Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Cell Death Dis ; 12(6): 602, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112753

RESUMEN

FKBP4 belongs to the family of immunophilins, which serve as a regulator for steroid receptor activity. Thus, FKBP4 has been recognized to play a critical role in several hormone-dependent cancers, including breast and prostate cancer. However, there is still no research to address the role of FKBP4 on lung adenocarcinoma (LUAD) progression. We found that FKBP4 expression was elevated in LUAD samples and predicted significantly shorter overall survival based on TCGA and our cohort of LUAD patients. Furthermore, FKBP4 robustly increased the proliferation, metastasis, and invasion of LUAD in vitro and vivo. Mechanistic studies revealed the interaction between FKBP4 and IKK kinase complex. We found that FKBP4 potentiated IKK kinase activity by interacting with Hsp90 and IKK subunits and promoting Hsp90/IKK association. Also, FKBP4 promotes the binding of IKKγ to IKKß, which supported the facilitation role in IKK complex assembly. We further identified that FKBP4 TPR domains are essential for FKBP4/IKK interaction since its association with Hsp90 is required. In addition, FKBP4 PPIase domains are involved in FKBP4/IKKγ interaction. Interestingly, the association between FKBP4 and Hsp70/RelA favors the transport of RelA toward the nucleus. Collectively, FKBP4 integrates FKBP4/Hsp90/IKK with FKBP4/Hsp70/RelA complex to potentiate the transcriptional activity and nuclear translocation of NF-κB, thereby promoting LUAD progression. Our findings suggest that FKBP4 may function as a prognostic biomarker of LUAD and provide a newly mechanistic insight into modulating IKK/NF-κB signaling.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Neoplasias Pulmonares/patología , Proteínas de Unión a Tacrolimus/fisiología , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Células HEK293 , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Quinasa I-kappa B/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , FN-kappa B/metabolismo , Transducción de Señal/fisiología , Factor de Transcripción ReIA/metabolismo
2.
J Clin Endocrinol Metab ; 106(1): 26-41, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32617576

RESUMEN

CONTEXT: Preeclampsia is a leading cardiovascular complication in pregnancy lacking effective diagnostic and treatment strategies. OBJECTIVE: To investigate the diagnostic and therapeutic target potential of the angiogenesis proteins, FK506-binding protein like (FKBPL) and CD44. DESIGN AND INTERVENTION: FKBPL and CD44 plasma concentration or placental expression were determined in women pre- or postdiagnosis of preeclampsia. Trophoblast and endothelial cell function was assessed following mesenchymal stem cell (MSC) treatment and in the context of FKBPL signaling. SETTINGS AND PARTICIPANTS: Human samples prediagnosis (15 and 20 weeks of gestation; n ≥ 57), or postdiagnosis (n = 18 for plasma; n = 4 for placenta) of preeclampsia were used to determine FKBPL and CD44 levels, compared to healthy controls. Trophoblast or endothelial cells were exposed to low/high oxygen, and treated with MSC-conditioned media (MSC-CM) or a FKBPL overexpression plasmid. MAIN OUTCOME MEASURES: Preeclampsia risk stratification and diagnostic potential of FKBPL and CD44 were investigated. MSC treatment effects and FKBPL-CD44 signaling in trophoblast and endothelial cells were assessed. RESULTS: The CD44/FKBPL ratio was reduced in placenta and plasma following clinical diagnosis of preeclampsia. At 20 weeks of gestation, a high plasma CD44/FKBPL ratio was independently associated with the 2.3-fold increased risk of preeclampsia (odds ratio = 2.3, 95% confidence interval [CI] 1.03-5.23, P = 0.04). In combination with high mean arterial blood pressure (>82.5 mmHg), the risk further increased to 3.9-fold (95% CI 1.30-11.84, P = 0.016). Both hypoxia and MSC-based therapy inhibited FKBPL-CD44 signaling, enhancing cell angiogenesis. CONCLUSIONS: The FKBPL-CD44 pathway appears to have a central role in the pathogenesis of preeclampsia, showing promising utilities for early diagnostic and therapeutic purposes.


Asunto(s)
Receptores de Hialuranos/fisiología , Trasplante de Células Madre Mesenquimatosas , Preeclampsia , Proteínas de Unión a Tacrolimus/fisiología , Adulto , Biomarcadores/análisis , Estudios de Casos y Controles , Células Cultivadas , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Receptores de Hialuranos/análisis , Receptores de Hialuranos/genética , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/fisiología , Terapia Molecular Dirigida/métodos , Neovascularización Patológica/diagnóstico , Neovascularización Patológica/genética , Neovascularización Patológica/terapia , Preeclampsia/diagnóstico , Preeclampsia/genética , Preeclampsia/terapia , Embarazo , Pronóstico , Medición de Riesgo , Transducción de Señal/genética , Proteínas de Unión a Tacrolimus/análisis , Proteínas de Unión a Tacrolimus/genética , Adulto Joven
3.
Biochem Soc Trans ; 47(6): 1815-1831, 2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31754722

RESUMEN

FKBP51 and FKBP52 are two iconic members of the family of peptidyl-prolyl-(cis/trans)-isomerases (EC: 5.2.1.8), which comprises proteins that catalyze the cis/trans isomerization of peptidyl-prolyl peptide bonds in unfolded and partially folded polypeptide chains and native state proteins. Originally, both proteins have been studied as molecular chaperones belonging to the steroid receptor heterocomplex, where they were first discovered. In addition to their expected role in receptor folding and chaperoning, FKBP51 and FKBP52 are also involved in many biological processes, such as signal transduction, transcriptional regulation, protein transport, cancer development, and cell differentiation, just to mention a few examples. Recent studies have revealed that both proteins are subject of post-translational modifications such as phosphorylation, SUMOlyation, and acetylation. In this work, we summarize recent advances in the study of these immunophilins portraying them as scaffolding proteins capable to organize protein heterocomplexes, describing some of their antagonistic properties in the physiology of the cell, and the putative regulation of their properties by those post-translational modifications.


Asunto(s)
Procesamiento Proteico-Postraduccional , Proteínas de Unión a Tacrolimus/fisiología , Acetilación , Humanos , Fosforilación , Unión Proteica , Transporte de Proteínas , Sumoilación , Proteínas de Unión a Tacrolimus/metabolismo , Factores de Transcripción/metabolismo
4.
Psychoneuroendocrinology ; 96: 118-125, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29936334

RESUMEN

Chronic inflammation caused by HIV infection may lead to deficient glucocorticoid (GC) signaling predisposing people living with HIV to depression and other psychiatric disorders linked to GC resistance. We hypothesized that comorbid HIV and depressive symptoms in women would synergistically associate with deficits in GC signaling. This cross-sectional study used samples obtained from the Women's Interagency HIV Study (WIHS). The Centers for Epidemiological Studies (CES-D) was used to define depression in four groups of women from the Women's Interagency HIV Study (WIHS): 1) HIV-negative, non-depressed (n = 37); 2) HIV-negative, depressed (n = 34); 3) HIV-positive, non-depressed (n = 38); and 4) HIV-positive, depressed (n = 38). To assess changes in GC signaling from peripheral blood mononuclear cells (PBMCs), we examined baseline and dexamethasone (Dex)-stimulated changes in the expression of the GC receptor (GR, gene: Nr3c1) and its negative regulator Fkbp5 via quantitative RT-PCR. GR sensitivity was evaluated in vitro by assessing the Dex inhibition of lipopolysaccharide (LPS)-stimulated IL-6 and TNF-α levels. Depressive symptoms and HIV serostatus were independently associated with elevated baseline expression of Fkbp5 and Nr3c1. Depressive symptoms, but not HIV status, was independently associated with reduced LPS-induced release of IL-6. Counter to predictions, there was no interactive association of depressive symptoms and HIV on any outcome. Comorbid depressive symptoms with HIV infection were associated with a gene expression and cytokine profile similar to that of healthy control women, a finding that may indicate further disruptions in disease adaptation.


Asunto(s)
Depresión/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Adulto , Estudios Transversales , Depresión/virología , Dexametasona/farmacología , Femenino , Glucocorticoides/metabolismo , Glucocorticoides/fisiología , VIH/patogenicidad , Infecciones por VIH/complicaciones , Infecciones por VIH/psicología , Humanos , Interleucina-6 , Errores Innatos del Metabolismo , Escalas de Valoración Psiquiátrica , Receptores de Glucocorticoides/deficiencia , Receptores de Glucocorticoides/fisiología , Transducción de Señal/fisiología , Proteínas de Unión a Tacrolimus/fisiología , Factor de Necrosis Tumoral alfa
5.
Nucleic Acids Res ; 46(5): 2459-2478, 2018 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-29361176

RESUMEN

FK506 binding proteins (FKBPs) catalyze the interconversion of cis-trans proline conformers in proteins. Importantly, FK506 drugs have anti-cancer and neuroprotective properties, but the effectors and mechanisms underpinning these properties are not well understood because the cellular function(s) of most FKBP proteins are unclear. FKBP25 is a nuclear prolyl isomerase that interacts directly with nucleic acids and is associated with several DNA/RNA binding proteins. Here, we show the catalytic FKBP domain binds microtubules (MTs) directly to promote their polymerization and stabilize the MT network. Furthermore, FKBP25 associates with the mitotic spindle and regulates entry into mitosis. This interaction is important for mitotic spindle dynamics, as we observe increased chromosome instability in FKBP25 knockdown cells. Finally, we provide evidence that FKBP25 association with chromatin is cell-cycle regulated by Protein Kinase C phosphorylation. This disrupts FKBP25-DNA contacts during mitosis while maintaining its interaction with the spindle apparatus. Collectively, these data support a model where FKBP25 association with chromatin and MTs is carefully choreographed to ensure faithful genome duplication. Additionally, they highlight that FKBP25 is a MT-associated FK506 receptor and potential therapeutic target in MT-associated diseases.


Asunto(s)
Ciclo Celular , Microtúbulos/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Línea Celular , ADN/metabolismo , Inestabilidad Genómica , Humanos , Mitosis , Isomerasa de Peptidilprolil/fisiología , Fosforilación , Polimerizacion , Proteína Quinasa C/metabolismo , Proteínas de Unión a Tacrolimus/fisiología
6.
J Biol Chem ; 289(38): 26263-26276, 2014 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-25104352

RESUMEN

Hsp90 binding immunophilins FKBP51 and FKBP52 modulate steroid receptor trafficking and hormone-dependent biological responses. With the purpose to expand this model to other nuclear factors that are also subject to nuclear-cytoplasmic shuttling, we analyzed whether these immunophilins modulate NF-κB signaling. It is demonstrated that FKBP51 impairs both the nuclear translocation rate of NF-κB and its transcriptional activity. The inhibitory action of FKBP51 requires neither the peptidylprolyl-isomerase activity of the immunophilin nor its association with Hsp90. The TPR domain of FKBP51 is essential. On the other hand, FKBP52 favors the nuclear retention time of RelA, its association to a DNA consensus binding sequence, and NF-κB transcriptional activity, the latter effect being strongly dependent on the peptidylprolyl-isomerase activity and also on the TPR domain of FKBP52, but its interaction with Hsp90 is not required. In unstimulated cells, FKBP51 forms endogenous complexes with cytoplasmic RelA. Upon cell stimulation with phorbol ester, the NF-κB soluble complex exchanges FKBP51 for FKBP52, and the NF-κB biological effect is triggered. Importantly, FKBP52 is functionally recruited to the promoter region of NF-κB target genes, whereas FKBP51 is released. Competition assays demonstrated that both immunophilins antagonize one another, and binding assays with purified proteins suggest that the association of RelA and immunophilins could be direct. These observations suggest that the biological action of NF-κB in different cell types could be positively regulated by a high FKBP52/FKBP51 expression ratio by favoring NF-κB nuclear retention, recruitment to the promoter regions of target genes, and transcriptional activity.


Asunto(s)
Proteínas de Unión a Tacrolimus/fisiología , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Regiones Promotoras Genéticas , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Ratas , Receptores de Glucocorticoides/metabolismo , Transcripción Genética , Activación Transcripcional
7.
Mol Endocrinol ; 28(8): 1265-75, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24933247

RESUMEN

Glucocorticoid receptor-α (GRα) and peroxisome proliferator-activated receptor-γ (PPARγ) are critical regulators of adipogenic responses. We have shown that FK506-binding protein 51 (FKBP51) represses the Akt-p38 kinase pathway to reciprocally inhibit GRα but stimulate PPARγ by targeting serine 112 (PPARγ) and serines 220 and 234 (GRα). Here, this mechanism is shown to be essential for GRα and PPARγ control of cellular adipogenesis. In 3T3-L1 cells, FKBP51 was a prominent marker of the differentiated state and knockdown of FKBP51 showed reduced lipid accumulation and expression of adipogenic genes. Compared with wild-type (WT), FKBP51 knockout (51KO) mouse embryonic fibroblasts (MEFs) showed dramatic resistance to differentiation, with almost no lipid accumulation and greatly reduced adipogenic gene expression. These features were rescued by reexpression of FKBP51 in 51KO cells. 51KO MEFs exhibited reduced fatty acid synthase activity, increased sensitivity to GRα-induced lipolysis, and reduced PPARγ activity at adipogenic genes (adiponectin, CD36, and perilipin) but elevated GRα transrepression at these same genes. A p38 kinase inhibitor increased lipid content in WT cells and also restored lipid levels in 51KO cells, showing that elevated p38 kinase activity is a major contributor to adipogenic resistance in the 51KO cells. In 51KO cells, the S112A mutant of PPARγ and the triple S212A/S220A/S234A mutant of GRα both increased lipid accumulation, identifying these residues as targets of the FKBP51/p38 axis. Our combined investigations have uncovered FKBP51 as a key regulator of adipogenesis via the Akt-p38 pathway and as a potential target in the treatment of obesity and related disorders.


Asunto(s)
Adipogénesis , PPAR gamma/metabolismo , Procesamiento Proteico-Postraduccional , Receptores de Glucocorticoides/metabolismo , Proteínas de Unión a Tacrolimus/fisiología , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología , Células 3T3-L1 , Animales , Regulación de la Expresión Génica , Metabolismo de los Lípidos , Sistema de Señalización de MAP Quinasas , Redes y Vías Metabólicas , Ratones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo
8.
Mol Endocrinol ; 28(8): 1254-64, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24933248

RESUMEN

FK506-binding protein 51 (FKBP51) is a negative regulator of glucocorticoid receptor-α (GRα), although the mechanism is unknown. We show here that FKBP51 is also a chaperone to peroxisome proliferator-activated receptor-γ (PPARγ), which is essential for activity, and uncover the mechanism underlying this differential regulation. In COS-7 cells, FKBP51 overexpression reduced GRα activity at a glucocorticoid response element-luciferase reporter, while increasing PPARγ activity at a peroxisome proliferator response element reporter. Conversely, FKBP51-deficient (knockout) (51KO) mouse embryonic fibroblasts (MEFs) showed elevated GRα but reduced PPARγ activities compared with those in wild-type MEFs. Phosphorylation is known to exert a similar pattern of reciprocal modulation of GRα and PPARγ. Knockdown of FKBP51 in 3T3-L1 preadipocytes increased phosphorylation of PPARγ at serine 112, a phospho-residue that inhibits activity. In 51KO cells, elevated phosphorylation of GRα at serines 220 and 234, phospho-residues that promote activity, was observed. Because FKBP51 is an essential chaperone to the Akt-specific phosphatase PH domain leucine-rich repeat protein phosphatase, Akt signaling was investigated. Elevated Akt activation and increased activation of p38 kinase, a downstream target of Akt that phosphorylates GRα and PPARγ, were seen in 51KO MEFs, causing activation and inhibition, respectively. Inactivation of p38 with PD169316 reversed the effects of FKBP51 deficiency on GRα and PPARγ activities and reduced PPARγ phosphorylation. Last, loss of FKBP51 caused a shift of PPARγ from cytoplasm to nucleus, as previously shown for GRα. A model is proposed in which FKBP51 loss reciprocally regulates GRα and PPARγ via 2 complementary mechanisms: activation of Akt-p38-mediated phosphorylation and redistribution of the receptors to the nucleus for direct targeting by p38.


Asunto(s)
PPAR gamma/metabolismo , Receptores de Glucocorticoides/metabolismo , Proteínas de Unión a Tacrolimus/fisiología , Células 3T3-L1 , Animales , Células COS , Núcleo Celular/metabolismo , Chlorocebus aethiops , Insulina/fisiología , Sistema de Señalización de MAP Quinasas , Ratones , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo
9.
Psychoneuroendocrinology ; 45: 67-76, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24845178

RESUMEN

BACKGROUND: Stress is a critical risk factor affecting both the development of and the relapse to drug addictions. Drug addictions are caused by genetic, environmental and drug-induced factors. The objective of this hypothesis-driven association study was to determine if genetic variants in stress-related genes are associated with heroin addiction. METHODS: 112 selected genetic variants in 26 stress-related genes were genotyped in 852 case subjects and 238 controls of predominantly European ancestry. The case subjects are former heroin addicts with a history of at least one year of daily multiple uses of heroin, treated at a methadone maintenance treatment program (MMTP). The two most promising SNPs were subsequently tested in an African-American sample comprising of 314 cases and 208 control individuals. RESULTS: Nineteen single nucleotide polymorphisms (SNPs) in 9 genes (AVP, AVPR1A, CRHR1, CRHR2, FKBP5, GAL, GLRA1, NPY1R and NR3C2) showed nominally significant association with heroin addiction. The associations of two FKBP5 SNPs that are part of one haplotype block, rs1360780 (intron 2) and rs3800373 (the 3' untranslated region), remained significant after correction for multiple testing (Pcorrected=0.03; OR=2.35, Pcorrected=0.0018; OR=2.85, respectively). The two SNPs also showed nominally significant association (P<0.05) with heroin addiction in an independent African-American cohort. FKBP5 is a co-chaperone that regulates glucocorticoid sensitivity. These FKBP5 SNPs were previously associated with diverse affective disorders and showed functional differences in gene expression and stress response. This study also supports our and others' previous reports of association of the GAL SNP rs694066 and the AVPR1A SNPs rs11174811, rs1587097 and rs10784339 with heroin and general drug addiction, respectively. CONCLUSIONS: This study suggests that variations in the FKBP5 gene contribute to the development of opiate addiction by modulating the stress response. These findings may enhance the understanding of the interaction between stress and heroin addiction.


Asunto(s)
Dependencia de Heroína/genética , Estrés Psicológico/genética , Proteínas de Unión a Tacrolimus/fisiología , Adulto , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Genotipo , Haplotipos , Dependencia de Heroína/epidemiología , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Estrés Psicológico/epidemiología , Proteínas de Unión a Tacrolimus/genética
10.
Cell Cycle ; 12(15): 2366-70, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23839048

RESUMEN

FK506-binding proteins (FKBP) belong to the immunophilin family and are best known for their ability to enable the immunosuppressive properties of FK506 and rapamycin. For rapamycin, this is achieved by inducing inhibitory ternary complexes with the kinase mTOR. The essential accessory protein for this gain-of-function was thought to be FKBP12. We recently showed that this view might be too restricted, since larger FK506-binding proteins can functionally substitute for FKBP12 in mammalian cells. Recent studies have also shown that FK506-binding proteins can modulate Akt-mTOR signaling in the absence of rapamycin. Here we discuss the role of FK506-binding proteins for the mechanism of rapamycin as well as their intrinsic actions on the Akt/mTOR pathway.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión a Tacrolimus/fisiología , Animales , Humanos , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
11.
Cell Mol Life Sci ; 70(18): 3243-75, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23224428

RESUMEN

From 5 to 12 FK506-binding proteins (FKBPs) are encoded in the genomes of disparate marine organisms, which appeared at the dawn of evolutionary events giving rise to primordial multicellular organisms with elaborated internal body plan. Fifteen FKBPs, several FKBP-like proteins and some splicing variants of them are expressed in humans. Human FKBP12 and some of its paralogues bind to different macrocyclic antibiotics such as FK506 or rapamycin and their derivatives. FKBP12/(macrocyclic antibiotic) complexes induce diverse pharmacological activities such as immunosuppression in humans, anticancerous actions and as sustainers of quiescence in certain organisms. Since the FKBPs bind to various assemblies of proteins and other intracellular components, their complexes with the immunosuppressive drugs may differentially perturb miscellaneous cellular functions. Sequence-structure relationships and pharmacological profiles of diverse FKBPs and their involvement in crucial intracellular signalization pathways and modulation of cryptic intercellular communication networks were discussed.


Asunto(s)
Proteínas de Unión a Tacrolimus/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Antibacterianos/uso terapéutico , Antineoplásicos/uso terapéutico , Comunicación Celular , Núcleo Celular/metabolismo , Humanos , Terapia de Inmunosupresión , Ligandos , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Sirolimus/uso terapéutico , Proteínas de Unión a Tacrolimus/genética
12.
Cardiovasc Ther ; 30(2): 109-16, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21176119

RESUMEN

It was first proposed that cyclic ADP-ribose (cADPR) could activate ryanodine receptors (RyR) in 1991. Following a subsequent report that cADPR could activate cardiac RyR (RyR2) reconstituted into artificial membranes and stimulate Ca(2+) -release from isolated cardiac SR, there has been a steadily mounting stockpile of publications proclaiming the physiological and pathophysiological importance of cADPR in the cardiovascular system. It was only 2 years earlier, in 1989, that cADPR was first identified as the active metabolite of nicotinamide adenine dinucleotide (NAD), responsible for triggering the release of Ca(2+) from crude homogenates of sea urchin eggs. Twenty years later, can we boast of being any closer to unraveling the mechanisms by which cADPR modulates intracellular Ca(2+) -release? This review sets out to examine the mechanisms underlying the effects of cADPR and ask whether cADPR is an important signaling molecule in the heart.


Asunto(s)
ADP-Ribosa Cíclica/fisiología , Corazón/fisiología , Óvulo/fisiología , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Erizos de Mar/fisiología , Animales , Señalización del Calcio/fisiología , ADP-Ribosa Cíclica/metabolismo , Fertilización , Humanos , NADP/análogos & derivados , NADP/metabolismo , Unión Proteica , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Proteínas de Unión a Tacrolimus/fisiología
13.
Circ Res ; 109(3): 281-90, 2011 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-21659649

RESUMEN

RATIONALE: Mutations in the cardiac type 2 ryanodine receptor (RyR2) have been linked to catecholaminergic polymorphic ventricular tachycardia (CPVT). CPVT-associated RyR2 mutations cause fatal ventricular arrhythmias in young individuals during ß-adrenergic stimulation. OBJECTIVE: This study sought to determine the effects of a novel RyR2-G230C mutation and whether this mutation and RyR2-P2328S alter the sensitivity of the channel to luminal calcium (Ca(2+)). METHODS AND RESULTS: Functional characterizations of recombinant human RyR2-G230C channels were performed under conditions mimicking stress. Human RyR2 mutant channels were generated by site-directed mutagenesis and heterologously expressed in HEK293 cells together with calstabin2. RyR2 channels were measured to examine the regulation of the channels by cytosolic versus luminal sarcoplasmic reticulum Ca(2+). A 50-year-old white man with repeated syncopal episodes after exercise had a cardiac arrest and harbored the mutation RyR2-G230C. cAMP-dependent protein kinase-phosphorylated RyR2-G230C channels exhibited a significantly higher open probability at diastolic Ca(2+) concentrations, associated with a depletion of calstabin2. The luminal Ca(2+) sensitivities of RyR2-G230C and RyR2-P2328S channels were WT-like. CONCLUSIONS: The RyR2-G230C mutant exhibits similar biophysical defects compared with previously characterized CPVT mutations: decreased binding of the stabilizing subunit calstabin2 and a leftward shift in the Ca(2+) dependence for activation under conditions that simulate exercise, consistent with a "leaky" channel. Both RyR2-G230C and RyR2-P2328S channels exhibit normal luminal Ca(2+) activation. Thus, diastolic sarcoplasmic reticulum Ca(2+) leak caused by reduced calstabin2 binding and a leftward shift in the Ca(2+) dependence for activation by diastolic levels of cytosolic Ca(2+) is a common mechanism underlying CPVT.


Asunto(s)
Calcio/fisiología , Muerte Súbita Cardíaca , Canal Liberador de Calcio Receptor de Rianodina/genética , Canal Liberador de Calcio Receptor de Rianodina/fisiología , Taquicardia Ventricular/fisiopatología , Catecolaminas/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Citosol/fisiología , Diástole/fisiología , Electrocardiografía , Células HEK293 , Humanos , Activación del Canal Iónico/fisiología , Masculino , Persona de Mediana Edad , Mutagénesis Sitio-Dirigida , Fenotipo , Mutación Puntual , Proteínas Recombinantes/genética , Taquicardia Ventricular/diagnóstico , Taquicardia Ventricular/genética , Proteínas de Unión a Tacrolimus/fisiología
14.
Cancer Cell ; 19(6): 792-804, 2011 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-21620777

RESUMEN

Alteration of the PTEN/PI3K pathway is associated with late-stage and castrate-resistant prostate cancer (CRPC). However, how PTEN loss is involved in CRPC development is not clear. Here, we show that castration-resistant growth is an intrinsic property of Pten null prostate cancer (CaP) cells, independent of cancer development stage. PTEN loss suppresses androgen-responsive gene expressions by modulating androgen receptor (AR) transcription factor activity. Conditional deletion of Ar in the epithelium promotes the proliferation of Pten null cancer cells, at least in part, by downregulating the androgen-responsive gene Fkbp5 and preventing PHLPP-mediated AKT inhibition. Our findings identify PI3K and AR pathway crosstalk as a mechanism of CRPC development, with potentially important implications for CaP etiology and therapy.


Asunto(s)
Orquiectomía , Fosfohidrolasa PTEN/fisiología , Neoplasias de la Próstata/patología , Animales , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Proteína Potenciadora del Homólogo Zeste 2 , Femenino , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Masculino , Ratones , Proteínas Nucleares/fisiología , Fosfoproteínas Fosfatasas/fisiología , Fosforilación , Complejo Represivo Polycomb 2 , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/prevención & control , Neoplasias de la Próstata/terapia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Receptores Androgénicos/fisiología , Proteínas de Unión a Tacrolimus/fisiología
15.
J Invest Dermatol ; 131(7): 1477-85, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21430698

RESUMEN

Immunophilin, FK506-binding protein 12 (FK506BP), is a receptor protein for the immunosuppressive drug FK506 by the FK506BP/FK506 complex. However, the precise function of FK506BP in inflammatory diseases remains unclear. Therefore, we examined the protective effects of FK506BP on atopic dermatitis (AD) in tumor necrosis factor-α (TNF-α)/interferon-γ (IFN-γ)-induced HaCaT cells and 2,4-dinitrofluorobenzene-induced AD-like dermatitis in Nishiki-nezumi Cinnamon/Nagoya (NC/Nga) mice using a cell-permeable PEP-1-FK506BP. Transduced PEP-1-FK506BP significantly inhibited the expression of cytokines, as well as the activation of NF-κB and mitogen-activated protein kinase (MAPK) in TNF-α/IFN-γ-induced HaCaT cells. Furthermore, topical application of PEP-1-FK506BP to NC/Nga mice markedly inhibited AD-like dermatitis as determined by a histological examination and assessment of serum IgE levels, as well as cytokines and chemokines. These results indicate that PEP-1-FK506BP inhibits NF-κB and MAPK activation in cells and AD-like skin lesions by reducing the expression levels of cytokines and chemokines, thus suggesting that PEP-1-FK506BP may be a potential therapeutic agent for AD.


Asunto(s)
Cisteamina/análogos & derivados , Dermatitis Atópica/tratamiento farmacológico , Péptidos/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas de Unión a Tacrolimus/uso terapéutico , Animales , Cisteamina/uso terapéutico , Dermatitis Atópica/etiología , Modelos Animales de Enfermedad , Inmunoglobulina E/sangre , Interferón gamma/antagonistas & inhibidores , Interferón gamma/genética , Sistema de Señalización de MAP Quinasas , Ratones , FN-kappa B/metabolismo , Péptidos/genética , Péptidos/fisiología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/fisiología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/genética
16.
Biochem Soc Trans ; 39(2): 663-8, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21428958

RESUMEN

FKBPs (FK506-binding proteins) have long been recognized as key regulators of the response to immunosuppressant drugs and as co-chaperones of steroid receptor complexes. More recently, evidence has emerged suggesting that this diverse protein family may also represent cancer biomarkers owing to their roles in cancer progression and response to treatment. FKBPL (FKBP-like) is a novel FKBP with roles in GR (glucocorticoid receptor), AR (androgen receptor) and ER (oestrogen receptor) signalling. FKBPL binds Hsp90 (heat-shock protein 90) and modulates translocation, transcriptional activation and phosphorylation of these steroid receptors. It has been proposed as a novel prognostic and predictive biomarker, where high levels predict for increased recurrence-free survival in breast cancer patients and enhanced sensitivity to endocrine therapy. Since this protein family has roles in a plethora of signalling pathways, its members represent novel prognostic markers and therapeutic targets for cancer diagnosis and treatment.


Asunto(s)
Biomarcadores de Tumor/fisiología , Inmunofilinas/fisiología , Neoplasias/diagnóstico , Proteínas de Unión a Tacrolimus/fisiología , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Humanos , Inmunofilinas/genética , Inmunofilinas/metabolismo , Modelos Biológicos , Neoplasias/genética , Neoplasias/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/fisiología , Transducción de Señal/fisiología , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/metabolismo
17.
Biochem Biophys Res Commun ; 406(2): 305-9, 2011 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-21320469

RESUMEN

The phosphatase of regenerating liver-3 (PRL-3) is a member of protein tyrosine phosphatases and whose deregulation is implicated in tumorigenesis and metastasis of many cancers. However, the underlying mechanism by which PRL-3 is regulated is not known. In this study, we identified the peptidyl prolyl cis/trans isomerase FK506-binding protein 38 (FKBP38) as an interacting protein of PRL-3 using a yeast two-hybrid system. FKBP38 specifically binds to PRL-3 in vivo, and that the N-terminal region of FKBP38 is crucial for binding with PRL-3. FKBP38 overexpression reduces endogenous PRL-3 expression levels, whereas the depletion of FKBP38 by siRNA increases the level of PRL-3 protein. Moreover, FKBP38 promotes degradation of endogenous PRL-3 protein via protein-proteasome pathway. Furthermore, FKBP38 suppresses PRL-3-mediated p53 activity and cell proliferation. These results demonstrate that FKBP38 is a novel regulator of the oncogenic protein PRL-3 abundance and that alteration in the stability of PRL-3 can have a dramatic impact on cell proliferation. Thus, FKBP38 may play a critical role in tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas de Unión a Tacrolimus/fisiología , Línea Celular Tumoral , Proliferación Celular , Estabilidad de Enzimas , Células HEK293 , Humanos , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas de Unión a Tacrolimus/genética
18.
Br J Cancer ; 104(1): 19-23, 2011 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-21119664

RESUMEN

FK506 binding protein 51 (FKBP51, also called FKBP5) belongs to a family of immunophilins, FK506 binding proteins (FKBPs). Members of this family are targets for drugs such as rapamycin and cyclosporine. Although FKBP5 shares characteristics with other FKBPs, it also has unique features, especially its role in the regulation of multiple signalling pathways and in tumourigenesis and chemoresistance. In this review, we will focus on the recently discovered role of FKBP5 in cancer aetiology and response to antineoplastic therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos , Proteínas HSP90 de Choque Térmico/fisiología , Neoplasias/tratamiento farmacológico , Neoplasias/etiología , Proteínas de Unión a Tacrolimus/fisiología , Humanos , Transducción de Señal
19.
Hum Mol Genet ; 19(14): 2804-16, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20442138

RESUMEN

The tyrosine kinase receptor RET51 is expressed in distinct families of neurons where it promotes different functions. FKBP52 is an immunophilin with neuroprotective effects on different kinds of neurons. In this paper, we demonstrate that RET51 activation by both glial cell line-derived neurotrophic factor (GDNF) and NGF triggers the formation of RET51/FKBP52 complex. The substitution of the tyrosine 905 of RET51, a key residue phosphorylated by both GDNF and NGF, disrupts the RET51/FKBP52 complex. NGF and GDNF have a functional role in dopaminergic (DA) neurons where RET51 and FKBP52 are expressed with a yet undefined function. To clarify if RET51/FKBP52 complex should exert its function in DA neurons, we used an indirect approach by screening the genes encoding for RET51 and FKBP52 in a group of 30 Parkinson's disease patients. The degeneration of DA neurons is the main feature of PD, which is associated to a complex multifactorial aetiology combining environmental, age-related and genetic factors. We found a compound heterozygous carrying two mutations in RET and FKBP52 that are sufficient to disrupt the RET51/FKBP52 complex, indicating its potential role in PD.


Asunto(s)
Enfermedad de Parkinson/etiología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Proteínas Proto-Oncogénicas c-ret/fisiología , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas de Unión a Tacrolimus/fisiología , Adulto , Línea Celular , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/fisiología , Factor de Crecimiento Nervioso/farmacología , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Fosforilación , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas/genética , Mapeo de Interacción de Proteínas , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-ret/química , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/genética , Adulto Joven
20.
Endocrinology ; 151(7): 3225-36, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20427484

RESUMEN

Although FK506-binding protein 52 (FKBP52) is an established positive regulator of glucocorticoid receptor (GR) activity, an in vivo role for FKBP52 in glucocorticoid control of metabolism has not been reported. To address this question, FKBP52(+/-) mice were placed on a high-fat (HF) diet known to induce obesity, hepatic steatosis, and insulin resistance. Tissue profiling of wild-type mice showed high levels of FKBP52 in the liver but little to no expression in muscle or adipose tissue, predicting a restricted pattern of FKBP52 effects on metabolism. In response to HF, FKBP52(+/-) mice demonstrated a susceptibility to hyperglycemia and hyperinsulinemia that correlated with reduced insulin clearance and reduced expression of hepatic CEACAM1 (carcinoembryonic antigen-related cell adhesion molecule 1), a mediator of clearance. Livers of HF-fed mutant mice had high lipid content and elevated expression of lipogenic genes (peroxisome proliferator-activated receptor gamma, fatty acid synthase, and sterol regulatory element-binding protein 1c) and inflammatory markers (TNFalpha). Interestingly, mutant mice under HF showed elevated serum corticosterone, but their steatotic livers had reduced expression of gluconeogenic genes (phosphoenolpyruvate carboxy kinase, glucose 6 phosphatase, and pyruvate dehydrogenase kinase 4), whereas muscle and adipose expressed normal to elevated levels of glucocorticoid markers. These data suggest a state of glucocorticoid resistance arising from liver-specific loss of GR activity. Consistent with this hypothesis, reduced expression of gluconeogenic genes and CEACAM1 was observed in dexamethasone-treated FKBP52-deficient mouse embryonic fibroblast cells. We propose a model in which FKBP52 loss reduces GR control of gluconeogenesis, predisposing the liver to steatosis under HF-diet conditions attributable to a shunting of metabolism from glucose production to lipogenesis.


Asunto(s)
Hígado Graso/genética , Receptores de Glucocorticoides/metabolismo , Proteínas de Unión a Tacrolimus/fisiología , Animales , Antígenos CD/genética , Western Blotting , Moléculas de Adhesión Celular/genética , Células Cultivadas , Corticosterona/sangre , Grasas de la Dieta/farmacología , Susceptibilidad a Enfermedades , Hígado Graso/inducido químicamente , Hiperglucemia/inducido químicamente , Hiperglucemia/genética , Hiperinsulinismo/inducido químicamente , Hiperinsulinismo/genética , Masculino , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores de Glucocorticoides/genética , Proteínas de Unión a Tacrolimus/genética , Factor de Necrosis Tumoral alfa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA