Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Biomed Pharmacother ; 177: 117038, 2024 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-39002441

RESUMO

INTRODUCTION: Dexmedetomidine (DEX), a highly selective α2-adrenergic receptor agonist, is widely used for sedation and anesthesia in patients undergoing hepatectomy. However, the effect of DEX on autophagic flux and liver regeneration remains unclear. OBJECTIVES: This study aimed to determine the role of DEX in hepatocyte autophagic flux and liver regeneration after PHx. METHODS: In mice, DEX was intraperitoneally injected 5 min before and 6 h after PHx. In vitro, DEX was co-incubated with culture medium for 24 h. Autophagic flux was detected by LC3-II and SQSTM1 expression levels in primary mouse hepatocytes and the proportion of red puncta in AML-12 cells transfected with FUGW-PK-hLC3 plasmid. Liver regeneration was assessed by cyclinD1 expression, Edu incorporation, H&E staining, ki67 immunostaining and liver/body ratios. Bafilomycin A1, si-GSK3ß and Flag-tagged GSK3ß, α2-ADR antagonist, GSK3ß inhibitor, AKT inhibitor were used to identify the role of GSK3ß in DEX-mediated autophagic flux and hepatocyte proliferation. RESULTS: Pre- and post-operative DEX treatment promoted liver regeneration after PHx, showing 12 h earlier than in DEX-untreated mice, accompanied by facilitated autophagic flux, which was completely abolished by bafilomycin A1 or α2-ADR antagonist. The suppression of GSK3ß activity by SB216763 and si-GSK3ß enhanced the effect of DEX on autophagic flux and liver regeneration, which was abolished by AKT inhibitor. CONCLUSION: Pre- and post-operative administration of DEX facilitates autophagic flux, leading to enhanced liver regeneration after partial hepatectomy through suppression of GSK3ß activity in an α2-ADR-dependent manner.

2.
J Nutr Biochem ; 126: 109587, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38262562

RESUMO

Calorie restriction (CR) mimetic, resveratrol (RSV), has the capacity of promoting phagocytosis. However, its role in hepatic ischemia and reperfusion injury (HIRI) remains poorly understood. This study aimed to investigate the effect of RSV on alleviating HIRI and explore the underlying mechanisms. RSV was intraperitoneally injected in mice HIRI model, while RSV was co-incubated with culture medium for 24 h in RAW 264.7 cells and kupffer cells. Macrophage efferocytosis was assessed by immunostaining of PI and F4/80. The clearance of apoptotic neutrophils in the liver was determined by immunostaining of Ly6-G and cleaved-caspase-3. HE staining, Suzuki's score, serum levels of ALT, AST, TNF-α and IL-1ß were analyzed to evaluate HIRI. The efferocytosis inhibitor, Cytochalasin D, was utilized to investigate the effect of RSV on HIRI. Western blot was employed to measure the levels of AMPKα, phospho-AMPKα, STAT3, phospho-STAT3 and S1PR1. SiSTAT3 and inhibitors targeting AMPK, STAT3 and S1PR1, respectively, were used to confirm the involvement of AMPK/STAT3/S1PR1 pathway in RSV-mediated efferocytosis and HIRI. RSV facilitated the clearance of apoptotic neutrophils and attenuated HIRI, which was impeded by Cytochalasin D. RSV boosted macrophage efferocytosis by up-regulating the levels of phospho-AMPKα, phospho-STAT3 and S1PR1, which was reversed by AMPK, STAT3 and S1PR1 inhibitors, respectively. Inhibition of STAT3 suppressed RSV-induced clearance of apoptotic neutrophils and exacerbated HIRI. CR mimetic, RSV, alleviates HIRI by promoting macrophages efferocytosis through AMPK/STAT3/S1PR1 pathway, providing valuable insights into the mechanisms underlying the protective effects of CR on attenuating HIRI.


Assuntos
Proteínas Quinases Ativadas por AMP , Traumatismo por Reperfusão , Camundongos , Animais , Resveratrol/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Eferocitose , Restrição Calórica , Citocalasina D/metabolismo , Fígado/metabolismo , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/metabolismo , Macrófagos/metabolismo , Isquemia
3.
Am J Respir Cell Mol Biol ; 70(4): 295-307, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38207123

RESUMO

The role of autophagy in pulmonary microvascular endothelial cells (PMVECs) is controversial in LPS-induced acute lung injury (ALI). Mixed lineage kinase domain-like pseudokinase (MLKL) has recently been reported to maintain cell survival by facilitating autophagic flux in response to starvation rather than its well-recognized role in necroptosis. Using a mouse PMVEC and LPS-induced ALI model, we showed that in PMVECs, MLKL was phosphorylated (p-MLKL) and autophagic flux was accelerated at the early stage of LPS stimulation (1-3 h), manifested by increases in concentrations of lipidated MAP1LC3B/LC3B (microtubule-associated protein 1 light chain 3 ß; LC3-II), decreases in concentrations of SQSTM1/p62 (sequestosome 1), and fusion of the autophagosome and lysosome by pHluorin-mKate2-human LC3 assay, which were all reversed by either MLKL inhibitor or siRNA MLKL. In mice, the inhibition of MLKL increased vascular permeability and aggravated mouse ALI upon 3-hour LPS stimulation. The p-MLKL induced by short-term LPS formed multimers to facilitate the closure of the phagophore by HaloTag-LC3 autophagosome completion assay. The charged multivesicular body protein 2A (CHMP2A) is essential in the process of phagophore closure into the nascent autophagosome. In agreement with the p-MLKL change, CHMP2A concentrations markedly increased during 1-3-hour LPS stimulation. CHMP2A knockdown blocked autophagic flux upon LPS stimulation, whereas CHMP2A overexpression boosted autophagic flux and attenuated mouse ALI even in the presence of MLKL inhibitor. We propose that the activated MLKL induced by short-term LPS facilitates autophagic flux by accelerating the closure of the phagophore via CHMP2A, thus protecting PMVECs and alleviating LPS-induced ALI.


Assuntos
Lesão Pulmonar Aguda , Células Endoteliais , Humanos , Lesão Pulmonar Aguda/metabolismo , Autofagia/genética , Proteínas de Transporte/metabolismo , Células Endoteliais/metabolismo , Lipopolissacarídeos , Pulmão/metabolismo , Proteínas Quinases/genética
4.
Autophagy ; 18(4): 726-744, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34282994

RESUMO

MLKL (mixed lineage kinase domain like pseudokinase) is a well-known core component of necrosome that executes necroptotic cell death upon phosphorylation by RIPK3 (receptor interacting serine/threonine kinase 3). Recent studies also implicate a role of MLKL in endosomal trafficking, which is not always dependent on RIPK3. Using mouse Neuro-2a and L929 as well as human HEK293 and HT29 cells, we show here that MLKL is phosphorylated in response to serum and amino acid deprivation from the culture medium, in a manner that depends on CAMK2/CaMKII (calcium/calmodulin dependent protein kinase II) but not RIPK3. The starvation-induced increase in MLKL phosphorylation was accompanied by decreases in levels of lipidated MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta; LC3-II) and SQSTM1/p62 (sequestosome 1), markers of autophagosomes. These changes were prevented by disrupting either MLKL or CAMK2 by pharmacology and genetic manipulations. Moreover, disrupting MLKL or CAMK2 also inhibited the incorporation of LC3-II into autolysosomes, demonstrating a role of the CAMK2-MLKL pathway in facilitating autophagic flux during short-term starvation, in contrast to necroptosis which suppressed autophagic flux. Furthermore, unlike the necroptotic pathway, the starvation-evoked CAMK2-mediated MLKL phosphorylation protected cells from starvation-induced death. We propose that upon nutrient deprivation, MLKL is activated by CAMK2, which in turn facilitates membrane scission needed for autophagosome maturation, allowing the proper fusion of the autophagosome with lysosome and the subsequent substance degradation. This novel function is independent of RIPK3 and is not involved in necroptosis, implicating new roles for this pseudokinase in cell survival, signaling and metabolism.Abbreviations: CAMK2/CaMKII: calcium/calmodulin dependent protein kinase II; DIABLO/SMAC: direct inhibitor of apoptosis-binding protein with low pI/second mitochondria-derived activator of caspase; ECS: extracellular solution; ESCRT: endosomal sorting complexes required for transport; FBS: fetal bovine serum; GSK3B: glycogen synthase kinase 3 beta; HBSS: Hanks' balanced salt solution; KO: knockout; LC3-II: lipidated microtubule associated protein 1 light chain 3 beta; LDH: lactate dehydrogenase; MLKL: mixed lineage kinase domain like pseudokinase; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; N2a: Neuro-2a neuroblastoma; Nec-1: necrostatin-1; NSA: necrosulfonamide; PBS: phosphate-buffered saline; PI: propidium iodide; PK-hLC3: pHluorin-mKate2-human LC3; RIPK1: receptor interacting serine/threonine kinase 1; RIPK3: receptor interacting serine/threonine kinase 3; ROS: reactive oxygen species; RPS6KB1/S6K: ribosomal protein S6 kinase B1; shRNA: short hairpin RNA; siRNA: small interference RNA; SQSTM1/p62: sequestosome 1; TBS: Tris-buffered saline; TNF/TNF-α: tumor necrosis factor; TSZ, treatment with TNF + DIABLO mimetics + z-VAD-FMK.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Cálcio , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Cálcio/metabolismo , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Células HEK293 , Humanos , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Quinases/metabolismo , RNA Interferente Pequeno/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Proteína Sequestossoma-1/metabolismo , Serina , Serina-Treonina Quinases TOR/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
5.
Brain Imaging Behav ; 14(6): 2500-2511, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32761563

RESUMO

Response inhibition is the ability to suppress automatic actions or behaviors that are not appropriate or are no longer adaptive to the situation. Although many studies have suggested regional brain activation, the nature of the reconfiguration of functional brain networks involved in response inhibition remains unknown. Here, we assessed brain changes associated with response inhibition using graph theoretical analysis applied to functional connectivity data acquired while subjects performed a simple stop-signal task. We identified several ways in which global network organization shifted to meet the demand for response inhibition. Increased demand for response inhibition was associated with a global network configuration with more efficient communication across the network (functional integration) and more specialized processing (functional segregation). Regions distributed in the frontoparietal network and attention networks were found to be highly efficient in the stop condition. Nodal efficiency was significantly associated with reaction time and showed a different pattern between the go and stop conditions. In addition, the conditional differences (stop vs. go) in nodal efficiency and regional task activation were common in the postcentral gyrus (PoCG) and superior temporal lobe gyrus (STG), and a negative correlation between these differences was found in the frontal and parietal lobes. These results provide compelling evidence that response inhibition is associated with truly global changes in brain functional connectivity and additional insights into how defects in response inhibition are associated with neurological or psychiatric difficulties.


Assuntos
Mapeamento Encefálico , Encéfalo , Encéfalo/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Rede Nervosa/diagnóstico por imagem , Lobo Parietal , Tempo de Reação
6.
Microvasc Res ; 128: 103941, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31678362

RESUMO

BACKGROUND: Activation of the proliferation of pulmonary microvascular endothelial cells (PMVECs) is a key step in the recovery of the integrity of endothelial monolayer, which helps to alleviate acute lung injury (ALI). Platelet endothelial aggregation receptor-1 (PEAR1), expressed on endothelial cells, was reported to inhibit the proliferation of vascular endothelial cells and angiogenesis. However, little is known about its role and mechanism in vascular endothelial disorders in ALI. OBJECTIVE: The aim of this study was to investigate the impact of PEAR1 on the proliferation of pulmonary microvascular endothelial cells in ALI. METHODS: We tested the expression level of PEAR1 in the lungs of WT mice in ALI model induced by intestinal IR. Primary human pulmonary microvascular endothelial cells (HPMECs) were stimulated by 1 mg/L LPS in vitro. We synthesized siPEAR1 and Flag-PEAR1 plasmid to verify the role of PEAR1 on regulating the proliferation of HPMECs under LPS condition and to explore related signaling pathways. RESULTS: The expression level of PEAR1 significantly increased in ALI induced by intestinal IR. PEAR1 knockdown enhanced the proliferation level of HPMECs, which, however, was inhibited by PEAR1 overexpression. PEAR1 knockdown activated PI3K/AKT pathway both in steady state and under LPS condition. PI3K inhibitor, LY294002, reversed the increasing proliferation level and cell progression of HPMECs induced by PEAR1 knockdown after LPS challenge. CONCLUSIONS: PEAR1 acts as a negative regulator in the proliferation of HPMECs in ALI model via the PI3K/AKT pathway.


Assuntos
Lesão Pulmonar Aguda/enzimologia , Proliferação de Células , Células Endoteliais/enzimologia , Pulmão/irrigação sanguínea , Microvasos/enzimologia , Receptores de Superfície Celular/metabolismo , Lesão Pulmonar Aguda/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/patologia , Humanos , Masculino , Camundongos Endogâmicos C57BL , Microvasos/patologia , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Superfície Celular/genética , Transdução de Sinais
7.
Cereb Cortex ; 29(11): 4889-4901, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-30810159

RESUMO

Structural and functional differences in brain hemispheric asymmetry have been well documented between female and male adults. However, potential differences in the connectivity patterns of the rich-club organization of hemispheric structural networks in females and males remain to be determined. In this study, diffusion tensor imaging was used to construct hemispheric structural networks in healthy subjects, and graph theoretical analysis approaches were applied to quantify hemisphere and gender differences in rich-club organization. The results showed that rich-club organization was consistently observed in both hemispheres of female and male adults. Moreover, a reduced level of connectivity was found in the left hemisphere. Notably, rightward asymmetries were mainly observed in feeder and local connections among one hub region and peripheral regions, many of which are implicated in visual processing and spatial attention functions. Additionally, significant gender differences were revealed in the rich-club, feeder, and local connections in rich-club organization. These gender-related hub and peripheral regions are involved in emotional, sensory, and cognitive control functions. The topological changes in rich-club organization provide novel insight into the hemisphere and gender effects on white matter connections and underlie a potential network mechanism of hemisphere- and gender-based differences in visual processing, spatial attention and cognitive control.


Assuntos
Cérebro/anatomia & histologia , Cérebro/fisiologia , Caracteres Sexuais , Substância Branca/anatomia & histologia , Substância Branca/fisiologia , Adulto , Encéfalo/anatomia & histologia , Encéfalo/fisiologia , Conectoma/métodos , Imagem de Tensor de Difusão , Feminino , Humanos , Masculino , Vias Neurais/anatomia & histologia , Vias Neurais/fisiologia , Adulto Jovem
8.
Int J Clin Exp Pathol ; 8(10): 13114-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26722509

RESUMO

Ischemia/reperfusion (I/R) injury often occurs, which is one of the major causes of acute kidney injury, thus increasing in-hospital mortality. HIF-2α has a protective role against ischemia of the kidney. Renal ischemia/reperfusion under sevoflurane anesthesia resulted in drastic improvements in renal function. We hypothesized that underlying mechanism responsible for renal protection from sevoflurane pretreatment involves the upregulation of HIF-2α. Sevoflurane pretreatment were performed on WT and HIF-2α knockout mice before renal ischemia/reperfusion. Levels of blood urea nitrogen (BUN) and serum creatinine (Cr) were determined with a standard clinical automatic analyzer. The left kidneys were taken for morphological examination. Expression of HIF-2α in kidney tissue was examined by western blotting. In WT mice, group I/R injury had significantly higher BUN and Cr levels than group control, whereas group I/R + Sev had significantly lower BUN and Cr levels than group I/R injury. Renal HIF-2α expression levels were significantly higher in WT mice of group I/R + Sev than group control and group I/R. In HIF-2α(-/-) mice, group I/R + Sev showed much higher BUN and Cr levels and severer histological damage than group I/R and group control. Renal HIF-2α expression levels were significantly higher in WT mice of group I/R + Sev than group control and group I/R. Our findings suggested that HIF-2α might contribute to the beneficial effect of sevoflurane in renal ischemia/reperfusion injury.


Assuntos
Injúria Renal Aguda/fisiopatologia , Anti-Inflamatórios/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Éteres Metílicos/farmacologia , Traumatismo por Reperfusão/fisiopatologia , Injúria Renal Aguda/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Testes de Função Renal , Camundongos , Camundongos Knockout , Traumatismo por Reperfusão/metabolismo , Sevoflurano , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA