Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 166
Filtrar
1.
Bioorg Chem ; 148: 107432, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38744169

RESUMEN

Adenylate kinase (AK) plays a crucial role in the metabolic monitoring of cellular adenine nucleotide homeostasis by catalyzing the reversible transfer of a phosphate group between ATP and AMP, yielding two ADP molecules. By regulating the nucleotide levels and energy metabolism, the enzyme is considered a disease modifier and potential therapeutic target for various human diseases, including malignancies and inflammatory and neurodegenerative disorders. However, lacking approved drugs targeting AK hinders broad studies on this enzyme's pathological importance and therapeutic potential. In this work, we determined the effect of a series of dinucleoside polyphosphate derivatives, commercially available (11 compounds) and newly synthesized (8 compounds), on the catalytic activity of human adenylate kinase isoenzyme 1 (hAK1). The tested compounds belonged to the following groups: (1) diadenosine polyphosphates with different phosphate chain lengths, (2) base-modified derivatives, and (3) phosphate-modified derivatives. We found that all the investigated compounds inhibited the catalytic activity of hAK1, yet with different efficiencies. Three dinucleoside polyphosphates showed IC50 values below 1 µM, and the most significant inhibitory effect was observed for P1-(5'-adenosyl) P5-(5'-adenosyl) pentaphosphate (Ap5A). To understand the observed differences in the inhibition efficiency of the tested dinucleoside polyphosphates, the molecular docking of these compounds to hAK1 was performed. Finally, we conducted a quantitative structure-activity relationship (QSAR) analysis to establish a computational prediction model for hAK1 modulators. Two PLS-regression-based models were built using kinetic data obtained from the AK1 activity analysis performed in both directions of the enzymatic reaction. Model 1 (AMP and ATP synthesis) had a good prediction power (R2 = 0.931, Q2 = 0.854, and MAE = 0.286), while Model 2 (ADP synthesis) exhibited a moderate quality (R2 = 0.913, Q2 = 0.848, and MAE = 0.370). These studies can help better understand the interactions between dinucleoside polyphosphates and adenylate kinase to attain more effective and selective inhibitors in the future.


Asunto(s)
Adenilato Quinasa , Fosfatos de Dinucleósidos , Relación Estructura-Actividad Cuantitativa , Humanos , Fosfatos de Dinucleósidos/química , Fosfatos de Dinucleósidos/síntesis química , Fosfatos de Dinucleósidos/farmacología , Fosfatos de Dinucleósidos/metabolismo , Cinética , Estructura Molecular , Adenilato Quinasa/metabolismo , Adenilato Quinasa/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química
2.
Dig Dis Sci ; 67(2): 524-535, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-33555515

RESUMEN

OBJECTIVE: Alcohol consumption is always the main cause of acute pancreatitis (AP). It has been reported that alcohol exerts direct damage to the pancreas. However, the specific role of alcohol during AP needs to be investigated. This study aims to examine the effects of alcohol in cerulein-induced AP and the role of the AMPK pathway. METHODS: Human subjects from operations, cerulein-induced AP rat, and cerulein-stimulated AR42J cell line were enrolled in this study. Electron microscopy was employed for observation of cell morphology, immunohistochemistry for identification of cells, ELISA for detection of inflammation factors, Annexin V/PI double staining for evaluation of cell apoptosis, immunofluorescence for assessment of autophagic flux, oil red O staining for examination of lipid droplet accumulation, and Western blot for measurement of expressions of proteins related to autophagy, apoptosis, and AMPK signal pathway. PI3K inhibitor 3-MA and AMPK inhibitor BML-275 were utilized for investigation of the relationship between impaired autophagic flux and the AMPK pathway by inhibiting or stimulating the formation of autophagosome. RESULTS: Alcohol consumption caused lipid droplet accumulation in the pancreas, and it also activated AMPK signaling pathway, thus aggravating the autophagic flux during AP. Alcohol up-regulated the expressions of anti-apoptotic proteins during the induction of AP to inhibit cell apoptosis and enhance cell necrosis. Inhibition of autophagosome formation by AMPK inhibitor BML-275 ameliorated the decreased cell viability caused by alcohol and cerulein in vitro. CONCLUSION: Alcohol aggravates AP progression by impairing autophagic flux and enhancing cell autophagy through the AMPK signaling pathway.


Asunto(s)
Adenilato Quinasa/metabolismo , Autofagia/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Etanol/farmacología , Páncreas/efectos de los fármacos , Pancreatitis Alcohólica/metabolismo , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/efectos de los fármacos , Animales , Línea Celular , Ceruletida/toxicidad , Humanos , Pancreatitis/inducido químicamente , Pancreatitis/metabolismo , Pancreatitis/patología , Pancreatitis Alcohólica/patología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Ratas , Transducción de Señal
3.
JCI Insight ; 6(19)2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34473647

RESUMEN

Herein, we report that Shroom3 knockdown, via Fyn inhibition, induced albuminuria with foot process effacement (FPE) without focal segmental glomerulosclerosis (FSGS) or podocytopenia. Interestingly, knockdown mice had reduced podocyte volumes. Human minimal change disease (MCD), where podocyte Fyn inactivation was reported, also showed lower glomerular volumes than FSGS. We hypothesized that lower glomerular volume prevented the progression to podocytopenia. To test this hypothesis, we utilized unilateral and 5/6th nephrectomy models in Shroom3-KD mice. Knockdown mice exhibited less glomerular and podocyte hypertrophy after nephrectomy. FYN-knockdown podocytes had similar reductions in podocyte volume, implying that Fyn was downstream of Shroom3. Using SHROOM3 or FYN knockdown, we confirmed reduced podocyte protein content, along with significantly increased phosphorylated AMPK, a negative regulator of anabolism. AMPK activation resulted from increased cytoplasmic redistribution of LKB1 in podocytes. Inhibition of AMPK abolished the reduction in glomerular volume and induced podocytopenia in mice with FPE, suggesting a protective role for AMPK activation. In agreement with this, treatment of glomerular injury models with AMPK activators restricted glomerular volume, podocytopenia, and progression to FSGS. Glomerular transcriptomes from MCD biopsies also showed significant enrichment of Fyn inactivation and Ampk activation versus FSGS glomeruli. In summary, we demonstrated the important role of AMPK in glomerular volume regulation and podocyte survival. Our data suggest that AMPK activation adaptively regulates glomerular volume to prevent podocytopenia in the context of podocyte injury.


Asunto(s)
Adenilato Quinasa/metabolismo , Glomérulos Renales/metabolismo , Proteínas de Microfilamentos/genética , Síndrome Nefrótico/genética , Podocitos/metabolismo , Adenilato Quinasa/antagonistas & inhibidores , Adolescente , Adulto , Anciano , Albuminuria/genética , Animales , Tamaño de la Célula , Supervivencia Celular/genética , Niño , Preescolar , Femenino , Técnicas de Silenciamiento del Gen , Glomerulonefritis Membranosa/genética , Glomerulonefritis Membranosa/patología , Glomeruloesclerosis Focal y Segmentaria/genética , Glomeruloesclerosis Focal y Segmentaria/patología , Humanos , Hipertrofia , Lactante , Glomérulos Renales/patología , Masculino , Ratones , Persona de Mediana Edad , Nefrectomía , Nefrosis Lipoidea/genética , Nefrosis Lipoidea/patología , Síndrome Nefrótico/patología , Podocitos/patología , Modelos de Riesgos Proporcionales , Proteínas Proto-Oncogénicas c-fyn/genética , Adulto Joven
4.
Acta Biochim Biophys Sin (Shanghai) ; 53(9): 1154-1165, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34355237

RESUMEN

Short-chain chlorinated paraffins (SCCPs) have been listed as a new class of persistent organic pollutants by the Stockholm Convention. SCCPs exhibit carcinogenic-, endocrine-, and metabolism-disrupting effects. However, the knowledge of the immunomodulatory effects of SCCPs and their underlying mechanisms, especially in specific immune cells, remains limited. In addition to SCCPs, C9-13-CPs have also been detected in humans. In this study, murine RAW264.7 macrophages were exposed to C9-13-CPs at environmentally relevant concentrations to investigate whether or how C9-13-CPs exhibit immunomodulatory effects. The results showed that the exposure of RAW264.7 cells to C9-13-CPs increased cell viability, as assayed by MTT analysis at 490 nm, and also promoted cell proliferation, as indicated by EdU uptake assay, which was measured at excitation and emission wavelengths of 488 and 512 nm, respectively. In addition, exposure to C9-13-CPs not only led to elevated ATP level and intracellular Ca2+ level but also caused AMPK signaling activation and NF-κB signaling inhibition. Moreover, molecular docking showed that the ß2-AR receptor could bind to C9-13-CPs. Taken together, these results suggest that the immune dysfunction of RAW264.7 cells caused by C9-13-CPs is closely related to the ß2-AR/AMPK/NF-κB signaling axis.


Asunto(s)
Hidrocarburos Clorados/inmunología , Hidrocarburos Clorados/toxicidad , Inmunomodulación/efectos de los fármacos , Macrófagos/efectos de los fármacos , Parafina/análogos & derivados , Parafina/toxicidad , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/genética , Adenilato Quinasa/metabolismo , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/genética , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Activación de Macrófagos/efectos de los fármacos , Macrófagos/citología , Ratones , Simulación del Acoplamiento Molecular , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Células RAW 264.7 , Receptores Adrenérgicos beta 2/metabolismo
5.
mSphere ; 6(4): e0053921, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34406854

RESUMEN

Treatment of invasive mold infections is limited by the lack of adequate drug options that are effective against these fatal infections. High-throughput screening of molds using traditional antifungal assays of growth is problematic and has greatly limited our ability to identify new mold-active agents. Here, we present a high-throughput screening platform for use with Aspergillus fumigatus, the most common causative agent of invasive mold infections, for the discovery of novel mold-active antifungals. This assay detects cell lysis through the release of the cytosolic enzyme adenylate kinase and, thus, is not dependent on changes in biomass or metabolism to detect antifungal activity. The ability to specifically detect cell lysis is a unique aspect of this assay that allows identification of molecules that disrupt fungal cell integrity, such as cell wall-active molecules. We also found that germinating A. fumigatus conidia release low levels of adenylate kinase and that a reduction in this background allowed us to identify molecules that inhibit conidial germination, expanding the potential for discovery of novel antifungal compounds. Here, we describe the validation of this assay and proof-of-concept pilot screens that identified a novel antifungal compound, PIK-75, that disrupts cell wall integrity. This screening assay provides a novel platform for high-throughput screens with A. fumigatus for the identification of anti-mold drugs. IMPORTANCE Fungal infections caused by molds have the highest mortality rates of human fungal infections. These devastating infections are hard to treat and available antifungal drugs are often not effective. Therefore, the identification of new antifungal drugs with mold activity is critical. Drug screening with molds is challenging and there are limited assays available to identify new antifungal compounds directly with these organisms. Here, we present an assay suitable for use for high-throughput screening with a common mold pathogen. This assay has exciting future potential for the identification of new drugs to treat these fatal infections.


Asunto(s)
Antifúngicos/farmacología , Aspergillus fumigatus/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Adenilato Quinasa/antagonistas & inhibidores , Aspergilosis/tratamiento farmacológico , Aspergillus fumigatus/enzimología , Aspergillus fumigatus/crecimiento & desarrollo , Pared Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos/métodos , Humanos , Prueba de Estudio Conceptual , Bibliotecas de Moléculas Pequeñas/farmacología , Esporas Fúngicas/efectos de los fármacos , Esporas Fúngicas/enzimología
6.
Acta Biochim Biophys Sin (Shanghai) ; 53(9): 1189-1197, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34357376

RESUMEN

Glucagon-like peptide-1 (GLP-1), a novel type of glucose-lowering agent, has been reported to exert cardioprotective effects. However, the cardioprotective mechanism of GLP-1 on spontaneous hypertension-induced cardiac hypertrophy has not been fully elucidated. In this study, we revealed that liraglutide or alogliptin treatment ameliorated spontaneous hypertension-induced cardiac hypertrophy, as evidenced by decreased levels of cardiac hypertrophic markers (atrial natriuretic peptide, brain natriuretic peptide, and ß-myosin heavy chain), as well as systolic blood pressure, diastolic blood pressure, mean arterial pressure, and histological changes. Both drugs significantly reduced the levels of angiotensin II (AngII) and AngII type 1 receptor (AT1R) and upregulated the levels of AngII type 2 receptor (AT2R) and angiotensin-converting enzyme 2 (ACE2), as indicated by a reduced AT1R/AT2R ratio. Simultaneously, treatment with liraglutide or alogliptin significantly increased GLP-1 receptor expression and adenosine monophosphate-activated protein kinase (AMPK) phosphorylation and downregulated the phosphorylation of mammalian target of rapamycin (mTOR), p70 ribosomal S6 protein kinase, and eukaryotic translation initiation factor 4E binding protein 1 in spontaneous hypertension rats. Furthermore, our data demonstrated that the AMPK inhibitor compound C or mTOR activator MHY1485 inhibited the anti-hypertrophic effect of GLP-1. In summary, our study suggests that liraglutide or alogliptin protects the heart against cardiac hypertrophy by regulating the expression of AngII/AT1R/ACE2 and activating the AMPK/mTOR pathway, and GLP-1 agonist can be used in the treatment of patients with cardiac hypertrophy.


Asunto(s)
Adenilato Quinasa/metabolismo , Angiotensina II/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Receptor de Angiotensina Tipo 1/metabolismo , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Adenilato Quinasa/antagonistas & inhibidores , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Cardiomegalia/prevención & control , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Péptido 1 Similar al Glucagón/uso terapéutico , Hipertensión/complicaciones , Liraglutida/farmacología , Liraglutida/uso terapéutico , Masculino , Morfolinas/farmacología , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/farmacología , Piperidinas/uso terapéutico , Ratas , Sistema Renina-Angiotensina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Triazinas/farmacología , Uracilo/análogos & derivados , Uracilo/farmacología , Uracilo/uso terapéutico
7.
Acta Biochim Biophys Sin (Shanghai) ; 53(9): 1177-1188, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34244711

RESUMEN

Stroke is the second leading cause of death and long-term disability worldwide, which lacks effective treatment. Perioperative stroke is associated with much higher rates of mortality and disability. The neuroprotective role of dexmedetomidine (Dex), a highly selective agonist of alpha2-adrenergic receptor, has been reported in a stroke rat model, and it was found that pretreatment of Dex before stroke could alleviate blood-brain barrier (BBB) breakdown. However, the underlying mechanisms are still unknown. As the brain endothelial cells are the main constituents of BBB and in high demand of energy, mitochondrial function of endothelial cells plays an important role in the maintenance of BBB. Given that dynamin-related protein 1 (Drp1) is a protein mediating mitochondrial fission, with mitochondrial fusion that balances mitochondrial morphology and ensures mitochondria function, the present study was designed to investigate the possible role of Drp1 in endothelial cells involved in the neuroprotective effects of Dex in ischemic stroke. Our results showed that preconditioning with Dex reduced infarction volume, alleviated brain water content and BBB damage, and improved neurological scores in middle cerebral artery occlusion rats. Meanwhile, Dex enhanced cell activity and decreased cell apoptosis in oxygen-glucose deprivation human brain microvascular endothelial cells in vitro. These protective effects of Dex were correlated with the mitochondrial morphology integrality of endothelial cells, mediated by increased phosphorylation of serine 637 in Drp1, and could be reversed by α2-adrenergic receptor antagonist Yohimbine and AMP-activated protein kinase inhibitor Compound C. These findings suggest new molecular pathways involved in the neuroprotective effects of Dex in ischemic stroke. As Dex is routinely used as a sedative drug clinically, our findings provide molecular evidence that it has perioperative neuroprotection from ischemic stroke.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Dexmedetomidina/farmacología , Dinaminas/metabolismo , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Mitocondrias/metabolismo , Fármacos Neuroprotectores/farmacología , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 2/farmacología , Antagonistas de Receptores Adrenérgicos alfa 2/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Línea Celular , Citocinas/metabolismo , Dexmedetomidina/uso terapéutico , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Alcaloides Indólicos/farmacología , Alcaloides Indólicos/uso terapéutico , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Accidente Cerebrovascular Isquémico/etiología , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Masculino , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Dinámicas Mitocondriales/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Proteína de la Zonula Occludens-1/metabolismo
8.
Int J Mol Sci ; 22(5)2021 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-33669058

RESUMEN

Differentiation-inducing factor-1 (DIF-1) is a chlorinated alkylphenone (a polyketide) found in the cellular slime mold Dictyostelium discoideum. DIF-1 and its derivative, DIF-1(3M) promote glucose consumption in vitro in mammalian cells and in vivo in diabetic rats; they are expected to be the leading antiobesity and antidiabetes compounds. In this study, we investigated the mechanisms underlying the actions of DIF-1 and DIF-1(3M). In isolated mouse liver mitochondria, these compounds at 2-20 µM promoted oxygen consumption in a dose-dependent manner, suggesting that they act as mitochondrial uncouplers, whereas CP-DIF-1 (another derivative of DIF-1) at 10-20 µM had no effect. In confluent mouse 3T3-L1 fibroblasts, DIF-1 and DIF-1(3M) but not CP-DIF-1 induced phosphorylation (and therefore activation) of AMP kinase (AMPK) and promoted glucose consumption and metabolism. The DIF-induced glucose consumption was reduced by compound C (an AMPK inhibitor) or AMPK knock down. These data suggest that DIF-1 and DIF-1(3M) promote glucose uptake, at least in part, via an AMPK-dependent pathway in 3T3-L1 cells, whereas cellular metabolome analysis revealed that DIF-1 and DIF-1(3M) may act differently at least in part.


Asunto(s)
Adenilato Quinasa/metabolismo , Dictyostelium/metabolismo , Glucosa/metabolismo , Hexanonas/farmacología , Hidrocarburos Clorados/farmacología , Metaboloma/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Pirazoles/farmacología , Pirimidinas/farmacología , Células 3T3 , Adenilato Quinasa/antagonistas & inhibidores , Animales , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Técnicas de Silenciamiento del Gen , Ratones , Mitocondrias/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Fosforilación , ARN Interferente Pequeño , Transducción de Señal/efectos de los fármacos
9.
J Med Chem ; 63(19): 11215-11234, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32914624

RESUMEN

Current pulmonary arterial hypertension (PAH) therapeutic strategies mainly focus on vascular relaxation with less emphasis on vascular remodeling, which results in poor prognosis. Hence, dual pathway regulators with vasodilation effect via soluble guanylate cyclase (sGC) stimulation and vascular remodeling regulation effect by AMP-activated protein kinase (AMPK) inhibition provide more advantages and potentialities. Herein, we designed and synthesized a series of novel pyrazolo[3,4-b] pyridine derivatives based on sGC stimulator and AMPK inhibitor scaffolds. In vitro, 2 exhibited moderate vasodilation activity and higher proliferation and migration suppressive effects compared to riociguat. In vivo, 2 significantly decreased right ventricular systolic pressure (RVSP), attenuated pulmonary artery medial thickness (PAMT), and right ventricular hypertrophy (RVH) in hypoxia-induced PAH rat models (i.g.). Given the unique advantages of significant vascular remodeling inhibition and moderate vascular relaxation based on the dual pathway regulation, we proposed 2 as a promising lead for anti-PAH drug discovery.


Asunto(s)
Diseño de Fármacos , Hipertensión Pulmonar/tratamiento farmacológico , Pirazoles/química , Piridinas/química , Remodelación Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/metabolismo , Animales , Línea Celular , Humanos , Pirazoles/farmacología , Piridinas/farmacología , Ratas , Relación Estructura-Actividad
10.
Eur J Clin Invest ; 50(6): e13247, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32307703

RESUMEN

BACKGROUND: The mechanism underlying the occurrence of the J wave in low temperature remains unclear. However, low temperature is associated with metabolic disorder and 5' AMP-activated protein kinase (AMPK), which modulates ionic currents and cardiac metabolism. This study investigated whether AMPK regulation can modulate the occurrence of the J wave at low temperature. METHODS: Unipolar and bipolar leads were used to record monophasic action potential (the endocardium and epicardium) and pseudo-electrocardiograms (inferior leads) to study the cardiac electrical activity. Measurements were taken in isolated Langendorff rabbit hearts at both 30℃ and 37℃ before and after administration of 4-aminopyridine (an ultrarapid delayed rectifier potassium current inhibitor, IKur , 50 µmol L-1 ), PF06409577 (an AMPK activator, 1 µmol L-1 ), compound C (an AMPK inhibitor, 10 µmol L-1 ) and glibenclamide (an ATP-sensitive inward rectifier potassium channel inhibitor, IKATP , 20 µmol L-1 ). RESULTS: The amplitude of the J wave (2.46 ± 0.34 mV vs. 1.11 ± 0.23 mV, P < .01) at 30℃ (n = 15) was larger than that at 37℃ (n = 15). PF06409577 (1 µmol L-1 ) increased the J waves at both 30℃ and 37℃. In contrast, compound C (10 µmol L-1 ) reduced J wave at both 37℃ and 30℃. Low-temperature-induced J waves were individually suppressed by 4-AP (50 µmol L-1 ) and glibenclamide (20 µmol L-1 ). CONCLUSIONS: AMPK inhibition reduces low-temperature-induced J waves and possible ventricular arrhythmogenesis by modulating IKATP and IKur channels.


Asunto(s)
Potenciales de Acción/fisiología , Adenilato Quinasa/metabolismo , Frío , Corazón/fisiopatología , Hipotermia/fisiopatología , Adenilato Quinasa/antagonistas & inhibidores , Aminopiridinas/farmacología , Animales , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Trastorno del Sistema de Conducción Cardíaco/fisiopatología , Electrocardiografía , Activadores de Enzimas/farmacología , Gliburida/farmacología , Corazón/efectos de los fármacos , Hipotermia/metabolismo , Preparación de Corazón Aislado , Canales KATP/metabolismo , Conejos
11.
Nat Metab ; 2(1): 41-49, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31993556

RESUMEN

Central to cellular metabolism and cell proliferation are highly conserved signalling pathways controlled by mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK)1,2, dysregulation of which are implicated in pathogenesis of major human diseases such as cancer and type 2 diabetes. AMPK pathways leading to reduced cell proliferation are well established and, in part, act through inhibition of TOR complex-1 (TORC1) activity. Here we demonstrate reciprocal regulation, specifically that TORC1 directly down-regulates AMPK signalling by phosphorylating the evolutionarily conserved residue Ser367 in the fission yeast AMPK catalytic subunit Ssp2, and AMPK α1Ser347/α2Ser345 in the mammalian homologs, which is associated with reduced phosphorylation of activation loop Thr172. Genetic or pharmacological inhibition of TORC1 signalling led to AMPK activation in the absence of increased AMP:ATP ratios; under nutrient stress conditions this was associated with growth limitation in both yeast and human cell cultures. Our findings reveal fundamental, bi-directional regulation between two major metabolic signalling networks and uncover new opportunity for cancer treatment strategies aimed at suppressing cell proliferation in the nutrient-poor tumor microenvironment.


Asunto(s)
Adenilato Quinasa/antagonistas & inhibidores , Proliferación Celular/fisiología , Diana Mecanicista del Complejo 1 de la Rapamicina/fisiología , Nutrientes/metabolismo , Estrés Fisiológico , Adenilato Quinasa/química , Adenilato Quinasa/metabolismo , Dominio Catalítico , Diabetes Mellitus Tipo 2/metabolismo , Regulación hacia Abajo , Activación Enzimática , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/efectos de los fármacos , Neoplasias/metabolismo , Fosforilación , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Transducción de Señal/fisiología
12.
IUBMB Life ; 71(12): 1929-1936, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31359579

RESUMEN

Colorectal cancer (CRC) is among the leading causes of cancer-related mortality, despite extensive efforts in the identification of new treatment options. Hence, there is a need for the development of novel agents with therapeutic potential in treatment of CRC. Dorsomorphin has demonstrated antiproliferative activity against different malignancies. Here we have investigated the pharmaceutical potential of dorsomorphin in two-dimensional and three-dimensional cell-culture models of CRC. The antiproliferative, antimigratory, apoptotic activity and effect of this agent on cell cycle was evaluated by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, wound healing assay, and flow cytometry, respectively, while the expression of genes involved in Wnt/Pi3K pathways was assessed at mRNA and/or proteins by reverse transcription polymerase chain reaction (RT-PCR) or western blot. Dorsomorphin inhibited CRC cell growth by modulating the cyclinD1, surviving and p-Akt. This agent was able to reduce the migratory behaviors of CRC cells, compared to control cells, through perturbation of E-cadherin. Also our data showed that dorsomorphin enhanced the percentage of the cells in sub-G1 and induced apoptosis in both late/early stages, as detected by annexin V. Also the regulatory effect of dorsomorphin on oxidant/antioxidant balance was assessed by cellular reactive oxygen species (ROS) generation. In particular, these data showed that dorsomorphin markedly increased the ROS production in CRC cells. Our finding demonstrated that dorsomorphin antagonizes cell growth and migration, through perturbation of Akt/mTOR/Wnt pathways in CRC, supporting further studies on the therapeutic potential of this novel anticancer agent in treatment of CRC.


Asunto(s)
Adenilato Quinasa/antagonistas & inhibidores , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
13.
FASEB J ; 33(9): 10089-10103, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31199678

RESUMEN

Sorafenib is a multikinase inhibitor that is effective in treating advanced liver cancer. Although its mechanism of action through several established cancer-related protein kinase targets is well-characterized, sorafenib induces variable responses among human tumors, and the cause for this variation is yet unknown. To investigate the underlying mechanisms, we applied mass spectrometry-based proteomic analysis to Huh7.5 human liver cancer cells and found that sorafenib significantly affected the expression of the key lipogenic enzymes, especially stearoyl coenzyme A desaturase 1 (SCD1), in these cells. Given that SCD1 catalyzes the most crucial and rate-limiting step in the synthesis of monounsaturated fatty acids (FAs), we performed a lipidomic analysis, which showed a dramatically altered lipid profile in sorafenib-treated cells. Detection and analysis of free FAs showed that the levels of monounsaturated FAs, including oleate, were significantly decreased in those cells treated by sorafenib. Addition of oleate protected liver cancer cells from sorafenib-induced death and alleviated the abnormalities of mitochondrial morphology and function caused by the drug. Treatment with sorafenib suppressed ATP production, resulting in AMPK activation via phosphorylation. Further secondary effects included reduction of the levels of sterol regulatory element-binding protein 1 (SREBP1) and the phosphorylation of mammalian target of rapamycin (mTOR) in liver cancer cells. These effects were partly abolished in the presence of compound C (an AMPK inhibitor) and ATP and adenosine, and SREBP1c overexpression also could be resistant to the effects of sorafenib, suggesting that the sorafenib-induced reduction in cell viability was mediated by the ATP-AMPK-mTOR-SREBP1 signaling pathway. Taken together, our results suggest that sorafenib's anticancer activity in liver cancer cells is based on the inhibition of ATP production, SCD1 expression, and monounsaturated FA synthesis. In addition, the decreased monounsaturated FA synthesis further triggered the more serious reduction of ATP production in sorafenib-treated cells. To our knowledge, this is the first evidence that sorafenib disrupts lipogenesis and triggers liver cancer cell death by targeting SCD1 through the ATP-AMPK-mTOR-SREBP1 pathway.-Liu, G., Kuang, S., Cao, R., Wang, J., Peng, Q., Sun, C. Sorafenib kills liver cancer cells by disrupting SCD1-mediated synthesis of monounsaturated fatty acids via the ATP-AMPK-mTOR- SREBP1 signaling pathway.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Antineoplásicos/farmacología , Carcinoma Hepatocelular/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Sorafenib/farmacología , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/fisiología , Animales , Antineoplásicos/uso terapéutico , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Humanos , Lipidómica , Neoplasias Hepáticas/patología , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/ultraestructura , Ácido Oléico/farmacología , Fosforilación , Inhibidores de Proteínas Quinasas/uso terapéutico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes/metabolismo , Sorafenib/uso terapéutico , Estearoil-CoA Desaturasa/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología , Serina-Treonina Quinasas TOR/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Commun Signal ; 17(1): 8, 2019 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-30683114

RESUMEN

BACKGROUND: Ghrelin modulates many physiological processes. However, the effects of intestinal ghrelin on hepatic glucose production (HGP) are still unclear. The current study was to explore the roles of intestinal ghrelin on glucose homeostasis and insulin signaling in the liver. METHODS: The system of intraduodenal infusion and intracerebral microinfusion into the nucleus of the solitary tract (NTS) in the normal chow-diet rats and pancreatic-euglycemic clamp procedure (PEC) combined with [3-3H] glucose as a tracer were used to analyze the effect of intestinal ghrelin. Intraduodenal co-infusion of ghrelin, tetracaine and Activated Protein Kinase (AMPK) activator (AICAR), or pharmacologic and molecular inhibitor of N-methyl-D-aspartate receptors within the dorsal vagal complex, or hepatic vagotomy in rats were used to explore the possible mechanism of the effect of intestinal ghrelin on HGP. RESULTS: Our results demonstrated that gut infusion of ghrelin inhibited duodenal AMP-dependent protein kinase (AMPK) signal pathways, increased HGP and expression of gluconeogenic enzymes, and decreased insulin signaling in the liver of the rat. Intraduodenal co-infusion of ghrelin receptor antagonist [D-Lys3]-GHRP-6 and AMPK agonist with ghrelin diminished gut ghrelin-induced increase in HGP and decrease in glucose infusion rate (GIR) and hepatic insulin signaling. The effects of gut ghrelin were also negated by co-infusion with tetracaine, or MK801, an N-methyl-D-aspartate (NMDA) receptor inhibitor, and adenovirus expressing the shRNA of NR1 subunit of NMDA receptors (Ad-shNR1) within the dorsal vagal complex, and hepatic vagotomy in rats. When ghrelin and lipids were co-infused into the duodenum, the roles of gut lipids in increasing the rate of glucose infusion (GIR) and lowering HGP were reversed. CONCLUSIONS: The current study provided evidence that intestinal ghrelin has an effect on HGP and identified a neural glucoregulatory function of gut ghrelin signaling.


Asunto(s)
Encéfalo/metabolismo , Tracto Gastrointestinal/metabolismo , Ghrelina/farmacología , Glucosa/biosíntesis , Insulina/metabolismo , Hígado/metabolismo , Transducción de Señal , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/metabolismo , Animales , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Ayuno , Tracto Gastrointestinal/efectos de los fármacos , Homeostasis , Mucosa Intestinal/metabolismo , Hígado/efectos de los fármacos , Hígado/enzimología , Masculino , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
15.
J Hematol Oncol ; 12(1): 12, 2019 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-30696468

RESUMEN

BACKGROUND: Adenylate kinase 4 (AK4) has been identified as a biomarker of metastasis in lung cancer. However, the impacts of AK4 on metabolic genes and its translational value for drug repositioning remain unclear. METHODS: Ingenuity upstream analyses were used to identify potential transcription factors that regulate the AK4 metabolic gene signature. The expression of AK4 and its upstream regulators in lung cancer patients was examined via immunohistochemistry. Pharmacological and gene knockdown/overexpression approaches were used to investigate the interplay between AK4 and its upstream regulators during epithelial-to-mesenchymal transition (EMT). Drug candidates that reversed AK4-induced gene expression were identified by querying a connectivity map. Orthotopic xenograft mouse models were established to evaluate the therapeutic efficacy of drug candidates for metastatic lung cancer. RESULTS: We found that HIF-1α is activated in the AK4 metabolic gene signature. IHC analysis confirmed this positive correlation, and the combination of both predicts worse survival in lung cancer patients. Overexpression of AK4 exaggerates HIF-1α protein expression by increasing intracellular ROS levels and subsequently induces EMT under hypoxia. Attenuation of ROS production with N-acetylcysteine abolishes AK4-induced invasion potential under hypoxia. Pharmacogenomics analysis of the AK4 gene signature revealed that withaferin-A could suppress the AK4-HIF-1α signaling axis and serve as a potent anti-metastatic agent in lung cancer. CONCLUSIONS: Overexpression of AK4 promotes lung cancer metastasis by enhancing HIF-1α stability and EMT under hypoxia. Reversing the AK4 gene signature with withaferin-A may serve as a novel therapeutic strategy to treat metastatic lung cancer.


Asunto(s)
Adenocarcinoma del Pulmón/metabolismo , Adenilato Quinasa/genética , Adenilato Quinasa/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/metabolismo , Estrés Oxidativo , Células A549 , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/patología , Adenilato Quinasa/antagonistas & inhibidores , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Transición Epitelial-Mesenquimal/genética , Estudios de Seguimiento , Técnicas de Silenciamiento del Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica/genética , Especies Reactivas de Oxígeno/metabolismo , Transfección , Hipoxia Tumoral , Witanólidos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
PLoS One ; 14(12): e0226940, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31891641

RESUMEN

Diets high in saturated fatty acids are linked to increased cardiovascular disease risk, whereas monounsaturated fatty acids have been associated with improved cardiovascular outcomes. Accordingly, cell culture studies have demonstrated that saturated fatty acids, particularly long chain saturated fatty acids such as palmitate, induce dysfunction and cell death in a variety of cell types, and monounsaturated fatty acids may confer protection against palmitate-mediated damage. The aim of the present study was to examine whether monounsaturated fatty acids could protect against palmitate-mediated cell death in endothelial cells, to determine if AMPK inactivation and activation (via compound C and AICAR, respectively) underlies both palmitate-induced damage and monounsaturated fatty acid-mediated protection, and to explore the role of ER stress in this context. Human umbilical vein endothelial cells were examined for cell viability and apoptosis following treatment for 24 hours with palmitate (0.25 and 0.5mM) alone or in combination with the monounsaturated fatty acids oleate or palmitoleate (0.25 and 0.5mM), AICAR, compound C, 4µ8C, or TUDCA. Compared to control cells, palmitate significantly decreased cell viability and increased apoptosis in a dose-dependent manner. The monounsaturated fatty acids oleate and palmitoleate completely prevented the cytotoxic effects of palmitate. Although palmitate induced markers of ER stress, chemical inhibition of ER stress did not prevent palmitate-induced lipoapoptosis. Conversely, the AMPK activator AICAR (0.1 and 0.5mM) conferred protection from palmitate mediated-alterations in viability, apoptosis and ER stress, whereas the AMPK inhibitor compound C (20 and 40µM) significantly exacerbated palmitate-mediated damage. Lastly, co-incubation with palmitate, monounsaturated fatty acids, and compound C significantly mitigated the protective effects of both oleate and palmitoleate. In conclusion, monounsaturated fatty acids confer protection against the cytotoxic effects of palmitate in vascular endothelial cells; and palmitate-mediated damage, as well as monounsaturated-mediated protection, are due in part to inactivation and activation, respectively, of the metabolic regulator AMPK. These results may have implications for understanding the deleterious effects of high saturated fat diets on cardiovascular dysfunction and disease risk.


Asunto(s)
Aminoimidazol Carboxamida/análogos & derivados , Apoptosis/efectos de los fármacos , Grasas de la Dieta/efectos adversos , Ácidos Grasos Monoinsaturados/administración & dosificación , Ácido Palmítico/efectos adversos , Ribonucleótidos/farmacología , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/metabolismo , Aminoimidazol Carboxamida/farmacología , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/prevención & control , Supervivencia Celular/efectos de los fármacos , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Ácidos Grasos Monoinsaturados/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ácido Palmítico/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología
17.
Biomed Pharmacother ; 107: 785-792, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30142540

RESUMEN

BACKGROUND: Thioredoxin-interacting protein (TXNIP) plays an important role in the development of diabetic nephropathy. In the present study, we investigated role of TXNIP on oxidative stress in glomerular mesangial cells (GMCs) cultured in high glucose or normal glucose, and explored the potential mechanism related to TXNIP as well. METHODS: Oxidative stress in GMCs under high or normal glucose was detected. TXNIP knockdown by specific siRNA or over expression by pcDNA3.0-TXNIP vector was performed to evaluate the role of TXNIP on injury of GMCs caused by oxidative stress. Activator of AMPK AICAR and AMPK inhibitor Compound C were treated the GMCs. Reactive oxygen species (ROS) and mitochondrial membrane potential were detected by flow cytometry. Activities of superoxide dismutase (SOD) and superoxide dismutase (CAT) were measured by ELISA. Activity of thioredoxin (Trx) was determined using Trx activity assay kit. mRNA expression of AMPK, TXNIP, Trx1 and Trx2 were tested by qRT-PCR. Expressions of P-AMPK, TXNIP and fibronectin proteins were detected by Western blotting. RESULTS: High glucose induced the increase of ROS level, activation of TXNIP, but restricted mitochondrial membrane potential and activities of p-AMPK, SOD and CAT, and Trx. TXNIP siRNA and AICAR inhibited high glucose-induced oxidative stress response in GMCs and fibronectin expression, but promoted cell viability. In contrast, pcDNA3.0-TXNIP and Compound C increased oxidative stress response in normal glucose cultured GMCs, but decreased cell viability. The combined effect of TXNIP siRNA and AICAR on the inhibition of oxidative stress was obviously stronger than that of single use of TXNIP siRNA. CONCLUSION: TXNIP facilitates the oxidative stress response in GMCs partially through AMPK pathway, which may provide potential therapeutic target for diabetic nephropathy treatment.


Asunto(s)
Adenilato Quinasa/metabolismo , Proteínas Portadoras/metabolismo , Células Mesangiales/metabolismo , Células Mesangiales/patología , Estrés Oxidativo , Transducción de Señal , Tiorredoxinas/metabolismo , Adenilato Quinasa/antagonistas & inhibidores , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Glucosa/toxicidad , Células Mesangiales/efectos de los fármacos , Ratones , Estrés Oxidativo/efectos de los fármacos , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , ARN Interferente Pequeño/metabolismo , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos
18.
J Biol Chem ; 293(37): 14276-14284, 2018 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-30030377

RESUMEN

The dimerization-driven paradoxical activation of RAF proto-oncogene Ser/Thr kinase (RAF) is the predominant cause of drug resistance and toxicity in cancer therapies with RAF inhibitors. The scaffold protein 14-3-3, which binds to the RAF C terminus, is essential for RAF activation under physiological conditions, but the molecular basis is unclear. Here we investigated whether and how 14-3-3 regulates the dimerization-driven transactivation of the RAF isoform CRAF by RAF inhibitors and affects drug resistance and toxicity by virtue of the dominant role of CRAF in these processes. We demonstrated that 14-3-3 enhances the dimerization-driven transactivation of CRAF by stabilizing CRAF dimers. Further, we identified AMP-activated protein kinase (AMPK) and CRAF itself as two putative kinases that redundantly phosphorylate CRAF's C terminus and thereby control its association with 14-3-3. Next, we determined whether the combinatory inhibition of AMPK and CRAF could overcome the paradoxical effect of RAF inhibitors. We found that the AMPK inhibitor (AMPKi) not only blocked the RAF inhibitor-driven paradoxical activation of ERK signaling and cellular overgrowth in Ras-mutated cancer cells by blocking phosphorylation of Ser-621 in CRAF but also reduced the formation of drug-resistant clones of BRAFV600E-mutated cancer cells. Last, we investigated whether 14-3-3 binding to the C terminus of CRAF is required for CRAF catalytic activity and observed that it was dispensable in vivo Altogether, our study unravels the molecular mechanism by which 14-3-3 regulates dimerization-driven RAF activation and identified AMPKi as a potential agent to counteract drug resistance and adverse effects of RAF inhibitors in cancer therapies.


Asunto(s)
Adenilato Quinasa/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-raf/antagonistas & inhibidores , Serina/metabolismo , Proteínas 14-3-3/metabolismo , Línea Celular Tumoral , Dimerización , Células HEK293 , Humanos , Fosforilación , Unión Proteica , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-raf/química , Proteínas Proto-Oncogénicas c-raf/metabolismo , Serina/química , Transducción de Señal
19.
Molecules ; 23(5)2018 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-29751552

RESUMEN

Multi-drug resistant microorganism infections with emerging problems that require not only a prevention strategy, but also the development of new inhibitory compounds. Six previously synthesized 5-arylidene-2-thioxothiazolidin-4-one derivatives 1a⁻f, were screened for inhibitory activity on adenylate kinases of different origins by molecular docking. The compounds 1c and 1d were the most efficient inhibitors of bacterial and some archean adenylate kinases. Hydrogen bond interactions were observed with the residues belonging to the ATP binding site. Moreover human adenylate kinases are poor targets, suggesting that this selectivity offers promising prospectives for refining the structure of our compounds.


Asunto(s)
Adenilato Quinasa/química , Antibacterianos/química , Antibacterianos/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Simulación del Acoplamiento Molecular , Tiazolidinas/química , Tiazolidinas/farmacología , Adenilato Quinasa/antagonistas & inhibidores , Secuencia de Aminoácidos , Sitios de Unión , Activación Enzimática , Humanos , Enlace de Hidrógeno , Conformación Molecular , Simulación de Dinámica Molecular , Unión Proteica , Relación Estructura-Actividad
20.
Mol Cell Endocrinol ; 476: 27-36, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-29655602

RESUMEN

C1q/tumor necrosis factor-related protein-3 (CTRP3) shows striking homologies of genomic structure to the adiponectin. In this study, we aimed to investigate the protective role of CTRP3 against sepsis-induced cardiomyopathy. Here, we overexpressed CTRP3 in myocardium by direct intramyocardial injection and constructed a model of lipopolysaccharide (LPS)-induced sepsis in mice. Our results demonstrated that cardiac-specific overexpression of CTRP3 remarkably attenuated myocardial dysfunction and increased the phosphorylation level of AMPKα during LPS-induced sepsis. The anti-inflammatory effects of CTRP3, as determined by decreased mRNA levels of TNF-α, IL-6 and a lower protein expression of phosphorylated NF-κB p65 and IκBα, was detected in mice following LPS treatment. Additionally, CTRP3 suppressed cardiac apoptosis induced by LPS in mice as indicated by terminal deoxynucleotidyl transferase nick-end labeling (TUNEL) staining and western blot for Cleaved-caspase3, Bax and Bcl-2. In conclusion, CTRP3 could protect against sepsis-induced myocardial dysfunction in mice. The cardioprotective effects of CTRP3 might be mediated by activating AMPKα signaling pathway and blunting inflammatory response and apoptosis.


Asunto(s)
Adipoquinas/metabolismo , Miocardio/metabolismo , Miocardio/patología , Sustancias Protectoras/metabolismo , Sepsis/complicaciones , Adenilato Quinasa/antagonistas & inhibidores , Adenilato Quinasa/metabolismo , Animales , Animales Recién Nacidos , Apoptosis , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Modelos Animales de Enfermedad , Activación Enzimática , Inflamación/patología , Lipopolisacáridos , Masculino , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Ratas , Transducción de Señal , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA