1B1 and promote malignant transformation. This novel hormone receptor-independent mechanism of malignant transformation suggests targeting AhR for TNBC prevention in the context of visceral adiposity.


Asunto(s)
Quinurenina , Neoplasias de la Mama Triple Negativas , Humanos , Adipocitos/metabolismo , Células Epiteliales/metabolismo , Hormonas/metabolismo , Quinurenina/metabolismo , Ligandos , Receptores de Hidrocarburo de Aril/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Triptófano/metabolismo
6.
Endocrinology ; 164(6)2023 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-37154098

RESUMEN

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.


Asunto(s)
Antineoplásicos , Neoplasias de la Próstata Resistentes a la Castración , Neoplasias de la Próstata , Humanos , Masculino , Antagonistas de Receptores Androgénicos/farmacología , Antagonistas de Receptores Androgénicos/uso terapéutico , Antineoplásicos/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal
7.
J Natl Cancer Inst Monogr ; 2023(61): 12-29, 2023 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-37139973

RESUMEN

The obesity pandemic currently affects more than 70 million Americans and more than 650 million individuals worldwide. In addition to increasing susceptibility to pathogenic infections (eg, SARS-CoV-2), obesity promotes the development of many cancer subtypes and increases mortality rates in most cases. We and others have demonstrated that, in the context of B-cell acute lymphoblastic leukemia (B-ALL), adipocytes promote multidrug chemoresistance. Furthermore, others have demonstrated that B-ALL cells exposed to the adipocyte secretome alter their metabolic states to circumvent chemotherapy-mediated cytotoxicity. To better understand how adipocytes impact the function of human B-ALL cells, we used a multi-omic RNA-sequencing (single-cell and bulk transcriptomic) and mass spectroscopy (metabolomic and proteomic) approaches to define adipocyte-induced changes in normal and malignant B cells. These analyses revealed that the adipocyte secretome directly modulates programs in human B-ALL cells associated with metabolism, protection from oxidative stress, increased survival, B-cell development, and drivers of chemoresistance. Single-cell RNA sequencing analysis of mice on low- and high-fat diets revealed that obesity suppresses an immunologically active B-cell subpopulation and that the loss of this transcriptomic signature in patients with B-ALL is associated with poor survival outcomes. Analyses of sera and plasma samples from healthy donors and those with B-ALL revealed that obesity is associated with higher circulating levels of immunoglobulin-associated proteins, which support observations in obese mice of altered immunological homeostasis. In all, our multi-omics approach increases our understanding of pathways that may promote chemoresistance in human B-ALL and highlight a novel B-cell-specific signature in patients associated with survival outcomes.


Asunto(s)
COVID-19 , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Animales , Ratones , Proteómica , SARS-CoV-2 , Obesidad/complicaciones , Obesidad/metabolismo
8.
Cell Death Differ ; 30(6): 1472-1487, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36966227

RESUMEN

The functionally differentiated mammary gland adapts to extreme levels of stress from increased demand for energy by activating specific protective mechanisms to support neonatal health. Here, we identify the breast tumor suppressor gene, single-minded 2 s (SIM2s) as a novel regulator of mitophagy, a key component of this stress response. Using tissue-specific mouse models, we found that loss of Sim2 reduced lactation performance, whereas gain (overexpression) of Sim2s enhanced and extended lactation performance and survival of mammary epithelial cells (MECs). Using an in vitro model of MEC differentiation, we observed SIM2s is required for Parkin-mediated mitophagy, which we have previously shown as necessary for functional differentiation. Mechanistically, SIM2s localizes to mitochondria to directly mediate Parkin mitochondrial loading. Together, our data suggest that SIM2s regulates the rapid recycling of mitochondria via mitophagy, enhancing the function and survival of differentiated MECs.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Mitofagia , Ratones , Femenino , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Células Epiteliales , Modelos Animales de Enfermedad , Ubiquitina-Proteína Ligasas/genética
9.
Breast Cancer Res ; 24(1): 42, 2022 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-35725493

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama , Animales , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/prevención & control , Femenino , Humanos , Ratones , Obesidad/complicaciones , Obesidad/metabolismo , Ovariectomía , Posmenopausia , Ratas , Roedores , Carga Tumoral , Aumento de Peso
10.
Biotech Histochem ; 97(1): 1-10, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34979848

RESUMEN

There are racial disparities in the outcome of triple negative breast cancer (TNBC) patients between women of African ancestry and women of European ancestry, even after accounting for lifestyle, socioeconomic and clinical factors. MicroRNA (miRNA) are non-coding molecules whose level of expression is associated with cancer suppression, proliferation and drug resistance; therefore, these have potential for biomarker applications in cancers including TNBC. Historically, miRNAs up-regulated in African American (AA) patients have received less attention than for patients of European ancestry. Using laser capture microdissection (LCM) to acquire ultrapure tumor cell samples, miRNA expression was evaluated in 15 AA and 15 European American (EA) TNBC patients. Tumor sections were evaluated using RNA extraction followed by miRNA analysis and profiling. Results were compared based on ethnicity and method of tissue fixation. miRNAs that showed high differential expression in AA TNBC patients compared to EA included: miR-19a, miR-192, miR-302a, miR-302b, miR-302c, miR-335, miR-520b, miR-520f and miR-645. LCM is a useful technique for isolation of tumor cells. We found a greater abundance of RNA in frozen samples compared to formalin fixed, paraffin embedded samples. miRNA appears to be a useful biomarker for TNBC to improve diagnosis and treatment.


Asunto(s)
MicroARNs , Neoplasias de la Mama Triple Negativas , Negro o Afroamericano/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Neoplasias de la Mama Triple Negativas/genética , Población Blanca/genética
12.
Endocrinology ; 162(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34410380

RESUMEN

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.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Antineoplásicos Hormonales/uso terapéutico , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Obesidad , Aumento de Peso/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Antineoplásicos Hormonales/farmacología , Inhibidores de la Aromatasa/administración & dosificación , Inhibidores de la Aromatasa/farmacología , Femenino , Humanos , Neoplasias Mamarias Experimentales/complicaciones , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Obesidad/patología , Tamoxifeno/administración & dosificación , Tamoxifeno/farmacología , Delgadez/complicaciones , Delgadez/tratamiento farmacológico , Delgadez/metabolismo , Delgadez/patología
13.
Annu Rev Nutr ; 41: 253-282, 2021 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-34357792

RESUMEN

Diet and nutrition are intricately related to cancer prevention, growth, and treatment response. Preclinical rodent models are a cornerstone to biomedical research and remain instrumental in our understanding of the relationship between cancer and diet and in the development of effective therapeutics. However, the success rate of translating promising findings from the bench to the bedside is suboptimal. Well-designed rodent models will be crucial to improving the impact basic science has on clinical treatment options. This review discusses essential experimental factors to consider when designing a preclinical cancer model with an emphasis on incorporatingthese models into studies interrogating diet, nutrition, and metabolism. The aims of this review are to (a) provide insight into relevant considerations when designing cancer models for obesity, nutrition, and metabolism research; (b) identify common pitfalls when selecting a rodent model; and (c) discuss strengths and limitations of available preclinical models.


Asunto(s)
Neoplasias , Roedores , Animales , Dieta , Humanos , Estado Nutricional , Obesidad/prevención & control
14.
Endocrinology ; 162(9)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34161568

RESUMEN

The selective estrogen receptor (ER) modulator tamoxifen is frequently used in preclinical studies to induce Cre recombinase and generate conditional transgenic mice. In addition, it is often prescribed to treat ER-positive breast cancer, which is diagnosed in approximately 150 000 people each year. In mice, protocols to activate Cre-ER transgenes require tamoxifen administration by several methods, including oral gavage, IP injection, or intragastric injection, spanning a wide range of doses to achieve transgene induction. As a result, the reported metabolic effects of tamoxifen treatment are not always consistent with anecdotal reports from breast cancer patients, or with expected outcomes based on the overall metabolically protective role of estrogen. A greater awareness of tamoxifen's adverse metabolic effects is critical to designing studies with appropriate controls, especially those investigations focused on metabolic outcomes.


Asunto(s)
Neoplasias de la Mama , Tamoxifeno , Animales , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Ratones , Ratones Transgénicos , Moduladores Selectivos de los Receptores de Estrógeno , Transgenes
15.
Diabetes Metab J ; 45(3): 285-311, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33775061

RESUMEN

The relative insufficiency of insulin secretion and/or insulin action causes diabetes. However, obesity and type 2 diabetes mellitus can be associated with an absolute increase in circulating insulin, a state known as hyperinsulinemia. Studies are beginning to elucidate the cause-effect relationships between hyperinsulinemia and numerous consequences of metabolic dysfunctions. Here, we review recent evidence demonstrating that hyperinsulinemia may play a role in inflammation, aging and development of cancers. In this review, we will focus on the consequences and mechanisms of excess insulin production and action, placing recent findings that have challenged dogma in the context of the existing body of literature. Where relevant, we elaborate on the role of specific signal transduction components in the actions of insulin and consequences of chronic hyperinsulinemia. By discussing the involvement of hyperinsulinemia in various metabolic and other chronic diseases, we may identify more effective therapeutics or lifestyle interventions for preventing or treating obesity, diabetes and cancer. We also seek to identify pertinent questions that are ripe for future investigation.


Asunto(s)
Diabetes Mellitus Tipo 2 , Hiperinsulinismo , Resistencia a la Insulina , Neoplasias , Diabetes Mellitus Tipo 2/complicaciones , Humanos , Hiperinsulinismo/complicaciones , Inflamación/complicaciones , Neoplasias/complicaciones , Neoplasias/epidemiología , Obesidad/complicaciones
16.
Am J Pathol ; 191(8): 1342-1352, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33639102

RESUMEN

The role of the adipocyte in the tumor microenvironment has received significant attention as a critical mediator of the obesity-cancer relationship. Current estimates indicate that 650 million adults have obesity, and thirteen cancers, including breast cancer, are estimated to be associated with obesity. Even in people with a normal body mass index, adipocytes are key players in breast cancer progression because of the proximity of tumors to mammary adipose tissue. Outside the breast microenvironment, adipocytes influence metabolic and immune function and produce numerous signaling molecules, all of which affect breast cancer development and progression. The current epidemiologic data linking obesity, and importantly adipose tissue, to breast cancer risk and prognosis, focusing on metabolic health, weight gain, and adipose distribution as underlying drivers of obesity-associated breast cancer is presented here. Bioactive factors produced by adipocytes, both normal and cancer associated, such as cytokines, growth factors, and metabolites, and the potential mechanisms through which adipocytes influence different breast cancer subtypes are highlighted.


Asunto(s)
Adipocitos/patología , Neoplasias de la Mama/patología , Microambiente Tumoral/fisiología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo/patología , Animales , Neoplasias de la Mama/metabolismo , Femenino , Humanos , Obesidad/complicaciones , Obesidad/metabolismo
17.
Clin Cancer Res ; 27(2): 585-597, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33097494

RESUMEN

PURPOSE: Thyroid disease is a frequent comorbidity in women with breast cancer, and many require thyroid hormone replacement therapy (THRT). We postulated that THRT has a deleterious clinical effect mechanistically through hormonal interactions, nuclear receptor cross-talk, and upregulation of high-risk breast cancer genes. EXPERIMENTAL DESIGN: Observational studies of patients with lymph node-negative (LN-) breast cancer (n = 820 and n = 160) were performed to test interactions between THRT and clinical, histologic, outcome, and treatment variables. Differences between the two cohorts include but are not limited to patient numbers, decades of treatment, duration of follow-up/treatment, tumor sizes, incidence, and type and dose/regimen of antihormonal and/or chemotherapeutic agents. In vivo and vitro models, in silico databases, and molecular methods were used to study interactions and define mechanisms underlying THRT effects. RESULTS: THRT significantly and independently reduced disease-free and breast cancer-specific overall survival of only the steroid receptor (SR)-positive (as compared with SR-negative) node-negative patients in both long-term observational studies. Patients with SR+ LN- breast cancer who received THRT and tamoxifen experienced the shortest survival of all treatment groups. A less potent interaction between THRT and aromatase inhibitors was noted in the second patient cohort. Using in vivo and in vitro models, TH administration enhanced estrogen and TH-associated gene expression and proliferation, nuclear colocalization of estrogen receptor and thyroid hormone receptor, and activation of genes used clinically to predict tumor aggression in SR+ breast cancer, including the IGF-IR, WNT, and TGFß pathways. CONCLUSIONS: We show clinically significant adverse interactions between THRT, estrogenic, and oncogenic signaling in patients with SR+ LN- breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Terapia de Reemplazo de Hormonas/métodos , Receptores de Estrógenos/metabolismo , Tamoxifeno/uso terapéutico , Hormonas Tiroideas/uso terapéutico , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Estudios de Cohortes , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Células MCF-7 , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Regulación hacia Arriba/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
18.
J Mammary Gland Biol Neoplasia ; 25(4): 237-253, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33146844

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/etiología , Mama/patología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Obesidad/complicaciones , Adiposidad/fisiología , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Carcinogénesis/inducido químicamente , Carcinogénesis/patología , Carcinógenos/administración & dosificación , Carcinógenos/toxicidad , Línea Celular Tumoral , Dieta Alta en Grasa/efectos adversos , Azúcares de la Dieta/administración & dosificación , Azúcares de la Dieta/efectos adversos , Femenino , Humanos , Glándulas Mamarias Animales/efectos de los fármacos , Neoplasias Mamarias Experimentales/etiología , Neoplasias Mamarias Experimentales/fisiopatología , Menopausia/fisiología , Ratones , Ratones Transgénicos , Obesidad/patología , Obesidad/fisiopatología , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Am J Physiol Regul Integr Comp Physiol ; 319(2): R171-R183, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32551825

RESUMEN

Exercise is often used as a strategy for weight loss maintenance. In preclinical models, we have shown that exercise may be beneficial because it counters the biological drive to regain weight. However, our studies have demonstrated sex differences in the response to exercise in this context. In the present study, we sought to better understand why females and males exhibit different compensatory food eating behaviors in response to regular exercise. Using a forced treadmill exercise paradigm, we measured weight gain, energy expenditure, food intake in real time, and the anorectic effects of leptin. The 4-wk exercise training resulted in reduced weight gain in males and sustained weight gain in females. In male rats, exercise decreased intake, whereas it increased food intake in females. Our results suggest that the anorectic effects of leptin were not responsible for these sex differences in appetite in response to exercise. If these results translate to the human condition, they may reveal important information for the use and application of regular exercise programs.


Asunto(s)
Apetito/fisiología , Peso Corporal/fisiología , Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Condicionamiento Físico Animal/fisiología , Animales , Ingestión de Energía/fisiología , Femenino , Masculino , Ratas
20.
J Biol Chem ; 294(52): 19950-19966, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31740580

RESUMEN

Porcupine O-acyltransferase (PORCN) is considered essential for Wnt secretion and signaling. However, we observed that PORCN inhibition does not phenocopy the effects of WNT4 knockdown in WNT4-dependent breast cancer cells. This suggests a unique relationship between PORCN and WNT4 signaling. To examine the role of PORCN in WNT4 signaling, here we overexpressed WNT4 or WNT3A in breast cancer, ovarian cancer, and fibrosarcoma cell lines. Conditioned media from these lines and co-culture systems were used to assess the dependence of Wnt secretion and activity on the critical Wnt secretion proteins PORCN and Wnt ligand secretion (WLS) mediator. We observed that WLS is universally required for Wnt secretion and paracrine signaling. In contrast, the dependence of WNT3A secretion and activity on PORCN varied across the cell lines, and WNT4 secretion was PORCN-independent in all models. Surprisingly, WNT4 did not exhibit paracrine activity in any tested context. Absent the expected paracrine activity of secreted WNT4, we identified cell-autonomous Wnt signaling activation by WNT4 and WNT3A, independent of PORCN or Wnt secretion. The PORCN-independent, cell-autonomous Wnt signaling demonstrated here may be critical in WNT4-driven cellular contexts or in those that are considered to have dysfunctional Wnt signaling.


Asunto(s)
Aciltransferasas/metabolismo , Proteínas de la Membrana/metabolismo , Vía de Señalización Wnt , Proteína Wnt3A/metabolismo , Proteína Wnt4/metabolismo , Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/genética , Línea Celular Tumoral , Proliferación Celular , Técnicas de Cocultivo , Medios de Cultivo Condicionados/química , Fulvestrant/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Comunicación Paracrina , Transporte de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt3A/antagonistas & inhibidores , Proteína Wnt3A/genética , Proteína Wnt4/antagonistas & inhibidores , Proteína Wnt4/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...