Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hum Exp Toxicol ; 41: 9603271211065972, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35238236

RESUMEN

Propofol is a commonly used drug for sedation and general anesthesia during cancer surgery. Previous studies indicate that propofol exerts anti-tumor effect in various cancers. The aim of this study was to investigate the underlying molecular mechanism of propofol in liver cancer. The effects of propofol on liver cancer cells were evaluated by cell viability assay, colony formation assay, and tumor xenograft model. Dysregulated lncRNAs of propofol-treated liver cancer cells were evaluated by transcriptome RNA sequencing. The underlying molecular mechanisms of lncRNA cancer susceptibility candidate 9 (CASC9) in propofol-induced anti-tumor effects were evaluated by western blot, quantitative real-time polymerase chain reaction (qRT-PCR), wound scratch healing assay, transwell cell migration and invasion assay, TUNEL staining, fluorescence in situ hybridization, RNA immunoprecipitation (RIP), and chromatin immunoprecipitation (ChIP). We found that propofol suppressed proliferation, migration, invasion, and tumor xenograft growth of liver cancer cells in a dose-dependent manner. Exosomes transfer from propofol-treated cells inhibited proliferation, migration, and invasion and promoted apoptosis of liver cancer cells. Transcriptional profiling of propofol-treated liver cancer cells identified CASC9 as significantly downregulated lncRNA in cells and exosomes. Enforced CASC9 expression partially rescued the inhibitory effects of propofol on liver cancer cells. Furthermore, CASC9 was found to interact directly with EZH2 and epigenetically regulated PTEN expression. Restoration of CASC9 partially abrogated the inhibition of propofol on Akt/mTOR signaling. Our results indicated that propofol exerted anti-tumor effects by downregulating CASC9, and subsequently suppressed Akt/mTOR signaling. Our findings provided a novel insight into propofol-induced anti-tumor effects in liver cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Línea Celular Tumoral/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Monoéster Fosfórico Hidrolasas/metabolismo , Propofol/uso terapéutico , Proteínas de Unión a Tacrolimus/metabolismo , Antineoplásicos/farmacología , Movimiento Celular/genética , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hepáticas/fisiopatología , Monoéster Fosfórico Hidrolasas/efectos de los fármacos , Monoéster Fosfórico Hidrolasas/genética , Propofol/metabolismo , Propofol/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/genética
2.
J Ethnopharmacol ; 290: 115101, 2022 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-35151834

RESUMEN

ETHNOPHARMACOLOGICAL RELEVANCE: Zexie Tang (ZXT), only two consists with Alismatis Rhizoma (AR) and Atractylodes macrocephala Rhizoma (AM), a classical Chinese medicine formula from Synopsis of the Golden Chamber with a history of 2000 years. Clinical observation in recent years has found that ZXT has excellent lipid-lowering effect. AIM OF THE STUDY: To explore the potential mechanism of ZXT ameliorates hyperlipidemia based on FKBP38/mTOR/SREBPs pathway. MATERIALS AND METHODS: WD-induced hyperlipidemia mice and oleic acid induced cell lipid accumulation model were used to investigate pharmacodynamic. The effect of ZXT on the transcriptional activity of SREBPs was detected by reporter gene assay. Proteins and downstream genes of mTOR/SREBPs pathway were detected in vivo and in vitro. Combined with network pharmacology and HPLC-Q-TOF/MS, the active ingredients were screened and identified. The interaction between active compounds of ZXT and FKBP38 protein were analyzed by docking analysis. RESULTS: ZXT decreased TC, TG and LDL-c levels in blood of WD-induced hyperlipidemia mouse model, and improved insulin resistance in vivo. ZXT also reduced TC, TG and lipid accumulation in cells line, and inhibited SREBPs luciferase activity, protein and its target genes expression such as FASN, HMGCR, etc. Meanwhile, ZXT inhibited protein expression levels of p-mTOR, p-S6K, etc in vitro and in vivo. Combined with network pharmacology and HPLC-Q-TOF/MS, 16 active ingredients were screened and identified. Docking results showed that active compounds of ZXT binding to FKBP38 and formed hydrogen bond. CONCLUSION: Our findings highlighted that ZXT ameliorates hyperlipidemia, in which FKBP/mTOR/SREBPs pathway might be the potential regulatory mechanism.


Asunto(s)
Hiperlipidemias/patología , Lípidos/sangre , Extractos Vegetales/farmacología , Proteínas de Unión a los Elementos Reguladores de Esteroles/efectos de los fármacos , Serina-Treonina Quinasas TOR/efectos de los fármacos , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Alismatales , Animales , Atractylodes , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Farmacología en Red
3.
Br J Cancer ; 122(3): 361-371, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31772325

RESUMEN

BACKGROUND: ALM201 is a therapeutic peptide derived from FKBPL that has previously undergone preclinical and clinical development for oncology indications and has completed a Phase 1a clinical trial in ovarian cancer patients and other advanced solid tumours. METHODS: In vitro, cancer stem cell (CSC) assays in a range of HGSOC cell lines and patient samples, and in vivo tumour initiation, growth delay and limiting dilution assays, were utilised. Mechanisms were determined by using immunohistochemistry, ELISA, qRT-PCR, RNAseq and western blotting. Endogenous FKBPL protein levels were evaluated using tissue microarrays (TMA). RESULTS: ALM201 reduced CSCs in cell lines and primary samples by inducing differentiation. ALM201 treatment of highly vascularised Kuramochi xenografts resulted in tumour growth delay by disruption of angiogenesis and a ten-fold decrease in the CSC population. In contrast, ALM201 failed to elicit a strong antitumour response in non-vascularised OVCAR3 xenografts, due to high levels of IL-6 and vasculogenic mimicry. High endogenous tumour expression of FKBPL was associated with an increased progression-free interval, supporting the protective role of FKBPL in HGSOC. CONCLUSION: FKBPL-based therapy can (i) dually target angiogenesis and CSCs, (ii) target the CD44/STAT3 pathway in tumours and (iii) is effective in highly vascularised HGSOC tumours with low levels of IL-6.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Diferenciación Celular/efectos de los fármacos , Células Madre Neoplásicas/efectos de los fármacos , Neovascularización Patológica/patología , Neoplasias Ováricas/patología , Péptidos/farmacología , Proteínas de Unión a Tacrolimus , Animales , Carcinoma Epitelial de Ovario/irrigación sanguínea , Carcinoma Epitelial de Ovario/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Receptores de Hialuranos/efectos de los fármacos , Receptores de Hialuranos/metabolismo , Técnicas In Vitro , Interleucina-6/metabolismo , Ratones , Ratones SCID , Neovascularización Patológica/metabolismo , Neoplasias Ováricas/irrigación sanguínea , Neoplasias Ováricas/metabolismo , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
4.
J Clin Endocrinol Metab ; 101(11): 4305-4312, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27459525

RESUMEN

CONTEXT: Endogenous Cushing's syndrome is caused by chronically elevated levels of cortisol. Mifepristone, a glucocorticoid receptor (GR) antagonist, is approved for the treatment of Cushing's syndrome. Currently there is an unmet clinical need for a direct biochemical method for monitoring the immediate effectiveness of mifepristone in patients with Cushing's syndrome. The glucocorticoid induction of FK506-binding protein 5 (FKBP5) expression is rapid and has been shown to be attenuated by GR antagonists in a range of in vitro and in vivo models. OBJECTIVE: The objective of the study was to develop a quantitative PCR assay for FKBP5 mRNA expression in blood and apply it to measure the inhibition of glucocorticoid-induced FKBP5 expression by GR antagonists in healthy human subjects. METHODS: Briefly, blood samples were acquired from a phase I study in which healthy human subjects were administered either a single dose of the GR agonist prednisone with and without coadministration of a single oral dose of mifepristone or glucocorticoid receptor antagonist (CORT125134) or multiple daily doses of CORT125134 over 14 days with coadministration of prednisone with the final dose. FKBP5 mRNA levels were analyzed by quantitative PCR in blood samples collected at selected time points. SETTING: The study was conducted at Quotient Clinical (Nottingham, United Kingdom). RESULTS: Oral administration of the glucocorticoid prednisone to healthy human subjects resulted in a time-dependent increase of FKBP5 mRNA to peak levels of approximately 12-fold compared with unstimulated levels within 4 hours of steroid administration, followed by a reduction to baseline levels within 24 hours. Furthermore, oral administration of mifepristone or the selective GR antagonist CORT125134 had the desired effect of inhibiting prednisone-mediated activation of GR as seen by a reduction of FKBP5 mRNA levels. CONCLUSIONS: The inhibition of FKBP5 mRNA expression by a selective GR antagonist is a potential clinical biomarker of GR antagonism.


Asunto(s)
Glucocorticoides/farmacología , Antagonistas de Hormonas/farmacología , Mifepristona/farmacología , Prednisona/farmacología , ARN Mensajero/sangre , Receptores de Glucocorticoides/antagonistas & inhibidores , Proteínas de Unión a Tacrolimus/sangre , Adulto , Animales , Bioensayo , Biomarcadores/sangre , Glucocorticoides/administración & dosificación , Antagonistas de Hormonas/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Mifepristona/administración & dosificación , Reacción en Cadena de la Polimerasa , Prednisona/administración & dosificación , ARN Mensajero/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Adulto Joven
5.
Psychopharmacology (Berl) ; 233(18): 3315-30, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27387895

RESUMEN

RATIONALE: Inflammation plays a key role in the pathogenesis of major depressive disorder (MDD) for a subset of depressed individuals. One of the possible routes by which cytokines can induce depressive symptoms is by promoting the dysregulation of hypothalamic-pituitary-adrenal (HPA) axis via altering glucocorticoid receptor (GR) function. OBJECTIVES: We investigated the mechanisms that finely tune the GR functioning upon lipopolysaccharide (LPS), i.e., subcellular localization of the GR, the levels of its co-chaperones FK506 binding protein 52 (FKBP4) and FK506 binding protein 51 (FKBP5), the receptor phosphorylation status along with its upstream kinases, as well as mRNA levels of GR-regulated genes in the prefrontal cortex (PFC) of male and female Wistar rats. RESULTS: We found that upon LPS treatment, animals of both sexes exhibited depressive-like behavior and elevated serum corticosterone. However, the nuclear translocation of the GR and both FKBPs was found only in males, together with elevated phosphorylation of the GR at serine 232 and 246 and the activation and nuclear translocation of all analyzed kinases. This activation of the GR in males was paralleled with altered expression of GR-related genes, particularly PTGS2 and BDNF. CONCLUSION: Our data suggest that LPS treatment produced alterations in the mechanisms that control the GR nuclear translocation in the PFC of males, and that these mechanisms may contribute to the sex-specific dysfunction of GR-related neurotrophic and neuroinflammatory processes in inflammation-associated depression.


Asunto(s)
Conducta Animal/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Depresión/metabolismo , Lipopolisacáridos/farmacología , Corteza Prefrontal/efectos de los fármacos , Receptores de Glucocorticoides/efectos de los fármacos , Animales , Núcleo Celular/metabolismo , Corticosterona/sangre , Depresión/genética , Trastorno Depresivo Mayor/metabolismo , Femenino , Sistema Hipotálamo-Hipofisario/metabolismo , Inflamación , Masculino , Fosforilación , Sistema Hipófiso-Suprarrenal/metabolismo , Corteza Prefrontal/metabolismo , Transporte de Proteínas , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Factores Sexuales , Transducción de Señal , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/metabolismo
6.
J Med Chem ; 59(6): 2410-22, 2016 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-26954324

RESUMEN

The FK506-binding protein 51 (FKBP51) is a key regulator of stress hormone receptors and an established risk factor for stress-related disorders. Drug development for FKBP51 has been impaired by the structurally similar but functionally opposing homologue FKBP52. High selectivity between FKBP51 and FKBP52 can be achieved by ligands that stabilize a recently discovered FKBP51-favoring conformation. However, drug-like parameters for these ligands remained unfavorable. In the present study, we replaced the potentially labile pipecolic ester group of previous FKBP51 ligands by various low molecular weight amides. This resulted in the first series of pipecolic acid amides, which had much lower molecular weights without affecting FKBP51 selectivity. We discovered a geminally substituted cyclopentyl amide as a preferred FKBP51-binding motif and elucidated its binding mode to provide a new lead structure for future drug optimization.


Asunto(s)
Ácidos Pipecólicos/síntesis química , Ácidos Pipecólicos/farmacología , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Amidas/síntesis química , Amidas/farmacología , Animales , Línea Celular , Descubrimiento de Drogas , Humanos , Modelos Moleculares , Conformación Molecular , Peso Molecular , Neuritas/efectos de los fármacos , Factores de Riesgo , Relación Estructura-Actividad , Difracción de Rayos X
7.
Nat Chem Biol ; 11(1): 33-7, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25436518

RESUMEN

The FK506-binding protein 51 (FKBP51, encoded by the FKBP5 gene) is an established risk factor for stress-related psychiatric disorders such as major depression. Drug discovery for FKBP51 has been hampered by the inability to pharmacologically differentiate against the structurally similar but functional opposing homolog FKBP52, and all known FKBP ligands are unselective. Here, we report the discovery of the potent and highly selective inhibitors of FKBP51, SAFit1 and SAFit2. This new class of ligands achieves selectivity for FKBP51 by an induced-fit mechanism that is much less favorable for FKBP52. By using these ligands, we demonstrate that selective inhibition of FKBP51 enhances neurite elongation in neuronal cultures and improves neuroendocrine feedback and stress-coping behavior in mice. Our findings provide the structural and functional basis for the development of mechanistically new antidepressants.


Asunto(s)
Proteínas de Unión a Tacrolimus/antagonistas & inhibidores , Adaptación Psicológica/efectos de los fármacos , Animales , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Sitios de Unión/efectos de los fármacos , Células Cultivadas , Descubrimiento de Drogas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Neuritas/efectos de los fármacos , Conformación Proteica , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/efectos de los fármacos
8.
Arterioscler Thromb Vasc Biol ; 33(10): 2425-31, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23887639

RESUMEN

OBJECTIVE: Sirolimus (SRL) is an immunosuppressant drug used to prevent rejection in organ transplantation and neointimal hyperplasia when delivered from drug-eluting stents. Major side effects of SRL include edema and local collection of intimal lipid deposits at drug-eluting stent sites, suggesting that SRL impairs endothelial barrier function (EBF). The aim of this study was to address the role of SRL on impaired EBF and the potential mechanisms involved. APPROACH AND RESULTS: Cultured human aortic endothelial cells (HAECs) and intact human and mouse endothelium was examined to determine the effect of SRL, which binds FKBP12.6 to inhibit the mammalian target of rapamycin, on EBF. EBF, measured by transendothelial electrical resistance, was impaired in HAECs when treated with SRL or small interfering RNA for FKBP12.6 and reversed when pretreated with ryanodine, a stabilizer of ryanodine receptor 2 intracellular calcium release channels. Intracellular calcium increased in HAECs treated with SRL and normalized with ryanodine pretreatment. SRL-treated HAECs demonstrated increases in protein kinase C-α phosphorylation, a calcium sensitive serine/threonine kinase important in vascular endothelial (VE) cadherin barrier function through its interaction with p120-catenin (p120). Immunostaining of HAECs, human coronary and mouse aortic endothelium treated with SRL showed disruption of p120-VE cadherin interaction treated with SRL. SRL impairment of HAEC EBF was reduced with protein kinase C-α small interfering RNA. Mice treated with SRL demonstrated increased vascular permeability by Evans blue albumin extravasation in the lungs, heart, and aorta. CONCLUSIONS: SRL-FKBP12.6 impairs EBF by activation of protein kinase C-α and downstream disruption of the p120-VE cadherin interaction in vascular endothelium. These data suggest this mechanism may be an important contributor of SRL side effects related to impaired EBF.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Permeabilidad Capilar/efectos de los fármacos , Cateninas/metabolismo , Células Endoteliales/efectos de los fármacos , Proteína Quinasa C-alfa/metabolismo , Sirolimus/toxicidad , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Células Endoteliales/enzimología , Activación Enzimática , Humanos , Ratones , Ratones Endogámicos C57BL , Fosforilación , Unión Proteica , Proteína Quinasa C-alfa/genética , Interferencia de ARN , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo , Factores de Tiempo , Transfección , Catenina delta
9.
J Med Chem ; 56(10): 3922-35, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23647266

RESUMEN

The design of efficient ligands remains a key challenge in drug discovery. In the quest for lead-like ligands for the FK506-binding protein 51 (FKBP51), we designed two new classes of bicyclic sulfonamides to probe the contribution of conformational energy in these ligands. The [4.3.1] scaffold had consistently higher affinity compared to the [3.3.1] or monocyclic scaffolds, which could be attributed to better preorganization of two key recognition motifs. Surprisingly, the binding of the rigid [4.3.1] scaffold was enthalpy-driven and entropically disfavored compared to the flexible analogues. Cocrystal structures at atomic resolution revealed that the sulfonamide nitrogen in the bicyclic scaffolds can accept an unusual hydrogen bond from Tyr(113) that mimics the putative FKBP transition state. This resulted in the first lead-like, functionally active ligand for FKBP51. Our work exemplifies how atom-efficient ligands can be achieved by careful conformational control even in very open and thus difficult binding sites such as FKBP51.


Asunto(s)
Inmunosupresores/química , Inmunosupresores/farmacología , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Sitios de Unión , Compuestos Bicíclicos con Puentes/síntesis química , Compuestos Bicíclicos con Puentes/farmacología , Calorimetría , Cromatografía Líquida de Alta Presión , Cristalografía , Diseño de Fármacos , Humanos , Indicadores y Reactivos , Ligandos , Conformación Proteica , Receptores de Gonadotropina/efectos de los fármacos , Receptores de Gonadotropina/metabolismo , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacología , Tacrolimus/química
10.
Pflugers Arch ; 465(3): 409-17, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23299847

RESUMEN

Chemically-inducible dimerization (CID) is a powerful tool that has proved useful in solving numerous problems in cell biology and related fields. In this review, we focus on case studies where CID was able to provide insight into otherwise refractory problems. Of particular interest are the cases of lipid second messengers and small GTPases, where the "signaling paradox" (how a small pool of signaling molecules can generate a large range of responses) can be at least partly explained through results gleaned from CID experiments. We also discuss several recent technical advances that provide improved specificity in CID action, novel CID substrates that allow simultaneous orthogonal manipulation of multiple systems in one cell, and several applications that move beyond the traditional CID technique of moving a protein of interest to a specific spatiotemporal location.


Asunto(s)
Multimerización de Proteína/efectos de los fármacos , Transducción de Señal , Animales , Humanos , Metabolismo de los Lípidos , Proteínas de Unión al GTP Monoméricas/metabolismo , Sirolimus/farmacología , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/efectos de los fármacos
11.
Acta Otolaryngol ; 132(1): 4-9, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22026503

RESUMEN

CONCLUSIONS: Quantitative, real-time RT-PCR demonstrated that intratympanic dexamethasone significantly up-regulates the expression of Fkbp5 in cochleae of mice in vivo. The immunohistochemistry results showed fundamentally ubiquitous expression of Fkbp5 in cochlear structures, with relatively strong expression in type 4 fibrocytes and weak signal in the inner hair cells. These data indicate that dexamethasone regulates gene expression at the level of transcription in vivo and that this process is basically ubiquitous in the cochlea. OBJECTIVES: To demonstrate that intratympanically applied dexamethasone up-regulates Fkbp5 in the cochlea in vivo. METHODS: Dexamethasone or control saline were intratympanically applied to adult C57/BL6 mice and dexamethasone-dependent changes in the levels of Fkbp5 expression in the cochlea were analyzed using quantitative real-time RT-PCR. The expression pattern of Fkbp5 in cochlea was investigated by immunohistochemistry in mice that were administered dexamethasone and in controls. RESULTS: Quantitative real-time RT-PCR demonstrated significant increases of Fkbp5 expression levels in cochleae of dexamethasone-treated mice as compared with controls at 12 h after application (244.8 ± 155.5, n = 5 vs 100.0 ± 3.0, n = 6, p < 0.01). Immunohistochemistry showed fundamentally ubiquitous expression of Fkbp5 in cochlear structures, with some strongly positive fibrocytes in the spiral ligaments and weak immunoreactivity in the inner hair cells. Distribution of Fkbp5 signaling was not different between the dexamethasone-treated group and controls.


Asunto(s)
Cóclea/metabolismo , Dexametasona/administración & dosificación , Pérdida Auditiva Sensorineural/genética , ARN Mensajero/genética , Proteínas de Unión a Tacrolimus/genética , Regulación hacia Arriba/genética , Administración Tópica , Animales , Cóclea/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Glucocorticoides/administración & dosificación , Proteínas HSP90 de Choque Térmico , Pérdida Auditiva Sensorineural/tratamiento farmacológico , Pérdida Auditiva Sensorineural/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas de Unión a Tacrolimus/biosíntesis , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Membrana Timpánica , Regulación hacia Arriba/efectos de los fármacos
12.
Acta Pharmacol Sin ; 32(2): 223-9, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21293474

RESUMEN

AIM: To investigate which endothelin receptors mediated isoproterenol (ISO)-induced downregulation of FKBP12.6/12 in cardiomyocytes and study whether argirhein, a novel compound containing rhein and L-arginine that has anti-inflammatory activity, could reverse the downregulation of FKBP12.6/12 induced by ISO. METHODS: Neonatal rat cardiomyocytes were incubated with ISO to downregulate FKBP12.6/12. Then the cells were treated with a selective ET(A) blocker (PD156707) and a ET(B) blocker (IRL1038), a dual ET(A)/ET(B) antagonist (CPU0213), and argirhein, respectively. FKBP12.6/12 expression was assayed by RT-PCR, Western blot, and immunocytochemistry. RESULTS: The expression of FKBP12.6 mRNA was reduced by 37.7% (P<0.01) and 28.9% (P<0.05) relative to the control by ISO 1 and 0.1 µmol/L, respectively, but no response to ISO 0.01 µmol/L was observed in vitro. FKBP12.6/12 protein expression was reduced by 47.2% (P<0.01) and 37.8% (P<0.05) by ISO 1 and 0.1 µmol/L, respectively. This decrease was reversed significantly by PD156707, or IRL1038, and CPU0213. CPU0213 was more potent than either PD156707 or IRL-1038. Argirhein 10 µmol/L blunted the downregulation of FKBP12.6/12 by ISO, as demonstrated by the rising mRNA and protein levels and by the fluorescent density of the ISO-incubated cardiomyocytes. CONCLUSION: In cardiomyocytes, the ISO induced downregulation of FKBP12.6/12 is modulated by both ET(A) and ET(B). A new compound, argirein, reversed the down-regulation of FKBP12.6/12 expression in myocardial cells stimulated with ISO.


Asunto(s)
Antraquinonas/farmacología , Antiinflamatorios/farmacología , Proteína 1A de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Animales , Animales Recién Nacidos , Western Blotting , Células Cultivadas , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Isoproterenol/administración & dosificación , Isoproterenol/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína 1A de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/genética
13.
J Pharm Pharmacol ; 62(1): 77-83, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20723002

RESUMEN

OBJECTIVES: The aim of this study was to determine if CPU228, a derivative of dofetilide, is more effective than dofetilide in attenuating isoproterenol-induced heart failure by recovering downregulated FK506 binding protein (FKBP12.6), and suppressing oxidative stress, upregulated NADPH oxidase and protein kinase C epsilon (PKC epsilon) hyperphosphorylation in the myocardium. METHODS: Heart failure was induced by isoproterenol (1 mg/kg s.c. for 5 days) in male Sprague-Dawley rats. Intervention with either CPU228 or dofetilide (2 mg/kg on Days 3-5) was then conducted in vivo and in vitro. KEY FINDINGS: Isoproterenol produced compromised left ventricular systolic pressure, left ventricular pressure rise (dp/dt(max)) and fall (dp/dt(min)), and left ventricular end-diastolic pressure, associated with oxidative stress, abnormal FKBP12.6, NADPH oxidase p67phox and PKC epsilon in the myocardium. CPU228 was more effective in attenuating these changes than dofetilide in vivo. Dofetilide produced a prolonged QTc to replace a shortened one. In primary neonatal cardiomyocytes, cultured with isoproterenol and treated with either CPU228 or dofetilide at 10(-8), 10(-7) and 10(-6) mol/l, isoproterenol produced a hyperadrenergic state characterized by downregulated FKBP12.6, upregulated NADPH oxidase p67phox and PKC epsilon in vitro. CPU228 was more effective than dofetilide in recovering these changes in a dose-dependent manner without a prolonged QTc. CONCLUSIONS: CPU228 was more effective than dofetilide in attenuating heart failure by normalizing isoproterenol-induced changes, including downregulation of FKBP12.6, upregulation of NADPH oxidase and PKC epsilon hyperphosphorylation in vivo and in vitro.


Asunto(s)
Insuficiencia Cardíaca/tratamiento farmacológico , Naftalenos/farmacología , Estrés Oxidativo/efectos de los fármacos , Fenetilaminas/farmacología , Sulfonamidas/farmacología , Animales , Antiarrítmicos/administración & dosificación , Antiarrítmicos/farmacología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo/efectos de los fármacos , Insuficiencia Cardíaca/fisiopatología , Isoproterenol , Masculino , NADPH Oxidasas/efectos de los fármacos , NADPH Oxidasas/genética , Naftalenos/administración & dosificación , Fenetilaminas/administración & dosificación , Fosforilación/efectos de los fármacos , Proteína Quinasa C-epsilon/efectos de los fármacos , Proteína Quinasa C-epsilon/metabolismo , Ratas , Ratas Sprague-Dawley , Sulfonamidas/administración & dosificación , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/genética , Regulación hacia Arriba/efectos de los fármacos
14.
J Biol Chem ; 285(27): 20870-81, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20439463

RESUMEN

Hepatitis C virus (HCV) often establishes a persistent infection that most likely involves a complex host-virus interplay. We previously reported that the HCV nonstructural protein 5A (NS5A) bound to cellular protein FKBP38 and resulted in apoptosis suppression in human hepatoma cell line Huh7. In the present research we further found that NS5A increased phosphorylation levels of two mTOR-targeted substrates, S6K1 and 4EBP1, in Huh7 in the absence of serum. mTOR inhibitor rapamycin or NS5A knockdown blocked S6K1 and 4EBP1 phosphorylation increase in NS5A-Huh7 and HCV replicon cells, suggesting that NS5A specifically regulated mTOR activation. Overexpression of NS5A and FKBP38 mutants or FKBP38 knockdown revealed this mTOR activation was dependent on NS5A-FKBP38 interaction. Phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 treatment in NS5A-Huh7 showed that the mTOR activation was independent of PI3K. Moreover, NS5A suppressed caspase 3 and poly(ADP-ribose) polymerase activation, which was abolished by NS5A knockdown or rapamycin, indicating NS5A inhibited apoptosis specifically through the mTOR pathway. Further analyses suggested that apoptotic inhibition exerted by NS5A via mTOR also required NS5A-FKBP38 interaction. Glutathione S-transferase pulldown and co-immunoprecipitation showed that NS5A disrupted the mTOR-FKBP38 association. Additionally, NS5A or FKBP38 mutants recovered the mTOR-FKBP38 interaction; this indicated that the impairment of mTOR-FKBP38 association was dependent on NS5A-FKBP38 binding. Collectively, our data demonstrate that HCV NS5A activates the mTOR pathway to inhibit apoptosis through impairing the interaction between mTOR and FKBP38, which may represent a pivotal mechanism for HCV persistence and pathogenesis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/farmacología , Anticuerpos Antivirales/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Caspasa 3/metabolismo , Línea Celular , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Activación Enzimática , Hepacivirus/genética , Hepacivirus/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/efectos de los fármacos , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Eliminación de Secuencia , Serina-Treonina Quinasas TOR , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Transfección , Proteínas no Estructurales Virales/genética , Replicación Viral/efectos de los fármacos
15.
J Med Chem ; 52(11): 3516-22, 2009 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-19422228

RESUMEN

We developed an in-cell NMR assay for screening small molecule interactor libraries (SMILI-NMR) for compounds capable of disrupting or enhancing specific interactions between two or more components of a biomolecular complex. The method relies on the formation of a well-defined biocomplex and utilizes in-cell NMR spectroscopy to identify the molecular surfaces involved in the interaction at atomic scale resolution. Changes in the interaction surface caused by a small molecule interfering with complex formation are used as a read-out of the assay. The in-cell nature of the experimental protocol insures that the small molecule is capable of penetrating the cell membrane and specifically engaging the target molecule(s). Utility of the method was demonstrated by screening a small dipeptide library against the FKBP-FRB protein complex involved in cell cycle arrest. The dipeptide identified by SMILI-NMR showed biological activity in a functional assay in yeast.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Espectroscopía de Resonancia Magnética/métodos , Bibliotecas de Moléculas Pequeñas , Bioensayo , Dipéptidos/química , Dipéptidos/farmacología , Modelos Moleculares , Saccharomyces cerevisiae/efectos de los fármacos , Tacrolimus/análogos & derivados , Tacrolimus/química , Proteínas de Unión a Tacrolimus/efectos de los fármacos
16.
J Pharm Pharmacol ; 60(12): 1687-94, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19000375

RESUMEN

Total triterpene acids (TTAs) isolated from Cornus officinalis Sieb., one of the herbs contained in Liuwei Dihuang decoction, were aimed at alleviating diabetic cardiomyopathy. We hypothesized that the benefits of TTAs may result from suppressing the endothelin-reactive oxidative species (ET-ROS) pathway in the myocardium. Diabetes was produced by a single injection of streptozotocin (STZ, 60 mg kg(-1), i.p.) in rats. Assessment of cardiac function, calcium handling proteins, endothelin-1 (ET-1) and redox system was conducted 8 weeks after STZ injection. Medication with TTAs (50 mg kg(-1), i.g.) was installed in the last 4 weeks. The compromised cardiac function was characterized by depressed contractility (LVSP and LV+dp/dt(max)) and relaxation (LVEDP and -LVdp/dt(min)) in association with hyperglycaemia (30.2 +/- 2.6 mmol L(-1)) in STZ-injected rats. Down-regulated expression of FKBP12.6 (calstabin 2), sarcoplasmic reticulum Ca(2+)-ATPase 2a (SERCA2a) and phospholamban (PLB) were also found. These changes occurred in connection with an increased ET-1, up-regulated mRNA of propreET-1 and endothelin converting enzyme (ECE), and a state of oxidant stress was found by increased malondialdehyde (MDA), decreased superoxide dismutase (SOD) and glutathione peroxidase (GSH-px) activity, and an enhanced activity and expression of inducible nitric oxide synthase (iNOS) in the diabetic myocardium. After 4 weeks of treatment with TTAs, these changes were alleviated dramatically despite a mild reduction in hyperglycaemia (26.9 +/- 3.4 mmol L(-1)). In conclusion, TTAs, as active ingredients of Liuwei Dihuang decoction, alleviated diabetic cardiomyopathy by normalizing the abnormality of FKBP12.6 and SERCA2a and ET-ROS pathway in the myocardium rather than by hypoglycaemic activity.


Asunto(s)
Cardiomiopatías/tratamiento farmacológico , Cornus/química , Complicaciones de la Diabetes/tratamiento farmacológico , Triterpenos/farmacología , Animales , Cardiomiopatías/etiología , Diabetes Mellitus Experimental/complicaciones , Endotelina-1/efectos de los fármacos , Endotelina-1/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Masculino , Miocardio/patología , Oxidación-Reducción/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Estreptozocina , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/metabolismo , Triterpenos/aislamiento & purificación
17.
Eur J Pharmacol ; 591(1-3): 211-8, 2008 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-18611397

RESUMEN

Downregulation of FKBP12.6 and sarcoplasmic reticulum Ca(2+) ATPase (SERCA2a) contributes to sudden cardiac death and heart failure. We aimed to test the hypothesis that (i) downregulation of FKBP12.6 and SERCA2a can be taken as molecular markers for drug interventions and (ii) such downregulation is produced by crosstalk between endothelin-reactive oxygen species and beta-adrenoceptors stimulation, mediated by hyperphosphorylation of protein kinase Cvarepsilon (PKCvarepsilon). Rat cardiomyocytes were incubated with isoproterenol (1 microM), endothelin-1 (0.1 microM) or hydrogen peroxide (10 microM) for 18 h, resulting in downregulation of mRNA and protein of FKBP12.6 and SERCA2a, as well as upregulation of PKCvarepsilon mRNA and phosphorylated PKCvarepsilon protein. These changes were reversed by an application of either propranolol (1 microM), endothelin receptor antagonist CPU0213 (1 microM) or vitamin E (1 microM). As indicated by the fluorescent dye Fluo3, diastolic [Ca(2+)](i) in rat ventricular myocytes was increased after incubation with isoproterenol (0.1 microM). The increased [Ca(2+)](i) in diastole was dramatically decreased by CPU0213. Thus, the downregulation of FKBP12.6 and SERCA2a, and hyperphosphorylation of PKCvarepsilon, appear to be related to crosstalk between over-activated endothelin-reactive oxygen species and a beta-adrenoceptor pathway. CPU0213 is beneficial in treating cardiac insufficiency and preventing cardiac arrhythmias possibly by normalizing hyperphosphorylation of PKCvarepsilon and abnormal FKBP12.6 and SERCA2a. The antioxidant activity of vitamin E was sufficient to normalize the levels of FKBP12.6 and SERCA2a and phosphorylation of PKCvarepsilon. Thus by testing with biomarkers FKBP12.6 and SERCA2a, we have shown that the endothelin receptor antagonist CPU0213 and the antioxidant vitamin E may relieve risk of lethal arrhythmias and heart failure by suppressing PKCvarepsilon.


Asunto(s)
Antagonistas de los Receptores de Endotelina , Regulación de la Expresión Génica/efectos de los fármacos , Pirazoles/farmacología , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Vitamina E/farmacología , Animales , Antioxidantes/farmacología , Biomarcadores Farmacológicos/metabolismo , Calcio/metabolismo , Muerte Súbita Cardíaca/prevención & control , Regulación hacia Abajo/efectos de los fármacos , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Fosforilación , Proteína Quinasa C-epsilon/efectos de los fármacos , Proteína Quinasa C-epsilon/metabolismo , Ratas , Ratas Sprague-Dawley , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/efectos de los fármacos , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo
18.
Eur J Immunol ; 37(7): 1730-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17523132

RESUMEN

The effect of tacrolimus (FK506) on down-regulation of IL-2 production by T cells is considered to be mainly responsible for its strong suppression of immunological events. In this study, we show that FK506 also has an affect on antigen presentation by antigen-presenting cells in vitro. FK506 was able to inhibit the presentation of endogenous MHC class II-restricted minor histocompatibility antigens in primary dendritic cells (DC) in vitro, but cyclosporine A (CsA) and rapamycin (RAP) were not. RNA interference (RNAi)-mediated reduction of endogenous FK506-binding protein (FKBP)51 expression resulted in a marked decrease in antigen presentation, suggesting that FKBP51 plays a role in endogenous MHC class II-restricted antigen presentation. Since our model used naturally expressed cytosolic antigens in primary DC, these effects might have been due to novel properties of the immunosuppressive drugs and may allow us to elucidate a new paradigm for the immunosuppressive mechanism of FK506.


Asunto(s)
Presentación de Antígeno/efectos de los fármacos , Antígenos de Histocompatibilidad Clase II/efectos de los fármacos , Inmunosupresores/farmacología , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Tacrolimus/farmacología , Animales , Western Blotting , Células COS , Chlorocebus aethiops , Ciclosporina/farmacología , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Citometría de Flujo , Expresión Génica/efectos de los fármacos , Antígenos de Histocompatibilidad Clase II/inmunología , Ratones , Ovalbúmina/inmunología , Interferencia de ARN , Sirolimus/farmacología , Proteínas de Unión a Tacrolimus/inmunología , Proteínas de Unión a Tacrolimus/metabolismo , Transfección
19.
Neuroscience ; 137(1): 231-9, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16289353

RESUMEN

BACKGROUND: The mechanisms underlying the neuroprotective effects of the immunosuppressant tacrolimus, observed in vivo, remain unclear. Here we quantify these effects in vitro, and evaluate the potential involvement of the glutamate and/or immunophilin FK506 binding protein 12 kDa in tacrolimus-induced neuroprotection. METHODS: Primary cultures of neurons and astrocytes from rat cerebral cortex were subjected to transient oxygen-glucose deprivation. Neuronal injury was evaluated by cell counting after immunostaining experiments, lactate dehydrogenase release and 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide reduction. The involvement of the immunophilin FK506 binding protein 12 kDa was explored using an anti-FK506 binding protein 12 kDa antibody, (3-3-pyridyl)-1-propyl(2 s)-1-(3,3-dimethyl-1,2-dioxopentyl)-2-pyrrolidine carboxylate and rapamycin. Extracellular glutamate and glutamate uptake were respectively measured by high performance liquid chromatography and l-[3H]glutamate incorporation. RESULTS: When added during either oxygen-glucose deprivation or reoxygenation, FK506 (50-500 pM) offered significant neuroprotection. During oxygen-glucose deprivation, it was able to reverse the oxygen-glucose deprivation-induced increase in extracellular glutamate and decrease in glutamate uptake and this effect was reversed in the presence of threo-3-methyl glutamate, a specific inhibitor of glutamate transporter-1. Blocking FK506 binding protein 12 kDa inhibited the neuroprotection induced by tacrolimus added during either oxygen-glucose deprivation or reoxygenation. Tacrolimus-induced neuroprotection was also reversed in the presence of rapamycin, an immunosuppressant FK506 binding protein 12 kDa ligand devoid of neuroprotective properties and (3-3-pyridyl)-1-propyl(2 s)-1-(3,3-dimethyl-1,2-dioxopentyl)-2-pyrrolidine carboxylate, a non-immunosuppressant ligand of FK506 binding protein 12 kDa, exerteing neuroprotective effects. CONCLUSION: The beneficial effects of tacrolimus during in vitro ischemia/reperfusion seem to indicate the restoration of a glutamate transporter-1-mediated activity and could be mediated by a FK506 binding protein 12 kDa pathway.


Asunto(s)
Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/tratamiento farmacológico , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Tacrolimus/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Cromatografía Líquida de Alta Presión , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Glucosa/deficiencia , Ácido Glutámico/metabolismo , Inmunohistoquímica , Técnicas In Vitro , Neuronas/patología , Ratas , Ratas Wistar , Proteínas de Unión a Tacrolimus/metabolismo
20.
Mol Endocrinol ; 19(11): 2859-70, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16020481

RESUMEN

To investigate the function of dimerization of the TRH receptor, a controlled dimerization system was developed. A variant FK506 binding protein (FKBP) domain was fused to the receptor C terminus and dimerization induced by incubating cells with dimeric FKBP ligand, AP20187. The TRH receptor-fusion bound hormone and signaled normally. Addition of dimerizer to cells expressing the receptor-FKBP fusion dramatically increased the fraction of receptor running as dimer on SDS-PAGE. AP20187 caused dimerization in a time- and concentration-dependent manner, acting within 1 min. Dimerizer had no effect on TRH receptors lacking the FKBP domain, and its effects were blocked by excess monomeric FKBP ligand. AP20187-induced dimerization did not cause receptor phosphorylation, inositol phosphate production, or ERK1/2 activation, and dimerizer did not alter signaling by TRH. Induced dimerization did, however, alter TRH receptor trafficking. TRH promoted greater receptor internalization in cells treated with AP20187 but not monomeric ligand, based on loss of surface binding sites and immunostaining. Dimerization increased the rate of internalization of TRH receptors and decreased the apparent rate of receptor recycling. AP20187 enhanced the small amount of TRH-induced receptor internalization when the receptor-FKBP fusion protein was expressed in cells lacking beta-arrestins. The results show that controlled dimerization of the TRH receptor potentiates hormone-induced receptor trafficking.


Asunto(s)
Receptores de Hormona Liberadora de Tirotropina/metabolismo , Hormona Liberadora de Tirotropina/metabolismo , Animales , Arrestinas/metabolismo , Células CHO , Calcio/metabolismo , Membrana Celular/química , Cricetinae , Dimerización , Activación Enzimática/efectos de los fármacos , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Transporte de Proteínas/efectos de los fármacos , Receptores de Hormona Liberadora de Tirotropina/análisis , Receptores de Hormona Liberadora de Tirotropina/efectos de los fármacos , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/efectos de los fármacos , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal/efectos de los fármacos , Tacrolimus/análogos & derivados , Tacrolimus/farmacología , Proteínas de Unión a Tacrolimus/análisis , Proteínas de Unión a Tacrolimus/efectos de los fármacos , Proteínas de Unión a Tacrolimus/metabolismo , Hormona Liberadora de Tirotropina/farmacología , Fosfolipasas de Tipo C/metabolismo , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...