Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
BMC Cancer ; 24(1): 1117, 2024 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-39251966

RESUMO

BACKGROUND/AIMS: Gastric cancer (GC) ranks among the prevalent types of cancer, and its progression is influenced by the tumor microenvironment (TME). A comprehensive comprehension of the TME associated with GC has the potential to unveil therapeutic targets of significance. METHODS: The complexity and heterogeneity of TME interactions were revealed through our investigation using an integrated analysis of single-cell and bulk-tissue sequencing data. RESULTS: We constructed a single-cell transcriptomic atlas of 150,913 cells isolated from GC patients. Our analysis revealed the intricate nature and heterogeneity of the GC TME and the metabolic properties of major cell types. Furthermore, two cell subtypes, LOX+ Fibroblasts and M2 Macrophages, were enriched in tumor tissue and related to the outcome of GC patients. In addition, LOX+ Fibroblasts were significantly associated with M2 macrophages. immunofluorescence double labeling indicated LOX+ Fibroblasts and M2 Macrophages were tightly localized in GC tissue. The two cell subpopulations strongly interacted in a hypoxic microenvironment, yielding an immunosuppressive phenotype. Our findings further suggest that LOX+ Fibroblasts may act as a trigger for inducing the differentiation of monocytes into M2 Macrophages via the IL6-IL6R signaling pathway. CONCLUSIONS: Our study revealed the intricate and interdependent communication network between the fibroblast and macrophage subpopulations, which could offer valuable insights for targeted manipulation of the tumor microenvironment.


Assuntos
Fibroblastos , Macrófagos , Análise de Célula Única , Neoplasias Gástricas , Microambiente Tumoral , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Neoplasias Gástricas/metabolismo , Humanos , Microambiente Tumoral/imunologia , Análise de Célula Única/métodos , Macrófagos/metabolismo , Macrófagos/imunologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Receptores Depuradores Classe E/metabolismo , Receptores Depuradores Classe E/genética , Comunicação Celular/imunologia , Proteína-Lisina 6-Oxidase/metabolismo , Proteína-Lisina 6-Oxidase/genética , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Transcriptoma , Transdução de Sinais
2.
J Cancer Res Clin Oncol ; 150(5): 230, 2024 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-38703300

RESUMO

OBJECTIVES: Gastric cancer (GC) is a prevalent malignant tumor widely distributed globally, exhibiting elevated incidence and fatality rates. The gene LAMC2 encodes the laminin subunit gamma-2 chain and is found specifically in the basement membrane of epithelial cells. Its expression is aberrant in multiple types of malignant tumors. This research elucidated a link between LAMC2 and the clinical characteristics of GC and investigated the potential involvement of LAMC2 in GC proliferation and advancement. MATERIALS AND METHODS: LAMC2 expressions were detected in GC cell lines and normal gastric epithelial cell lines via qRT-PCR. Silencing and overexpression of the LAMC2 were conducted by lentiviral transfection. A xenograft mouse model was also developed for in vivo analysis. Cell functional assays were conducted to elucidate the involvement of LAMC2 in cell growth, migration, and penetration. Further, immunoblotting was conducted to investigate the impact of LAMC2 on the activation of signal pathways after lentiviral transfection. RESULTS: In the findings, LAMC2 expression was markedly upregulated in GC cell lines as opposed to normal gastric epithelial cells. In vitro analysis showed that sh-LAMC2 substantially inhibited GC cell growth, migration, and invasion, while oe-LAMC2 displayed a contrasting effect. Xenograft tumor models demonstrated that oe-LAMC2 accelerated tumor growth via high expression of Ki-67. Immunoblotting analysis revealed a substantial decrease in various signaling pathway proteins, PI3K, p-Akt, and Vimentin levels upon LAMC2 knockdown, followed by increased E-cadherin expression. Conversely, its overexpression exhibited contrasting effects. Besides, epithelial-mesenchymal transition (EMT) was accelerated by LAMC2. CONCLUSION: This study provides evidence indicating that LAMC2, by stimulating signaling pathways, facilitated EMT and stimulated the progression of GC cells in laboratory settings and mouse models. Research also explored that the abnormal LAMC2 expression acts as a biomarker for GC.


Assuntos
Proliferação de Células , Laminina , Invasividade Neoplásica , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Neoplasias Gástricas , Neoplasias Gástricas/patologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Humanos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Camundongos , Laminina/metabolismo , Linhagem Celular Tumoral , Camundongos Nus , Transição Epitelial-Mesenquimal , Movimento Celular , Feminino , Masculino , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Ensaios Antitumorais Modelo de Xenoenxerto , Regulação Neoplásica da Expressão Gênica
3.
Arch Biochem Biophys ; 754: 109896, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38417691

RESUMO

AIMS: The purpose of this study was to explore the role of RAE1 in the invasion and metastasis of gastric cancer (GC) cells. MATERIALS AND METHODS: RAE1 expression in GC cells was determined by reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting (WB). Cell models featuring RAE1 gene silencing and overexpression were constructed by lentiviral transfection; The proliferation, migration, and invasion ability of cells were detected by cell counting, colony formation assay, would healing assay, and transwell invasion and migration test. WB analysis of ERK/MAPK signaling pathway (ERK1/2, p-ERK1/2, c-Myc) and EMT-related molecules (ZEB1, E-cadherin, N-cadherin, and Vimentin). RESULTS: The expression level of RAE1 in GC was notably higher than in adjacent tissues. Elevated RAE1 expression correlated with an unfavorable prognosis for GC patients. Knockdown of RAE1, as compared to the control group, resulted in a significant inhibition of proliferation, migration, and invasion abilities in GC cell lines. Furthermore, RAE1 knockdown led to a substantial decrease in the expression of N-cadherin, vimentin, ZEB1, p-ERK1/2, and c-Myc proteins, coupled with a marked increase in E-cadherin expression. The biological effects of RAE1 in GC cells were effectively reversed by the inhibition of the ERK/MAPK signaling pathway using SCH772984. Additionally, RAE1 knockdown demonstrated a suppressive effect on GC tumor size in vivo. Immunohistochemistry (IHC) results revealed significantly lower expression of Ki-67 in RAE1 knockout mice compared to the control group. CONCLUSIONS: RAE1 promotes GC cell migration and invasion through the ERK/MAPK pathway and is a potential therapeutic target for GC therapy.


Assuntos
Transição Epitelial-Mesenquimal , Neoplasias Gástricas , Animais , Humanos , Camundongos , Caderinas/genética , Caderinas/metabolismo , Carcinogênese , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Invasividade Neoplásica/genética , Proteínas Associadas à Matriz Nuclear/genética , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Vimentina/genética , Vimentina/metabolismo
4.
BMC Pediatr ; 24(1): 82, 2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38279097

RESUMO

BACKGROUND: Severe neonatal hyperbilirubinemia could lead to kernicterus and neonatal death. This study aimed to analyze the association between single nucleotide polymorphisms in genes involved in bilirubin metabolism and the incidence of severe hyperbilirubinemia. METHODS: A total of 144 neonates with severe hyperbilirubinemia and 50 neonates without or mild hyperbilirubinemia were enrolled in 3 institutions between 2019 and 2020. Twelve polymorphisms of 5 genes (UGT1A1, SLCO1B1, SLCO1B3, BLVRA, and HMOX1) were analyzed by PCR amplification of genomic DNA. Genotyping was performed using an improved multiplex ligation detection reaction technique based on ligase detection reaction. RESULTS: The frequencies of the A allele in UGT1A1-rs4148323 and the C allele in SLCO1B3-rs2417940 in the severe hyperbilirubinemia group (30.2% and 90.6%, respectively) were significantly higher than those in the controls (30.2% vs.13.0%, 90.6% vs. 78.0%, respectively, both p < 0.05). Haplotype analysis showed the ACG haplotype of UGT1A1 were associated with an increased hyperbilirubinemia risk (OR 3.122, p = 0.001), whereas the GCG haplotype was related to a reduced risk (OR 0.523, p = 0.018). CONCLUSION: The frequencies of the A allele in rs4148323 and the C allele in rs2417940 are highly associated with the incidence of severe hyperbilirubinemia in Chinese Han neonates. TRIAL REGISTRATION: Trial registration number:ChiCTR1800020424; Date of registration:2018-12-29.


Assuntos
Hiperbilirrubinemia Neonatal , Polimorfismo de Nucleotídeo Único , Recém-Nascido , Humanos , Transportador 1 de Ânion Orgânico Específico do Fígado/genética , Alelos , Hiperbilirrubinemia Neonatal/genética , Glucuronosiltransferase/genética , China/epidemiologia , Membro 1B3 da Família de Transportadores de Ânion Orgânico Carreador de Soluto/genética , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo
5.
Front Med (Lausanne) ; 9: 923280, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35721080

RESUMO

Purpose: To investigate the indirect effects of intense pulsed light (IPL) on morphological and pathological changes of the meibomian glands (MGs) in apolipoprotein E knockout (ApoE-/- ) mice and explore the underlying mechanisms. Methods: ApoE-/- mice were treated with or without IPL three times below the lower eyelids and MGs were not directly exposed to irradiation. The eyelids and ocular surface were observed under a stereoscope. The morphology of MGs was examined by photographing and hematoxylin and eosin staining. Lipid droplets in MGs were examined by Oil Red O staining. The ultrastructure of meibocytes and mitochondria was observed under transmission electron microscopy. The relative gene and protein expression in MGs of upper eyelids was determined by immunostaining, Western blot, and qRT-PCR. Results: Three IPL treatments decreased the toothpaste-like plugging of orifices and thickening and irregularity of the upper and lower eyelid margins in ApoE-/- mice. The morphology of some MGs improved after IPL treatments, accompanied by increased proliferation of acinar basal cells and decreased ductal keratinization. Furthermore, the accumulation of hyperchromatic lipid droplets in the acini increased, and the lipid droplets distributed in the cells around the acini were round and small. Compared with untreated ApoE-/- mice, oxidative stress and apoptosis were downregulated by IPL treatment, accompanied by the improvements in mitochondrial structure. Further research showed that IPL treatments reduced the levels of tumor necrosis factor-alpha (TNF-α), interleukin (IL)-17A, IL-6 in MGs and inactivated nuclear factor kappa B (NF-κ B). Conclusion: Collectively, the results demonstrate that indirect effects of IPL can improve the structure and function of MGs and mitigate the progression of MGD, which may be related to the indirect effects of photobiomodulation.

6.
Acta Biochim Biophys Sin (Shanghai) ; 54(7): 893-903, 2022 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-35713317

RESUMO

The presence of senescent cells is associated with renal fibrosis. This study aims to investigate the effect of albumin-induced premature senescence on tubulointerstitial fibrosis and its possible mechanism in vitro. Different concentrations of bovine serum albumim (BSA) with or without si-p21 are used to stimulate HK-2 cells for 72 h, and SA-ß-gal activity, senescence-associated secretory phenotypes (SASPs), LaminB1 are used as markers of senescence. Immunofluorescence staining is performed to characterize the G2/M phase arrest between the control and BSA groups. Alterations in the DNA damage marker γ-H2AX, fibrogenesis, and associated proteins at the G2/M phase, such as p21, p-CDC25C and p-CDK1, are evaluated. Compared with those in the control group, the SA-ß-gal activity, SASP, and γ-H2AX levels are increased in the BSA group, while the level of LaminB1 is decreased. Meanwhile, HK-2 cells blocked at the G2/M phase are significantly increased under the stimulation of BSA, and the levels of p21, p-CDC25C and p-CDK1, as well as fibrogenesis are also increased. When p21 expression is inhibited, the levels of p-CDC25C and p-CDK1 are decreased and the G2/M phase arrest is improved, which decreases the production of fibrogenesis. In conclusion, BSA induces renal tubular epithelial cell premature senescence, which regulates the G2/M phase through the CDC25C/CDK1 pathway, leading to tubulointerstitial fibrosis.


Assuntos
Senescência Celular , Nefropatias , Albuminas/farmacologia , Células Epiteliais/metabolismo , Fibrose , Humanos , Rim/metabolismo , Nefropatias/metabolismo
7.
Cell Death Dis ; 13(5): 495, 2022 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-35614068

RESUMO

Infiltration of eosinophils is associated with and contributes to liver regeneration. Chemotaxis of eosinophils is orchestrated by the eotaxin family of chemoattractants. We report here that expression of eotaxin-1 (referred to as eotaxin hereafter), but not that of either eotaxin-2 or eotaxin-3, were elevated, as measured by quantitative PCR and ELISA, in the proliferating murine livers compared to the quiescent livers. Similarly, exposure of primary murine hepatocytes to hepatocyte growth factor (HGF) stimulated eotaxin expression. Liver specific deletion of Brahma-related gene 1 (Brg1), a chromatin remodeling protein, attenuated eosinophil infiltration and down-regulated eotaxin expression in mice. Brg1 deficiency also blocked HGF-induced eotaxin expression in cultured hepatocytes. Further analysis revealed that Brg1 could directly bind to the proximal eotaxin promoter to activate its transcription. Mechanistically, Brg1 interacted with nuclear factor kappa B (NF-κB)/RelA to activate eotaxin transcription. NF-κB knockdown or pharmaceutical inhibition disrupted Brg1 recruitment to the eotaxin promoter and blocked eotaxin induction in hepatocytes. Adenoviral mediated over-expression of eotaxin overcame Brg1 deficiency caused delay in liver regeneration in mice. On the contrary, eotaxin depletion with RNAi or neutralizing antibodies retarded liver regeneration in mice. More important, Brg1 expression was detected to be correlated with eotaxin expression and eosinophil infiltration in human liver specimens. In conclusion, our data unveil a novel role of Brg1 as a regulator of eosinophil trafficking by activating eotaxin transcription.


Assuntos
Quimiocina CCL11 , DNA Helicases , Regeneração Hepática , Proteínas Nucleares , Fatores de Transcrição , Animais , Células Cultivadas , Quimiocina CCL11/genética , Quimiocina CCL11/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Camundongos , NF-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional
8.
Clin Sci (Lond) ; 136(3): 223-238, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35084460

RESUMO

Extracellular matrix (ECM) accumulation is considered an important pathological feature of diabetic kidney disease (DKD). Histone deacetylase (HDAC) inhibitors protect against kidney injury. However, the potential mechanisms of HDACs in DKD are still largely unknown. Here, we describe a novel feedback loop composed of HDAC2 and miR-205 that regulates ECM production in tubular epithelial cells in individuals with DKD. We found that HDAC2 mRNA expression in peripheral blood was markedly higher in patients with DKD than in patients with diabetes. Nuclear HDAC2 protein expression was increased in TGFß1-stimulated tubular epithelial cells and db/db mice. We also found that miR-205 was regulated by HDAC2 and down-regulated in TGFß1-treated HK2 cells and db/db mice. In addition, HDAC2 reduced histone H3K9 acetylation in the miR-205 promoter region to inhibit its promoter activity and subsequently suppressed miR-205 expression through an SP1-mediated pathway. Furthermore, miR-205 directly targeted HDAC2 and inhibited HDAC2 expression. Intriguingly, miR-205 also regulated its own transcription by inhibiting HDAC2 and increasing histone H3K9 acetylation in its promoter, forming a feedback regulatory loop. Additionally, the miR-205 agonist attenuated ECM production in HK2 cells and renal interstitial fibrosis in db/db mice. In conclusion, the HDAC2/SP1/miR-205 feedback loop may be crucial for the pathogenesis of DKD.


Assuntos
Nefropatias Diabéticas/patologia , Células Epiteliais/metabolismo , Histona Desacetilases/metabolismo , MicroRNAs/metabolismo , Animais , Linhagem Celular , Complicações do Diabetes/enzimologia , Complicações do Diabetes/metabolismo , Complicações do Diabetes/patologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/etiologia , Células Epiteliais/enzimologia , Proteínas da Matriz Extracelular/metabolismo , Retroalimentação , Histona Desacetilase 2/metabolismo , Inibidores de Histona Desacetilases/metabolismo , Humanos , Camundongos , MicroRNAs/genética , Fator de Crescimento Transformador beta1/metabolismo
9.
Front Cell Dev Biol ; 9: 697614, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34631698

RESUMO

Intestinal fibrosis is one of the common pathophysiological processes in inflammatory bowel diseases (IBDs). Previously it has been demonstrated that epithelial-mesenchymal transition (EMT) can contribute to the development of intestinal fibrosis. Here we report that conditional ablation of SIRT1, a class III lysine deacetylase, in intestinal epithelial cells exacerbated 2, 4, 6-trinitro-benzene sulfonic acid (TNBS) induced intestinal fibrosis in mice. SIRT1 activity, but not SIRT1 expression, was down-regulated during EMT likely due to up-regulation of its inhibitor deleted in breast cancer 1 (DBC1). TGF-ß augmented the recruitment of KDM4A, a histone H3K9 demethylase, to the DBC1 promoter in cultured intestinal epithelial cells (IEC-6) leading to DBC1 trans-activation. KDM4A depletion or inhibition abrogated DBC1 induction by TGF-ß and normalized SIRT1 activity. In addition, KDM4A deficiency attenuated TGF-ß induced EMT in IEC-6 cells. In conclusion, our data identify a KDM4-DBC1-SIRT1 pathway that regulates EMT to contribute to intestinal fibrosis.

10.
Front Cell Dev Biol ; 9: 680344, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34621736

RESUMO

Liver fibrosis is mediated by myofibroblasts, a specialized cell type involved in wound healing and extracellular matrix production. Hepatic stellate cells (HSC) are the major source of myofibroblasts in the fibrotic livers. In the present study we investigated the involvement of CXXC-type zinc-finger protein 5 (CXXC5) in HSC activation and the underlying mechanism. Down-regulation of CXXC5 was observed in activated HSCs compared to quiescent HSCs both in vivo and in vitro. In accordance, over-expression of CXXC5 suppressed HSC activation. RNA-seq analysis revealed that CXXC5 influenced multiple signaling pathways to regulate HSC activation. The proto-oncogene MYCL1 was identified as a novel target for CXXC5. CXXC5 bound to the proximal MYCL1 promoter to repress MYCL1 transcription in quiescent HSCs. Loss of CXXC5 expression during HSC activation led to the removal of CpG methylation and acquisition of acetylated histone H3K9/H3K27 on the MYCL1 promoter resulting in MYCL1 trans-activation. Finally, MYCL1 knockdown attenuated HSC activation whereas MYCL1 over-expression partially relieved the blockade of HSC activation by CXXC5. In conclusion, our data unveil a novel transcriptional mechanism contributing to HSC activation and liver fibrosis.

11.
Redox Biol ; 46: 102079, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34454163

RESUMO

Liver injuries induced by various stimuli share in common an acute inflammatory response, in which circulating macrophages home to the liver parenchyma to participate in the regulation of repair, regeneration, and fibrosis. In the present study we investigated the role of hepatocyte-derived C-C motif ligand 7 (CCL7) in macrophage migration during liver injury focusing on its transcriptional regulation. We report that CCL7 expression was up-regulated in the liver by lipopolysaccharide (LPS) injection (acute liver injury) or methionine-and-choline-deficient (MCD) diet feeding (chronic liver injury) paralleling increased macrophage infiltration. CCL7 expression was also inducible in hepatocytes, but not in hepatic stellate cells or in Kupffer cells, by LPS treatment or exposure to palmitate in vitro. Hepatocyte-specific deletion of Brahma-related gene 1 (BRG1), a chromatin remodeling protein, resulted in a concomitant loss of CCL7 induction and macrophage infiltration in the murine livers. Of interest, BRG1-induced CCL7 transcription and macrophage migration was completely blocked by the antioxidant N-acetylcystine. Further analyses revealed that BRG1 interacted with activator protein 1 (AP-1) to regulate CCL7 transcription in hepatocytes in a redox-sensitive manner mediated in part by casein kinase 2 (CK2)-catalyzed phosphorylation of BRG1. Importantly, a positive correlation between BRG1/CCL7 expression and macrophage infiltration was identified in human liver biopsy specimens. In conclusion, our data unveil a novel role for BRG1 as a redox-sensitive activator of CCL7 transcription.


Assuntos
DNA Helicases , Proteínas Nucleares , Animais , Células Cultivadas , Quimiocina CCL7/metabolismo , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Camundongos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oxirredução , Fatores de Transcrição
12.
Exp Mol Pathol ; 122: 104671, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34358519

RESUMO

This study aims to investigate the role of lncRNA RHPN1-AS1 in NPC and its potential regulatory mechanism. The expression of RHPN1-AS1 in tissues and cells was measured by qRT-PCR. The effect of RHPN1-AS1 silencing on biological functions of NPC cells was detected by CCK-8, colony formation, flow cytometry, wound healing, and transwell assays. The protein expression was measured by western blot. The RBPs of RHPN1-AS1 were predicted by Starbase and LncTar, and verify by RIP assay. ESTIMATE was used to analyze the relationship between CELF2 expression and tumor purity. GSEA was used to analyze the downstream signaling pathway of CELF2. In our study, RHPN1-AS1 was up-regulated in NPC tissues and cells. RHPN1-AS1 silencing inhibited cell viability, capacity of proliferation, migration and invasion, promoted apoptosis, decreased protein expression of Bcl-2, MMP2/9, increased protein expression of Bax, caspase-3, and TIMP2 of NPC cells. CELF2 was a target of RHPN1-AS1 and was down-regulated in NPC tissues and cells. CELF2 level was associated with tumor purity negatively. Low expression of CELF2 activated mTORC1 signaling pathway and increased protein expression of p-mTORC1/mTORC1 and p-P70S6K/P70S6K. RHPN1-AS1 silencing eliminated the activating effect of CELF2 silencing on mTORC1 signaling pathway. Moreover, CELF2 silencing reversed the inhibitory effect of RHPN1-AS1 on NPC progression. In conclusion, our findings indicated that RHPN1-AS1 plays an important role in NPC via activating mTORC1 signaling which is modulated by CELF2. RHPN1-AS1 may serve as a potential therapeutic target for NPC treatment.


Assuntos
Proteínas CELF/genética , Carcinogênese/genética , Carcinoma Nasofaríngeo/genética , Proteínas do Tecido Nervoso/genética , RNA Longo não Codificante/genética , Adulto , Idoso , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/patologia , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas de Ligação a RNA/genética
13.
Mol Med Rep ; 24(1)2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34080657

RESUMO

Nasopharyngeal carcinoma (NPC) is a common malignant tumor in South China and is characterized by a high death rate. Ophiopogonin B (OP­B) is a bioactive component of Radix Ophiopogon japonicus, which is frequently used in traditional Chinese medicine to treat cancer. The present study aimed to examine the anti­cancer properties of OP­B on NPC cells. Cell viability and cell proliferation were measured using MTT and EdU assays. Flow cytometry was used to measure cell apoptosis, reactive oxygen species and mitochondrial membrane potential. Western blotting was used to investigate the expression of apoptosis and Hippo signaling pathway proteins. OP­B inhibited the proliferation of NPC cells by inducing apoptosis and disturbing the mitochondrial integrity. OP­B enhanced ROS accumulation. In addition, OP­B promoted the expression of mammalian STE20­like kinase 1, large tumor suppressor 1 and phosphorylated yes­associated protein (YAP) and suppressed the expression of YAP and transcriptional enhanced associate domain in NPC cells. OP­B increased the expression of forkhead box transcription factor O1 in the nuclear fraction. In conclusion, OP­B has therapeutic potential and feasibility in the development of novel YAP inhibitors for NPC.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Saponinas/farmacologia , Espirostanos/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Via de Sinalização Hippo , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neoplasias Nasofaríngeas/patologia , Transdução de Sinais/efeitos dos fármacos
14.
Front Cell Dev Biol ; 9: 617073, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33816466

RESUMO

Multiple organ failure is one of the most severe consequences in patients with septic shock. Liver injury is frequently observed during this pathophysiological process. In the present study we investigated the contribution of Brahma related gene 1 (BRG1), a chromatin remodeling protein, to septic shock induced liver injury. When wild type (WT) and liver conditional BRG1 knockout (LKO) mice were injected with lipopolysaccharide (LPS), liver injury was appreciably attenuated in the LKO mice compared to the WT mice as evidenced by plasma ALT/AST levels, hepatic inflammation and apoptosis. Of interest, there was a down-regulation of sterol response element binding protein 1a (SREBP1a), known to promote liver injury, in the LKO livers compared to the WT livers. BRG1 did not directly bind to the SREBP1a promoter. Instead, BRG1 was recruited to the toll-like receptor 4 (TLR4) promoter and activated TLR4 transcription. Ectopic TLR4 restored SREBP1a expression in BRG1-null hepatocytes. Congruently, adenovirus carrying TLR4 or SREBP1a expression vector normalized liver injury in BRG1 LKO mice injected with LPS. Finally, a positive correlation between BRG1 and TLR4 expression was detected in human liver biopsy specimens. In conclusion, our data demonstrate that a BRG1-TLR4-SREBP1a axis that mediates LPS-induced liver injury in mice.

15.
Cell Death Dis ; 10(12): 899, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31776330

RESUMO

Excessive fibrogenic response in the liver disrupts normal hepatic anatomy and function heralding such end-stage liver diseases as hepatocellular carcinoma and cirrhosis. Sinusoidal endothelial cells contribute to myofibroblast activation and liver fibrosis by undergoing endothelial-mesenchymal transition (EndMT). The underlying mechanism remains poorly defined. Here we report that inhibition or endothelial-specific deletion of MKL1, a transcriptional modulator, attenuated liver fibrosis in mice. MKL1 inhibition or deletion suppressed EndMT induced by TGF-ß. Mechanistically, MKL1 was recruited to the promoter region of TWIST1, a master regulator of EndMT, and activated TWIST1 transcription in a STAT3-dependent manner. A small-molecule STAT3 inhibitor (C188-9) alleviated EndMT in cultured cells and bile duct ligation (BDL) induced liver fibrosis in mice. Finally, direct inhibition of TWIST1 by a small-molecule compound harmine was paralleled by blockade of EndMT in cultured cells and liver fibrosis in mice. In conclusion, our data unveil a novel mechanism underlying EndMT and liver fibrosis and highlight the possibility of targeting the STAT3-MKL1-TWIST1 axis in the intervention of aberrant liver fibrogenesis.


Assuntos
Endotélio/patologia , Cirrose Hepática/genética , Cirrose Hepática/patologia , Mesoderma/patologia , Transativadores/metabolismo , Transcrição Gênica , Proteína 1 Relacionada a Twist/genética , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Ductos Biliares/patologia , Tetracloreto de Carbono , Células Endoteliais/metabolismo , Células HEK293 , Humanos , Ligadura , Cirrose Hepática/sangue , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Fator de Transcrição STAT3/metabolismo , Transativadores/deficiência , Proteína 1 Relacionada a Twist/metabolismo
16.
Front Immunol ; 10: 3020, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32082297

RESUMO

Systemic lupus erythematosus (SLE) is characterized by high levels of autoantibodies and multiorgan tissue damage. The pathogenesis of splenomegaly in SLE remains unknown. In this study, the role of immunoglobulin G (IgG) generation and deposition in the inflammation of the spleen and associated dysfunction in SLE was investigated. In the lupus mice, we observed the development of spontaneous splenomegaly, and we found that lupus serum IgG is an important pathological factor involved in the initiation of inflammation and further germinal center (GC) and plasma cell formation. We discovered that macrophages of the splenic marginal zone are dispensable for the GC response induced by lupus IgG, but red pulp macrophages are important for GC responses. Furthermore, we found that pathogenic lupus IgG promotes inflammation and GC formation through the macrophage-mediated secretion of TNF-α. Syk inhibitor treatment suppressed the changes in the histopathology of the spleen induced by lupus IgG. This study will contribute to the understanding of the pathogenesis of splenomegaly in lupus and promote the development of an effective therapeutic strategy for SLE.


Assuntos
Imunoglobulina G/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Esplenomegalia/imunologia , Animais , Feminino , Centro Germinativo/imunologia , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Plasmócitos/imunologia , Baço/imunologia , Esplenomegalia/genética , Esplenomegalia/patologia
17.
Artigo em Inglês | MEDLINE | ID: mdl-30056131

RESUMO

Excessive reactive oxygen species (ROS) causes irreparable damages to cells and commit cells to programmed cell death or apoptosis. A panel of well-documented pro-apoptotic genes, including p53 apoptosis effector related to PMP-22 (PERP), are up-regulated and collectively mediate ROS induced apoptosis. The epigenetic mechanism whereby ROS stimulates PERP transcription, however, lacks in-depth characterization. Here we report that the transcriptional modulator megakaryocytic leukemia 1 (MKL1) is activated by H2O2 treatment in skeletal muscle cells (C2C12). Small interfering RNA (siRNA) mediated silencing or small-molecule compound (CCG-1423) mediated inhibition of MKL1 attenuated H2O2 induced apoptosis of C2C12 cells. Over-expression of MKL1 potentiated trans-activation of PERP whereas MKL1 ablation/inhibition abrogated the induction of PERP by H2O2 in C2C12 cells. Mechanistically, MKL1 interacted with and was recruited to the PERP promoter by the transcription factor E2F1. Once bound to the PERP promoter, MKL1 engaged the histone demethylase KDM3A to modulate the chromatin structure surrounding the PERP promoter thereby leading to PERP trans-activation. Depletion of either E2F1 or KDM3A blocked the induction of PERP by H2O2. In conclusion, our data illustrate a novel epigenetic pathway that links PERP transcription to ROS-induced apoptosis in skeletal muscle cells.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA