Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Brain Behav Immun ; 111: 365-375, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37196785

RESUMEN

Microglia is a heterogeneous population that mediates neuroinflammation in the central nervous system (CNS) and plays a crucial role in developing neuropathic pain. FKBP5 facilitates the assembly of the IκB kinase (IKK) complex for the activation of NF-κB, which arises as a novel target for treating neuropathic pain. In this study, cannabidiol (CBD), a main active component of Cannabis, was identified as an antagonist of FKBP5. In vitro protein intrinsic fluorescence titration showed that CBD directly bound to FKBP5. Cellular thermal shift assay (CETSA) indicated that CBD binding increased the FKBP5 stability, which implies that FKBP5 is the endogenous target of CBD. CBD was found to inhibit the assembly of the IKK complex and the activation of NF-κB, therefore blocking LPS-induced NF-κB downstream pro-inflammatory factors NO, IL-1ß, IL-6 and TNF-α. Stern-Volmer analysis and protein thermal shift assay revealed that tyrosine 113 (Y113) of FKBP5 was critical for FKBP5 interacting with CBD, which is consistent with in silico molecular docking simulation. FKBP5 Y113 mutation (Y113A) alleviated the effect of CBD inhibiting LPS-induced pro-inflammatory factors overproduction. Furthermore, systemic administration of CBD inhibited chronic constriction injury (CCI)-induced microglia activation and FKBP5 overexpression in lumbar spinal cord dorsal horn. These data imply that FKBP5 is an endogenous target of CBD.


Asunto(s)
Cannabidiol , Neuralgia , Proteínas de Unión a Tacrolimus , Animales , Ratas , Cannabidiol/farmacología , Lipopolisacáridos/farmacología , Simulación del Acoplamiento Molecular , Neuralgia/tratamiento farmacológico , Neuralgia/metabolismo , Enfermedades Neuroinflamatorias , FN-kappa B/metabolismo , Ratas Sprague-Dawley , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores
2.
Mol Psychiatry ; 27(12): 4928-4938, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36104438

RESUMEN

Stress-related psychiatric disorders such as depression are among the leading causes of morbidity and mortality. Considering that many individuals fail to respond to currently available antidepressant drugs, there is a need for antidepressants with novel mechanisms. Polymorphisms in the gene encoding FK506-binding protein 51 (FKBP51), a co-chaperone of the glucocorticoid receptor, have been linked to susceptibility to stress-related psychiatric disorders. Whether this protein can be targeted for their treatment remains largely unexplored. The aim of this work was to investigate whether inhibition of FKBP51 with SAFit2, a novel selective inhibitor, promotes hippocampal neuron outgrowth and neurogenesis in vitro and stress resilience in vivo in a mouse model of chronic psychosocial stress. Primary hippocampal neuronal cultures or hippocampal neural progenitor cells (NPCs) were treated with SAFit2 and neuronal differentiation and cell proliferation were analyzed. Male C57BL/6 mice were administered SAFit2 while concurrently undergoing a chronic stress paradigm comprising of intermittent social defeat and overcrowding, and anxiety and depressive -related behaviors were evaluated. SAFit2 increased neurite outgrowth and number of branch points to a greater extent than brain derived neurotrophic factor (BDNF) in primary hippocampal neuronal cultures. SAFit2 increased hippocampal NPC neurogenesis and increased neurite complexity and length of these differentiated neurons. In vivo, chronic SAFit2 administration prevented stress-induced social avoidance, decreased anxiety in the novelty-induced hypophagia test, and prevented stress-induced anxiety in the open field but did not alter adult hippocampal neurogenesis in stressed animals. These data warrant further exploration of inhibition of FKBP51 as a strategy to treat stress-related disorders.


Asunto(s)
Hipocampo , Resiliencia Psicológica , Estrés Psicológico , Proteínas de Unión a Tacrolimus , Animales , Masculino , Ratones , Antidepresivos/farmacología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Depresión/tratamiento farmacológico , Hipocampo/metabolismo , Ratones Endogámicos C57BL , Neurogénesis/efectos de los fármacos , Resiliencia Psicológica/efectos de los fármacos , Estrés Psicológico/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/metabolismo
3.
Cell Mol Life Sci ; 79(3): 175, 2022 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-35244772

RESUMEN

FK506-binding protein 51 (encoded by Fkpb51, also known as Fkbp5) has been associated with stress-related mental illness. To investigate its function, we studied the morphological consequences of Fkbp51 deletion. Artificial Intelligence-assisted morphological analysis revealed that male Fkbp51 knock-out (KO) mice possess more elongated dentate gyrus (DG) but shorter hippocampal height in coronal sections when compared to WT. Primary cultured Fkbp51 KO hippocampal neurons were shown to exhibit larger dendritic outgrowth than wild-type (WT) controls and pharmacological manipulation experiments suggest that this may occur through the regulation of microtubule-associated protein. Both in vitro primary culture and in vivo labeling support a role for FKBP51 in the regulation of microtubule-associated protein expression. Furthermore, Fkbp51 KO hippocampi exhibited decreases in ßIII-tubulin, MAP2, and Tau protein levels, but a greater than 2.5-fold increase in Parkin protein. Overexpression and knock-down FKBP51 demonstrated that FKBP51 negatively regulates Parkin in a dose-dependent and ubiquitin-mediated manner. These results indicate a potential novel post-translational regulatory mechanism of Parkin by FKBP51 and the significance of their interaction on disease onset. KO has more flattened hippocampus using AI-assisted measurement Both pyramidal cell layer (PCL) of CA and granular cell layer (GCL) of DG distinguishable as two layers: deep cell layer and superficial layer. Distinct MAP2 expression between deep and superficial layer between KO and WT, Higher Parkin expression in KO brain Mechanism of FKBP51 inhibition resulting in Parkin, MAP2, Tau, and Tubulin expression differences between KO and WT mice, and resulting neurite outgrowth differences.


Asunto(s)
Hipocampo/metabolismo , Proteínas de Unión a Tacrolimus/genética , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Encéfalo/anatomía & histología , Células Cultivadas , Hipocampo/anatomía & histología , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/deficiencia , Proteínas de Unión a Tacrolimus/metabolismo , Tubulina (Proteína)/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba , Proteínas tau/metabolismo
4.
Biochim Biophys Acta Mol Cell Res ; 1869(5): 119212, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35090967

RESUMEN

Autophagy is a homeostatic process by which misfolded proteins, organelles and cytoplasmic material are engulfed in autophagosomal vesicles and degraded through a lisosomal pathway. FKBP8 is a member of the FK506-binding proteins family (FKBP) usually found in mitochondria and the endoplasmic reticulum. This protein plays a critical role in cell functions such as protein trafficking and folding. In the present report we demonstrate that the depletion of FKBP8 abrogated autophagy activation induced by starvation, whereas the overexpression of this protein triggered the autophagy cascade. We found that FKBP8 co-localizes with ATG14L and BECN1, both members of the VPS34 lipid kinase complex, which regulates the initial steps in the autophagosome formation process. We have also demonstrated that FKBP8 is necessary for VPS34 activity. Our findings indicate that the regulatory function of FKBP8 in the autophagy process depends of its transmembrane domain. Surprisingly, this protein was not found in autophagosomal vesicles, which reinforces the notion that the FKBP8 only participates in the initial steps of the autophagosome formation process. Taken together, our data provide evidence that FKBP8 modulates the early steps of the autophagosome formation event by interacting with the VPS34 lipid kinase complex. SUMMARY: In this article, the protein FKBP38 is reported to be a novel modulator of the initial steps of the autophagic pathway, specifically in starvation-induced autophagy. FKBP38 interacts with the VPS34 lipid kinase complex, with the transmembrane domain of FKBP38 being critical for its biological function.


Asunto(s)
Autofagia , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Autofagosomas/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Beclina-1/química , Beclina-1/metabolismo , Proteínas Portadoras/metabolismo , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética
5.
Biochem Pharmacol ; 194: 114814, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34688635

RESUMEN

Colorectal cancer (CRC) remains the most frequently diagnosed malignancy and also a major contributor to cancer-related death throughout the world. Here, we first revealed the role of histone lysine-specific demethylase 5D (KDM5D) in CRC in males. KDM5D expression in tumor and adjacent tissues of male CRC patients was investigated using immunohistochemistry and RT-qPCR, and the correlation between its expression and patients' prognosis was analyzed. Downregulation of KDM5D in CRC patients was associated with poor prognoses. Overexpression of KDM5D significantly inhibited the growth and metastasis of CRC in vitro and in vivo. The downstream mechanism of KDM5D in CRC was investigated using bioinformatics analysis, and the regulatory relationship was confirmed by ChIP-qPCR and luciferase reporter assays. KDM5D suppressed E2F1 expression by mediating H3K4me3 demethylation. E2F1, highly expressed in CRC, promoted the expression of FKBP4 at the transcriptional level by binding to the FKBP4 promoter. Finally, rescue experiments revealed that overexpression of FKBP4 significantly reversed the inhibitory effect of KDM5D on CRC growth and metastasis. Collectively, KDM5D exerted an anti-tumor and anti-metastatic in CRC through demethylation in E2F1 and suppression of FKBP4 transcription, which might represent a novel target in CRC treatment in male.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Factor de Transcripción E2F1/biosíntesis , Regulación Neoplásica de la Expresión Génica , Histona Demetilasas/biosíntesis , Antígenos de Histocompatibilidad Menor/biosíntesis , Proteínas de Unión a Tacrolimus/biosíntesis , Activación Transcripcional/fisiología , Anciano , Animales , Neoplasias Colorrectales/genética , Bases de Datos Genéticas , Factor de Transcripción E2F1/antagonistas & inhibidores , Células HCT116 , Histona Demetilasas/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor/genética , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
J Med Chem ; 64(6): 3320-3349, 2021 03 25.
Artículo en Inglés | MEDLINE | ID: mdl-33666419

RESUMEN

The FK506-binding protein 51 (FKBP51) emerged as a key player in several diseases like stress-related disorders, chronic pain, and obesity. Linear analogues of FK506 called SAFit were shown to be highly selective for FKBP51 over its closest homologue FKBP52, allowing the proof-of-concept studies in animal models. Here, we designed and synthesized the first macrocyclic FKBP51-selective ligands to stabilize the active conformation. All macrocycles retained full FKBP51 affinity and selectivity over FKBP52 and the incorporation of polar functionalities further enhanced affinity. Six high-resolution crystal structures of macrocyclic inhibitors in complex with FKBP51 confirmed the desired selectivity-enabling binding mode. Our results show that macrocyclization is a viable strategy to target the shallow FKBP51 binding site selectively.


Asunto(s)
Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Cristalografía por Rayos X , Diseño de Fármacos , Humanos , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/metabolismo
7.
Naunyn Schmiedebergs Arch Pharmacol ; 394(5): 941-954, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33284399

RESUMEN

Binary enterotoxins Clostridioides difficile CDT toxin, Clostridium botulinum C2 toxin, and Clostridium perfringens iota toxin consist of two separate protein components. The B-components facilitate receptor-mediated uptake into mammalian cells and form pores into endosomal membranes through which the enzymatic active A-components translocate into the cytosol. Here, the A-components ADP-ribosylate G-actin which leads to F-actin depolymerization followed by rounding of cells which causes clinical symptoms. The protein folding helper enzymes Hsp90, Hsp70, and peptidyl-prolyl cis/trans isomerases of the cyclophilin (Cyp) and FK506 binding protein (FKBP) families are required for translocation of A-components of CDT, C2, and iota toxins from endosomes to the cytosol. Here, we demonstrated that simultaneous inhibition of these folding helpers by specific pharmacological inhibitors protects mammalian, including human, cells from intoxication with CDT, C2, and iota toxins, and that the inhibitor combination displayed an enhanced effect compared to application of the individual inhibitors. Moreover, combination of inhibitors allowed a concentration reduction of the individual compounds as well as decreasing of the incubation time with inhibitors to achieve a protective effect. These results potentially have implications for possible future therapeutic applications to relieve clinical symptoms caused by bacterial toxins that depend on Hsp90, Hsp70, Cyps, and FKBPs for their membrane translocation into the cytosol of target cells.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Toxinas Bacterianas/toxicidad , Toxinas Botulínicas/toxicidad , Enterotoxinas/toxicidad , Animales , Células CACO-2 , Chlorocebus aethiops , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/metabolismo , Proteínas HSP70 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/metabolismo , Células Vero
8.
Drug Res (Stuttg) ; 70(7): 325-332, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32403135

RESUMEN

SAFit-1 and SAFit-2 are selective FKBP51 (FK506-binding protein 51) ligands. In this paper, we present the development and validation data of an LC-MS/MS method for the simultaneous quantitation of SAFit-1 and SAFit-2 in mice plasma as per FDA regulatory guideline. SAFit-1 and SAFit-2 along with internal standard were extracted from mice plasma using liquid-liquid extraction method. Chromatographic resolution of SAFit-1, SAFit-2 and the internal standard (warfarin) was achieved on an X-Terra phenyl column using 0.2% formic acid:acetonitrile (20:80, v/v) as an eluent, which was delivered at a flow-rate of 0.9 mL/min. The MS/MS ion transitions monitored were m/z 748.4→420.4, 803.7→384.3 and 309.2 →163.2 for SAFit-1, SAFit-2 and the internal standard, respectively. The linearity range was 2.45-2446 ng/mL for both SAFit-1 and SAFit-2. The intra- and inter-day accuracy and intra- and inter-day precision were in the range of 0.90-1.07 and 2.38-10.8%, respectively for SAFit-1; 0.97-1.15 and 0.23-12.5%, respectively for SAFit-2. Both SAFit-1 and SAFit-2 were found to be stable in stability studies (up to three freeze-thaw cycles and for long-term at -80°C for 30 days) and processed (bench-top for 3 h and in in-injector for 16 h) samples. The application of the validated method was shown in a pharmacokinetic study in mice.


Asunto(s)
Antidepresivos/análisis , Química Farmacéutica/métodos , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Administración Oral , Animales , Antidepresivos/administración & dosificación , Antidepresivos/farmacocinética , Cromatografía Líquida de Alta Presión , Masculino , Ratones , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem
9.
Alcohol Clin Exp Res ; 44(5): 1025-1036, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32154593

RESUMEN

BACKGROUND: Chronic alcohol exposure can alter glucocorticoid receptor (GR) function in some brain areas that promotes escalated and compulsive-like alcohol intake. GR antagonism can prevent dependence-induced escalation in drinking, but very little is known about the role of GR in regulating high-risk nondependent alcohol intake. Here, we investigate the role of GR in regulating binge-like drinking and aversive responses to alcohol in the High Drinking in the Dark (HDID-1) mice, which have been selectively bred for high blood ethanol (EtOH) concentrations (BECs) in the Drinking in the Dark (DID) test, and in their founder line, the HS/NPT. METHODS: In separate experiments, male and female HDID-1 mice were administered one of several compounds that inhibited GR or its negative regulator, FKBP51 (mifepristone [12.5, 25, 50, 100 mg/kg], CORT113176 [20, 40, 80 mg/kg], and SAFit2 [10, 20, 40 mg/kg]) during a 2-day DID task. EtOH consumption and BECs were measured. EtOH conditioned taste and place aversion (CTA and CPA, respectively) were measured in separate HDID-1 mice after mifepristone administration to assess GR's role in regulating the conditioned aversive effects of EtOH. Lastly, HS/NPT mice were administered CORT113176 during DID to assess whether dissimilar effects from those of HDID-1 would be observed, which could suggest that selective breeding had altered sensitivity to the effects of GR antagonism on binge-like drinking. RESULTS: GR antagonism (with both mifepristone and CORT113176) selectively reduced binge-like EtOH intake and BECs in the HDID-1 mice, while inhibition of FKBP51 did not alter intake or BECs. In contrast, GR antagonism had no effect on EtOH intake or BECs in the HS/NPT mice. Although HDID-1 mice exhibit attenuated EtOH CTA, mifepristone administration did not enhance the aversive effects of EtOH in either a CTA or CPA task. CONCLUSION: These data suggest that the selection process increased sensitivity to GR antagonism on EtOH intake in the HDID-1 mice, and support a role for the GR as a genetic risk factor for high-risk alcohol intake.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/fisiopatología , Etanol/administración & dosificación , Receptores de Glucocorticoides/antagonistas & inhibidores , Receptores de Glucocorticoides/fisiología , Consumo de Bebidas Alcohólicas/tratamiento farmacológico , Consumo de Bebidas Alcohólicas/genética , Animales , Agentes Aversivos , Consumo Excesivo de Bebidas Alcohólicas/genética , Consumo Excesivo de Bebidas Alcohólicas/prevención & control , Femenino , Isoquinolinas/farmacología , Masculino , Ratones , Mifepristona/farmacología , Pirazoles/farmacología , Receptores de Glucocorticoides/genética , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores
10.
J Med Chem ; 63(1): 231-240, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31800244

RESUMEN

Selective inhibition of FKBP51 has emerged as possible novel treatment for diseases like major depressive disorder, obesity, chronic pain, and certain cancers. The current FKBP51 inhibitors are rather large, flexible, and have to be further optimized. By using a structure-based rigidification strategy, we hereby report the design and synthesis of a novel promising bicyclic scaffold for FKBP51 ligands. The structure-activity analysis revealed the decalin scaffold as the best moiety for the selectivity-enabling subpocket of FBKP51. The resulting compounds retain high potency for FKBP51 and excellent selectivity over the close homologue FKBP52. With the cocrystal structure of an advanced ligand in this novel series, we show how the decalin locks the key selectivity-inducing cyclohexyl moiety of the ligand in a conformation typical for FKBP51-selective binding. The best compound 29 produces cell death in a HeLa-derived KB cell line, a cellular model of cervical adenocarcinoma, where FKBP51 is highly overexpressed. Our results show how FKBP51 inhibitors can be rigidified and extended while preserving FKBP51 selectivity. Such inhibitors might be novel tools in the treatment of human cancers with deregulated FKBP51.


Asunto(s)
Naftalenos/farmacología , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Sitios de Unión , Células HeLa , Humanos , Ligandos , Estructura Molecular , Naftalenos/síntesis química , Naftalenos/metabolismo , Unión Proteica , Relación Estructura-Actividad , Proteínas de Unión a Tacrolimus/metabolismo
11.
Addict Biol ; 25(3): e12758, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31173432

RESUMEN

There is still no widely effective pharmacotherapy for alcohol addiction available in the clinic. FK506-binding protein 51 (FKBP51) is a negative regulator of the glucocorticoid receptor signaling pathway that regulates the stress-induced glucocorticoid feedback circuit. Here we asked whether selective inhibitors of FKBP51, exemplified by SAFit2, may serve as a new pharmacological strategy to reduce alcohol consumption and conditioned alcohol effects in a mouse model. We report that a relatively short treatment with SAFit2 (20 mg/kg, ip) reduces ongoing 16 vol% alcohol consumption when administered during free access to alcohol in a two-bottle free-choice test. SAFit2 was also able to reduce alcohol consumption when given during an abstinence period immediately before relapse. In contrast, SAFit2 did not affect alcohol consumption when given during a relapse period after repeated withdrawal from alcohol. SAFit2 (10 and 20 mg/kg, ip) showed no effects when used in an intermittent drinking schedule. When 20 vol% alcohol was only available every other day, SAFit2 had no effect on drinking, no matter whether given during a drinking episode or the day before. SAFit2 (2 and 20 mg/kg, ip) did not affect the expression of an alcohol-induced conditioned place preference (CPP). However, SAFit2 was able to inhibit alcohol-induced reinstatement of an extinguished CPP in a dose-dependent way. Altogether, these data may suggest pharmacological inhibition of FKBP51 as a viable strategy to reduce alcohol seeking and consumption.


Asunto(s)
Consumo de Bebidas Alcohólicas , Conducta Animal/efectos de los fármacos , Depresores del Sistema Nervioso Central/administración & dosificación , Condicionamiento Clásico/efectos de los fármacos , Etanol/administración & dosificación , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Alcoholismo , Animales , Modelos Animales de Enfermedad , Masculino , Ratones , Autoadministración
12.
Drug Des Devel Ther ; 13: 3137-3149, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31564830

RESUMEN

PURPOSE: Intracellular calcium ([Ca2+]i) overload is a major cause of cell injury during myocardial ischemia/reperfusion (I/R). Dexmedetomidine (DEX) has been shown to exert anti-inflammatory and organ protective effects. This study aimed to investigate whether pretreatment with DEX could protect H9c2 cardiomyocytes against oxygen-glucose deprivation/reoxygenation (OGD/R) injury through regulating the Ca2+ signaling. METHODS: H9c2 cardiomyocytes were subjected to OGD for 12 h, followed by 3 h of reoxygenation. DEX was administered 1 h prior to OGD/R. Cell viability, lactate dehydrogenase (LDH) release, level of [Ca2+]i, cell apoptosis, and the expression of 12.6-kd FK506-binding protein/ryanodine receptor 2 (FKBP12.6/RyR2) and caspase-3 were assessed. RESULTS: Cells exposed to OGD/R had decreased cell viability, increased LDH release, elevated [Ca2+]i level and apoptosis rate, down-regulated expression of FKBP12.6, and up-regulated expression of phosphorylated-Ser2814-RyR2 and cleaved caspase-3. Pretreatment with DEX significantly blocked the above-mentioned changes, alleviating the OGD/R-induced injury in H9c2 cells. Moreover, knockdown of FKBP12.6 by small interfering RNA abolished the protective effects of DEX. CONCLUSION: This study indicates that DEX pretreatment protects the cardiomyocytes against OGD/R-induced injury by inhibiting [Ca2+]i overload and cell apoptosis via regulating the FKBP12.6/RyR2 signaling. DEX may be used for preventing cardiac I/R injury in the clinical settings.


Asunto(s)
Agonistas de Receptores Adrenérgicos alfa 2/farmacología , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Dexmedetomidina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Oxígeno/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Calcio/administración & dosificación , Calcio/análisis , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Glucosa/metabolismo , Humanos , Miocitos Cardíacos/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/genética , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
13.
Essays Biochem ; 63(3): 377-387, 2019 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-31324706

RESUMEN

The hallmark of fibrosis is an excessive accumulation of collagen, ultimately leading to organ failure. It has become evident that the deposited collagen also exhibits qualitative modifications. A marked modification is the increased cross-linking, leading to a stabilization of the collagen network and limiting fibrosis reversibility. Not only the level of cross-linking is increased, but also the composition of cross-linking is altered: an increase is seen in hydroxyallysine-derived cross-links at the expense of allysine cross-links. This results in irreversible fibrosis, as collagen cross-linked by hydroxyallysine is more difficult to degrade. Hydroxyallysine is derived from a hydroxylysine in the telopeptides of collagen. The expression of lysyl hydroxylase (LH) 2 (LH2), the enzyme responsible for the formation of telopeptidyl hydroxylysine, is universally up-regulated in fibrosis. It is expected that inhibition of this enzyme will lead to reversible fibrosis without interfering with the normal repair process. In this review, we discuss the molecular basis of collagen modifications and cross-linking, with an emphasis on LH2-mediated hydroxyallysine cross-links, and their implications for the pathogenesis and treatment of fibrosis.


Asunto(s)
Colágeno/metabolismo , Fibrosis/etiología , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Ácido 2-Aminoadípico/análogos & derivados , Ácido 2-Aminoadípico/química , Animales , Colágeno/química , Inhibidores Enzimáticos/farmacología , Fibrosis/tratamiento farmacológico , Humanos , Hidroxilación , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/antagonistas & inhibidores , Proteína-Lisina 6-Oxidasa/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores
14.
EBioMedicine ; 41: 526-537, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30737086

RESUMEN

BACKGROUND: Skin atrophy is a major adverse effect of topical glucocorticoids. We recently reported that REDD1 (regulated in development and DNA damage 1) and FKBP51 (FK506 binding protein 5), negative regulators of mTOR/Akt signaling, are induced by glucocorticoids in mouse and human skin and are central drivers of steroid skin atrophy. Thus, we hypothesized that REDD1/FKBP51 inhibitors could protect skin against catabolic effects of glucocorticoids. METHODS: Using drug repurposing approach, we screened LINCS library (http://lincsproject.org/LINCS/) to identify repressors of REDD1/FKBP51 expression. Candidate compounds were tested for their ability to inhibit glucocorticoid-induced REDD1/FKBP51 expression in human primary/immortalized keratinocytes and in mouse skin. Reporter gene expression, microarray, and chromatin immunoprecipitation were employed to evaluate effect of these inhibitors on the glucocorticoid receptor (GR) signaling. FINDINGS: Bioinformatics analysis unexpectedly identified phosphoinositide-3-kinase (PI3K)/mTOR/Akt inhibitors as a pharmacological class of REDD1/FKBP51 repressors. Selected PI3K/mTOR/Akt inhibitors-Wortmannin (WM), LY294002, AZD8055, and two others indeed blocked REDD1/FKBP51expression in human keratinocytes. PI3K/mTOR/Akt inhibitors also modified global effect of glucocorticoids on trascriptome, shifting it towards therapeutically important transrepression; negatively impacted GR phosphorylation; nuclear translocation; and GR loading on REDD1/FKBP51 gene promoters. Further, topical application of LY294002 together with glucocorticoid fluocinolone acetonide (FA) protected mice against FA-induced proliferative block and skin atrophy but did not alter the anti-inflammatory activity of FA in ear edema test. INTERPRETATION: Our results built a strong foundation for development of safer GR-targeted therapies for inflammatory skin diseases using combination of glucocorticoids with PI3K/mTOR/Akt inhibitors. FUND: Work is supported by NIH grants R01GM112945, R01AI125366, and HESI-THRIVE foundation.


Asunto(s)
Glucocorticoides/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Piel/efectos de los fármacos , Animales , Atrofia , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Humanos , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Glucocorticoides/metabolismo , Piel/patología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcriptoma/efectos de los fármacos , Wortmanina/farmacología
15.
Biochemistry ; 57(45): 6452-6459, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30366497

RESUMEN

The DNA-binding specificity of genome editing tools can be applied to gene regulation. Recently, multiple artificial transcription factors (ATFs) were shown to synergistically and efficiently regulate gene expression. Chemically triggered protein associations are useful for functional regulation at specific timings. A combination of several inducible protein association systems could enable the regulation of multiple genes at different loci with independent timing. We applied the FKBP-rapamycin-FRB and GAI-gibberellin-GID systems for gene regulation using multiple TALEs and dCas9. By the combined use of currently available systems, reporter gene assays were performed; the results indicated that gene expression was regulated by rapamycin or gibberellin in the presence of the FRB or GAI effector domains, respectively. Furthermore, the activation of endogenous genes was differentially regulated by the system. This success suggests the usability of the chemically inducible multiple ATFs for the time-dependent regulation of multiple genes, such as the case for cellular phenomena that are dependent on the programmable timing of expression and the differential expression of multiple genes.


Asunto(s)
Sistemas CRISPR-Cas , Regulación de la Expresión Génica/efectos de los fármacos , Giberelinas/farmacología , Luciferasas/metabolismo , Sirolimus/farmacología , Factores de Transcripción/metabolismo , Activación Transcripcional , Antibióticos Antineoplásicos/farmacología , Edición Génica , Humanos , Reguladores del Crecimiento de las Plantas/farmacología , Regiones Promotoras Genéticas , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
16.
Chem Pharm Bull (Tokyo) ; 66(6): 602-607, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29863061

RESUMEN

Dolastatin 16 is a cyclic depsipeptide isolated from the marine invertebrates and cyanobacterium Lyngbya majuscula, however, its bioactivity has been a historical question. In this study, peptidyl-prolyl cis-trans isomerase FKBP1A (FKBP12) was predicted as a potential target of dolastatin 16 via PharmMapper as well as verified using chemical-protein interactome (CPI) and molecular docking. FKBP1A has been previously identified as a target for the natural polyketide FK506 (tacrolimus), an immune suppressor inhibiting the rejection of organ transplantation in clinical use. The comparison study via the reverse pharmacophore screening and molecular docking of dolastatin 16 and FK506 indicated the good consistency of analysis with the computational approach. As the results, the lowest binding energy of dolastatin 16-FKBP1A complex was -7.4 kcal/mol and FK506-FKBP1A complex was -8.7 kcal/mol. The ligand dolastatin 16 formed three hydrogen bonds vs. four of FK506, as well as seven hydrophobic interactions vs. six of FK506 within the active site residues. These functional residues are highly repetitive and consistent with previously reported active site of model of FK506-FKBP1A complex, and the pharmacophore model was shown feasibly matching with the molecular feature of dolastatin 16.


Asunto(s)
Depsipéptidos/farmacología , Simulación del Acoplamiento Molecular , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Depsipéptidos/química , Evaluación Preclínica de Medicamentos , Humanos , Modelos Moleculares , Conformación Molecular , Tacrolimus/química , Tacrolimus/farmacología
17.
ACS Chem Biol ; 13(8): 2288-2299, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29893552

RESUMEN

Genetic and epigenetic alterations in FK506-binding protein 5 ( FKBP5) have been associated with increased risk for psychiatric disorders, including post-traumatic stress disorder (PTSD). Some of these common variants can increase the expression of FKBP5, the gene that encodes FKBP51. Excess FKBP51 promotes hypothalamic-pituitary-adrenal (HPA) axis dysregulation through altered glucocorticoid receptor (GR) signaling. Thus, we hypothesized that GR activity could be restored by perturbing FKBP51. Here, we screened 1280 pharmacologically active compounds and identified three compounds that rescued FKBP51-mediated suppression of GR activity without directly activating GR. One of the three compounds, benztropine mesylate, disrupted the association of FKBP51 with the GR/Hsp90 complex in vitro. Moreover, we show that removal of FKBP51 from this complex by benztropine restored GR localization in ex vivo brain slices and primary neurons from mice. In conclusion, we have identified a novel disruptor of the FKBP51/GR/Hsp90 complex. Targeting this complex may be a viable approach to developing treatments for disorders related to aberrant FKBP51 expression.


Asunto(s)
Benzotropina/farmacología , Depresión/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/metabolismo , Receptores de Glucocorticoides/metabolismo , Trastornos por Estrés Postraumático/tratamiento farmacológico , Proteínas de Unión a Tacrolimus/metabolismo , Animales , Benzotropina/química , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Depresión/metabolismo , Descubrimiento de Drogas , Humanos , Ratones , Terapia Molecular Dirigida , Unión Proteica/efectos de los fármacos , Trastornos por Estrés Postraumático/metabolismo , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores
18.
J Med Chem ; 61(8): 3660-3673, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29578710

RESUMEN

FK506-binding proteins (FKBPs) are evolutionarily conserved proteins that display peptidyl-prolyl isomerase activities and act as coreceptors for immunosuppressants. Microbial macrophage-infectivity-potentiator (Mip)-type FKBPs can enhance infectivity. However, developing druglike ligands for FKBPs or Mips has proven difficult, and many FKBPs and Mips still lack biologically useful ligands. To explore the scope and potential of C5-substituted [4.3.1]-aza-bicyclic sulfonamides as a broadly applicable class of FKBP inhibitors, we developed a new synthesis method for the bicyclic core scaffold and used it to prepare an FKBP- and Mip-focused library. This allowed us to perform a systematic structure-activity-relationship analysis across key human FKBPs and microbial Mips, yielding highly improved inhibitors for all the FKBPs studied. A cocrystal structure confirmed the molecular-binding mode of the core structure and explained the affinity gained as a result of the preferred substituents. The best FKBP and Mip ligands showed promising antimalarial, antileginonellal, and antichlamydial properties in cellular models of infectivity, suggesting that substituted [4.3.1]-aza-bicyclic sulfonamides could be a novel class of anti-infectives.


Asunto(s)
Compuestos de Azabiciclo/farmacología , Sulfonamidas/farmacología , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Compuestos de Azabiciclo/síntesis química , Compuestos de Azabiciclo/química , Compuestos de Azabiciclo/metabolismo , Candida albicans/efectos de los fármacos , Chlamydia trachomatis/efectos de los fármacos , Células HeLa , Humanos , Legionella pneumophila/efectos de los fármacos , Estructura Molecular , Plasmodium falciparum/efectos de los fármacos , Unión Proteica , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo
19.
Protein Eng Des Sel ; 31(12): 489-498, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31120120

RESUMEN

The FK506-binding protein of Plasmodium knowlesi (Pk-FKBP35) is considerably a viable antimalarial drug target, which belongs to the peptidyl-prolyl cis-trans isomerase (PPIase) protein family member. Structurally, this protein consists of an N-terminal FK506-binding domain (FKBD) and a C-terminal tetratricopeptide repeat domain (TPRD). This study aims to decipher functional properties of these domains as a platform for development of novel antimalarial drugs. Accordingly, full-length Pk-FKBP35 as well as its isolated domains, Pk-FKBD and Pk-TPRD were overexpressed, purified, and characterized. The results showed that catalytic PPIase activity was confined to the full-length Pk-FKBP35 and Pk-FKBD, suggesting that the catalytic activity is structurally regulated by the FKBD. Meanwhile, oligomerization analysis revealed that Pk-TPRD is essential for dimerization. Asp55, Arg60, Trp77 and Phe117 in the Pk-FKBD were considerably important for catalysis as underlined by significant reduction of PPIase activity upon mutations at these residues. Further, inhibition activity of Pk-FKBP35 towards calcineurin phosphatase activity revealed that the presence of FKBD is essential for the inhibitory property, while TPRD may be important for efficient binding to calcineurin. We then discussed possible roles of FKBP35 in Plasmodium cells and proposed mechanisms by which the immunosuppressive drug, FK506, interacts with the protein.


Asunto(s)
Plasmodium knowlesi/enzimología , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/metabolismo , Calcineurina/metabolismo , Expresión Génica , Humanos , Plasmodium knowlesi/genética , Dominios Proteicos , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/genética
20.
Proteins ; 86(1): 43-56, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29023988

RESUMEN

As co-chaperones of the 90-kDa heat shock protein(HSP90), FK506 binding protein 51 (FKBP51) and FK506 binding protein 52 (FKBP52) modulate the maturation of steroid hormone receptor through their specific FK1 domains (FKBP12-like domain 1). The inhibitors targeting FK1 domains are potential therapies for endocrine-related physiological disorders. However, the structural conservation of the FK1 domains between FKBP51 and FKBP52 make it difficult to obtain satisfactory selectivity in FK506-based drug design. Fortunately, a series of iFit ligands synthesized by Hausch et al exhibited excellent selectivity for FKBP51, providing new opportunity for design selective inhibitors. We performed molecular dynamics simulation, binding free energy calculation and unbinding pathway analysis to reveal selective mechanism for the inhibitor iFit4 binding with FKBP51 and FKBP52. The conformational stability evaluation of the "Phe67-in" and "Phe67-out" states implies that FKBP51 and FKBP52 have different preferences for "Phe67-in" and "Phe67-out" states, which we suggest as the determinant factor for the selectivity for FKBP51. The binding free energy calculations demonstrate that nonpolar interaction is favorable for the inhibitors binding, while the polar interaction and entropy contribution are adverse for the inhibitors binding. According to the results from binding free energy decomposition, the electrostatic difference of residue 85 causes the most significant thermodynamics effects on the binding of iFit4 to FKBP51 and FKBP52. Furthermore, the importance of substructure units on iFit4 were further evaluated by unbinding pathway analysis and residue-residue contact analysis between iFit4 and the proteins. The results will provide new clues for the design of selective inhibitors for FKBP51.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas de Unión a Tacrolimus/química , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Enlace de Hidrógeno , Ligandos , Estructura Molecular , Unión Proteica , Conformación Proteica , Isoformas de Proteínas/antagonistas & inhibidores , Isoformas de Proteínas/química , Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...