Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cardiovasc Res ; 118(1): 305-315, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33119054

RESUMEN

AIMS: The pulmonary vascular tone and hypoxia-induced alterations of the pulmonary vasculature may be regulated by the mitochondrial membrane permeability transition pore (mPTP) that controls mitochondrial calcium load and apoptosis. We thus investigated, if the mitochondrial proteins p66shc and cyclophilin D (CypD) that regulate mPTP opening affect the pulmonary vascular tone. METHODS AND RESULTS: Mice deficient for p66shc (p66shc-/-), CypD (CypD-/-), or both proteins (p66shc/CypD-/-) exhibited decreased pulmonary vascular resistance (PVR) compared to wild-type mice determined in isolated lungs and in vivo. In contrast, systemic arterial pressure was only lower in CypD-/- mice. As cardiac function and pulmonary vascular remodelling did not differ between genotypes, we determined alterations of vascular contractility in isolated lungs and calcium handling in pulmonary arterial smooth muscle cells (PASMC) as underlying reason for decreased PVR. Potassium chloride (KCl)-induced pulmonary vasoconstriction and KCl-induced cytosolic calcium increase determined by Fura-2 were attenuated in all gene-deficient mice. In contrast, KCl-induced mitochondrial calcium increase determined by the genetically encoded Mito-Car-GECO and calcium retention capacity were increased only in CypD-/- and p66shc/CypD-/- mitochondria indicating that decreased mPTP opening affected KCl-induced intracellular calcium peaks in these cells. All mouse strains showed a similar pulmonary vascular response to chronic hypoxia, while acute hypoxic pulmonary vasoconstriction was decreased in gene-deficient mice indicating that CypD and p66shc regulate vascular contractility but not remodelling. CONCLUSIONS: We conclude that p66shc specifically regulates the pulmonary vascular tone, while CypD also affects systemic pressure. However, only CypD acts via regulation of mPTP opening and mitochondrial calcium regulation.


Asunto(s)
Presión Arterial , Calcio/metabolismo , Hipertensión Pulmonar/enzimología , Mitocondrias/enzimología , Peptidil-Prolil Isomerasa F/deficiencia , Arteria Pulmonar/enzimología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Vasoconstricción , Animales , Señalización del Calcio , Proliferación Celular , Células Cultivadas , Peptidil-Prolil Isomerasa F/genética , Modelos Animales de Enfermedad , Eliminación de Gen , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/fisiopatología , Hipoxia/complicaciones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Poro de Transición de la Permeabilidad Mitocondrial/metabolismo , Arteria Pulmonar/fisiopatología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Remodelación Vascular , Resistencia Vascular
2.
Drug Des Devel Ther ; 13: 3199-3206, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31686782

RESUMEN

BACKGROUND: p66Shc, a Src homologue and collagen homologue (Shc) adaptor protein, mediates oxidative stress signaling. The p66Shc-null mice have increased lifespan and enhanced resistance to oxidative stress. Studies have also indicated its potential role in inner ear aging, which can lead to deafness. OBJECTIVE: The aim of this study was to determine the effects of p66Shc down-regulation on the marginal cells (MCs) of the inner ear stria vascularis. METHODS: Primary MCs were isolated from neonatal rats and treated with glucose oxidase to induce oxidative stress. The cells were transduced with adenovirus expressing siRNA, and the knockdown was verified by Western blotting. The reactive oxygen species (ROS) levels and apoptosis were analyzed using the DCFH-DA probe and Annexin-V/7-AAD staining respectively. The ultrastructure of the differentially-treated cells was examined by transmission electron microscopy (TEM).Results: The in vitro oxidative stress model was established successfully in rat MCs. Knockdown of p66Shc alleviated the high ROS levels and apoptosis in the glucose oxidase-treated cells. In addition, glucose oxidase significantly increased the number of peroxisomes in the MCs, which was decreased by p66Shc inhibition. CONCLUSION: Oxidative stress increases p66Shc levels in the marginal cells of the inner ear, which aggravates ROS production and cellular injury. Blocking p66Shc expression can effectively reduce oxidative stress and protect the MCs.


Asunto(s)
Apoptosis , Modelos Animales de Enfermedad , Regulación hacia Abajo , Estrés Oxidativo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Estría Vascular/metabolismo , Animales , Células Cultivadas , Células Epiteliales/metabolismo , Ratas , Ratas Sprague-Dawley , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo
3.
Haematologica ; 104(10): 2040-2052, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30819907

RESUMEN

The Shc family adaptor p66Shc acts as a negative regulator of proliferative and survival signals triggered by the B-cell receptor and, by enhancing the production of reactive oxygen species, promotes oxidative stress-dependent apoptosis. Additionally, p66Shc controls the expression and function of chemokine receptors that regulate lymphocyte traffic. Chronic lymphocytic leukemia cells have a p66Shc expression defect which contributes to their extended survival and correlates with poor prognosis. We analyzed the impact of p66Shc ablation on disease severity and progression in the Eµ-TCL1 mouse model of chronic lymphocytic leukemia. We showed that Eµ-TCL1/p66Shc-/- mice developed an aggressive disease that had an earlier onset, occurred at a higher incidence and led to earlier death compared to that in Eµ-TCL1 mice. Eµ-TCL1/p66Shc-/- mice displayed substantial leukemic cell accumulation in both nodal and extranodal sites. The target organ selectivity correlated with upregulation of chemokine receptors whose ligands are expressed therein. This also applied to chronic lymphocytic leukemia cells, where chemokine receptor expression and extent of organ infiltration were found to correlate inversely with these cells' level of p66Shc expression. p66Shc expression declined with disease progression in Eµ-TCL1 mice and could be restored by treatment with the Bruton tyrosine kinase inhibitor ibrutinib. Our results highlight p66Shc deficiency as an important factor in the progression and severity of chronic lymphocytic leukemia and underscore p66Shc expression as a relevant therapeutic target.


Asunto(s)
Carcinogénesis/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/metabolismo , Receptores de Quimiocina/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Animales , Carcinogénesis/genética , Carcinogénesis/patología , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Receptores de Quimiocina/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo
4.
Am J Physiol Renal Physiol ; 315(6): F1833-F1842, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30207172

RESUMEN

The p66ShcA protein controls cellular responses to oxidative stress, senescence, and apoptosis. Here, we test the hypothesis that aging phenotype(s) commonly associated with the broad category of chronic kidney disease are accelerated in diabetic kidneys and linked to the p66ShcA locus. At the organ level, tissue stem cells antagonize senescent phenotypes by replacing old dysfunctional cells. Using established methods, we isolated a highly purified population of stem cell antigen-1-positive mesenchymal stem cells (Sca-1+ MSCs) from kidneys of wild-type (WT) and p66 knockout (p66 KO) mice. Cells were plated in culture medium containing normal glucose (NG) or high glucose (HG). Reactive oxygen species (ROS) metabolism was substantially increased in WT MSCs in HG medium in association with increased cell death by apoptosis and acquisition of the senescent phenotype. DNA microarray analysis detected striking differences in the expression profiles of WT and p66 KO-MSCs in HG medium. Unexpectedly, the analysis for p66 KO-MSCs revealed upregulation of Wnt genes implicated in self-renewal and differentiation. To test the in vivo consequences of constitutive p66 expression in diabetic kidneys, we crossed the Akita diabetic mouse with the p66KO mouse. Homozygous mutation at the p66 locus delays or prevents aging phenotype(s) in the kidney that may be precursors to diabetic nephropathy.


Asunto(s)
Envejecimiento/metabolismo , Nefropatías Diabéticas/metabolismo , Riñón/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Factores de Edad , Envejecimiento/genética , Envejecimiento/patología , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Senescencia Celular , Nefropatías Diabéticas/genética , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Glucosa/metabolismo , Riñón/patología , Células Madre Mesenquimatosas/patología , Ratones Noqueados , Fenotipo , Especies Reactivas de Oxígeno/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Nicho de Células Madre , Vía de Señalización Wnt
5.
Int J Cardiol ; 268: 179-186, 2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30047409

RESUMEN

AIMS: Hyperglycaemia-induced reactive oxygen species (ROS) are key mediators of cardiac dysfunction. Intensive glycaemic control (IGC) has failed to reduce risk of heart failure in patients with diabetes but the underlying mechanisms remain to be elucidated. The present study investigates whether epigenetic regulation of the pro-oxidant adaptor p66Shc contributes to persistent myocardial dysfunction despite IGC. METHODS AND RESULTS: p66Shc expression was increased in the heart of diabetic mice, and 3-week IGC by slow-release insulin implants did not revert this phenomenon. Sustained p66Shc upregulation was associated with oxidative stress, myocardial inflammation and left ventricular dysfunction, as assessed by conventional and 2D speckle-tracking echocardiography. In vivo gene silencing of p66Shc, performed during IGC, inhibited ROS production and restored cardiac function. Furthermore, we show that dysregulation of methyltransferase DNMT3b and deacetylase SIRT1 causes CpG demethylation and histone 3 acetylation on p66Shc promoter, leading to persistent transcription of the adaptor. Altered DNMT3b/SIRT1 axis in the diabetic heart was explained by upregulation of miR-218 and miR-34a. Indeed, in human cardiomyocytes exposed to high glucose, inhibition of these miRNAs restored the expression of DNMT3b and SIRT1 and erased the adverse epigenetic signatures on p66Shc promoter. Consistently, reprogramming miR-218 and miR-34a attenuated persistent p66Shc expression and ROS generation. CONCLUSIONS: In diabetic left ventricular dysfunction, a complex epigenetic mechanism linking miRNAs and chromatin modifying enzymes drives persistent p66Shc transcription and ROS generation. Our results set the stage for pharmacological targeting of epigenetic networks to alleviate the clinical burden of diabetic cardiomyopathy.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Epigénesis Genética/fisiología , Hiperglucemia/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/biosíntesis , Disfunción Ventricular Izquierda/metabolismo , Animales , Diabetes Mellitus Experimental/genética , Hiperglucemia/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Disfunción Ventricular Izquierda/genética
6.
Sci Rep ; 8(1): 3213, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29453337

RESUMEN

Accumlating evidence have suggested that diabetes mellitus links dementia, notably of Alzheimer's disease (AD). However, the underlying mechanism remains unclear. Several studies have shown oxidative stress (OS) to be one of the major factors in the pathogenesis of diabetic complications. Here we show OS involvement in brain damage in a diabetic animal model that is at least partially mediated through an AD-pathology-independent mechanism apart from amyloid-ß accumulation. We investigated the contribution of the p66Shc signaling pathway to diabetes-related cognitive decline using p66Shc knockout (-/-) mice. p66Shc (-/-) mice have less OS in the brain and are resistant to diabetes-induced brain damage. Moreover, p66Shc (-/-) diabetic mice show significantly less cognitive dysfunction and decreased levels of OS and the numbers of microglia. This study postulates a p66Shc-mediated inflammatory cascade leading to OS as a causative pathogenic mechanism in diabetes-associated cognitive impairment that is at least partially mediated through an AD-pathology-independent mechanism.


Asunto(s)
Disfunción Cognitiva/etiología , Diabetes Mellitus Experimental/complicaciones , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Animales , Inflamación/etiología , Ratones , Ratones Noqueados , Microglía/citología , Estrés Oxidativo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética
7.
Blood ; 130(18): 2006-2017, 2017 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-28931525

RESUMEN

The high proportion of long-term nonprogressors among chronic lymphocytic leukemia (CLL) patients suggests the existence of a regulatory network that restrains the proliferation of tumor B cells. The identification of molecular determinants composing such network is hence fundamental for our understanding of CLL pathogenesis. Based on our previous finding establishing a deficiency in the signaling adaptor p66Shc in CLL cells, we undertook to identify unique phenotypic traits caused by this defect. Here we show that a lack of p66Shc shapes the transcriptional profile of CLL cells and leads to an upregulation of the surface receptor ILT3, the immunoglobulin-like transcript 3 that is normally found on myeloid cells. The ectopic expression of ILT3 in CLL was a distinctive feature of neoplastic B cells and hematopoietic stem cells, thus identifying ILT3 as a selective marker of malignancy in CLL and the first example of phenotypic continuity between mature CLL cells and their progenitors in the bone marrow. ILT3 expression in CLL was found to be driven by Deltex1, a suppressor of antigen receptor signaling in lymphocytes. Triggering of ILT3 inhibited the activation of Akt kinase upon B-cell receptor (BCR) stimulation. This effect was achieved through the dynamic coalescence of ILT3, BCRs, and phosphatidylinositol-3,4,5-trisphosphate 5-phosphatase 1 into inhibitory clusters at the cell surface. Collectively, our findings identify ILT3 as a signature molecule of p66Shc deficiency in CLL and indicate that ILT3 may functionally contribute to a regulatory network controlling tumor progression by suppressing the Akt pathway.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/enzimología , Leucemia Linfocítica Crónica de Células B/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Superficie Celular/metabolismo , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Activación Enzimática , Regulación Leucémica de la Expresión Génica , Humanos , Inmunomodulación/genética , Glicoproteínas de Membrana , Receptores de Superficie Celular/genética , Receptores Inmunológicos , Transducción de Señal , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Células Madre/metabolismo , Transcriptoma/genética , Ubiquitina-Proteína Ligasas/metabolismo , Regulación hacia Arriba/genética
8.
Arterioscler Thromb Vasc Biol ; 36(12): 2394-2403, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27789474

RESUMEN

OBJECTIVE: Diabetes mellitus causes vascular endothelial dysfunction and alters vascular microRNA expression. We investigated whether endothelial microRNA-34a (miR-34a) leads to diabetic vascular dysfunction by targeting endothelial sirtuin1 (Sirt1) and asked whether the oxidative stress protein p66Shc governs miR-34a expression in the diabetic endothelium. APPROACH AND RESULTS: MiR-34a is upregulated, and Sirt1 downregulated, in aortic endothelium of db/db and streptozotocin-induced diabetic mice. Systemic administration of miR-34a inhibitor, or endothelium-specific knockout of miR-34a, prevents downregulation of aortic Sirt1 and rescues impaired endothelium-dependent aortic vasorelaxation induced by diabetes mellitus. Moreover, overexpression of Sirt1 mitigates impaired endothelium-dependent vasorelaxation caused by miR-34a mimic ex vivo. Systemic infusion of miR-34a inhibitor or genetic ablation of endothelial miR-34a prevents downregulation of endothelial Sirt1 by high glucose. MiR-34a is upregulated, Sirt1 is downregulated, and oxidative stress (hydrogen peroxide) is induced in endothelial cells incubated with high glucose or the free fatty acid palmitate in vitro. Increase of hydrogen peroxide and induction of endothelial miR-34a by high glucose or palmitate in vitro is suppressed by knockdown of p66shc. In addition, overexpression of wild-type but not redox-deficient p66Shc upregulates miR-34a in endothelial cells. P66Shc-stimulated upregulation of endothelial miR-34a is suppressed by cell-permeable antioxidants. Finally, mice with global knockdown of p66Shc are protected from diabetes mellitus-induced upregulation of miR-34a and downregulation of Sirt1 in the endothelium. CONCLUSIONS: These data show that hyperglycemia and elevated free fatty acids in the diabetic milieu recruit p66Shc to upregulate endothelial miR-34a via an oxidant-sensitive mechanism, which leads to endothelial dysfunction by targeting Sirt1.


Asunto(s)
Aorta/enzimología , Angiopatías Diabéticas/enzimología , Endotelio Vascular/enzimología , MicroARNs/metabolismo , Sirtuina 1/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Vasodilatación , Animales , Antioxidantes/farmacología , Aorta/efectos de los fármacos , Aorta/fisiopatología , Células Cultivadas , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/genética , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/fisiopatología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Metabolismo Energético , Genotipo , Glucosa/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Estrés Oxidativo , Ácido Palmítico/metabolismo , Fenotipo , Interferencia de ARN , Transducción de Señal , Sirtuina 1/genética , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/genética , Transfección , Proteína p53 Supresora de Tumor/metabolismo , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
9.
Sci Rep ; 6: 25450, 2016 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-27157635

RESUMEN

p66(shc), a member of the ShcA protein family, is essential for cellular response to oxidative stress, and elicits the formation of mitochondrial Reactive Oxygen Species (ROS), thus promoting vasomotor dysfunction and inflammation. Accordingly, mice lacking the p66 isoform display increased resistance to oxidative tissue damage and to cardiovascular disorders. Oxidative stress also contributes to noise-induced hearing loss (NIHL); we found that p66(shc) expression and serine phosphorylation were induced following noise exposure in the rat cochlea, together with markers of oxidative stress, inflammation and ischemia as indicated by the levels of the hypoxic inducible factor (HIF) and the vascular endothelial growth factor (VEGF) in the highly vascularised cochlear lateral region and spiral ganglion. Importantly, p66(shc) knock-out (p66 KO) 126 SvEv adult mice were less vulnerable to acoustic trauma with respect to wild type controls, as shown by preserved auditory function and by remarkably lower levels of oxidative stress and ischemia markers. Of note, decline of auditory function observed in 12 month old WT controls was markedly attenuated in p66KO mice consistent with delayed inner ear senescence. Collectively, we have identified a pivotal role for p66(shc) -induced vascular dysfunction in a common pathogenic cascade shared by noise-induced and age-related hearing loss.


Asunto(s)
Cóclea/irrigación sanguínea , Cóclea/fisiopatología , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Provocada por Ruido/fisiopatología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Animales , Cóclea/metabolismo , Inflamación/patología , Isquemia/metabolismo , Isquemia/patología , Isquemia/fisiopatología , Masculino , Ratones Noqueados , Neovascularización Fisiológica , Oxidación-Reducción , Estrés Oxidativo , Fosforilación , Ratas Wistar , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia
10.
Aging Cell ; 15(3): 446-54, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26968134

RESUMEN

Oxidative stress and telomere attrition are considered the driving factors of aging. As oxidative damage to telomeric DNA favors the erosion of chromosome ends and, in turn, telomere shortening increases the sensitivity to pro-oxidants, these two factors may trigger a detrimental vicious cycle. To check whether limiting oxidative stress slows down telomere shortening and related progeria, we have investigated the effect of p66SHC deletion, which has been shown to reduce oxidative stress and mitochondrial apoptosis, on late-generation TERC (telomerase RNA component)-deficient mice having short telomeres and reduced lifespan. Double mutant (TERC(-/-) p66SHC(-/-) ) mice were generated, and their telomere length, fertility, and lifespan investigated in different generations. Results revealed that p66SHC deletion partially rescues sterility and weight loss, as well as organ atrophy, of TERC-deficient mice, but not their short lifespan and telomere erosion. Therefore, our data suggest that p66SHC-mediated oxidative stress and telomere shortening synergize in some tissues (including testes) to accelerate aging; however, early mortality of late-generation mice seems to be independent of any link between p66SHC-mediated oxidative stress and telomere attrition.


Asunto(s)
Fertilidad/genética , Eliminación de Gen , Longevidad , Progeria/genética , Progeria/patología , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo , Telomerasa/deficiencia , Animales , Daño del ADN , Genotipo , Ratones Endogámicos C57BL , Estrés Oxidativo , Fenotipo , ARN/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/deficiencia , Análisis de Supervivencia , Telomerasa/metabolismo , Acortamiento del Telómero , Pérdida de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA