Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 298(2): 101554, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34973337

RESUMEN

The mitochondrial pyruvate carrier (MPC) is an inner mitochondrial membrane complex that plays a critical role in intermediary metabolism. Inhibition of the MPC, especially in liver, may have efficacy for treating type 2 diabetes mellitus. Herein, we examined the antidiabetic effects of zaprinast and 7ACC2, small molecules which have been reported to act as MPC inhibitors. Both compounds activated a bioluminescence resonance energy transfer-based MPC reporter assay (reporter sensitive to pyruvate) and potently inhibited pyruvate-mediated respiration in isolated mitochondria. Furthermore, zaprinast and 7ACC2 acutely improved glucose tolerance in diet-induced obese mice in vivo. Although some findings were suggestive of improved insulin sensitivity, hyperinsulinemic-euglycemic clamp studies did not detect enhanced insulin action in response to 7ACC2 treatment. Rather, our data suggest acute glucose-lowering effects of MPC inhibition may be due to suppressed hepatic gluconeogenesis. Finally, we used reporter sensitive to pyruvate to screen a chemical library of drugs and identified 35 potentially novel MPC modulators. Using available evidence, we generated a pharmacophore model to prioritize which hits to pursue. Our analysis revealed carsalam and six quinolone antibiotics, as well as 7ACC1, share a common pharmacophore with 7ACC2. We validated that these compounds are novel inhibitors of the MPC and suppress hepatocyte glucose production and demonstrated that one quinolone (nalidixic acid) improved glucose tolerance in obese mice. In conclusion, these data demonstrate the feasibility of therapeutic targeting of the MPC for treating diabetes and provide scaffolds that can be used to develop potent and novel classes of MPC inhibitors.


Asunto(s)
Proteínas de Transporte de Anión , Proteínas de Transporte de Membrana Mitocondrial , Transportadores de Ácidos Monocarboxílicos , Obesidad , Quinolonas , Animales , Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Dieta , Glucosa/metabolismo , Ratones , Ratones Obesos , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Transportadores de Ácidos Monocarboxílicos/metabolismo , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Ácido Pirúvico/metabolismo , Quinolonas/farmacología
2.
Cell Metab ; 33(3): 629-648.e10, 2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33333007

RESUMEN

The metabolic rewiring of cardiomyocytes is a widely accepted hallmark of heart failure (HF). These metabolic changes include a decrease in mitochondrial pyruvate oxidation and an increased export of lactate. We identify the mitochondrial pyruvate carrier (MPC) and the cellular lactate exporter monocarboxylate transporter 4 (MCT4) as pivotal nodes in this metabolic axis. We observed that cardiac assist device-induced myocardial recovery in chronic HF patients was coincident with increased myocardial expression of the MPC. Moreover, the genetic ablation of the MPC in cultured cardiomyocytes and in adult murine hearts was sufficient to induce hypertrophy and HF. Conversely, MPC overexpression attenuated drug-induced hypertrophy in a cell-autonomous manner. We also introduced a novel, highly potent MCT4 inhibitor that mitigated hypertrophy in cultured cardiomyocytes and in mice. Together, we find that alteration of the pyruvate-lactate axis is a fundamental and early feature of cardiac hypertrophy and failure.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Cardiomegalia/patología , Insuficiencia Cardíaca/patología , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Animales , Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/genética , Cardiomegalia/inducido químicamente , Cardiomegalia/complicaciones , Insuficiencia Cardíaca/etiología , Corazón Auxiliar , Humanos , Ácido Láctico/metabolismo , Potencial de la Membrana Mitocondrial , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/antagonistas & inhibidores , Proteínas de Transporte de Membrana Mitocondrial/genética , Transportadores de Ácidos Monocarboxílicos/antagonistas & inhibidores , Transportadores de Ácidos Monocarboxílicos/genética , Proteínas Musculares/antagonistas & inhibidores , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Ácido Pirúvico/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Izquierda/fisiología
3.
Biol Pharm Bull ; 43(11): 1792-1798, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33132325

RESUMEN

Xanthine and hypoxanthine are intermediate metabolites of uric acid and a source of reactive oxidative species (ROS) by xanthine oxidoreductase (XOR), suggesting that facilitating their elimination is beneficial. Since they are reabsorbed in renal proximal tubules, we investigated their reabsorption mechanism by focusing on the renal uric acid transporters URAT1 and GLUT9, and examined the effect of clinically used URAT1 inhibitor on their renal clearance when their plasma concentration is increased by XOR inhibitor. Uptake study for [3H]xanthine and [3H]hypoxanthine was performed using URAT1- and GLUT9-expressing Xenopus oocytes. Transcellular transport study for [3H]xanthine was carried out using Madin-Darby canine kidney (MDCK)II cells co-expressing URAT1 and GLUT9. In in vivo pharmacokinetic study, renal clearance of xanthine was estimated based on plasma concentration and urinary recovery. Uptake by URAT1- and GLUT9-expressing oocytes demonstrated that xanthine is a substrate of URAT1 and GLUT9, while hypoxanthine is not. Transcellular transport of xanthine in MDCKII cells co-expressing URAT1 and GLUT9 was significantly higher than those in mock cells and cells expressing URAT1 or GLUT9 alone. Furthermore, dotinurad, a URAT1 inhibitor, increased renal clearance of xanthine in rats treated with topiroxostat to inhibit XOR. It was suggested that xanthine is reabsorbed in the same manner as uric acid through URAT1 and GLUT9, while hypoxanthine is not. Accordingly, it is expected that treatment with XOR and URAT1 inhibitors will effectively decrease purine pools in the body and prevent cell injury due to ROS generated during XOR-mediated reactions.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Proteínas de Transporte de Monosacáridos/metabolismo , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Xantina/farmacocinética , Animales , Proteínas de Transporte de Anión/antagonistas & inhibidores , Benzotiazoles/administración & dosificación , Perros , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Células de Riñón Canino Madin Darby , Modelos Animales , Nitrilos/administración & dosificación , Oocitos , Transportadores de Anión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/genética , Piridinas/administración & dosificación , Ratas , Ratas Wistar , Especies Reactivas de Oxígeno , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Eliminación Renal/efectos de los fármacos , Ácido Úrico/metabolismo , Xantina/sangre , Xantina/metabolismo , Xantina/orina , Xantina Deshidrogenasa/antagonistas & inhibidores , Xantina Deshidrogenasa/metabolismo , Xenopus laevis
4.
Int J Mol Sci ; 21(11)2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32545297

RESUMEN

Proteins are relevant antimicrobial drug targets, and among them, enzymes represent a significant group, since most of them catalyze reactions essential for supporting the central metabolism, or are necessary for the pathogen vitality. Genomic exploration of pathogenic and non-pathogenic microorganisms has revealed genes encoding for a superfamily of metalloenzymes, known as carbonic anhydrases (CAs, EC 4.2.1.1). CAs catalyze the physiologically crucial reversible reaction of the carbon dioxide hydration to bicarbonate and protons. Herein, we investigated the sulfonamide inhibition profile of the recombinant ß-CA (CynT2) identified in the genome of the Gram-negative bacterium Escherichia coli. This biocatalyst is indispensable for the growth of the microbe at atmospheric pCO2. Surprisingly, this enzyme has not been investigated for its inhibition with any class of CA inhibitors. Here, we show that CynT2 was strongly inhibited by some substituted benzene-sulfonamides and the clinically used inhibitor sulpiride (KIs in the range of 82-97 nM). This study may be relevant for identifying novel CA inhibitors, as well as for another essential part of the drug discovery pipeline, such as the structure-activity relationship for this class of enzyme inhibitors.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Antibacterianos/farmacología , Inhibidores de Anhidrasa Carbónica/farmacología , Anhidrasas Carbónicas/metabolismo , Proteínas de Escherichia coli/metabolismo , Sulfonamidas/química , Sulfonamidas/farmacología , Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/genética , Antibacterianos/química , Benceno/química , Dióxido de Carbono/química , Dióxido de Carbono/metabolismo , Inhibidores de Anhidrasa Carbónica/química , Anhidrasas Carbónicas/genética , Evaluación Preclínica de Medicamentos/métodos , Proteínas de Escherichia coli/antagonistas & inhibidores , Proteínas de Escherichia coli/genética , Humanos , Relación Estructura-Actividad
5.
Int J Biol Macromol ; 159: 570-576, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32442571

RESUMEN

Volume-regulated anion channel (VRAC) is ubiquitously expressed in vertebrate cells and in various types of cancer cells. Leucine-rich repeat containing 8A (LRRC8A) and its four homologous family members (LRRC8B-E) assemble into heterogeneous VRAC complexes of ~800 kDa. The main components of VRAC, LRRC8A and LRRC8D have been implicated in the proliferation, migration, death, and multidrug resistance of cancer cells through their involvement in various signal pathways. This review summarizes recent findings concerning the involvement of VRAC in cancer development and progression, including the molecular structure, function, and regulation of VRAC and its roles in various cancers, and highlights the remaining challenges in the field. Our aim is to evaluate the potential of VRAC as a therapeutic target for cancer therapies and to discuss the major problems to be solved.


Asunto(s)
Proteínas de Transporte de Anión/metabolismo , Biomarcadores de Tumor , Neoplasias/etiología , Neoplasias/metabolismo , Animales , Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/química , Proteínas de Transporte de Anión/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Unión Proteica , Multimerización de Proteína , Transducción de Señal/efectos de los fármacos
6.
Am J Physiol Renal Physiol ; 318(4): F870-F877, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-31984792

RESUMEN

Adenosine plays an important role in various aspects of kidney physiology, but the specific targets and mechanisms of actions are not completely understood. The collecting duct has the highest expression of adenosine receptors, particularly adenosine A1 receptors (A1Rs). Interstitial adenosine levels are greatly increased up to a micromolar range in response to dietary salt loading. We have previously shown that the basolateral membrane of principal cells has primarily K+ conductance mediated by Kir4.1/5.1 channels to mediate K+ recycling and to set up a favorable driving force for Na+/K+ exchange (47). Intercalated cells express the Cl- ClC-K2/b channel mediating transcellular Cl- reabsorption. Using patch-clamp electrophysiology in freshly isolated mouse collecting ducts, we found that acute application of adenosine reversely inhibits ClC-K2/b open probability from 0.31 ± 0.04 to 0.17 ± 0.06 and to 0.10 ± 0.05 for 1 and 10 µM, respectively. In contrast, adenosine (10 µM) had no measureable effect on Kir4.1/5.1 channel activity in principal cells. The inhibitory effect of adenosine on ClC-K2/b was abolished in the presence of the A1R blocker 8-cyclopentyl-1,3-dipropylxanthine (10 µM). Consistently, application of the A1R agonist N6-cyclohexyladenosine (1 µM) recapitulated the inhibitory action of adenosine on ClC-K2/b open probability. The effects of adenosine signaling in the collecting duct were independent from its purinergic counterpartner, ATP, having no measurable actions on ClC-K2/b and Kir4.1/5.1. Overall, we demonstrated that adenosine selectively inhibits ClC-K2/b activity in intercalated cells by targeting A1Rs. We propose that inhibition of transcellular Cl- reabsorption in the collecting duct by adenosine would aid in augmenting NaCl excretion during high salt intake.


Asunto(s)
Agonistas del Receptor de Adenosina A1/farmacología , Adenosina/farmacología , Proteínas de Transporte de Anión/antagonistas & inhibidores , Canales de Cloruro/antagonistas & inhibidores , Cloruros/metabolismo , Túbulos Renales Colectores/efectos de los fármacos , Receptor de Adenosina A1/efectos de los fármacos , Reabsorción Renal/efectos de los fármacos , Animales , Proteínas de Transporte de Anión/metabolismo , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Canales de Cloruro/metabolismo , Túbulos Renales Colectores/citología , Túbulos Renales Colectores/metabolismo , Masculino , Potenciales de la Membrana/efectos de los fármacos , Ratones Endogámicos C57BL , Receptor de Adenosina A1/metabolismo
7.
Physiol Rep ; 7(23): e14232, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31833218

RESUMEN

Slc4a11, a member of the Slc4 HCO3- transporter family, has a wide tissue distribution. In mouse salivary glands, the expression of Slc4a11 mRNA was more than eightfold greater than the other nine members of the Slc4 gene family. The Slc4a11 protein displayed a diffuse subcellular distribution in both the acinar and duct cells of mouse submandibular glands (SMG). Slc4a11 disruption induced a significant increase in the Na+ and Cl- concentrations of stimulated SMG saliva, whereas it did not affect the fluid secretion rate in response to either ß-adrenergic or cholinergic receptor stimulation. Heterologous expressed mouse Slc4a11 acted as a H+ /OH- transporter that was uncoupled of Na+ or Cl- movement, and this activity was blocked by ethyl-isopropyl amiloride (EIPA) but not 4,4'-Diisothiocyanato-2,2'-stilbenedisulfonic acid (DIDS). Slc4a11 disruption revealed that Slc4a11 does not play a major role in intracellular pH regulation in mouse salivary gland cells. In contrast, NaCl reabsorption was impaired in the SMG saliva of female compared to male Slc4a11 null mice, which correlated with the loss of duct cells and a decrease in expression of the duct-cell-specific transcription factor Ascl3. Together, our results suggest that Slc4a11 expression regulates the number of ducts cells in the mouse SMG and consequently NaCl reabsorption.


Asunto(s)
Absorción Fisiológica , Proteínas de Transporte de Anión/metabolismo , Protones , Cloruro de Sodio/metabolismo , Glándula Submandibular/metabolismo , Simportadores/metabolismo , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Amilorida/análogos & derivados , Amilorida/farmacología , Animales , Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/genética , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Glándula Submandibular/citología , Simportadores/antagonistas & inhibidores , Simportadores/genética
8.
Mol Nutr Food Res ; 63(12): e1801402, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30913372

RESUMEN

SCOPE: Conjugated linoleic acid (CLA), a bioactive substance predominantly found in ruminant products, improves insulin resistance and exhibits anti-inflammatory activity. The chief objective of the study is to investigate the effects and potential mechanisms of CLA on high fructose-induced hyperuricemia and renal inflammation. METHODS AND RESULTS: Hyperuricemia and renal inflammation are induced in rats by 10% fructose. Hyperuricemia, insulin resistance, and renal inflammation are evaluated. CLA potently ameliorates fructose-induced hyperuricemia with insulin resistance and significantly reduces the levels of inflammation factors in serum and kidney. It reverses fructose-induced upregulation of glucose transporter 9 (GLUT9) and urate transporter 1 (URAT1) in the kidney. Moreover, CLA dramatically inhibits the activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. Additionally, CLA suppresses toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MyD88) signaling activation to inhibit nuclear factor-kB (NF-kB) signaling in the kidney of fructose-fed rats. CONCLUSION: CLA ameliorates hyperuricemia along with insulin resistance and renal inflammatory, which may be associated with the suppression of renal GLUT9 and URAT1 in fructose-fed rats. Its molecular mechanism may be related to the inhibition of NLRP3 inflammasome and TLR4/MyD88 signaling pathway. Therefore, CLA may be a promising candidate for preventing fructose-induced hyperuricemia and renal inflammation.


Asunto(s)
Fructosa/administración & dosificación , Hiperuricemia/tratamiento farmacológico , Inflamasomas/fisiología , Inflamación/tratamiento farmacológico , Riñón/efectos de los fármacos , Ácidos Linoleicos Conjugados/farmacología , Proteína con Dominio Pirina 3 de la Familia NLR/fisiología , Receptor Toll-Like 4/fisiología , Animales , Proteínas de Transporte de Anión/antagonistas & inhibidores , Ácidos Linoleicos Conjugados/uso terapéutico , Masculino , Proteínas de Transporte de Monosacáridos/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
9.
Cancer Res ; 78(10): 2513-2523, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29510993

RESUMEN

Glycolysis and fatty acid synthesis are highly active in cancer cells through cytosolic citrate metabolism, with intracellular citrate primarily derived from either glucose or glutamine via the tricarboxylic acid cycle. We show here that extracellular citrate is supplied to cancer cells through a plasma membrane-specific variant of the mitochondrial citrate transporter (pmCiC). Metabolomic analysis revealed that citrate uptake broadly affected cancer cell metabolism through citrate-dependent metabolic pathways. Treatment with gluconate specifically blocked pmCiC and decreased tumor growth in murine xenografts of human pancreatic cancer. This treatment altered metabolism within tumors, including fatty acid metabolism. High expression of pmCiC was associated with invasion and advanced tumor stage across many human cancers. These findings support the exploration of extracellular citrate transport as a novel potential target for cancer therapy.Significance: Uptake of extracellular citrate through pmCiC can be blocked with gluconate to reduce tumor growth and to alter metabolic characteristics of tumor tissue. Cancer Res; 78(10); 2513-23. ©2018 AACR.


Asunto(s)
Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/metabolismo , Proliferación Celular/efectos de los fármacos , Ácido Cítrico/metabolismo , Gluconatos/farmacología , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Células Epiteliales/metabolismo , Ácidos Grasos/biosíntesis , Glucólisis/fisiología , Humanos , Masculino , Ratones , Transportadores de Anión Orgánico , Próstata/citología , Próstata/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética
10.
J Cell Physiol ; 233(2): 1414-1423, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28543431

RESUMEN

Existing studies on the mechanism of cell volume regulation are mainly relevant to ion channels and osmosis in extracellular fluid. Recently, accumulating evidence has shown that cellular mechanical microenvironment also influences the cell volume. Herein, we investigated the regulation of substrate stiffness on the cell volume homeostasis of MCF-7 cells and their following migration behaviors. We found that cell volume increases with increasing substrate stiffness, which could be affected by blocking the cell membrane anion permeability and dopamine receptor. In addition, the cell migration is significantly inhibited by decreasing the cell volume using tamoxifen and such inhibition effect on migration is enhanced by increasing substrate stiffness. The cell membrane anion permeability might be the linker between cellular mechanical microenvironment and cellular volume homeostasis regulation. This work revealed the regulation of substrate stiffness on cell volume homeostasis for the first time, which would provide a new perspective into the understanding of cancer metastasis and a promising anti-cancer therapy through regulation of cell volume homeostasis.


Asunto(s)
Resinas Acrílicas/química , Neoplasias de la Mama/metabolismo , Membrana Celular/metabolismo , Tamaño de la Célula , Osmorregulación , Ácido 4,4'-Diisotiocianostilbeno-2,2'-Disulfónico/farmacología , Proteínas de Transporte de Anión/antagonistas & inhibidores , Proteínas de Transporte de Anión/metabolismo , Antineoplásicos Hormonales/farmacología , Benzazepinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Adhesión Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/patología , Permeabilidad de la Membrana Celular , Movimiento Celular , Tamaño de la Célula/efectos de los fármacos , Colágeno/metabolismo , Femenino , Humanos , Hidrogeles , Soluciones Hipotónicas/farmacología , Células MCF-7 , Osmorregulación/efectos de los fármacos , Porosidad , Receptores de Dopamina D1/antagonistas & inhibidores , Receptores de Dopamina D1/metabolismo , Tamoxifeno/farmacología , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...