Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gastroenterology ; 166(1): 139-154, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37739089

RESUMEN

BACKGROUND & AIMS: The dismal prognosis of pancreatic ductal adenocarcinoma (PDAC) is linked to the presence of pancreatic cancer stem-like cells (CSCs) that respond poorly to current chemotherapy regimens. The epigenetic mechanisms regulating CSCs are currently insufficiently understood, which hampers the development of novel strategies for eliminating CSCs. METHODS: By small molecule compound screening targeting 142 epigenetic enzymes, we identified that bromodomain-containing protein BRD9, a component of the BAF histone remodeling complex, is a key chromatin regulator to orchestrate the stemness of pancreatic CSCs via cooperating with the TGFß/Activin-SMAD2/3 signaling pathway. RESULTS: Inhibition and genetic ablation of BRD9 block the self-renewal, cell cycle entry into G0 phase and invasiveness of CSCs, and improve the sensitivity of CSCs to gemcitabine treatment. In addition, pharmacological inhibition of BRD9 significantly reduced the tumorigenesis in patient-derived xenografts mouse models and eliminated CSCs in tumors from pancreatic cancer patients. Mechanistically, inhibition of BRD9 disrupts enhancer-promoter looping and transcription of stemness genes in CSCs. CONCLUSIONS: Collectively, the data suggest BRD9 as a novel therapeutic target for PDAC treatment via modulation of CSC stemness.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animales , Humanos , Ratones , Proteínas que Contienen Bromodominio , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/patología , Gemcitabina , Células Madre Neoplásicas/patología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteína Smad2/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Breast Cancer Res ; 26(1): 108, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38951862

RESUMEN

BACKGROUND: Metastasis, the spread, and growth of malignant cells at secondary sites within a patient's body, accounts for over 90% of cancer-related mortality. Breast cancer is the most common tumor type diagnosed and the leading cause of cancer lethality in women in the United States. It is estimated that 10-16% breast cancer patients will have brain metastasis. Current therapies to treat patients with breast cancer brain metastasis (BCBM) remain palliative. This is largely due to our limited understanding of the fundamental molecular and cellular mechanisms through which BCBM progresses, which represents a critical barrier for the development of efficient therapies for affected breast cancer patients. METHODS: Previous research in BCBM relied on co-culture assays of tumor cells with rodent neural cells or rodent brain slice ex vivo. Given the need to overcome the obstacle for human-relevant host to study cell-cell communication in BCBM, we generated human embryonic stem cell-derived cerebral organoids to co-culture with human breast cancer cell lines. We used MDA-MB-231 and its brain metastatic derivate MDA-MB-231 Br-EGFP, other cell lines of MCF-7, HCC-1806, and SUM159PT. We leveraged this novel 3D co-culture platform to investigate the crosstalk of human breast cancer cells with neural cells in cerebral organoid. RESULTS: We found that MDA-MB-231 and SUM159PT breast cancer cells formed tumor colonies in human cerebral organoids. Moreover, MDA-MB-231 Br-EGFP cells showed increased capacity to invade and expand in human cerebral organoids. CONCLUSIONS: Our co-culture model has demonstrated a remarkable capacity to discern the brain metastatic ability of human breast cancer cells in cerebral organoids. The generation of BCBM-like structures in organoid will facilitate the study of human tumor microenvironment in culture.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Técnicas de Cocultivo , Organoides , Humanos , Organoides/patología , Neoplasias Encefálicas/secundario , Neoplasias Encefálicas/patología , Femenino , Neoplasias de la Mama/patología , Línea Celular Tumoral , Encéfalo/patología , Comunicación Celular
3.
Arterioscler Thromb Vasc Biol ; 41(2): 783-795, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33297755

RESUMEN

OBJECTIVE: Vascular endothelial cells (ECs) play a critical role in maintaining vascular homeostasis. Aberrant EC metabolism leads to vascular dysfunction and metabolic diseases. TFEB (transcription factor EB), a master regulator of lysosome biogenesis and autophagy, has protective effects on vascular inflammation and atherosclerosis. However, the role of endothelial TFEB in metabolism remains to be explored. In this study, we sought to investigate the role of endothelial TFEB in glucose metabolism and underlying molecular mechanisms. Approach and Results: To determine whether endothelial TFEB is critical for glucose metabolism in vivo, we utilized EC-selective TFEB knockout and EC-selective TFEB transgenic mice fed a high-fat diet. EC-selective TFEB knockout mice exhibited significantly impaired glucose tolerance compared with control mice. Consistently, EC-selective TFEB transgenic mice showed improved glucose tolerance. In primary human ECs, small interfering RNA-mediated TFEB knockdown blunts Akt (AKT serine/threonine kinase) signaling. Adenovirus-mediated overexpression of TFEB consistently activates Akt and significantly increases glucose uptake in ECs. Mechanistically, TFEB upregulates IRS1 and IRS2 (insulin receptor substrate 1 and 2). TFEB increases IRS2 transcription measured by reporter gene and chromatin immunoprecipitation assays. Furthermore, we found that TFEB increases IRS1 protein via downregulation of microRNAs (miR-335, miR-495, and miR-548o). In vivo, Akt signaling in the skeletal muscle and adipose tissue was significantly impaired in EC-selective TFEB knockout mice and consistently improved in EC-selective TFEB transgenic mice on high-fat diet. CONCLUSIONS: Our data revealed a critical role of TFEB in endothelial metabolism and suggest that TFEB constitutes a potential molecular target for the treatment of vascular and metabolic diseases.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Glucemia/metabolismo , Células Endoteliales/metabolismo , Intolerancia a la Glucosa/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Tejido Adiposo/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Glucemia/efectos de los fármacos , Células Cultivadas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Femenino , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/tratamiento farmacológico , Intolerancia a la Glucosa/genética , Humanos , Hipoglucemiantes/farmacología , Insulina/sangre , Insulina/farmacología , Proteínas Sustrato del Receptor de Insulina/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
4.
Circulation ; 142(5): 483-498, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32354235

RESUMEN

BACKGROUND: Abdominal aortic aneurysm (AAA) is a severe aortic disease with a high mortality rate in the event of rupture. Pharmacological therapy is needed to inhibit AAA expansion and prevent aneurysm rupture. Transcription factor EB (TFEB), a master regulator of autophagy and lysosome biogenesis, is critical to maintain cell homeostasis. In this study, we aim to investigate the role of vascular smooth muscle cell (VSMC) TFEB in the development of AAA and establish TFEB as a novel target to treat AAA. METHODS: The expression of TFEB was measured in human and mouse aortic aneurysm samples. We used loss/gain-of-function approaches to understand the role of TFEB in VSMC survival and explored the underlying mechanisms through transcriptome and functional studies. Using VSMC-selective Tfeb knockout mice and different mouse AAA models, we determined the role of VSMC TFEB and a TFEB activator in AAA in vivo. RESULTS: We found that TFEB is downregulated in both human and mouse aortic aneurysm lesions. TFEB potently inhibits apoptosis in VSMCs, and transcriptome analysis revealed that TFEB regulates apoptotic signaling pathways, especially apoptosis inhibitor B-cell lymphoma 2. B-cell lymphoma 2 is significantly upregulated by TFEB and is required for TFEB to inhibit VSMC apoptosis. We consistently observed that TFEB deficiency increases VSMC apoptosis and promotes AAA formation in different mouse AAA models. Furthermore, we demonstrated that 2-hydroxypropyl-ß-cyclodextrin, a clinical agent used to enhance the solubility of drugs, activates TFEB and inhibits AAA formation and progression in mice. Last, we found that 2-hydroxypropyl-ß-cyclodextrin inhibits AAA in a VSMC TFEB-dependent manner in mouse models. CONCLUSIONS: Our study demonstrated that TFEB protects against VSMC apoptosis and AAA. TFEB activation by 2-hydroxypropyl-ß-cyclodextrin may be a promising therapeutic strategy for the prevention and treatment of AAA.


Asunto(s)
2-Hidroxipropil-beta-Ciclodextrina/uso terapéutico , Aneurisma de la Aorta Abdominal/prevención & control , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/fisiología , Modelos Animales de Enfermedad , Músculo Liso Vascular/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , 2-Hidroxipropil-beta-Ciclodextrina/farmacología , Aminopropionitrilo/toxicidad , Aneurisma Roto/etiología , Angiotensina II/toxicidad , Animales , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Apoptosis/efectos de los fármacos , Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/biosíntesis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/deficiencia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Colesterol/metabolismo , Regulación hacia Abajo , Evaluación Preclínica de Medicamentos , Mutación con Ganancia de Función , Regulación de la Expresión Génica , Vectores Genéticos/toxicidad , Humanos , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/fisiología , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Transcriptoma/efectos de los fármacos
5.
Arterioscler Thromb Vasc Biol ; 40(6): 1533-1542, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32268786

RESUMEN

OBJECTIVE: Clopidogrel is a commonly used P2Y12 inhibitor to treat and prevent arterial thrombotic events. Clopidogrel is a prodrug that requires bioactivation by CYP (cytochrome P450) enzymes to exert antiplatelet activity. Diabetes mellitus is associated with an increased risk of ischemic events, and impaired ability to generate the active metabolite (AM) from clopidogrel. The objective of this study is to identify the mechanism of clopidogrel resistance in a murine model of diet-induced obesity (DIO). Approach and Results: C57BL/6J mice and IL-1R-/- mice were given high-fat diet for 10 weeks to generate a murine model of diet-induced obesity. Platelet aggregation and carotid arterial thrombosis were assessed in response to clopidogrel treatment. Wild-type DIO mice exhibited resistance to antiplatelet and antithrombotic effects of clopidogrel that was associated with reduced hepatic expression of CYP genes and reduced generation of the AM. IL (Interleukin)-1 receptor-deficient DIO (IL1R-/- DIO) mice showed no resistance to clopidogrel. Lack of resistance was accompanied by increased exposure of the clopidogrel AM. This resistance was also absent when wild-type DIO mice were treated with the conjugate of the clopidogrel AM, DT-678. CONCLUSIONS: These findings indicate that antiplatelet effects of clopidogrel may be impaired in the setting of diabetes mellitus due to reduced prodrug bioactivation related to IL-1 receptor signaling. Therapeutic targeting of P2Y12 in patients with diabetes mellitus using the conjugate of clopidogrel AM may lead to improved outcomes.


Asunto(s)
Clopidogrel/farmacocinética , Clopidogrel/uso terapéutico , Resistencia a Medicamentos , Obesidad/complicaciones , Receptores de Interleucina-1/fisiología , Animales , Trombosis de las Arterias Carótidas/prevención & control , Clopidogrel/farmacología , Sistema Enzimático del Citocromo P-450/genética , Diabetes Mellitus , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Fibrinolíticos , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Microsomas Hepáticos/enzimología , Obesidad/etiología , Obesidad/metabolismo , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria , Profármacos/farmacocinética , Profármacos/uso terapéutico , Receptores de Interleucina-1/deficiencia
6.
Arterioscler Thromb Vasc Biol ; 40(10): 2494-2507, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32787523

RESUMEN

OBJECTIVE: Currently, there are no approved drugs for abdominal aortic aneurysm (AAA) treatment, likely due to limited understanding of the primary molecular mechanisms underlying AAA development and progression. BAF60a-a unique subunit of the SWI/SNF (switch/sucrose nonfermentable) chromatin remodeling complex-is a novel regulator of metabolic homeostasis, yet little is known about its function in the vasculature and pathogenesis of AAA. In this study, we sought to investigate the role and underlying mechanisms of vascular smooth muscle cell (VSMC)-specific BAF60a in AAA formation. Approach and Results: BAF60a is upregulated in human and experimental murine AAA lesions. In vivo studies revealed that VSMC-specific knockout of BAF60a protected mice from both Ang II (angiotensin II)-induced and elastase-induced AAA formation with significant suppression of vascular inflammation, monocyte infiltration, and elastin fragmentation. Through RNA sequencing and pathway analysis, we found that the expression of inflammatory response genes in cultured human aortic smooth muscle cells was significantly downregulated by small interfering RNA-mediated BAF60a knockdown while upregulated upon adenovirus-mediated BAF60a overexpression. BAF60a regulates VSMC inflammation by recruiting BRG1 (Brahma-related gene-1)-a catalytic subunit of the SWI/SNF complex-to the promoter region of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) target genes. Furthermore, loss of BAF60a in VSMCs prevented the upregulation of the proteolytic enzyme cysteine protease CTSS (cathepsin S), thus ameliorating ECM (extracellular matrix) degradation within the vascular wall in AAA. CONCLUSIONS: Our study demonstrated that BAF60a is required to recruit the SWI/SNF complex to facilitate the epigenetic regulation of VSMC inflammation, which may serve as a potential therapeutic target in preventing and treating AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal/prevención & control , Aortitis/prevención & control , Proteínas Cromosómicas no Histona/deficiencia , Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Remodelación Vascular , Animales , Aorta Abdominal/metabolismo , Aorta Abdominal/patología , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Aortitis/genética , Aortitis/metabolismo , Aortitis/patología , Estudios de Casos y Controles , Catepsinas/metabolismo , Células Cultivadas , Proteínas Cromosómicas no Histona/genética , Modelos Animales de Enfermedad , Matriz Extracelular/patología , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Transducción de Señal
7.
Cardiovasc Drugs Ther ; 35(5): 939-951, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-32671602

RESUMEN

PURPOSE: Abdominal aortic aneurysm (AAA) is one of the leading causes of death in the developed world and is currently undertreated due to the complicated nature of the disease. Herein, we aimed to address the therapeutic potential of a novel class of pleiotropic mediators, specifically a new drug candidate, nitro-oleic acid (NO2-OA), on AAA, in a well-characterized murine AAA model. METHODS: We generated AAA using a mouse model combining AAV.PCSK9-D377Y induced hypercholesterolemia with angiotensin II given by chronic infusion. Vehicle control (PEG-400), oleic acid (OA), or NO2-OA were subcutaneously delivered to mice using an osmotic minipump. We characterized the effects of NO2-OA on pathophysiological responses and dissected the underlying molecular mechanisms through various in vitro and ex vivo strategies. RESULTS: Subcutaneous administration of NO2-OA significantly decreased the AAA incidence (8/28 mice) and supra-renal aorta diameters compared to mice infused with either PEG-400 (13/19, p = 0.0117) or OA (16/23, p = 0.0078). In parallel, the infusion of NO2-OA in the AAA model drastically decreased extracellular matrix degradation, inflammatory cytokine levels, and leucocyte/macrophage infiltration in the vasculature. Administration of NO2-OA reduced inflammation, cytokine secretion, and cell migration triggered by various biological stimuli in primary and macrophage cell lines partially through activation of the peroxisome proliferator-activated receptor-gamma (PPARγ). Moreover, the protective effect of NO2-OA relies on the inhibition of macrophage prostaglandin E2 (PGE2)-induced PGE2 receptor 4 (EP4) cAMP signaling, known to participate in the development of AAA. CONCLUSION: Administration of NO2-OA protects against AAA formation and multifactorial macrophage activation. With NO2-OA currently undergoing FDA approved phase II clinical trials, these findings may expedite the use of this nitro-fatty acid for AAA therapy.


Asunto(s)
Aneurisma de la Aorta Abdominal/fisiopatología , Activación de Macrófagos/efectos de los fármacos , Nitrocompuestos/farmacología , Ácidos Oléicos/farmacología , Angiotensina II/farmacología , Animales , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
8.
Circ Res ; 122(7): 945-957, 2018 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-29467198

RESUMEN

RATIONALE: Postischemic angiogenesis is critical to limit the ischemic tissue damage and improve the blood flow recovery. The regulation and the underlying molecular mechanisms of postischemic angiogenesis are not fully unraveled. TFEB (transcription factor EB) is emerging as a master gene for autophagy and lysosome biogenesis. However, the role of TFEB in vascular disease is less understood. OBJECTIVE: We aimed to determine the role of endothelial TFEB in postischemic angiogenesis and its underlying molecular mechanism. METHODS AND RESULTS: In primary human endothelial cells (ECs), serum starvation induced TFEB nuclear translocation. VEGF (vascular endothelial growth factor) increased TFEB expression level and nuclear translocation. Utilizing genetically engineered EC-specific TFEB transgenic and KO (knockout) mice, we investigated the role of TFEB in postischemic angiogenesis in the mouse hindlimb ischemia model. We observed improved blood perfusion and increased capillary density in the EC-specific TFEB transgenic mice compared with the wild-type littermates. Furthermore, blood flow recovery was attenuated in EC-TFEB KO mice compared with control mice. In aortic ring cultures, the TFEB transgene significantly increased vessel sprouting, whereas TFEB deficiency impaired the vessel sprouting. In vitro, adenovirus-mediated TFEB overexpression promoted EC tube formation, migration, and survival, whereas siRNA-mediated TFEB knockdown had the opposite effect. Mechanistically, TFEB activated AMPK (AMP-activated protein kinase)-α signaling and upregulated autophagy. Through inactivation of AMPKα or inhibition of autophagy, we demonstrated that the AMPKα and autophagy are necessary for TFEB to regulate angiogenesis in ECs. Finally, the positive effect of TFEB on AMPKα activation and EC tube formation was mediated by TFEB-dependent transcriptional upregulation of MCOLN1 (mucolipin-1). CONCLUSIONS: In summary, our data demonstrate that TFEB is a positive regulator of angiogenesis through activation of AMPKα and autophagy, suggesting that TFEB constitutes a novel molecular target for ischemic vascular disease.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Endotelio Vascular/metabolismo , Isquemia Miocárdica/metabolismo , Neovascularización Fisiológica , Quinasas de la Proteína-Quinasa Activada por el AMP , Transporte Activo de Núcleo Celular , Animales , Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Núcleo Celular/metabolismo , Células Cultivadas , Endotelio Vascular/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Miocitos Cardíacos/metabolismo , Proteínas Quinasas/metabolismo , Regeneración , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 39(3): 402-412, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30602303

RESUMEN

Objective- Mutations in Krüppel like factor-11 ( KLF11), a gene also known as maturity-onset diabetes mellitus of the young type 7, contribute to the development of diabetes mellitus. KLF11 has anti-inflammatory effects in endothelial cells and beneficial effects on stroke. However, the function of KLF11 in the cardiovascular system is not fully unraveled. In this study, we investigated the role of KLF11 in vascular smooth muscle cell biology and arterial thrombosis. Approach and Results- Using a ferric chloride-induced thrombosis model, we found that the occlusion time was significantly reduced in conventional Klf11 knockout mice, whereas bone marrow transplantation could not rescue this phenotype, suggesting that vascular KLF11 is critical for inhibition of arterial thrombosis. We further demonstrated that vascular smooth muscle cell-specific Klf11 knockout mice also exhibited significantly reduced occlusion time. The expression of tissue factor (encoded by the F3 gene), a main initiator of the coagulation cascade, was increased in the artery of Klf11 knockout mice, as determined by real-time quantitative polymerase chain reaction and immunofluorescence. Furthermore, vascular smooth muscle cells isolated from Klf11 knockout mouse aortas showed increased tissue factor expression, which was rescued by KLF11 overexpression. In human aortic smooth muscle cells, small interfering RNA-mediated knockdown of KLF11 increased tissue factor expression. Consistent results were observed on adenovirus-mediated overexpression of KLF11. Mechanistically, KLF11 downregulates F3 at the transcriptional level as determined by reporter and chromatin immunoprecipitation assays. Conclusions- Our data demonstrate that KLF11 is a novel transcriptional suppressor of F3 in vascular smooth muscle cells, constituting a potential molecular target for inhibition of arterial thrombosis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Represoras/fisiología , Tromboplastina/biosíntesis , Trombosis/prevención & control , Animales , Antitrombina III/análisis , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Proteínas Reguladoras de la Apoptosis/deficiencia , Proteínas Reguladoras de la Apoptosis/genética , Trasplante de Médula Ósea , Células Cultivadas , Cloruros/toxicidad , Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Femenino , Compuestos Férricos/toxicidad , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptido Hidrolasas/análisis , Agregación Plaquetaria , Interferencia de ARN , Proteínas Recombinantes/metabolismo , Proteínas Represoras/antagonistas & inhibidores , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Tromboplastina/genética , Trombosis/inducido químicamente , Transcripción Genética
10.
Physiol Genomics ; 51(6): 224-233, 2019 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-31074702

RESUMEN

Endothelial cell (EC) dysfunction is a crucial initiation event in the development of atherosclerosis and is associated with diabetes mellitus, hypertension, and heart failure. Both digestive and oxidative inflammatory conditions lead to the endogenous formation of nitrated derivatives of unsaturated fatty acids (FAs) upon generation of the proximal nitrating species nitrogen dioxide (·NO2) by nitric oxide (·NO) and nitrite-dependent reactions. Nitro-FAs (NO2-FAs) such as nitro-oleic acid (NO2-OA) and nitro-linoleic acid (NO2-LA) potently inhibit inflammation and oxidative stress, regulate cellular functions, and maintain cardiovascular homeostasis. Recently, conjugated linoleic acid (CLA) was identified as the preferential FA substrate of nitration in vivo. However, the functions of nitro-CLA (NO2-CLA) in ECs remain to be explored. In the present study, a distinct transcriptome regulated by NO2-CLA was revealed in primary human coronary artery endothelial cells (HCAECs) through RNA sequencing. Differential gene expression and pathway enrichment analysis identified numerous regulatory networks including those related to the modulation of inflammation, oxidative stress, cell cycle, and hypoxic responses by NO2-CLA, suggesting a diverse impact of NO2-CLA and other electrophilic nitrated FAs on cellular processes. These findings extend the understanding of the protective actions of NO2-CLA in cardiovascular diseases and provide new insight into the underlying mechanisms that mediate the pleiotropic cellular responses to NO2-CLA.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Ácidos Linoleicos Conjugados/farmacología , Adulto , Sistema Cardiovascular/efectos de los fármacos , Células Cultivadas , Redes Reguladoras de Genes/genética , Homeostasis/efectos de los fármacos , Homeostasis/genética , Humanos , Inflamación/genética , Masculino , Óxido Nítrico/genética , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
11.
Circulation ; 138(1): 67-79, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29371216

RESUMEN

BACKGROUND: The perivascular adipose tissue (PVAT) surrounding vessels constitutes a distinct functional integral layer of the vasculature required to preserve vascular tone under physiological conditions. However, there is little information on the relationship between PVAT and blood pressure regulation, including its potential contributions to circadian blood pressure variation. METHODS: Using unique brown adipocyte-specific aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1) and angiotensinogen knockout mice, we determined the vasoactivity of homogenized PVAT in aortic rings and how brown adipocyte peripheral expression of Bmal1 and angiotensinogen in PVAT regulates the amplitude of diurnal change in blood pressure in mice. RESULTS: We uncovered a peripheral clock in PVAT and demonstrated that loss of Bmal1 in PVAT reduces blood pressure in mice during the resting phase, leading to a superdipper phenotype. PVAT extracts from wild-type mice significantly induced contractility of isolated aortic rings in vitro in an endothelium-independent manner. This property was impaired in PVAT from brown adipocyte-selective Bmal1-deficient (BA-Bmal1-KO) mice. The PVAT contractile properties were mediated by local angiotensin II, operating through angiotensin II type 1 receptor-dependent signaling in the isolated vessels and linked to PVAT circadian regulation of angiotensinogen. Indeed, angiotensinogen mRNA and angiotensin II levels in PVAT of BA-Bmal1-KO mice were significantly reduced. Systemic infusion of angiotensin II, in turn, reduced Bmal1 expression in PVAT while eliminating the hypotensive phenotype during the resting phase in BA-Bmal1-KO mice. Angiotensinogen, highly expressed in PVAT, shows circadian expression in PVAT, and selective deletion of angiotensinogen in brown adipocytes recapitulates the phenotype of selective deletion of Bmal1 in brown adipocytes. Furthermore, angiotensinogen is a transcriptional target of Bmal1 in PVAT. CONCLUSIONS: These data indicate that local Bmal1 in PVAT regulates angiotensinogen expression and the ensuing increase in angiotensin II, which acts on smooth muscle cells in the vessel walls to regulate vasoactivity and blood pressure in a circadian fashion during the resting phase. These findings will contribute to a better understanding of the cardiovascular complications of circadian disorders, alterations in the circadian dipping phenotype, and cross-talk between systemic and peripheral regulation of blood pressure.


Asunto(s)
Factores de Transcripción ARNTL/metabolismo , Tejido Adiposo Pardo/metabolismo , Angiotensinógeno/metabolismo , Aorta Torácica/metabolismo , Presión Sanguínea , Ritmo Circadiano , Sistema Renina-Angiotensina , Transcripción Genética , Factores de Transcripción ARNTL/deficiencia , Factores de Transcripción ARNTL/genética , Angiotensinógeno/deficiencia , Angiotensinógeno/genética , Animales , Presión Sanguínea/genética , Ritmo Circadiano/genética , Genotipo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Sistema Renina-Angiotensina/genética , Descanso , Transducción de Señal , Factores de Tiempo , Vasoconstricción
12.
Arterioscler Thromb Vasc Biol ; 38(8): 1738-1747, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29954752

RESUMEN

Objective- Perivascular adipose tissue (PVAT) contributes to vascular homeostasis by producing paracrine factors. Previously, we reported that selective deletion of PPARγ (peroxisome proliferator-activated receptor γ) in vascular smooth muscle cells resulted in concurrent loss of PVAT and enhanced atherosclerosis in mice. To address the causal relationship between loss of PVAT and atherosclerosis, we used BA-PPARγ-KO (brown adipocyte-specific PPARγ knockout) mice. Approach and Results- Deletion of PPARγ in brown adipocytes did not affect PPARγ in white adipocytes or vascular smooth muscle cells or PPARα and PPARδ expression in brown adipocytes. However, development of PVAT and interscapular brown adipose tissue was remarkably impaired, associated with reduced expression of genes encoding lipogenic enzymes in the BA-PPARγ-KO mice. Thermogenesis in brown adipose tissue was significantly impaired with reduced expression of thermogenesis genes in brown adipose tissue and compensatory increase in subcutaneous and gonadal white adipose tissues. Remarkably, basal expression of inflammatory genes and macrophage infiltration in PVAT and brown adipose tissue were significantly increased in the BA-PPARγ-KO mice. BA-PPARγ-KO mice were crossbred with ApoE KO (apolipoprotein E knockout) mice to investigate the development of atherosclerosis. Flow cytometry analysis confirmed increased systemic and PVAT inflammation. Consequently, atherosclerotic lesions were significantly increased in mice with impaired PVAT development, thus indicating that the lack of normal PVAT is sufficient to drive increased atherosclerosis. Conclusions- PPARγ is required for functional PVAT development. PPARγ deficiency in PVAT, while still expressed in vascular smooth muscle cell, enhances atherosclerosis and results in vascular and systemic inflammation, providing new insights on the specific roles of PVAT in atherosclerosis and cardiovascular disease at large.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Enfermedades de la Aorta/metabolismo , Aterosclerosis/metabolismo , PPAR gamma/deficiencia , Adipocitos Marrones/patología , Tejido Adiposo Pardo/patología , Tejido Adiposo Pardo/fisiopatología , Tejido Adiposo Blanco/patología , Tejido Adiposo Blanco/fisiopatología , Adiposidad , Animales , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Enfermedades de la Aorta/fisiopatología , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/fisiopatología , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Lipogénesis/genética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , PPAR gamma/genética , Placa Aterosclerótica , Transducción de Señal , Termogénesis
13.
Arterioscler Thromb Vasc Biol ; 37(6): 1068-1075, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28428219

RESUMEN

OBJECTIVE: CETP (cholesteryl ester transfer protein) plays an important role in lipoprotein metabolism; however, whether inhibition of CETP activity can prevent cardiovascular disease remains controversial. APPROACH AND RESULTS: We generated CETP knockout (KO) rabbits by zinc finger nuclease gene editing and compared their susceptibility to cholesterol diet-induced atherosclerosis to that of wild-type (WT) rabbits. On a chow diet, KO rabbits showed higher plasma levels of high-density lipoprotein (HDL) cholesterol than WT controls, and HDL particles of KO rabbits were essentially rich in apolipoprotein AI and apolipoprotein E contents. When challenged with a cholesterol-rich diet for 18 weeks, KO rabbits not only had higher HDL cholesterol levels but also lower total cholesterol levels than WT rabbits. Analysis of plasma lipoproteins revealed that reduced plasma total cholesterol in KO rabbits was attributable to decreased apolipoprotein B-containing particles, while HDLs remained higher than that in WT rabbits. Both aortic and coronary atherosclerosis was significantly reduced in KO rabbits compared with WT rabbits. Apolipoprotein B-depleted plasma isolated from CETP KO rabbits showed significantly higher capacity for cholesterol efflux from macrophages than that from WT rabbits. Furthermore, HDLs isolated from CETP KO rabbits suppressed tumor necrosis factor-α-induced vascular cell adhesion molecule 1 and E-selectin expression in cultured endothelial cells. CONCLUSIONS: These results provide evidence that genetic ablation of CETP activity protects against cholesterol diet-induced atherosclerosis in rabbits.


Asunto(s)
Enfermedades de la Aorta/prevención & control , Aterosclerosis/prevención & control , Proteínas de Transferencia de Ésteres de Colesterol/deficiencia , Colesterol en la Dieta , Enfermedad de la Arteria Coronaria/prevención & control , Errores Innatos del Metabolismo Lipídico/metabolismo , Macrófagos/metabolismo , Animales , Animales Modificados Genéticamente , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/metabolismo , Enfermedades de la Aorta/patología , Apolipoproteína A-I/sangre , Apolipoproteínas B/sangre , Apolipoproteínas E/sangre , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Línea Celular , Proteínas de Transferencia de Ésteres de Colesterol/sangre , Proteínas de Transferencia de Ésteres de Colesterol/genética , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , HDL-Colesterol/sangre , Enfermedad de la Arteria Coronaria/genética , Enfermedad de la Arteria Coronaria/metabolismo , Enfermedad de la Arteria Coronaria/patología , Modelos Animales de Enfermedad , Selectina E/metabolismo , Femenino , Edición Génica , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Errores Innatos del Metabolismo Lipídico/sangre , Errores Innatos del Metabolismo Lipídico/genética , Masculino , Ratones , Conejos , Factores de Tiempo , Factor de Necrosis Tumoral alfa/farmacología , Molécula 1 de Adhesión Celular Vascular/metabolismo
14.
Pharm Biol ; 56(1): 567-572, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30460863

RESUMEN

CONTEXT: TangGanJian (TGJ) has a curative effect in the clinical treatment of nonalcoholic fatty liver disease (NAFLD) with type 2 diabetes mellitus (T2DM), while the mechanism involved in the treatment process remains unclear. OBJECTIVE: This study details the mechanism of TGJ on the treatment of NAFLD with T2DM. MATERIALS AND METHODS: NAFLD was induced in T2DM rat model. Male Wistar rats were assigned into six groups: Group I (control), Group II (model), Group III (pioglitazone, 0.5 mg/kg), Group IV (high dose of TGJ, 24.8 g/kg), Group V (middle dose of TGJ, 12.4 g/kg) and Group VI (low dose of TGJ, 6.2 g/kg). All rats in each group were treated with the corresponding drugs by gavage for 8 weeks. Haematoxylin and eosin analysis was conducted. The indicators of inflammatory and oxidative stress were analysed utilizing one-way ANOVA. RESULTS: The contents of TNF-α (15.794 ± 3.302 pg/mL), IL-6 (76.801 ± 8.491 pg/mL), IL-1ß (100.101 ± 13.150 pg/mL), CRP (1.052 ± 0.079 pg/mL) and MDA (3.972 ± 0.159 pg/mL) were obviously elevated in NAFLD with T2DM rats compared to controls. Except for the IL-6, the levels of other markers declined in a dose-dependent manner after treatment with TGJ. The SOD (14.139 ± 1.479 U/mgprot) and GSH-PX (81.511 ± 5.276 U/mgprot) levels significantly decreased in NAFLD with T2DM rats, while the levels of these indicators increased after treatment with TGJ. CONCLUSIONS: TGJ may be a therapy for the NAFLD with T2DM rats by modulating the inflammatory response and the oxidative stress capacity.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Medicamentos Herbarios Chinos/farmacología , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Animales , Glucemia/metabolismo , Proteína C-Reactiva/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/inducido químicamente , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Insulina/sangre , Interleucina-1beta , Interleucina-6/sangre , Hígado/efectos de los fármacos , Masculino , Enfermedad del Hígado Graso no Alcohólico/complicaciones , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Pioglitazona/farmacología , Ratas , Ratas Wistar , Factor de Necrosis Tumoral alfa/sangre
15.
Gastroenterology ; 150(5): 1208-1218, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26774178

RESUMEN

BACKGROUND & AIMS: The rs58542926 C>T variant of the transmembrane 6 superfamily member 2 gene (TM6SF2), encoding an E167K amino acid substitution, has been correlated with reduced total cholesterol (TC) and cardiovascular disease. However, little is known about the role of TM6SF2 in metabolism. We investigated the long-term effects of altered TM6SF2 levels in cholesterol metabolism. METHODS: C57BL/6 mice (controls), mice that expressed TM6SF2 specifically in the liver, and mice with CRISPR/Cas9-mediated knockout of Tm6sf2 were fed chow or high-fat diets. Blood samples were collected from all mice and plasma levels of TC, low-density lipoprotein cholesterol (LDL-c), high-density lipoprotein cholesterol, and triglycerides were measured. Liver tissues were collected and analyzed by histology, real-time polymerase chain reaction, and immunoblot assays. Adenovirus vectors were used to express transgenes in cultured Hep3B hepatocytes. RESULTS: Liver-specific expression of TM6SF2 increased plasma levels of TC and LDL-c, compared with controls, and altered liver expression of genes that regulate cholesterol metabolism. Tm6sf2-knockout mice had decreased plasma levels of TC and LDL-c, compared with controls, and consistent changes in expression of genes that regulate cholesterol metabolism. Expression of TM6SF2 promoted cholesterol biosynthesis in hepatocytes. CONCLUSIONS: TM6SF2 regulates cholesterol metabolism in mice and might be a therapeutic target for cardiovascular disease.


Asunto(s)
Colesterol/sangre , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Biomarcadores/sangre , Línea Celular , HDL-Colesterol/sangre , LDL-Colesterol/sangre , Dieta Alta en Grasa , Femenino , Regulación de la Expresión Génica , Genotipo , Humanos , Masculino , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Factores de Tiempo , Transfección , Triglicéridos/sangre
16.
Metab Brain Dis ; 30(2): 401-10, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24338065

RESUMEN

Transcription factors play an important role in the pathophysiology of many neurological disorders, including stroke. In the past three decades, an increasing number of transcription factors and their related gene signaling networks have been identified, and have become a research focus in the stroke field. Krüppel-like factors (KLFs) are members of the zinc finger family of transcription factors with diverse regulatory functions in cell growth, differentiation, proliferation, migration, apoptosis, metabolism, and inflammation. KLFs are also abundantly expressed in the brain where they serve as critical regulators of neuronal development and regeneration to maintain normal brain function. Dysregulation of KLFs has been linked to various neurological disorders. Recently, there is emerging evidence that suggests KLFs have an important role in the pathogenesis of stroke and provide endogenous vaso-or neuro-protection in the brain's response to ischemic stimuli. In this review, we summarize the basic knowledge and advancement of these transcriptional mediators in the central nervous system, highlighting the novel roles of KLFs in stroke.


Asunto(s)
Enfermedades del Sistema Nervioso Central/fisiopatología , Sistema Nervioso Central/fisiología , Factores de Transcripción de Tipo Kruppel/fisiología , Enfermedades del Sistema Nervioso/fisiopatología , Accidente Cerebrovascular/fisiopatología , Animales , Enfermedades del Sistema Nervioso Central/genética , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Regeneración Nerviosa , Enfermedades del Sistema Nervioso/genética , Accidente Cerebrovascular/genética
17.
J Biol Chem ; 288(7): 4625-36, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23264620

RESUMEN

Proliferation of vascular smooth muscle cells (VSMCs) in response to vascular injury plays a critical role in vascular lesion formation. Emerging data suggest that peroxisome proliferator-activated receptor γ coactivator 1 (PGC-1) is a key regulator of energy metabolism and other biological processes. However, the physiological role of PGC-1ß in VSMCs remains unknown. A decrease in PGC-1ß expression was observed in balloon-injured rat carotid arteries. PGC-1ß overexpression substantially inhibited neointima formation in vivo and markedly inhibited VSMC proliferation and induced cell cycle arrest at the G(1)/S transition phase in vitro. Accordingly, overexpression of PGC-1ß decreased the expression of minichromosome maintenance 4 (MCM4), which leads to a decreased loading of the MCM complex onto chromatin at the replication origins and decreased cyclin D1 levels, whereas PGC-1ß loss of function by adenovirus containing PGC-1ß shRNA resulted in the opposite effect. The transcription factor AP-1 was involved in the down-regulation of MCM4 expression. Furthermore, PGC-1ß is up-regulated by metformin, and metformin-associated anti-proliferative activity in VSMCs is at least partially dependent on PGC-1ß. Our data show that PGC-1ß is a critical component in regulating DNA replication, VSMC proliferation, and vascular lesion formation, suggesting that PGC-1ß may emerge as a novel therapeutic target for control of proliferative vascular diseases.


Asunto(s)
Proteínas Portadoras/metabolismo , Cromatina/metabolismo , Regulación de la Expresión Génica , Proteína 1 de Mantenimiento de Minicromosoma/metabolismo , Miocitos del Músculo Liso/citología , Proteínas de Unión al ARN/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Arterias Carótidas/metabolismo , Núcleo Celular/metabolismo , Proliferación Celular , Diabetes Mellitus/metabolismo , Técnicas de Transferencia de Gen , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Ratas
18.
Brain ; 136(Pt 4): 1274-87, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23408111

RESUMEN

Peroxisome proliferator-activated receptor gamma (PPARγ) is emerging as a major regulator in neurological diseases. However, the role of (PPARγ) and its co-regulators in cerebrovascular endothelial dysfunction after stroke is unclear. Here, we have demonstrated that (PPARγ) activation by pioglitazone significantly inhibited both oxygen-glucose deprivation-induced cerebral vascular endothelial cell death and middle cerebral artery occlusion-triggered cerebrovascular damage. Consistent with this finding, selective (PPARγ) genetic deletion in vascular endothelial cells resulted in increased cerebrovascular permeability and brain infarction in mice after focal ischaemia. Moreover, we screened for (PPARγ) co-regulators using a genome-wide and high-throughput co-activation system and revealed KLF11 as a novel (PPARγ) co-regulator, which interacted with (PPARγ) and regulated its function in mouse cerebral vascular endothelial cell cultures. Interestingly, KLF11 was also found as a direct transcriptional target of (PPARγ). Furthermore, KLF11 genetic deficiency effectively abolished pioglitazone cytoprotection in mouse cerebral vascular endothelial cell cultures after oxygen-glucose deprivation, as well as pioglitazone-mediated cerebrovascular protection in a mouse middle cerebral artery occlusion model. Mechanistically, we demonstrated that KLF11 enhanced (PPARγ) transcriptional suppression of the pro-apoptotic microRNA-15a (miR-15a) gene, resulting in endothelial protection in cerebral vascular endothelial cell cultures and cerebral microvasculature after ischaemic stimuli. Taken together, our data demonstrate that recruitment of KLF11 as a novel (PPARγ) co-regulator plays a critical role in the cerebrovascular protection after ischaemic insults. It is anticipated that elucidating the coordinated actions of KLF11 and (PPARγ) will provide new insights into understanding the molecular mechanisms underlying (PPARγ) function in the cerebral vasculature and help to develop a novel therapeutic strategy for the treatment of stroke.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Células Endoteliales/metabolismo , PPAR gamma/metabolismo , Proteínas Represoras/fisiología , Accidente Cerebrovascular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Infarto Encefálico/metabolismo , Proteínas de Ciclo Celular/deficiencia , Proteínas de Ciclo Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos , MicroARNs/genética , PPAR gamma/deficiencia , PPAR gamma/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/fisiopatología
19.
Biochem J ; 451(1): 55-60, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23360436

RESUMEN

Endothelial activation characterized by the expression of multiple chemokines and adhesive molecules is a critical initial step of vascular inflammation, which results in recruitment of leucocytes into the sub-endothelial layer of the vascular wall and triggers vascular inflammatory diseases such as atherosclerosis. Although inhibiting endothelial inflammation has already been well recognized as a therapeutic strategy in vascular inflammatory diseases, the therapeutic targets are still elusive. In the present study we found that Zc3h12c (zinc finger CCCH-type-containing 12C), a recently discovered CCCH zinc finger-containing protein, significantly inhibited the endothelial cell inflammatory response in vitro. Overexpression of Zc3h12c significantly attenuated TNFα (tumour necrosis factor α)-induced expression of chemokines and adhesive molecules, and thus reduced monocyte adherence to HUVECs (human umbilical vein endothelial cells). Conversely, siRNA (small interfering RNA)-mediated knockdown of Zc3h12c increased the TNFα-induced expression of chemokines and adhesive molecules in HUVECs. Furthermore, forced expression of Zc3h12c decreased TNFα-induced IKKα/ß [IκB (inhibitor of nuclear factor κB) kinase α/ß], IκBα phosphorylation and p65 nuclear translocation, suggesting that Zc3h12c exerted its anti-inflammatory function probably by suppressing the NF-κB (nuclear factor κB) pathway. Thus Zc3h12c is an endogenous inhibitor of TNFα-induced inflammatory signalling in HUVECs and might be a therapeutic target in vascular inflammatory diseases.


Asunto(s)
Núcleo Celular/metabolismo , Regulación de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Vasculitis/metabolismo , Transporte Activo de Núcleo Celular/genética , Núcleo Celular/genética , Núcleo Celular/patología , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Factor de Transcripción ReIA/genética , Factores de Transcripción/genética , Factor de Necrosis Tumoral alfa/genética , Vasculitis/genética , Vasculitis/patología
20.
Artículo en Inglés | MEDLINE | ID: mdl-37622706

RESUMEN

OBJECTIVE: The lung is one of the target organs of diabetes. This study aimed to probe the protective mechanism of Jiangtang Tongmai Prescription (JTTMP) against diabetic lung injury. METHODS: JTTMP-containing serum was collected, and a high glucose and high-fat diabetic cell model was established. The cells were treated with a drug-containing serum or a CAV1-associated vector. Transfection efficiency was measured by qRT-PCR and western blot, the cell proliferative capacity was tested by CCK-8 assay, and the expression of autophagosome marker LC3B was measured by immunophluorescence assay. Expression levels of the autophagy markers LC3B, p62, and Beclin-1, and the expression levels of the fibrosis markers α-SMA, FN-1, and TGF-ß1 were determined by western blot, and the levels of inflammatory factors TNF-α and IL-1ß in the supernatants were assessed by ELISA. RESULTS: In high glucose and high fat-induced MRC-5 cells, JTTMP-containing serum impeded the abnormal cell proliferation and the expression levels of autophagy markers, fibrosis markers, as well as inflammatory factors. CAV1 expression was decreased in MRC-5 cells treated with JTTMP-containing serum. In MRC-5 cells upon transfection with the CAV1 overexpression vector and treatment with JTTMP-containing serum, increased cell proliferation, increased LC3B, p62, Beclin-1, α-SMA, FN-1, and TGF-ß1, TNF-α, and IL-1ß levels were found compared with cells treated with JTTMP-containing serum alone. CONCLUSION: This study suggests that JTTMP suppresses CAV1 expression to attenuate diabetic lung injury by reducing abnormal proliferation and autophagy, and reducing levels of fibrosis and inflammation.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Lesión Pulmonar , Humanos , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Lesión Pulmonar/metabolismo , Beclina-1/metabolismo , Fibrosis , Pulmón/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Hiperglucemia/metabolismo , Glucosa/metabolismo , Fibroblastos/metabolismo , Autofagia , Diabetes Mellitus/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA