Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Oncotarget ; 7(36): 57955-57969, 2016 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-27462784

RESUMO

Loss of progesterone-receptors (PR) expression is associated with breast cancer progression. Herein we provide evidence that OHPg/PR-B through Beclin-1 evoke autophagy-senescence transition, in breast cancer cells. Specifically, OHPg increases Beclin-1 expression through a transcriptional mechanism due to the occupancy of Beclin-1 promoter by PR-B, together with the transcriptional coactivator SRC-2. This complex binds at a canonical half progesterone responsive element, which is fundamental for OHPg effects, as shown by site-directed mutagenesis. Beside, OHPg via non-genomic action rapidly activates JNK, which phosphorylates Bcl-2, producing the functional release from Beclin-1 interaction. This is not linked to an efficient autophagic flux, since p62 levels, marker of degradation via lysosomes, were not reduced after sustained OHPg stimulus. Instead, the cell cycle inhibitor p27 was induced, together with an irreversible G1 arrest, hallmark of cellular senescence. Specifically the increase of senescence-associated ß-galactosidase activity was blocked by Bcl-2 siRNA but also by Beclin-1 siRNA. Collectively these findings support the importance of PR-B expression in breast cancer cells, thus targeting PR-B may be a useful strategy to provide additional approaches to existing therapies for breast cancer patients.


Assuntos
Autofagia , Proteína Beclina-1/metabolismo , Neoplasias da Mama/metabolismo , Senescência Celular , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Progesterona/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genômica , Humanos , Ligantes , Células MCF-7 , Mutagênese Sítio-Dirigida , Plasmídeos/metabolismo , Progesterona/metabolismo , Regiões Promotoras Genéticas , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , beta-Galactosidase/metabolismo
2.
Oncol Rep ; 35(1): 568-76, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26459431

RESUMO

Alterations in cellular metabolism are among the most consistent hallmarks of cancer. Herein, after a comprehensive metabolic phenotype characterization of MCF7 and ZR75 breast cancer cells, we investigated the activity of bergapten (Bg), a plant-derived compound, against breast cancer. The study of different biochemical pathways involved in cell metabolism revealed that the two cell lines have different bioenergetic phenotypes: MCF7 cells express a glycolytic phenotype only partially oxidative, while ZR75 cells mainly have an oxidative phenotype. In both cell lines, Bg blocked glycolysis and significantly decreased glucose-6-phosphate dehydrogenase (G6PDH) activity promoting glucose accumulation; modulated bioenergetic requirements altering the expression of oxidative phosphorylation (OXPHOS) complexes and ATP production; and induced a lipid-lowering effect since an increased lipase activity concomitantly to a reduction in triglyceride levels was observed. Quantitative data of different metabolites and enzymatic activities were presented. Treatment with Bg resulted in an alteration in different metabolic pathways inducing death in the cells. We report a novel action of the natural product Bg on breast cancer, since it induced metabolic reprogramming by disrupting the interconnected network of different metabolic mechanisms. Bg can be used in combination with other forms of targeted chemotherapy to improve cancer treatment outcomes.


Assuntos
Neoplasias da Mama/metabolismo , Metabolismo Energético/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Metoxaleno/análogos & derivados , 5-Metoxipsoraleno , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucosefosfato Desidrogenase/metabolismo , Humanos , Lipase/metabolismo , Células MCF-7 , Metoxaleno/farmacologia
3.
Mol Cancer ; 14: 130, 2015 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-26148846

RESUMO

BACKGROUND: Bergapten (5-methoxypsoralen), a natural psoralen derivative present in many fruits and vegetables, has shown antitumoral effects in a variety of cell types. In this study, it has been addressed how Bergapten in breast cancer cells induces autophagic process. RESULTS: In MCF7 and ZR-75 breast cancer cells Bergapten exhibited anti-survival response by inducing the autophagic process increasing Beclin1, PI3KIII, UVRAG, AMBRA expression and conversion of LC3-I to LC3-II. LC3-GFP, Acridine orange assay and transmission electron microscopy even confirmed the increased autophagosome formations in treated cells. Bergapten-induced autophagy is dependent by PTEN up-regulation, since silencing this gene, the induction of Beclin1 and the p-AKT/p-mTOR signal down-regulation were reversed. PTEN is transcriptionally regulated by Bergapten through the involvement of p38MAPK/NF-Y, as evidenced by the use of p38MAPK inhibitor SB203580, site-direct mutagenesis of NF-Y element and NF-Y siRNA. Furthermore NF-Y knockdown prevented Bergapten-induced acid vesicular organelle accumulations (AVOs), strengthening the role of this element in mediating autophagy. CONCLUSIONS: Our data indicate PTEN as a key target of Bergapten action in breast cancer cells for the induction of autophagy. These findings add further details on the mechanism of action of Bergapten, therefore suggesting that phytochemical compounds may be implemented in the novel strategies for breast cancer treatment.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Metoxaleno/análogos & derivados , PTEN Fosfo-Hidrolase/genética , 5-Metoxipsoraleno , Autofagia/efeitos dos fármacos , Autofagia/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Feminino , Expressão Gênica , Genes Reporter , Humanos , Células MCF-7 , Metoxaleno/farmacologia , Fenótipo , Regiões Promotoras Genéticas , Regulação para Cima
4.
Histochem Cell Biol ; 144(1): 67-76, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25850410

RESUMO

The most common cause of male infertility is the testicular varicocele, a condition that impairs production and decreases quality of sperm. Male fertility also strictly depends on androgens acting through their own receptor. The enzyme 5α-reductase (SRD5A) is involved in the conversion of testosterone to 5α-dihydrotestosterone, both required for the development and maintenance of male reproductive function. Here, we evaluated, by western blotting analysis, the presence of SRD5A in human ejaculated spermatozoa and evidenced differences in sperm SRD5A content between healthy donors and varicocele-affected patients. Additionally, SRD5A sperm ultrastructural localization was also assessed by transmission electron microscopy and immunogold assay. We evidenced that SRD5A enzyme is present in the human spermatozoa and that its cellular content is lowered in sperm samples from varicocele patients compared to healthy subjects. The presence of SRD5A in human ejaculated spermatozoa highlights the potential role of this enzyme in sperm physiopathology suggesting that the decrease in its content, by affecting the conversion of testosterone into 5α-dihydrotestosterone, may be an important additional mechanism involved in the harmful effect of varicocele in male fertility.


Assuntos
3-Oxo-5-alfa-Esteroide 4-Desidrogenase/metabolismo , Infertilidade/etiologia , Proteínas de Membrana/metabolismo , Espermatozoides/enzimologia , Varicocele/enzimologia , Adulto , Humanos , Imuno-Histoquímica , Masculino , Microscopia Eletrônica de Transmissão , Microscopia Imunoeletrônica , Espermatozoides/fisiologia , Espermatozoides/ultraestrutura , Testosterona/metabolismo , Varicocele/fisiopatologia
5.
Appl Immunohistochem Mol Morphol ; 23(5): 374-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24992177

RESUMO

Varicocele, an abnormal tortuosity and dilation of veins of the pampiniform plexus, is the most common identifiable and correctable cause of male infertility. It is now becoming apparent that signaling through vitamin A metabolites, such as all-trans retinoic acid (ATRA), is indispensable for spermatogenesis and disruption of retinoic acid receptor-α (RARα) function may result in male sterility and aberrant spermatogenesis. Herein, we investigated by Western blot and immunogold electron microscopy the expression profiles and subcellular localization of RARα in healthy and varicocele human sperm; in addition, we analyzed the effects of ATRA on cholesterol efflux and sperm survival utilizing enzymatic colorimetric CHOD-PAP method and Eosin Y technique, respectively. In varicocele samples, a strong reduction of RARα expression was observed. Immunogold labeling evidenced cellular location of RARα also confirming its reduced expression in "varicocele" samples. Sperm responsiveness to ATRA treatment was reduced in varicocele sperm. Our study showed that RARα is expressed in human sperm probably with a dual role in promoting both cholesterol efflux and survival. RARα might be involved in the pathogenesis of varicocele as its expression is reduced in pathologic samples. Thus, ATRA administration in procedures for artificial insemination or dietary vitamin A supplementation might represent a promising therapeutic approach for the management of male infertility.


Assuntos
Expressão Gênica , Receptores do Ácido Retinoico/genética , Espermatozoides/metabolismo , Espermatozoides/ultraestrutura , Varicocele/genética , Transporte Biológico , Western Blotting , Células Cultivadas , Colesterol/metabolismo , Amarelo de Eosina-(YS) , Humanos , Imuno-Histoquímica , Masculino , Microscopia Eletrônica , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Cordão Espermático/metabolismo , Cordão Espermático/patologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/patologia , Tretinoína/metabolismo , Tretinoína/farmacologia , Varicocele/diagnóstico , Varicocele/metabolismo , Varicocele/patologia
6.
J Cell Mol Med ; 18(11): 2252-65, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25216078

RESUMO

The tumour suppressor activity of the phosphatase and tensin homologue on chromosome 10 (PTEN) is subject of intense investigative efforts, although limited information on its regulation in breast cancer is available. Herein, we report that, in breast cancer cells, progesterone (OHPg), through its cognate receptor PR-B, positively modulates PTEN expression by inducing its mRNA and protein levels, and increasing PTEN-promoter activity. The OHPg-dependent up-regulation of PTEN gene activity requires binding of the PR-B to an Sp1-rich region within the PTEN gene promoter. Indeed, ChIP and EMSA analyses showed that OHPg treatment induced the occupancy of PTEN promoter by PR and Sp1 together with transcriptional coactivators such as SRC1 and CBP. PR-B isoform knockdown abolished the complex formation indicating its specific involvement. The OHPg/PR-B dependent induction of PTEN causes the down-regulation of PI3K/AKT signal, switching on the autophagy process through an enhanced expression of UVRAG and leading to a reduced cell survival. Altogether these findings highlight a novel functional connection between OHPg/PR-B and tumour suppressor pathways in breast cancer.


Assuntos
Neoplasias da Mama/genética , Proteína Oncogênica v-akt/genética , PTEN Fosfo-Hidrolase/biossíntese , Progesterona/genética , Receptores de Progesterona/biossíntese , Autofagia/genética , Neoplasias da Mama/patologia , Sobrevivência Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , Progesterona/metabolismo , Transdução de Sinais/genética
7.
Stereotact Funct Neurosurg ; 92(4): 211-7, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25096235

RESUMO

BACKGROUND: Restoration of functions in Huntington's disease (HD) by neurotransplantation stems from the formation of a striatum-like structure capable of establishing host connections as a result of grafted striatal neuroblast maturation. For the first time, we demonstrated some developmental steps accomplished by progenitor cells in the brain of an HD patient and analysed the molecular asset of the human primordium. CASE REPORT: Surgery involved bilateral (two sessions) stereotactic, caudate-putaminal transplantation of whole ganglionic eminence fragments from single legally aborted fetuses. MRI showed that the tissue deposits of the left hemisphere grew and joined to constitute a single tissue mass that remodelled basal ganglia anatomy and remained stable in size over time. No evidence of graft growth was observed contralaterally. PET demonstrated increased striatal and stable cortical metabolism. Unified Huntington's Disease Rating Scale assessments demonstrated improvement of motor performances, which faded over the 36-month follow-up. Cognitive performance tended to decrease at a lower rate than before transplantation. CONCLUSION: The striatal primordium grew into the host brain and this process was associated with metabolic change and some clinical benefit. The study suggests the plasticity and reparative potential of un-manipulated primordium in an era where promising cell-based therapies are still in their infancy.


Assuntos
Transplante de Tecido Encefálico , Corpo Estriado/patologia , Transplante de Tecido Fetal , Doença de Huntington/cirurgia , Plasticidade Neuronal , Telencéfalo/transplante , Adulto , Transplante de Tecido Encefálico/métodos , Fármacos do Sistema Nervoso Central/uso terapêutico , Transtornos Cognitivos/etiologia , Terapia Combinada , Corpo Estriado/diagnóstico por imagem , Transplante de Tecido Fetal/métodos , Seguimentos , Perfilação da Expressão Gênica , Sobrevivência de Enxerto , Humanos , Doença de Huntington/tratamento farmacológico , Doença de Huntington/patologia , Doença de Huntington/psicologia , Itália , Imageamento por Ressonância Magnética , Masculino , Neuroimagem , Tomografia por Emissão de Pósitrons , Robótica , Índice de Gravidade de Doença , Técnicas Estereotáxicas , Telencéfalo/embriologia , Telencéfalo/metabolismo
8.
Reproduction ; 147(5): 589-98, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24429393

RESUMO

The study of androgens involved in male reproduction has been object of intense efforts, while their reported action on human male gametes is limited. We previously described the presence of androgen receptor (AR) in sperm with a role related to the modulation of the PI3K pathway. In the present study, we investigated the expression of AR and its ultrastructural location in normal sperm as well as in spermatozoa obtained from varicocele patients. We observed a reduced AR content in varicocele sperm with respect to healthy sperm by western blot analysis and transmission electron microscopy (TEM). The ultrastructural location of AR was detected mainly on the head membrane as well as in the nucleus, neck, and mitochondria. Influence of dihydrotestosterone (DHT) treatment on cholesterol efflux was increased in normal sperm, while it was reduced or absent in varicocele sperm. To better understand DHT/AR significance in human male gametes, we evaluated triglyceride content and lipase, acyl-CoA dehydrogenase, and glucose-6-phosphate dehydrogenase activities upon DHT treatment. The metabolic outcome glimpsed in normal sperm was an increased metabolic rate, while 'varicocele' sperm economized energy. Taken together, our results reveal DHT and AR as new players in sperm endocrinology, indicating that varicocele sperm may have difficulty in switching to the capacitated status. A decreased AR expression and a consequent reduced responsiveness to DHT in sperm may represent molecular mechanisms involved in the pathophysiology of varicocele leading to male infertility. This study revealed new detrimental effects of varicocele on sperm at the molecular level.


Assuntos
Receptores Androgênicos/fisiologia , Espermatozoides/patologia , Varicocele/patologia , Varicocele/fisiopatologia , Células Cultivadas , Di-Hidrotestosterona/farmacologia , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/fisiologia , Masculino , Transdução de Sinais/fisiologia , Capacitação Espermática/efeitos dos fármacos , Capacitação Espermática/fisiologia , Espermatozoides/efeitos dos fármacos , Espermatozoides/ultraestrutura
9.
Asian J Androl ; 15(6): 835-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23728590

RESUMO

Insulin (Ins) has recently been demonstrated to have the ability to induce the capacitation process in pig spermatozoa. In various mammalian species, capacitation has been linked to the nitric oxide (NO) signalling; therefore, this study investigated NO production in Ins-treated pig spermatozoa by fluorescence-activated cell sorting. For the same samples, sperm capacitation was evaluated by chlortetracycline staining, protein tyrosine phosphorylation pattern and acrosomal status. A significant increase of the intrasperm NO level and the activation of three capacitation indices were detected in response to Ins treatment. Conversely, sperm preincubation with an NO synthase inhibitor (N-nitro-L-arginine methyl ester) or with the anti-Ins receptor ß (IRß) antibody reversed all of the Ins-related effects. These results suggest that Ins has the capacity to enhance intracellular NO concentrations in pig spermatozoa and indicate a possible NO implication upon Ins promotion of capacitation.


Assuntos
Insulina/fisiologia , Óxido Nítrico/fisiologia , Espermatozoides/fisiologia , Animais , Masculino , Suínos
10.
Ultrastruct Pathol ; 37(3): 176-82, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23634797

RESUMO

Abstract Recent reports support the possible role of PI3K in sperm capacitation and acrosome reaction, although studies regarding PI3K identity in human sperm, under certain disease states such as varicocele, are still lacking. The authors, therefore, examined the expression profile and ultrastructural localization of PI3K in human semen samples, comparing healthy donors and patients with varicocele. The results obtained performing western blotting assay showed decreased PI3K expression in varicocele with respect to the "healthy" sperm. Immunogold labeling revealed human sperm cellular compartments containing PI3K, evidencing it in the head at both the membrane and nucleus and the entire tail, from the middle to the end piece of normal sperm. In varicocele PI3K label was confined to the head, with a strong reduction of specific reaction in the neck, middle piece, and tail. In conclusion, the data suggest that PI3K may play a role in the maintenance of male factor infertility associated with varicocele, and it may be further exploited as an additional molecular marker for the diagnosis of male infertility disorders.


Assuntos
Infertilidade Masculina/enzimologia , Fosfatidilinositol 3-Quinase/análise , Espermatozoides/enzimologia , Varicocele/enzimologia , Biomarcadores/análise , Western Blotting , Estudos de Casos e Controles , Humanos , Imuno-Histoquímica , Infertilidade Masculina/etiologia , Infertilidade Masculina/patologia , Masculino , Microscopia Eletrônica de Transmissão , Valor Preditivo dos Testes , Análise do Sêmen/métodos , Espermatozoides/ultraestrutura , Varicocele/complicações , Varicocele/patologia
11.
Mol Reprod Dev ; 80(2): 155-65, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23280600

RESUMO

Myricetin is a natural flavonoid, particularly enriched in red wines, whose occurrence is widespread among plants. Despite extensive research, the beneficial effects of Myricetin on human health are still controversial. Here, we tested the estrogen-like effect of the phytoestrogen Myricetin on human ejaculated sperm biology. To this aim, human normozoospermic samples were exposed to increasing concentrations (10 nM, 100 nM, and 1 µM) of Myricetin. Motility, viability, capacitation-associated biochemical changes (i.e., cholesterol efflux and tyrosine phosphorylation), acrosin activity, as well as glucose utilization and fatty-acid oxidation (i.e., glucose and lipid metabolism) were all significantly increased by low doses of Myricetin. Importantly, both estrogen receptors α and ß (ERs) and phosphatidylinositol-3-OH kinase (PI3K)/AKT signaling are activated in the presence of Myricetin since these were both abrogated by specific inhibitors of each pathway. Our results show how Myricetin, through ERs and PI3K/AKT signalings, potentiates sperm function. This effect is dose-dependent at low concentrations of Myricetin (up to 100 nM), whereas higher amounts do not seem to improve any further sperm motility, viability, or other tested features, and, in some cases, they reduced or even abrogated the efficacy exerted by lower doses. Further studies are needed to elucidate if high levels of Myricetin, which could be attained even with moderate wine consumption, could synergize with endogenous estrogens in the female reproductive tract, interfering with the physiological sperm fertilization process.


Assuntos
Flavonoides/farmacologia , Fitoestrógenos/farmacologia , Espermatozoides/fisiologia , Vinho/análise , Acrosina/metabolismo , Análise de Variância , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácidos Graxos/metabolismo , Feminino , Flavonoides/análise , Glucose/metabolismo , Humanos , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Fitoestrógenos/análise , Capacitação Espermática/efeitos dos fármacos , Motilidade dos Espermatozoides/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos
12.
Mol Nutr Food Res ; 56(11): 1655-64, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22976781

RESUMO

SCOPE: Green tea and its major constituent epigallocatechin gallate (EGCG) have been extensively studied as potential treatment for a variety of diseases. We assessed the influence of EGCG on male fertilizing potential by analyzing different features of human sperm involved in capacitation process. METHODS AND RESULTS: Using human normozoospermic samples, we evaluated the effect of EGCG (2 µM, 20 µM, 60 µM) on sperm activities. Our results showed that lower doses of EGCG (from 2 to 20 µM) increased cholesterol efflux and tyrosine phosphorylation through the estrogen receptor (ER), since ICI 182,780, a specific ER antagonist, abrogated 20 µM EGCG effects. Besides, we evidenced that EGCG at similar concentrations, increased sperm motility, viability, and phosphorylation of proteins controlling cell survival such as Bcl2, Akt, and Src, via ER. Furthermore, we observed reduction of triglycerides content, induction of lipase, as well as the G6PDH activity. These results address to an increase in energy expenditure. On the contrary, treatment of 60 µM EGCG produced opposite effects that still appear after ICI cotreatment. CONCLUSION: These results provide a novel mechanism involving ERs through which low doses of EGCG exerted benefits to sperm physiology, also detected data evidence the adverse action of high EGCG concentrations probably related to its prooxidant and antiestrogenic potential.


Assuntos
Catequina/análogos & derivados , Espermatozoides/efeitos dos fármacos , Espermatozoides/metabolismo , Catequina/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Colesterol/metabolismo , Relação Dose-Resposta a Droga , Estradiol/análogos & derivados , Estradiol/farmacologia , Fulvestranto , Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Fosforilação/efeitos dos fármacos , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Estrogênio/antagonistas & inibidores , Receptores de Estrogênio/metabolismo , Motilidade dos Espermatozoides/efeitos dos fármacos , Chá/química , Triglicerídeos/metabolismo , Tirosina/metabolismo , Quinases da Família src/metabolismo
13.
Oncotarget ; 3(8): 798-810, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22878233

RESUMO

Recent studies have suggested that cancer cells behave as metabolic parasites, by inducing oxidative stress in adjacent normal fibroblasts. More specifically, oncogenic mutations in cancer cells lead to ROS production and the "secretion" of hydrogen peroxide species. Oxidative stress in stromal fibroblasts then induces their metabolic conversion into cancer-associated fibroblasts. Such oxidative stress drives the onset of autophagy, mitophagy, and aerobic glycolysis in fibroblasts, resulting in the local production of high-energy mitochondrial fuels (such as L-lactate, ketone bodies, and glutamine). These recycled nutrients are then transferred to cancer cells, where they are efficiently burned via oxidative mitochondrial metabolism (OXPHOS). We have termed this new energy-transfer mechanism "Two-Compartment Tumor Metabolism", to reflect that the production and consumption of nutrients (L-lactate and other catabolites) is highly compartmentalized. Thus, high-energy onco-catabolites are produced by the tumor stroma. Here, we used a genetic approach to stringently test this energy-transfer hypothesis. First, we generated hTERT-immortalized fibroblasts which were genetically re-programmed towards catabolic metabolism. Metabolic re-programming towards glycolytic metabolism was achieved by the recombinant over-expression of MFF (mitochondrial fission factor). MFF over-expression results in extensive mitochondrial fragmentation, driving mitochondrial dysfunction. Our results directly show that MFFfibroblasts undergo oxidative stress, with increased ROS production, and the onset of autophagy and mitophagy, both catabolic processes. Mechanistically, oxidative stress induces autophagy via NF-kB activation, also providing a link with inflammation. As a consequence MFF-fibroblasts showed intracellular ATP depletion and the extracellular secretion of L-lactate, a critical onco-catabolite. MFF-fibroblasts also showed signs of myofibroblast differentiation, with the expression of SMA and calponin. Importantly, MFF-fibroblasts signficantly promoted early tumor growth (up to 6.5-fold), despite a 20% overall reduction in angiogenesis. Thus, catabolic metabolism in cancer-associated fibroblasts may be a critical event during tumor intiation, allowing accelerated tumor growth, especially prior to the onset of neoangiogenesis.


Assuntos
Transformação Celular Neoplásica/metabolismo , Glicólise , Ácido Láctico/metabolismo , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Proteínas Mitocondriais/metabolismo , Células Estromais/metabolismo , Trifosfato de Adenosina/metabolismo , Autofagia , Linhagem Celular Tumoral , Metabolismo Energético , Humanos , Proteínas de Membrana/biossíntese , Proteínas Mitocondriais/biossíntese , Mitofagia , Miofibroblastos/metabolismo , NF-kappa B/biossíntese , NF-kappa B/metabolismo , Neoplasias , Neovascularização Patológica , Fosforilação Oxidativa , Estresse Oxidativo , Espécies Reativas de Oxigênio , Microambiente Tumoral
14.
Cell Cycle ; 11(16): 3019-35, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22874531

RESUMO

We have previously shown that a loss of stromal Cav-1 is a biomarker of poor prognosis in breast cancers. Mechanistically, a loss of Cav-1 induces the metabolic reprogramming of stromal cells, with increased autophagy/mitophagy, mitochondrial dysfunction and aerobic glycolysis. As a consequence, Cav-1-low CAFs generate nutrients (such as L-lactate) and chemical building blocks that fuel mitochondrial metabolism and the anabolic growth of adjacent breast cancer cells. It is also known that a loss of Cav-1 is associated with hyperactive TGF-ß signaling. However, it remains unknown whether hyperactivation of the TGF-ß signaling pathway contributes to the metabolic reprogramming of Cav-1-low CAFs. To address these issues, we overexpressed TGF-ß ligands and the TGF-ß receptor I (TGFß-RI) in stromal fibroblasts and breast cancer cells. Here, we show that the role of TGF-ß in tumorigenesis is compartment-specific, and that TGF-ß promotes tumorigenesis by shifting cancer-associated fibroblasts toward catabolic metabolism. Importantly, the tumor-promoting effects of TGF-ß are independent of the cell type generating TGF-ß. Thus, stromal-derived TGF-ß activates signaling in stromal cells in an autocrine fashion, leading to fibroblast activation, as judged by increased expression of myofibroblast markers, and metabolic reprogramming, with a shift toward catabolic metabolism and oxidative stress. We also show that TGF-ß-activated fibroblasts promote the mitochondrial activity of adjacent cancer cells, and in a xenograft model, enhancing the growth of breast cancer cells, independently of angiogenesis. Conversely, activation of the TGF-ß pathway in cancer cells does not influence tumor growth, but cancer cell-derived-TGF-ß ligands affect stromal cells in a paracrine fashion, leading to fibroblast activation and enhanced tumor growth. In conclusion, ligand-dependent or cell-autonomous activation of the TGF-ß pathway in stromal cells induces their metabolic reprogramming, with increased oxidative stress, autophagy/mitophagy and glycolysis, and downregulation of Cav-1. These metabolic alterations can spread among neighboring fibroblasts and greatly sustain the growth of breast cancer cells. Our data provide novel insights into the role of the TGF-ß pathway in breast tumorigenesis, and establish a clear causative link between the tumor-promoting effects of TGF-ß signaling and the metabolic reprogramming of the tumor microenvironment.


Assuntos
Neoplasias da Mama/metabolismo , Fibroblastos/metabolismo , Ácido Láctico/metabolismo , Comunicação Parácrina , Fator de Crescimento Transformador beta/metabolismo , Animais , Comunicação Autócrina , Autofagia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Reprogramação Celular , Técnicas de Cocultura , Feminino , Fibroblastos/patologia , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Imuno-Histoquímica , Ligantes , Camundongos , Camundongos Nus , Mitocôndrias/genética , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neovascularização Patológica/metabolismo , Fosforilação Oxidativa , Estresse Oxidativo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Fator de Crescimento Transformador beta/genética , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Cell Cycle ; 11(15): 2911-21, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22810004

RESUMO

Testicular germ cell tumors are the most common tumor in male and the least studied. We focused on human seminoma using the TCAM2 cell line. Through ERß, 10 nM estradiol (E2) was able to induce PTEN gene expression and promoter transactivation. Transient transfections, ChIP and EMSA assays evidenced the 5'-flanking region of PTEN gene promoter E2-responsive. The ERß binding to the Sp1 on PTEN promoter decreased cell survival. The presence of ERß or PTEN is necessary to induce the loss of cell survival upon E2, addressing their cooperation in this action. pAKT and AKT expression decreased under E2 and DPN, while known apoptotic markers appeared to be unchanged. The PI3K/AKT pathway inhibition also leads to autophagy: E2 and DPN enhanced the expression of autophagy-related markers such as PI3III, Beclin 1, AMBRA and UVRAG. MDC and TEM assays confirmed E2-induced autophagy. The absence of DNA fragmentation, caspase 9 and PARP1 cleavages suggested that necroptosis and/or parthanatos may occur. FACS analysis, LDH assay and RIP1 expression attested this hypothesis. Our study reveals a unique mechanism through which ERß/PTEN signaling induces cell death in TCAM2 by autophagy and necroptosis. These data, supporting estrogen-dependency of human seminoma, propose ERß ligands for therapeutic use in the treatment of this pathological condition.


Assuntos
Autofagia , Receptor beta de Estrogênio/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Seminoma/genética , Neoplasias Testiculares/genética , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Reguladoras de Apoptose/biossíntese , Proteína Beclina-1 , Caspase 9/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular , Estradiol , Humanos , Masculino , Proteínas de Membrana/biossíntese , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/biossíntese , Inibidores de Fosfoinositídeo-3 Quinase , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/biossíntese , Proteínas Proto-Oncogênicas c-akt/genética , Seminoma/metabolismo , Seminoma/patologia , Transdução de Sinais/genética , Transcrição Gênica , Proteínas Supressoras de Tumor/biossíntese
16.
Cell Cycle ; 11(12): 2285-302, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22684298

RESUMO

Senescent fibroblasts are known to promote tumor growth. However, the exact mechanism remains largely unknown. An important clue comes from recent studies linking autophagy with the onset of senescence. Thus, autophagy and senescence may be part of the same physiological process, known as the autophagy-senescence transition (AST). To test this hypothesis, human fibroblasts immortalized with telomerase (hTERT-BJ1) were stably transfected with autophagy genes (BNIP3, CTSB or ATG16L1). Their overexpression was sufficient to induce a constitutive autophagic phenotype, with features of mitophagy, mitochondrial dysfunction and a shift toward aerobic glycolysis, resulting in L-lactate and ketone body production. Autophagic fibroblasts also showed features of senescence, with increased p21(WAF1/CIP1), a CDK inhibitor, cellular hypertrophy and increased ß-galactosidase activity. Thus, we genetically validated the existence of the autophagy-senescence transition. Importantly, autophagic-senescent fibroblasts promoted tumor growth and metastasis, when co-injected with human breast cancer cells, independently of angiogenesis. Autophagic-senescent fibroblasts stimulated mitochondrial metabolism in adjacent cancer cells, when the two cell types were co-cultured, as visualized by MitoTracker staining. In particular, autophagic ATG16L1 fibroblasts, which produced large amounts of ketone bodies (3-hydroxy-butyrate), had the strongest effects and promoted metastasis by up to 11-fold. Conversely, expression of ATG16L1 in epithelial cancer cells inhibited tumor growth, indicating that the effects of autophagy are compartment-specific. Thus, autophagic-senescent fibroblasts metabolically promote tumor growth and metastasis, by paracrine production of high-energy mitochondrial fuels. Our current studies provide genetic support for the importance of "two-compartment tumor metabolism" in driving tumor growth and metastasis via a simple energy transfer mechanism. Finally, ß-galactosidase, a known lysosomal enzyme and biomarker of senescence, was localized to the tumor stroma in human breast cancer tissues, providing in vivo support for our hypothesis. Bioinformatic analysis of genome-wide transcriptional profiles from tumor stroma, isolated from human breast cancers, also validated the onset of an autophagy-senescence transition. Taken together, these studies establish a new functional link between host aging, autophagy, the tumor microenvironment and cancer metabolism.


Assuntos
Autofagia , Senescência Celular , Fibroblastos/metabolismo , Corpos Cetônicos/metabolismo , Proteínas Relacionadas à Autofagia , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Catepsina B/genética , Catepsina B/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Técnicas de Cocultura , Feminino , Glicólise , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo
17.
Cell Cycle ; 11(12): 2272-84, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22684333

RESUMO

Previous studies have demonstrated that loss of caveolin-1 (Cav-1) in stromal cells drives the activation of the TGF-ß signaling, with increased transcription of TGF-ß target genes, such as connective tissue growth factor (CTGF). In addition, loss of stromal Cav-1 results in the metabolic reprogramming of cancer-associated fibroblasts, with the induction of autophagy and glycolysis. However, it remains unknown if activation of the TGF-ß / CTGF pathway regulates the metabolism of cancer-associated fibroblasts. Therefore, we investigated whether CTGF modulates metabolism in the tumor microenvironment. For this purpose, CTGF was overexpressed in normal human fibroblasts or MDA-MB-231 breast cancer cells. Overexpression of CTGF induces HIF-1α-dependent metabolic alterations, with the induction of autophagy/mitophagy, senescence, and glycolysis. Here, we show that CTGF exerts compartment-specific effects on tumorigenesis, depending on the cell-type. In a xenograft model, CTGF overexpressing fibroblasts promote the growth of co-injected MDA-MB-231 cells, without any increases in angiogenesis. Conversely, CTGF overexpression in MDA-MB-231 cells dramatically inhibits tumor growth in mice. Intriguingly, increased extracellular matrix deposition was seen in tumors with either fibroblast or MDA-MB-231 overexpression of CTGF. Thus, the effects of CTGF expression on tumor formation are independent of its extracellular matrix function, but rather depend on its ability to activate catabolic metabolism. As such, CTGF-mediated induction of autophagy in fibroblasts supports tumor growth via the generation of recycled nutrients, whereas CTGF-mediated autophagy in breast cancer cells suppresses tumor growth, via tumor cell self-digestion. Our studies shed new light on the compartment-specific role of CTGF in mammary tumorigenesis, and provide novel insights into the mechanism(s) generating a lethal tumor microenvironment in patients lacking stromal Cav-1. As loss of Cav-1 is a stromal marker of poor clinical outcome in women with primary breast cancer, dissecting the downstream signaling effects of Cav-1 are important for understanding disease pathogenesis, and identifying novel therapeutic targets.


Assuntos
Autofagia , Senescência Celular , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Fibroblastos/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Fator de Crescimento do Tecido Conjuntivo/genética , Feminino , Glicólise , Humanos , Camundongos , Estresse Oxidativo , Células Estromais/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Transplante Heterólogo , Microambiente Tumoral
18.
Endocrinology ; 153(4): 1743-54, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22355067

RESUMO

The mechanisms through which sperm manage their energy metabolism are poorly understood. The present study provides biochemical and morphological evidence that mitochondrial citrate carrier (CIC) is present in ejaculated human sperm and is restricted to the midpiece. The inhibition of CIC with the specific substrate analog 1,2,3-benzenetricarboxylate resulted in the reduction of cholesterol efflux, protein tyrosine phosphorylation, phospho-AKT, phospho-p60src, hyperactivated motility and acrosome reaction, suggesting a role for this mitochondrial carrier in sperm physiology. Furthermore, inhibition of CIC by 1,2,3-benzenetricarboxylate resulted in a reduction of glucose-stimulated insulin secretion and autocrine insulin secretion by sperm. Remarkably, blocking CIC also reduced glucose-6-phosphate dehydrogenase activity, probably in accordance with its regulation on insulin secretion. Capacitation and glucose metabolism were stimulated by glucose as well as citrate, the specific substrate of CIC, implying a similar action because glucose and citrate both induced insulin secretion by sperm. In the present finding, we discovered a new site of action for CIC in the regulation of metabolism, and it may be assumed that CIC works with other factors in the regulation of sperm energy metabolism to sustain capacitation process and acrosome reaction.


Assuntos
Proteínas de Transporte/fisiologia , Metabolismo Energético/fisiologia , Insulina/metabolismo , Mitocôndrias/fisiologia , Espermatozoides/fisiologia , Reação Acrossômica/fisiologia , Colesterol/metabolismo , Humanos , Secreção de Insulina , Masculino , Proteínas Tirosina Quinases/metabolismo , Capacitação Espermática/fisiologia , Motilidade dos Espermatozoides/fisiologia
19.
Cell Cycle ; 10(23): 4065-73, 2011 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-22129993

RESUMO

Increasing chronological age is the most significant risk factor for cancer. Recently, we proposed a new paradigm for understanding the role of the aging and the tumor microenvironment in cancer onset. In this model, cancer cells induce oxidative stress in adjacent stromal fibroblasts. This, in turn, causes several changes in the phenotype of the fibroblast including mitochondrial dysfunction, hydrogen peroxide production, and aerobic glycolysis, resulting in high levels of L-lactate production. L-lactate is then transferred from these glycolytic fibroblasts to adjacent epithelial cancer cells and used as "fuel" for oxidative mitochondrial metabolism.  Here, we created a new pre-clinical model system to directly test this hypothesis experimentally. To synthetically generate glycolytic fibroblasts, we genetically-induced mitochondrial dysfunction by knocking down TFAM using an sh-RNA approach.  TFAM is mitochondrial transcription factor A, which is important in functionally maintaining the mitochondrial respiratory chain. Interestingly, TFAM-deficient fibroblasts showed evidence of mitochondrial dysfunction and oxidative stress, with the loss of certain mitochondrial respiratory chain components, and the over-production of hydrogen peroxide and L-lactate. Thus, TFAM-deficient fibroblasts underwent metabolic reprogramming towards aerobic glycolysis.  Most importantly, TFAM-deficient fibroblasts significantly promoted tumor growth, as assayed using a human breast cancer (MDA-MB-231) xenograft model. These increases in glycolytic fibroblast driven tumor growth were independent of tumor angiogenesis. Mechanistically, TFAM-deficient fibroblasts increased the mitochondrial activity of adjacent epithelial cancer cells in a co-culture system, as seen using MitoTracker. Finally, TFAM-deficient fibroblasts also showed a loss of caveolin-1 (Cav-1), a known breast cancer stromal biomarker. Loss of stromal fibroblast Cav-1 is associated with early tumor recurrence, metastasis, and treatment failure, resulting in poor clinical outcome in breast cancer patients. Thus, this new experimental model system, employing glycolytic fibroblasts, may be highly clinically relevant. These studies also have implications for understanding the role of hydrogen peroxide production in oxidative damage and "host cell aging," in providing a permissive metabolic microenvironment for promoting and sustaining tumor growth.


Assuntos
Neoplasias da Mama/patologia , Senescência Celular , Ácido Láctico/metabolismo , Mitocôndrias/metabolismo , Estresse Oxidativo , Animais , Neoplasias da Mama/metabolismo , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Técnicas de Cocultura , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Técnicas de Silenciamento de Genes , Glicólise , Humanos , Peróxido de Hidrogênio/metabolismo , Neoplasias Mamárias Experimentais , Camundongos , Camundongos Nus , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
20.
Reprod Biol Endocrinol ; 9: 133, 2011 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-21970701

RESUMO

BACKGROUND: Nitric oxide (NO) is a signaling molecule produced by intracellular nitric oxide synthase (NOS) enzymes. This free radical appears to affect sperm capacitation, a maturation step preceding acrosome reaction. Recent studies have reported leptin ability to promote capacitation and acrosome reaction in pig male gametes. METHODS: This study has investigated nitric oxide production in leptin-treated pig spermatozoa by fluorescence-activated cell sorting, while the intracellular NOS isoforms were assessed by Western blot analysis. In addition, acrosome status of treated-spermatozoa was evaluated by FITC-PNA staining. RESULTS: Significant increases of nitric oxide levels and acrosome reaction extent were detected in leptin-treated spermatozoa, but both the effects were reversed in presence of L-NAME. Furthermore, the immunoblots of sperm extracts have evidenced three bands of ~160 Kd(bNOS), ~130 Kd (iNOS) and ~135 Kd (eNOS). CONCLUSIONS: The identification of the three intracellular NOS isoforms suggests that pig spermatozoa could produce NO, while the augmented nitric oxide levels in leptin-treated male gametes indicates the capacity of the hormone to induce nitric oxide production. Furthermore, the inhibitory effect of L-NAME and of Ab-ObR on the promotion of acrosome reaction triggered by leptin suggests a possible involvement of NO in the hormone action.


Assuntos
Reação Acrossômica , Leptina/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Óxido Nítrico Sintase Tipo I/metabolismo , Óxido Nítrico/metabolismo , Espermatozoides/metabolismo , Reação Acrossômica/efeitos dos fármacos , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Citometria de Fluxo , Fluoresceínas/metabolismo , Masculino , Sondas Moleculares/metabolismo , Peso Molecular , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/química , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II/química , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/química , Aglutinina de Amendoim/metabolismo , Receptores para Leptina/antagonistas & inibidores , Espermatozoides/citologia , Espermatozoides/efeitos dos fármacos , Sus scrofa , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA