Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Commun ; 12(1): 3299, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-34083537

RESUMO

Bioenergetic perturbations driving neoplastic growth increase the production of reactive oxygen species (ROS), requiring a compensatory increase in ROS scavengers to limit oxidative stress. Intervention strategies that simultaneously induce energetic and oxidative stress therefore have therapeutic potential. Phenformin is a mitochondrial complex I inhibitor that induces bioenergetic stress. We now demonstrate that inflammatory mediators, including IFNγ and polyIC, potentiate the cytotoxicity of phenformin by inducing a parallel increase in oxidative stress through STAT1-dependent mechanisms. Indeed, STAT1 signaling downregulates NQO1, a key ROS scavenger, in many breast cancer models. Moreover, genetic ablation or pharmacological inhibition of NQO1 using ß-lapachone (an NQO1 bioactivatable drug) increases oxidative stress to selectively sensitize breast cancer models, including patient derived xenografts of HER2+ and triple negative disease, to the tumoricidal effects of phenformin. We provide evidence that therapies targeting ROS scavengers increase the anti-neoplastic efficacy of mitochondrial complex I inhibitors in breast cancer.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Fenformin/farmacologia , Fator de Transcrição STAT1/metabolismo , Animais , Antineoplásicos/administração & dosagem , Linhagem Celular Tumoral , Sinergismo Farmacológico , Complexo I de Transporte de Elétrons/antagonistas & inibidores , Metabolismo Energético/efeitos dos fármacos , Feminino , Glutationa/antagonistas & inibidores , Glutationa/biossíntese , Humanos , Interferon gama/administração & dosagem , Interferon gama/deficiência , Interferon gama/metabolismo , Células MCF-7 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos SCID , NAD(P)H Desidrogenase (Quinona)/antagonistas & inibidores , NAD(P)H Desidrogenase (Quinona)/metabolismo , Naftoquinonas/administração & dosagem , Estresse Oxidativo/efeitos dos fármacos , Fenformin/administração & dosagem , Poli I-C/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/agonistas , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Invest Dermatol ; 137(5): 1135-1143, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28143781

RESUMO

Inactivation of the tumor suppressor neurofibromin 1 (NF1) presents a newly characterized melanoma subtype, for which currently no targeted therapies are clinically available. Preclinical studies suggest that extracellular signal-regulated kinase (ERK) inhibitors are likely to provide benefit, albeit with limited efficacy as a single agent; therefore, there is a need for rationally designed combination therapies. Here, we evaluate the combination of the ERK inhibitor SCH772984 and the biguanide phenformin. A combination of both compounds showed potent synergy in cell viability assays and cooperatively induced apoptosis. Treatment with both drugs was required to fully suppress mechanistic target of rapamycin signaling, a known effector of NF1 loss. Mechanistically, SCH772984 increased the oxygen consumption rate, indicating that these cells relied more on oxidative phosphorylation upon treatment. Consistently, SCH772984 increased expression of the mitochondrial transcriptional coactivator peroxisome proliferator-activated receptor gamma, coactivator 1-α. In contrast, cotreatment with phenformin, an inhibitor of complex I of the respiratory chain, decreased the oxygen consumption rate. SCH772984 also promoted the expansion of the H3K4 demethylase KDM5B (also known as JARID1B)-positive subpopulation of melanoma cells, which are slow-cycling and treatment-resistant. Importantly, phenformin suppressed this KDM5B-positive population, which reduced the emergence of SCH772984-resistant clones in long-term cultures. Our results warrant the clinical investigation of this combination therapy in patients with NF1 mutant melanoma.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Indazóis/farmacologia , Melanoma/tratamento farmacológico , Neurofibromina 1/genética , Fenformin/farmacologia , Piperazinas/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Humanos , Indazóis/administração & dosagem , Melanoma/genética , Melanoma/patologia , Mutação , Consumo de Oxigênio/efeitos dos fármacos , Fenformin/administração & dosagem , Piperazinas/administração & dosagem
3.
Oncotarget ; 7(47): 77664-77682, 2016 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-27765910

RESUMO

Mutation of p53 occasionally results in a gain of function, which promotes tumor growth. We asked whether destabilizing the gain-of-function protein would kill tumor cells. Downregulation of the gene reduced cell proliferation in p53-mutant cells, but not in p53-null cells, indicating that the former depended on the mutant protein for survival. Moreover, phenformin and 2-deoxyglucose suppressed cell growth and simultaneously destabilized mutant p53. The AMPK pathway, MAPK pathway, chaperone proteins and ubiquitination all contributed to this process. Interestingly, phenformin and 2-deoxyglucose also reduced tumor growth in syngeneic mice harboring the p53 mutation. Thus, destabilizing mutant p53 protein in order to kill cells exhibiting "oncogene addiction" could be a promising strategy for combatting p53 mutant tumors.


Assuntos
Desoxiglucose/administração & dosagem , Mutação , Neoplasias/patologia , Fenformin/administração & dosagem , Proteína Supressora de Tumor p53/genética , Células A549 , Animais , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desoxiglucose/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Metástase Neoplásica , Fenformin/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Oncotarget ; 7(31): 49397-49410, 2016 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-27384481

RESUMO

Among ALDH isoforms, ALDH1L1 in the folate pathway showed highly increased expression in non-small-cell lung cancer cells (NSCLC). Based on the basic mechanism of ALDH converting aldehyde to carboxylic acid with by-product NADH, we suggested that ALDH1L1 may contribute to ATP production using NADH through oxidative phosphorylation. ALDH1L1 knockdown reduced ATP production by up to 60% concomitantly with decrease of NADH in NSCLC. ALDH inhibitor, gossypol, also reduced ATP production in a dose dependent manner together with decrease of NADH level in NSCLC. A combination treatment of gossypol with phenformin, mitochondrial complex I inhibitor, synergized ATP depletion, which efficiently induced cell death. Pre-clinical xenograft model using human NSCLC demonstrated a remarkable therapeutic response to the combined treatment of gossypol and phenformin.


Assuntos
Trifosfato de Adenosina/metabolismo , Aldeído Desidrogenase/antagonistas & inibidores , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Gossipol/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Fenformin/administração & dosagem , Potenciais de Ação , Aldeído Desidrogenase/metabolismo , Animais , Ácido Aspártico/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Citosol/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Malatos/metabolismo , Potencial da Membrana Mitocondrial , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/metabolismo , NAD/metabolismo , NADP/metabolismo , Transplante de Neoplasias , Fosforilação Oxidativa , Oxirredutases atuantes sobre Doadores de Grupo CH-NH , RNA Interferente Pequeno/metabolismo
5.
Free Radic Res ; 50(8): 929-37, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27328723

RESUMO

Metformin (N,N-dimethylbiguanide), buformin (1-butylbiguanide), and phenformin (1-phenethylbiguanide) are anti-diabetic biguanide drugs, expected to having anti-cancer effect. The mechanism of anti-cancer effect by these drugs is not completely understood. In this study, we demonstrated that these drugs dramatically enhanced oxidative DNA damage under oxidative condition. Metformin, buformin, and phenformin enhanced generation of 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-oxodG) in isolated DNA reacted with hydrogen peroxide (H2O2) and Cu(II), although these drugs did not form 8-oxodG in the absence of H2O2 or Cu(II). An electron paramagnetic resonance (EPR) study, utilizing alpha-(4-pyridyl-1-oxide)-N-tert-butylnitrone and 3,3,5,5-tetramethyl-1-pyrroline-N-oxide as spin trapping agents, showed that nitrogen-centered radicals were generated from biguanides in the presence of Cu(II) and H2O2, and that these radicals were decreased by the addition of DNA. These results suggest that biguanides enhance Cu(II)/H2O2-mediated 8-oxodG generation via nitrogen-centered radical formation. The enhancing effect on oxidative DNA damage may play a role on anti-cancer activity.


Assuntos
Buformina/farmacologia , Dano ao DNA/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Fenformin/farmacologia , Animais , Buformina/administração & dosagem , Bovinos , Dano ao DNA/genética , Humanos , Hipoglicemiantes/administração & dosagem , Metformina/administração & dosagem , Oxirredução , Fenformin/administração & dosagem , Espécies Reativas de Oxigênio
6.
Nanomedicine (Lond) ; 10(18): 2819-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26377155

RESUMO

AIM: Phenformin-loaded micelles (Phen M) were used in combination with gemcitabine-loaded micelles (Gem M) to study their combined effect against H460 human lung cancer cells and cancer stem cells (CSCs) in vitro and in vivo. MATERIALS & METHODS: Gem M and Phen M were prepared via self-assembly of a mixture of a diblock copolymer of PEG and urea-functionalized polycarbonate (PEG-PUC) and a diblock copolymer of PEG and acid-functionalized polycarbonate (PEG-PAC) through hydrogen bonding and ionic interactions. Gem M and Phen M were characterized and tested for efficacy both in vitro and in vivo against cancer cells and CSCs. RESULTS: The combination of Gem M/Phen M exhibited higher cytotoxicity against CSCs and non-CSCs than Gem M and Phen M alone, and showed significant cell cycle growth arrest in vitro. The combination therapy had superior tumor suppression and apoptosis in vivo without inducing toxicity to liver and kidney. CONCLUSION: The combination of Gem M and Phen M may be potentially used in lung cancer therapy.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Desoxicitidina/análogos & derivados , Portadores de Fármacos/química , Neoplasias Pulmonares/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Fenformin/administração & dosagem , Animais , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/administração & dosagem , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Sistemas de Liberação de Medicamentos , Sinergismo Farmacológico , Feminino , Humanos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Micelas , Células-Tronco Neoplásicas/patologia , Fenformin/farmacologia , Fenformin/uso terapêutico , Cimento de Policarboxilato/química , Polietilenoglicóis/química , Ureia/análogos & derivados , Gencitabina
7.
Oncol Rep ; 34(3): 1620-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26133123

RESUMO

Human hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide particularly in Asia. Deregulation of cellular energetics was recently included as one of the cancer hallmarks. Compounds that target the mitochondria in cancer cells were proposed to have therapeutic potential. Biguanide drugs which inhibit mitochondrial complex I and repress mTOR signaling are clinically used to treat type 2 diabetes mellitus patients (T2DM) and were recently found to reduce the risk of HCC in T2DM patients. However, whether alteration of energy metabolism is involved in regulating the sensitivity of HCC to biguanide drugs is still unclear. In the present study, we treated four HCC cell lines with mitochondrial inhibitors (rotenone and oligomycin) and biguanide drugs (metformin and phenformin), and found that the HCC cells which had a higher mitochondrial respiration rate were more sensitive to these treatments; whereas the HCC cells which exhibited higher glycolysis were more resistant. When glucose was replaced by galactose in the medium, the altered energy metabolism from glycolysis to mitochondrial respiration in the HCC cells enhanced the cellular sensitivity to mitochondrial inhibitors and biguanides. The energy metabolism change enhanced AMP-activated protein kinase (AMPK) activation, mTOR repression and downregulation of cyclin D1 and Mcl-1 in response to the mitochondrial inhibitors and biguanides. In conclusion, our results suggest that increased mitochondrial oxidative metabolism upregulates the sensitivity of HCC to biguanide drugs. Enhancing the mitochondrial oxidative metabolism in combination with biguanide drugs may be a therapeutic strategy for HCC.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Metabolismo Energético/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Trifosfato de Adenosina/metabolismo , Apoptose/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Glicólise/efeitos dos fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Metformina/administração & dosagem , Mitocôndrias/metabolismo , Oligomicinas/administração & dosagem , Consumo de Oxigênio/efeitos dos fármacos , Fenformin/administração & dosagem , Rotenona/administração & dosagem
9.
Cancer Res ; 74(24): 7521-33, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25377470

RESUMO

Metformin, a biguanide widely used in the treatment of type II diabetes, clearly exhibits antineoplastic activity in experimental models and has been reported to reduce cancer incidence in diabetics. There are ongoing clinical trials to evaluate its antitumor properties, which may relate to its fundamental activity as an inhibitor of oxidative phosphorylation. Here, we show that serine withdrawal increases the antineoplastic effects of phenformin (a potent biguanide structurally related to metformin). Serine synthesis was not inhibited by biguanides. Instead, metabolic studies indicated a requirement for serine to allow cells to compensate for biguanide-induced decrease in oxidative phosphorylation by upregulating glycolysis. Furthermore, serine deprivation modified the impact of metformin on the relative abundance of metabolites within the citric acid cycle. In mice, a serine-deficient diet reduced serine levels in tumors and significantly enhanced the tumor growth-inhibitory actions of biguanide treatment. Our results define a dietary manipulation that can enhance the efficacy of biguanides as antineoplastic agents that target cancer cell energy metabolism.


Assuntos
Biguanidas/administração & dosagem , Neoplasias/tratamento farmacológico , Fenformin/administração & dosagem , Serina/metabolismo , Animais , Linhagem Celular Tumoral , Glicólise/efeitos dos fármacos , Humanos , Metformina , Camundongos , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação Oxidativa/efeitos dos fármacos , Serina/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Proc Natl Acad Sci U S A ; 110(45): 18226-31, 2013 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-24145418

RESUMO

Biguanides, such as the diabetes therapeutics metformin and phenformin, have demonstrated antitumor activity both in vitro and in vivo. The energy-sensing AMP-activated protein kinase (AMPK) is known to be a major cellular target of biguanides. Based on our discovery of cross-talk between the AMPK and v-Raf murine sarcoma viral oncogene homolog B1 (BRAF) signaling pathways, we investigated the antitumor effects of combining phenformin with a BRAF inhibitor PLX4720 on the proliferation of BRAF-mutated melanoma cells in vitro and on BRAF-driven tumor growth in vivo. Cotreatment of BRAF-mutated melanoma cell lines with phenformin and PLX4720 resulted in synergistic inhibition of cell viability, compared with the effects of the single agent alone. Moreover, treatment with phenformin significantly delayed the development of resistance to PLX4720 in cultured melanoma cells. Biochemical analyses showed that phenformin and PLX4720 exerted cooperative effects on inhibiting mTOR signaling and inducing apoptosis. Noticeably, phenformin selectively targeted subpopulations of cells expressing JARID1B, a marker for slow cycling melanoma cells, whereas PLX4720 selectively targeted JARID1B-negative cells. Finally, in contrast to their use as single agents, the combination of phenformin and PLX4720 induced tumor regression in both nude mice bearing melanoma xenografts and in a genetically engineered BRAF(V600E)/PTEN(null)-driven mouse model of melanoma. These results strongly suggest that significant therapeutic advantage may be achieved by combining AMPK activators such as phenformin with BRAF inhbitors for the treatment of melanoma.


Assuntos
Indóis/farmacologia , Melanoma/tratamento farmacológico , Fenformin/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Sulfonamidas/farmacologia , Análise de Variância , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Imuno-Histoquímica , Indóis/administração & dosagem , Histona Desmetilases com o Domínio Jumonji/metabolismo , Melanoma/genética , Camundongos , Mutação de Sentido Incorreto/genética , Fenformin/administração & dosagem , Proteínas Proto-Oncogênicas B-raf/genética , Sulfonamidas/administração & dosagem
11.
Anticancer Res ; 31(2): 421-6, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21378320

RESUMO

A report that effects of butyrate on some cells may be mediated by activation of AMP-activated protein kinase (AMPK) prompted this study which examines if other AMPK activators can induce differentiation and inhibit proliferation of colon cancer cells in a manner similar to butyrate. Using induction of alkaline phosphatase as a marker, it was observed that compound C, an AMPK inhibitor, is able to reduce the differentiating effect of butyrate on SW1116 and Caco-2 colon cancer cells. Metformin was observed to be less effective than butyrate in the induction of alkaline phosphatase but was more effective as a growth inhibitor. Phenformin was found to be a more potent growth inhibitor than metformin and both compounds cause acidification of the medium when incubated with colon cancer cells. Combined incubation of 2-deoxyglucose with either of the biguanides prevented the acidification of the medium but enhanced the growth inhibitory effects.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias do Colo/tratamento farmacológico , Desoxiglucose/farmacologia , Fenformin/farmacologia , Adenilato Quinase/metabolismo , Fosfatase Alcalina/biossíntese , Fosfatase Alcalina/metabolismo , Butiratos/farmacologia , Células CACO-2 , Processos de Crescimento Celular/efeitos dos fármacos , Neoplasias do Colo/enzimologia , Neoplasias do Colo/patologia , Desoxiglucose/administração & dosagem , Sinergismo Farmacológico , Ativação Enzimática/efeitos dos fármacos , Indução Enzimática/efeitos dos fármacos , Células HT29 , Humanos , Concentração de Íons de Hidrogênio , Fenformin/administração & dosagem
12.
Toxicol Appl Pharmacol ; 233(2): 203-10, 2008 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-18817800

RESUMO

As a class, the biguanides induce lactic acidosis, a hallmark of mitochondrial impairment. To assess potential mitochondrial impairment, we evaluated the effects of metformin, buformin and phenformin on: 1) viability of HepG2 cells grown in galactose, 2) respiration by isolated mitochondria, 3) metabolic poise of HepG2 and primary human hepatocytes, 4) activities of immunocaptured respiratory complexes, and 5) mitochondrial membrane potential and redox status in primary human hepatocytes. Phenformin was the most cytotoxic of the three with buformin showing moderate toxicity, and metformin toxicity only at mM concentrations. Importantly, HepG2 cells grown in galactose are markedly more susceptible to biguanide toxicity compared to cells grown in glucose, indicating mitochondrial toxicity as a primary mode of action. The same rank order of potency was observed for isolated mitochondrial respiration where preincubation (40 min) exacerbated respiratory impairment, and was required to reveal inhibition by metformin, suggesting intramitochondrial bio-accumulation. Metabolic profiling of intact cells corroborated respiratory inhibition, but also revealed compensatory increases in lactate production from accelerated glycolysis. High (mM) concentrations of the drugs were needed to inhibit immunocaptured respiratory complexes, supporting the contention that bioaccumulation is involved. The same rank order was found when monitoring mitochondrial membrane potential, ROS production, and glutathione levels in primary human hepatocytes. In toto, these data indicate that biguanide-induced lactic acidosis can be attributed to acceleration of glycolysis in response to mitochondrial impairment. Indeed, the desired clinical outcome, viz., decreased blood glucose, could be due to increased glucose uptake and glycolytic flux in response to drug-induced mitochondrial dysfunction.


Assuntos
Buformina/toxicidade , Hipoglicemiantes/toxicidade , Metformina/toxicidade , Mitocôndrias Hepáticas/efeitos dos fármacos , Fenformin/toxicidade , Acidose Láctica/induzido quimicamente , Animais , Glicemia/efeitos dos fármacos , Buformina/administração & dosagem , Linhagem Celular , Respiração Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meios de Cultura , Glutationa/efeitos dos fármacos , Glutationa/metabolismo , Glicólise/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Hipoglicemiantes/administração & dosagem , Ácido Láctico/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Metformina/administração & dosagem , Oxirredução/efeitos dos fármacos , Fenformin/administração & dosagem , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
13.
Am J Physiol Heart Circ Physiol ; 293(1): H457-66, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17369473

RESUMO

AMP-activated protein kinase (AMPK) acts as a cellular energy sensor: it responds to an increase in AMP concentration ([AMP]) or the AMP-to-ATP ratio (AMP/ATP). Metformin and phenformin, which are biguanides, have been reported to increase AMPK activity without increasing AMP/ATP. This study tests the hypothesis that these biguanides increase AMPK activity in the heart by increasing cytosolic [AMP]. Groups of isolated rat hearts (n = 5-7 each) were perfused with Krebs-Henseleit buffer with or without 0.2 mM phenformin or 10 mM metformin, and (31)P-NMR-measured phosphocreatine, ATP, and intracellular pH were used to calculate cytosolic [AMP]. At various times, hearts were freeze-clamped and assayed for AMPK activity, phosphorylation of Thr(172) on AMPK-alpha, and phosphorylation of Ser(79) on acetyl-CoA carboxylase, an AMPK target. In hearts treated with phenformin for 18 min and then perfused for 20 min with Krebs-Henseleit buffer, [AMP] began to increase at 26 min and AMPK activity was elevated at 36 min. In hearts treated with metformin, [AMP] was increased at 50 min and AMPK activity, phosphorylated AMPK, and phosphorylated acetyl-CoA carboxylase were elevated at 61 min. In metformin-treated hearts, HPLC-measured total AMP content and total AMP/ATP did not increase. In summary, phenformin and metformin increase AMPK activity and phosphorylation in the isolated heart. The increase in AMPK activity was always preceded by and correlated with increased cytosolic [AMP]. Total AMP content and total AMP/ATP did not change. Cytosolic [AMP] reported metabolically active AMP, which triggered increased AMPK activity, but measures of total AMP did not.


Assuntos
Monofosfato de Adenosina/metabolismo , Adenilato Quinase/metabolismo , Citosol/metabolismo , Metformina/administração & dosagem , Miocárdio/metabolismo , Fenformin/administração & dosagem , Transdução de Sinais/fisiologia , Animais , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Hipoglicemiantes/administração & dosagem , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos
14.
Pol Arch Med Wewn ; 81(4): 231-9, 1989 Apr.
Artigo em Polonês | MEDLINE | ID: mdl-2626349

RESUMO

The authors made a retrospective analysis of late ineffectiveness of sulphonylurea derivatives and its correlation with the age of diabetes onset, duration of the disease, sex, body mass, smoking cigarettes, diabetes history in the family, coexistence with some diabetic complications as retinopathy, ischemic heart disease, arterial hypertension and vascular diseases in lower limbs. The study was carried out in a group of 220 patients with diabetes type 2 lasting from 1 up to 25 years, aged 30-77 undergoing therapy in the Department of Gastroenterology and Metabolic Diseases in the years 1976-1987 in whom a late ineffectiveness appeared to sulphonylurea derivatives. The average time of effective therapy by means of sulphonylurea derivatives in the group examined was 8.7 +/- 4.9 years (9.2 +/- 5.2 in men and 8.3 +/- 4.6 in women). The time of effective therapy by means of the above drugs was inversely proportional to the age of patients when diabetes type 2 appeared. The time of effective therapy by means of sulphonylurea derivatives in obese men was significantly longer than in obese women. No correlation was found between the appearance of late ineffectiveness of sulphonylurea derivatives and diabetes history in the family, smoking ischemic heart disease, arterial hypertension, retinopathy and vascular diseases of lower limbs.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hiperglicemia/etiologia , Hipoglicemiantes/administração & dosagem , Adulto , Fatores Etários , Idoso , Clorpropamida/administração & dosagem , Diabetes Mellitus Tipo 2/sangue , Resistência a Medicamentos , Quimioterapia Combinada , Feminino , Hospitalização , Humanos , Masculino , Metformina/administração & dosagem , Pessoa de Meia-Idade , Fenformin/administração & dosagem , Fatores de Tempo
15.
Vopr Onkol ; 26(12): 28-31, 1980.
Artigo em Russo | MEDLINE | ID: mdl-7467216

RESUMO

Some preliminary results of using phenformin and miscleron in 17 breast cancer patients, previously radically operated upon, have been assessed as being characterized by metabolic disturbances associated with the reduced cell immunity level. The treatment of patients during 15--7 months was shown to result both in the correction of endocrine-metabolic disorders and stimulation of delayed hypersensitivity response, determined by skin tests with dinitro-chlorbenzene, tuberculin and Candida. The necessity to liquidate metabolic metabolic immunodepression with the aim of prophylaxis and treatment of the tuumor process is discussed. Some other routes of metabolic immunotherapy in addition to using phenformin and miscleron are considered. It seems rational to study the effect of a continuous administration of such kind of drugs on the results of treatment of oncological patients.


Assuntos
Neoplasias da Mama/imunologia , Clofibrato/imunologia , Hipersensibilidade Tardia/imunologia , Hipolipemiantes/imunologia , Fenformin/imunologia , Neoplasias da Mama/tratamento farmacológico , Clofibrato/administração & dosagem , Dinitroclorobenzeno , Quimioterapia Combinada , Feminino , Humanos , Hipersensibilidade Tardia/tratamento farmacológico , Hipolipemiantes/administração & dosagem , Pessoa de Meia-Idade , Fenformin/administração & dosagem , Testes Cutâneos , Fatores de Tempo
16.
Cancer Lett ; 7(6): 357-61, 1979 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-509414

RESUMO

It was shown that 1-phenylethylbiguanide (phenformin) administered orally to mice at a dose of 2 mg/day potentiated the antitumor effect of cyclophosphamide on transplantable squamous cell cervical carcinoma, hepatoma-22a and Lewis lung tumor, but did not alter the effect of cyclophosphamide on sarcoma-180 and L-1210. Oral administration of phenformin (5 mg/day) to rats with transplanted Walker 256 carcinoma enhanced the antitumor effect of hydrazine sulfate.


Assuntos
Ciclofosfamida/administração & dosagem , Hidrazinas/administração & dosagem , Neoplasias Experimentais/tratamento farmacológico , Fenformin/administração & dosagem , Animais , Carcinoma 256 de Walker/tratamento farmacológico , Sinergismo Farmacológico , Quimioterapia Combinada , Feminino , Neoplasias Hepáticas Experimentais/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Endogâmicos , Sarcoma 180/tratamento farmacológico , Neoplasias do Colo do Útero/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA