Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Nature ; 618(7963): 159-168, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37225977

RESUMEN

Harnessing the potential beneficial effects of kinase signalling through the generation of direct kinase activators remains an underexplored area of drug development1-5. This also applies to the PI3K signalling pathway, which has been extensively targeted by inhibitors for conditions with PI3K overactivation, such as cancer and immune dysregulation. Here we report the discovery of UCL-TRO-1938 (referred to as 1938 hereon), a small-molecule activator of the PI3Kα isoform, a crucial effector of growth factor signalling. 1938 allosterically activates PI3Kα through a distinct mechanism by enhancing multiple steps of the PI3Kα catalytic cycle and causes both local and global conformational changes in the PI3Kα structure. This compound is selective for PI3Kα over other PI3K isoforms and multiple protein and lipid kinases. It transiently activates PI3K signalling in all rodent and human cells tested, resulting in cellular responses such as proliferation and neurite outgrowth. In rodent models, acute treatment with 1938 provides cardioprotection from ischaemia-reperfusion injury and, after local administration, enhances nerve regeneration following nerve crush. This study identifies a chemical tool to directly probe the PI3Kα signalling pathway and a new approach to modulate PI3K activity, widening the therapeutic potential of targeting these enzymes through short-term activation for tissue protection and regeneration. Our findings illustrate the potential of activating kinases for therapeutic benefit, a currently largely untapped area of drug development.


Asunto(s)
Regeneración Nerviosa , Humanos , Neoplasias/tratamiento farmacológico , Regeneración Nerviosa/efectos de los fármacos , Isoformas de Proteínas/agonistas , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasa Clase I/química , Fosfatidilinositol 3-Quinasa Clase I/efectos de los fármacos , Cardiotónicos/farmacología , Animales , Biocatálisis/efectos de los fármacos , Conformación Proteica/efectos de los fármacos , Neuritas/efectos de los fármacos , Daño por Reperfusión/prevención & control , Compresión Nerviosa , Proliferación Celular/efectos de los fármacos
2.
Mol Ther ; 28(5): 1263-1275, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32145202

RESUMEN

Tumor-targeting oncolytic viruses such as vaccinia virus (VV) are attractive cancer therapeutic agents that act through multiple mechanisms to provoke both tumor lysis and anti-tumor immune responses. However, delivery of these agents remains restricted to intra-tumoral administration, which prevents effective targeting of inaccessible and disseminated tumor cells. In the present study we have identified transient pharmacological inhibition of the leukocyte-enriched phosphoinositide 3-kinase δ (PI3Kδ) as a novel mechanism to potentiate intravenous delivery of oncolytic VV to tumors. Pre-treatment of immunocompetent mice with the PI3Kδ-selective inhibitor IC87114 or the clinically approved idelalisib (CAL-101), prior to intravenous delivery of a tumor-tropic VV, dramatically improved viral delivery to tumors. This occurred via an inhibition of viral attachment to, but not internalization by, systemic macrophages through perturbation of signaling pathways involving RhoA/ROCK, AKT, and Rac. Pre-treatment using PI3Kδ-selective inhibitors prior to intravenous delivery of VV resulted in enhanced anti-tumor efficacy and significantly prolonged survival compared to delivery without PI3Kδ inhibition. These results indicate that effective intravenous delivery of oncolytic VV may be clinically achievable and could be useful in improving anti-tumor efficacy of oncolytic virotherapy.


Asunto(s)
Adenina/análogos & derivados , Administración Intravenosa/métodos , Antineoplásicos/uso terapéutico , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Inmunoterapia/métodos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/inmunología , Purinas/uso terapéutico , Quinazolinas/uso terapéutico , Quinazolinonas/uso terapéutico , Virus Vaccinia/inmunología , Adenina/farmacología , Adenina/uso terapéutico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Supervivencia Celular , Terapia Combinada/métodos , Femenino , Ratones , Ratones Endogámicos BALB C , Purinas/farmacología , Quinazolinas/farmacología , Quinazolinonas/farmacología , Trasplante Homólogo , Resultado del Tratamiento , Carga Tumoral
3.
Nat Commun ; 8(1): 1804, 2017 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-29180704

RESUMEN

Vps34 PI3K is thought to be the main producer of phosphatidylinositol-3-monophosphate, a lipid that controls intracellular vesicular trafficking. The organismal impact of systemic inhibition of Vps34 kinase activity is not completely understood. Here we show that heterozygous Vps34 kinase-dead mice are healthy and display a robustly enhanced insulin sensitivity and glucose tolerance, phenotypes mimicked by a selective Vps34 inhibitor in wild-type mice. The underlying mechanism of insulin sensitization is multifactorial and not through the canonical insulin/Akt pathway. Vps34 inhibition alters cellular energy metabolism, activating the AMPK pathway in liver and muscle. In liver, Vps34 inactivation mildly dampens autophagy, limiting substrate availability for mitochondrial respiration and reducing gluconeogenesis. In muscle, Vps34 inactivation triggers a metabolic switch from oxidative phosphorylation towards glycolysis and enhanced glucose uptake. Our study identifies Vps34 as a new drug target for insulin resistance in Type-2 diabetes, in which the unmet therapeutic need remains substantial.


Asunto(s)
Resistencia a la Insulina , Mitocondrias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia/fisiología , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasas Clase III , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Técnicas de Sustitución del Gen , Glucosa/análisis , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Glucólisis/fisiología , Hepatocitos , Heterocigoto , Humanos , Insulina/metabolismo , Hígado/citología , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Mioblastos , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Cultivo Primario de Células
4.
J Mol Med (Berl) ; 94(1): 5-11, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26658520

RESUMEN

The PI3K lipid kinases are involved in signal transduction and intracellular vesicular traffic, endowing these enzymes with multiple cellular functions and important roles in normal physiology and disease. In this mini-review, we aim to distill from the vast PI3K literature the key relevant concepts for successful targeting of this pathway in disease. Of the eight isoforms of PI3K, the class I PI3Ks have been implicated in the aetiology and maintenance of various diseases, most prominently cancer, overgrowth syndromes, inflammation and autoimmunity, with emerging potential roles in metabolic and cardiovascular disorders. The development of class I PI3K inhibitors, mainly for use in cancer and inflammation, is a very active area of drug development. In 2014, an inhibitor of the p110δ isoform of PI3K was approved for the treatment of specific human B cell malignancies. The key therapeutic indications of targeting each class I PI3K isoform are summarized and discussed.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Inflamación/tratamiento farmacológico , Neoplasias/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Humanos , Fosforilación , Isoformas de Proteínas/genética , Transducción de Señal
5.
PLoS Genet ; 11(7): e1005304, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26132308

RESUMEN

The organismal roles of the ubiquitously expressed class I PI3K isoform p110ß remain largely unknown. Using a new kinase-dead knockin mouse model that mimics constitutive pharmacological inactivation of p110ß, we document that full inactivation of p110ß leads to embryonic lethality in a substantial fraction of mice. Interestingly, the homozygous p110ß kinase-dead mice that survive into adulthood (maximum ~26% on a mixed genetic background) have no apparent phenotypes, other than subfertility in females and complete infertility in males. Systemic inhibition of p110ß results in a highly specific blockade in the maturation of spermatogonia to spermatocytes. p110ß was previously suggested to signal downstream of the c-kit tyrosine kinase receptor in germ cells to regulate their proliferation and survival. We now report that p110ß also plays a germ cell-extrinsic role in the Sertoli cells (SCs) that support the developing sperm, with p110ß inactivation dampening expression of the SC-specific Androgen Receptor (AR) target gene Rhox5, a homeobox gene critical for spermatogenesis. All extragonadal androgen-dependent functions remain unaffected by global p110ß inactivation. In line with a crucial role for p110ß in SCs, selective inactivation of p110ß in these cells results in male infertility. Our study is the first documentation of the involvement of a signalling enzyme, PI3K, in the regulation of AR activity during spermatogenesis. This developmental pathway may become active in prostate cancer where p110ß and AR have previously been reported to functionally interact.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fertilidad/fisiología , Infertilidad Masculina/genética , Receptores Androgénicos/metabolismo , Células de Sertoli/metabolismo , Animales , Blastocisto/citología , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/genética , Femenino , Proteínas de Homeodominio/genética , Infertilidad Femenina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mórula/citología , Receptores Androgénicos/genética , Transducción de Señal/genética , Espermatogénesis/genética , Factores de Transcripción/genética , Transcripción Genética/genética
6.
Blood ; 126(9): 1128-37, 2015 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-26109204

RESUMEN

The physiologic roles of the class II phosphoinositide 3-kinases (PI3Ks) and their contributions to phosphatidylinositol 3-monophosphate (PI3P) and PI(3,4)P2 production remain elusive. Here we report that mice heterozygous for a constitutively kinase-dead PI3K-C2α display aberrant platelet morphology with an elevated number of barbell-shaped proplatelets, a recently discovered intermediate stage in the final process of platelet production. Platelets with heterozygous PI3K-C2α inactivation have critical defects in α-granules and membrane structure that are associated with modifications in megakaryocytes. These platelets are more rigid and unable to form filopodia after stimulation. Heterozygous PI3K-C2α inactivation in platelets led to a significant reduction in the basal pool of PI3P and a mislocalization of several membrane skeleton proteins known to control the interactions between the plasma membrane and cytoskeleton. These alterations had repercussions on the performance of platelet responses with delay in the time of arterial occlusion in an in vivo model of thrombosis and defect in thrombus formation in an ex vivo blood flow system. These data uncover a key role for PI3K-C2α activity in the generation of a basal housekeeping PI3P pool and in the control of membrane remodeling, critical for megakaryocytopoiesis and normal platelet production and function.


Asunto(s)
Plaquetas/patología , Membrana Celular/patología , Mutación , Fosfatidilinositol 3-Quinasas/genética , Animales , Plaquetas/citología , Plaquetas/metabolismo , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Técnicas de Sustitución del Gen , Heterocigoto , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Trombopoyesis
7.
Nat Immunol ; 13(11): 1045-1054, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23023391

RESUMEN

Lipopolysaccharide activates plasma-membrane signaling and endosomal signaling by Toll-like receptor 4 (TLR4) through the TIRAP-MyD88 and TRAM-TRIF adaptor complexes, respectively, but it is unclear how the signaling switch between these cell compartments is coordinated. In dendritic cells, we found that the p110δ isoform of phosphatidylinositol-3-OH kinase (PI(3)K) induced internalization of TLR4 and dissociation of TIRAP from the plasma membrane, followed by calpain-mediated degradation of TIRAP. Accordingly, inactivation of p110δ prolonged TIRAP-mediated signaling from the plasma membrane, which augmented proinflammatory cytokine production while decreasing TRAM-dependent endosomal signaling that generated anti-inflammatory cytokines (interleukin 10 and interferon-ß). In line with that altered signaling output, p110δ-deficient mice showed enhanced endotoxin-induced death. Thus, by controlling the 'topology' of TLR4 signaling complexes, p110δ balances overall homeostasis in the TLR4 pathway.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase Ia/inmunología , Células Dendríticas/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Animales , Calpaína/farmacología , Compartimento Celular/inmunología , Membrana Celular/efectos de los fármacos , Membrana Celular/genética , Membrana Celular/inmunología , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Endosomas/efectos de los fármacos , Endosomas/genética , Endosomas/inmunología , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Interferón beta/biosíntesis , Interferón beta/inmunología , Interleucina-10/biosíntesis , Interleucina-10/inmunología , Isoenzimas/genética , Isoenzimas/inmunología , Lipopolisacáridos/farmacología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Noqueados , Receptores de Interleucina/genética , Receptores de Interleucina/inmunología , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/inmunología , Choque Séptico/genética , Choque Séptico/inmunología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Receptor Toll-Like 4/genética
8.
Biochem J ; 443(3): 857-67, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22375552

RESUMEN

PI3Ks (phosphoinositide 3-kinases) are signalling molecules and drug targets with important biological functions, yet the regulation of PI3K gene expression is poorly understood. Key PI3Ks are the class IA PI3Ks that consist of a catalytic subunit (p110α, p110ß and p110δ) in complex with a p85 regulatory subunit. Whereas p110α and p110ß are ubiquitously expressed, high levels of p110δ are mainly found in white blood cells, with most non-leucocytes expressing low levels of p110δ. In the present paper we report that TNFα (tumour necrosis factor α) stimulation induces p110δ expression in human ECs (endothelial cells) and synovial fibroblasts, but not in leucocytes, through transcription start sites located in a novel promoter region in the p110δ gene (PIK3CD). This promoter is used in all cell types, including solid tumour cell lines that express p110δ, and is activated by TNFα in ECs and synovial fibroblasts. We further present a detailed biochemical and bioinformatic characterization of p110δ gene regulation, demonstrating that PIK3CD has distinct promoters, some of which can be dynamically activated by pro-inflammatory mediators. This is the first molecular identification of a PI3K promoter under the control of acute extracellular stimulation.


Asunto(s)
Fosfatidilinositol 3-Quinasas/genética , Regiones Promotoras Genéticas , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I , Humanos
9.
Environ Toxicol Chem ; 27(11): 2341-5, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18476752

RESUMEN

The nonsteroidal anti-inflammatory drug diclofenac is extremely toxic to Old World Gyps vultures (median lethal dose -0.1-0.2 mg/kg), evoking visceral gout, renal necrosis, and mortality within a few days of exposure. Unintentional secondary poisoning of vultures that fed upon carcasses of diclofenac-treated livestock decimated populations in the Indian subcontinent. Because of the widespread use of diclofenac and other cyclooxygenase-2 inhibiting drugs, a toxicological study was undertaken in turkey vultures (Cathartes aura) as an initial step in examining sensitivity of New World scavenging birds. Two trials were conducted entailing oral gavage of diclofenac at doses ranging from 0.08 to 25 mg/kg body weight. Birds were observed for 7 d, blood samples were collected for plasma chemistry (predose and 12, 24, and 48 h and 7 d postdose), and select individuals were necropsied. Diclofenac failed to evoke overt signs of toxicity, visceral gout, renal necrosis, or elevate plasma uric acid at concentrations greater than 100 times the estimated median lethal dose reported for Gyps vultures. For turkey vultures receiving 8 or 25 mg/kg, the plasma half-life of diclofenac was estimated to be 6 h, and it was apparently cleared after several days as no residues were detectable in liver or kidney at necropsy. Differential sensitivity among avian species is a hallmark of cyclooxygenase-2 inhibitors, and despite the tolerance of turkey vultures to diclofenac, additional studies in related scavenging species seem warranted.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Diclofenaco/toxicidad , Animales , Aves , Tolerancia a Medicamentos , Femenino , Masculino , Especificidad de la Especie
10.
EMBO J ; 25(12): 2757-67, 2006 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-16724108

RESUMEN

Small carboxyl-terminal domain (CTD) phosphatase 2 (SCP2) was identified and verified as a protein that interacts with the androgen receptor (AR). Ectopic expression of SCP2 or two other family members, SCP1 and SCP3, attenuated AR transcriptional activity in LNCaP cells and were recruited in an androgen- and AR-dependent fashion onto the prostate-specific antigen (PSA) promoter. Silencing SCP2 and SCP1 by short hairpin RNAs increased androgen-dependent transcription of the PSA gene and augmented AR loading onto the PSA promoter and enhancer. SCP2 also attenuated glucocorticoid receptor (GR) function, and its silencing increased dexamethasone-mediated PSA mRNA accumulation and GR loading onto the PSA enhancer in LNCaP 1F5 cells. SCP2 silencing was accompanied by augmented recruitment and earlier cycling of RNA polymerase II on the promoter. Ser 5 phosphorylation of the RNA polymerase II CTD, a process necessary for initiation of transcription elongation, occurred significantly earlier in SCP2-silenced than parental LNCaP cells. Collectively, our results suggest that SCP2 is involved in promoter clearance during steroid-activated transcription.


Asunto(s)
Andrógenos/farmacología , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Receptores Androgénicos/metabolismo , Transcripción Genética/efectos de los fármacos , Animales , Células COS , Proteínas de Ciclo Celular , Células Cultivadas , Chlorocebus aethiops , Proteínas de Unión al ADN , Elementos de Facilitación Genéticos/genética , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Células HeLa , Humanos , Masculino , Fosfoserina/metabolismo , Regiones Promotoras Genéticas/genética , Antígeno Prostático Específico/genética , Proteína Fosfatasa 2 , ARN Polimerasa II/metabolismo , Activación Transcripcional/genética
11.
Protein Expr Purif ; 45(2): 335-42, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16202624

RESUMEN

LACTB is a mammalian mitochondrial protein sharing sequence similarity to the beta-lactamase/penicillin-binding protein family of serine proteases that are involved in bacterial cell wall metabolism. The physiological role of LACTB is unclear. In this study we have subcloned the cDNA of mouse LACTB (mLACTB) and produced recombinant mLACTB protein in Escherichia coli. When mLACTB was expressed as an N-terminal GST fusion protein (GST-mLACTB), full-length GST-mLACTB protein was recovered by glutathione-agarose affinity chromatography as determined by MALDI-TOF mass spectrometry and immunoblotting. Expression of mLACTB as a C-terminal GST fusion protein or with either an N- or C-terminal His6-tag resulted in proteolytic degradation of the protein and we were not able to detect full-length mLACTB. Analysis of GST-mLACTB by Fourier transform infrared spectrometry revealed the presence of alpha-helices, beta-sheets and turns, consistent with a well-defined secondary structure. These results show that mLACTB can be expressed as a GST fusion protein in E. coli and suggest that GST-mLACTB was properly folded.


Asunto(s)
Escherichia coli/metabolismo , Proteínas de la Membrana , Mitocondrias/enzimología , Proteínas Recombinantes de Fusión , Proteínas Ribosómicas , Secuencia de Aminoácidos , Animales , Escherichia coli/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/metabolismo , Ratones , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/aislamiento & purificación , Proteínas Ribosómicas/metabolismo , Alineación de Secuencia , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectroscopía Infrarroja por Transformada de Fourier , beta-Lactamasas/genética , beta-Lactamasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA