Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cancer Sci ; 114(7): 2882-2894, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37137487

RESUMEN

ANP32B, a member of the acidic leucine-rich nuclear phosphoprotein 32 kDa (ANP32) family of proteins, is critical for normal development because its constitutive knockout mice are perinatal lethal. It is also shown that ANP32B acts as a tumor-promoting gene in some kinds of cancer such as breast cancer and chronic myelogenous leukemia. Herein, we observe that ANP32B is lowly expressed in B-cell acute lymphoblastic leukemia (B-ALL) patients, which correlates with poor prognosis. Furthermore, we utilized the N-myc or BCR-ABLp190 -induced B-ALL mouse model to investigate the role of ANP32B in B-ALL development. Intriguingly, conditional deletion of Anp32b in hematopoietic cells significantly promotes leukemogenesis in two B-ALL mouse models. Mechanistically, ANP32B interacts with purine rich box-1 (PU.1) and enhances the transcriptional activity of PU.1 in B-ALL cells. Overexpression of PU.1 dramatically suppresses B-ALL progression, and highly expressed PU.1 significantly reverses the accelerated leukemogenesis in Anp32b-deficient mice. Collectively, our findings identify ANP32B as a suppressor gene and provide novel insight into B-ALL pathogenesis.


Asunto(s)
Linfoma de Burkitt , Leucemia Mieloide , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Ratones , Proteínas Nucleares/genética , Ratones Noqueados , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas de Fusión bcr-abl , Proteínas del Tejido Nervioso/metabolismo , Proteínas de Ciclo Celular/metabolismo
2.
Blood ; 138(24): 2485-2498, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34359074

RESUMEN

Proper regulation of p53 signaling is critical for the maintenance of hematopoietic stem cells (HSCs) and leukemic stem cells (LSCs). The hematopoietic cell-specific mechanisms regulating p53 activity remain largely unknown. Here, we demonstrate that conditional deletion of acidic leucine-rich nuclear phosphoprotein 32B (ANP32B) in hematopoietic cells impairs repopulation capacity and postinjury regeneration of HSCs. Mechanistically, ANP32B forms a repressive complex with p53 and thus inhibits the transcriptional activity of p53 in hematopoietic cells, and p53 deletion rescues the functional defect in Anp32b-deficient HSCs. Of great interest, ANP32B is highly expressed in leukemic cells from patients with chronic myelogenous leukemia (CML). Anp32b deletion enhances p53 transcriptional activity to impair LSC function in a murine CML model and exhibits synergistic therapeutic effects with tyrosine kinase inhibitors in inhibiting CML propagation. In summary, our findings provide a novel strategy to enhance p53 activity in LSCs by inhibiting ANP32B and identify ANP32B as a potential therapeutic target in treating CML.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Células Madre Neoplásicas/patología , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Ratones , Células Madre Neoplásicas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Proteína p53 Supresora de Tumor/genética
3.
Basic Res Cardiol ; 108(4): 364, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23744058

RESUMEN

Nkx2.5 plays protective roles in cardiac homeostasis and survival in the postnatal hearts. However, the underlying molecular mechanisms that mediate the protective functions of Nkx2.5 remain unknown. Here, we showed that Nkx2.5 was downregulated in response to various stresses and was required for protection against the stress-induced apoptosis of cardiomyocytes. SIRT1, a member of the sirtuin family of proteins, was found to be a direct transcriptional target of Nkx2.5 and was required for the Nkx2.5-mediated protection of cardiomyocytes from doxorubicin (DOX)-induced apoptosis. Moreover, using chromatin immunoprecipitation assays, we found that Nkx2.5 was able to bind to the SIRT1 promoter and that this binding was significantly decreased in DOX-treated mouse hearts. Furthermore, the cardiac-specific overexpression of SIRT1 decreased the DOX-induced apoptosis of cardiomyocytes in SIRT1 transgenic mouse hearts compared with the hearts of their wild-type littermates. These findings demonstrate that SIRT1 acts as a direct transcriptional target of Nkx2.5 that maintains cardiomyocyte homeostasis and survival.


Asunto(s)
Proteínas de Homeodominio/fisiología , Miocitos Cardíacos/fisiología , Sirtuina 1/fisiología , Estrés Fisiológico/fisiología , Factores de Transcripción/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Doxorrubicina/farmacología , Proteína Homeótica Nkx-2.5 , Homeostasis/fisiología , Ratones , Ratones Transgénicos , Modelos Animales , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Sirtuina 1/genética , Regulación hacia Arriba/fisiología
4.
Circ Res ; 109(6): 639-48, 2011 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-21778425

RESUMEN

RATIONALE: Inactivation of the p66Shc adaptor protein confers resistance to oxidative stress and protects mice from aging-associated vascular diseases. However, there is limited information about the negative regulating mechanisms of p66Shc expression in the vascular system. OBJECTIVE: In this study, we investigated the role of SIRT1, a class III histone deacetylase, in the regulation of p66Shc expression and hyperglycemia-induced endothelial dysfunction. METHODS AND RESULTS: Expressions of p66Shc gene transcript and protein were significantly increased by different kinds of class III histone deacetylase (sirtuin) inhibitors in human umbilical vein endothelial cells and 293A cells. Adenoviral overexpression of SIRT1 inhibited high-glucose-induced p66Shc upregulation in human umbilical vein endothelial cells. Knockdown of SIRT1 increased p66Shc expression and also increased the expression levels of plasminogen activator inhibitor-1 expression, but decreased manganese superoxide dismutase expression in high-glucose conditions. However, knockdown of p66Shc significantly reversed the effects of SIRT1 knockdown. In addition, p66Shc overexpression significantly decreased manganese superoxide dismutase expression and increased plasminogen activator inhibitor-1 expression in high-glucose conditions, which were recovered by SIRT1 overexpression. Moreover, compared to streptozotocin-induced wild-type diabetic mice, endothelium-specific SIRT1 transgenic diabetic mice had decreased p66Shc expression at both the mRNA and the protein levels, improved endothelial function, and reduced accumulation of nitrotyrosine and 8-OHdG (markers of oxidative stress). We further found that SIRT1 was able to bind to the p66Shc promoter (-508 bp to -250 bp), resulting in a decrease in the acetylation of histone H3 bound to the p66Shc promoter region. CONCLUSION: Our findings indicate that repression of p66Shc expression by SIRT1 contributes to the protection of hyperglycemia-induced endothelial dysfunction.


Asunto(s)
Regulación hacia Abajo/genética , Endotelio Vascular/metabolismo , Hiperglucemia/genética , Proteínas Adaptadoras de la Señalización Shc/antagonistas & inhibidores , Sirtuina 1/fisiología , Envejecimiento/genética , Animales , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Endotelio Vascular/patología , Células HEK293 , Humanos , Hiperglucemia/patología , Hiperglucemia/prevención & control , Inmunidad Innata/genética , Masculino , Ratones , Ratones Transgénicos , Estrés Oxidativo/genética , Estabilidad Proteica , Proteínas Adaptadoras de la Señalización Shc/biosíntesis , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src
5.
Circ Res ; 108(10): 1180-9, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21474819

RESUMEN

RATIONALE: Vascular smooth muscle cell (VSMC) proliferation and migration are crucial events involved in the pathophysiology of vascular diseases. Sirtuin 1 (SIRT1), a class III histone deacetylase (HDAC), has been reported to have the function of antiatherosclerosis, but its role in neointima formation remains unknown. OBJECTIVE: The present study was designed to investigate the role of SIRT1 in the regulation of neointima formation and to elucidate the underlying mechanisms. METHODS AND RESULTS: A decrease in SIRT1 expression was observed following carotid artery ligation. smooth muscle cell (SMC)-specific human SIRT1 transgenic (Tg) mice were generated. SIRT1 overexpression substantially inhibited neointima formation after carotid artery ligation or carotid artery wire injury. In the intima of injured carotid arteries, VSMC proliferation (proliferating cell nuclear antigen (PCNA)-positive cells) was significantly reduced. SIRT1 overexpression markedly inhibited VSMC proliferation and migration and induced cell cycle arrest at G1/S transition in vitro. Accordingly, SIRT1 overexpression decreased the induction of cyclin D1 and matrix metalloproteinase-9 (MMP-9) expression by treatment with serum and TNF-α, respectively, whereas RNAi knockdown of SIRT1 resulted in the opposite effect. Decreased cyclin D1 and MMP-9 expression/activity were also observed in injured carotid arteries from SMC-SIRT1 Tg mice. Furthermore, 2 targets of SIRT1, c-Fos and c-Jun, were involved in the downregulation of cyclin D1 and MMP-9 expression. CONCLUSIONS: Our findings demonstrate the inhibitory effect of SIRT1 on the VSMC proliferation and migration that underlie neointima formation and implicate SIRT1 as a potential target for intervention in vascular diseases.


Asunto(s)
Traumatismos de las Arterias Carótidas/metabolismo , Neointima/etiología , Neointima/metabolismo , Sirtuina 1/fisiología , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Células Cultivadas , Humanos , Ligadura , Masculino , Ratones , Ratones Transgénicos , Neointima/patología , Ratas , Ratas Sprague-Dawley , Sirtuina 1/biosíntesis
6.
J Hematol Oncol ; 16(1): 9, 2023 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-36774506

RESUMEN

BACKGROUND: Selectively targeting leukemia stem cells (LSCs) is a promising approach in treating acute myeloid leukemia (AML), for which identification of such therapeutic targets is critical. Increasing lines of evidence indicate that FBXO22 plays a critical role in solid tumor development and therapy response. However, its potential roles in leukemogenesis remain largely unknown. METHODS: We established a mixed lineage leukemia (MLL)-AF9-induced AML model with hematopoietic cell-specific FBXO22 knockout mice to elucidate the role of FBXO22 in AML progression and LSCs regulation, including self-renewal, cell cycle, apoptosis and survival analysis. Immunoprecipitation combined with liquid chromatography-tandem mass spectrometry analysis, Western blotting and rescue experiments were performed to study the mechanisms underlying the oncogenic role of FBXO22. RESULTS: FBXO22 was highly expressed in AML, especially in MLL-rearranged (MLLr) AML. Upon FBXO22 knockdown, human MLLr leukemia cells presented markedly increased apoptosis. Although conditional deletion of Fbxo22 in hematopoietic cells did not significantly affect the function of hematopoietic stem cells, MLL-AF9-induced leukemogenesis was dramatically abrogated upon Fbxo22 deletion, together with remarkably reduced LSCs after serial transplantations. Mechanistically, FBXO22 promoted degradation of BACH1 in MLLr AML cells, and overexpression of BACH1 suppressed MLLr AML progression. In line with this, heterozygous deletion of BACH1 significantly reversed delayed leukemogenesis in Fbxo22-deficient mice. CONCLUSIONS: FBXO22 promotes MLLr AML progression by targeting BACH1 and targeting FBXO22 might be an ideal strategy to eradicate LSCs without influencing normal hematopoiesis.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico , Proteínas F-Box , Leucemia Mieloide Aguda , Receptores Citoplasmáticos y Nucleares , Animales , Humanos , Ratones , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Ciclo Celular , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/patología , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Células Madre Neoplásicas/patología , Receptores Citoplasmáticos y Nucleares/metabolismo
7.
J Biol Chem ; 285(10): 7097-110, 2010 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-20042607

RESUMEN

SIRT1 (Sirtuin type 1), a mammalian orthologue of yeast SIR2 (silent information regulator 2), has been shown to mediate a variety of calorie restriction (CR)-induced physiological events, such as cell fate regulation via deacetylation of the substrate proteins. However, whether SIRT1 deacetylates activator protein-1 (AP-1) to influence its transcriptional activity and target gene expression is still unknown. Here we demonstrate that SIRT1 directly interacts with the basic leucine zipper domains of c-Fos and c-Jun, the major components of AP-1, by which SIRT1 suppressed the transcriptional activity of AP-1. This process requires the deacetylase activity of SIRT1. Notably, SIRT1 reduced the expression of COX-2, a typical AP-1 target gene, and decreased prostaglandin E(2) (PGE(2)) production of peritoneal macrophages (pMPhis). pMPhis with SIRT1 overexpression displayed improved phagocytosis and tumoricidal functions, which are associated with depressed PGE(2). Furthermore, SIRT1 protein level was up-regulated in CR mouse pMPhis, whereas elevated SIRT1 decreased COX-2 expression and improved PGE(2)-related macrophage functions that were reversed following inhibition of SIRT1 deacetylase activity. Thus, our results indicate that SIRT1 may be a mediator of CR-induced macrophage regulation, and its deacetylase activity contributes to the inhibition of AP-1 transcriptional activity and COX-2 expression leading to amelioration of macrophage function.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Regulación de la Expresión Génica , Macrófagos/fisiología , Sirtuina 1/metabolismo , Factor de Transcripción AP-1/metabolismo , Animales , Restricción Calórica , Línea Celular , Ciclooxigenasa 2/genética , Humanos , Leucina Zippers , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-fos/química , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/química , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Distribución Aleatoria , Sirtuina 1/genética , Factor de Transcripción AP-1/genética , Transcripción Genética
8.
Circ Res ; 104(10): 1160-8, 2009 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-19359600

RESUMEN

The paraoxonase (PON) gene cluster consists of the PON1, PON2, and PON3 genes, each of which can individually inhibit atherogenesis. To analyze the functions of the PON gene cluster (PC) in atherogenesis and plaque stability, human PC transgenic (Tg) mice were generated using bacterial artificial chromosome. The high-density lipoprotein from Tg mice exhibited increased paraoxonase activity. When crossed to the ApoE-null background and challenged by high-fat diet, PC Tg/ApoE-null mice formed significantly fewer atherosclerotic lesions. However overexpression of the PC transgene had no additive effect on atherosclerosis compared to the overexpression of the single PON1 or PON3 transgene. Plaques from PC Tg/ApoE-null mice exhibited increased levels of collagen and smooth muscle cells, and reduced levels of macrophages and lipid, compared with those from ApoE-null mice, indicating lesions of PC Tg/ApoE-null mice had characteristics of more stable plaques than those of ApoE-null mice. PC transgene enhanced high-density lipoprotein ability to protect low-density lipoprotein against oxidation in vitro. Serum intercellular adhesion molecule-1 and monocyte chemoattractant protein-1 were also repressed by PC transgene. Proatherogenic reactions of Tg mouse peritoneal macrophages induced by oxidized low-density lipoprotein were inhibited by PC transgene, as indicated by reduced reactive oxygen species generation, inflammation, matrix metalloproteinase-9 expression, and foam cell formation. Our results demonstrate that the PC transgene not only represses atherogenesis but also promotes atherosclerotic plaque stability in vivo. PC may therefore be a useful target for atherosclerosis treatment.


Asunto(s)
Apolipoproteínas E/metabolismo , Arildialquilfosfatasa/genética , Aterosclerosis/fisiopatología , Familia de Multigenes/genética , Animales , Apolipoproteínas E/genética , Aterosclerosis/etiología , Aterosclerosis/genética , Quimiocina CCL2/sangre , Grasas de la Dieta/efectos adversos , Modelos Animales de Enfermedad , Humanos , Molécula 1 de Adhesión Intercelular/sangre , Lipoproteínas HDL/metabolismo , Lipoproteínas LDL/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Oxidación-Reducción
9.
Genesis ; 48(5): 343-50, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20143346

RESUMEN

Wnt signaling regulates cortical and hippocampal development. In a previous study we found that a particular Wnt receptor, Frizzled9 (Fzd9), was selectively expressed in both the developing and adult hippocampus. Taking advantage of the specificity of this promoter, we generated a transgenic cre mouse line using the putative control elements of the Fzd9 gene. In the Fzd9-cre mice, Cre is mainly detected in the developing cortex and hippocampus and is confined to the CA fields and dentate gyrus in adults. Furthermore, by crossing the Fzd9-cre mouse with the ROSA26 reporter line, we examined the activity of Cre and found that it has very high recombination efficiency. Thus, this mouse line will likely prove to be a useful tool for studying cortical and hippocampal development via activation or inactivation of interesting genes.


Asunto(s)
Corteza Cerebral/embriología , Embrión de Mamíferos/embriología , Hipocampo/embriología , Integrasas/metabolismo , Animales , Corteza Cerebral/metabolismo , Embrión de Mamíferos/metabolismo , Receptores Frizzled , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/metabolismo , Inmunohistoquímica , Integrasas/genética , Operón Lac/genética , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Regiones Promotoras Genéticas/genética , Proteínas/genética , Proteínas/metabolismo , ARN no Traducido , Receptores de Neurotransmisores/genética , Factores de Tiempo , beta-Galactosidasa/metabolismo
10.
Biochem Biophys Res Commun ; 397(3): 569-75, 2010 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-20617556

RESUMEN

The proinflammatory cytokine TNF-alpha plays an important role in stimulating inflammatory responses of vascular smooth muscle cells (VSMCs). The anti-inflammatory function of Sirtuin 1 (SIRT1), a NAD-dependent class III histone/protein deacetylase, has been well documented, but how SIRT1 is regulated under inflammatory conditions is largely unknown. In the present research, we showed that levels of SIRT1 mRNA and protein expression increased in TNF-alpha-treated VSMCs. Overexpression of the p65/RelA subunit of NF-kappaB, a TNF-alpha-activated inflammatory transcription factor, in A7r5 cells, upregulated SIRT1 mRNA and protein expression as well as SIRT1 promoter activity, while knockdown of endogenous p65/RelA expression by RNAi not only led to a decrease in SIRT1's basal protein expression and promoter activity, but almost abolished the TNF-alpha-induced elevation of SIRT1 protein expression and SIRT1 promoter activity. Furthermore, using promoter deletion analysis and chromatin immunoprecipitation assays, we found that p65/RelA bound to the SIRT1 promoter at a consensus NF-kappaB binding site. Our study indicates that p65/RelA mediates the TNF-alpha-induced elevated expression of SIRT1 in VSMCs, shedding new light on the regulation of SIRT1 under inflammatory conditions.


Asunto(s)
Inflamación/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Sirtuina 1/biosíntesis , Factor de Transcripción ReIA/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Humanos , Inflamación/genética , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Biosíntesis de Proteínas , ARN Mensajero/biosíntesis , Sirtuina 1/genética , Factor de Transcripción ReIA/genética , Factor de Necrosis Tumoral alfa/genética
11.
Zhonghua Yi Xue Za Zhi ; 89(1): 48-53, 2009 Jan 06.
Artículo en Zh | MEDLINE | ID: mdl-19489245

RESUMEN

OBJECTIVE: To study the morphological behavior, the expression of MCP-1 and PDGF-B of distal anastomotic neointima after expanded polytetrafluoroethylene (ePTFE) bypass grafting. And to identify the effects of Batroxobin (BX) on distal anastomotic intimal hyperplasia. METHODS: 12 adult mongrel dogs underwent end to side bypass grafting in left common carotid artery using ePTFE vascular graft (6mm in diameter, 5cm in length) were divided randomly into two groups. All 12 bypass grafts were removed 28 days after grafting and distal anastomotic parts were obtained. Using HE, Masson, IHC, RT-PCR and western blot, we analyzed the thickness, relative content of ECM, SMC PCNA index, expression of MCP-1 and PDGF-B in distal anastomotic neointima. And we compared the differences between the two groups. RESULTS: 28 day after grafting, the neointima was formed by SMC and ECM and was covered by epithelial cells. After using BX, the thickness of neointima was reduced [( 381.3 +/- 144.7) microm versus (213.8 +/- 29.0) microm, P < 0.05] and the relative content of ECM were significantly decreased (P = 0.006). The PCNA index (P = 0.109) and PDGF-B level (P = 0.055 by RT-PCR and P = 0.337 by WB) between the two groups had no significant difference. MCP-1 level was markedly decreased (P = 0.025 by RT-PCR and P = 0.016 by WB). CONCLUSIONS: Using BX after ePTFE grafting could effectively reduce the relative content of ECM, reduce the expression of MCP-1 and decrease the thickness of neointima in the early time.


Asunto(s)
Batroxobina/farmacología , Arteria Carótida Común/cirugía , Estomas Quirúrgicos/patología , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Anastomosis Quirúrgica , Animales , Prótesis Vascular , Quimiocina CCL2/metabolismo , Perros , Femenino , Hiperplasia , Masculino , Politetrafluoroetileno , Proteínas Proto-Oncogénicas c-sis/metabolismo , Procedimientos Quirúrgicos Vasculares
12.
Cardiovasc Res ; 80(2): 191-9, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18689793

RESUMEN

AIMS: Hazardous environmental and genetic factors can damage endothelial cells to induce atherosclerotic vascular disease. Recent studies suggest that class III deacetylase SIRT1 may promote cell survival via novel antioxidative mechanisms. The current study tested the hypothesis that SIRT1, specifically overexpressed in the endothelium, is atheroprotective. METHODS AND RESULTS: Human umbilical vein endothelial cells (HUVECs) were used to study the effects of oxidized low-density lipoprotein (LDL) on SIRT1 expression. Endothelial cell-specific SIRT1 transgenic (SIRT1-Tg) mice were used to study the effects of SIRT1 on aortic vascular tone. SIRT1-Tg mice were crossed with apolipoprotein E null (apoE(-/-)) mice to obtain SIRT1-Tg/apoE(-/-) mice for the analysis of atherogenesis in the presence of endothelial overexpression of SIRT1. SIRT1 expression in HUVECs was increased by the treatment with oxidative LDL. Adenoviral-mediated overexpression of SIRT1 was protective of apoptosis of HUVECs. Calorie restriction increased, whereas high-fat diet decreased, the SIRT1 expression in mouse aortas. In SIRT1-Tg mice, high fat-induced impairment in endothelium-dependent vasorelaxation was improved compared with that of wild-type littermates. This was accompanied by an upregualtion of aortic endothelial nitric oxide synthase expression in the SIRT1-Tg mice. The SIRT1-Tg/apoE(-/-) mice had less atherosclerotic lesions compared with apoE(-/-) controls, without affecting blood lipids and glucose levels. CONCLUSION: These results suggest that endothelium-specific SIRT1 overexpression likely suppresses atherogenesis via improving endothelial cell survival and function.


Asunto(s)
Apolipoproteínas E/deficiencia , Aterosclerosis/prevención & control , Endotelio Vascular/enzimología , Sirtuinas/metabolismo , Animales , Apolipoproteínas E/genética , Apoptosis , Aterosclerosis/enzimología , Aterosclerosis/etiología , Aterosclerosis/fisiopatología , Glucemia/metabolismo , Restricción Calórica , Células Cultivadas , Grasas de la Dieta , Modelos Animales de Enfermedad , Endotelio Vascular/patología , Endotelio Vascular/fisiopatología , Humanos , Lípidos/sangre , Lipoproteínas LDL/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III , Sirtuina 1 , Sirtuinas/genética , Regulación hacia Arriba , Vasodilatación
13.
Genesis ; 46(10): 523-9, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18802960

RESUMEN

Wnt signaling plays an important role in regulating cortical and hippocampal development, but many of the other molecular mechanisms underlying dorsal telencephalic development are largely unknown. We are taking advantage of the highly regionalized expression patterns of signaling components of the Wnt pathway to generate new mouse lines that will be useful for studying forebrain development. Here, we describe a transgenic mouse line where Cre is driven by the promoter of the Wnt receptor, Frizzled10. In these mice, Cre activity is mainly detected in the dorsal telencephalon during development and is confined to the pyramidal cell fields in the adult hippocampus. The Cre recombinase has very high efficiency when assayed by crossing the transgenic line with the ROSA26 reporter line. Thus, this Cre line will be useful for the study of dorsal telencephalic development and conditional inactivation of target genes in the cortex and hippocampus.


Asunto(s)
Receptores Frizzled/genética , Integrasas/genética , Receptores Acoplados a Proteínas G/genética , Telencéfalo/embriología , Telencéfalo/enzimología , Animales , Receptores Frizzled/fisiología , Hipocampo/enzimología , Hipocampo/fisiología , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas/genética , Células Piramidales/enzimología , Células Piramidales/fisiología , Receptores Acoplados a Proteínas G/fisiología , Telencéfalo/fisiología , Transgenes
14.
Circulation ; 115(14): 1885-94, 2007 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-17389268

RESUMEN

BACKGROUND: A20 was originally characterized as a tumor necrosis factor-inducible gene in human umbilical vein endothelial cells. As an inhibitor of nuclear factor-kappaB signaling, A20 protects against apoptosis, inflammation, and cardiac hypertrophy. In the present study, we tested the hypothesis that cardiac-specific overexpression of A20 could protect the heart from myocardial infarction. METHODS AND RESULTS: We investigated the role of constitutive human A20 expression in acute myocardial infarction using a transgenic model. Transgenic mice containing the human A20 gene under the control of the alpha-myosin heavy chain promoter were constructed. Myocardial infarction was produced by coronary ligation in A20 transgenic mice and control animals. The extent of infarction was then quantified by 2-dimensional and M-mode echocardiography and by molecular and pathological analyses of heart samples in infarct and remote heart regions 7 days after myocardial infarction. Constitutive overexpression of A20 in the murine heart resulted in attenuated infarct size and improved cardiac function 7 days after myocardial infarction. Significantly, we found a decrease in nuclear factor-kappaB signaling and apoptosis, as well as proinflammatory response, cardiac remodeling, and interstitial fibrosis, in noninfarct regions in the hearts of constitutive A20-expressing animals compared with control animals. CONCLUSIONS: Cardiac-specific overexpression of A20 improves cardiac function and inhibits cardiac remodeling, apoptosis, inflammation, and fibrosis after acute myocardial infarction.


Asunto(s)
Ventrículos Cardíacos/fisiopatología , Hipertrofia Ventricular Izquierda/prevención & control , Péptidos y Proteínas de Señalización Intracelular/fisiología , Infarto del Miocardio/patología , FN-kappa B/antagonistas & inhibidores , Proteínas Nucleares/fisiología , Disfunción Ventricular Izquierda/prevención & control , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Citocinas/sangre , Proteínas de Unión al ADN , Fibrosis , Genes Sintéticos , Humanos , Hipertrofia Ventricular Izquierda/etiología , Quinasa I-kappa B/análisis , Inflamación/etiología , Mediadores de Inflamación/análisis , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Transgénicos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/patología , FN-kappa B/fisiología , Péptidos Natriuréticos/análisis , Neutrófilos/patología , Proteínas Nucleares/genética , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal , Método Simple Ciego , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Factor de Necrosis Tumoral alfa/fisiología , Ultrasonografía , Disfunción Ventricular Izquierda/etiología , Miosinas Ventriculares/genética , Remodelación Ventricular/fisiología
15.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 29(3): 441-7, 2007 Jun.
Artículo en Zh | MEDLINE | ID: mdl-17633479

RESUMEN

The silent information regulator protein 2 (Sir2) and its homologues play an important role in the regulation of cellular physiological processes such as survival, apoptosis, and aging. SIRT1, the mammalian Sir 2 homologue, has been shown to deacetylate a wide range of non-histone substrates and histone substrates. It has been constantly reported that SIRT1 may be associated with the occurrence of metabolic syndrome, genomic homeostasis, tumors, and neurodegenerative diseases. Calorie restriction may mitigate many major diseases in rodent models by SIRT1-mediated deacetylase activity and prolong the life expectancies in these animals. Therefore, SIRT1 may be emphasized as a new therapy target for many different diseases.


Asunto(s)
Sirtuina 1/fisiología , Animales , Restricción Calórica , Humanos , Longevidad , Sirtuina 1/genética , Especificidad por Sustrato
16.
Free Radic Biol Med ; 40(10): 1756-75, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16767845

RESUMEN

Cardiac hypertrophy is a major cause of morbidity and mortality worldwide. Recent in vitro and in vivo studies have suggested that reactive oxygen species (ROS) may play an important role in cardiac hypertrophy. It was therefore thought to be of particular value to examine the effects of antioxidants on cardiac hypertrophy. Epigallocatechin-3-gallate (EGCG) is a major bioactive polyphenol present in green tea and a potent antioxidant. The current study was designed to test the hypothesis that EGCG inhibits cardiac hypertrophy in vitro and in vivo. In this study, we investigated the effects of EGCG on angiotensin II- (Ang II) and pressure-overload-induced cardiac hypertrophy. Our results showed that EGCG attenuated Ang II- and pressure-overload-mediated cardiac hypertrophy. Both reactive oxygen species generation and NADPH oxidase expressions induced by Ang II and pressure overload were suppressed by EGCG. The increased hypertension by pressure overload was almost completely blocked after EGCG treatment. Further studies showed that EGCG inhibited Ang II-induced NF-kappaB and AP-1 activation. Inhibition of the activity of NF-kappaB was through blocking ROS-dependent p38 and JNK signaling pathways, whereas inhibition of AP-1 activation was via blocking EGFR transactivation and its downstream events ERKs/PI3K/Akt/mTOR/p70(S6K). The combination of these actions resulted in repressing the reactivation of ANP and BNP, and ultimately preventing the progress of cardiac hypertrophy. These findings indicated that EGCG prevents the development of cardiac hypertrophy through ROS-dependent and -independent mechanisms involving inhibition of different intracellular signaling transductional pathways.


Asunto(s)
Antioxidantes/farmacología , Cardiomegalia/prevención & control , Catequina/análogos & derivados , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Northern Blotting , Western Blotting , Cardiomegalia/etiología , Catequina/farmacología , Ensayo de Cambio de Movilidad Electroforética , Activación Enzimática/efectos de los fármacos , Receptores ErbB/efectos de los fármacos , Receptores ErbB/metabolismo , Hipertensión/inducido químicamente , Hipertensión/complicaciones , MAP Quinasa Quinasa 4/efectos de los fármacos , MAP Quinasa Quinasa 4/metabolismo , Masculino , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología , Factor de Transcripción AP-1/efectos de los fármacos , Factor de Transcripción AP-1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
J Agric Food Chem ; 61(40): 9666-71, 2013 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-24020431

RESUMEN

Utilization of curcumin has been limited due to its poor oral bioavailability. Oral bioavailability of hydrophobic compounds might be elevated via encapsulation in artificial seed oil bodies. This study aimed to improve oral bioavailability of curcumin via this encapsulation. Unfortunately, curcumin was indissoluble in various seed oils. A mixed dissolvent formula was used to dissolve curcumin, and the admixture was successfully encapsulated in artificial oil bodies stabilized by recombinant sesame caleosin. The artificial oil bodies of relatively small sizes (150 nm) were stably solidified in the forms of powder and tablet. Oral bioavailability of curcumin with or without encapsulation in artificial oil bodies was assessed in Sprague-Dawley male rats. The results showed that encapsulation of curcumin significantly elevated its bioavailability and provided the highest maximum whole blood concentration (Cmax), 37 ± 28 ng/mL, in the experimental animals 45 ± 17 min (t(max)) after oral administration. Relative bioavailability calculated on the basis of the area under the plasma concentration-time curve (AUC) was increased by 47.7 times when curcumin was encapsulated in the artificial oil bodies. This novel formulation of artificial oil bodies seems to possess great potential to encapsulate hydrophobic drugs for oral administration.


Asunto(s)
Química Farmacéutica , Curcumina/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Proteínas de Unión al Calcio/metabolismo , Curcumina/administración & dosificación , Masculino , Tamaño de la Partícula , Proteínas de Plantas/metabolismo , Polvos/administración & dosificación , Polvos/química , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo , Aceite de Sésamo/química , Comprimidos/administración & dosificación , Comprimidos/química
19.
Cardiovasc Res ; 84(2): 292-9, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19541669

RESUMEN

AIMS: Hypertension is one of the major risk factors for cardiovascular diseases. Endothelial cells (ECs) exert important functions in the regulation of blood pressure. A novel gene, IC53, as an isoform of the cyclin-dependent kinase (CDK)-binding protein gene C53, is mainly expressed in vascular ECs and is upregulated in the failing heart of rats. Overexpression of IC53 promotes proliferation of ECs. To examine whether IC53 plays a role in the regulation of vascular tone and blood pressure, we constructed a transgenic (tg) mouse model of the IC53 gene and studied its phenotypes relevant to vascular function. METHODS AND RESULTS: IC53 cDNA was cloned from a human aorta cDNA library. Using the endothelium-specific VE-cadherin promoter, we constructed tg mice in which IC53 was specifically overexpressed in vascular endothelia and found that the tg mice exhibit elevated systolic blood pressure (SBP) in contrast to the wild-type (wt) controls. Further studies revealed impaired endothelium-dependent vasodilation, reduced nitric oxide (NO) production and decreased endothelial NO synthase (eNOS) expression, and activity in the tg mice. Inhibition of IC53 in human umbilical vein ECs induces upregulation of eNOS activity. CONCLUSION: Our results indicate that IC53 participates in the regulation of vascular homeostasis. Endothelium-specific overexpression of IC53 is associated with elevated SBP, which may be in part attributed to the downregulation of eNOS signalling.


Asunto(s)
Presión Sanguínea , Células Endoteliales/enzimología , Hipertensión/enzimología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Vasodilatación , Animales , Antígenos CD/genética , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/genética , Cadherinas/genética , Proteínas de Ciclo Celular , Células Cultivadas , Clonación Molecular , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Células Endoteliales/efectos de los fármacos , Endotelina-1/sangre , Regulación Enzimológica de la Expresión Génica , Genotipo , Humanos , Hipertensión/genética , Hipertensión/fisiopatología , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Óxido Nítrico/sangre , Óxido Nítrico Sintasa de Tipo III/genética , Fenotipo , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Mensajero/metabolismo , Sístole , Proteínas Supresoras de Tumor , Vasodilatación/efectos de los fármacos , Vasodilatación/genética , Vasodilatadores/farmacología
20.
J Biol Chem ; 283(42): 28436-44, 2008 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-18678879

RESUMEN

The apolipoprotein (apo) AI/CIII/AIV/AV cluster genes are expressed at different levels in the liver and intestine. The apoCIII enhancer, a common regulatory element, regulates the tissue-specific expression of apoAI, apoCIII, and apoAIV but not apoAV. To study this regulation at the chromatin level, the histone modifications and intergenic transcription in the human apoAI/CIII/AIV/AV cluster were investigated in HepG2 and Caco-2 cells and in the livers of transgenic mice carrying the human gene cluster constructs with or without the apoCIII enhancer. We found that both the promoters and the intergenic regions of the apoAI/CIII/AIV genes were hyperacetylated and formed an open subdomain that did not include the apoAV gene. Hepatic and intestinal intergenic transcripts were identified to transcribe bidirectionally with strand preferences along the cluster. The deletion of the apoCIII enhancer influenced both histone modification and intergenic transcription in the apoAI/CIII/AIV gene region. These results demonstrate that the apoCIII enhancer contributes to the maintenance of an active chromatin subdomain of the apoAI/CIII/AIV genes, but not apoAV.


Asunto(s)
Apolipoproteína A-I/biosíntesis , Apolipoproteína C-III/química , Apolipoproteína C-III/genética , Apolipoproteína C-III/fisiología , Apolipoproteínas A/biosíntesis , Histonas/metabolismo , Animales , Apolipoproteína A-I/química , Apolipoproteína A-V , Apolipoproteínas A/química , Células CACO-2 , Línea Celular Tumoral , Humanos , Ratones , Ratones Transgénicos , Modelos Biológicos , Modelos Genéticos , Familia de Multigenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA