Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Med Res Rev ; 41(3): 1499-1538, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33274768

RESUMO

Prostate cancer (PCa), one of the most commonly diagnosed cancers worldwide, still presents important unmet clinical needs concerning treatment. In the last years, the metabolic reprogramming and the specificities of tumor cells emerged as an exciting field for cancer therapy. The unique features of PCa cells metabolism, and the activation of specific metabolic pathways, propelled the use of metabolic inhibitors for treatment. The present work revises the knowledge of PCa metabolism and the metabolic alterations that underlie the development and progression of the disease. A focus is given to the role of bioenergetic sources, namely, glucose, lipids, and glutamine sustaining PCa cell survival and growth. Moreover, it is described as the action of oncogenes/tumor suppressors and sex steroid hormones in the metabolic reprogramming of PCa. Finally, the status of PCa treatment based on the inhibition of metabolic pathways is presented. Globally, this review updates the landscape of PCa metabolism, highlighting the critical metabolic alterations that could have a clinical and therapeutic interest.


Assuntos
Neoplasias da Próstata , Humanos , Masculino , Redes e Vias Metabólicas , Oncogenes , Neoplasias da Próstata/tratamento farmacológico
2.
Prostate ; 75(9): 923-35, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25786656

RESUMO

BACKGROUND: Imatinib mesylate is a chemotherapeutic drug that inhibits the tyrosine kinase activity of c-KIT and has been successfully used to treat leukemias and some solid tumors. However, its application for treatment of hormone-refractory prostate cancer (HRPC) has shown modest effectiveness and did not follow the outcomes in cultured cells or animal models. Moreover, the molecular pathways by which imatinib induces cytotoxicity in prostate cancer cells are poorly characterized. METHODS: Two cell line models of HRPC (DU145 and PC3) were exposed to 20 µM of imatinib for 6-72 hr. MTS assay was used to assess cell viability during the course of experiment. Gene expression analysis of c-KIT, cell-cycle and apoptosis regulators, and angiogenic factors was determined by means of real-time PCR, western blot, and/or immunocytochemistry. The enzymatic activity of the apoptosis effector, caspase-3, was determined by a colorimetric assay. RESULTS: Imatinib significantly decreased the viability of DU145 cells but paradoxically augmented the viability of PC3 cells. DU145 cells displayed diminished expression of anti-apoptotic Bcl-2 protein and augmented levels of caspase-8 and -9, as well as, increased enzymatic activity of caspase-3 in response to imatinib. No differences existed on the expression levels of apoptosis-related proteins in PC3 cells treated with imatinib, though the activity of caspase-3 was decreased. The mRNA levels of angiogenic factor VEGF were decreased in DU145-treated cells, whereas an opposite effect was seen in PC3. In addition, it was shown that DU145 and PC3 cells present a differential expression of c-KIT protein variants. CONCLUSION: DU145 and PC3 cells displayed a contradictory behavior in response to imatinib, which was underpinned by a distinct expression pattern (or activity) of target regulators of cell-cycle, apoptosis, and angiogenesis. The paradoxical effect of imatinib in PC3 cells may be related with the differential expression of c-KIT protein variants. Moreover, the present findings helped to understand the discrepancies in the efficacy of imatinib as therapeutic option in HRPC.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Piperazinas/farmacologia , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Proteínas Proto-Oncogênicas c-kit/biossíntese , Pirimidinas/farmacologia , Western Blotting , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Imuno-Histoquímica , Masculino , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Quinases Ativadas por p21/biossíntese , Quinases Ativadas por p21/genética
3.
Mol Reprod Dev ; 81(12): 1064-79, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25359157

RESUMO

Maintaining the delicate balance between cell survival and death is of the utmost importance for the proper development of germ cells and subsequent fertility. On the other hand, the fine regulation of tissue homeostasis by mechanisms that control cell fate is a factor that can prevent carcinogenesis. c-KIT is a type III receptor tyrosine kinase activated by its ligand, stem cell factor (SCF). c-KIT signaling plays a crucial role in cell fate decisions, specifically controlling cell proliferation, differentiation, survival, and apoptosis. Indeed, deregulating the SCF/c-KIT system by attenuation or overactivation of its signaling strength is linked to male infertility and cancer, and rebalancing its activity via c-KIT inhibitors has proven beneficial in treating human tumors that contain gain-of-function mutations or overexpress c-KIT. This review addresses the roles of SCF and c-KIT in the male reproductive tract, and discusses the potential application of c-KIT target therapies in disorders of the reproductive system.


Assuntos
Fertilidade/fisiologia , Genitália/fisiologia , Modelos Biológicos , Neoplasias/fisiopatologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais/fisiologia , Fator de Células-Tronco/metabolismo , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Genitália/metabolismo , Humanos , Masculino , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética
4.
Pathol Res Pract ; 240: 154181, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36327818

RESUMO

BACKGROUND: Androgens, the known drivers of prostate cancer (PCa), have been indicated as important metabolic regulators with a relevant role in stimulating lipid metabolism. Also, the relationship between obesity and the aggressiveness of PCa has been established. However, it is unknown if the androgenic hormonal environment may alter the response of PCa cells to lipid availability. PURPOSE: The present study evaluated the effect of 5α-dihydrotestosterone (DHT) in regulating lipid metabolism, and the interplay between this hormone and low-density lipoprotein (LDL)-cholesterol in modulating PCa cells fate. METHODS: Non-neoplastic and neoplastic PCa cells were treated with 10 nM DHT, and the expression of fatty acids transporter, fatty acid synthase (FASN), and carnitine palmitoyltransferase 1A (CPT1A) evaluated. PCa cells were also exposed to LDL (100 µg/ml) in the presence or absence of DHT. RESULTS: Treatment with DHT upregulated the expression of FASN and CPT1A in androgen-sensitive PCa cells. In contrast, LDL supplementation suppressed FASN expression regardless of the presence of DHT, whereas augmenting CPT1A levels. Our results also showed that LDL-cholesterol increased PCa cells viability, proliferation, and migration dependently on the presence of DHT. Moreover, LDL and DHT synergistically enhanced the accumulation of lipid droplets in PCa cells. CONCLUSIONS: The obtained results show that androgens deregulate lipid metabolism and enhance the effects of LDL increasing PCa cells viability, proliferation and migration. The present findings support clinical data linking obesity with PCa and first implicate androgens in this relationship. Also, they sustain the application of pharmacological approaches targeting cholesterol availability and androgens signaling simultaneously.


Assuntos
Androgênios , Neoplasias da Próstata , Masculino , Humanos , Androgênios/farmacologia , LDL-Colesterol/uso terapêutico , Neoplasias da Próstata/metabolismo , Di-Hidrotestosterona/farmacologia , Obesidade , Receptores Androgênicos/metabolismo
5.
Cell Oncol (Dordr) ; 44(2): 385-403, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33464483

RESUMO

PURPOSE: Resistance to androgen-deprivation therapies and progression to so-called castrate-resistant prostate cancer (CRPC) remain challenges in prostate cancer (PCa) management and treatment. Among other alterations, CRPC has been associated with metabolic reprogramming driven by androgens. Here, we investigated the role of androgens in regulating glutaminolysis in PCa cells and determined the relevance of this metabolic route in controlling the survival and growth of androgen-sensitive (LNCaP) and CRPC (DU145 and PC3) cells. METHODS: PCa cells (LNCaP, DU145 and PC3) and 3-month old rats were treated with 5α-dihydrotestosterone (DHT). Alternatively, LNCaP cells were exposed to the glutaminase inhibitor BPTES, alone or in combination with the anti-androgen bicalutamide. Biochemical, Western blot and extracellular flux assays were used to evaluate the viability, proliferation, migration and metabolism of PCa cells in response to DHT treatment or glutaminase inhibition. RESULTS: We found that DHT up-regulated the expression of the glutamine transporter ASCT2 and glutaminase, both in vitro in LNCaP cells and in vivo in rat prostate cells. BPTES diminished the viability and migration of PCa cells, while increasing caspase-3 activity. CRPC cells were found to be more dependent on glutamine and more sensitive to glutaminase inhibition. BPTES and bicalutamide co-treatment had an additive effect on suppressing LNCaP cell viability. Finally, we found that inhibition of glutaminolysis differentially affected glycolysis and lipid metabolism in both androgen-sensitive and CRPC cells. CONCLUSION: Our data reveal glutaminolysis as a central metabolic route controlling PCa cell fate and highlight the relevance of targeting glutaminase for CRPC treatment.


Assuntos
Di-Hidrotestosterona/farmacologia , Glutamina/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Sistema ASC de Transporte de Aminoácidos/genética , Sistema ASC de Transporte de Aminoácidos/metabolismo , Androgênios/farmacologia , Anilidas/farmacologia , Animais , Caspase 3/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Glutaminase/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Ácido Láctico/biossíntese , Metabolismo dos Lipídeos/efeitos dos fármacos , Masculino , Antígenos de Histocompatibilidade Menor/genética , Antígenos de Histocompatibilidade Menor/metabolismo , Modelos Biológicos , Proteínas de Neoplasias/metabolismo , Nitrilas/farmacologia , Neoplasias de Próstata Resistentes à Castração/patologia , Ratos , Sulfetos/farmacologia , Tiadiazóis/farmacologia , Compostos de Tosil/farmacologia
6.
Eur J Med Chem ; 171: 343-363, 2019 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-30928707

RESUMO

The last decade has witnessed the peculiarities of metabolic reprogramming in tumour onset and progression, and their relevance in cancer therapy. Also, it has been indicated that the metastatic process may depend on the metabolic rewiring and adaptation of cancer cells to the pressure of tumour microenvironment and limiting nutrient availability. The present review gatherers the existent knowledge on the influence of tumour microenvironment and metabolic routes driving metastasis. A focus will be given to glycolysis, fatty acid metabolism, glutaminolysis, and amino acid handling. In addition, the role of metabolic waste driving metastasization will be explored. Finally, we discuss the status of cancer treatment approaches targeting metabolism. This knowledge revision will highlight the critical metabolic targets in metastasis and the chemicals already used in preclinical studies and clinical trials, providing clues that would be further exploited in medicinal chemistry research.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Antineoplásicos/química , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Neoplasias/metabolismo , Relação Estrutura-Atividade , Microambiente Tumoral/efeitos dos fármacos
7.
Life Sci ; 218: 274-283, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30605651

RESUMO

AIMS: The tyrosine kinase inhibitor imatinib has been used in prostate cancer treatment with outcomes that did not follow the in vitro findings. The glycolytic environment has been shown to influence the efficacy of anti-cancer drugs. This study aimed to evaluate the effect of imatinib on cell viability, apoptosis, and metabolism in cell line models of castrate-resistant prostate cancer (CRPC) under hyperglycemic and hypoglycemic conditions. MAIN METHODS: DU145 and PC3 CRPC cell lines were exposed to 20 µM imatinib under 5 mM (hypoglycemia) or 30 mM glucose (hyperglycemia) for 48-72 h. Cell viability was assessed by the MTS assay. The expression of apoptosis regulators and glycolytic metabolism-related proteins was analysed by Western blot, and the activity of caspase-3 and lactate dehydrogenase (LDH) was determined spectrophotometrically. Glucose consumption and lactate production were determined using biochemical assays. KEY FINDINGS: Imatinib decreased CRPC cells viability, whereas increasing apoptosis; effects only observed in hyperglycemic conditions. Glucose consumption and lactate production were significantly increased in imatinib-treated DU145 and PC3 cells, and independently of glucose availability. Accordingly, LDH expression and activity were significantly increased in response to imatinib. SIGNIFICANCE: Higher glucose availability improved the effectiveness of imatinib suppressing survival and growth of CRPC cells. It was also shown that imatinib treatment stimulated the glycolytic metabolism of CRPC cells. This study first demonstrated that a glucose-enriched environment intensifies the effect of imatinib, which stimulates the interest for testing this compound into the clinical setting, namely in hyperglycemia conditions (diabetic patients) or in co-administration with inhibitors of glycolytic metabolism.


Assuntos
Glucose/metabolismo , Hiperglicemia/patologia , Mesilato de Imatinib/farmacologia , Ácido Láctico/metabolismo , Neoplasias de Próstata Resistentes à Castração/patologia , Inibidores de Proteínas Quinases/farmacologia , Apoptose , Proliferação de Células , Glicólise , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Masculino , Neoplasias de Próstata Resistentes à Castração/tratamento farmacológico , Neoplasias de Próstata Resistentes à Castração/metabolismo , Células Tumorais Cultivadas
8.
J Cell Commun Signal ; 11(4): 297-307, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28656507

RESUMO

The stem cell factor (SCF) is a cytokine that specifically binds the tyrosine kinase receptor c-KIT. The SCF/c-KIT interaction leads to receptor dimerization, activation of kinase activity and initiation of several signal transduction pathways that control cell proliferation, apoptosis, differentiation and migration in several tissues. The activity of SCF/c-KIT system is linked with the phosphatidylinositol 3-kinase (PI3-K), the Src, the Janus kinase/signal transducers and activators of transcription (JAK/STAT), the phospholipase-C (PLC-γ) and the mitogen-activated protein kinase (MAPK) pathways. Moreover, it has been reported that cancer cases display an overactivation of c-KIT due to the presence of gain-of-function mutations or receptor overexpression, which renders c-KIT a tempting target for cancer treatment. In the case of male cancers the most documented activated pathways are the PI3-K and Src, both enhancing abnormal cell proliferation. It is also known that the Src activity in prostate cancer cases depends on the presence of tr-KIT, the cytoplasmic truncated variant of c-KIT that is specifically expressed in tumour tissues and, thus, a very interesting target for drug development. The present review provides an overview of the signalling pathways activated by SCF/c-KIT and discusses the potential application of c-KIT inhibitors for treatment of testicular and prostatic cancers.

9.
Cancer Lett ; 405: 10-21, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28751268

RESUMO

The tyrosine kinase receptor c-KIT and its ligand, the stem cell factor (SCF) are expressed in several tissues of male and female reproductive tract, playing an important role in the regulation of basic biological processes. The activation of c-KIT by SCF controls, cell survival and death, cell differentiation and migration. Also, the SCF/c-KIT system has been implicated in carcinogenesis of reproductive tissues due to its altered expression pattern or overactivation in consequence of gain-of-functions mutations. Over the years, it has also been shown that hormones, the primary regulators of reproductive function and causative agents in the case of hormone-dependent cancers, are also able to control the SCF/c-KIT tissue levels. Therefore, it is liable to suppose that disturbed SCF/c-KIT expression driven by (de)regulated hormone actions can be a relevant step towards carcinogenesis. The present review describes the SCF and c-KIT expression in cancers of reproductive tissues, discussing the implications of the hormonal regulation of the SCF/c-KIT system in cancer development. Understanding the relationship between hormonal imbalance and the SCF/c-KIT expression and activity would be relevant in the context of novel therapeutic approaches in reproductive cancers.


Assuntos
Neoplasias da Mama/metabolismo , Carcinogênese/metabolismo , Hormônios Esteroides Gonadais/fisiologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/metabolismo , Neoplasias Urogenitais/metabolismo , Mama/metabolismo , Neoplasias da Mama/genética , Feminino , Humanos , Masculino , Próstata/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Fator de Células-Tronco/genética , Testículo/metabolismo , Neoplasias Urogenitais/genética , Útero/metabolismo
10.
Biochem Pharmacol ; 99: 73-87, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26592659

RESUMO

The development of prostate cancer (PCa) is intimately associated with the hormonal environment, and the sex steroids estrogens have been implicated in prostate malignancy. However, if some studies identified estrogens as causative agents of PCa, others indicated that these steroids have a protective role counteracting prostate overgrowth. The tyrosine kinase receptor c-KIT and its ligand, the stem cell factor (SCF), have been associated with the control of cell proliferation/apoptosis and prostate carcinogenesis, and studies show that estrogens regulate their expression in different tissues, though, in the case of prostate this remains unknown. The present study aims to evaluate the role of 17ß-estradiol (E2) in regulating the expression of SCF/c-KIT in human prostate cell lines and rat prostate, and to investigate the consequent effects on prostate cell proliferation and apoptosis. qPCR, Western Blot, and immuno(cito)histochemistry analysis showed that E2-treatment decreased the expression of SCF and c-KIT both in human prostate cells and rat prostate. Furthermore, the diminished expression of SCF/c-KIT was underpinned by the diminished prostate weight and reduced proliferation index. On the other hand, the results of TUNEL labelling, the increased activity of caspase-3, and the augmented expression of caspase-8 and Fas system in the prostate of E2-treated animals indicated augmented apoptosis in response to E2. The obtained results demonstrated that E2 down-regulated the expression of SCF/c-KIT system in prostate cells, which was associated with antiproliferative and proapoptotic effects. Moreover, these findings support the protective role of estrogens in PCa and open new perspectives on the application of estrogen-based therapies.


Assuntos
Apoptose/fisiologia , Proliferação de Células/fisiologia , Estrogênios/farmacologia , Próstata/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Fator de Células-Tronco/metabolismo , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Humanos , Masculino , Próstata/citologia , Próstata/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/antagonistas & inibidores , Ratos , Ratos Wistar , Fator de Células-Tronco/antagonistas & inibidores
11.
Med Oncol ; 32(9): 228, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26255063

RESUMO

Androgens have been associated with the development of normal breast, and their role in mammary gland carcinogenesis has also been described. Several studies reported that androgens inhibit breast cancer cell growth, whereas others linked their action with the modulation of calcium (Ca(2+)) pumps, Ca(2+) channels and Ca(2+)-binding proteins. Also, it is known that deregulated Ca(2+) homeostasis has been implicated in the pathophysiology of breast. The L-type Ca(2+) channels (LTCCs) were found to be up-regulated in colon, colorectal and prostate cancer, but their presence in breast tissues remains uncharacterized. On the other hand, regucalcin (RGN) is a Ca(2+)-binding protein involved in the control of mammary gland cell proliferation, which has been identified as an androgen target gene in distinct tissues except breast. This study aimed to confirm the expression and activity of LTCCs in human breast cancer cells and investigate the effect of androgens in regulating the expression of α1C subunit (Cav1.2) of LTCCs and Ca(2+)-binding protein RGN. PCR, Western blot, immunofluorescence and electrophysiological experiments demonstrated the expression and activity of Cav1.2 subunit in MCF-7 cells. The MCF-7 cells were treated with 1, 10 or 100 nM of 5α-dihydrotestosterone (DHT) for 24-72 h. The obtained results showed that 1 nM DHT up-regulated the expression of Cav1.2 subunit while diminishing RGN protein levels, which was underpinned by reduced cell viability. These findings first confirmed the presence of LTCCs in breast cancer cells and opened new perspectives for the development of therapeutic approaches targeting Ca(2+) signaling.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Di-Hidrotestosterona/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Canais de Cálcio Tipo L/análise , Canais de Cálcio Tipo L/genética , Proteínas de Ligação ao Cálcio/análise , Proteínas de Ligação ao Cálcio/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Di-Hidrotestosterona/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/análise , Peptídeos e Proteínas de Sinalização Intracelular/genética
12.
Prog Histochem Cytochem ; 49(1-3): 1-19, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25451758

RESUMO

The c-KIT, a tyrosine kinase receptor, and its ligand the stem cell factor (SCF) play an important role in the production of male and female gametes. The interaction of SCF with c-KIT is required for germ cell survival and growth, and abnormalities in the activity of the SCF/c-KIT system have been associated with human infertility. Recently, it was demonstrated that gonadotropic and sex steroid hormones, among others, regulate the expression of SCF and c-KIT in testicular and ovarian cells. Therefore, the hormonal (de)regulation of SCF/c-KIT system in the testis and ovary may be a cause underpinning infertility. In the present review, we will discuss the effects of hormones modulating the expression levels of SCF and c-KIT in the human gonads. In addition, the implications of hormonal regulation of SCF/c-KIT system for germ cell development and fertility will be highlighted.


Assuntos
Infertilidade/fisiopatologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Feminino , Gonadotropinas/fisiologia , Humanos , Ligantes , Masculino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA