Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell ; 57(4): 721-734, 2015 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-25699712

RESUMO

MicroRNAs (miRNAs) regulate the translational potential of their mRNA targets and control many cellular processes. The key step in canonical miRNA biogenesis is the cleavage of the primary transcripts by the nuclear RNase III enzyme Drosha. Emerging evidence suggests that the miRNA biogenic cascade is tightly controlled. However, little is known whether Drosha is regulated. Here, we show that Drosha is targeted by stress. Under stress, p38 MAPK directly phosphorylates Drosha at its N terminus. This reduces its interaction with DiGeorge syndrome critical region gene 8 and promotes its nuclear export and degradation by calpain. This regulatory mechanism mediates stress-induced inhibition of Drosha function. Reduction of Drosha sensitizes cells to stress and increases death. In contrast, increase in Drosha attenuates stress-induced death. These findings reveal a critical regulatory mechanism by which stress engages p38 MAPK pathway to destabilize Drosha and inhibit Drosha-mediated cellular survival.


Assuntos
Ribonuclease III/fisiologia , Estresse Fisiológico , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Transporte Ativo do Núcleo Celular , Sobrevivência Celular , Células HEK293 , Humanos , Fosforilação , Proteólise , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/fisiologia , Ribonuclease III/genética , Ribonuclease III/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
J Neuroinflammation ; 18(1): 295, 2021 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-34930303

RESUMO

BACKGROUND: Parkinson's disease (PD) is characterized by degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc), accompanied by accumulation of α-synuclein, chronic neuroinflammation and autophagy dysfunction. Previous studies suggested that misfolded α-synuclein induces the inflammatory response and autophagy dysfunction in microglial cells. The NLRP3 inflammasome signaling pathway plays a crucial role in the neuroinflammatory process in the central nervous system. However, the relationship between autophagy deficiency and NLRP3 activation induced by α-synuclein accumulation is not well understood. METHODS: Through immunoblotting, immunocytochemistry, immunofluorescence, flow cytometry, ELISA and behavioral tests, we investigated the role of p38-TFEB-NLRP3 signaling pathways on neuroinflammation in the α-synuclein A53T PD models. RESULTS: Our results showed that increased protein levels of NLRP3, ASC, and caspase-1 in the α-synuclein A53T PD models. P38 is activated by overexpression of α-synuclein A53T mutant, which inhibited the master transcriptional activator of autophagy TFEB. And we found that NLRP3 was degraded by chaperone-mediated autophagy (CMA) in microglial cells. Furthermore, p38-TFEB pathways inhibited CMA-mediated NLRP3 degradation in Parkinson's disease. Inhibition of p38 had a protective effect on Parkinson's disease model via suppressing the activation of NLRP3 inflammasome pathway. Moreover, both p38 inhibitor SB203580 and NLRP3 inhibitor MCC950 not only prevented neurodegeneration in vivo, but also alleviated movement impairment in α-synuclein A53T-tg mice model of Parkinson's disease. CONCLUSION: Our research reveals p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease, which could be a potential therapeutic strategy for PD. p38-TFEB pathways promote microglia activation through inhibiting CMA-mediated NLRP3 degradation in Parkinson's disease. In this model, p38 activates NLRP3 inflammasome via inhibiting TFEB in microglia. TFEB signaling negatively regulates NLRP3 inflammasome through increasing LAMP2A expression, which binds to NLRP3 and promotes its degradation via chaperone-mediated autophagy (CMA). NLRP3-mediated microglial activation promotes the death of dopaminergic neurons.


Assuntos
Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Autofagia Mediada por Chaperonas/fisiologia , Microglia/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Doença de Parkinson/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Autofagia Mediada por Chaperonas/efeitos dos fármacos , Imidazóis/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/efeitos dos fármacos , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Doença de Parkinson/genética , Proteólise/efeitos dos fármacos , Piridinas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
3.
Biochem Biophys Res Commun ; 509(2): 390-394, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30594390

RESUMO

Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra. Prevailing evidence suggests that abnormal autophagy and mitochondrial dysfunction participate in the process of PD. However, many damages of neuronal functions are regulated by intracellular Ca2+ signaling and the contribution of mitochondrial Ca2+ to the process of neurodegeneration is still unclear. MPP+, the metabolite of a neurotoxin MPTP, causes symptom of PD in animal models by selectively destroying dopaminergic neurons in substantia nigra. Here we report that mitochondrial Ca2+ uniporter (MCU) participated in MPP+-induced autophagic cell death in SH-SY5Y cells. Pharmacological agonist of MCU or exogenous expressed MCU can partially reduce MPP+-induced autophagic cell death. Down-regulation of MCU enhanced autophagic cell death via AMPK activation, which was independent of Beclin1 and PI3K. These findings show that the mitochondrial calcium dyshomeostasis contributes to MPP+-induced neuronal degeneration, and MCU may be a potential therapeutic target of PD through the prevention of pathological autophagy.


Assuntos
1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/efeitos dos fármacos , Cálcio/metabolismo , Mitocôndrias/efeitos dos fármacos , Neurônios/efeitos dos fármacos , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagia/genética , Proteína Beclina-1/genética , Proteína Beclina-1/metabolismo , Biotransformação , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Sinalização do Cálcio , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Humanos , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neurônios/metabolismo , Neurônios/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo
4.
Cell Commun Signal ; 17(1): 28, 2019 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-30902093

RESUMO

BACKGROUND: Macroautophagy (hereafter autophagy) is a tightly regulated process that delivers cellular components to lysosomes for degradation. Damage-regulated autophagy modulator 1 (DRAM1) induces autophagy and is necessary for p53-mediated apoptosis. However, the signalling pathways regulated by DRAM1 are not fully understood. METHODS: HEK293T cells were transfected with FLAG-DRAM1 plasmid. Autophagic proteins (LC3 and p62), phosphorylated p53 and the phosphorylated proteins of the class I PI3K-Akt-mTOR-ribosomal protein S6 (rpS6) signalling pathway were detected with Western blot analysis. Cellular distribution of DRAM1 was determined with immunostaining. DRAM1 was knocked down in HEK293T cells using siRNA oligos which is confirmed by quantitative RT-PCR. Cells were serum starved for 18 h after overexpression or knockdown of DRAM1 to decrease the rpS6 activity to the basal level, and then the cells were stimulated with insulin growth factor, epidermal growth factor or serum. rpS6 phosphorylation and rpS6 were detected with Western blotting. Similarly, after overexpression or knockdown of DRAM1, phosphorylation of IGF-1Rß and IGF-1R were examined with Western blotting. Cell viability was determined with CCK-8 assay and colony formation assay. Finally, human cancer cells Hela, SW480, and HCT116 were transfected with the FLAG-DRAM1 plasmid and phosphorylated rpS6 and rpS6 were detected with Western blot analysis. RESULTS: DRAM1 induced autophagy and inhibited rpS6 phosphorylation in an mTORC1-dependent manner in HEK293T cells. DRAM1 didn't affect the phosphorylated and total levels of p53. Furthermore, DRAM1 inhibited the activation of the PI3K-Akt pathway stimulated with growth factors or serum. DRAM1 was localized at the plasma membrane and regulate the phosphorylation of IGF-1 receptor. DRAM1 decreased cell viability and colony numbers upon serum starvation. Additionally, DRAM1 inhibited rpS6 phosphorylation in several human cancer cells. CONCLUSIONS: Here we provided evidence that DRAM1 inhibited rpS6 phosphorylation in multiple cell types. DRAM1 inhibited the phosphorylation of Akt and the activation of Akt-rpS6 pathway stimulated with growth factors and serum. Furthermore, DRAM1 regulated the activation of IGF-1 receptor. Thus, our results identify that the class I PI3K-Akt-rpS6 pathway is regulated by DRAM1 and may provide new insight into the potential role of DRAM1 in human cancers.


Assuntos
Autofagia/fisiologia , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Proteínas de Membrana , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor IGF Tipo 1/metabolismo , Proteína S6 Ribossômica/metabolismo , Apoptose , Proliferação de Células , Sobrevivência Celular , Células HCT116 , Células HEK293 , Células HeLa , Humanos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/fisiologia , Fosforilação , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo
5.
J Biol Chem ; 287(8): 5797-805, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22215669

RESUMO

Synaptic and extrasynaptic NMDA receptors (NMDARs) appear to play opposite roles in neuronal survival and death. Here we report the new findings on the dysregulation of survival factor, myocyte enhancer factor 2D (MEF2D), by extrasynaptic NMDARs. Excitotoxicity led to the NMDAR-dependent degradation of MEF2D protein and inhibition of its transactivation activity in mature cortical neurons. The activation of extrasynaptic NMDARs alone was sufficient for degradation of MEF2D. Calpain directly cleaved MEF2D in vitro and blocking this protease activity greatly attenuated NMDAR signaled degradation of MEF2D in neurons. Consistently, inhibition of calpain protected cortical neurons from NMDA-induced excitotoxicity. Furthermore, knockdown of MEF2D sensitized neurons to NMDA-induced excitotoxicity, which was not protected by calpain inhibition. Collectively, these findings suggest that dysregulation of MEF2D by calpain may mediate excitotoxicity via an extrasynaptic NMDAR-dependent manner.


Assuntos
Calpaína/metabolismo , Neurotoxinas/toxicidade , Proteólise/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Fatores de Transcrição/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Glucose/deficiência , Fatores de Transcrição MEF2 , N-Metilaspartato/toxicidade , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/enzimologia , Neurônios/metabolismo , Ratos , Transdução de Sinais/efeitos dos fármacos , Sinapses/enzimologia
6.
J Biol Chem ; 287(41): 34246-55, 2012 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-22891246

RESUMO

Parkinson disease (PD) is characterized by the selective demise of dopaminergic (DA) neurons in the substantial nigra pars compacta. Dysregulation of transcriptional factor myocyte enhancer factor 2D (MEF2D) has been implicated in the pathogenic process in in vivo and in vitro models of PD. Here, we identified a small molecule bis(3)-cognitin (B3C) as a potent activator of MEF2D. We showed that B3C attenuated the toxic effects of neurotoxin 1-methyl-4-phenylpyridinium (MPP(+)) by activating MEF2D via multiple mechanisms. B3C significantly reduced MPP(+)-induced oxidative stress and potentiated Akt to down-regulate the activity of MEF2 inhibitor glycogen synthase kinase 3ß (GSK3ß) in a DA neuronal cell line SN4741. Furthermore, B3C effectively rescued MEF2D from MPP(+)-induced decline in both nucleic and mitochondrial compartments. B3C offered SN4741 cells potent protection against MPP(+)-induced apoptosis via MEF2D. Interestingly, B3C also protected SN4741 cells from wild type or mutant A53T α-synuclein-induced cytotoxicity. Using the in vivo PD model of C57BL/6 mice treated with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine hydrochloride (MPTP), we showed that B3C maintained redox homeostasis, promoted Akt function activity, and restored MEF2D level in midbrain neurons. Moreover, B3C greatly prevented the loss of tyrosine hydroxylase signal in substantial nigra pars compacta DA neurons and ameliorated behavioral impairments in mice treated with MPTP. Collectedly, our studies identified B3C as a potent neuroprotective agent whose effectiveness relies on its ability to effectively up-regulate MEF2D in DA neurons against toxic stress in models of PD in vitro and in vivo.


Assuntos
Apoptose/efeitos dos fármacos , Neurônios Dopaminérgicos/metabolismo , Fatores de Regulação Miogênica/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fármacos Neuroprotetores/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Transtornos Parkinsonianos/tratamento farmacológico , Tacrina/análogos & derivados , 1-Metil-4-Fenil-1,2,3,6-Tetra-Hidropiridina , 1-Metil-4-fenilpiridínio/efeitos adversos , 1-Metil-4-fenilpiridínio/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Linhagem Celular , Dopaminérgicos/efeitos adversos , Dopaminérgicos/farmacologia , Neurônios Dopaminérgicos/patologia , Herbicidas/efeitos adversos , Herbicidas/farmacologia , Fatores de Transcrição MEF2 , Masculino , Camundongos , Transtornos Parkinsonianos/metabolismo , Transtornos Parkinsonianos/patologia , Substância Negra/metabolismo , Substância Negra/patologia , Tacrina/farmacologia , Tirosina 3-Mono-Oxigenase/metabolismo
7.
Autophagy ; 19(3): 822-838, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-35941759

RESUMO

TFEB (transcription factor EB) regulates multiple genes involved in the process of macroautophagy/autophagy and plays a critical role in lifespan determination. However, the detailed mechanisms that regulate TFEB activity are not fully clear. In this study, we identified a role for HSP90AA1 in modulating TFEB. HSP90AA1 was phosphorylated by CDK5 at Ser 595 under basal condition. This phosphorylation inhibited HSP90AA1, disrupted its binding to TFEB, and impeded TFEB's nuclear localization and subsequent autophagy induction. Pro-autophagy signaling attenuated CDK5 activity and enhanced TFEB function in an HSP90AA1-dependent manner. Inhibition of HSP90AA1 function or decrease in its expression significantly attenuated TFEB's nuclear localization and transcriptional function following autophagy induction. HSP90AA1-mediated regulation of a TFEB ortholog was involved in the extended lifespan of Caenorhabditis elegans in the absence of its food source bacteria. Collectively, these findings reveal that this regulatory process plays an important role in modulation of TFEB, autophagy, and longevity.Abbreviations : AL: autolysosome; AP: autophagosome; ATG: autophagy related; BafA1: bafilomycin A1; CDK5: cyclin-dependent kinase 5; CDK5R1: cyclin dependent kinase 5 regulatory subunit 1; CR: calorie restriction; FUDR: 5-fluorodeoxyuridine; HSP90AA1: heat shock protein 90 alpha family class A member 1; MAP1LC3: microtubule associated protein 1 light chain 3; NB: novobiocin sodium; SQSTM1: sequestosome 1; TFEB: transcription factor EB; WT: wild type.


Assuntos
Autofagia , Longevidade , Animais , Autofagia/genética , Núcleo Celular/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Autofagossomos/metabolismo , Transdução de Sinais/genética , Chaperonas Moleculares/metabolismo , Caenorhabditis elegans/metabolismo , Lisossomos/metabolismo
8.
J Biol Chem ; 286(18): 16504-15, 2011 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-21454572

RESUMO

Neuroprotective strategies, including free radical scavengers, ion channel modulators, and anti-inflammatory agents, have been extensively explored in the last 2 decades for the treatment of neurological diseases. Unfortunately, none of the neuroprotectants has been proved effective in clinical trails. In the current study, we demonstrated that methylene blue (MB) functions as an alternative electron carrier, which accepts electrons from NADH and transfers them to cytochrome c and bypasses complex I/III blockage. A de novo synthesized MB derivative, with the redox center disabled by N-acetylation, had no effect on mitochondrial complex activities. MB increases cellular oxygen consumption rates and reduces anaerobic glycolysis in cultured neuronal cells. MB is protective against various insults in vitro at low nanomolar concentrations. Our data indicate that MB has a unique mechanism and is fundamentally different from traditional antioxidants. We examined the effects of MB in two animal models of neurological diseases. MB dramatically attenuates behavioral, neurochemical, and neuropathological impairment in a Parkinson disease model. Rotenone caused severe dopamine depletion in the striatum, which was almost completely rescued by MB. MB rescued the effects of rotenone on mitochondrial complex I-III inhibition and free radical overproduction. Rotenone induced a severe loss of nigral dopaminergic neurons, which was dramatically attenuated by MB. In addition, MB significantly reduced cerebral ischemia reperfusion damage in a transient focal cerebral ischemia model. The present study indicates that rerouting mitochondrial electron transfer by MB or similar molecules provides a novel strategy for neuroprotection against both chronic and acute neurological diseases involving mitochondrial dysfunction.


Assuntos
Complexo de Proteínas da Cadeia de Transporte de Elétrons/metabolismo , Azul de Metileno/farmacologia , Mitocôndrias/enzimologia , Fármacos Neuroprotetores/farmacologia , Doença de Parkinson Secundária/tratamento farmacológico , Animais , Linhagem Celular , Complexo de Proteínas da Cadeia de Transporte de Elétrons/antagonistas & inibidores , Inibidores Enzimáticos/farmacologia , Glicólise/efeitos dos fármacos , Masculino , Neurônios/enzimologia , Neurônios/patologia , Consumo de Oxigênio/efeitos dos fármacos , Doença de Parkinson Secundária/induzido quimicamente , Doença de Parkinson Secundária/enzimologia , Doença de Parkinson Secundária/patologia , Doença de Parkinson Secundária/fisiopatologia , Ratos , Ratos Sprague-Dawley , Rotenona/efeitos adversos , Rotenona/farmacologia , Substância Negra/enzimologia , Substância Negra/patologia , Substância Negra/fisiopatologia , Desacopladores/efeitos adversos , Desacopladores/farmacologia
9.
J Neurochem ; 122(6): 1203-10, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22764880

RESUMO

The myocyte enhancer factor 2A-D (MEF2) proteins are members of the MCM1-agamous-deficiens-serum response factor family of transcription factors. Various MEF2 isoform proteins are enriched in neurons and exhibit distinct patterns of expression in different regions of the brain. In neurons, MEF2 functions as a converging factor to regulate many neuronal functions including survival. MEF2 activities are tightly controlled in neurons in response to stress. Whether stress signal may differentially regulate MEF2s remains largely unknown. In this work, we showed that MEF2A, but not MEF2C or MEF2D, was modified by ubiquitination in dopaminergic neuronal cell line SN4741 cells. MEF2A was ubiquitinated at its N'-terminus, and the ubiquitination of MEF2A was first detectable in the nuclear compartment and later in the cytoplasm. Ubiquitination of MEF2A correlated with reduced DNA-binding activity and transcriptional activity. More importantly, disturbing the degradation of ubiquitinated MEF2A through proteasome pathway by neurotoxins known to induce Parkinson's disease features in model animals caused accumulation of ubiquitinated MEF2A, reduced MEF2 activity, and impaired cellular viability. Our work thus provides the first evidence to demonstrate an isoforms-specific regulation of MEF2s by ubiquitination-proteasome pathway in dopaminergic neuronal cell by neurotoxins, suggesting that stress signal and cellular context-dependent dysregulation of MEF2s may underlie the loss of neuronal viability.


Assuntos
Neurônios Dopaminérgicos/metabolismo , Fatores de Regulação Miogênica/metabolismo , Estresse Fisiológico/fisiologia , Ubiquitinação/fisiologia , Animais , Linhagem Celular , Neurônios Dopaminérgicos/efeitos dos fármacos , Neurônios Dopaminérgicos/fisiologia , Fatores de Transcrição MEF2 , Camundongos , Fatores de Regulação Miogênica/fisiologia , Neurotoxinas/toxicidade , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Estresse Fisiológico/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos
10.
Front Oncol ; 12: 852076, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35978813

RESUMO

Background: Amide proton transfer (APT) imaging as an emerging MRI approach has been used for distinguishing tumor recurrence (TR) and treatment effects (TEs) in glioma patients, but the initial results from recent studies are different. Aim: The aim of this study is to systematically review and quantify the diagnostic performance of APT in assessing treatment response in patients with post-treatment gliomas. Methods: A systematic search in PubMed, EMBASE, and the Web of Science was performed to retrieve related original studies. For the single and added value of APT imaging in distinguishing TR from TEs, we calculated pooled sensitivity and specificity by using Bayesian bivariate meta-analyses. Results: Six studies were included, five of which reported on single APT imaging parameters and four of which reported on multiparametric MRI combined with APT imaging parameters. For single APT imaging parameters, the pooled sensitivity and specificity were 0.85 (95% CI: 0.75-0.92) and 0.88 (95% CI: 0.74-0.97). For multiparametric MRI including APT, the pooled sensitivity and specificity were 0.92 (95% CI: 0.85-0.97) and 0.83 (95% CI: 0.55-0.97), respectively. In addition, in the three studies reported on both single and added value of APT imaging parameters, the combined imaging parameters further improved diagnostic performance, yielding pooled sensitivity and specificity of 0.91 (95% CI: 0.80-0.97) and 0.92 (95% CI: 0.79-0.98), respectively, but the pooled sensitivity was 0.81 (95% CI: 0.65-0.93) and specificity was 0.82 (95% CI: 0.61-0.94) for single APT imaging parameters. Conclusion: APT imaging showed high diagnostic performance in assessing treatment response in patients with post-treatment gliomas, and the addition of APT imaging to other advanced MRI techniques can improve the diagnostic accuracy for distinguishing TR from TE.

11.
Radiat Res ; 197(4): 324-331, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35104874

RESUMO

The purpose of this study was to use a 3-dimensional arterial spin labeling (3D ASL) magnetic resonance (MR) method to measure cerebral blood flow (CBF) before and after radiotherapy, and correlate changes with time after receiving radiotherapy and cognitive function. Patients with nasopharyngeal carcinoma receiving radiotherapy at our institution were recruited for the study. Participants were divided into three groups: Pre-radiotherapy control (PC) group, acute reaction period (ARP) group, and delayed reaction period (DRP)group. Thirty-four patients were included in the study. Compared with the PC group, the ARP group exhibited significantly decreased perfusion in the left anterior cingulate cortex (ACC) and right putamen, and increased perfusion in the right cerebellum (Crus 1), right inferior occipital gyrus, left lingual gyrus, left precuneus, and left calcarine gyrus. in the DRP group, increased perfusion was noted in the right cerebellum (Crus 1) and decreased perfusion in the left superior frontal gyrus. CBF differences were observed in several brain areas in the DRP group as compared to the ARP group (P < 0.001). Total Montreal Cognitive Assessment score, and subdomain language and delayed memory recall scores were significantly lower in the ARP and DRP groups than in the PC group (P < 0.05). Data suggest that ASL allows for non-invasive detection of radiation-induced whole-brain CBF changes, which is transient, dynamic and complicated and may be a factor contributing to cognitive impairment induced by radiotherapy for nasopharyngeal carcinoma.


Assuntos
Disfunção Cognitiva , Neoplasias Nasofaríngeas , Encéfalo/diagnóstico por imagem , Mapeamento Encefálico , Disfunção Cognitiva/diagnóstico por imagem , Disfunção Cognitiva/etiologia , Humanos , Imageamento por Ressonância Magnética/métodos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/diagnóstico por imagem , Neoplasias Nasofaríngeas/radioterapia , Perfusão
12.
Crit Care Med ; 39(6): 1467-73, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21336112

RESUMO

OBJECTIVE: Adjunctive therapies that reduce the cerebral edema in bacterial meningitis include osmotic agents. There is a lack of information comparing mannitol vs. hypertonic saline as an osmotic agent for adjunctive therapy of bacterial meningitis. We attempted to elucidate the impact of hypertonic saline in cerebral edema in the setting of bacterial meningitis as well as to explore potential mechanisms of action. DESIGN: Randomized controlled in vivo study. SETTING: University research laboratory. SUBJECTS: Rabbits. INTERVENTIONS: A rabbit model of bacterial meningitis was used comparing 3% hypertonic saline with 20% mannitol as adjunctive therapy. MEASUREMENTS AND MAIN RESULTS: Adjunctive 3% hypertonic saline treatment persistently elevated mean arterial pressure as compared with the model or ampicillin group (p < .01). Although both 20% mannitol and 3% hypertonic saline efficiently elevated serum osmolality for almost 5 hrs (p < .01), 20% mannitol lowered intracranial pressure for only a short time (<2 hrs) and did not elevate cerebral perfusion pressure. Three percent hypertonic saline treatment efficiently lowered intracranial pressure and elevated cerebral perfusion pressure for almost 5 hrs (p < .01). Furthermore, 3% hypertonic saline treatment efficiently elevated serum Na+ concentration for >5 hrs (p < .01). Three percent hypertonic saline treatment was superior to 20% mannitol in lowering leukocyte number and protein content in cerebrospinal fluid (p < .01). Three percent hypertonic saline treatment reduced water content and Evans blue incorporation in the brain (p < .01). Three percent hypertonic saline treatment inhibited aquaporin 4 expression (p < .01) and attenuated pathologic brain damage more efficiently compared with adjuvant 20% mannitol treatment (p < .01). CONCLUSIONS: Adjunctive 3% hypertonic saline treatment significantly elevated mean arterial pressure, reduced intracranial pressure, greatly improved cerebral perfusion pressure, inhibited brain aquaporin 4 expression, reduced cerebral edema, and attenuated brain damage with a superior effect over 20% mannitol in a rabbit bacterial meningitis model.


Assuntos
Edema Encefálico/microbiologia , Edema Encefálico/prevenção & controle , Diuréticos Osmóticos/uso terapêutico , Manitol/uso terapêutico , Meningite devida a Escherichia coli/complicações , Solução Salina Hipertônica/uso terapêutico , Ampicilina/uso terapêutico , Animais , Antibacterianos/uso terapêutico , Quimioterapia Adjuvante , Modelos Animais de Doenças , Meningite devida a Escherichia coli/terapia , Coelhos
13.
Cell Biol Toxicol ; 27(1): 41-7, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20661636

RESUMO

Pulmonary surfactant is a lipoprotein complex on the alveolar surface. It reduces the surface tension at the air-water interface and stabilizes the alveoli during expiration. Surfactant deficiency or dysfunction is associated with occurrence and development of many pulmonary diseases. Family members of CTP:phosphocholine cytidylyltransferase are rate-limiting enzymes for surfactant phospholipid synthesis. We had reported recently that the expression of CTP:phosphocholine cytidylyltransferase alpha (CCT-α) was inhibited during N-methyl-D: -aspartic acid (NMDA)-induced lung injury. But the molecular mechanism underlining remains elusive. In this work, we reported that NMDA induced nitric oxide synthase (NOS) activation and nuclear factor-kB (NF-κB) subunit p65 nuclear translocation in A549 cells, which were responsible for decreased (CCT-α) expression. Furthermore, NOS activation and elevated NO production are upstream regulators for p65 nuclear translocation and (CCT-α) expression inhibition. Our results provided important clues for further elucidating the mechanisms underlying glutamate-induced lung injury.


Assuntos
Colina-Fosfato Citidililtransferase/biossíntese , N-Metilaspartato/farmacologia , NF-kappa B/fisiologia , Óxido Nítrico/fisiologia , Regulação Enzimológica da Expressão Gênica , Humanos , Transdução de Sinais , Células Tumorais Cultivadas
14.
Abdom Radiol (NY) ; 46(3): 894-908, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32975646

RESUMO

PURPOSE: To evaluate and compare the diagnostic performance of percentage changes in apparent diffusion coefficient (∆ADC%) and slow diffusion coefficient (∆D%) for assessing pathological complete response (pCR) to neoadjuvant therapy in patients with locally advanced rectal cancer (LARC). METHODS: A systematic search in PubMed, EMBASE, the Web of Science, and the Cochrane Library was performed to retrieve related original studies. For each parameter (∆ADC% and ∆D%), we pooled the sensitivity, specificity and calculated the area under summary receiver operating characteristic curve (AUROC) values. Meta-regression and subgroup analyses were performed to explore heterogeneity among the studies on ∆ADC%. RESULTS: 15 original studies (804 patients with 805 lesions, 15 studies on ∆ADC%, 4 of the studies both on ∆ADC% and ∆D%) were included. pCR was observed in 213 lesions (26.46%). For the assessment of pCR, the pooled sensitivity, specificity and AUROC of ∆ADC% were 0.83 (95% confidence intervals [CI] 0.76, 0.89), 0.74 (95% CI 0.66, 0.81), 0.87 (95% CI 0.83, 0.89), and ∆D% were 0.70 (95% CI 0.52, 0.84), 0.81 (95% CI 0.65, 0.90), 0.81 (95% CI 0.77, 0.84), respectively. In the four studies on the both metrics, ∆ADC% yielded an equivalent diagnostic performance (AUROC 0.80 [95% CI 0.76, 0.83]) to ∆D%, but lower than in the studies (n = 11) only on ∆ADC% (AUROC 0.88 [95% CI 0.85, 0.91]). Meta-regression and subgroup analyses showed no significant factors affecting heterogeneity. CONCLUSIONS: Our meta-analysis confirms that ∆ADC% could reliably evaluate pCR in patients with LARC after neoadjuvant therapy. ∆D% may not be superior to ∆ADC%, which deserves further investigation.


Assuntos
Terapia Neoadjuvante , Neoplasias Retais , Imagem de Difusão por Ressonância Magnética , Humanos , Neoplasias Retais/diagnóstico por imagem , Neoplasias Retais/terapia , Reto , Resultado do Tratamento
15.
J Biol Chem ; 284(47): 32619-26, 2009 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-19801631

RESUMO

Glycogen synthase kinase 3beta (GSK3beta) has been identified to play important roles in neuronal death. Evidence from both in vitro and in vivo studies indicates that increased GSK3beta activity contributes to neurodegeneration and to the pathogenesis of Alzheimer disease. But the molecular mechanisms that underlie GSK3beta-mediated neurotoxicity remain poorly understood. We reported here that myocyte enhancer factor 2D (MEF2D), a nuclear transcription factor known to promote neuronal survival, is directly phosphorylated by GSK3beta. Our data showed that phosphorylation of MEF2D by GSK3beta at three specific residues in its transactivation domain inhibits MEF2D transcriptional activity. Withdrawal of neuronal activity in cerebellar granule neurons activated GSK3beta in the nucleus, leading to GSK3beta-dependent inhibition of MEF2 function. This inhibition contributed to GSK3beta-mediated neuronal toxicity. Overexpression of MEF2D mutant that is resistant to GSK3beta inhibition protected cerebellar granule neurons from either GSK3beta activation- or neuronal activity deprivation-induced toxicity. These results identify survival factor MEF2D as a novel downstream effector targeted by GSK3beta and define a molecular link between activation of GSK3beta and neuronal survival machinery which may underlie in part GSK3beta-mediated neurotoxicity.


Assuntos
Apoptose , Quinase 3 da Glicogênio Sintase/metabolismo , Fatores de Regulação Miogênica/metabolismo , Neurônios/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Citoplasma/metabolismo , Glicogênio Sintase Quinase 3 beta , Fatores de Transcrição MEF2 , Modelos Biológicos , Fosforilação , Potássio/metabolismo , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , Transdução de Sinais , Transfecção
16.
CNS Neurol Disord Drug Targets ; 18(5): 366-371, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30963986

RESUMO

BACKGROUND & OBJECTIVE: Amyotrophic lateral sclerosis is a progressive neurodegenerative disease that specifically affects motor neurons in the brain and in the spinal cord. Patients with amyotrophic lateral sclerosis usually die from respiratory failure within 3 to 5 years from when the symptoms first appear. Currently, there is no cure for amyotrophic lateral sclerosis. Accumulating evidence suggests that dismantling of neuromuscular junction is an early event in the pathogenesis of amyotrophic lateral sclerosis. CONCLUSION: It is starting to realized that macrophage malfunction contributes to the disruption of neuromuscular junction. Modulation of macrophage activation states may stabilize neuromuscular junction and provide protection against motor neuron degeneration in amyotrophic lateral sclerosis.


Assuntos
Esclerose Lateral Amiotrófica/tratamento farmacológico , Sistemas de Liberação de Medicamentos/métodos , Macrófagos/efeitos dos fármacos , Animais , Humanos , Junção Neuromuscular/efeitos dos fármacos
17.
Artigo em Inglês | MEDLINE | ID: mdl-29732413

RESUMO

Autophagy and inflammation are two processes vital for immune cells to perform their functions. Their proper interplay upon signal is pivotal for proper response to stress. The stress kinase p38α MAPK in microglia senses inflammatory cue LPS, directly phosphorylates ULK1, relieves the autophagic inhibition on the inflammatory machinery, and thus allows for a full immune response.

18.
Autophagy ; 14(6): 1097-1098, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29749797

RESUMO

Macroautophagy/autophagy and inflammation are 2 intertwined processes vital for immune cells to perform their functions. Under resting conditions, autophagy acts as a brake to suppress inflammation in microglia. Upon signal stimulation, their fine-tuned interplay is pivotal for proper response to stress. How inflammatory signals remove this autophagy brake on inflammation remains unclear. In a recent study, we showed that the stress kinase MAPK14/p38α in microglia senses the inflammatory cue lipopolysaccharide (LPS), directly phosphorylates and inhibits ULK1, relieves the autophagic inhibition on the inflammatory machinery, and thus allows for a full immune response.


Assuntos
Autofagia , Proteína Quinase 14 Ativada por Mitógeno , Proteína Homóloga à Proteína-1 Relacionada à Autofagia , Humanos , Inflamação , Peptídeos e Proteínas de Sinalização Intracelular , Microglia , Proteínas Quinases p38 Ativadas por Mitógeno
19.
Curr Pharmacol Rep ; 4(3): 261-275, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34540559

RESUMO

PURPOSE OF THE REVIEW: To reason that targeting chaperone-mediated autophagy (CMA) represents a promising approach for disease therapy, we will summarize advances in researches on the relationship between CMA and diseases and discuss relevant strategies for disease therapy by targeting the CMA process. RECENT FINDINGS: CMA is a unique kind of selective autophagy in lysosomes. Under physiological conditions, CMA participates in the maintenance of cellular homeostasis by protein quality control, bioenergetics, and timely regulated specific substrate-associated cellular processes. Under pathological conditions, CMA interplays with various disease conditions. CMA makes adaptive machinery to address stress, while disease-associated proteins alter CMA which is involved in pathogeneses of diseases. As more proteins are identified as CMA substrates and regulators, dysregulation of CMA has been implicated in an increasing number of diseases, while rectifying CMA alteration may be a benefit for these diseases. SUMMARY: Alterations of CMA in diseases mainly including neurodegenerative diseases and many cancers raise the possibility of targeting CMA to recover cellular homeostasis as one potential strategy for therapy of relevant diseases.

20.
J Cell Biol ; 217(1): 315-328, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29196462

RESUMO

Inflammation and autophagy are two critical cellular processes. The relationship between these two processes is complex and includes the suppression of inflammation by autophagy. However, the signaling mechanisms that relieve this autophagy-mediated inhibition of inflammation to permit a beneficial inflammatory response remain unknown. We find that LPS triggers p38α mitogen-activated protein kinase (MAPK)-dependent phosphorylation of ULK1 in microglial cells. This phosphorylation inhibited ULK1 kinase activity, preventing it from binding to the downstream effector ATG13, and reduced autophagy in microglia. Consistently, p38α MAPK activity is required for LPS-induced morphological changes and the production of IL-1ß by primary microglia in vitro and in the brain, which correlates with the p38α MAPK-dependent inhibition of autophagy. Furthermore, inhibition of ULK1 alone was sufficient to promote an inflammatory response in the absence of any overt inflammatory stimulation. Thus, our study reveals a molecular mechanism that enables the initial TLR4-triggered signaling pathway to inhibit autophagy and optimize inflammatory responses, providing new understanding into the mechanistic basis of the neuroinflammatory process.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Autofagia/fisiologia , Inflamação/patologia , Microglia/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Animais , Proteína Homóloga à Proteína-1 Relacionada à Autofagia/antagonistas & inibidores , Encéfalo/metabolismo , Caspase 1/metabolismo , Linhagem Celular , Células HEK293 , Humanos , Interleucina-1beta/biossíntese , Lipopolissacarídeos , Camundongos , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/genética , Fosforilação , Células RAW 264.7 , Interferência de RNA , RNA Interferente Pequeno/genética , Ratos , Ratos Long-Evans , Receptor 4 Toll-Like/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA