Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 128
Filtrar
1.
Theranostics ; 12(2): 929-943, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34976221

RESUMO

Background: Bone is a frequent site of metastases from breast cancer, but existing therapeutic options are not satisfactory. Although osteoblasts have active roles in cancer progression by assisting the vicious bone-destructive cycle, we employed a counterintuitive approach of activating pro-tumorigenic Wnt signaling and examined the paradoxical possibility of developing osteoblast-derived tumor-suppressive, bone-protective secretomes. Methods: Wnt signaling was activated by the overexpression of Lrp5 and ß-catenin in osteoblasts as well as a pharmacological agent (BML284), and the therapeutic effects of their conditioned medium (CM) were evaluated using in vitro cell cultures, ex vivo breast cancer tissues, and a mouse model of osteolysis. To explore the unconventional regulatory mechanism of the action of Wnt-activated osteoblasts, whole-genome proteomics analysis was conducted, followed by immunoprecipitation and gain- and loss-of-function assays. Results: While osteoblasts did not present any innate tumor-suppressing ability, we observed that the overexpression of Lrp5 and ß-catenin in Wnt signaling made their CM tumor-suppressive and bone-protective. The growth of breast cancer cells and tissues was inhibited by Lrp5-overexpressing CM (Lrp5 CM), which suppressed mammary tumors and tumor-driven bone destruction in a mouse model. Lrp5 CM also inhibited the differentiation and maturation of bone-resorbing osteoclasts by downregulating NFATc1 and cathepsin K. The overexpression of Lrp5 upregulated osteopontin that enriched Hsp90ab1 (Hsp90 beta) and moesin (MSN) in Lrp5 CM. Hsp90ab1 and MSN are atypical tumor-suppressing proteins since they are multi-tasking, moonlighting proteins that promote tumorigenesis in tumor cells. Importantly, Hsp90ab1 immuno-precipitated latent TGFß and inactivated TGFß, whereas MSN interacted with CD44, a cancer stem-cell marker, as well as fibronectin 1, an ECM protein. Furthermore, Hsp90ab1 and MSN downregulated KDM3A that demethylated histones, together with PDL1 that inhibited immune responses. Conclusion: In contrast to inducing tumor-enhancing secretomes and chemoresistance in general by inhibiting varying oncogenic pathways in chemotherapy, this study presented the unexpected outcome of generation tumor-suppressive secretomes by activating the pro-tumorigenic Wnt pathway. The results shed light on the contrasting role of oncogenic signaling in tumor cells and osteoblast-derived secretomes, suggesting a counterintuitive option for the treatment of breast cancer-associated bone metastasis.


Assuntos
Neoplasias da Mama/complicações , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas dos Microfilamentos/metabolismo , Osteoblastos/metabolismo , Osteólise/prevenção & controle , Proteínas Supressoras de Tumor/metabolismo , Via de Sinalização Wnt , Animais , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Fibronectinas/antagonistas & inibidores , Fibronectinas/metabolismo , Humanos , Receptores de Hialuronatos/antagonistas & inibidores , Receptores de Hialuronatos/metabolismo , Neoplasias Mamárias Experimentais/complicações , Neoplasias Mamárias Experimentais/terapia , Camundongos , Osteoclastos/metabolismo , Osteogênese , Osteólise/metabolismo , Proteoma/metabolismo , Secretoma , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo
2.
Endocrine ; 75(3): 949-958, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34797509

RESUMO

PURPOSE: Circulating estrogens in breast cancer patients and survivors are often extremely low due to menopause and estrogen-reducing cancer treatments. Simultaneously, circulating inflammatory markers, and inflammatory proteins in brains of rodent tumor models, can be elevated and correlate with debilitating neurological and psychological comorbidities. Because estrogen has anti-inflammatory properties in the brain, we hypothesized that mammary tumor-induced neuroinflammation is driven, in part, by reduced brain estrogen signaling. METHODS: An ovariectomized mouse model of postmenopausal breast cancer utilizing the ERα-positive 67NR mammary tumor cell line was used for these experiments. A novel, orally bioavailable, and brain penetrant ERß agonist was administered daily via oral gavage. Following treatment, estrogen-responsive genes were measured in brain regions. Central and circulating inflammatory markers were measured via RT-qPCR and a multiplex cytokine array, respectively. RESULTS: We present novel findings that peripheral mammary tumors alter estrogen signaling genes including receptors and aromatase in the hypothalamus, hippocampus, and frontal cortex. Mammary tumors induced peripheral and central inflammation, however, pharmacological ERß activation was not sufficient to reduce this inflammation. CONCLUSIONS: Data presented here suggest that compensating for low circulating estrogen with ERß brain activation is not sufficient to attenuate mammary tumor-induced neuroinflammation, and is therefore not a likely candidate for the treatment of behavioral symptoms in patients. The novel finding that mammary tumors alter estrogen signaling-related genes is a clinically relevant advancement to the understanding of how peripheral tumor biology modulates neurobiology. This is necessary to predict and prevent behavioral comorbidities (e.g., cognitive impairment) prevalent in cancer patients and survivors.


Assuntos
Neoplasias da Mama , Receptor beta de Estrogênio , Neoplasias Mamárias Experimentais , Doenças Neuroinflamatórias , Animais , Neoplasias da Mama/complicações , Modelos Animais de Doenças , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/agonistas , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Estrogênios/metabolismo , Feminino , Neoplasias Mamárias Experimentais/complicações , Camundongos , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/metabolismo
3.
Endocrinology ; 162(11)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34410380

RESUMO

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.


Assuntos
Tecido Adiposo/efeitos dos fármacos , Antineoplásicos Hormonais/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Obesidade , Aumento de Peso/efeitos dos fármacos , Tecido Adiposo/metabolismo , Animais , Antineoplásicos Hormonais/farmacologia , Inibidores da Aromatase/administração & dosagem , Inibidores da Aromatase/farmacologia , Feminino , Humanos , Neoplasias Mamárias Experimentais/complicações , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , Obesidade/complicações , Obesidade/tratamento farmacológico , Obesidade/metabolismo , Obesidade/patologia , Tamoxifeno/administração & dosagem , Tamoxifeno/farmacologia , Magreza/complicações , Magreza/tratamento farmacológico , Magreza/metabolismo , Magreza/patologia
4.
Pharmacol Res ; 152: 104576, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31790822

RESUMO

Breast carcinoma causes severe pain, which decreases the quality of life of patients. Current treatments produce adverse effects and have limited efficacy. Transient potential receptor ankyrin 1 (TRPA1) is related to the onset of cancer and neuropathic pain. The aim of this study was to evaluate the involvement of TRPA1 in a model of breast carcinoma. We injected 4T1 cells in the fourth caudal mammary fat pad of female BALB/c mice, and after 20 days we observed mechanical and cold allodynia and spontaneous nociception behavior (mouse grimace scale detection, MGS). TRPA1 selective antagonist (HC-030031 or A-967079) administration or intrathecal administration of TRPA1 antisense (AS) oligonucleotide was performed. The activity of NADPH oxidase, superoxide dismutase (SOD) and hydrogen peroxide (H2O2) levels were evaluated. The chemical hyperalgesia produced by a TRPA1 agonist (allyl isothiocyanate, AITC) was also detected. The administration of TRPA1 antagonists, TRPA1 AS, or antioxidant, transiently attenuated MGS, or mechanical and cold allodynia. Intraplantar injection of AITC also caused nociception. NADPH oxidase or SOD activity and H2O2 levels were increased in the sciatic nerve and hind paw skin samples. The 4T1 cells did not express TRPA1, and the use of HC-030031 or α-lipoic acid did not reduce the cytotoxic effect of a chemotherapeutic drug (paclitaxel). Thus, TRPA1 could be investigated as a target for breast carcinoma pain treatment.


Assuntos
Dor do Câncer , Neoplasias Mamárias Experimentais , Canal de Cátion TRPA1 , Acetanilidas/farmacologia , Acetanilidas/uso terapêutico , Analgésicos/uso terapêutico , Animais , Antineoplásicos Fitogênicos/farmacologia , Dor do Câncer/tratamento farmacológico , Dor do Câncer/etiologia , Dor do Câncer/genética , Dor do Câncer/metabolismo , Linhagem Celular Tumoral , Feminino , Peróxido de Hidrogênio/metabolismo , Hiperalgesia/tratamento farmacológico , Neoplasias Mamárias Experimentais/complicações , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos Endogâmicos BALB C , NADPH Oxidases/metabolismo , Nociceptividade/efeitos dos fármacos , Oximas/uso terapêutico , Paclitaxel/farmacologia , Purinas/farmacologia , Purinas/uso terapêutico , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Pele/metabolismo , Superóxido Dismutase/metabolismo , Canal de Cátion TRPA1/antagonistas & inibidores , Canal de Cátion TRPA1/genética , Ácido Tióctico/uso terapêutico
5.
Front Immunol ; 10: 2088, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31552036

RESUMO

Cancer patients are at increased risk of developing thrombosis, comorbidity that has been associated with increased neutrophil counts and the formation of neutrophil extracellular traps (NETs). Interleukin-1ß (IL-1ß) modulates the expression of granulocyte colony-stimulating factor (G-CSF), a cytokine that promotes cancer-associated neutrophilia and NET generation. Herein, we combined a murine breast cancer model with a flow-restriction thrombosis model to evaluate whether the IL-1ß blockade could interfere with cancer-associated thrombosis. Mice bearing metastatic 4T1 tumors exhibited high neutrophil counts as well as elevated expression of G-CSF and IL-1ß in their tumors. On the other hand, mice bearing non-metastatic 67NR tumors showed no elevation in neutrophil counts and displayed low expression levels of G-CSF and IL-1ß in their tumors. 4T1 tumor-bearing mice but not 67NR tumor-bearing mice exhibited a NET-dependent prothrombotic state. Pharmacological blockade of IL-1 receptor (IL-1R) decreased the primary growth of 4T1 tumors and reduced the systemic levels of myeloperoxidase, cell-free DNA (cfDNA) and G-CSF, without interfering with the neutrophil counts. Most remarkably, the blockade of IL-1R abolished the prothrombotic state observed in 4T1 tumor-bearing mice. Overall, our results demonstrate that IL-1ß might be a feasible target to attenuate cancer-associated thrombosis, particularly in cancer types that rely on increased G-CSF production and involvement of NET formation.


Assuntos
Armadilhas Extracelulares/efeitos dos fármacos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/antagonistas & inibidores , Neoplasias Mamárias Experimentais/complicações , Receptores de Interleucina-1/antagonistas & inibidores , Trombose/prevenção & controle , Animais , Neoplasias da Mama/complicações , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Armadilhas Extracelulares/metabolismo , Feminino , Fator Estimulador de Colônias de Granulócitos/genética , Fator Estimulador de Colônias de Granulócitos/metabolismo , Humanos , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Contagem de Leucócitos , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos Endogâmicos BALB C , Neutrófilos/metabolismo , Peroxidase/metabolismo , Receptores de Interleucina-1/metabolismo , Trombose/complicações , Trombose/metabolismo , Carga Tumoral/efeitos dos fármacos
6.
Nutrients ; 11(9)2019 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-31480307

RESUMO

Breast cancer and cardiovascular diseases (CVD) have shared risk factors and mechanisms of pathogenicity, as proven by increased cardiac risk in breast cancer patients receiving anticancerogenic therapies and in cancer survivors. A growing mammary tumor may cause heart injury in cancer patients who have not yet been treated. This study aimed to evaluate the effect of conjugated linoleic acid (CLA) supplementation of female rats with 7,12-dimethylbenz(a)anthracene (DMBA)-induced cancerogenesis on fatty acids (FAs), conjugated FAs (CFAs), malondialdehyde (MDA), cholesterol and oxysterols content in cardiac tissue. FAs, cholesterol and oxysterols contents were determined by gas chromatography coupled with mass spectrometry, while the contents of CFAs and MDA were determined by high performance liquid chromatography with photodiode detection. Our results indicate that both CLA supplementation and the presence of tumors influence the lipid biomarkers of CVD. A significant interaction of both experimental factors was observed in the content of polyunsaturated FAs (PUFAs), n-6 PUFAs and CFAs. CLA supplementation significantly inhibited PUFA oxidation, as evidenced by the lower content of MDA in rats' hearts, while the cancerous process intensified the oxidation of cholesterol, as confirmed by the elevated levels of 7-ketocholesterol in DMBA-treated rats. These results may significantly expand knowledge about CLA properties in terms of the prevention of co-existing non-communicable diseases.


Assuntos
Suplementos Nutricionais , Cardiopatias/prevenção & controle , Ácidos Linoleicos Conjugados/farmacologia , Peroxidação de Lipídeos/efeitos dos fármacos , Neoplasias Mamárias Experimentais/complicações , 9,10-Dimetil-1,2-benzantraceno , Animais , Feminino , Coração/efeitos dos fármacos , Cardiopatias/etiologia , Isomerismo , Ácidos Linoleicos Conjugados/química , Neoplasias Mamárias Experimentais/induzido quimicamente , Oxirredução/efeitos dos fármacos , Ratos
7.
JCI Insight ; 4(7)2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30944244

RESUMO

Paraneoplastic neurological disorders result from an autoimmune response against neural self-antigens that are ectopically expressed in neoplastic cells. In paraneoplastic disorders associated to autoantibodies against intracellular proteins, such as paraneoplastic cerebellar degeneration (PCD), current data point to a major role of cell-mediated immunity. In an animal model, in which a neo-self-antigen was expressed in both Purkinje neurons and implanted breast tumor cells, immune checkpoint blockade led to complete tumor control at the expense of cerebellum infiltration by T cells and Purkinje neuron loss, thereby mimicking PCD. Here, we identify 2 potential therapeutic targets expressed by cerebellum-infiltrating T cells in this model, namely α4 integrin and IFN-γ. Mice with PCD were treated with anti-α4 integrin antibodies or neutralizing anti-IFN-γ antibodies at the onset of neurological signs. Although blocking α4 integrin had little or no impact on disease development, treatment using the anti-IFN-γ antibody led to almost complete protection from PCD. These findings strongly suggest that the production of IFN-γ by cerebellum-invading T cells plays a major role in Purkinje neuron death. Our successful preclinical use of neutralizing anti-IFN-γ antibody for the treatment of PCD offers a potentially new therapeutic opportunity for cancer patients at the onset of paraneoplastic neurological disorders.


Assuntos
Interferon gama/antagonistas & inibidores , Neoplasias Mamárias Experimentais/complicações , Degeneração Paraneoplásica Cerebelar/tratamento farmacológico , Células de Purkinje/patologia , Linfócitos T/efeitos dos fármacos , Animais , Antígenos de Neoplasias/imunologia , Autoanticorpos/imunologia , Autoantígenos/imunologia , Linhagem Celular Tumoral/transplante , Feminino , Integrina alfa4/antagonistas & inibidores , Integrina alfa4/imunologia , Integrina alfa4/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Neoplasias Mamárias Experimentais/imunologia , Camundongos , Camundongos Knockout , Degeneração Paraneoplásica Cerebelar/imunologia , Degeneração Paraneoplásica Cerebelar/patologia , Células de Purkinje/imunologia , Células de Purkinje/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
8.
Pharmacol Res ; 142: 140-150, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30797069

RESUMO

Sinomenine (SIN) is an anti-inflammatory and antiarthritic alkaloid derived from Sinomenium acutum, and the product Zhengqing Fengtongning produced from SIN has been marketed in China for treating rheumatoid arthritis (RA). Interestingly, we recently found that SIN could significantly ameliorate bone destruction induced by breast cancer cells in mice. Micro-CT examination showed that bone loss of the trabecular bones in tumor-bearing mice was markedly decreased by i.p. treatment of SIN at 150 mg/kg body weight. A mechanistic study demonstrated that SIN could suppress osteoclast formation and bone absorption induced by both MDA-MB-231 cells and MDA-MB-231 cell-conditioned medium (MDA-MB-231 CM) in preosteoclastic RAW264.7 cells. The MDA-MB-231 CM-induced osteoclast-related genes TRAP and OSCAR were obviously downregulated by SIN. In addition, mRNA expression of c-Fos and NFATc1 and nuclear translocation of c-Fos and NFATc1 protein were inhibited by SIN during MDA-MB-231 CM-induced osteoclastogenesis, while NF-κB signaling was not impacted by SIN. More interestingly, SIN was demonstrated to decrease hIL-8 mRNA expression in cultured MDA-MB-231 cells and to inhibit hIL-8 protein expression in MDA-MB-231 cells cocultured with preosteoclastic RAW264.7 cells while simultaneously downregulating CXCR1, the ligand of IL-8 related to bone destruction, during MDA-MB-231 CM-induced osteoclastogenesis. Previously, IL-8/CXCR1 was reported to be associated with the pathogenesis and progression of RA, and SIN was observed to markedly ameliorate bone erosion of RA patients. Our current findings may extend the utilization of SIN to preventing osteoclastogenesis and bone destruction in breast cancer patients and may enable IL-8/CXCR1 to serve as new targets for both anticancer and antiarthritic drug discovery.


Assuntos
Interleucina-8/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , Morfinanos/farmacologia , Fatores de Transcrição NFATC/metabolismo , Osteólise/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Receptores de Interleucina-8A/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Interleucina-8/genética , Neoplasias Mamárias Experimentais/complicações , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Morfinanos/uso terapêutico , Fatores de Transcrição NFATC/genética , Osteoclastos/efeitos dos fármacos , Osteoclastos/fisiologia , Osteogênese/efeitos dos fármacos , Osteólise/tratamento farmacológico , Osteólise/etiologia , Osteólise/genética , Proteínas Proto-Oncogênicas c-fos/genética , Células RAW 264.7 , Receptores de Interleucina-8A/genética , Transdução de Sinais/efeitos dos fármacos
9.
Sci Rep ; 7(1): 6438, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28743887

RESUMO

Cancer patients are at an increased risk of developing thromboembolic complications. Several mechanisms have been proposed to explain cancer-associated thrombosis including the release of tumor-derived extracellular vesicles and the activation of host vascular cells. It was proposed that neutrophil extracellular traps (NETs) contribute to the prothrombotic phenotype in cancer. In this study, we evaluated the possible cooperation between tumor-derived exosomes and NETs in cancer-associated thrombosis. Female BALB/c mice were orthotopically injected with 4T1 breast cancer cells. The tumor-bearing animals exhibited increased levels of plasma DNA and myeloperoxidase in addition to significantly increased numbers of circulating neutrophils. Mice were subjected to either Rose Bengal/laser-induced venous thrombosis or ferric chloride-induced arterial thrombosis models. The tumor-bearing mice exhibited accelerated thrombus formation in both models compared to tumor-free animals. Treatment with recombinant human DNase 1 reversed the prothrombotic phenotype of tumor-bearing mice in both models. Remarkably, 4T1-derived exosomes induced NET formation in neutrophils from mice treated with granulocyte colony-stimulating factor (G-CSF). In addition, tumor-derived exosomes interacted with NETs under static conditions. Accordingly, the intravenous administration of 4T1-derived exosomes into G-CSF-treated mice significantly accelerated venous thrombosis in vivo. Taken together, our observations suggest that tumor-derived exosomes and neutrophils may act cooperatively in the establishment of cancer-associated thrombosis.


Assuntos
Exossomos/patologia , Neoplasias Mamárias Experimentais/patologia , Neutrófilos/patologia , Trombose/etiologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Armadilhas Extracelulares , Feminino , Fator Estimulador de Colônias de Granulócitos/farmacologia , Neoplasias Mamárias Experimentais/complicações , Camundongos Endogâmicos BALB C , Trombose/tratamento farmacológico , Trombose Venosa/tratamento farmacológico , Trombose Venosa/etiologia
10.
J Nucl Med ; 58(5): 815-820, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28126891

RESUMO

Solid tumor perfusion is a proven variable of interest for predicting cancer aggression and response to therapy. Current methods for noninvasively imaging tumor perfusion with PET are limited by restricted accessibility and short half-lives of perfusion radiotracers. This study presents 2-18F-fluoroethanol (2-18F-FEtOH) as a perfusion reporter that can distinguish between tumors of varying perfusion levels and can be applied to screening drugs that modify tumor perfusion. Methods: Uptake of 2-18F-FEtOH in 4T1 and 67NR murine mammary carcinoma tumors grown in mice was measured using ex vivo radiography as well as static and dynamic PET imaging. 2-18F-FEtOH uptake was directly compared with the 14C-iodoantipyrine perfusion reporter, and the perfusion-modifying drugs nicotinamide, pentoxifylline, and hydralazine were used to manipulate tumor perfusion before 2-18F-FEtOH quantification. Results: Uptake of 2-18F-FEtOH in 4T1 and 67NR tumors was consistent with known perfusion differences within and between these tumors. 2-18F-FEtOH uptake corresponded well with 14C-iodoantipyrine and reflected the tumor perfusion-modifying effects of each drug. Conclusion: 2-18F-FEtOH is a novel 18F-based radiotracer for investigating tumor perfusion with PET imaging. Quantification of 2-18F-FEtOH uptake can be used to distinguish between tumors of varying perfusion and to screen the efficacy of blood flow-modifying drugs for use as adjuvants to existing cancer therapies.


Assuntos
Etanol/análogos & derivados , Neoplasias Mamárias Experimentais/diagnóstico por imagem , Neoplasias Mamárias Experimentais/metabolismo , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/metabolismo , Tomografia por Emissão de Pósitrons/métodos , Animais , Linhagem Celular Tumoral , Diagnóstico Diferencial , Etanol/farmacocinética , Feminino , Neoplasias Mamárias Experimentais/complicações , Taxa de Depuração Metabólica , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias , Neovascularização Patológica/etiologia , Compostos Radiofarmacêuticos/farmacocinética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Distribuição Tecidual
11.
Acta Physiol (Oxf) ; 219(4): 803-813, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27228549

RESUMO

AIM: Exercise training has been suggested as a non-pharmacological approach to prevent skeletal muscle wasting and improve muscle function in cancer cachexia. However, little is known about the molecular mechanisms underlying such beneficial effect. In this study, we aimed to, firstly, examine the contribution of TWEAK signalling to cancer-induced skeletal muscle wasting and, secondly, evaluate whether long-term exercise alters TWEAK signalling and prevents muscle wasting. METHODS: Female Sprague-Dawley rats were randomly assigned to control and exercise groups. Fifteen animals from each group were exposed to N-Methyl-N-nitrosourea carcinogen. Animals in exercise groups were submitted to moderate treadmill exercise for 35 weeks. After the experimental period, animals were killed and gastrocnemius muscles were harvested for morphological and biochemical analysis. RESULTS: We verified that exercise training prevented tumour-induced TWEAK/NF-κB signalling in skeletal muscle with a beneficial impact in fibre cross-sectional area and metabolism. Indeed, 35 weeks of exercise training promoted the upregulation of PGC-1α and oxidative phosphorylation complexes. This exercise-induced muscle remodelling in tumour-bearing animals was associated with less malignant mammary lesions. CONCLUSION: Data support the benefits of an active lifestyle for the prevention of muscle wasting secondary to breast cancer, highlighting TWEAK/NF- κB signalling as a potential therapeutic target for the preservation of muscle mass.


Assuntos
Proteínas Reguladoras de Apoptose/biossíntese , Caquexia/metabolismo , Neoplasias Mamárias Experimentais/complicações , Proteínas de Membrana/biossíntese , Condicionamento Físico Animal/métodos , Transdução de Sinais , Fatores de Necrose Tumoral/biossíntese , Animais , Caquexia/etiologia , Citocina TWEAK , Modelos Animais de Doenças , Feminino , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Atrofia Muscular/metabolismo , NF-kappa B/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
12.
Nat Commun ; 7: 13007, 2016 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-27708283

RESUMO

Obesity is associated with an increased risk of developing breast cancer and is also associated with worse clinical prognosis. The mechanistic link between obesity and breast cancer progression remains unclear, and there has been no development of specific treatments to improve the outcome of obese cancer patients. Here we show that obesity-associated NLRC4 inflammasome activation/ interleukin (IL)-1 signalling promotes breast cancer progression. The tumour microenvironment in the context of obesity induces an increase in tumour-infiltrating myeloid cells with an activated NLRC4 inflammasome that in turn activates IL-1ß, which drives disease progression through adipocyte-mediated vascular endothelial growth factor A (VEGFA) expression and angiogenesis. Further studies show that treatment of mice with metformin inhibits obesity-associated tumour progression associated with a marked decrease in angiogenesis. This report provides a causal mechanism by which obesity promotes breast cancer progression and lays out a foundation to block NLRC4 inflammasome activation or IL-1ß signalling transduction that may be useful for the treatment of obese cancer patients.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Inflamassomos/metabolismo , Interleucina-1beta/metabolismo , Obesidade/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Mama/metabolismo , Neoplasias da Mama/complicações , Linhagem Celular Tumoral/metabolismo , Progressão da Doença , Feminino , Humanos , Linfócitos do Interstício Tumoral/citologia , Neoplasias Mamárias Experimentais/complicações , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/metabolismo , Transplante de Neoplasias , Obesidade/complicações , Transdução de Sinais
13.
Radiat Res ; 185(5): 505-15, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27135968

RESUMO

A detailed understanding of the relationship between radiation-induced breast cancer and obesity is needed for appropriate risk management and to prevent the development of a secondary cancer in patients who have been treated with radiation. Our goal was to develop an animal model to study the relationship by combining two existing Sprague-Dawley rat models of radiation-induced mammary carcinogenesis and diet-induced obesity. Female rats were fed a high-fat diet for 4 weeks and categorized as obesity prone or obesity resistant based on their body weight at 7 weeks of age, at which time the rats were irradiated with 4 Gy. Control rats were fed a standard diet and irradiated at the same time and in the same manner. All rats were maintained on their initial diets and assessed for palpable mammary cancers once a week for the next 30 weeks. The obesity-prone rats were heavier than those in the other groups. The obesity-prone rats were also younger than the other animals at the first detection of mammary carcinomas and their carcinoma weights were greater. A tendency toward higher insulin and leptin blood levels were observed in the obesity-prone rats compared to the other two groups. Blood angiotensin II levels were elevated in the obesity-prone and obesity-resistant rats. Genes related to translation and oxidative phosphorylation were upregulated in the carcinomas of obesity-prone rats. Expression profiles from human breast cancers were used to validate this animal model. As angiotensin is potentially an important factor in obesity-related morbidities and breast cancer, a second set of rats was fed in a similar manner, irradiated and then treated with an angiotensin-receptor blocker, losartan and candesartan. Neither blocker altered mammary carcinogenesis; analyses of losartan-treated animals indicated that expression of renin in the renal cortex and of Agtr1a (angiotensin II receptor, type 1) in cancer tissue was significantly upregulated, suggesting the presence of compensating mechanisms for blocking angiotensin-receptor signaling. Thus, obesity-related elevation of insulin and leptin blood levels and an increase in available energy may facilitate sustained protein synthesis in cancer cells, which is required for rapid cancer development.


Assuntos
Carcinogênese/efeitos da radiação , Dieta/efeitos adversos , Neoplasias Mamárias Experimentais/complicações , Neoplasias Induzidas por Radiação/complicações , Obesidade/complicações , Obesidade/etiologia , Trifosfato de Adenosina/biossíntese , Animais , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica/efeitos da radiação , Hormônios/sangue , Humanos , Obesidade/sangue , Ratos , Ratos Sprague-Dawley , Risco
14.
Endocrinology ; 157(4): 1341-7, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26934299

RESUMO

Patients with breast cancer (BCa) frequently have preexisting vitamin D deficiency (low serum 25-hydroxyvitamin D) when their cancer develops. A number of epidemiological studies show an inverse association between BCa risk and vitamin D status in humans, although some studies have failed to find an association. In addition, several studies have reported that BCa patients with vitamin D deficiency have a more aggressive molecular phenotype and worse prognostic indicators. However, it is unknown whether this association is mechanistically causative and, if so, whether it results from systemic or tumor autonomous effects of vitamin D signaling. We found that ablation of vitamin D receptor expression within BCa cells accelerates primary tumor growth and enables the development of metastases, demonstrating a tumor autonomous effect of vitamin D signaling to suppress BCa metastases. We show that vitamin D signaling inhibits the expression of the tumor progression gene Id1, and this pathway is abrogated in vitamin D deficiency in vivo in 2 murine models of BCa. These findings are relevant to humans, because we discovered that the mechanism of VDR regulation of Inhibitor of differentiation 1 (ID1) is conserved in human BCa cells, and there is a negative correlation between serum 25-hydroxyvitamin D levels and the level of ID1 in primary tumors from patients with BCa.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Receptores de Calcitriol/metabolismo , Transdução de Sinais , Deficiência de Vitamina D/metabolismo , Animais , Western Blotting , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Neoplasias Mamárias Experimentais/complicações , Neoplasias Mamárias Experimentais/genética , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Receptores de Calcitriol/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Vitamina D/análogos & derivados , Vitamina D/sangue , Vitamina D/metabolismo , Deficiência de Vitamina D/complicações , Deficiência de Vitamina D/genética
15.
Chem Biol Interact ; 244: 1-8, 2016 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-26646421

RESUMO

Epirubicin is widely used for the treatment of various breast cancers; however, it has serious adverse side effects, such as hepatotoxicity, which require dose-adjustment or therapy substitution. Paeonol, an active component from Moutan Cortex, has a variety of biological activities, including preventing or reducing various toxicities induced by antineoplastics. Protection by paeonol against hepatotoxicity induced by epirubicin and the underlying mechanism of action were investigated in this study. Cytosolic enzymes in the serum and oxidative stress indices in the liver were determined. The protective effects were determined using the MTT assay in vitro or by evaluating the expression of apoptotic factors and crucial proteins in the PI3K/Akt/NF-kB pathway using western blot analysis. It is concluded that paeonol alleviates epirubicin-induced hepatotoxicity in 4T1-tumor bearing mice by inhibiting the PI3K/Akt/NF-kB pathway.


Assuntos
Acetofenonas/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Epirubicina/efeitos adversos , Neoplasias Mamárias Experimentais/tratamento farmacológico , NF-kappa B/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/complicações , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Medicamentos de Ervas Chinesas/química , Feminino , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Neoplasias Mamárias Experimentais/complicações , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Paeonia/química , Substâncias Protetoras/farmacologia , Relação Estrutura-Atividade
16.
São Paulo; s.n; s.n; 2016. 99 p. tab, graf, ilus.
Tese em Inglês | LILACS | ID: biblio-846595

RESUMO

Breast cancer is an important public health problem. As mammary gland development is a dynamic process that initiates in embryonic life, recent evidence show that in-utero life exposure to maternal nutritional factors can alter mammary gland development and program breast cancer risk in adult life. Even tough studies focus on maternal nutrition, recent evidence show that paternal nutritional factors in-utero and during preconception also affects their female offspring mammary gland development and breast cancer susceptibility in adult life. Studies highlight epigenetic modulation of gene expression in the mammary gland as possible breast cancer programming underlying mechanisms. Selenium is a micronutrient with essential role in central aspects of embryogenesis, male fertility and that has been extensively studied as a chemopreventive agent in several breast cancer models. Among selenium possible mechanisms of action, modulation of cell proliferation, apoptosis, DNA damage, gene expression and epigenetic marks are highlighted. Thus, a rat experiment was conducted to evaluate whether paternal selenium deficiency or supplementation during preconception could affect mammary gland development and breast cancer risk, as well as possible molecular mechanisms involved. Four-week old male Sprague-Dawley rats were exposed to experimental diets (AIN93G) containing 0.15 (control), 0.05 (deficient) and 1ppm (supplemented) of selenium as sodium selenate for 9 weeks and mated with control females. At 7-week old, mammary carcinogenesis was induced in their female offspring by oral administration of 7,12 dymethylbenz[a] anthracene and mammary neoplasia development was evaluated. Paternal selenium deficiency during preconception altered mammary gland development as increased terminal end buds (TEBs) number, epithelial elongation and cell proliferation and decreased apoptosis that were associated with increased breast cancer risk (higher incidence and grade tumors). In addition, paternal selenium deficiency during preconception induced molecular alterations in the mammary gland of the female offspring such as global DNA hypomethylation, increased global levels of H3K27me3 and altered expression of genes related to early life and mammary gland development, apoptosis, cell cycle control, and DNA damage repair. Paternal selenium supplementation during preconception on the other hand did not influence breast cancer programing. Our data show that breast cancer risk can be determined in early-life stages trough the male germline molecular modulation and preconception as an important window of opportunity to start breast cancer prevention strategies. Assuring and adequate selenium intake by men could be a possible starting point


O câncer de mama é um importante problema de saúde pública. O desenvolvimento da glândula mamária é um processo dinâmico que se inicia na vida intrauterina e evidências recentes mostram que a exposição do feto a fatores nutricionais maternos altera o desenvolvimento da glândula mamária e a susceptibilidade ao câncer de mama na vida adulta. Mesmo com um maior foco na nutrição materna, evidências recentes apontam que a nutrição paterna no período intrauterino e de preconcepção também afetam o desenvolvimento da glândula mamária e o risco de câncer de mama da sua prole feminina na vida adulta. Estudos apontam a modulação epigenética da expressão de genes na glândula mamária como possíveis mecanismos envolvidos na programação do câncer de mama. O selênio é um micronutriente com papel essencial em aspectos centrais da embriogênese, fertilidade masculina e que tem sido extensivamente estudado como um agente quimiopreventivo em diferentes modelos de câncer de mama. Dentre os possíveis mecanismos de ação do selênio, destacam-se a capacidade de modulação da proliferação celular, apoptose, danos do DNA e da expressão de genes e mecanismos epigenéticos. Dessa forma, foi conduzido um experimento em ratos para avaliar se a deficiência ou suplementação paterna com selênio durante o período de preconcepção poderia afetar na prole feminina o desenvolvimento da glândula mamária e o risco ao câncer de mama na vida adulta, assim como possíveis mecanismos moleculares envolvidos. Ratos machos da linhagem Sprague-Dawley com 4 semanas de vida foram submetidos à dieta experimental AIN93G contendo 0,15 (controle); 0,05 (deficiente) e 1ppm (suplementada) com selênio na forma de selenato de sódio por 9 semanas e acasalados com fêmeas controle. Com 7 semanas de vida, a carcinogênese mamária foi iniciada na prole feminina através da administração oral do carcinógeno químico 7,12 dimetilbenz[a] antraceno e o desenvolvimento das neoplasias mamárias foi avaliado. A deficiência paterna de selênio causou alterações no desenvolvimento da glândula mamária da prole feminina como aumento no número de terminal end buds (TEBs), aumento da elongação do epitélio mamário, aumento da proliferação celular e diminuição da apoptose que foram associados ao aumento do risco do câncer de mama (maior incidência e agressividade das lesões). Além disso, a deficiência paterna de selênio causou alterações de nível molecular na glândula mamária da prole feminina como hipometilação global, aumento dos níveis globais de H3K27me3 e alteração na expressão de genes relacionados ao desenvolvimento no início da vida e da glândula mamária, apoptose, controle de ciclo celular e reparo de danos no DNA. A suplementação paterna com selênio não foi influenciou o desenvolvimento da glândula mamária e o risco ao câncer de mama na vida adulta. Nossos resultados mostram que o risco do câncer de mama pode ser determinado no início da vida através de influências paternas por meio da modulação de mecanismos moleculares e que o período de preconcepção se caracteriza como uma importante janela de susceptibilidade para iniciar estratégias de diminuição do risco do câncer de mama. Assegurar uma ingestão adequada de selênio por homens pode ser um possível ponto de partida


Assuntos
Masculino , Feminino , Ratos , Selênio/efeitos adversos , Neoplasias da Mama/prevenção & controle , Suscetibilidade a Doenças , Carcinogênese/induzido quimicamente , Neoplasias Mamárias Experimentais/complicações
19.
Oncol Rep ; 34(1): 477-87, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25998578

RESUMO

In previous studies, we demonstrated that the green tea Camellia sinensis (CS) water extract had potent antitumor and antimetastatic effects on 4T1 breast cancer. The metronomic regimen (0.0125 mg/kg twice a week for 4 weeks) of zoledronate (ZOL) was found to be effective in decreasing tumor burden and metastasis as compared with conventional regimen. The aim of the present study was to investigate the antitumor, antimetastatic and anti-osteolytic effects of the combined use of CS water extract and metronomic ZOL against 4T1 breast carcinoma in vitro and in vivo. The results demonstrated that the combination of CS+ZOL exerted a more potent effect on lung and liver by decreasing tumor burden and metastasis, when compared to CS or metronomic ZOL as monotherapies. The combination of CS+ZOL demonstrated optimal bone protection against breast cancer-induced osteolysis for the three groups of CS, ZOL and CS+ZOL. The in vitro results further demonstrated that ZOL enhanced CS-induced apoptosis in 4T1 cells as assessed by the Annexin V-FITC/PI staining and caspase-3 activity assays. In addition, the combined use of CS+ZOL significantly inhibited 4T1 cell migration. Mechanistic studies showed that the enzyme levels of matrix metalloproteinases (MMP)-2 and MMP-9 were suppressed significantly by CS+ZOL. In conclusion, to the best of our knowledge, this is the first study to investigate the novel combined application of herbal extract CS and chemotherapy ZOL in 4T1 breast cancer. The combination of CS plus metronomic ZOL demonstrated significant antitumor, antimetastatic and anti-osteolytic effects against breast cancer, and suggested potential clinical application for breast cancer patients.


Assuntos
Camellia sinensis/química , Difosfonatos/administração & dosagem , Imidazóis/administração & dosagem , Neoplasias Mamárias Experimentais/tratamento farmacológico , Osteólise/tratamento farmacológico , Extratos Vegetais/administração & dosagem , Administração Metronômica , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Difosfonatos/farmacologia , Quimioterapia Combinada , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Imidazóis/farmacologia , Neoplasias Mamárias Experimentais/complicações , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Metástase Neoplásica , Osteólise/metabolismo , Extratos Vegetais/farmacologia , Ácido Zoledrônico
20.
PLoS One ; 10(3): e0121921, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25822717

RESUMO

Anemia is a common complication of cancer; a role of spleen in tumor-stress erythropoiesis has been suggested. However, the molecular mechanisms involved in the splenic erythropoiesis following tumor maintenance remain poorly understood. Here we show that tumor development blocks medullar erythropoiesis by granulocyte colony-stimulating factor (G-CSF) and then causes anemia in murine 4T1 breast tumor-bearing mice. Meanwhile, tumor-stress promotes splenic erythropoiesis. Splenectomy worsened tumor-induced anemia, and reduced tumor volume and tumor weight, indicating the essential role of spleen in tumor-stress erythropoiesis and tumor growth. Tumor progression of these mice led to increased amounts of bone morphogenetic protein 4 (BMP4) in spleen. The in vivo role of macrophages in splenic erythropoiesis under tumor-stress conditions was investigated. Macrophage depletion by injecting liposomal clodronate decreased the expression of BMP4, inhibited splenic erythropoiesis, aggravated the tumor-induced anemia and suppressed tumor growth. Our results provide insight that macrophages and BMP4 are positive regulators of splenic erythropoiesis in tumor pathological situations. These findings reveal that during the tumor-stress period, the microenvironment of the spleen is undergoing changes, which contributes to adopt a stress erythropoietic fate and supports the expansion and differentiation of stress erythroid progenitors, thereby replenishing red blood cells and promoting tumor growth.


Assuntos
Eritropoese , Macrófagos/patologia , Neoplasias Mamárias Experimentais/patologia , Baço/patologia , Anemia/sangue , Anemia/etiologia , Anemia/patologia , Animais , Proteína Morfogenética Óssea 4/biossíntese , Proteína Morfogenética Óssea 4/genética , Linhagem Celular Tumoral , Ácido Clodrônico/farmacologia , Progressão da Doença , Eritropoetina/biossíntese , Feminino , Fator Estimulador de Colônias de Granulócitos/sangue , Macrófagos/efeitos dos fármacos , Neoplasias Mamárias Experimentais/sangue , Neoplasias Mamárias Experimentais/complicações , Camundongos , Camundongos Endogâmicos BALB C , Esplenectomia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA