Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Nitric Oxide ; 142: 47-57, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38049061

RESUMO

BACKGROUND: Endothelial-mesenchymal transition (EndMT) induced by low shear stress plays an important role in the development of atherosclerosis. However, little is known about the correlation between hydrogen sulfide (H2S), a protective gaseous mediator in atherosclerosis and the process of EndMT. METHODS: We constructed a stable low-shear-stress-induced(2 dyn/cm2) EndMT model, acombined with the pretreatment method of hydrogen sulfide slow release agent(GYY4137). The level of MEST was detected in the common carotid artery of ApoE-/- mice with local carotid artery ligation. The effect of MEST on atherosclerosis development in vivo was verified using ApoE-/- mice were given tail-vein injection of endothelial-specific overexpressed and knock-down MEST adeno-associated virus (AAV). RESULTS: These findings confirmed that MEST is up-regulated in low-shear-stress-induced EndMT and atherosclerosis. In vivo experiments showed that MEST gene overexpression significantly promoted EndMT and aggravated the development of atherosclerotic plaques and MEST gene knockdown significantly inhibited EndMT and delayed the process of atherosclerosis. In vitro, H2S inhibits the expression of MEST and EndMT induced by low shear stress and inhibits EndMT induced by MEST overexpression. Knockdown of NFIL3 inhibit the up regulation of MEST and EndMT induced by low shear stress in HUVECs. CHIP-qPCR assay and Luciferase Reporter assay confirmed that NFIL3 binds to MEST DNA, increases its transcription and H2S inhibits the binding of NFIL3 and MEST DNA, weakening NFIL3's transcriptional promotion of MEST. Mechanistically, H2S increased the sulfhydrylation level of NFIL3, an important upstream transcription factors of MEST. In part, transcription factor NFIL3 restrain its binding to MEST DNA by sulfhydration. CONCLUSIONS: H2S negatively regulate the expression of MEST by sulfhydrylation of NFIL3, thereby inhibiting low-shear-stress-induced EndMT and atherosclerosis.


Assuntos
Aterosclerose , Sulfeto de Hidrogênio , Camundongos , Animais , Humanos , Sulfeto de Hidrogênio/farmacologia , Sulfeto de Hidrogênio/metabolismo , Transição Endotélio-Mesênquima , Aterosclerose/genética , Aterosclerose/metabolismo , Endotélio/metabolismo , DNA/metabolismo , Apolipoproteínas E/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Transição Epitelial-Mesenquimal
2.
Pestic Biochem Physiol ; 201: 105859, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38685213

RESUMO

The efficient use of pesticides has long been a topic of public concern, necessitating a thorough understanding of their movement in plants. This study investigates the translocation and distribution of penthiopyrad in pakchoi plants cultivated both in hydroponic and soil-cultivated conditions. Results indicate that penthiopyrad predominantly accumulates in the roots, with concentrations of 11.3-53.9 mg/kg following root application, and in the leaves, with concentrations of 2.0-17.1 mg/kg following foliar application. The bioconcentration factor exceeded 1, with values ranging from 1.2 to 23.9 for root application and 6.4 to 164.0 for foliar application, indicating a significant role in the absorption and accumulation processes. The translocation factor data, which were <1, suggest limited the translocations within pakchoi plants. The limitation may be attributed to the hydrophobic properties of penthiopyrad (log Kow = 3.86), as evidenced by its predominant distribution in the subcellular solid fractions of pakchoi tissues, accounting for 93.1% to 99.5% of the total proportion. Six metabolites (753-A-OH, M12, 754-T-DO, M11, PCA, and PAM) were identified in this study as being formed during this process. These findings provide valuable insights into the absorption, translocation, and metabolism of penthiopyrad in pakchoi.


Assuntos
Hidroponia , Raízes de Plantas , Solo , Solo/química , Raízes de Plantas/metabolismo , Folhas de Planta/metabolismo , Poluentes do Solo/metabolismo , Transporte Biológico
3.
Nitric Oxide ; 127: 18-25, 2022 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-35839994

RESUMO

Hydrogen sulfide (H2S) is the third gaseous signaling molecule discovered in the body after NO and CO and plays an important organismal protective role in various diseases. Within adipose tissue, related catalytic enzymes (cystathionine-ß-synthetase, cystathionine-γ-lyase, and 3-mercaptopyruvate transsulfuration enzyme) can produce and release endogenous H2S. Atherosclerosis (As) is a pathological change in arterial vessels that is closely related to abnormal glucose and lipid metabolism and a chronic inflammatory response. Previous studies have shown that H2S can act on the cardiovascular system, exerting effects such as improving disorders of glycolipid metabolism, alleviating insulin resistance, protecting the function of vascular endothelial cells, inhibiting vascular smooth muscle cell proliferation and migration, regulating vascular tone, inhibiting the inflammatory response, and antagonizing the occurrence and development of As.


Assuntos
Aterosclerose , Sulfeto de Hidrogênio , Tecido Adiposo/metabolismo , Aterosclerose/patologia , Cistationina gama-Liase/metabolismo , Células Endoteliais/metabolismo , Humanos , Sulfeto de Hidrogênio/metabolismo , Sulfeto de Hidrogênio/farmacologia
4.
Mol Cell Biochem ; 477(1): 255-265, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34687394

RESUMO

Diabetic cardiomyopathy (DCM) is a cardiovascular complication that tends to occur in patients with diabetes, obesity, or insulin resistance, with a higher late mortality rate. Sustained hyperglycemia, increased free fatty acids, or insulin resistance induces metabolic disorders in cardiac tissues and cells, leading to myocardial fibrosis, left ventricular hypertrophy, diastolic and/or systolic dysfunction, and finally develop into congestive heart failure. The close connection between all signaling pathways and the complex pathogenesis of DCM cause difficulties in finding effective targets for the treatment of DCM. It reported that hydrogen sulfide (H2S) could regulate cell energy substrate metabolism, reduce insulin resistance, protect cardiomyocytes, and improve myocardial function by acting on related key proteins such as differentiation cluster 36 (CD36) and glucose transporter 4 (GLUT4). In this article, the relative mechanisms of H2S in alleviating metabolic disorders of DCM were reviewed, and how H2S can better prevent and treat DCM in clinical practice will be discussed.


Assuntos
Cardiomiopatias Diabéticas/metabolismo , Metabolismo Energético , Sulfeto de Hidrogênio/metabolismo , Resistência à Insulina , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Humanos
5.
Circulation ; 142(5): 483-498, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32354235

RESUMO

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.


Assuntos
2-Hidroxipropil-beta-Ciclodextrina/uso terapêutico , Aneurisma da Aorta Abdominal/prevenção & controle , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/fisiologia , Modelos Animais de Doenças , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , 2-Hidroxipropil-beta-Ciclodextrina/farmacologia , Aminopropionitrilo/toxicidade , Aneurisma Roto/etiologia , Angiotensina II/toxicidade , Animais , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Apoptose/efeitos dos fármacos , Autofagia , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/biossíntese , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/deficiência , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/genética , Colesterol/metabolismo , Regulação para Baixo , Avaliação Pré-Clínica de Medicamentos , Mutação com Ganho de Função , Regulação da Expressão Gênica , Vetores Genéticos/toxicidade , Humanos , Mutação com Perda de Função , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Transcriptoma/efeitos dos fármacos
6.
Arterioscler Thromb Vasc Biol ; 40(10): 2494-2507, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787523

RESUMO

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.


Assuntos
Aneurisma da Aorta Abdominal/prevenção & controle , Aortite/prevenção & controle , Proteínas Cromossômicas não Histona/deficiência , Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Remodelação Vascular , Animais , Aorta Abdominal/metabolismo , Aorta Abdominal/patologia , Aneurisma da Aorta Abdominal/genética , Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/patologia , Aortite/genética , Aortite/metabolismo , Aortite/patologia , Estudos de Casos e Controles , Catepsinas/metabolismo , Células Cultivadas , Proteínas Cromossômicas não Histona/genética , Modelos Animais de Doenças , Matriz Extracelular/patologia , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Transdução de Sinais
7.
Cardiovasc Drugs Ther ; 35(5): 939-951, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-32671602

RESUMO

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.


Assuntos
Aneurisma da Aorta Abdominal/fisiopatologia , Ativação de Macrófagos/efeitos dos fármacos , Nitrocompostos/farmacologia , Ácidos Oleicos/farmacologia , Angiotensina II/farmacologia , Animais , Movimento Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos
8.
Circulation ; 138(1): 67-79, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29371216

RESUMO

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.


Assuntos
Fatores de Transcrição ARNTL/metabolismo , Tecido Adiposo Marrom/metabolismo , Angiotensinogênio/metabolismo , Aorta Torácica/metabolismo , Pressão Sanguínea , Ritmo Circadiano , Sistema Renina-Angiotensina , Transcrição Gênica , Fatores de Transcrição ARNTL/deficiência , Fatores de Transcrição ARNTL/genética , Angiotensinogênio/deficiência , Angiotensinogênio/genética , Animais , Pressão Sanguínea/genética , Ritmo Circadiano/genética , Genótipo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Sistema Renina-Angiotensina/genética , Descanso , Transdução de Sinais , Fatores de Tempo , Vasoconstrição
9.
Arterioscler Thromb Vasc Biol ; 38(8): 1738-1747, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29954752

RESUMO

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.


Assuntos
Adipócitos Marrons/metabolismo , Adipogenia , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/metabolismo , Doenças da Aorta/metabolismo , Aterosclerose/metabolismo , PPAR gama/deficiência , Adipócitos Marrons/patologia , Tecido Adiposo Marrom/patologia , Tecido Adiposo Marrom/fisiopatologia , Tecido Adiposo Branco/patologia , Tecido Adiposo Branco/fisiopatologia , Adiposidade , Animais , Doenças da Aorta/genética , Doenças da Aorta/patologia , Doenças da Aorta/fisiopatologia , Aterosclerose/genética , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Mediadores da Inflamação/metabolismo , Lipogênese/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout para ApoE , PPAR gama/genética , Placa Aterosclerótica , Transdução de Sinais , Termogênese
10.
Cardiovasc Diabetol ; 17(1): 134, 2018 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-30305178

RESUMO

Perivascular adipose tissue (PVAT), the adipose tissue that surrounds most of the vasculature, has emerged as an active component of the blood vessel wall regulating vascular homeostasis and affecting the pathogenesis of atherosclerosis. Although PVAT characteristics resemble both brown and white adipose tissues, recent evidence suggests that PVAT develops from its own distinct precursors implying a closer link between PVAT and vascular system. Under physiological conditions, PVAT has potent anti-atherogenic properties mediated by its ability to secrete various biologically active factors that induce non-shivering thermogenesis and metabolize fatty acids. In contrast, under pathological conditions (mainly obesity), PVAT becomes dysfunctional, loses its thermogenic capacity and secretes pro-inflammatory adipokines that induce endothelial dysfunction and infiltration of inflammatory cells, promoting atherosclerosis development. Since PVAT plays crucial roles in regulating key steps of atherosclerosis development, it may constitute a novel therapeutic target for the prevention and treatment of atherosclerosis. Here, we review the current literature regarding the roles of PVAT in the pathogenesis of atherosclerosis.


Assuntos
Adipocinas/metabolismo , Tecido Adiposo/metabolismo , Aterosclerose/metabolismo , Vasos Sanguíneos/metabolismo , Mediadores da Inflamação/metabolismo , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/patologia , Tecido Adiposo/fisiopatologia , Adiposidade , Animais , Anti-Inflamatórios/uso terapêutico , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Aterosclerose/prevenção & controle , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Vasos Sanguíneos/fisiopatologia , Fármacos Cardiovasculares/uso terapêutico , Metabolismo Energético , Humanos , Fatores de Proteção , Fatores de Risco , Transdução de Sinais , Termogênese
11.
Int J Cancer ; 136(8): 1769-80, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25257795

RESUMO

Aberrant PDGF-PDGFR signaling and its effects on downstream effectors have been implicated in glioma development. A crucial AKT regulator, ACK1 (TNK2) has been shown to be a downstream mediator of PDGF signaling; however, the exact underlying mechanisms in gliomas remain elusive. Here, we report that in glioma cells, PDGFR-ß activation enhanced the interaction between ACK1 and AKT, resulting in AKT activation. PDGF treatment consistently promoted the formation of complexes containing PDGFR-ß and ACK1. Mutational analysis suggested that Y635 of ACK1 is a PDGFR-ß phosphorylation site and that the ACK1 Y635F mutant abrogated the sequential activation of AKT. Moreover, PDK1 interacted with ACK1 during PDGF stimulation, which is required for the binding of ACK1 to PDGFR-ß. Further mutational analysis showed that T325 of ACK1 was crucial for the ACK1 and PDK1 interaction. ACK1 Y635F or T325A mutants abolished PDGFR-ß-induced AKT activation, the subsequent nuclear translocation of ß-catenin and the expression of cyclin D1. Glioma cell cycle progression, proliferation and tumorigenesis were accordingly blocked by ACK1 Y635F or T325A. In glioblastoma multiforme samples from 51 patients, increased ACK1 tyrosine phosphorylation correlated with upregulated PDGFR-ß activity and AKT activation. Taken together, our data demonstrate that ACK1 plays a pivotal role in PDGF-PDGFR-induced AKT signaling in glioma tumorigenesis. This knowledge contributes to our understanding of glioma progression and may facilitate the identification of novel therapeutic targets for future glioma treatment.


Assuntos
Carcinogênese/genética , Glioblastoma/genética , Glioblastoma/patologia , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Transdução de Sinais/genética , Carcinogênese/patologia , Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Ciclina D1/genética , Ciclina D1/metabolismo , Glioblastoma/metabolismo , Células HEK293 , Humanos , Fosforilação/genética , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Regulação para Cima/genética , beta Catenina/genética , beta Catenina/metabolismo
12.
Front Plant Sci ; 15: 1450786, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39301163

RESUMO

Submerged plants are an important part of aquatic ecosystems, and the restoration of submerged plants is a key step in the reconstruction of aquatic ecosystems. However, little is known about the role of modified sediments in helping submerged plants recover under low light. In this study, we set up four sediment types and two light intensities to explore the effects of modified sediments on the growth of Vallisneria natans under two low light conditions. The results showed that the independent and interactive effects of light intensity and sediment type significantly affected the biomass, morphology, photosynthetic pigment content and antioxidant enzyme activity of V. natans. At 5% and 20% natural light intensity, the sediment modified with 40% peat soil had a larger root biomass and the highest leaf and root C/N ratio, the sediment modified with 40% vermiculite had a longer root length and more ramets. At 5% natural light intensity, the sediments modified with fly ash had shorter root length and smaller leaf biomass. The sediments modified with fly ash had the greatest chlorophyll content at 20% natural light intensity. It can be concluded that the addition of 40% peat soil or 40% vermiculite in sediment is conducive to the growth of V. natans under low light conditions. Our study indicates the positive effects of the modified sediment on the growth of V. natans under low light conditions, and our study will provide a reference for the restoration of submerged plants in aquatic ecosystems.

13.
Atherosclerosis ; 390: 117430, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38301602

RESUMO

BACKGROUND AND AIMS: Tripartite motif (TRIM65) is an important member of the TRIM protein family, which is a newly discovered E3 ligase that interacts with and ubiquitinates various substrates and is involved in diverse pathological processes. However, the function of TRIM65 in atherosclerosis remains unarticulated. In this study, we investigated the role of TRIM65 in the pathogenesis of atherosclerosis, specifically in vascular smooth muscle cells (VSMCs) phenotype transformation, which plays a crucial role in formation of atherosclerotic lesions. METHODS AND RESULTS: Both non-atherosclerotic and atherosclerotic lesions during autopsy were collected singly or pairwise from each individual (n = 16) to investigate the relationship between TRIM65 and the development of atherosclerosis. In vivo, Western diet-fed ApoE-/- mice overexpressing or lacking TRIM65 were used to assess the physiological function of TRIM65 on VSMCs phenotype, proliferation and atherosclerotic lesion formation. In vitro, VSMCs phenotypic transformation was induced by platelet-derived growth factor-BB (PDGF-BB). TRIM65-overexpressing or TRIM65-abrogated primary mouse aortic smooth muscle cells (MOASMCs) and human aortic smooth muscle cells (HASMCs) were used to investigate the mechanisms underlying the progression of VSMCs phenotypic transformation, proliferation and migration. Increased TRIM65 expression was detected in α-SMA-positive cells in the medial and atherosclerotic lesions of autopsy specimens. TRIM65 overexpression increased, whereas genetic knockdown of TRIM65 remarkably inhibited, atherosclerotic plaque development. Mechanistically, TRIM65 overexpression activated PI3K/Akt/mTOR signaling, resulting in the loss of the VSMCs contractile phenotype, including calponin, α-SMA, and SM22α, as well as cell proliferation and migration. However, opposite phenomena were observed when TRIM65 was deficient in vivo or in vitro. Moreover, in cultured PDGF-BB-induced TRIM65-overexpressing VSMCs, inhibition of PI3K by treatment with the inhibitor LY-294002 for 24 h markedly attenuated PI3K/Akt/mTOR activation, regained the VSMCs contractile phenotype, and blocked the progression of cell proliferation and migration. CONCLUSIONS: TRIM65 overexpression enhances atherosclerosis development by promoting phenotypic transformation of VSMCs from contractile to synthetic state through activation of the PI3K/Akt/mTOR signal pathway.


Assuntos
Aterosclerose , Proteínas Proto-Oncogênicas c-akt , Humanos , Camundongos , Animais , Becaplermina/genética , Becaplermina/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Músculo Liso Vascular/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Movimento Celular , Transdução de Sinais , Proliferação de Células , Serina-Treonina Quinases TOR/metabolismo , Aterosclerose/patologia , Miócitos de Músculo Liso/patologia , Fenótipo , Células Cultivadas , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética
14.
Mol Nutr Food Res ; 68(5): e2300784, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38314939

RESUMO

SCOPE: Premature ovarian insufficiency (POI) is a common female infertility problem, with its pathogenesis remains unknown. The NOD-like receptor family pyrin domain-containing 3 (NLRP3)-mediated pyroptosis has been proposed as a possible mechanism in POI. This study investigates the therapeutic effect of α-ketoglutarate (AKG) on ovarian reserve function in POI rats and further explores the potential molecular mechanisms. METHODS AND RESULTS: POI rats are caused by administration of cyclophosphamide (CTX) to determine whether AKG has a protective effect. AKG treatment increases the ovarian index, maintains both serum hormone levels and follicle number, and improves the ovarian reserve function in POI rats, as evidence by increased the level of lactate and the expression of rate-limiting enzymes of glycolysis in the ovaries, additionally reduced the expression of NLRP3, Gasdermin D (GSDMD), Caspase-1, Interleukin-18 (IL-18), and Interleukin-1 beta (IL-1ß). In vitro, KGN cells are treated with LPS and nigericin to mimic pyroptosis, then treated with AKG and MCC950. AKG inhibits inflammatory and pyroptosis factors such as NLRP3, restores the glycolysis process in vitro, meanwhile inhibition of NLRP3 has the same effect. CONCLUSION: AKG ameliorates CTX-induced POI by inhibiting NLRP3-mediated pyroptosis, which provides a new therapeutic strategy and drug target for clinical POI patients.


Assuntos
Reserva Ovariana , Insuficiência Ovariana Primária , Humanos , Ratos , Feminino , Animais , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ácidos Cetoglutáricos/farmacologia , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/tratamento farmacológico , Piroptose , Células da Granulosa/metabolismo , Inflamassomos/metabolismo
15.
Curr Med Chem ; 30(40): 4554-4568, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36476437

RESUMO

An abdominal aortic aneurysm (AAA) is a progressive dilatation of the vascular wall occurring below the aortic fissure, preferably occurring below the renal artery. The molecular mechanism of AAA has not yet been elucidated. In the past few decades, research on abdominal aortic aneurysm has been mainly focused on the vessel wall, and it is generally accepted that inflammation and middle layer fracture of the vessel wall is the core steps in the development of AAA. However, perivascular adipose tissue plays a non-negligible role in the occurrence and development of AAA. The position of PVAT plays a supporting and protective role on the vascular wall, but the particularity of the location makes it not only have the physiological function of visceral fat; but also can regulate the vascular function by secreting a large number of adipokines and cytokines. An abdominal aortic aneurysm is getting higher and higher, with a vascular rupture, low rescue success rate, and extremely high lethality rate. At present, there is no drug to control the progression or reverse abdominal aortic aneurysm. Therefore, it is critical to deeply explore the mechanism of abdominal aortic aneurysms and find new therapeutic ways to inhibit abdominal aortic aneurysm formation and disease progression. An abdominal aortic aneurysm is mainly characterized by inflammation of the vessel wall and matrix metalloprotein degradation. In this review, we mainly focus on the cytokines released by the perivascular adipose tissue, summarize the mechanisms involved in the regulation of abdominal aortic aneurysms, and provide new research directions for studying abdominal aortic aneurysms.


Assuntos
Aneurisma da Aorta Abdominal , Motivação , Humanos , Aneurisma da Aorta Abdominal/metabolismo , Tecido Adiposo/metabolismo , Inflamação/metabolismo , Citocinas/metabolismo
16.
Lab Invest ; 92(11): 1623-34, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22964852

RESUMO

The blood-brain barrier (BBB) opening following traumatic brain injury (TBI) provides a chance for therapeutic agents to cross the barrier, yet the reduction of the cerebral microvascular perfusion after TBI may limit the intervention. Meanwhile, optimizing the cerebral capillary perfusion by the strategies such as fluid administration may cause brain edema due to the BBB opening post trauma. To guide the TBI therapy, we characterized the relationship between the changes in the cerebral capillary perfusion and BBB permeability after TBI. First, we observed the changes of the cerebral capillary perfusion by the intracardiac perfusion of Evans Blue and the BBB disruption with magnetic resonance imaging (MRI) in the rat subjected to lateral fluid percussion (FP) brain injury. The correlation between two variables was next evaluated with the correlation analysis. Since related to BBB breakdown, matrix metalloproteinase-9 (MMP-9) activity was finally detected by gelatin zymography. We found that the ratios of the perfused microvessel numbers in the lesioned cortices were significantly reduced at 0 and 1 h post trauma compared with that in the normal cortex, which then dramatically recovered at 4 and 24 h after injury, and that the BBB permeability was greatly augmented in the ipsilateral parts at 4, 12, and 24 h, and in the contralateral area at 24 h after injury compared with that in the uninjured brain. The correlation analysis showed that the BBB permeability increase was related to the restoration of the cerebral capillary perfusion over a 24-h period post trauma. Moreover, the gelatin zymography analysis indicated that the MMP-9 activity in the injured brain increased at 4 h and significantly elevated at 12 and 24 h as compared to that at 0 or 1 h after TBI. Our findings demonstrate that the 4 h post trauma is a critical turning point during the development of TBI, and, importantly, the correlation analysis may guide us how to treat TBI.


Assuntos
Barreira Hematoencefálica , Lesões Encefálicas/metabolismo , Lesões Encefálicas/fisiopatologia , Circulação Cerebrovascular , Microcirculação , Animais , Lesões Encefálicas/enzimologia , Permeabilidade Capilar , Azul Evans , Imageamento por Ressonância Magnética , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Ratos , Ratos Sprague-Dawley
17.
Cancer Sci ; 103(2): 181-90, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22093097

RESUMO

The Notch signaling pathway has been implicated in both developmental processes and tumorigenesis. Aberrant Notch signaling has been repeatedly demonstrated to facilitate the proliferation and survival of glioma cells by regulating downstream effectors or other signaling pathways. In glioblastoma multiforme specimens from 59 patients, Notch1 was highly expressed in tumor tissues compared with normal brain tissues, and this expression was correlated with elevated AKT phosphorylation and Snail expression. Increased nuclear localization of ß-catenin and p50 as well as enhanced IKKα/AKT interaction were also observed in glioma tissues. In U87MG cells, the activation of Notch1 by DLL4 stimulation or by the overexpression of Notch intracellular domain (NICD) resulted in AKT activation and thereby promoted ß-catenin activity and NF-κB signaling. Inhibition of EGFR partially blocked the ß-catenin and NF-κB signaling stimulated by Notch1 activation. Furthermore, NICD overexpression in U87MG cells led to the upregulated expression of several metastasis-associated molecules, which could be abrogated by the knockdown of either ß-catenin or p50. In U87MG and U251 cells, DLL4-induced cellular migration and invasion could be inhibited by either ß-catenin or a p50 inhibitor. Collectively, these results indicate that Notch activation could stimulate ß-catenin and NF-κB signaling through AKT activation in glioma cells. Thus, Notch activation-stimulated ß-catenin and NF-κB signaling synergistically promote the migratory and invasive properties of glioma cells.


Assuntos
Glioblastoma/metabolismo , Glioblastoma/patologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Notch1/metabolismo , Transdução de Sinais , beta Catenina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Encéfalo/patologia , Proteínas de Ligação ao Cálcio , Linhagem Celular Tumoral , Movimento Celular , Complexo Dinactina , Receptores ErbB/antagonistas & inibidores , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Humanos , Quinase I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Proteínas Associadas aos Microtúbulos/antagonistas & inibidores , Proteínas Associadas aos Microtúbulos/biossíntese , NF-kappa B/genética , Invasividade Neoplásica , Proteínas Proto-Oncogênicas c-akt/genética , Interferência de RNA , RNA Interferente Pequeno , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , beta Catenina/biossíntese , beta Catenina/genética
18.
Curr Pharm Des ; 28(16): 1321-1328, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35974674

RESUMO

Hippo, an evolutionarily conserved kinase cascade reaction in organisms, can respond to a set of signals, such as mechanical signals and cell metabolism, to maintain cell growth, differentiation, tissue/organ development, and homeostasis. In the past ten years, Hippo has controlled the development of tissues and organs by regulating the process of cell proliferation, especially in the field of cardiac regeneration after myocardial infarction. This suggests that Hippo signaling is closely linked to cardiovascular disease. Atherosclerosis is the most common disease of the cardiovascular system. It is characterised by chronic inflammation of the vascular wall, mainly involving dysfunction of endothelial cells, smooth muscle cells, and macrophages. Oxidized Low density lipoprotein (LDL) damages the barrier function of endothelial cells, which enter the middle membrane of the vascular wall, accelerate the formation of foam cells, and promote the occurrence and development of atherosclerosis. Autophagy is associated with the development of atherosclerosis. However, the mechanism of Hippo regulation of atherosclerosis has not meant to be clarified. In view of the pivotal role of this signaling pathway in maintaining cell growth, proliferation, and differentiation, the imbalance of Hippo is related to atherosclerosis and related diseases. In this review, we emphasized Hippo as a hub for regulating atherosclerosis and discussed its potential targets in pathophysiology, human diseases, and related pharmacology.


Assuntos
Aterosclerose , Células Endoteliais , Aterosclerose/metabolismo , Células Endoteliais/metabolismo , Células Espumosas/metabolismo , Humanos , Macrófagos/metabolismo , Transdução de Sinais
19.
DNA Cell Biol ; 41(3): 262-275, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35180350

RESUMO

Inflammation is a double-edged sword. The moderate inflammatory response is a fundamental defense mechanism produced by the body's resistance to dangerous stimuli and a repair process of the body itself. Increasing studies have confirmed that the overactivation of the inflammasome is involved in the occurrence and development of inflammatory diseases. Strictly controlling the overactivation of the inflammasome and preventing excessive inflammatory response have always been the research focus on inflammatory diseases. However, the endogenous regulatory mechanism of inflammasome is not completely clear. The tripartite motif (TRIM) protein is one of the members of E3 ligases in the process of ubiquitination. The universality and importance of the functions of TRIM members are recognized, including the regulation of inflammatory response. This article will focus on research on the relationship between TRIMs and NLRP3 Inflammasome, which may help us make some references for future related research and the discovery of treatment methods.


Assuntos
Inflamassomos/fisiologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/fisiologia , Proteínas com Motivo Tripartido/fisiologia , Animais , Inflamação/etiologia , Inflamação/fisiopatologia , Modelos Biológicos , Transdução de Sinais/fisiologia
20.
Genes Dis ; 9(6): 1701-1715, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36157502

RESUMO

GTPase-activating SH3 domain-binding protein 2 (G3BP2) is a mediator that responds to environmental stresses through stress granule formation and is involved in the progression of chronic diseases. However, no studies have examined the contribution of G3BP2 in the oscillatory shear stress (OSS)-induced endothelial dysfunction. Here we assessed the effects of G3BP2 in endothelial cells (ECs) function and investigated the underlying mechanism. Using shear stress apparatus and partial ligation model, we identified that stress granule-related genes in ECs could be induced by OSS with RNA-seq, and then confirmed that G3BP2 was highly and specifically expressed in athero-susceptible endothelia in the OSS regions. G3bp2 -/- Apoe -/- mice had significantly decreased atherosclerotic lesions associated with deficiency of G3BP2 in protecting endothelial barrier function, decreasing monocyte adhesion to ECs and inhibiting the proinflammatory cytokine levels. Furthermore, loss of G3BP2 diminished OSS-induced inflammation in ECs by increasing YAP nucleocytoplasmic shuttling and phosphorylation. These data demonstrate that G3BP2 is a critical OSS regulated gene in regulating ECs function and that G3BP2 inhibition in ECs is a promising atheroprotective therapeutic strategy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA