Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 7(9)2022 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-35358093

RESUMEN

Arterial stiffness predicts cardiovascular disease and all-cause mortality, but its treatment remains challenging. Mice treated with angiotensin II (Ang II) develop hypertension, arterial stiffness, vascular dysfunction, and a downregulation of Rho-related BTB domain-containing protein 1 (RhoBTB1) in the vasculature. RhoBTB1 is associated with blood pressure regulation, but its function is poorly understood. We tested the hypothesis that restoring RhoBTB1 can attenuate arterial stiffness, hypertension, and vascular dysfunction in Ang II-treated mice. Genetic complementation of RhoBTB1 in the vasculature was achieved using mice expressing a tamoxifen-inducible, smooth muscle-specific RhoBTB1 transgene. RhoBTB1 restoration efficiently and rapidly alleviated arterial stiffness but not hypertension or vascular dysfunction. Mechanistic studies revealed that RhoBTB1 had no substantial effect on several classical arterial stiffness contributors, such as collagen deposition, elastin content, and vascular smooth muscle remodeling. Instead, Ang II increased actin polymerization in the aorta, which was reversed by RhoBTB1. Changes in the levels of 2 regulators of actin polymerization, cofilin and vasodilator-stimulated phosphoprotein, in response to RhoBTB1 were consistent with an actin depolymerization mechanism. Our study reveals an important function of RhoBTB1, demonstrates its vital role in antagonizing established arterial stiffness, and further supports a functional and mechanistic separation among hypertension, vascular dysfunction, and arterial stiffness.


Asunto(s)
Hipertensión , Rigidez Vascular , Actinas/genética , Actinas/metabolismo , Angiotensina II/metabolismo , Animales , Hipertensión/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Remodelación Vascular
2.
JCI Insight ; 52019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31184598

RESUMEN

Patients with mutations in Cullin-3 (CUL3) exhibit severe early onset hypertension but the contribution of the smooth muscle remains unclear. Conditional genetic ablation of CUL3 in vascular smooth muscle (S-CUL3KO) causes progressive impairment in responsiveness to nitric oxide (NO), rapid development of severe hypertension, and increased arterial stiffness. Loss of CUL3 in primary aortic smooth muscle cells or aorta resulted in decreased expression of the NO receptor, soluble guanylate cyclase (sGC), causing a marked reduction in cGMP production and impaired vasodilation to cGMP analogues. Vasodilation responses to a selective large conductance Ca2+-activated K+-channel activator were normal suggesting that downstream signals which promote smooth muscle-dependent relaxation remained intact. We conclude that smooth muscle specific CUL3 ablation impairs both cGMP production and cGMP responses and that loss of CUL3 function selectively in smooth muscle is sufficient to cause severe hypertension by interfering with the NO-sGC-cGMP pathway. Our study provides compelling evidence for the sufficiency of vascular smooth muscle CUL3 as a major regulator of BP. CUL3 mutations cause severe vascular dysfunction, arterial stiffness and hypertension due to defects in vascular smooth muscle.


Asunto(s)
Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Predisposición Genética a la Enfermedad/genética , Hipertensión/genética , Hipertensión/metabolismo , Músculo Liso/metabolismo , Animales , Aorta/metabolismo , Aorta/patología , GMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Hipertensión/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Mutación , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico , Guanilil Ciclasa Soluble/metabolismo , Transcriptoma , Rigidez Vascular , Vasodilatación
3.
J Clin Invest ; 129(6): 2318-2332, 2019 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-30896450

RESUMEN

Mice selectively expressing PPARγ dominant negative mutation in vascular smooth muscle exhibit RhoBTB1-deficiency and hypertension. Our rationale was to employ genetic complementation to uncover the mechanism of action of RhoBTB1 in vascular smooth muscle. Inducible smooth muscle-specific restoration of RhoBTB1 fully corrected the hypertension and arterial stiffness by improving vasodilator function. Notably, the cardiovascular protection occurred despite preservation of increased agonist-mediated contraction and RhoA/Rho kinase activity, suggesting RhoBTB1 selectively controls vasodilation. RhoBTB1 augmented the cGMP response to nitric oxide by restraining the activity of phosphodiesterase 5 (PDE5) by acting as a substrate adaptor delivering PDE5 to the Cullin-3 E3 Ring ubiquitin ligase complex for ubiquitination inhibiting PDE5. Angiotensin-II infusion also caused RhoBTB1-deficiency and hypertension which was prevented by smooth muscle specific RhoBTB1 restoration. We conclude that RhoBTB1 protected from hypertension, vascular smooth muscle dysfunction, and arterial stiffness in at least two models of hypertension.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/metabolismo , Hipertensión/prevención & control , Músculo Liso Vascular/metabolismo , Rigidez Vascular , Vasodilatación , Proteínas de Unión al GTP rho/metabolismo , Angiotensina II/efectos adversos , Angiotensina II/farmacología , Animales , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5/genética , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/metabolismo , Ratones , Ratones Transgénicos , Músculo Liso Vascular/patología , Óxido Nítrico/genética , Óxido Nítrico/metabolismo , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
4.
J Clin Invest ; 129(4): 1641-1653, 2019 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-30721156

RESUMEN

Hyperactivated AKT/mTOR signaling is a hallmark of pancreatic neuroendocrine tumors (PNETs). Drugs targeting this pathway are used clinically, but tumor resistance invariably develops. A better understanding of factors regulating AKT/mTOR signaling and PNET pathogenesis is needed to improve current therapies. We discovered that RABL6A, a new oncogenic driver of PNET proliferation, is required for AKT activity. Silencing RABL6A caused PNET cell-cycle arrest that coincided with selective loss of AKT-S473 (not T308) phosphorylation and AKT/mTOR inactivation. Restoration of AKT phosphorylation rescued the G1 phase block triggered by RABL6A silencing. Mechanistically, loss of AKT-S473 phosphorylation in RABL6A-depleted cells was the result of increased protein phosphatase 2A (PP2A) activity. Inhibition of PP2A restored phosphorylation of AKT-S473 in RABL6A-depleted cells, whereas PP2A reactivation using a specific small-molecule activator of PP2A (SMAP) abolished that phosphorylation. Moreover, SMAP treatment effectively killed PNET cells in a RABL6A-dependent manner and suppressed PNET growth in vivo. The present work identifies RABL6A as a new inhibitor of the PP2A tumor suppressor and an essential activator of AKT in PNET cells. Our findings offer what we believe is a novel strategy of PP2A reactivation for treatment of PNETs as well as other human cancers driven by RABL6A overexpression and PP2A inactivation.


Asunto(s)
Carcinoma Neuroendocrino/enzimología , Proteínas Oncogénicas/metabolismo , Neoplasias Pancreáticas/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Carcinoma Neuroendocrino/genética , Carcinoma Neuroendocrino/patología , Línea Celular Tumoral , Activadores de Enzimas/farmacología , Fase G1/efectos de los fármacos , Fase G1/genética , Humanos , Proteínas Oncogénicas/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas de Unión al GTP rab/genética
5.
Physiol Genomics ; 49(11): 653-658, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-28916634

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are a family of conserved ligand-activated nuclear receptor transcription factors heterogeneously expressed in mammalian tissues. PPARγ is recognized as a master regulator of adipogenesis, fatty acid metabolism, and glucose homeostasis, but genetic evidence also supports the concept that PPARγ regulates the cardiovascular system, particularly vascular function and blood pressure. There is now compelling evidence that the beneficial blood pressure-lowering effects of PPARγ activation are due to its activity in vascular smooth muscle and endothelium, through its modulation of nitric oxide-dependent vasomotor function. Endothelial PPARγ regulates the production and bioavailability of nitric oxide, while PPARγ in the smooth muscle regulates the vasomotor response to nitric oxide. We recently identified retinol binding protein 7 (RBP7) as a PPARγ target gene that is specifically and selectively expressed in the endothelium. In this review, we will discuss the evidence that RBP7 is required to mediate the antioxidant effects of PPARγ and mediate PPARγ target gene selectivity in the endothelium.


Asunto(s)
Antioxidantes/metabolismo , Endotelio Vascular/metabolismo , PPAR gamma/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , Animales , Humanos , Modelos Biológicos , Unión Proteica
6.
Hypertension ; 70(1): 174-182, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28507170

RESUMEN

Selective expression of dominant negative (DN) peroxisome proliferator-activated receptor γ (PPARγ) in vascular smooth muscle cells (SMC) results in hypertension, atherosclerosis, and increased nuclear factor-κB (NF-κB) target gene expression. Mesenteric SMC were cultured from mice designed to conditionally express wild-type (WT) or DN-PPARγ in response to Cre recombinase to determine how SMC PPARγ regulates expression of NF-κB target inflammatory genes. SMC-specific overexpression of WT-PPARγ or agonist-induced activation of endogenous PPARγ blunted tumor necrosis factor α (TNF-α)-induced NF-κB target gene expression and activity of an NF-κB-responsive promoter. TNF-α-induced gene expression responses were enhanced by DN-PPARγ in SMC. Although expression of NF-κB p65 was unchanged, nuclear export of p65 was accelerated by WT-PPARγ and prevented by DN-PPARγ in SMC. Leptomycin B, a nuclear export inhibitor, blocked p65 nuclear export and inhibited the anti-inflammatory action of PPARγ. Consistent with a role in facilitating p65 nuclear export, WT-PPARγ coimmunoprecipitated with p65, and WT-PPARγ was also exported from the nucleus after TNF-α treatment. Conversely, DN-PPARγ does not bind to p65 and was retained in the nucleus after TNF-α treatment. Transgenic mice expressing WT-PPARγ or DN-PPARγ specifically in SMC (S-WT or S-DN) were bred with mice expressing luciferase controlled by an NF-κB-responsive promoter to assess effects on NF-κB activity in whole tissue. TNF-α-induced NF-κB activity was decreased in aorta and carotid artery from S-WT but was increased in vessels from S-DN mice. We conclude that SMC PPARγ blunts expression of proinflammatory genes by inhibition of NF-κB activity through a mechanism promoting nuclear export of p65, which is abolished by DN mutation in PPARγ.


Asunto(s)
Hipertensión , Músculo Liso Vascular , FN-kappa B , PPAR gamma/genética , Factor de Transcripción ReIA/metabolismo , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Antiinflamatorios/farmacología , Núcleo Celular/metabolismo , Células Cultivadas , Ácidos Grasos Insaturados/farmacología , Hipertensión/genética , Hipertensión/metabolismo , Inflamación/genética , Inflamación/metabolismo , Ratones , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatología , Mutación , FN-kappa B/antagonistas & inhibidores , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
7.
JCI Insight ; 2(6): e91738, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28352663

RESUMEN

Impaired PPARγ activity in endothelial cells causes oxidative stress and endothelial dysfunction which causes a predisposition to hypertension, but the identity of key PPARγ target genes that protect the endothelium remain unclear. Retinol-binding protein 7 (RBP7) is a PPARγ target gene that is essentially endothelium specific. Whereas RBP7-deficient mice exhibit normal endothelial function at baseline, they exhibit severe endothelial dysfunction in response to cardiovascular stressors, including high-fat diet and subpressor angiotensin II. Endothelial dysfunction was not due to differences in weight gain, impaired glucose homeostasis, or hepatosteatosis, but occurred through an oxidative stress-dependent mechanism which can be rescued by scavengers of superoxide. RNA sequencing revealed that RBP7 was required to mediate induction of a subset of PPARγ target genes by rosiglitazone in the endothelium including adiponectin. Adiponectin was selectively induced in the endothelium of control mice by high-fat diet and rosiglitazone, whereas RBP7 deficiency abolished this induction. Adiponectin inhibition caused endothelial dysfunction in control vessels, whereas adiponectin treatment of RBP7-deficient vessels improved endothelium-dependent relaxation and reduced oxidative stress. We conclude that RBP7 is required to mediate the protective effects of PPARγ in the endothelium through adiponectin, and RBP7 is an endothelium-specific PPARγ target and regulator of PPARγ activity.


Asunto(s)
Adiponectina/metabolismo , Antioxidantes/metabolismo , Endotelio Vascular/metabolismo , PPAR gamma/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , Adiponectina/genética , Animales , Dieta Alta en Grasa , Endotelio Vascular/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , PPAR gamma/genética , ARN Mensajero/genética , Proteínas Celulares de Unión al Retinol/genética
8.
JCI Insight ; 1(19): e91015, 2016 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-27882355

RESUMEN

Cullin-3 (CUL3) mutations (CUL3Δ9) were previously identified in hypertensive patients with pseudohypoaldosteronism type-II (PHAII), but the mechanism causing hypertension and whether this is driven by renal tubular or extratubular mechanisms remains unknown. We report that selective expression of CUL3Δ9 in smooth muscle acts by interfering with expression and function of endogenous CUL3, resulting in impaired turnover of the CUL3 substrate RhoA, increased RhoA activity, and augmented RhoA/Rho kinase signaling. This caused vascular dysfunction and increased arterial pressure under baseline conditions and a marked increase in arterial pressure, collagen deposition, and vascular stiffness in response to a subpressor dose of angiotensin II, which did not cause hypertension in control mice. Inhibition of total cullin activity increased the level of CUL3 substrates cyclin E and RhoA, and expression of CUL3Δ9 decreased the level of the active form of endogenous CUL3 in human aortic smooth muscle cells. These data indicate that selective expression of the Cul3Δ9 mutation in vascular smooth muscle phenocopies the hypertension observed in Cul3Δ9 human subjects and suggest that mutations in CUL3 cause human hypertension in part through a mechanism involving smooth muscle dysfunction initiated by a loss of CUL3-mediated degradation of RhoA.


Asunto(s)
Proteínas Cullin/genética , Hipertensión/genética , Músculo Liso Vascular/fisiopatología , Rigidez Vascular , Animales , Células Cultivadas , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Miocitos del Músculo Liso/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 310(1): H39-48, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26566726

RESUMEN

Loss of peroxisome proliferator-activated receptor (PPAR)-γ function in the vascular endothelium enhances atherosclerosis and NF-κB target gene expression in high-fat diet-fed apolipoprotein E-deficient mice. The mechanisms by which endothelial PPAR-γ regulates inflammatory responses and protects against atherosclerosis remain unclear. To assess functional interactions between PPAR-γ and inflammation, we used a model of IL-1ß-induced aortic dysfunction in transgenic mice with endothelium-specific overexpression of either wild-type (E-WT) or dominant negative PPAR-γ (E-V290M). IL-1ß dose dependently decreased IκB-α, increased phospho-p65, and increased luciferase activity in the aorta of NF-κB-LUC transgenic mice. IL-1ß also dose dependently reduced endothelial-dependent relaxation by ACh. The loss of ACh responsiveness was partially improved by pretreatment of the vessels with the PPAR-γ agonist rosiglitazone or in E-WT. Conversely, IL-1ß-induced endothelial dysfunction was worsened in the aorta from E-V290M mice. Although IL-1ß increased the expression of NF-κB target genes, NF-κB p65 inhibitor did not alleviate endothelial dysfunction induced by IL-1ß. Tempol, a SOD mimetic, partially restored ACh responsiveness in the IL-1ß-treated aorta. Notably, tempol only modestly improved protection in the E-WT aorta but had an increased protective effect in the E-V290M aorta compared with the aorta from nontransgenic mice, suggesting that PPAR-γ-mediated protection involves antioxidant effects. IL-1ß increased ROS and decreased the phospho-endothelial nitric oxide synthase (Ser(1177))-to-endothelial nitric oxide synthase ratio in the nontransgenic aorta. These effects were completely abolished in the aorta with endothelial overexpression of WT PPAR-γ but were worsened in the aorta with E-V290M even in the absence of IL-1ß. We conclude that PPAR-γ protects against IL-1ß-mediated endothelial dysfunction through a reduction of oxidative stress responses but not by blunting IL-1ß-mediated NF-κB activity.


Asunto(s)
Aorta/efectos de los fármacos , Enfermedades de la Aorta/prevención & control , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Interleucina-1beta/farmacología , Estrés Oxidativo/efectos de los fármacos , PPAR gamma/metabolismo , Animales , Antioxidantes/farmacología , Aorta/metabolismo , Aorta/patología , Aorta/fisiopatología , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Relación Dosis-Respuesta a Droga , Células Endoteliales/metabolismo , Células Endoteliales/patología , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Femenino , Regulación de la Expresión Génica , Genotipo , Humanos , Proteínas I-kappa B/metabolismo , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibidor NF-kappaB alfa , Óxido Nítrico Sintasa de Tipo III/metabolismo , PPAR gamma/agonistas , PPAR gamma/genética , Fenotipo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Transcripción ReIA/metabolismo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
10.
J Biol Chem ; 290(31): 19208-17, 2015 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-26100637

RESUMEN

Cullin-Ring ubiquitin ligases regulate protein turnover by promoting the ubiquitination of substrate proteins, targeting them for proteasomal degradation. It has been shown previously that mutations in Cullin3 (Cul3) causing deletion of 57 amino acids encoded by exon 9 (Cul3Δ9) cause hypertension. Moreover, RhoA activity contributes to vascular constriction and hypertension. We show that ubiquitination and degradation of RhoA is dependent on Cul3 in HEK293T cells in which Cul3 expression is ablated by either siRNA or by CRISPR-Cas9 genome editing. The latter was used to generate a Cul3-null cell line (HEK293T(Cul3KO)). When expressed in these cells, Cul3Δ9 supported reduced ubiquitin ligase activity toward RhoA compared with equivalent levels of wild-type Cul3 (Cul3WT). Consistent with its reduced activity, binding of Cul3Δ9 to the E3 ubiquitin ligase Rbx1 and neddylation of Cul3Δ9 were impaired significantly compared with Cul3WT. Conversely, Cul3Δ9 bound to substrate adaptor proteins more efficiently than Cul3WT. Cul3Δ9 also forms unstable dimers with Cul3WT, disrupting dimers of Cul3WT complexes that are required for efficient ubiquitination of some substrates. Indeed, coexpression of Cul3WT and Cul3Δ9 in HEK293T(Cul3KO) cells resulted in a decrease in the active form of Cul3WT. We conclude that Cul3Δ9-associated ubiquitin ligase activity toward RhoA is impaired and suggest that Cul3Δ9 mutations may act dominantly by sequestering substrate adaptors and disrupting Cul3WT complexes.


Asunto(s)
Proteínas Cullin/genética , Hipertensión/genética , Ubiquitinación , Proteína de Unión al GTP rhoA/metabolismo , Proteínas Portadoras , Expresión Génica , Células HEK293 , Humanos , Hipertensión/enzimología , Unión Proteica , Multimerización de Proteína
11.
Cancer Res ; 74(22): 6661-70, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25273089

RESUMEN

Mechanisms of neuroendocrine tumor (NET) proliferation are poorly understood, and therapies that effectively control NET progression and metastatic disease are limited. We found amplification of a putative oncogene, RABL6A, in primary human pancreatic NETs (PNET) that correlated with high-level RABL6A protein expression. Consistent with those results, stable silencing of RABL6A in cultured BON-1 PNET cells revealed that it is essential for their proliferation and survival. Cells lacking RABL6A predominantly arrested in G1 phase with a moderate mitotic block. Pathway analysis of microarray data suggested activation of the p53 and retinoblastoma (Rb1) tumor-suppressor pathways in the arrested cells. Loss of p53 had no effect on the RABL6A knockdown phenotype, indicating that RABL6A functions independent of p53 in this setting. By comparison, Rb1 inactivation partially restored G1 to S phase progression in RABL6A-knockdown cells, although it was insufficient to override the mitotic arrest and cell death caused by RABL6A loss. Thus, RABL6A promotes G1 progression in PNET cells by inactivating Rb1, an established suppressor of PNET proliferation and development. This work identifies RABL6A as a novel negative regulator of Rb1 that is essential for PNET proliferation and survival. We suggest RABL6A is a new potential biomarker and target for anticancer therapy in PNET patients.


Asunto(s)
Proliferación Celular , Fase G1 , Tumores Neuroendocrinos/patología , Proteínas Oncogénicas/fisiología , Neoplasias Pancreáticas/patología , Proteína de Retinoblastoma/fisiología , Fase S , Proteínas de Unión al GTP rab/fisiología , Línea Celular Tumoral , Humanos , Mitosis
13.
Cell Cycle ; 13(8): 1288-98, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24621507

RESUMEN

The p53 tumor suppressor is controlled by an interactive network of factors that stimulate or inhibit its transcriptional activity. Within that network, Mdm2 functions as the major antagonist of p53 by promoting its ubiquitylation and degradation. Conversely, Tip60 activates p53 through direct association on target promoters as well as acetylation of p53 at lysine 120 (K120). This study examines the functional relationship between Mdm2 and Tip60 with a novel p53 regulator, NIAM (nuclear interactor of ARF and Mdm2). Previous work showed NIAM can suppress proliferation and activate p53 independently of ARF, indicating that other factors mediate those activities. Here, we demonstrate that NIAM is a chromatin-associated protein that binds Tip60. NIAM can promote p53 K120 acetylation, although that modification is not required for NIAM to inhibit proliferation or induce p53 transactivation of the p21 promoter. Notably, Tip60 silencing showed it contributes to but is not sufficient for NIAM-mediated p53 activation, suggesting other mechanisms are involved. Indeed, growth-inhibitory forms of NIAM also bind to Mdm2, and increased NIAM expression levels disrupt p53-Mdm2 association, inhibit p53 polyubiquitylation, and prevent Mdm2-mediated inhibition of p53 transcriptional activity. Importantly, loss of NIAM significantly impairs p53 activation. Together, these results show that NIAM activates p53 through multiple mechanisms involving Tip60 association and Mdm2 inhibition. Thus, NIAM regulates 2 critical pathways that control p53 function and are altered in human cancers, implying an important role for NIAM in tumorigenesis.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Animales , Línea Celular , Línea Celular Tumoral , Proliferación Celular/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Histona Acetiltransferasas/metabolismo , Humanos , Lisina Acetiltransferasa 5 , Ratones , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Activación Transcripcional , Ubiquitinación
14.
Circ Res ; 112(3): 411-4, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23371897
15.
Mol Cancer Res ; 7(8): 1294-303, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19671690

RESUMEN

The PI3K/AKT signaling pathway contributes to cell cycle progression of cytokine-dependent hematopoietic cells under normal conditions, and it is absolutely required to override DNA damage-induced cell cycle arrest checkpoints in these cells. Phosphatidylinositol-3-kinase (PI3K)/AKT activity also correlates with Cdk2 activity in hematopoietic cells, suggesting that Cdk2 activation may be a relevant end point for this signaling pathway. However, mediators downstream of AKT in this pathway have not been defined. The forkhead transcription factor O (FOXO) family are negatively regulated by AKT-dependent phosphorylation and are known regulators of genes affecting cell cycle progression. We show that enhanced FOXO activity replicates the effect of PI3K inhibitors in enforcing G(1) and G(2) phase arrest after DNA damage. Conversely, knockdown of endogenous FOXO proteins increased Cdk2 activity and overrode DNA damage checkpoints in cells lacking PI3K activity. Moreover, loss of FOXO activity caused an increase in sensitivity to cisplatin-induced cell death, which was associated with failure to arrest cell cycle progression in the face of DNA damage caused by this chemotherapeutic agent. These cell cycle arrests were dependent on p27 expression when mediated by FOXO3a alone, but also involve p27-independent mechanisms when promoted by endogenous FOXO proteins. Together, these observations show that FOXO proteins enforce DNA damage-induced cell cycle arrest in hematopoietic cells. Inhibition of FOXO activity by cytokine-induced PI3K/AKT signaling is sufficient to override these DNA damage-induced cell cycle checkpoints, but may negatively impact hematopoietic cell viability.


Asunto(s)
Ciclo Celular , Daño del ADN , Factores de Transcripción Forkhead/metabolismo , Sistema Hematopoyético/citología , Sistema Hematopoyético/metabolismo , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/efectos de la radiación , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Cisplatino/farmacología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Resistencia a Medicamentos/efectos de los fármacos , Resistencia a Medicamentos/efectos de la radiación , Rayos gamma , Técnicas de Silenciamiento del Gen , Sistema Hematopoyético/efectos de los fármacos , Sistema Hematopoyético/efectos de la radiación , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3
16.
Cell Cycle ; 7(18): 2877-85, 2008 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-18769155

RESUMEN

DNA damage activates arrest checkpoints to halt cell cycle progression in G(1) and G(2) phases. These checkpoints can be overridden in hematopoietic cells by cytokines, such as erythropoietin, through the activation of a phosphoinositide 3-kinase (PI3K) signaling pathway. Here, we show that PI3K activity specifically overrides delayed mechanisms effecting permanent G(1) and G(2) phase arrests, but does not affect transient checkpoints arresting cells up to 10 hours after gamma-irradiation. Assessing the status of cell cycle regulators in hematopoietic cells arrested after gamma-irradiation, we show that Cdk2 activity is completely inhibited in both G(1) and G(2) arrested cells. Despite the absence of Cdk2 activity, cells arrested in G(2) phase did retain detectable levels of Cdk1 activity in the absence of PI3K signaling. However, reactivation of PI3K promoted robust increases in both Cdk1 and Cdk2 activity in G(2)-arrested cells. Reactivation of Cdks was accompanied by a resumption of cell cycling, but with strikingly different effectiveness in G(1) and G(2) phase arrested cells. Specifically, G(1)-arrested cells resumed normal cell cycle progression with little loss in viability when PI3K was activated after gamma-irradiation. Conversely, PI3K activation in G(2)-arrested cells promoted endoreduplication and death of the entire population. These observations show that cytokine-induced PI3K signaling pathways promote Cdk activation and override permanent cell cycle arrest checkpoints in hematopoietic cells. While this activity can rescue irradiated cells from permanent G(1) phase arrest, it results in aberrant cell cycling and death when activated in hematopoietic cells arrested at the G(2) phase DNA damage checkpoint.


Asunto(s)
Daño del ADN , Fase G2 , Sistema Hematopoyético/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Animales , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de la radiación , Línea Celular , Fase G2/efectos de la radiación , Rayos gamma , Sistema Hematopoyético/efectos de la radiación , Humanos , Metafase/efectos de la radiación , Ratones , Transducción de Señal/efectos de la radiación
17.
Immunol Res ; 39(1-3): 173-84, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17917064

RESUMEN

The proliferation of many myeloid and lymphoid cell populations is directly controlled by cytokine growth factors acting through a related family of cytokine receptors. This regulation implies that the signaling pathways activated by cytokine receptors must communicate with mechanisms that control mammalian cell cycle progression. Evidence for how these signaling pathways promote hematopoietic cell proliferation is considered along with their likely targets among the cell cycle regulators.


Asunto(s)
Ciclo Celular/fisiología , Citocinas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores de Citocinas/metabolismo , Transducción de Señal , Animales , Proliferación Celular , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Redes y Vías Metabólicas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Factores de Transcripción STAT/metabolismo
18.
Mol Cell Biol ; 25(4): 1258-71, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15684379

RESUMEN

The ARF tumor suppressor is a nucleolar protein that activates p53-dependent checkpoints by binding Mdm2, a p53 antagonist. Despite persuasive evidence that ARF can bind and inactivate Mdm2 in the nucleoplasm, the prevailing view is that ARF exerts its growth-inhibitory activities from within the nucleolus. We suggest ARF primarily functions outside the nucleolus and provide evidence that it is sequestered and held inactive in that compartment by a nucleolar phosphoprotein, nucleophosmin (NPM). Most cellular ARF is bound to NPM regardless of whether cells are proliferating or growth arrested, indicating that ARF-NPM association does not correlate with growth suppression. Notably, ARF binds NPM through the same domains that mediate nucleolar localization and Mdm2 binding, suggesting that NPM could control ARF localization and compete with Mdm2 for ARF association. Indeed, NPM knockdown markedly enhanced ARF-Mdm2 association and diminished ARF nucleolar localization. Those events correlated with greater ARF-mediated growth suppression and p53 activation. Conversely, NPM overexpression antagonized ARF function while increasing its nucleolar localization. These data suggest that NPM inhibits ARF's p53-dependent activity by targeting it to nucleoli and impairing ARF-Mdm2 association.


Asunto(s)
Nucléolo Celular/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células COS , Nucléolo Celular/genética , Proliferación Celular , Chlorocebus aethiops , Inhibidor p16 de la Quinasa Dependiente de Ciclina , Humanos , Ratones , Células 3T3 NIH , Nucleofosmina , Unión Proteica , Transporte de Proteínas/fisiología , Proteínas Proto-Oncogénicas c-mdm2 , Células Tumorales Cultivadas , Proteína p14ARF Supresora de Tumor/genética
19.
Int J Oncol ; 26(2): 509-14, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15645137

RESUMEN

Heregulin (HRG), a ligand of ErbB receptor tyrosine kinases, is a potent mitogenic factor for breast cancer cells. Prolactin (PRL) has also been reported to regulate proliferation in breast cancer cells through its receptor, a member of the type I cytokine receptor family. Cytokine receptors are potent mitogens in hematopoietic cells, where they also override DNA damage-induced growth arrest checkpoints through activation of a phosphatidylinositol-3 kinase (PI3K) signaling pathway. In this study, we assessed the effect of gamma-irradiation on the mitogenic activity of HRG and PRL in breast cancer cells. HRG and PRL enhanced the proliferation of non-irradiated breast cancer cell lines in association with their ability to activate PI3K signaling pathways. Both growth factors also overrode irradiation-induced growth arrest in T47D cells, which resulted in decreased viability after irradiation. An inhibitor of PI3K, LY294002, abrogated growth factor-induced proliferation and the activity of cell cycle-dependent kinases in non-irradiated and irradiated cells. Thus, growth factors acting through distinct receptor families share a similar PI3K-dependent ability to promote proliferation and override DNA damage-induced growth arrest in breast cancer cells. These observations also suggest that selective activation of PI3K-dependent signaling can enhance radiosensitivity in breast cancer cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Neurregulina-1/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Prolactina/fisiología , Western Blotting , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Cromonas/farmacología , ADN/metabolismo , Sustancias de Crecimiento , Humanos , Morfolinas/farmacología , Fosforilación , Transducción de Señal
20.
Exp Cell Res ; 299(1): 257-66, 2004 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-15302592

RESUMEN

Cytokine growth factors regulate the proliferation of hematopoietic cells through activation of several distinct signaling pathways. We have assessed the contribution of phosphoinositide 3-kinase (PI3K) pathways to erythropoietin (Epo) and interleukin (IL)-3-induced proliferation of factor-dependent hematopoietic cells. Lack of cytokine-induced PI3K activation caused by receptor mutation or treatment with a specific inhibitor (LY294002) did not prevent proliferation but resulted in an increase in the G1 phase content and doubling time of cell cultures. The reduced proliferation of cells lacking cytokine-induced PI3K activity could be partially restored by overexpressing constitutively active Akt. Inhibition of PI3K activity decreased the proportion of cytokine-treated cells entering S phase and was associated with a significant reduction in cytokine-induced phosphorylation and activation of Cdk2. By contrast, Cdk4 activity and p27(Kip1) expression were not significantly altered by inhibition of PI3K. Together, these observations identify a mechanism through which cytokine-activated PI3K contributes to G1 to S phase progression in factor-dependent hematopoietic cells by enhancing the phosphorylation and activation of Cdk2.


Asunto(s)
Quinasas CDC2-CDC28/metabolismo , Citocinas/metabolismo , Células Madre Hematopoyéticas/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Proteínas de Ciclo Celular/metabolismo , División Celular/efectos de los fármacos , División Celular/fisiología , Línea Celular , Quinasa 2 Dependiente de la Ciclina , Quinasa 4 Dependiente de la Ciclina , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Quinasas Ciclina-Dependientes/metabolismo , Citocinas/farmacología , Inhibidores Enzimáticos/farmacología , Eritropoyetina/metabolismo , Eritropoyetina/farmacología , Fase G1/efectos de los fármacos , Fase G1/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Interleucina-3/metabolismo , Interleucina-3/farmacología , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Fase S/efectos de los fármacos , Fase S/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA