Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Mol Endocrinol ; 62(3): R223-R237, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30620710

RESUMO

Breast cancer is a heterogeneous disease, and the complexity of breast carcinogenesis is associated with epigenetic modification. There are several major classes of epigenetic enzymes that regulate chromatin activity. This review will focus on the nine mammalian protein arginine methyltransferases (PRMTs) and the dysregulation of PRMT expression and function in breast cancer. This class of enzymes catalyse the mono- and (symmetric and asymmetric) di-methylation of arginine residues on histone and non-histone target proteins. PRMT signalling (and R methylation) drives cellular proliferation, cell invasion and metastasis, targeting (i) nuclear hormone receptor signalling, (ii) tumour suppressors, (iii) TGF-ß and EMT signalling and (iv) alternative splicing and DNA/chromatin stability, influencing the clinical and survival outcomes in breast cancer. Emerging reports suggest that PRMTs are also implicated in the development of drug/endocrine resistance providing another prospective avenue for the treatment of hormone resistance and associated metastasis. The complexity of PRMT signalling is further underscored by the degree of alternative splicing and the scope of variant isoforms (with distinct properties) within each PRMT family member. The evolution of PRMT inhibitors, and the ongoing clinical trials of PRMT inhibitors against a subgroup of solid cancers, coupled to the track record of lysine methyltransferases inhibitors in phase I/II clinical trials against cancer underscores the potential therapeutic utility of targeting PRMT epigenetic enzymes to improve survival outcomes in aggressive and metastatic breast cancer.


Assuntos
Arginina/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Epigênese Genética/genética , Animais , Feminino , Humanos , Metilação
2.
Endocrinology ; 158(3): 431-447, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-27983866

RESUMO

Breast cancer is a heterogeneous disease and its complexity has hindered the development of efficacious treatments targeting all breast cancer subtypes. Many studies have linked the diversity of breast carcinogenesis and metastasis to aberrant epigenetic signaling and control. Here, we focus on the current state of the discipline and review the major epigenetic enzymes controlling chromatin structure and function in the context of breast cancer, including (1) DNA methyltransferases, (2) lysine methyltransferases and demethylases, (3) protein arginine methyltransferases, and (4) histone acetyltransferases and deacetylases. Moreover, therapeutic drugs targeting these epigenetic enzymes are rapidly emerging and/or undergoing clinical trials. Therefore, we discuss the pharmacological manipulation of epigenetic enzymes for breast cancer treatment and present new clinical and survival outcome analysis on epigenetic factors that have evaded analysis to date. Understanding and pharmacologically exploiting epigenetic regulation in breast cancer promises to be an essential aspect of next-generation drug development and adjuvant therapies targeting advanced disease and treatment-resistant tumors.


Assuntos
Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Epigênese Genética , Animais , Carcinogênese , Metilases de Modificação do DNA/metabolismo , Feminino , Histona Desacetilases/metabolismo , Histona Desmetilases/metabolismo , Humanos , Lisina Acetiltransferases/metabolismo
3.
EBioMedicine ; 6: 59-72, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27211549

RESUMO

We have previously reported that RORγ expression was decreased in ER-ve breast cancer, and increased expression improves clinical outcomes. However, the underlying RORγ dependent mechanisms that repress breast carcinogenesis have not been elucidated. Here we report that RORγ negatively regulates the oncogenic TGF-ß/EMT and mammary stem cell (MaSC) pathways, whereas RORγ positively regulates DNA-repair. We demonstrate that RORγ expression is: (i) decreased in basal-like subtype cancers, and (ii) inversely correlated with histological grade and drivers of carcinogenesis in breast cancer cohorts. Furthermore, integration of RNA-seq and ChIP-chip data reveals that RORγ regulates the expression of many genes involved in TGF-ß/EMT-signaling, DNA-repair and MaSC pathways (including the non-coding RNA, LINC00511). In accordance, pharmacological studies demonstrate that an RORγ agonist suppresses breast cancer cell viability, migration, the EMT transition (microsphere outgrowth) and mammosphere-growth. In contrast, RNA-seq demonstrates an RORγ inverse agonist induces TGF-ß/EMT-signaling. These findings suggest pharmacological modulation of RORγ activity may have utility in breast cancer.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Reparo do DNA , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Fator de Crescimento Transformador beta/genética , Benzamidas/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Metástase Neoplásica , Piperazinas/farmacologia , Propanóis/farmacologia , Análise de Sequência de RNA , Transdução de Sinais
4.
Mol Endocrinol ; 30(6): 660-76, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27144290

RESUMO

Skeletal muscle remodels metabolic capacity, contractile and exercise phenotype in response to physiological demands. This adaptive remodeling response to physical activity can ameliorate/prevent diseases associated with poor diet and lifestyle. Our previous work demonstrated that skeletal muscle-specific transgenic expression of the neuron-derived orphan nuclear receptor, Nor-1 drives muscle reprogramming, improves exercise endurance, and oxidative metabolism. The current manuscript investigates the association between exercise, Nor-1 expression and the role of Nor-1 in adaptive remodeling. We demonstrate that Nor-1 expression is induced by exercise and is dependent on calcium/calcineurin signaling (in vitro and in vivo). Analysis of fatigue-resistant transgenic mice that express Nor-1 in skeletal muscle revealed increased hypertrophy and vascularization of muscle tissue. Moreover, we demonstrate that transgenic Nor-1 expression is associated with increased intracellular recycling, ie, autophagy, involving 1) increased expression of light chain 3A or LC3A-II, autophagy protein 5, and autophagy protein 12 in quadriceps femoris muscle extracts from Tg-Nor-1 (relative to Wild-type (WT) littermates); 2) decreased p62 expression indicative of increased autophagolysosome assembly; and 3) decreased mammalian target of rapamycin complex 1 activity. Transfection of LC3A-GFP-RFP chimeric plasmid demonstrated that autophagolysosome formation was significantly increased by Nor-1 expression. Furthermore, we demonstrated a single bout of exercise induced LC3A-II expression in skeletal muscle from C57BL/6 WT mice. This study, when combined with our previous studies, demonstrates that Nor-1 expression drives multiple physiological changes/pathways that are critical to the beneficial responses of muscle to exercise and provides insights into potential pharmacological manipulation of muscle reprogramming for the treatment of lifestyle induced chronic diseases.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Condicionamento Físico Animal , Receptores de Esteroides/metabolismo , Receptores dos Hormônios Tireóideos/metabolismo , Animais , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagia/efeitos dos fármacos , Calcineurina/metabolismo , Cálcio/metabolismo , Linhagem Celular , Proteínas de Ligação a DNA/genética , Hipertrofia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos/metabolismo , Modelos Biológicos , Músculo Esquelético/irrigação sanguínea , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/parasitologia , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Esteroides/genética , Receptores dos Hormônios Tireóideos/genética , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia
5.
J Biol Chem ; 283(26): 18411-21, 2008 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-18441015

RESUMO

Homozygous staggerer mice (sg/sg) display decreased and dysfunctional retinoic acid receptor-related orphan receptor alpha (RORalpha) expression. We observed decreases in serum (and liver) triglycerides and total and high density lipoprotein serum cholesterol in sg/sg mice. Moreover, the sg/sg mice were characterized by reduced adiposity (associated with decreased fat pad mass and adipocyte size). Candidate-based expression profiling demonstrated that the dyslipidemia in sg/sg mice is associated with decreased hepatic expression of SREBP-1c, and the reverse cholesterol transporters, ABCA1 and ABCG1. This is consistent with the reduced serum lipids. The molecular mechanism did not involve aberrant expression of LXR and/or ChREBP. However, ChIP and transfection analyses revealed that RORalpha is recruited to and regulates the activity of the SREBP-1c promoter. Furthermore, the lean phenotype in sg/sg mice is also characterized by significantly increased expression of PGC-1alpha, PGC-1beta, and lipin1 mRNA in liver and white and brown adipose tissue from sg/sg mice. In addition, we observed a significant 4-fold increase in beta(2)-adrenergic receptor mRNA in brown adipose tissue. Finally, dysfunctional RORalpha expression protects against diet-induced obesity. Following a 10-week high fat diet, wild-type but not sg/sg mice exhibited a approximately 20% weight gain, increased hepatic triglycerides, and notable white and brown adipose tissue accumulation. In summary, these changes in gene expression (that modulate lipid homeostasis) in metabolic tissues are involved in decreased adiposity and resistance to diet-induced obesity in the sg/sg mice, despite hyperphagia. In conclusion, we suggest this orphan nuclear receptor is a key modulator of fat accumulation and that selective ROR modulators may have utility in the treatment of obesity.


Assuntos
Tecido Adiposo/metabolismo , Regulação da Expressão Gênica , Lipídeos/química , Obesidade/genética , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/fisiologia , Transativadores/genética , Transativadores/fisiologia , Ração Animal , Animais , Células COS , Chlorocebus aethiops , Heterozigoto , Metabolismo dos Lipídeos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Membro 1 do Grupo F da Subfamília 1 de Receptores Nucleares , Triglicerídeos/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA