Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Breast Cancer Res ; 24(1): 42, 2022 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-35725493

RESUMO

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.


Assuntos
Neoplasias da Mama , Animais , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/prevenção & controle , Feminino , Humanos , Camundongos , Obesidade/complicações , Obesidade/metabolismo , Ovariectomia , Pós-Menopausa , Ratos , Roedores , Carga Tumoral , Aumento de Peso
2.
Am J Physiol Regul Integr Comp Physiol ; 317(5): R684-R695, 2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31553623

RESUMO

Exercise is a potent facilitator of long-term weight loss maintenance (WLM), whereby it decreases appetite and increases energy expenditure beyond the cost of the exercise bout. We have previously shown that exercise may amplify energy expenditure through energetically expensive nutrient deposition. Therefore, we investigated the effect of exercise on hepatic de novo lipogenesis (DNL) during WLM and relapse to obesity. Obese rats were calorically restricted with (EX) or without (SED) treadmill exercise (1 h/day, 6 days/wk, 15 m/min) to induce and maintain weight loss. After 6 wk of WLM, subsets of WLM-SED and WLM-EX rats were allowed ad libitum access to food for 1 day to promote relapse (REL). An energy gap-matched group of sedentary, relapsing rats (REL-GM) were provided a diet matched to the positive energy imbalance of the REL-EX rats. During relapse, exercise increased enrichment of hepatic DN-derived lipids and induced hepatic molecular adaptations favoring DNL compared with the gap-matched controls. In the liver, compared with both REL-SED and REL-GM rats, REL-EX rats had lower hepatic expression of genes required for cholesterol biosynthesis; greater hepatic expression of genes that mediate very low-density lipoprotein synthesis and secretion; and greater mRNA expression of Cyp27a1, which encodes an enzyme involved in the biosynthesis of bile acids. Altogether, these data provide compelling evidence that the liver has an active role in exercise-mediated potentiation of energy expenditure during early relapse.


Assuntos
Colesterol/biossíntese , Metabolismo Energético , Lipogênese , Fígado/metabolismo , Obesidade/terapia , Condicionamento Físico Animal , Aumento de Peso , Redução de Peso , Animais , Ácidos e Sais Biliares/biossíntese , Restrição Calórica , Modelos Animais de Doenças , Metabolismo Energético/genética , Regulação Enzimológica da Expressão Gênica , Insulina/sangue , Lipogênese/genética , Masculino , Obesidade/genética , Obesidade/metabolismo , Obesidade/fisiopatologia , Recidiva , Corrida , Fatores de Tempo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
3.
Dig Dis Sci ; 64(3): 855-862, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30311152

RESUMO

BACKGROUND: Infliximab is a chimeric anti-tumor necrosis factor alpha (TNF-α) monoclonal antibody that ameliorates inflammation when it binds to and neutralizes TNF-α. It is often used in patients with Crohn's disease and ulcerative colitis to reduce the severity of disease symptoms and induce disease remission. Infusions are generally administered in the hospital setting due to concerns over patient safety, and limited data exist regarding the incidence and management of infusion reactions (IRs) in an alternate care setting without direct physician oversight. AIMS: The aim of this study was to evaluate the incidence of IRs following administration of infliximab and associated management approaches in an alternate care setting. METHODS: A retrospective chart review of 796 patients with Crohn's disease or ulcerative colitis that received a combined 5581 infusions with one home infusion provider between January 2014 and November 2016 was conducted. Timing, severity, management approach, and outcomes of IRs were abstracted and analyzed. RESULTS: A total of 109 infusion reactions (2.0% of all infusions) were recorded in 62 patients (7.8% of all patients). The majority of these reactions were acute and mild or moderate in severity and resolved with rate adjustments and/or medication. Emergency room visits were required in 0.1% of all infusions, and 0.3% of all infusions were not completed due to a reaction. CONCLUSIONS: IRs to infliximab were uncommon and mostly mild or moderate in severity. Resolution of the IR and continuation of therapy was achieved in most patients through a management approach that included prompt recognition and initial treatment via rate adjustments and medications according to physician's orders.


Assuntos
Anti-Inflamatórios/efeitos adversos , Produtos Biológicos/efeitos adversos , Colite Ulcerativa/tratamento farmacológico , Doença de Crohn/tratamento farmacológico , Serviços de Assistência Domiciliar , Infliximab/efeitos adversos , Reação no Local da Injeção/terapia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Anti-Inflamatórios/administração & dosagem , Produtos Biológicos/administração & dosagem , Criança , Colite Ulcerativa/diagnóstico , Colite Ulcerativa/imunologia , Doença de Crohn/diagnóstico , Doença de Crohn/imunologia , Feminino , Humanos , Incidência , Infliximab/administração & dosagem , Infusões Intravenosas , Reação no Local da Injeção/diagnóstico , Reação no Local da Injeção/epidemiologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento , Fator de Necrose Tumoral alfa/imunologia , Adulto Jovem
4.
Horm Cancer ; 8(5-6): 269-285, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28741260

RESUMO

The androgen receptor (AR) has context-dependent roles in breast cancer growth and progression. Overall, high tumor AR levels predict a favorable patient outcome, but several studies have established a tumor promotional role for AR, particularly in supporting the growth of estrogen receptor positive (ER-positive) breast cancers after endocrine therapy. Our previous studies have demonstrated that obesity promotes mammary tumor progression after ovariectomy (OVX) in a rat model of postmenopausal breast cancer. Here, we investigated a potential role for AR in obesity-associated post-OVX mammary tumor progression following ovarian estrogen loss. In this model, we found that obese but not lean rats had nuclear localized AR in tumors that progressed 3 weeks after OVX, compared to those that regressed. AR nuclear localization is consistent with activation of AR-dependent transcription. Longer-term studies (8 weeks post-OVX) showed that AR nuclear localization and expression were maintained in tumors that had progressed, but AR expression was nearly lost in tumors that were regressing. The anti-androgen enzalutamide effectively blocked tumor progression in obese rats by promoting tumor necrosis and also prevented the formation of new tumors after OVX. Neither circulating nor mammary adipose tissue levels of the AR ligand testosterone were elevated in obese compared to lean rats; however, IL-6, which we previously reported to be higher in plasma from obese versus lean rats, sensitized breast cancer cells to low levels of testosterone. Our study demonstrates that, in the context of obesity, AR plays a role in driving ER-positive mammary tumor progression in an environment of low estrogen availability, and that circulating factors unique to the obese host, including IL-6, may influence how cancer cells respond to steroid hormones.


Assuntos
Neoplasias da Mama/etiologia , Neoplasias da Mama/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Ovário/metabolismo , Receptores Androgênicos/metabolismo , Tecido Adiposo/metabolismo , Animais , Antineoplásicos/farmacologia , Benzamidas , Biomarcadores , Neoplasias da Mama/sangue , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Cromatografia Líquida , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Imuno-Histoquímica , Interleucina-6/metabolismo , Interleucina-6/farmacologia , Neoplasias Mamárias Experimentais , Espectrometria de Massas , Nitrilas , Obesidade/sangue , Ovariectomia , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Pós-Menopausa , Ratos , Esteroides/sangue , Esteroides/metabolismo , Testosterona/metabolismo , Testosterona/farmacologia
5.
Cancer Prev Res (Phila) ; 10(3): 198-207, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28154203

RESUMO

Several epidemiologic studies have associated metformin treatment with a reduction in breast cancer incidence in prediabetic and type II diabetic populations. Uncertainty exists regarding which patient populations and/or tumor subtypes will benefit from metformin treatment, and most preclinical in vivo studies have given little attention to the cellular pharmacology of intratumoral metformin uptake. Epidemiologic reports consistently link western-style high fat diets (HFD), which drive overweight and obesity, with increased risk of breast cancer. We used a rat model of HFD-induced overweight and mammary carcinogenesis to define intratumoral factors that confer metformin sensitivity. Mammary tumors were initiated with 1-methyl-1-nitrosourea, and rats were randomized into metformin-treated (2 mg/mL drinking water) or control groups (water only) for 8 weeks. Two-thirds of existing mammary tumors responded to metformin treatment with decreased tumor volumes (P < 0.05), reduced proliferative index (P < 0.01), and activated AMPK (P < 0.05). Highly responsive tumors accumulated 3-fold greater metformin amounts (P < 0.05) that were positively correlated with organic cation transporter-2 (OCT2) protein expression (r = 0.57; P = 0.038). Importantly, intratumoral metformin concentration negatively associated with tumor volume (P = 0.03), and each 10 pmol increase in intratumoral metformin predicted >0.11 cm3 reduction in tumor volume. Metformin treatment also decreased proinflammatory arachidonic acid >1.5-fold in responsive tumors (P = 0.023). Collectively, these preclinical data provide evidence for a direct effect of metformin in vivo and suggest that OCT2 expression may predict metformin uptake and tumor response. Cancer Prev Res; 10(3); 198-207. ©2017 AACR.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Mamárias Experimentais/patologia , Metformina/farmacologia , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Feminino , Hipoglicemiantes/farmacologia , Transportador 2 de Cátion Orgânico , Ratos , Ratos Wistar
6.
Cancer Prev Res (Phila) ; 7(1): 54-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24196830

RESUMO

In the present study, the ability of metformin to inhibit skin tumor promotion by 12-O-tetradecanoylphorbol-13-acetate (TPA) was analyzed in mice maintained on either an overweight control diet or an obesity-inducing diet. Rapamycin was included for comparison, and a combination of metformin and rapamycin was also evaluated. Metformin (given in the drinking water) and rapamycin (given topically) inhibited development of both papillomas and squamous cell carcinomas in overweight and obese mice in a dose-dependent manner. A low-dose combination of these two compounds displayed an additive inhibitory effect on tumor development. Metformin treatment also reduced the size of papillomas. Interestingly, all treatments seemed to be at least as effective for inhibiting tumor formation in obese mice, and both metformin and rapamycin were more effective at reducing tumor size in obese mice compared with overweight control mice. The effect of metformin on skin tumor development was associated with a significant reduction in TPA-induced epidermal hyperproliferation. Furthermore, treatment with metformin led to activation of epidermal AMP-activated protein kinase (AMPK) and attenuated signaling through mTOR complex (mTORC)-1 and p70S6K. Combinations of metformin and rapamycin were more effective at blocking epidermal mTORC1 signaling induced by TPA consistent with the greater inhibitory effect on skin tumor promotion. Collectively, the current data demonstrate that metformin given in the drinking water effectively inhibited skin tumor promotion in both overweight and obese mice and that the mechanism involves activation of epidermal AMPK and attenuated signaling downstream of mTORC1.


Assuntos
Metformina/farmacologia , Neoplasias Cutâneas/prevenção & controle , Adenilato Quinase/metabolismo , Adiponectina/metabolismo , Animais , Peso Corporal , Carcinogênese , Carcinoma de Células Escamosas/prevenção & controle , Dieta , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Insulina/metabolismo , Fator de Crescimento Insulin-Like I/metabolismo , Leptina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Obesos , Complexos Multiproteicos/metabolismo , Neoplasias Experimentais/induzido quimicamente , Neoplasias Experimentais/prevenção & controle , Obesidade/complicações , Sobrepeso/complicações , Papiloma/prevenção & controle , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Neoplasias Cutâneas/induzido quimicamente , Serina-Treonina Quinases TOR/metabolismo , Acetato de Tetradecanoilforbol
7.
Ann N Y Acad Sci ; 1229: 7-17, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21793833

RESUMO

Obesity affects more than one third of the U.S. population and is associated with increased risk and/or disease severity for several chronic diseases, including cancer. In contrast, calorie restriction (CR) consistently inhibits cancer across species and cancer types. Differential effects on globally active circulatory proteins, particularly insulin-like growth factor-1 (IGF-1), provide a plausible mechanistic explanation for the energy balance-cancer link. Diet-induced changes in circulating IGF-1 modulate IGF-1R/EGFR activation and downstream signaling to Akt and mTOR. These dietary energy balance effects on signaling ultimately modulate the levels and/or activity of cell cycle regulatory proteins, regulating proliferation, and modulating susceptibility to tumor development. Selective targeting of mTORC1 potently inhibits tumorigenesis in several model systems producing CR mimetic effects. Targeting this and other pathways modulated by dietary energy balance may lead to the development of strategies for cancer chemoprevention and for reversing the effects of obesity on cancer development and progression.


Assuntos
Restrição Calórica , Transformação Celular Neoplásica , Células Epiteliais/metabolismo , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Animais , Metabolismo Energético/fisiologia , Células Epiteliais/patologia , Humanos , Serina-Treonina Quinases TOR/metabolismo
8.
Cancer Prev Res (Phila) ; 4(7): 1011-20, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21733825

RESUMO

Aberrant activation of phosphoinositide-3-kinase (PI3K)/Akt signaling has been implicated in the development and progression of multiple human cancers. During the process of skin tumor promotion induced by treatment with the phorbol ester 12-O-tetradecanoylphorbol-13-acetate (TPA), activation of epidermal Akt occurs as well as several downstream effectors of Akt, including the activation of mTORC1. Rapamycin, an established mTORC1 inhibitor, was used to further explore the role of mTORC1 signaling in epithelial carcinogenesis, specifically during the tumor promotion stage. Rapamycin blocked TPA-induced activation of mTORC1 as well as several downstream targets. In addition, TPA-induced epidermal hyperproliferation and hyperplasia were inhibited in a dose-dependent manner with topical rapamycin treatments. Immunohistochemical analyses of the skin from mice in this multiple treatment experiment revealed that rapamycin also significantly decreased the number of infiltrating macrophages, T cells, neutrophils, and mast cells seen in the dermis following TPA treatment. Using a two-stage skin carcinogenesis protocol with 7,12-dimethylbenz(a)anthracene (DMBA) as initiator and TPA as the promoter, rapamycin (5-200 nmol per mouse given topically 30 minutes prior to TPA) exerted a powerful antipromoting effect, reducing both tumor incidence and tumor multiplicity. Moreover, topical application of rapamycin to existing papillomas induced regression and/or inhibited further growth. Overall, the data indicate that rapamycin is a potent inhibitor of skin tumor promotion and suggest that signaling through mTORC1 contributes significantly to the process of skin tumor promotion. The data also suggest that blocking this pathway either alone or in combination with other agents targeting additional pathways may be an effective strategy for prevention of epithelial carcinogenesis.


Assuntos
Antibióticos Antineoplásicos/uso terapêutico , Anticarcinógenos/uso terapêutico , Carcinógenos/toxicidade , Papiloma/prevenção & controle , Neoplasias Cutâneas/prevenção & controle , Acetato de Tetradecanoilforbol/toxicidade , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Western Blotting , Humanos , Hiperplasia/induzido quimicamente , Hiperplasia/tratamento farmacológico , Camundongos , Estadiamento de Neoplasias , Papiloma/induzido quimicamente , Papiloma/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Sirolimo/uso terapêutico , Neoplasias Cutâneas/induzido quimicamente , Neoplasias Cutâneas/patologia , Serina-Treonina Quinases TOR/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...