Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 223
Filtrar
1.
Annu Rev Biochem ; 85: 265-90, 2016 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-27294439

RESUMEN

Transcription factor IIH (TFIIH) is a multiprotein complex involved in both transcription and DNA repair, revealing a striking functional link between these two processes. Some of its subunits also belong to complexes involved in other cellular processes, such as chromosome segregation and cell cycle regulation, emphasizing the multitasking capabilities of this factor. This review aims to depict the structure of TFIIH and to dissect the roles of its subunits in different cellular mechanisms. Our understanding of the biochemistry of TFIIH has greatly benefited from studies focused on diseases related to TFIIH mutations. We address the etiology of these disorders and underline the fact that TFIIH can be considered a promising target for therapeutic strategies.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Factor de Transcripción TFIIH/genética , Transcripción Genética/efectos de los fármacos , Síndromes de Tricotiodistrofia/genética , Xerodermia Pigmentosa/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Segregación Cromosómica , ADN/genética , ADN/metabolismo , Daño del ADN , Humanos , Modelos Moleculares , Terapia Molecular Dirigida , Mutación , Fenilendiaminas/uso terapéutico , Subunidades de Proteína/antagonistas & inhibidores , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Pirimidinas/uso terapéutico , Espironolactona/uso terapéutico , Factor de Transcripción TFIIH/antagonistas & inhibidores , Factor de Transcripción TFIIH/metabolismo , Síndromes de Tricotiodistrofia/tratamiento farmacológico , Síndromes de Tricotiodistrofia/metabolismo , Síndromes de Tricotiodistrofia/patología , Xerodermia Pigmentosa/tratamiento farmacológico , Xerodermia Pigmentosa/metabolismo , Xerodermia Pigmentosa/patología
2.
Cell ; 159(5): 1126-1139, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25416950

RESUMEN

The MYC oncoproteins are thought to stimulate tumor cell growth and proliferation through amplification of gene transcription, a mechanism that has thwarted most efforts to inhibit MYC function as potential cancer therapy. Using a covalent inhibitor of cyclin-dependent kinase 7 (CDK7) to disrupt the transcription of amplified MYCN in neuroblastoma cells, we demonstrate downregulation of the oncoprotein with consequent massive suppression of MYCN-driven global transcriptional amplification. This response translated to significant tumor regression in a mouse model of high-risk neuroblastoma, without the introduction of systemic toxicity. The striking treatment selectivity of MYCN-overexpressing cells correlated with preferential downregulation of super-enhancer-associated genes, including MYCN and other known oncogenic drivers in neuroblastoma. These results indicate that CDK7 inhibition, by selectively targeting the mechanisms that promote global transcriptional amplification in tumor cells, may be useful therapy for cancers that are driven by MYC family oncoproteins.


Asunto(s)
Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Modelos Animales de Enfermedad , Neuroblastoma/tratamiento farmacológico , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Fenilendiaminas/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/uso terapéutico , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Quinasas Ciclina-Dependientes/metabolismo , Humanos , Proteína Proto-Oncogénica N-Myc , Transcripción Genética/efectos de los fármacos , Quinasa Activadora de Quinasas Ciclina-Dependientes
3.
Brain ; 146(4): 1554-1560, 2023 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-36718088

RESUMEN

Recurrent episodes of weakness in periodic paralysis are caused by intermittent loss of muscle fibre excitability, as a consequence of sustained depolarization of the resting potential. Repolarization is favoured by increasing the fibre permeability to potassium. Based on this principle, we tested the efficacy of retigabine, a potassium channel opener, to suppress the loss of force induced by a low-K+ challenge in hypokalaemic periodic paralysis (HypoPP). Retigabine can prevent the episodic loss of force in HypoPP. Knock-in mutant mouse models of HypoPP (Cacna1s p.R528H and Scn4a p.R669H) were used to determine whether pre-treatment with retigabine prevented the loss of force, or post-treatment hastened recovery of force for a low-K+ challenge in an ex vivo contraction assay. Retigabine completely prevents the loss of force induced by a 2 mM K+ challenge (protection) in our mouse models of HypoPP, with 50% inhibitory concentrations of 0.8 ± 0.13 µM and 2.2 ± 0.42 µM for NaV1.4-R669H and CaV1.1-R528H, respectively. In comparison, the effective concentration for the KATP channel opener pinacidil was 10-fold higher. Application of retigabine also reversed the loss of force (rescue) for HypoPP muscle maintained in 2 mM K+. Our findings show that retigabine, a selective agonist of the KV7 family of potassium channels, is effective for the prevention of low-K+ induced attacks of weakness and to enhance recovery from an ongoing loss of force in mouse models of type 1 (Cacna1s) and type 2 (Scn4a) HypoPP. Substantial protection from the loss of force occurred in the low micromolar range, well within the therapeutic window for retigabine.


Asunto(s)
Parálisis Periódica Hipopotasémica , Ratones , Animales , Músculo Esquelético , Carbamatos/farmacología , Carbamatos/uso terapéutico , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico
4.
Clin Sci (Lond) ; 136(1): 45-60, 2022 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-34918039

RESUMEN

Cold storage/rewarming is an inevitable process for kidney transplantation from deceased donors, which correlates closely with renal ischemia-reperfusion injury (IRI) and the occurrence of delayed graft function. Histone deacetylases (HDAC) are important epigenetic regulators, but their involvement in cold storage/rewarming injury in kidney transplantation is unclear. In the present study, we showed a dynamic change of HDAC3 in a mouse model of kidney cold storage followed by transplantation. We then demonstrated that the selective HDAC3 inhibitor RGFP966 could reduce acute tubular injury and cell death after prolonged cold storage with transplantation. RGFP966 also improved renal function, kidney repair and tubular integrity when the transplanted kidney became the sole life-supporting graft in the recipient mouse. In vitro, cold storage of proximal tubular cells followed by rewarming induced remarkable cell death, which was suppressed by RGFP966 or knockdown of HDAC3 with shRNA. Inhibition of HDAC3 decreased the mitochondrial pathway of apoptosis and preserved mitochondrial membrane potential. Collectively, HDAC3 plays a pathogenic role in cold storage/rewarming injury in kidney transplantation, and its inhibition may be a therapeutic option.


Asunto(s)
Acrilamidas/uso terapéutico , Histona Desacetilasas/efectos de los fármacos , Trasplante de Riñón , Fenilendiaminas/uso terapéutico , Daño por Reperfusión/prevención & control , Aloinjertos , Animales , Apoptosis , Frío , Técnicas de Silenciamiento del Gen , Histona Desacetilasas/genética , Túbulos Renales Proximales/patología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones Endogámicos C57BL , Preservación de Órganos/efectos adversos , ARN Interferente Pequeño
5.
Oral Dis ; 28(3): 611-620, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33503275

RESUMEN

BACKGROUND: Cyclin-dependent kinase 7 (CDK7) has been critically linked to human cancer. However, the roles of CDK7 in head and neck squamous cell carcinoma (HNSCC) remain incompletely known. Here, we sought to dissect the functions of CDK7 underlying HNSCC tumorigenesis and explore whether pharmacological inhibition of CDK7 could induce anti-cancer effects. METHODS: CDK7 expression was measured in a panel of HNSCC cell lines with p53 mutation and 20 pairs of HNSCC samples and adjacent non-tumor tissues. Genetic targeting and pharmacological inhibition of CDK7 were conducted to dissect the biological roles of CDK7 in p53-mutated HNSCC cells. An HNSCC xenograft model was developed to determine the therapeutic effects of THZ1 in vivo. Potential genes and pathways responsible for therapeutic effects of THZ1 were identified by genome-wide RNA-sequencing and bioinformatics interrogations. RESULTS: CDK7 expression was significantly elevated in cancerous cells and samples as compared with their adjacent non-tumor counterparts. Impaired cell proliferation, migration, and invasion as well increased apoptosis were observed in cells upon CDK7 knockdown or THZ1 exposure. THZ1 administration potently inhibited tumor overgrowth in vivo. Mechanistically, hundreds of genes enriched in cell proliferation, apoptosis, and cancer-related categories were identified to be potentially mediated the therapeutic effects of THZ1 in HNSCC. CONCLUSION: Our findings reveal that CDK7 might serve as a novel putative pro-oncogenic gene underlying HNSCC tumorigenesis and therapeutic targeting of CDK7 might be a promising strategy for p53-mutated HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello , Proteína p53 Supresora de Tumor , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Quinasas Ciclina-Dependientes/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/genética , Humanos , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Fosfotransferasas (Aceptor de Grupo Alcohol) , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Proteína p53 Supresora de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Activadora de Quinasas Ciclina-Dependientes
6.
Proc Natl Acad Sci U S A ; 116(47): 23426-23436, 2019 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-31685616

RESUMEN

As a central feature of neuroinflammation, microglial dysfunction has been increasingly considered a causative factor of neurodegeneration implicating an intertwined pathology with amyloidogenic proteins. Herein, we report the smallest synthetic molecule (N,N'-diacetyl-p-phenylenediamine [DAPPD]), simply composed of a benzene ring with 2 acetamide groups at the para position, known to date as a chemical reagent that is able to promote the phagocytic aptitude of microglia and subsequently ameliorate cognitive defects. Based on our mechanistic investigations in vitro and in vivo, 1) the capability of DAPPD to restore microglial phagocytosis is responsible for diminishing the accumulation of amyloid-ß (Aß) species and significantly improving cognitive function in the brains of 2 types of Alzheimer's disease (AD) transgenic mice, and 2) the rectification of microglial function by DAPPD is a result of its ability to suppress the expression of NLRP3 inflammasome-associated proteins through its impact on the NF-κB pathway. Overall, our in vitro and in vivo investigations on efficacies and molecular-level mechanisms demonstrate the ability of DAPPD to regulate microglial function, suppress neuroinflammation, foster cerebral Aß clearance, and attenuate cognitive deficits in AD transgenic mouse models. Discovery of such antineuroinflammatory compounds signifies the potential in discovering effective therapeutic molecules against AD-associated neurodegeneration.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antiinflamatorios/farmacología , Cognición/efectos de los fármacos , Microglía/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fagocitosis/efectos de los fármacos , Fenilendiaminas/farmacología , Enfermedad de Alzheimer/psicología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Antiinflamatorios/uso terapéutico , Evaluación Preclínica de Medicamentos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamasomas/efectos de los fármacos , Inflamasomas/genética , Aprendizaje por Laberinto , Ratones , Ratones Transgénicos , Microglía/fisiología , Estructura Molecular , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Fármacos Neuroprotectores/uso terapéutico , Fragmentos de Péptidos/genética , Fenilendiaminas/química , Fenilendiaminas/uso terapéutico , Presenilina-1/genética , Memoria Espacial/efectos de los fármacos
7.
Proc Natl Acad Sci U S A ; 116(26): 12986-12995, 2019 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-31182587

RESUMEN

The aberrant hedgehog (Hh) pathway plays important roles in multiple cancer types, therefore serving as a promising drug target. Current clinically available hedgehog-targeted drugs act mostly by antagonizing the upstream component smoothened; however, both primary and acquired resistance to FDA-approved smoothened inhibitor (SMOi) drugs have been described. We have recently demonstrated that the BET inhibitor effectively suppresses SMOi-resistant Hh-driven cancers through antagonizing transcription of GLI1 and GLI2, the core transcriptional factors of Hh pathway, suggesting epigenetic or transcriptional targeted therapy represents an anti-Hh therapeutic strategy that can overcome SMOi resistance. Here we performed an unbiased screening of epigenetic or transcriptional targeted small molecules to test their inhibitory effects on GLI1 and GLI2 transcription or cell viability of Hh-driven tumor lines. THZ1, a covalent inhibitor of cyclin-dependent kinase 7 (CDK7), is identified as the top hit in our screening. We then confirmed that antagonizing CDK7 by either small-molecule inhibitors or the CRISPR-Cas9 approach causes substantial suppression of GLI1 and GLI2 transcription, resulting in effective inhibition of Hh-driven cancers in vitro and in vivo. More importantly, antagonizing CDK7 retains inhibitory activity against Hh-driven cancers with almost all so-far described primary or acquired SMOi resistance. Furthermore, we reveal a synergy between CDK7 inhibition and BET inhibition on antagonizing aberrant Hh pathway and Hh-driven cancers that are either responsive or resistant to SMOi. Our results illustrate transcriptional inhibition through targeting CDK7 as a promising therapeutic strategy for treating Hh-driven cancers, especially those with primary or acquired resistance to SMOi drugs.


Asunto(s)
Antineoplásicos/farmacología , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Resistencia a Antineoplásicos/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Fenilendiaminas/farmacología , Pirimidinas/farmacología , Receptor Smoothened/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral/trasplante , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Quinasas Ciclina-Dependientes/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Ensayos de Selección de Medicamentos Antitumorales , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Células 3T3 NIH , Neoplasias/genética , Proteínas Nucleares/genética , Fenilendiaminas/uso terapéutico , Cultivo Primario de Células , Pirimidinas/uso terapéutico , RNA-Seq , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transcripción Genética/efectos de los fármacos , Proteína con Dedos de Zinc GLI1/genética , Proteína Gli2 con Dedos de Zinc/genética , Quinasa Activadora de Quinasas Ciclina-Dependientes
8.
J Am Soc Nephrol ; 32(11): 2759-2776, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34716241

RESUMEN

BACKGROUND: Autosomal dominant polycystic kidney disease (ADPKD), the most common inherited kidney disease, is regulated by different forms of cell death, including apoptosis and autophagy. However, the role in ADPKD of ferroptosis, a recently discovered form of cell death mediated by iron and lipid metabolism, remains elusive. METHODS: To determine a pathophysiologic role of ferroptosis in ADPKD, we investigated whether the absence of Pkd1 (encoding polycystin-1) affected the expression of key factors involved in the process of ferroptosis, using Western blot and qRT-PCR analysis in Pkd1 mutant renal cells and tissues. We also examined whether treatment with erastin, a ferroptosis inducer, and ferrostain-1, a ferroptosis inhibitor, affected cyst growth in Pkd1 mutant mouse models. RESULTS: We found that kidney cells and tissues lacking Pkd1 exhibit extensive metabolic abnormalities, including reduced expression of the system Xc- amino acid antiporter (critical for import of cystine), of iron exporter (ferroportin), and of GPX4 (a key and negative regulator of ferroptosis). The abnormalities also include increased expression of iron importers (TfR1, DMT1) and HO-1, which in turn result in high iron levels, low GSH and GPX4 activity, increased lipid peroxidation, and propensity to ferroptosis. We further found that erastin increased, and ferrostatin-1 inhibited ferroptotic cell death and proliferation of Pkd1-deficient cells in kidneys from Pkd1 mutant mice. A lipid peroxidation product increased in Pkd1-deficient cells, 4HNE, promoted the proliferation of survived Pkd1 mutant cells via activation of Akt, S6, Stat3, and Rb during the ferroptotic process, contributing to cyst growth. CONCLUSION: These findings indicate that ferroptosis contributes to ADPKD progression and management of ferroptosis may be a novel strategy for ADPKD treatment.


Asunto(s)
Ferroptosis , Riñón Poliquístico Autosómico Dominante/patología , Animales , Ciclo Celular , Células Cultivadas , Ciclohexilaminas/farmacología , Ciclohexilaminas/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Hierro/metabolismo , Peroxidación de Lípido , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Interferencia de ARN , Esferoides Celulares , Canales Catiónicos TRPP/deficiencia , Canales Catiónicos TRPP/genética , Transcriptoma
9.
Artículo en Inglés | MEDLINE | ID: mdl-33139276

RESUMEN

In the treatment of hookworm infections, pharmacotherapy has been only moderately successful and drug resistance is a threat. Therefore, novel treatment options including combination therapies should be considered, in which tribendimidine could play a role. Our aims were to (i) characterize the pharmacokinetics of tribendimidine's metabolites in adolescents receiving tribendimidine monotherapy or in combination with ivermectin or oxantel pamoate, (ii) evaluate possible drug-drug interactions (DDI), (iii) link exposure to response, and (iv) identify a treatment strategy associated with high efficacy, i.e., >90% cure rates (CRs), utilizing model-based simulations. A population pharmacokinetic model was developed for tribendimidine's primary and secondary metabolites, dADT and adADT, in 54 hookworm-positive adolescents, with combination therapy evaluated as a possible covariate. Subsequently, an exposure-response analysis was performed utilizing CRs as response markers. Simulations were performed to identify a treatment strategy to achieve >90% CRs. A two-compartmental model best described metabolite disposition. No pharmacokinetic DDI was identified with ivermectin or oxantel pamoate. All participants receiving tribendimidine plus ivermectin were cured. For the monotherapy arm and the arm including the combination with oxantel pamoate, Emax models adequately described the correlation between dADT exposure and probability of being cured, with required exposures to achieve 50% of maximum effect of 39.6 and 15.6 nmol/ml·h, respectively. Based on our simulations, an unrealistically high monotherapy tribendimidine dose would be necessary to achieve CRs of >90%, while combination therapy with ivermectin would meet this desired target product profile. Further clinical studies should be launched to develop this combination for the treatment of hookworm and other helminth infections.


Asunto(s)
Antihelmínticos , Infecciones por Uncinaria , Adolescente , Ancylostomatoidea , Animales , Antihelmínticos/uso terapéutico , Infecciones por Uncinaria/tratamiento farmacológico , Humanos , Fenilendiaminas/uso terapéutico
10.
Biochem Biophys Res Commun ; 548: 27-34, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-33631670

RESUMEN

Degenerative retinal diseases, including age-related macular degeneration, are serious diseases that may lead to irreversible retinal neuron damage and permanent vision impairment. There are currently no effective treatments for these diseases due to our incomplete understanding of the underlying pathological mechanisms. Ferroptosis, a newly identified iron-dependent mode of cell death, is implicated in various diseases. However, it is unknown whether ferroptosis is involved in light-induced retinal degeneration. In this study, we found that light exposure significantly reduced the viability of photoreceptor cells in vitro and induced pro-ferroptotic changes, including iron accumulation, mitochondrial shrinkage, glutathione depletion, increased malondialdehyde (MDA), and decreased protein expression of SLC7A11 and GPX4. The effects of light exposure on ferroptosis were attenuated by ferrostatin-1. Consistently, the results of in vivo studies demonstrated that ferrostatin-1 protected against light-induced ferroptosis. And it exerted therapeutic effects by inhibiting neuroinflammation and prevented the effects of light exposure on the structure and function of the retina. The findings reveal an important role of ferroptosis in the pathogenesis of light-induced retinal degeneration and suggest that ferroptosis may be a novel treatment target for preventing retinal degeneration.


Asunto(s)
Ciclohexilaminas/uso terapéutico , Ferroptosis/efectos de los fármacos , Luz , Fármacos Neuroprotectores/uso terapéutico , Fenilendiaminas/uso terapéutico , Retina/patología , Degeneración Retiniana/tratamiento farmacológico , Animales , Línea Celular , Ciclohexilaminas/farmacología , Masculino , Fármacos Neuroprotectores/farmacología , Fenilendiaminas/farmacología , Ratas Sprague-Dawley , Retina/efectos de los fármacos , Retina/ultraestructura , Degeneración Retiniana/patología
11.
Mol Psychiatry ; 25(6): 1323-1333, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-30385872

RESUMEN

Major depressive disorder (MDD) is a leading cause of disability worldwide, yet current treatment strategies remain limited in their mechanistic diversity. Recent evidence has highlighted a promising novel pharmaceutical target-the KCNQ-type potassium channel-for the treatment of depressive disorders, which may exert a therapeutic effect via functional changes within the brain reward system, including the ventral striatum. The current study assessed the effects of the KCNQ channel opener ezogabine (also known as retigabine) on reward circuitry and clinical symptoms in patients with MDD. Eighteen medication-free individuals with MDD currently in a major depressive episode were enrolled in an open-label study and received ezogabine up to 900 mg/day orally over the course of 10 weeks. Resting-state functional magnetic resonance imaging data were collected at baseline and posttreatment to examine brain reward circuitry. Reward learning was measured using a computerized probabilistic reward task. After treatment with ezogabine, subjects exhibited a significant reduction of depressive symptoms (Montgomery-Asberg Depression Rating Scale score change: -13.7 ± 9.7, p < 0.001, d = 2.08) and anhedonic symptoms (Snaith-Hamilton Pleasure Scale score change: -6.1 ± 5.3, p < 0.001, d = 1.00), which remained significant even after controlling for overall depression severity. Improvement in depression was associated with decreased functional connectivity between the ventral caudate and clusters within the mid-cingulate cortex and posterior cingulate cortex (n = 14, voxel-wise p < 0.005). In addition, a subgroup of patients tested with a probabilistic reward task (n = 9) showed increased reward learning following treatment. These findings highlight the KCNQ-type potassium channel as a promising target for future drug discovery efforts in mood disorders.


Asunto(s)
Carbamatos/farmacología , Carbamatos/uso terapéutico , Trastorno Depresivo Mayor/tratamiento farmacológico , Activación del Canal Iónico/efectos de los fármacos , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Estriado Ventral/efectos de los fármacos , Trastorno Depresivo Mayor/metabolismo , Femenino , Humanos , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/metabolismo , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Recompensa , Estriado Ventral/metabolismo
12.
Biol Pharm Bull ; 44(2): 169-180, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33239494

RESUMEN

Chronic stress-induced brain injury (CSBI) is the organic damage of brain tissue caused by long-term psychological and environmental stress. However, there is no effective drug for the treatment of CSBI. The present study aimed to investigate possible mechanisms of CSBI and to explore related therapeutic targets. A rat model of CSBI was established by combining chronic restraint and cold water immersion. Our CSBI model was validated via Nissl staining, Western blotting, and behavioral tests. RNA sequencing (RNA-seq) was used to identify differentially expressed genes (DEGs) within brain tissue during CSBI. Both Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) analyses were performed to determine signaling pathways associated with CSBI-induced DEGs. Agonists/antagonists were used to validate the pharmacodynamics of potential therapeutic targets. A combination of chronic restraint and cold water immersion successfully induced a rat model of CSBI, as indicated by various markers of brain injury and cell apoptosis that were verified via Nissl staining, Western blotting, and behavioral tests. RNA-seq analysis identified 1131 DEGs in CSBI rats. Of these DEGs, 553 genes were up-regulated and 778 genes were down-regulated. GO and KEGG pathway analyses revealed that significant DEGs were predominantly related to membrane-bound ion channels, among which the potassium channel function was found to be significantly affected. Pharmacological experiments revealed that retigabine, a voltage-gated potassium channel opener, demonstrated a protective effect in CSBI rats. Taken together, our findings suggest that potassium channel function is disrupted in CSBI, and that potassium channel regulators may function as anti-CSBI drugs.


Asunto(s)
Lesiones Encefálicas/etiología , Fármacos Neuroprotectores/farmacología , Canales de Potasio/metabolismo , Estrés Psicológico/complicaciones , Animales , Encéfalo/efectos de los fármacos , Encéfalo/patología , Lesiones Encefálicas/patología , Lesiones Encefálicas/prevención & control , Carbamatos/farmacología , Carbamatos/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Fármacos Neuroprotectores/uso terapéutico , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/agonistas , Canales de Potasio/genética , RNA-Seq , Ratas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
13.
J Pharmacol Exp Ther ; 373(1): 72-80, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31969383

RESUMEN

Spinal cord injury (SCI) usually leads to acute neuronal death and delayed secondary degeneration, resulting in sensory dysfunction, paralysis, and chronic pain. Excessive excitation is one of the critical factors leading to secondary neural damage initiated by various insults. KCNQ/Kv7 channels are highly expressed in spinal neurons and axons and play an important role in controlling their excitability. Enhancing KCNQ channel activity by using its specific opener retigabine could thus be a plausible treatment strategy to reduce the pathology after SCI. We produced contusive SCI at T10 in adult male rats, which then received 10 consecutive days' treatment with retigabine or vehicle starting 3 hours or 3 days after contusion. Two different concentrations and two different delivery methods were applied. Delivery of retigabine via Alzet osmotic pumps, but not intraperitoneal injections 3 hours after contusion, promoted recovery of locomotor function. Remarkably, retigabine delivery in both methods significantly attenuated the development of mechanical stimuli-induced hyperreflexia and spontaneous pain; however, no significant difference in the thermal threshold was observed. Although retigabine delivered 3 days after contusion significantly attenuated the development of mechanical hypersensitivity and spontaneous pain, the locomotor function is not improved by the delayed treatments. Finally, we found that early application of retigabine attenuates the inflammatory activity in the spinal cord and increases the survival of white matter after SCI. Our results suggest that decreasing neuronal excitability by targeting KCNQ/Kv7 channels at acute stage aids the recovery of locomotor function and attenuates the development of neuropathic pain after SCI. SIGNIFICANCE STATEMENT: Several pharmacological interventions have been proposed for spinal cord injury (SCI) treatment, but none have been shown to be both effective and safe in clinical trials. Necrotic neuronal death and chronic pain are often the cost of pathological neural excitation after SCI. We show that early, brief application of retigabine could aid locomotor and sensory neurobehavioral recovery after SCI, supporting the use of this drug in the clinic to promote motor and sensory function in patients with SCI.


Asunto(s)
Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/metabolismo , Locomoción/fisiología , Recuperación de la Función/fisiología , Traumatismos de la Médula Espinal/metabolismo , Animales , Carbamatos/farmacología , Carbamatos/uso terapéutico , Locomoción/efectos de los fármacos , Masculino , Moduladores del Transporte de Membrana/farmacología , Moduladores del Transporte de Membrana/uso terapéutico , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Recuperación de la Función/efectos de los fármacos , Traumatismos de la Médula Espinal/tratamiento farmacológico , Vértebras Torácicas/lesiones
14.
J Autoimmun ; 113: 102515, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32703754

RESUMEN

The nucleotide-binding oligomerization domain (NOD)-like receptor containing pyrin domain 3 (NLRP3) inflammasome is a high-molecular-weight complex mediated by the activation of pattern-recognition receptors (PRRs) seed in innate immunity. Once NLRP3 is activated, the following recruitment of the adapter apoptosis-associated speck-like protein containing a caspase recruitment domain (CARD) (ASC) and procaspase-1 would be initiated. Cleavage of procaspase-1 into active caspase-1 then leads to the maturation of the precursor forms of interleukin (IL)-1ß and IL-18 into biologically active IL-1ß and IL-18. The activation of NLRP3 inflammasome is thought to be tightly associated with a regulator never in mitosis A (NIMA)-related kinase 7 (NEK7), apart from other signaling events such as K+ efflux and reactive oxygen species (ROS). Plus, the NLRP3 inflammasome has been linked to various metabolic disorders, chronic inflammation and other diseases. In this review, we firstly describe the cellular/molecular mechanisms of the NEK7-licensed NLRP3 inflammasome activation. Then we detail the potential inhibitors that can selectively and effectively modulate either the NEK7-NLRP3 complex itself or the related molecular/cellular events. Finally, we describe some inhibitors as promising therapeutic strategies for diverse diseases driven by NLRP3 inflammasome.


Asunto(s)
Inflamasomas/inmunología , Inflamación/inmunología , Enfermedades Metabólicas/inmunología , Quinasas Relacionadas con NIMA/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Quinasa de Linfoma Anaplásico/metabolismo , Animales , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Enfermedad Crónica/tratamiento farmacológico , Modelos Animales de Enfermedad , Diterpenos de Tipo Kaurano/farmacología , Diterpenos de Tipo Kaurano/uso terapéutico , Humanos , Inflamasomas/antagonistas & inhibidores , Inflamasomas/metabolismo , Inflamación/tratamiento farmacológico , Enfermedades Metabólicas/tratamiento farmacológico , Ratones , Quinasas Relacionadas con NIMA/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Fenilendiaminas/farmacología , Fenilendiaminas/uso terapéutico , Unión Proteica/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
15.
Toxicol Appl Pharmacol ; 406: 115189, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32800772

RESUMEN

The potential therapeutic effect of histone deacetylase 3 (HDAC3) pharmacologic inhibition on diabetes has been focused recently. RGFP966, as a highly-selective HDAC3 inhibitor, its possible roles and underlying mechanism in the treatment of diabetes needs to be clarified. In this study, low-dose streptozotocin (STZ)-induced pre-diabetic mice were used to test the regulatory ability of RGFP966 in blood glucose and insulin. We isolated the islets both from normal C57BL/6 J mice and KKAy mice with spontaneous type 2 diabetes to determine the potency of RGFP966 on glucose-stimulated insulin secretion. NIT-1 pancreatic ß-cells induced by sodium palmitate (PA) were applied to identify the protective effects of RGFP966 against ß-cell apoptosis. The results showed that administration of RGFP966 in the pre-diabetic mice not only significantly reduced hyperglycemia, promoted phase I insulin secretion, improved morphology of islets, but also increased glucose infusion rate (GIR) during hyperglycemic clamp test. When treated in vitro, RGFP966 enhanced insulin secretion and synthesis in islets of normal C57BL/6J mice and diabetic KKAy mice. In addition, it partially attenuated PA-induced apoptosis in NIT-1 cells. Therefore, our research suggests that RGFP966, probably through selective inhibition of HDAC3, might serve as a novel potential preventive and therapeutic candidate for diabetes.


Asunto(s)
Acrilamidas/uso terapéutico , Diabetes Mellitus Experimental/tratamiento farmacológico , Intolerancia a la Glucosa/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/metabolismo , Hipoglucemiantes/uso terapéutico , Fenilendiaminas/uso terapéutico , Acrilamidas/farmacología , Animales , Línea Celular , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Expresión Génica/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Hipoglucemiantes/farmacología , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Fenilendiaminas/farmacología
16.
Pharmacol Res ; 160: 105200, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32942014

RESUMEN

De novo variants in KCNQ2 encoding for Kv7.2 voltage-dependent neuronal potassium (K+) channel subunits are associated with developmental epileptic encephalopathy (DEE). We herein describe the clinical and electroencephalographic (EEG) features of a child with early-onset DEE caused by the novel KCNQ2 p.G310S variant. In vitro experiments demonstrated that the mutation induces loss-of-function effects on the currents produced by channels incorporating mutant subunits; these effects were counteracted by the selective Kv7 opener retigabine and by gabapentin, a recently described Kv7 activator. Given these data, the patient started treatment with gabapentin, showing a rapid and sustained clinical and EEG improvement over the following months. Overall, these results suggest that gabapentin can be regarded as a precision therapy for DEEs due to KCNQ2 loss-of-function mutations.


Asunto(s)
Anticonvulsivantes/uso terapéutico , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Gabapentina/uso terapéutico , Canal de Potasio KCNQ2/genética , Edad de Inicio , Animales , Células CHO , Carbamatos/uso terapéutico , Células Cultivadas , Niño , Cricetinae , Cricetulus , Electroencefalografía , Femenino , Humanos , Mutación , Fenilendiaminas/uso terapéutico , Medicina de Precisión , Ratas , Resultado del Tratamiento
17.
Epilepsy Behav ; 103(Pt A): 106670, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31864943

RESUMEN

Evidence indicates that ferrostain-1 (Fer-1), a specific inhibitor of ferroptosis, could ameliorate cognitive dysfunction of rats with kainic acid (KA)-induced temporal lobe epilepsy (TLE) by suppressing ferroptosis processes. Recent studies suggest that P38 mitogen-activated protein kinase (MAPK) pathway could be mediated by ferroptosis processes. The activation of P38 MAPK results in cognitive impairment by suppressing the expression of synaptic plasticity-related proteins. However, it is unclear whether Fer-1 can mitigate cognitive impairment of rats with KA-induced TLE by inhibiting P38 MAPK activation. In the present study, treatment with Fer-1 blocked the activation of P38 MAPK, which resulted in an increased expression of synaptophysin (SYP) and postsynaptic density protein 95 (PSD-95) in the hippocampus of rats with KA-induced TLE, hence, ameliorating their cognitive impairment. Also, P38 MAPK activation in the hippocampus of the rats reduced the expression of both PSD-95 and SYP proteins. Treatment of the rats with SB203580, a P38 MAPK-specific inhibitor, prevented the activation of P38 MAPK, which resulted in an increase in SYP and PSD95 protein levels in the hippocampus. These results suggest that Fer-1 could mitigate the cognitive impairment by suppressing P38 MAPK activation thus restoring the expression of synaptic proteins. Ferroptosis processes might be involved in suppressing synaptic protein expression.


Asunto(s)
Disfunción Cognitiva/prevención & control , Ciclohexilaminas/uso terapéutico , Epilepsia del Lóbulo Temporal/tratamiento farmacológico , Fármacos Neuroprotectores/uso terapéutico , Fenilendiaminas/uso terapéutico , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Biomarcadores/metabolismo , Disfunción Cognitiva/metabolismo , Ciclohexilaminas/farmacología , Epilepsia del Lóbulo Temporal/complicaciones , Epilepsia del Lóbulo Temporal/metabolismo , Epilepsia del Lóbulo Temporal/psicología , Ferroptosis/efectos de los fármacos , Ferroptosis/fisiología , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Fármacos Neuroprotectores/farmacología , Fenilendiaminas/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Cell Mol Biol Lett ; 25: 10, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32161620

RESUMEN

BACKGROUND: Ferroptosis is a newly recognized type of cell death, which is different from traditional necrosis, apoptosis or autophagic cell death. However, the position of ferroptosis in lipopolysaccharide (LPS)-induced acute lung injury (ALI) has not been explored intensively so far. In this study, we mainly analyzed the relationship between ferroptosis and LPS-induced ALI. METHODS: In this study, a human bronchial epithelial cell line, BEAS-2B, was treated with LPS and ferrostatin-1 (Fer-1, ferroptosis inhibitor). The cell viability was measured using CCK-8. Additionally, the levels of malondialdehyde (MDA), 4-hydroxynonenal (4-HNE), and iron, as well as the protein level of SLC7A11 and GPX4, were measured in different groups. To further confirm the in vitro results, an ALI model was induced by LPS in mice, and the therapeutic action of Fer-1 and ferroptosis level in lung tissues were evaluated. RESULTS: The cell viability of BEAS-2B was down-regulated by LPS treatment, together with the ferroptosis markers SLC7A11 and GPX4, while the levels of MDA, 4-HNE and total iron were increased by LPS treatment in a dose-dependent manner, which could be rescued by Fer-1. The results of the in vivo experiment also indicated that Fer-1 exerted therapeutic action against LPS-induced ALI, and down-regulated the ferroptosis level in lung tissues. CONCLUSIONS: Our study indicated that ferroptosis has an important role in the progression of LPS-induced ALI, and ferroptosis may become a novel target in the treatment of ALI patients.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Ciclohexilaminas/uso terapéutico , Ferroptosis/efectos de los fármacos , Fenilendiaminas/uso terapéutico , Lesión Pulmonar Aguda/tratamiento farmacológico , Lesión Pulmonar Aguda/patología , Aldehídos/metabolismo , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclohexilaminas/farmacología , Ferroptosis/inmunología , Humanos , Hierro/metabolismo , Lipopolisacáridos/farmacología , Masculino , Malondialdehído/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenilendiaminas/farmacología , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo
19.
Pharmacology ; 105(7-8): 471-476, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32062659

RESUMEN

INTRODUCTION: Gout arthritis is an inflammatory disease characterized by severe acute pain. The goal of pharmacological gout arthritis treatments is to reduce pain, and thereby increase the patient's quality of life. The Kv7/M channel activators retigabine and flupirtine show analgesic efficacy in animal models of osteoarthritic pain. We hypothesized that these drugs may also alleviate gout arthritis pain. OBJECTIVE: To determine the effects of retigabine and flupirtine on pain behavior associated with monosodium urate (MSU)-induced gout arthritis. METHODS: The gout arthritis model was established with an intra-articular injection of MSU into the right ankle joint, animals were treated with retigabine or flupirtine, and pain-related behaviors were assessed. RESULTS: Retigabine and flupirtine significantly increased the mechanical threshold and prolonged the paw withdrawal latency in a rat model of gout arthritis pain in a dose-dependent manner. The antinociceptive effects of retigabine and flupirtine were fully antagonized by the Kv7/M channel blocker XE991. CONCLUSION: Retigabine and flupirtine showed antinociceptive effects for MSU-induced gout pain at different times during pain development.


Asunto(s)
Aminopiridinas/farmacología , Analgésicos/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Gotosa/tratamiento farmacológico , Carbamatos/farmacología , Dolor/tratamiento farmacológico , Fenilendiaminas/farmacología , Aminopiridinas/uso terapéutico , Analgésicos/uso terapéutico , Animales , Artritis Experimental/inducido químicamente , Artritis Gotosa/inducido químicamente , Conducta Animal/efectos de los fármacos , Carbamatos/uso terapéutico , Modelos Animales de Enfermedad , Hiperalgesia/tratamiento farmacológico , Canales de Potasio KCNQ/agonistas , Canales de Potasio KCNQ/efectos de los fármacos , Masculino , Dolor/inducido químicamente , Fenilendiaminas/uso terapéutico , Ratas , Ratas Sprague-Dawley , Ácido Úrico/toxicidad
20.
Artículo en Inglés | MEDLINE | ID: mdl-30718244

RESUMEN

Opisthorchiasis, caused by the foodborne trematode Opisthorchis viverrini, affects more than 8 million people in Southeast Asia. In the framework of a phase 2b clinical trial conducted in Lao People's Democratic Republic, pharmacokinetic samples were obtained from 125 adult and adolescent O. viverrini-infected patients treated with 400 mg tribendimidine following the design of a sparse sampling scheme at 20 min and 2, 7.75, 8, and 30 h after treatment using dried blood spot sampling. Pharmacokinetic data for the metabolites deacetylated amidantel (dADT) and acetylated dADT (adADT) were pooled with data from two previous ascending-dose trials and evaluated using nonlinear mixed-effects modeling. The observed pharmacokinetic data were described using a flexible transit absorption model for the active metabolite dADT, followed by one-compartment disposition models for both metabolites. Significant covariates were age, body weight, formulation, and breaking of the enteric coating on the tablets. There were significant associations between O. viverrini cure and both the dADT maximum concentration and the area under the concentration-time curve (P < 0.001), with younger age being associated with a higher probability of cure. Modeling and simulation of exposures in patients with different weight and age combinations showed that an oral single dose of 400 mg tribendimidine attained therapeutic success in over 90% of adult patients. Our data confirmed that tribendimidine could be a valuable novel alternative to the standard treatment, praziquantel, for the treatment of O. viverrini infections.


Asunto(s)
Opistorquiasis/tratamiento farmacológico , Fenilendiaminas/farmacocinética , Adolescente , Adulto , Anciano , Animales , Antiplatelmínticos/sangre , Antiplatelmínticos/farmacocinética , Antiplatelmínticos/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Opisthorchis , Fenilendiaminas/sangre , Fenilendiaminas/uso terapéutico , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA