Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 97(6): e0005323, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37255470

RESUMO

Macrophages can serve as a reservoir for human immunodeficiency-1 (HIV-1) virus in host cells, constituting a barrier to eradication, even in patients who are receiving antiretroviral therapy. Although many noncoding RNAs have been characterized as regulators in HIV-1/AIDS-induced immune response and pathogenesis, only a few long noncoding RNAs (lncRNAs) have demonstrated a close association with HIV-1 replication, and the molecular mechanisms remain unknown. In this study, we investigated how lncRNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), related microRNAs, and key inflammatory genes alter HIV-1 replication in macrophages. Our data show that HIV-1 infection modulates the expression of miR-155 and miR-150-5p in a time-dependent manner, which is regulated by MALAT1. MALAT1 induced suppressor of cytokine signaling 1 (SOCS1) expression by sponging miR-150-5p in HIV-1-infected macrophages and stimulated inflammatory mediators triggering receptor expressed on myeloid cells/cold inducible RNA binding protein (TREM 1/CIRP) ligand/receptor. The RNA immunoprecipitation (RIP) assay validated the direct interaction within the MALAT1/miR-150-5p/SOCS1 axis. HIV-1 infection-mediated upregulation of MALAT1, SOCS1, and HIV-1 Gag was attenuated by SN50 (an NF-кB p50 inhibitor). MALAT1 antisense oligonucleotides (ASOs) suppressed HIV-1 p24 production and HIV-1 Gag gene expression and decreased expression of miR-155 and SOCS1, as well as the production of proinflammatory cytokines by HIV-1-infected macrophages. In conclusion, HIV-1 infection induces MALAT1, which attenuates miR-150-5p expression and increases SOCS1 expression, promoting HIV-1 replication and reactivation. These data provide new insights into how MALAT1 alters the macrophage microenvironment and subsequently promotes viral replication and suggest a potential role for targeting MALAT1 as a therapeutic approach to eliminate HIV-1 reservoirs. IMPORTANCE Viral reservoirs constitute an obstacle to curing HIV-1 diseases, despite antiretroviral therapy. Macrophages serve as viral reservoirs in HIV infection by promoting long-term replication and latency. Recent studies have shown that lncRNAs can modulate virus-host interactions, but the underlying mechanisms are not fully understood. In this study, we demonstrate how lncRNA MALAT1 contributes to HIV-1 replication through modulation of the miR-150/SOCS1 axis in human macrophages. Our findings have the potential to identify new therapies for eliminating HIV-1 reservoirs in immune cells.


Assuntos
Infecções por HIV , MicroRNAs , RNA Longo não Codificante , Replicação Viral , Humanos , Infecções por HIV/genética , Macrófagos/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , HIV-1/fisiologia
2.
J Gastroenterol Hepatol ; 37(3): 507-517, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34676588

RESUMO

BACKGROUND AND AIM: Esophageal squamous cell carcinoma (ESCC) is the most significant subtype of esophageal cancer featured with high occurrence. Long noncoding RNAs (lncRNAs) have been proved to modulate the biological properties of cancer cells, including cell proliferation, invasion, migration, and apoptosis. LncRNA protein tyrosine phosphatase receptor type G-antisense RNA 1 (PTPRG-AS1) has been reported to play as an oncogene in diverse cancers. However, the detailed function PTPRG-AS1 may exert in ESCC is unclear. METHODS: PTPRG-AS1 expression in ESCC cells was investigated via quantitative reverse transcription real-time polymerase chain reaction (RT-qPCR). The effects of PTPRG-AS1 on ESCC cell proliferation, migration, glycolysis, and stemness were verified through functional assays. Mechanism assays including RIP assay, RNA pull down assay, and luciferase reporter assays were performed to verify the molecular mechanism of PTPRG-AS1. RESULTS: PTPRG-AS1 silencing hindered the proliferation, migration, glycolysis and stemness of ESCC cells. PTPRG-AS1 regulated pyruvate dehydrogenase kinase 1 (PDK1) expression via sponging miR-599. The PTPRG-AS1/miR-599/PDK1 axis was further verified to aggravate the progression of ESCC cells. CONCLUSION: PTPRG-AS1 sponged miR-599 to up-regulate PDK1 expression, thereby promoting the proliferation and migration as well as glycolysis and stemness properties of ESCC cells.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Monoéster Fosfórico Hidrolases , RNA Longo não Codificante , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/genética , Carcinoma de Células Escamosas do Esôfago/metabolismo , Glicólise/genética , Humanos , MicroRNAs/metabolismo , Monoéster Fosfórico Hidrolases/genética , Monoéster Fosfórico Hidrolases/metabolismo , Piruvato Desidrogenase Quinase de Transferência de Acetil/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Transdução de Sinais
3.
Biochem Genet ; 60(6): 2570-2586, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35639219

RESUMO

Recently, long non-coding RNA (lncRNA) is proved to play critical roles in non-small cell lung cancer (NSCLC) progression. However, the detailed effects of LINC01426 in NSCLC and its functional mechanism remain unknown. The expression of LINC01426, microRNA-143-3p (miR-143-3p), and Ubiquitin-specific peptidase 28 (USP28) was assessed by quantitative real-time polymerase chain reaction (RT-qPCR). The colony-forming ability was determined by colony-forming assay. 5-ethynyl-2'-deoxyuridine (EdU) staining assay was performed to evaluate cell proliferation. The migrated and invaded abilities of cells were measured by transwell assays. Flow cytometry was used to examine cell apoptosis. The protein expression was analyzed by Western blot analysis. The glycolysis ability was analyzed by commercial kits. Dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay, and RNA pull-down assay were used to confirm relationship among LINC01426, miR-143-3p, and USP28. A xenograft experiment was conducted to explore the effects of LINC01426 inhibition in vivo. Our results confirmed that LINC01426 and USP28 expression were increased, while miR-143-3p expression was decreased in NSCLC tissues and cells. Further functional experiments demonstrated that LINC01426 inhibition markedly impaired cell proliferation, migration, invasion, autophagy, and glycolysis while induced apoptosis in NSCLC cells, and LINC01426 derived malignant behaviors of NSCLC cells by sponging miR-143-3p. Additionally, LINC01426 regulated USP28 expression by sponging miR-143-3p. USP28 overexpression partly overturned the inhibitory effect of miR-143-3p on NSCLC progression. Consistently, silencing of LINC01426 significantly inhibited the growth of NSCLC tumor in vivo. LINC01426 accelerated the malignant progression of NSCLC. Mechanistically, LINC01426 acted as a competing endogenous RNA (ceRNA) for miR-143-3p to upregulate USP28 expression.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma Pulmonar de Células não Pequenas/patologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Neoplasias Pulmonares/patologia , MicroRNAs/metabolismo , Proliferação de Células , Linhagem Celular Tumoral , Ubiquitina Tiolesterase
4.
Neurobiol Dis ; 124: 322-334, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30528256

RESUMO

Neural stem/progenitor cells (NPCs) are known to have potent therapeutic effects in neurological disorders through the secretion of extracellular vesicles (EVs). Despite the therapeutic potentials, the numbers of NPCs are limited in the brain, curbing the further use of EVs in the disease treatment. To overcome the limitation of NPC numbers, we used a three transcription factor (Brn2, Sox2, and Foxg1) somatic reprogramming approach to generate induced NPCs (iNPCs) from mouse fibroblasts and astrocytes. The resulting iNPCs released significantly higher numbers of EVs compared with wild-type NPCs (WT-NPCs). Furthermore, iNPCs-derived EVs (iNPC-EVs) promoted NPC function by increasing the proliferative potentials of WT-NPCs. Characterizations of EV contents through proteomics analysis revealed that iNPC-EVs contained higher levels of growth factor-associated proteins that were predicted to activate the down-stream extracellular signal-regulated kinase (ERK) pathways. As expected, the proliferative effects of iNPC-derived EVs on WT-NPCs can be blocked by an ERK pathway inhibitor. Our data suggest potent therapeutic effects of iNPC-derived EVs through the promotion of NPC proliferation, release of growth factors, and activation of ERK pathways. These studies will help develop highly efficient cell-free therapeutic strategies for the treatment of neurological diseases.


Assuntos
Proliferação de Células/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Vesículas Extracelulares/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Técnicas de Reprogramação Celular/métodos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/citologia , Transdução de Sinais/fisiologia
5.
Cell Commun Signal ; 17(1): 96, 2019 08 16.
Artigo em Inglês | MEDLINE | ID: mdl-31419975

RESUMO

Neural stem/progenitor cells (NPCs) are known to have potent therapeutic effects in neurological disorders through secreting exosomes. The limited numbers of NPCs in adult brain and the decline of NPC pool in many neurological disorders restrain the further use of exosomes in treating these diseases. The direct conversion of somatic cells into induced NPCs (iNPCs) provides abundant NPC-like cells to study the therapeutic effects of NPCs-originated exosomes (EXOs). Our recent study demonstrated that iNPCs-derived exosomes (iEXOs) exhibit distinct potential in facilitating the proliferation of NPCs, compared to EXOs, indicating the importance to investigate the effects of EXOs and iEXOs on the differentiation of NPCs, which remains unknown. Here, our results suggest that EXOs, but not iEXOs, promoted neuronal differentiation and neither of them had effect on glial generation. Microarray analysis revealed different miRNA signatures in EXOs and iEXOs, in which miR-21a was highly enriched in EXOs. Perturbation of function assay demonstrated the key roles of miR-21a in the generation of neurons and mediating the neurogenic potential of exosomes. Our data suggest that EXOs and iEXOs may achieve their therapeutic effects in promoting neurogenesis through transferring key miRNAs, which sheds light on the development of highly efficient cell-free therapeutic strategies for treating neurological diseases.


Assuntos
Exossomos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , MicroRNAs/metabolismo , Células-Tronco Neurais/metabolismo , Animais , Camundongos , Neurogênese , Transdução de Sinais
6.
J Neuroinflammation ; 15(1): 79, 2018 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-29540215

RESUMO

BACKGROUND: Extracellular vesicles (EVs) are important in the intercellular communication of the central nervous system, and their release is increased during neuroinflammation. Our previous data demonstrated an increased release of EVs during HIV-1 infection and immune activation in glial cells. However, the molecular mechanism by which infection and inflammation increase EV release remains unknown. In the current study, we investigated the role of glutaminase 1 (GLS1)-mediated glutaminolysis and the production of a key metabolic intermediate α-ketoglutarate on EV release. METHODS: Human monocyte-derived macrophage primary cultures and a BV2 microglia cell line were used to represent the innate immune cells in the CNS. Transmission electron microscopy, nanoparticle tracking analysis, and Western blots were used to determine the EV regulation. GLS1 overexpression was performed using an adenovirus vector in vitro and transgenic mouse models in vivo. Data were evaluated statistically by ANOVA, followed by the Bonferroni post-test for paired observations. RESULTS: Our data revealed an increased release of EVs in GLS1-overexpressing HeLa cells. In HIV-1-infected macrophages and immune-activated microglia BV2 cells, treatment with bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) or CB839, two specific GLS inhibitors, significantly decreased EV release, suggesting a critical role of GLS1 in EV release. Furthermore, addition of α-ketoglutarate or ceramide rescued EV release during BPTES treatment, implicating α-ketoglutarate and ceramide as critical downstream effectors for GLS inhibitors. These findings were further corroborated with the investigation of brain tissues in GLS1-transgenic mice. The EV levels were significantly higher in GLS1 transgenic mice than those in control mice, suggesting that GLS1 increases EV release in vivo. CONCLUSIONS: These findings suggest that GLS1-mediated glutaminolysis and its downstream production of α-ketoglutarate are essential in regulating EV release during HIV-1 infection and immune activation. These new mechanistic regulations may help understand how glutamine metabolism shapes EV biogenesis and release during neuroinflammation.


Assuntos
Vesículas Extracelulares/metabolismo , Glutamatos/metabolismo , Glutaminase/metabolismo , Compostos de Anilina/farmacologia , Benzenoacetamidas/farmacologia , Compostos de Benzilideno/farmacologia , Encéfalo/efeitos dos fármacos , Encéfalo/ultraestrutura , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Sistema Nervoso Central/citologia , Ceramidas/farmacologia , Relação Dose-Resposta a Droga , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Inibidores Enzimáticos/farmacologia , Vesículas Extracelulares/efeitos dos fármacos , Glutamina/metabolismo , Infecções por HIV/patologia , Infecções por HIV/fisiopatologia , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/ultraestrutura , Macrófagos/virologia , Proteínas de Membrana/metabolismo , Microglia/ultraestrutura , Microglia/virologia , Sulfetos/farmacologia , Tiadiazóis/farmacologia
7.
J Neuroinflammation ; 14(1): 87, 2017 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-28427419

RESUMO

BACKGROUND: Extracellular vesicles (EVs) are membrane-contained vesicles shed from cells. EVs contain proteins, lipids, and nucleotides, all of which play important roles in intercellular communication. The release of EVs is known to increase during neuroinflammation. Glutaminase, a mitochondrial enzyme that converts glutamine to glutamate, has been implicated in the biogenesis of EVs. We have previously demonstrated that TNF-α promotes glutaminase expression in neurons. However, the expression and the functionality of glutaminase in astrocytes during neuroinflammation remain unknown. We posit that TNF-α can promote the release of EVs in astrocytes through upregulation of glutaminase expression. RESULTS: Release of EVs, which was demonstrated by electron microscopy, nanoparticle tracking analysis (NTA), and Western Blot, increased in mouse astrocytes when treated with TNF-α. Furthermore, TNF-α treatment significantly upregulated protein levels of glutaminase and increased the production of glutamate, suggesting that glutaminase activity is increased after TNF-α treatment. Interestingly, pretreatment with a glutaminase inhibitor blocked TNF-α-mediated generation of reactive oxygen species in astrocytes, which indicates that glutaminase activity contributes to stress in astrocytes during neuroinflammation. TNF-α-mediated increased release of EVs can be blocked by either the glutaminase inhibitor, antioxidant N-acetyl-L-cysteine, or genetic knockout of glutaminase, suggesting that glutaminase plays an important role in astrocyte EV release during neuroinflammation. CONCLUSIONS: These findings suggest that glutaminase is an important metabolic factor controlling EV release from astrocytes during neuroinflammation.


Assuntos
Astrócitos/metabolismo , Vesículas Extracelulares/metabolismo , Glutaminase/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Regulação para Cima
8.
Brain Behav Immun ; 66: 135-145, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28624534

RESUMO

Glutaminolysis, a metabolic process that converts glutamine to glutamate, is particularly important for the central nervous system since glutamate is the major transmitter of excitatory synapses. Glutaminase is the mitochondrial enzyme that catalyzes the first step of glutaminolysis. Two genes encode at least four isoforms of glutaminase in humans. Gls1 gene encodes isoforms kidney-type glutaminase (KGA) and glutaminase C (GAC) through alternative splicing, whereas Gls2 gene encodes liver-type glutaminase isoforms. KGA and GAC have been associated with several neurological diseases. However, it remains unclear whether changes in their expressions can directly cause brain abnormalities. Using a transgenic approach, we generated mice that overexpressed GAC in the brain. The resulting transgenic mice had severe impairments in spatial and fear learning compared with littermate controls. The learning deficits were consistent with diminished hippocampal long-term potentiation in the hippocampal slices of the GAC transgenic mice. Furthermore, we found increases in astrocyte and microglia markers, inflammatory factors, and a decrease in synapse marker synaptophysin, suggesting neuroinflammation and synaptic changes in the GAC transgenic mouse brains. In conclusion, these findings provide the first evidence that GAC overexpression in the brain has deleterious effects on learning and synaptic integrity in vivo.


Assuntos
Encéfalo/enzimologia , Condicionamento Clássico/fisiologia , Encefalite/enzimologia , Glutaminase/fisiologia , Aprendizagem em Labirinto/fisiologia , Sinapses/enzimologia , Animais , Apoptose , Encefalite/etiologia , Medo , Glutaminase/metabolismo , Hipocampo/enzimologia , Hipocampo/fisiologia , Potenciação de Longa Duração , Camundongos , Camundongos Transgênicos , Neuroglia/enzimologia
9.
Stem Cells ; 33(8): 2574-85, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25833331

RESUMO

Neural progenitor cell (NPC) migration is an essential process for brain development, adult neurogenesis, and neuroregeneration after brain injury. Stromal cell-derived factor-1 (SDF-1, CXCL12) and its traditional receptor CXCR4 are well known to regulate NPC migration. However, the discovery of CXCR7, a newly identified CXCL12 receptor, adds to the dynamics of the existing CXCL12/CXCR4 pair. Antagonists for either CXCR4 or CXCR7 blocked CXCL12-mediated NPC migration in a transwell chemotaxis assay, suggesting that both receptors are required for CXCL12 action. We derived NPC cultures from Cxcr4 knockout (KO) mice and used transwell and stripe assays to determine the cell migration. NPCs derived from Cxcr4 KO mice polarized and migrated in response to CXCL12 gradient, suggesting that CXCR7 could serve as an independent migration receptor. Furthermore, Cxcr4 KO NPCs transplanted into the adult mouse striatum migrated in response to the adjacent injection of CXCL12, an effect that was blocked by a CXCR7 antagonist, suggesting that CXCR7 also mediates NPC migration in vivo. Molecular mechanism studies revealed that CXCR7 interact with Rac1 in the leading edge of the polarized NPCs in the absence of CXCR4. Both CXCR7 and Rac1 are required for extracellular signal-regulated kinases (ERK) 1/2 activation and subsequent NPC migration, indicating that CXCR7 could serve as a functional receptor in CXCL12-mediated NPC migration independent of CXCR4. Together these results reveal an essential role of CXCR7 for CXCL12-mediated NPC migration that will be important to understand neurogenesis during development and in adulthood.


Assuntos
Quimiocina CXCL12/metabolismo , Quimiotaxia , Sistema de Sinalização das MAP Quinases , Células-Tronco Neurais/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Animais , Quimiocina CXCL12/genética , Camundongos , Camundongos Knockout , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Células-Tronco Neurais/citologia , Neuropeptídeos/genética , Neuropeptídeos/metabolismo , Receptores CXCR/genética , Receptores CXCR4/genética , Proteínas rac1 de Ligação ao GTP/genética , Proteínas rac1 de Ligação ao GTP/metabolismo
10.
J Neurochem ; 134(2): 315-26, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25913161

RESUMO

Exposure to atmospheric particulate matter PM2.5 (aerodynamic diameter ≤ 2.5 µm) has been epidemiologically associated with respiratory illnesses. However, recent data have suggested that PM2.5 is able to infiltrate into circulation and elicit a systemic inflammatory response. Potential adverse effects of air pollutants to the central nervous system (CNS) have raised concerns, but whether PM2.5 causes neurotoxicity remains unclear. In this study, we have demonstrated that PM2.5 impairs the tight junction of endothelial cells and increases permeability and monocyte transmigration across endothelial monolayer in vitro, indicating that PM2.5 is able to disrupt blood-brain barrier integrity and gain access to the CNS. Exposure of primary neuronal cultures to PM2.5 resulted in decrease in cell viability and loss of neuronal antigens. Furthermore, supernatants collected from PM2.5 -treated macrophages and microglia were also neurotoxic. These macrophages and microglia significantly increased extracellular levels of glutamate following PM2.5 exposure, which were negatively correlated with neuronal viability. Pre-treatment with NMDA receptor antagonist MK801 alleviated neuron loss, suggesting that PM2.5 neurotoxicity is mediated by glutamate. To determine the potential source of excess glutamate production, we investigated glutaminase, the main enzyme for glutamate generation. Glutaminase was reduced in PM2.5 -treated macrophages and increased in extracellular vesicles, suggesting that PM2.5 induces glutaminase release through extracellular vesicles. In conclusion, these findings indicate PM2.5 as a potential neurotoxic factor, crucial to understanding the effects of air pollution on the CNS.


Assuntos
Ácido Glutâmico/biossíntese , Glutaminase/metabolismo , Macrófagos/metabolismo , Neurônios/patologia , Material Particulado/toxicidade , Poluentes Atmosféricos/toxicidade , Animais , Western Blotting , Permeabilidade Capilar/fisiologia , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Endotélio Vascular/patologia , Imunofluorescência , Humanos , Síndromes Neurotóxicas/etiologia , Síndromes Neurotóxicas/metabolismo , Síndromes Neurotóxicas/fisiopatologia , Ratos , Ratos Sprague-Dawley
11.
Sci Rep ; 14(1): 9167, 2024 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-38649770

RESUMO

Syndecan-binding protein (SDCBP) was reported to stimulate the advancement of esophageal squamous cell carcinoma (ESCC) and could potentially be a target for ESCC treatment. There is a growing corpus of research on the anti-tumor effects of iron chelators; however, very few studies have addressed the involvement of dexrazoxane in cancer. In this study, structure-based virtual screening was employed to select drugs targeting SDCBP from the Food and Drug Administration (FDA)-approved drug databases. The sepharose 4B beads pull-down assay revealed that dexrazoxane targeted SDCBP by interacting with its PDZ1 domain. Additionally, dexrazoxane inhibited ESCC cell proliferation and anchorage-independent colony formation via SDCBP. ESCC cell apoptosis and G2 phase arrest were induced as measured by the flow cytometry assay. Subsequent research revealed that dexrazoxane attenuated the binding ability between SDCBP and EGFR in an immunoprecipitation assay. Furthermore, dexrazoxane impaired EGFR membrane localization and inactivated the EGFR/PI3K/Akt pathway. In vivo, xenograft mouse experiments indicated that dexrazoxane suppressed ESCC tumor growth. These data indicate that dexrazoxane might be established as a potential anti-cancer agent in ESCC by targeting SDCBP.


Assuntos
Proliferação de Células , Receptores ErbB , Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Sinteninas , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/metabolismo , Carcinoma de Células Escamosas do Esôfago/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores ErbB/metabolismo , Animais , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patologia , Proliferação de Células/efeitos dos fármacos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinteninas/metabolismo , Linhagem Celular Tumoral , Apoptose/efeitos dos fármacos , Camundongos Nus , Antineoplásicos/farmacologia
12.
J Neurochem ; 125(6): 897-908, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23578284

RESUMO

Glutaminase 1 is the main enzyme responsible for glutamate production in mammalian cells. The roles of macrophage and microglia glutaminases in brain injury, infection, and inflammation are well documented. However, little is known about the regulation of neuronal glutaminase, despite neurons being a predominant cell type of glutaminase expression. Using primary rat and human neuronal cultures, we confirmed that interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α), two pro-inflammatory cytokines that are typically elevated in neurodegenerative disease states, induced neuronal death and apoptosis in vitro. Furthermore, both intracellular and extracellular glutamate levels were significantly elevated following IL-1ß and/or TNF-α treatment. Pre-treatment with N-Methyl-D-aspartate (NMDA) receptor antagonist MK-801 blocked cytokine-induced glutamate production and alleviated the neurotoxicity, indicating that IL-1ß and/or TNF-α induce neurotoxicity through glutamate. To determine the potential source of excess glutamate production in the culture during inflammation, we investigated the neuronal glutaminase and found that treatment with IL-1ß or TNF-α significantly upregulated the kidney-type glutaminase (KGA), a glutaminase 1 isoform, in primary human neurons. The up-regulation of neuronal glutaminase was also demonstrated in situ in a murine model of HIV-1 encephalitis. In addition, IL-1ß or TNF-α treatment increased the levels of KGA in cytosol and TNF-α specifically increased KGA levels in the extracellular fluid, away from its main residence in mitochondria. Together, these findings support neuronal glutaminase as a potential component of neurotoxicity during inflammation and that modulation of glutaminase may provide therapeutic avenues for neurodegenerative diseases.


Assuntos
Ácido Glutâmico/biossíntese , Glutaminase/metabolismo , Interleucina-1beta/metabolismo , Neurônios/citologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Morte Celular , Córtex Cerebral/citologia , Citosol/enzimologia , Encefalite Viral/enzimologia , Encefalite Viral/virologia , Espaço Extracelular/metabolismo , HIV-1 , Humanos , Interleucina-1beta/toxicidade , Espaço Intracelular/metabolismo , Masculino , Camundongos , Camundongos SCID , Mitocôndrias/enzimologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/toxicidade , Regulação para Cima
13.
Stem Cells ; 30(11): 2571-83, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22987307

RESUMO

Chemokine CXCL12 is widely expressed in the central nervous system and essential for the proper functioning of human neural progenitor cells (hNPCs). Although CXCL12 is known to function through its receptor CXCR4, recent data have suggested that CXCL12 binds to chemokine receptor CXCR7 with higher affinity than to CXCR4. However, little is known about the function of CXCR7 in hNPCs. Using a primary hNPC culture system, we demonstrated that CXCL12 promotes hNPC survival in the events of camptothecin-induced apoptosis or growth factor deprivation, and that this effect requires both CXCR7 and CXCR4. Through fluorescence-activated cell sorting analysis and immunocytochemistry, we determined that CXCR7 is mainly localized in the early endosome, while CXCR4 is more broadly expressed at the cell surface and on both early and recycling endosomes. Furthermore, we found that endocytosis is required for the prosurvival function of CXCL12. Using dual-color total internal reflection fluorescence microscopy and immunoprecipitation, we demonstrated that CXCR7 quickly trafficks to plasma membrane in mediating CXCL12 endocytosis and colocalizes with CXCR4 after CXCL12 treatment. Investigating the molecular mechanisms, we found that ERK1/2 endocytotic signaling pathway is essential for hNPC survival upon apoptotic challenges. Consistent with these findings, a significantly higher number of apoptotic NPCs were found in the developing brain of CXCR7 knockout mice. In conclusion, CXCL12 protects hNPCs from apoptotic challenges through CXCR7- and CXCR4-mediated endocytotic signaling. Since survival of hNPCs is important for neurogenesis, CXCR7 may become a new therapeutic target to properly regulate critical processes of brain development.


Assuntos
Quimiocina CXCL12/fisiologia , Endocitose , Células-Tronco Neurais/fisiologia , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Animais , Apoptose/efeitos dos fármacos , Camptotecina/farmacologia , Sobrevivência Celular , Células Cultivadas , Cérebro/embriologia , Cérebro/metabolismo , Endossomos/metabolismo , Técnicas de Silenciamento de Genes , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Knockout , Células-Tronco Neurais/metabolismo , Ligação Proteica , RNA Interferente Pequeno/genética , Receptores CXCR/genética , Receptores CXCR/fisiologia , Receptores CXCR4/genética , Receptores CXCR4/fisiologia
14.
Oncol Rep ; 49(6)2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37083097

RESUMO

Aurora kinase A (AURKA), a serine/threonine kinase that regulates mitotic processes, has garnered significant interest given its association with the development of several types of cancer. In the present study, it was shown that AURKA expression was significantly upregulated in esophageal squamous cell carcinoma (ESCC) and could serve as a diagnostic and prognostic indicator based on data obtained from The Cancer Genome Atlas (TCGA) and immunohistochemical analysis. In addition, AURKA was functionally associated with ESCC cell proliferation and colony formation in vitro and knockdown of AURKA inhibited ESCC tumor growth in vivo. Both bioinformatics analysis and pull­down assays demonstrated that TPX2 interacted with AURKA, and their expression was correlated. AURKA cooperated with TPX2 to regulate ESCC progression via the PI3K/Akt pathway. Furthermore, AURKA or TPX2 expression levels were negatively associated with the infiltration of cytotoxic cells, CD8+ T cells and mast cells, but positively associated with Th2 cells. The present study provided a relatively comprehensive understanding of the oncogenic roles of AURKA in ESCC based on data obtained from TCGA combined with experimental analysis.


Assuntos
Neoplasias Esofágicas , Carcinoma de Células Escamosas do Esôfago , Humanos , Carcinoma de Células Escamosas do Esôfago/genética , Aurora Quinase A , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Linfócitos T CD8-Positivos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Linhagem Celular Tumoral , Biologia Computacional , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
15.
J Neurosci ; 31(42): 15195-204, 2011 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-22016553

RESUMO

Microglia represent the main cellular targets of HIV-1 in the brain. Infected and/or activated microglia play a pathogenic role in HIV-associated neurocognitive disorders (HAND) by instigating primary dysfunction and subsequent death of neurons. Although microglia are known to secrete neurotoxins when infected with HIV-1, the detailed mechanism of neurotoxicity remains unclear. Using a human microglia primary culture system and macrophage-tropic HIV-1 strains, we have now demonstrated that HIV-1 infection of microglia resulted in a significant increase in extracellular glutamate concentrations and elevated levels of neurotoxicity. RNA and protein analysis revealed upregulation of the glutamate-generating enzyme glutaminase isoform glutaminase C in HIV-1-infected microglia. The clinical relevance of these findings was further corroborated with investigation of postmortem brain tissues. The glutaminase C levels in the brain tissues of HIV dementia individuals were significantly higher than HIV serum-negative control and correlated with elevated concentrations of glutamate. When glutaminase was subsequently inhibited by siRNA or by a small molecular inhibitor, the HIV-induced glutamate production and the neuronal loss was diminished. In conclusion, these findings support glutaminase as a potential component of the HAND pathogenic process as well as a novel therapeutic target in their treatment.


Assuntos
Glutaminase/metabolismo , Infecções por HIV/patologia , Microglia/enzimologia , Microglia/virologia , Análise de Variância , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Encéfalo/patologia , Caspase 3/metabolismo , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , Desintegrinas/farmacologia , Maleato de Dizocilpina/farmacologia , Ensaio de Imunoadsorção Enzimática/métodos , Antagonistas de Aminoácidos Excitatórios/farmacologia , Feto , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Ácido Glutâmico/metabolismo , Glutaminase/genética , Glutamina/metabolismo , HIV-1/genética , HIV-1/metabolismo , Humanos , Microglia/efeitos dos fármacos , Proteínas Associadas aos Microtúbulos/metabolismo , RNA Interferente Pequeno/farmacologia , Receptores de Superfície Celular/metabolismo , Sais de Tetrazólio , Tiazóis , Fatores de Tempo
16.
Glia ; 59(4): 641-54, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21294163

RESUMO

Reactive astrogliosis is one of the pathological hallmarks of neurodegenerative diseases. Inflammatory cytokines, such as TNF-α and IL-1ß, have been shown to mediate the reactive astrogliosis in neurodegenerative diseases; however, the molecular mechanism remains unclear. In this study, we investigated the role of transcription factor FOXO3a on astrocyte proliferation, one primary aspect of severe reactive astrogliosis. Our results confirmed that TNF-α and IL-1ß increased astrocyte proliferation, as determined by Ki67 and BrdU immunostaining. Furthermore, we found that cytokine-mediated astrocyte proliferation was accompanied by an increase of the phosphorylation and reduced nuclear expression of FOXO3a. Intracranial injection of TNF-α and IL-1ß induced astrocyte proliferation and hypertrophy, which was associated with reduced nuclear expression of Foxo3a in astrocytes. To determine the function of FOXO3a in astrocyte proliferation, wild type FOXO3a was overexpressed with adenovirus, which subsequently upregulated p27Kip1 and Gadd45α, and significantly inhibited cytokine-induced astrocyte proliferation. In contrast, overexpression of dominant negative FOXO3a decreased p27Kip1, upregulated cyclin D1 and promoted astrocyte proliferation. Along the same line, astrocytes isolated from Foxo3a-null mice have higher proliferative potential. In response to intracranial injection of cytokines, Foxo3a-null mice manifested severe astrogliosis in vivo. In conclusion, FOXO3a is important in restraining astrocyte proliferation during proinflammatory cytokine stimulation and loss of function of FOXO3a may be responsible for the proliferation of astrocytes in the severe form of reactive astrogliosis. Understanding the key regulatory role of FOXO3a in reactive astrogliosis may provide a novel therapeutic target during neuroinflammation.


Assuntos
Astrócitos/metabolismo , Proliferação de Células/efeitos dos fármacos , Fatores de Transcrição Forkhead/metabolismo , Gliose/metabolismo , Interleucina-1beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Análise de Variância , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/patologia , Western Blotting , Células Cultivadas , Proteína Forkhead Box O3 , Gliose/patologia , Humanos , Imuno-Histoquímica , Interleucina-1beta/farmacologia , Masculino , Camundongos , Fosforilação , Fator de Necrose Tumoral alfa/farmacologia
17.
J Vis Exp ; (172)2021 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-34279498

RESUMO

Controversies have always existed in research related to reading abilities; on whether printed words are perceived in a feedforward manner based on orthographic information after which, other representations, such as phonology and semantics are activated, or whether these are fully interactive and high-level semantic information affects early processing. An interference paradigm was implemented in the presented protocol of phonological and semantic judgment tasks that utilized the same precede-target pairs to explore the relative order of phonological and semantic activation. The high- and low-frequency target words were preceded with three conditions: semantically related, phonological-related (homophones), or unrelated. The results showed that the induced P200 component of low-frequency word pairs was significantly greater than high-frequency words in both the semantic and phonological tasks. In addition, both the homophones in the semantic task and the semantically related pairs in the phonological task caused reduction in N400 when compared to the the control condition, word frequency-independently. It is worth noting that for the low-frequency pairs in the phonological judgment task, the P200 released by the semantically related word pairs was significantly larger than that in the control condition. Overall, semantic processing in phonological tasks and phonological processing in semantic tasks were found in both high- and low-frequency words, suggesting that the interaction between semantics and phonology may operate in a task-independent manner. However, the specific time this interaction occurred may have been affected by the task and frequency.


Assuntos
Eletroencefalografia , Semântica , Eletrofisiologia , Potenciais Evocados , Feminino , Humanos , Masculino , Fonética , Tempo de Reação , Leitura
18.
Front Psychol ; 12: 608379, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33679524

RESUMO

The present study aims to delineate the working mechanism of prediction in sentence comprehension, by disentangling the influence of the facilitated general memory retrieval from the coexistent influence of the predicted language-specific semantic and/or syntactic information for the first time. The results support that prediction might influence the downstream cognitive processing in two aspects: (1) the pre-activated information facilitates the retrieval of a matched input in memory and, (2) the pre-activated information interacts with higher-level semantic/syntactic processing. More importantly, the present findings suggest that these two types of influences seem to occur at different stages of sentence comprehension: the facilitated memory retrieval of the input modulates N400 amplitude and the latency of post-N400 late central-parietal positivity/P600, while the predicted semantic/syntactic information and/or their interactions modulate the amplitude of the late positivity. The present findings would be helpful for interpreting the underlying mechanism of observed effects in prediction studies.

19.
Front Psychol ; 12: 637238, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33716906

RESUMO

Unlike in English, the role of phonology in word recognition in Chinese is unclear. In this event-related potential experiment, we investigated the role of phonology in reading both high- and low-frequency two-character compound Chinese words. Participants executed semantic and homophone judgment tasks of the same precede-target pairs. Each pair of either high- or low-frequency words were either unrelated (control condition) or related semantically or phonologically (homophones). The induced P200 component was greater for low- than for high-frequency word-pairs both in semantic and phonological tasks. Homophones in the semantic judgment task and semantically-related words in the phonology task both elicited a smaller N400 than the control condition, word frequency-independently. However, for low-frequency words in the phonological judgment task, it was found that the semantically related pairs released a significantly larger P200 than the control condition. Thus, the semantic activation of both high- and low-frequency words may be no later than phonological activation.

20.
Transl Cancer Res ; 10(6): 2653-2662, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35116578

RESUMO

BACKGROUND: Esophagectomy is the standard treatment for early-stage esophageal cancer but is associated with high morbidity and mortality. Thus, endoscopic resection is increasingly used as an alternative option. However, the literature is inconsistent regarding the efficacy of these treatments. This meta-analysis aimed to compare the efficacy and safety of these two treatments. METHODS: A systematic electronic search of PubMed, EMBASE, and the Cochrane Library was performed for studies comparing endoscopic resection and surgery for early-stage esophageal cancer. The overall survival, tumor recurrence, major adverse events, procedure-related mortality, and R0 resection rates were investigated. Forest plots were constructed based on the random-effects model. RESULTS: We found 15 studies involving 2,467 and 2,264 patients who underwent endoscopic resection and surgery, respectively. The meta-analysis showed that patients undergoing endoscopic resection had significantly fewer major adverse events (relative risk, 0.46; 95% confidence interval, 0.33-0.64) and a lower procedure-related mortality rate (relative risk, 0.27; 95% confidence interval, 0.10-0.73) than those undergoing surgery. The number of postprocedural stricture events did not significantly differ between the two treatments (relative risk, 0.89; 95% confidence interval, 0.53-1.49). Endoscopic resection was associated with higher recurrence rates (relative risk, 1.69; 95% confidence interval, 0.99-2.89) and lower R0 resection rates (relative risk, 0.92; 95% confidence interval, 0.86-0.98) than surgery. There may be some advantage conferred by esophagectomy in the long-term survival outcomes (hazard ratio, 1.21; 95% confidence interval, 1.02-1.43). DISCUSSION: Endoscopic resection is a minimally invasive and safe treatment for early-stage esophageal cancer. However, esophagectomy may be associated with better long-term survival.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA