Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
1.
Nat Commun ; 12(1): 3299, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083537

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Fenformina/farmacología , Factor de Transcripción STAT1/metabolismo , Animales , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Sinergismo Farmacológico , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Metabolismo Energético/efectos de los fármacos , Femenino , Glutatión/antagonistas & inhibidores , Glutatión/biosíntesis , Humanos , Interferón gamma/administración & dosificación , Interferón gamma/deficiencia , Interferón gamma/metabolismo , Células MCF-7 , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones SCID , NAD(P)H Deshidrogenasa (Quinona)/antagonistas & inhibidores , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Naftoquinonas/administración & dosificación , Estrés Oxidativo/efectos de los fármacos , Fenformina/administración & dosificación , Poli I-C/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT1/agonistas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Biomolecules ; 10(10)2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33019635

RESUMEN

Damage to cerebral mitochondria, particularly opening of mitochondrial permeability transition pore (MPTP), is a key mechanism of ischemic brain injury, therefore, modulation of MPTP may be a potential target for a neuroprotective strategy in ischemic brain pathologies. The aim of this study was to investigate whether biguanides-metformin and phenformin as well as other inhibitors of Complex I of the mitochondrial electron transfer system may protect against ischemia-induced cell death in brain slice cultures by suppressing MPTP, and whether the effects of these inhibitors depend on the age of animals. Experiments were performed on brain slice cultures prepared from 5-7-day (premature) and 2-3-month old (adult) rat brains. In premature brain slice cultures, simulated ischemia (hypoxia plus deoxyglucose) induced necrosis whereas in adult rat brain slice cultures necrosis was induced by hypoxia alone and was suppressed by deoxyglucose. Phenformin prevented necrosis induced by simulated ischemia in premature and hypoxia-induced-in adult brain slices, whereas metformin was protective in adult brain slices cultures. In premature brain slices, necrosis was also prevented by Complex I inhibitors rotenone and amobarbital and by MPTP inhibitor cyclosporine A. The latter two inhibitors were protective in adult brain slices as well. Short-term exposure of cultured neurons to phenformin, metformin and rotenone prevented ionomycin-induced MPTP opening in intact cells. The data suggest that, depending on the age, phenformin and metformin may protect the brain against ischemic damage possibly by suppressing MPTP via inhibition of mitochondrial Complex I.


Asunto(s)
Lesiones Encefálicas/tratamiento farmacológico , Metformina/administración & dosificación , Mitocondrias/efectos de los fármacos , Fenformina/administración & dosificación , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Complejo I de Transporte de Electrón/genética , Humanos , Masculino , Mitocondrias/genética , Poro de Transición de la Permeabilidad Mitocondrial/antagonistas & inhibidores , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Necrosis por Permeabilidad de la Transmembrana Mitocondrial/efectos de los fármacos , Necrosis/tratamiento farmacológico , Necrosis/metabolismo , Necrosis/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/administración & dosificación , Fosforilación Oxidativa/efectos de los fármacos , Ratas
3.
Drug Dev Res ; 81(4): 390-401, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-31916629

RESUMEN

Diabetes mellitus is a serious metabolic disorder affecting millions of people worldwide. Phenformin and metformin are biguanide antidiabetic agents that are conveniently synthesized in a single-step chemical reaction. Phenformin was once used to lower blood glucose levels, but later withdrawn from the market in several countries because it was frequently associated with lactic acidosis. Metformin is still a widely prescribed medication for the treatment of type 2 diabetes despite the introduction of several newer antidiabetic agents. Metformin is administered orally and has desirable pharmacokinetics. Incidence of metformin-induced lactic acidosis is serious but very rare. Imeglimin, a novel molecule being investigated by Poxel and Sumitomo Dainippon Pharma in Japan, is currently in clinical trials for the treatment of type 2 diabetes. Unlike metformin, imeglimin is a cyclic molecule containing a triazine ring. However, like metformin, imeglimin is also a basic small molecule. Imeglimin is synthesized from metformin as a precursor via a single step chemical reaction. Recent mechanism of action studies suggests that imeglimin improves mitochondria function, when given in combination with metformin it helps achieve better glycemic control in patients with type 2 diabetes. We herein describe and compare the current status, synthesis, physicochemical properties, pharmacokinetic parameters, mechanism of action, and preclinical/clinical studies of metformin and imeglimin.


Asunto(s)
Metformina/administración & dosificación , Fenformina/administración & dosificación , Triazinas/administración & dosificación , Acidosis Láctica/inducido químicamente , Animales , Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/efectos adversos , Hipoglucemiantes/farmacocinética , Metformina/efectos adversos , Metformina/farmacocinética , Fenformina/efectos adversos , Fenformina/farmacocinética , Triazinas/efectos adversos , Triazinas/farmacocinética
5.
J Invest Dermatol ; 137(5): 1135-1143, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28143781

RESUMEN

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.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Indazoles/farmacología , Melanoma/tratamiento farmacológico , Neurofibromina 1/genética , Fenformina/farmacología , Piperazinas/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Indazoles/administración & dosificación , Melanoma/genética , Melanoma/patología , Mutación , Consumo de Oxígeno/efectos de los fármacos , Fenformina/administración & dosificación , Piperazinas/administración & dosificación
6.
Oncotarget ; 7(47): 77664-77682, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27765910

RESUMEN

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.


Asunto(s)
Desoxiglucosa/administración & dosificación , Mutación , Neoplasias/patología , Fenformina/administración & dosificación , Proteína p53 Supresora de Tumor/genética , Células A549 , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desoxiglucosa/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Sustitución del Gen , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Metástasis de la Neoplasia , Fenformina/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 7(31): 49397-49410, 2016 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-27384481

RESUMEN

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.


Asunto(s)
Adenosina Trifosfato/metabolismo , Aldehído Deshidrogenasa/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Gosipol/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Fenformina/administración & dosificación , Potenciales de Acción , Aldehído Deshidrogenasa/metabolismo , Animales , Ácido Aspártico/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Citosol/metabolismo , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Malatos/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo , NAD/metabolismo , NADP/metabolismo , Trasplante de Neoplasias , Fosforilación Oxidativa , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , ARN Interferente Pequeño/metabolismo
8.
Free Radic Res ; 50(8): 929-37, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27328723

RESUMEN

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.


Asunto(s)
Buformina/farmacología , Daño del ADN/efectos de los fármacos , Hipoglucemiantes/farmacología , Metformina/farmacología , Fenformina/farmacología , Animales , Buformina/administración & dosificación , Bovinos , Daño del ADN/genética , Humanos , Hipoglucemiantes/administración & dosificación , Metformina/administración & dosificación , Oxidación-Reducción , Fenformina/administración & dosificación , Especies Reactivas de Oxígeno
9.
Nanomedicine (Lond) ; 10(18): 2819-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26377155

RESUMEN

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.


Asunto(s)
Antimetabolitos Antineoplásicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Desoxicitidina/análogos & derivados , Portadores de Fármacos/química , Neoplasias Pulmonares/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Fenformina/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Desoxicitidina/administración & dosificación , Desoxicitidina/farmacología , Desoxicitidina/uso terapéutico , Sistemas de Liberación de Medicamentos , Sinergismo Farmacológico , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones Endogámicos BALB C , Ratones Desnudos , Micelas , Células Madre Neoplásicas/patología , Fenformina/farmacología , Fenformina/uso terapéutico , Cemento de Policarboxilato/química , Polietilenglicoles/química , Urea/análogos & derivados , Gemcitabina
10.
Oncol Rep ; 34(3): 1620-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26133123

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Metabolismo Energético/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Glucólisis/efectos de los fármacos , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Metformina/administración & dosificación , Mitocondrias/metabolismo , Oligomicinas/administración & dosificación , Consumo de Oxígeno/efectos de los fármacos , Fenformina/administración & dosificación , Rotenona/administración & dosificación
12.
Cancer Res ; 74(24): 7521-33, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25377470

RESUMEN

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.


Asunto(s)
Biguanidas/administración & dosificación , Neoplasias/tratamiento farmacológico , Fenformina/administración & dosificación , Serina/metabolismo , Animales , Línea Celular Tumoral , Glucólisis/efectos de los fármacos , Humanos , Metformina , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Fosforilación Oxidativa/efectos de los fármacos , Serina/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Drug Res (Stuttg) ; 64(9): 454-61, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24307270

RESUMEN

A computational study has suggested that phenformin, an oral hypoglycaemic drug, may bind to the active sites of the monoamine oxidase (MAO) A and B enzymes. The present study therefore investigates the MAO inhibitory properties of phenformin. Pentamidine, a structurally related diamidine compound, has previously been reported to be a MAO inhibitor and was included in this study as a reference compound. Using recombinant human MAO-A and MAO-B, this study finds that phenformin acts as a moderately potent MAO-A selective inhibitor with an IC50 value of 41 µM. Pentamidine, on the other hand, potently inhibits both MAO-A and MAO-B with IC50 values of 0.61 µM and 0.22 µM, respectively. An examination of the recoveries of the enzymatic activities after dilution and dialysis of the enzyme-inhibitor complexes shows that both compounds interact reversibly with the MAO enzymes. A kinetic analysis suggests that pentamidine acts as a competitive inhibitor with estimated Ki values of 0.41 µM and 0.22 µM for the inhibition of MAO-A and MAO-B, respectively. Phenformin also exhibited a competitive mode of MAO-A inhibition with an estimated Ki value of 65 µM. This study concludes that biguanide and amidine functional groups are most likely important structural features for the inhibition of the MAOs by phenformin and pentamidine, and compounds containing these and closely related functional groups should be considered as potential MAO inhibitors. Furthermore, the biguanide and amidine functional groups may act as useful moieties in the future design of MAO inhibitors.


Asunto(s)
Inhibidores de la Monoaminooxidasa/farmacología , Monoaminooxidasa/metabolismo , Pentamidina/farmacología , Fenformina/farmacología , Humanos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/farmacología , Concentración 50 Inhibidora , Pentamidina/administración & dosificación , Fenformina/administración & dosificación , Proteínas Recombinantes
14.
Proc Natl Acad Sci U S A ; 110(45): 18226-31, 2013 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-24145418

RESUMEN

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.


Asunto(s)
Indoles/farmacología , Melanoma/tratamiento farmacológico , Fenformina/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/farmacología , Análisis de Varianza , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Inmunohistoquímica , Indoles/administración & dosificación , Histona Demetilasas con Dominio de Jumonji/metabolismo , Melanoma/genética , Ratones , Mutación Missense/genética , Fenformina/administración & dosificación , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/administración & dosificación
15.
Zhongguo Zhong Yao Za Zhi ; 37(19): 2960-3, 2012 Oct.
Artículo en Chino | MEDLINE | ID: mdl-23270242

RESUMEN

OBJECTIVE: To study the effect of phenformin hydrochloride that may be illegally added in traditional Chinese medicine preparations on the pharmacokinetics of puerarin in rats. METHOD: Rats were randomly divided into the single pueraria group and the phenformin hydrochloride combined with pueraria group. After oral administration in the two groups, their bloods were sampled at different time points to determine the drug concentration of puerarin in rat blood and calculate pharmacokinetic parameters. RESULT: After oral administration with pueraria extracts and phenformin hydrochloride combined with pueraria extracts, the two groups showed main pharmacokinetic parameters as follows: Cmax were (2.39 +/- 1.01), (1.03 +/- 0.35) mg x L(-1), respectively; Tmax were (0.50 +/- 0.09), (1.5 +/- 0.5) h, respectively; Ke were (0.153 +/- 0.028), (0.172 +/- 0.042) h(-1), respectively; t(1/2) were (4.65 +/- 0.86), (4.20 +/- 0.81) h, respectively; AUC(0-t), were (5.73 +/- 2.60), (5.45 +/- 1.81) mg x h x L(-1), respectively; AUC(0-infinity) were (6.72 +/- 2.89), (6.26 +/- 1.88) mg x h x L(-1), respectively. Compared with the single puerarin group, the Cmax was significantly decreased (P < 0.05) and the Tmax was markedly longer (P < 0.01) than the hydrochloride combined with pueraria group. CONCLUSION: Phenformin hydrochloride can slow down the absorption process of puerarin and change the pharmacokinetic process of puerarin to some extent.


Asunto(s)
Hipoglucemiantes/farmacología , Isoflavonas/farmacocinética , Fenformina/farmacología , Vasodilatadores/farmacocinética , Administración Oral , Animales , Interacciones Farmacológicas , Hipoglucemiantes/administración & dosificación , Isoflavonas/administración & dosificación , Masculino , Fenformina/administración & dosificación , Ratas , Ratas Wistar , Vasodilatadores/administración & dosificación
16.
Anticancer Res ; 31(2): 421-6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21378320

RESUMEN

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.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Colon/tratamiento farmacológico , Desoxiglucosa/farmacología , Fenformina/farmacología , Adenilato Quinasa/metabolismo , Fosfatasa Alcalina/biosíntesis , Fosfatasa Alcalina/metabolismo , Butiratos/farmacología , Células CACO-2 , Procesos de Crecimiento Celular/efectos de los fármacos , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Desoxiglucosa/administración & dosificación , Sinergismo Farmacológico , Activación Enzimática/efectos de los fármacos , Inducción Enzimática/efectos de los fármacos , Células HT29 , Humanos , Concentración de Iones de Hidrógeno , Fenformina/administración & dosificación
17.
Toxicol Appl Pharmacol ; 233(2): 203-10, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18817800

RESUMEN

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.


Asunto(s)
Buformina/toxicidad , Hipoglucemiantes/toxicidad , Metformina/toxicidad , Mitocondrias Hepáticas/efectos de los fármacos , Fenformina/toxicidad , Acidosis Láctica/inducido químicamente , Animales , Glucemia/efectos de los fármacos , Buformina/administración & dosificación , Línea Celular , Respiración de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Medios de Cultivo , Glutatión/efectos de los fármacos , Glutatión/metabolismo , Glucólisis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hipoglucemiantes/administración & dosificación , Ácido Láctico/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metformina/administración & dosificación , Oxidación-Reducción/efectos de los fármacos , Fenformina/administración & dosificación , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo
18.
J Diabetes Complications ; 21(2): 108-17, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17331859

RESUMEN

The present study aimed to investigate the actions of several classes of oral hypoglycemic agents [e.g., sulfonylureas (SUs), biguanides (BGs) and thiazolidinediones (TZDs)] in an in vitro model of lipid binding based on the "response to retention" hypothesis of atherogenesis. The incorporation of [(35)S]-SO(4) into proteoglycans synthesized by human vascular smooth muscle cells (VSMCs) was assessed by cetylpyridinium chloride (CPC) precipitation method, proteoglycan electrophoretic mobility was evaluated by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE), and binding to low-density lipoprotein (LDL) was assessed by gel mobility shift assay (GMSA). The SUs evaluated showed no effect on [(35)S]-SO(4) incorporation into proteoglycans. Only one BG, phenformin, caused a concentration-related inhibition of proteoglycan synthesis under basal conditions and in the presence of transforming growth factor-beta1 (TGF-beta1), caused by an inhibition of proteoglycan core protein synthesis secondary to a reduction in total protein synthesis. However, neither metformin nor phenformin (30-300 micromol/l) had any effect on the electrophoretic mobility of proteoglycans. The TZDs--troglitazone (TRO), rosiglitazone (ROS), and pioglitazone (PIO) (10, 30, and 30 micromol/l, respectively)--inhibited proteoglycan biosynthesis and stimulated total proteoglycan core protein synthesis, while TRO alone inhibited overall protein synthesis. All three TZDs moderately reduced the electrophoretic mobility of synthesized proteoglycans assessed by SDS-PAGE, reduced the sizes of cleaved glycosaminoglycan (GAG) chains assessed by size exclusion chromatography, and significantly reduced binding to LDL. The data indicate that TZDs show anti-atherogenic actions through the modification of proteoglycan structure, leading to a possible reduction in lipid retention in the vessel wall.


Asunto(s)
Hipoglucemiantes/farmacología , Músculo Liso Vascular/efectos de los fármacos , Proteoglicanos/metabolismo , Administración Oral , Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Células Cultivadas , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Ensayo de Cambio de Movilidad Electroforética , Gliburida/farmacología , Glicosaminoglicanos/metabolismo , Humanos , Hipoglucemiantes/administración & dosificación , Lipoproteínas LDL/metabolismo , Metformina/administración & dosificación , Metformina/farmacología , Músculo Liso Vascular/metabolismo , Fenformina/administración & dosificación , Fenformina/farmacología , Unión Proteica , Sulfatos/antagonistas & inhibidores , Sulfatos/metabolismo , Compuestos de Sulfonilurea/administración & dosificación , Compuestos de Sulfonilurea/farmacología , Radioisótopos de Azufre , Tiazolidinedionas/farmacología , Factor de Crecimiento Transformador beta1/farmacología
19.
Am J Physiol Heart Circ Physiol ; 293(1): H457-66, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17369473

RESUMEN

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.


Asunto(s)
Adenosina Monofosfato/metabolismo , Adenilato Quinasa/metabolismo , Citosol/metabolismo , Metformina/administración & dosificación , Miocardio/metabolismo , Fenformina/administración & dosificación , Transducción de Señal/fisiología , Animales , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Hipoglucemiantes/administración & dosificación , Masculino , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
20.
J Physiol ; 566(Pt 3): 781-92, 2005 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15919715

RESUMEN

Active re-absorption of Na+ across the alveolar epithelium is essential to maintain lung fluid balance. Na+ entry at the luminal membrane is predominantly via the amiloride-sensitive Na+ channel (ENaC) down its electrochemical gradient. This gradient is generated and maintained by basolateral Na+ extrusion via Na+,K+-ATPase an energy-dependent process. Several kinases and factors that activate them are known to regulate these processes; however, the role of AMP-activated protein kinase (AMPK) in the lung is unknown. AMPK is an ultra-sensitive cellular energy sensor that monitors energy consumption and down-regulates ATP-consuming processes when activated. The biguanide phenformin has been shown to independently decrease ion transport processes, influence cellular metabolism and activate AMPK. The AMP mimetic drug 5-aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside (AICAR) also activates AMPK in intact cells. Western blotting revealed that both the alpha1 and alpha2 catalytic subunits of AMPK are present in Na+ transporting H441 human lung epithelial cells. Phenformin and AICAR increased AMPK activity in H441 cells in a dose-dependent fashion, stimulating the kinase maximally at 5-10 mm (P = 0.001, n = 3) and 2 mm (P < 0.005, n = 3), respectively. Both agents significantly decreased basal ion transport (measured as short circuit current) across H441 monolayers by approximately 50% compared with that of controls (P < 0.05, n = 4). Neither treatment altered the resistance of the monolayers. Phenformin and AICAR significantly reduced amiloride-sensitive transepithelial Na+ transport compared with controls (P < 0.05, n = 4). This was a result of both decreased Na+,K+-ATPase activity and amiloride-sensitive apical Na+ conductance. Transepithelial Na+ transport decreased with increasing concentrations of phenformin (0.1-10 mm) and showed a significant correlation with AMPK activity. Taken together, these results show that phenformin and AICAR suppress amiloride-sensitive Na+ transport across H441 cells via a pathway that includes activation of AMPK and inhibition of both apical Na+ entry through ENaC and basolateral Na+ extrusion via the Na+,K+-ATPase. These are the first studies to provide a cellular signalling mechanism for the action of phenformin on ion transport processes, and also the first studies showing AMPK as a regulator of Na+ absorption in the lung.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Pulmón/metabolismo , Complejos Multienzimáticos/efectos de los fármacos , Complejos Multienzimáticos/metabolismo , Fenformina/administración & dosificación , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Ribonucleótidos/administración & dosificación , Sodio/metabolismo , Proteínas Quinasas Activadas por AMP , Aminoimidazol Carboxamida/administración & dosificación , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Línea Celular , Relación Dosis-Respuesta a Droga , Activación Enzimática , Humanos , Pulmón/efectos de los fármacos , Mucosa Respiratoria/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...