Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Biol Chem ; 300(5): 107260, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38582447

RESUMO

Thoracic aortic dissection (TAD) is a highly dangerous cardiovascular disorder caused by weakening of the aortic wall, resulting in a sudden tear of the internal face. Progressive loss of the contractile apparatus in vascular smooth muscle cells (VSMCs) is a major event in TAD. Exploring the endogenous regulators essential for the contractile phenotype of VSMCs may aid the development of strategies to prevent TAD. Krüppel-like factor 15 (KLF15) overexpression was reported to inhibit TAD formation; however, the mechanisms by which KLF15 prevents TAD formation and whether KLF15 regulates the contractile phenotype of VSMCs in TAD are not well understood. Therefore, we investigated these unknown aspects of KLF15 function. We found that KLF15 expression was reduced in human TAD samples and ß-aminopropionitrile monofumarate-induced TAD mouse model. Klf15KO mice are susceptible to both ß-aminopropionitrile monofumarate- and angiotensin II-induced TAD. KLF15 deficiency results in reduced VSMC contractility and exacerbated vascular inflammation and extracellular matrix degradation. Mechanistically, KLF15 interacts with myocardin-related transcription factor B (MRTFB), a potent serum response factor coactivator that drives contractile gene expression. KLF15 silencing represses the MRTFB-induced activation of contractile genes in VSMCs. Thus, KLF15 cooperates with MRTFB to promote the expression of contractile genes in VSMCs, and its dysfunction may exacerbate TAD. These findings indicate that KLF15 may be a novel therapeutic target for the treatment of TAD.


Assuntos
Aneurisma da Aorta Torácica , Dissecção da Aorta Torácica , Fatores de Transcrição Kruppel-Like , Miócitos de Músculo Liso , Fatores de Transcrição , Animais , Humanos , Masculino , Camundongos , Angiotensina II/metabolismo , Angiotensina II/farmacologia , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/genética , Aneurisma da Aorta Torácica/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/metabolismo , Miócitos de Músculo Liso/patologia , Fenótipo , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética
2.
J Mol Cell Cardiol ; 189: 25-37, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38395296

RESUMO

Aortic dissection (AD) is the most catastrophic vascular disease with a high mortality rate. Trimethylamine N-oxide (TMAO), a gut microbial metabolite, has been implicated in the pathogenesis of cardiovascular diseases. However, the role of TMAO in AD and the underlying mechanisms remain unclear. This study aimed to explore the effects of TMAO on AD. Plasma and fecal samples from patients with AD and healthy individuals were collected to analyze TMAO levels and gut microbial species, respectively. The plasma levels of TMAO were significantly higher in 253 AD patients compared with those in 98 healthy subjects (3.47, interquartile range (IQR): 2.33 to 5.18 µM vs. 1.85, IQR: 1.40 to 3.35 µM; p < 0.001). High plasma TMAO levels were positively associated with AD severity. An increase in the relative abundance of TMA-producing genera in patients with AD was revealed using 16S rRNA sequencing. In the angiotensin II or ß-aminopropionitrile-induced rodent model of AD, mice fed a TMAO-supplemented diet were more likely to develop AD compared to mice fed a normal diet. Conversely, TMAO depletion mitigated AD formation in the BAPN model. RNA sequencing of aortic endothelial cells isolated from mice administered TMAO revealed significant upregulation of genes involved in inflammatory pathways. The in vitro experiments verified that TMAO promotes endothelial dysfunction and activates nuclear factor (NF)-κB signaling. The in vivo BAPN-induced AD model confirmed that TMAO increased aortic inflammation. Our study demonstrates that the gut microbial metabolite TMAO aggravates the development of AD at least in part by inducing endothelial dysfunction and inflammation. This study provides new insights into the etiology of AD and ideas for its management.


Assuntos
Dissecção Aórtica , Microbioma Gastrointestinal , Metilaminas , Humanos , Camundongos , Animais , Microbioma Gastrointestinal/fisiologia , RNA Ribossômico 16S , Aminopropionitrilo , Células Endoteliais , Inflamação , Dissecção Aórtica/etiologia
3.
J Biol Chem ; 299(10): 105226, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37673339

RESUMO

Successful muscle regeneration following injury is essential for functional homeostasis of skeletal muscles. Krüppel-like factor 15 (KLF15) is a metabolic transcriptional regulator in the muscles. However, little is known regarding its function in muscle regeneration. Here, we examined microarray datasets from the Gene Expression Omnibus database, which indicated downregulated KLF15 in muscles from patients with various muscle diseases. Additionally, we found that Klf15 knockout (Klf15KO) impaired muscle regeneration following injury in mice. Furthermore, KLF15 expression was robustly induced during myoblast differentiation. Myoblasts with KLF15 deficiency showed a marked reduction in their fusion capacity. Unbiased transcriptome analysis of muscles on day 7 postinjury revealed downregulated genes involved in cell differentiation and metabolic processes in Klf15KO muscles. The FK506-binding protein 51 (FKBP5), a positive regulator of myoblast differentiation, was ranked as one of the most strongly downregulated genes in the Klf15KO group. A mechanistic search revealed that KLF15 binds directly to the promoter region of FKBP5 and activates FKBP5 expression. Local delivery of FKBP5 rescued the impaired muscle regeneration in Klf15KO mice. Our findings reveal a positive regulatory role of KLF15 in myoblast differentiation and muscle regeneration by activating FKBP5 expression. KLF15 signaling may be a novel therapeutic target for muscle disorders associated with injuries or diseases.


Assuntos
Mioblastos , Proteínas de Ligação a Tacrolimo , Animais , Humanos , Camundongos , Diferenciação Celular/genética , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos Knockout , Músculo Esquelético/metabolismo , Mioblastos/metabolismo , Regeneração/genética , Proteínas de Ligação a Tacrolimo/metabolismo , Masculino , Camundongos Endogâmicos C57BL
4.
Circ Res ; 128(1): 8-23, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33092471

RESUMO

RATIONALE: Thoracic aortic aneurysm (TAA) leads to substantial mortality worldwide. Familial and syndromic TAAs are highly correlated with genetics. However, the incidence of sporadic isolated TAA (iTAA) is much higher, and the genetic contribution is not yet clear. OBJECTIVE: Here, we examined the genetic characteristics of sporadic iTAA. METHODS AND RESULTS: We performed a genetic screen of 551 sporadic iTAA cases and 1071 controls via whole-exome sequencing. The prevalence of pathogenic mutations in known causal genes was 5.08% in the iTAA cohort. We selected 100 novel candidate genes using a strict strategy, and the suspected functional variants of these genes were significantly enriched in cases compared with controls and carried by 60.43% of patients. We found more severe phenotypes and a lower proportion of hypertension in cases with pathogenic mutations or suspected functional variants. Among the candidate genes, Testin (TES), which encodes a focal adhesion scaffold protein, was identified as a potential TAA causal gene, accounting for 4 patients with 2 missense variants in the LIM1 domain (c.751T>C encoding p.Y251H; c.838T>C encoding p.Y280H) and highly expressed in the aorta. The 2 variants led to a decrease in TES expression. The thoracic aorta was spontaneously dilated in the TesY249H knock-in and Tes-/- mice. Mechanistically, the p.Y249H variant or knockdown of TES led to the repression of vascular smooth muscle cell contraction genes and disturbed the vascular smooth muscle cell contractile phenotype. Interestingly, suspected functional variants of other focal adhesion scaffold genes, including TLN1 (Talin-1) and ZYX (zyxin), were also significantly enriched in patients with iTAA; moreover, their knockdown resulted in decreased contractility of vascular smooth muscle cells. CONCLUSIONS: For the first time, this study revealed the genetic landscape across iTAA and showed that the focal adhesion scaffold genes are critical in the pathogenesis of iTAA.


Assuntos
Aneurisma da Aorta Torácica/genética , Dissecção Aórtica/genética , Proteínas do Citoesqueleto/genética , Adesões Focais/genética , Mutação de Sentido Incorreto , Polimorfismo de Nucleotídeo Único , Proteínas de Ligação a RNA/genética , Adulto , Dissecção Aórtica/diagnóstico por imagem , Dissecção Aórtica/metabolismo , Dissecção Aórtica/fisiopatologia , Animais , Aorta Torácica/diagnóstico por imagem , Aorta Torácica/metabolismo , Aorta Torácica/fisiopatologia , Aneurisma da Aorta Torácica/diagnóstico por imagem , Aneurisma da Aorta Torácica/metabolismo , Aneurisma da Aorta Torácica/fisiopatologia , Estudos de Casos e Controles , Células Cultivadas , Proteínas do Citoesqueleto/metabolismo , Feminino , Adesões Focais/metabolismo , Predisposição Genética para Doença , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Músculo Liso Vascular/diagnóstico por imagem , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiopatologia , Fenótipo , Proteínas de Ligação a RNA/metabolismo , Talina/genética , Talina/metabolismo , Vasoconstrição , Sequenciamento do Exoma , Zixina/genética , Zixina/metabolismo
5.
J Biol Chem ; 295(30): 10212-10223, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32493731

RESUMO

After injury, the coordinated balance of pro- and anti-inflammatory factors in the microenvironment contribute to skeletal muscle regeneration. However, the underlying molecular mechanisms regulating this balance remain incompletely understood. In this study, we examined the roles of microRNAs (miRNAs) in inflammation and muscle regeneration. miRNA-Seq transcriptome analysis of mouse skeletal muscle revealed that miR-223-3p is upregulated in the early stage of muscle regeneration after injury. miR-223-3p knockout resulted in increased inflammation, impaired muscle regeneration, and increased interstitial fibrosis. Mechanistically, we found that myeloid-derived miR-223-3p suppresses the target gene interleukin-6 (Il6), associated with the maintenance of the proinflammatory macrophage phenotype during injury. Administration of IL-6-neutralizing antibody in miR-223-3p-knockout muscle could rescue the impaired regeneration ability and reduce the fibrosis. Together, our results reveal that miR-223-3p improves muscle regeneration by regulating inflammation, indicating that miRNAs can participate in skeletal muscle regeneration by controlling the balance of pro- and anti-inflammatory factors in the skeletal muscle microenvironment.


Assuntos
MicroRNAs/biossíntese , Músculo Esquelético , Regeneração , Regulação para Cima , Animais , Inflamação/genética , Inflamação/metabolismo , Interleucina-6/biossíntese , Interleucina-6/genética , Camundongos , Camundongos Knockout , MicroRNAs/genética , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , RNA-Seq
6.
J Pathol ; 245(2): 222-234, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29537081

RESUMO

Colorectal cancer (CRC) is the third most common cancer worldwide, with more than 1.3 million new cases and 690 000 deaths each year. In China, the incidence of CRC has increased dramatically due to dietary and lifestyle changes, to become the fifth leading cause of cancer-related death. Here, we performed whole-exome sequencing in 50 rectal cancer cases among the Chinese population as part of the International Cancer Genome Consortium research project. Frequently mutated genes and enriched pathways were identified. Moreover, a previously unreported gene, PCDHB3, was found frequently mutated in 5.19% cases. Additionally, PCDHB3 expression was found decreased in 81.6% of CRC tissues and all eight CRC cell lines tested. Low expression and cytoplasmic localization of PCDHB3 predict poor prognosis in advanced CRC. Copy number decrease and/or CpG island hypermethylation contributes to the pervasive decreased expression of PCDHB3. PCDHB3 inhibits CRC cell proliferation, migration, and epithelial-mesenchymal transition. The tumor-suppressive effects of PCDHB3 are partially due to inhibition of NF-κB transcriptional activity through K63 deubiquitination of p50 at lysine 244/252, which increases the binding affinity of inactive p50 homodimer to κB DNA, resulting in competitive inhibition of the transcription of NF-κB target genes by p65 dimers. Our study identified PCDHB3 as a novel tumor suppressor in CRC via inhibition of the NF-κB pathway, and its expression and localization may serve as prognostic markers for advanced CRC. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Biomarcadores Tumorais/genética , Caderinas/genética , Neoplasias Colorretais/genética , Sequenciamento do Exoma , Inativação Gênica , Genes Supressores de Tumor , Mutação , Adulto , Idoso , Animais , Povo Asiático/genética , Biomarcadores Tumorais/metabolismo , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , China , Neoplasias Colorretais/etnologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ilhas de CpG , Metilação de DNA , Regulação para Baixo , Feminino , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , NF-kappa B/genética , NF-kappa B/metabolismo , Fenótipo , Protocaderinas
7.
Biochem Biophys Res Commun ; 488(1): 189-195, 2017 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-28499869

RESUMO

Persistent or excess activation of NF-κB leads to cancer, autoimmune and inflammatory diseases. Therefore, activated NF-κB needs to be terminated after induction, which highlights the physiological significance of NF-κB-negative regulators. However, the molecular mechanisms that negatively regulate NF-κB are not well understood. Here, we report that Ring Finger Protein 8 (RNF8), an E3 ubiquitin ligase, inhibits TNFα-mediated NF-κB activation by targeting IκB kinase (IKK). Upon TNFα stimulation, RNF8 binds to the catalytic subunits of IKK complex, resulting in inhibition of IKKα/ß phosphorylation and subsequent NF-κB activation. RNF8 targets the IKK complex in a manner independent of its RING domain. We further provide evidence that the silencing of RNF8 results in enhanced TNFα-induced IKK activation, and an increase expression of NF-κB-induced inflammatory cytokine IL-8. Our study identifies a previously unrecognized role for RNF8 in the negative regulation of NF-κB activation by targeting and deactivating the IKK complex.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Quinase I-kappa B/metabolismo , Inflamação/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Linhagem Celular Tumoral , Células HEK293 , Humanos , Fator de Necrose Tumoral alfa/metabolismo , Ubiquitina-Proteína Ligases
8.
J Virol ; 89(1): 300-11, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25320310

RESUMO

UNLABELLED: Influenza A viruses (IAVs) rely on host factors to support their life cycle, as viral proteins hijack or interact with cellular proteins to execute their functions. Identification and understanding of these factors would increase our knowledge of the molecular mechanisms manipulated by the viruses. In this study, we searched for novel binding partners of the influenza A virus NS2 protein, the nuclear export protein responsible for overcoming host range restriction, by a yeast two-hybrid screening assay and glutathione S-transferase-pulldown and coimmunoprecipitation assays and identified AIMP2, a potent tumor suppressor that usually functions to regulate protein stability, as one of the major NS2-binding candidates. We found that the presence of NS2 protected AIMP2 from ubiquitin-mediated degradation in NS2-transfected cells and AIMP2 functioned as a positive regulator of IAV replication. Interestingly, AIMP2 had no significant effect on NS2 but enhanced the stability of the matrix protein M1. Further, we provide evidence that AIMP2 recruitment switches the modification of M1 from ubiquitination to SUMOylation, which occurs on the same attachment site (K242) on M1 and thereby promotes M1-mediated viral ribonucleoprotein complex nuclear export to increase viral replication. Collectively, our results reveal a new mechanism of AIMP2 mediation of influenza virus replication. IMPORTANCE: Although the ubiquitination of M1 during IAV infection has been observed, the precise modification site and the molecular consequences of this modification remain obscure. Here, we demonstrate for the first time that ubiquitin and SUMO compete for the same lysine (K242) on M1 and the interaction of NS2 with AIMP2 facilitates the switch of the M1 modification from ubiquitination to SUMOylation, thus increasing viral replication.


Assuntos
Vírus da Influenza A/fisiologia , Proteínas Nucleares/metabolismo , Sumoilação , Ubiquitinação , Proteínas da Matriz Viral/metabolismo , Proteínas não Estruturais Virais/metabolismo , Replicação Viral , Linhagem Celular , Centrifugação , Interações Hospedeiro-Patógeno , Humanos , Imunoprecipitação , Ligação Proteica , Mapeamento de Interação de Proteínas , Técnicas do Sistema de Duplo-Híbrido
9.
Cell Physiol Biochem ; 37(1): 243-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26302821

RESUMO

BACKGROUND/AIMS: MicroRNA (miRNA) is a small non-coding RNA molecule that functions in regulation of gene expression by targeting mRNA to affect its stability and/or translation. The aim of this study was to evaluate the miRNAs involvement in gestational diabetes mellitus (GDM), a well known risk factor for fetal overgrowth. METHODS: Differential microRNA expression in placental tissues of normal controls and women with GDM were identified by miRNA micorarray analysis and further confirmed by quantitative real-time PCR (qRT-PCR) on an independent set of normal and GDM placental tissues. Target genes of microRNAs were bioinformatically predicted and verified in vitro by Western blotting. RESULTS: Our results uncovered 9 miRNAs that were significantly deregulated in GDM samples: miR-508-3p was up-regulated and miR-27a, miR-9, miR-137, miR-92a, miR-33a, miR-30d, miR-362-5p and miR-502-5p were down-regulated. Bioinformatic approaches revealed that the microRNAs signature identifies gene targets involved in EGFR (epidermal growth factor receptor)-PI3K (phosphoinositide 3-Kinase)-Akt (also known as protein kinase B) pathway, a signal cascade which plays important roles in placental development and fetal growth. We found that the protein levels of EGFR, PI3K and phospho-Akt were up-regulated and PIKfyve (a FYVE finger-containing phosphoinositide kinase), a negative regulator of EGFR signaling, was down-regulated significantly in GDM tissues. We also confirmed PIKfyve was a direct target of miR-508-3p. CONCLUSION: Our data identified a miRNA signature involvement in GDM which may contribute to macrosomia through enhancing EGFR signaling.


Assuntos
Diabetes Gestacional/genética , Macrossomia Fetal/etiologia , Macrossomia Fetal/genética , MicroRNAs/genética , Adulto , Estudos de Casos e Controles , Regulação para Baixo/genética , Receptores ErbB/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Fosfatidilinositol 3-Quinases/genética , Placenta , Gravidez , Proteínas Proto-Oncogênicas c-akt , Risco , Transdução de Sinais/genética , Regulação para Cima/genética , Adulto Jovem
10.
J Virol ; 88(4): 1924-34, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24284319

RESUMO

DNA vaccines offer advantage over conventional vaccines, as they are safer to use, easier to produce, and able to induce humoral as well cellular immune responses. Unfortunately, no DNA vaccines have been licensed for human use for the difficulties in developing an efficient and safe in vivo gene delivery system. In vivo electroporation (EP)-based DNA delivery has attracted great attention for its potency to enhance cellular uptake of DNA vaccines and function as an adjuvant. Minicircle DNA (a new form of DNA containing only a gene expression cassette and lacking a backbone of bacterial plasmid DNA) is a powerful candidate of gene delivery in terms of improving the levels and the duration of transgene expression in vivo. In this study, as a novel vaccine delivery system, we combined in vivo EP and the minicircle DNA carrying a codon-optimized HIV-1 gag gene (minicircle-gag) to evaluate the immunogenicity of this system. We found that minicircle-gag conferred persistent and high levels of gag expression in vitro and in vivo. The use of EP delivery further increased minicircle-based gene expression. Moreover, when delivered by EP, minicircle-gag vaccination elicited a 2- to 3-fold increase in cellular immune response and a 1.5- to 3-fold augmentation of humoral immune responses compared with those elicited by a pVAX1-gag positive control. Increased immunogenicity of EP-assisted minicircle-gag may benefit from increasing local antigen expression, upregulating inflammatory genes, and recruiting immune cells. Collectively, in vivo EP of minicircle DNA functions as a novel vaccine platform that can enhance efficacy and immunogenicity of DNA vaccines.


Assuntos
DNA de Cinetoplasto/imunologia , Sistemas de Liberação de Medicamentos/métodos , Eletroporação/métodos , HIV-1/genética , Vacinas de DNA/administração & dosagem , Análise de Variância , Animais , Western Blotting , Ensaio de Imunoadsorção Enzimática , ELISPOT , Imunidade Humoral/genética , Imuno-Histoquímica , Luciferases , Camundongos , Camundongos Endogâmicos BALB C , Reação em Cadeia da Polimerase em Tempo Real , Produtos do Gene gag do Vírus da Imunodeficiência Humana/genética , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo
11.
J Immunol ; 191(5): 2614-23, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23918984

RESUMO

The intracellular RIG-I-like receptors recognize 5'-triphosphate viral genomic RNA and initiate the production of cytokines through mitochondria adaptor VISA. The regulation of this signal pathway is largely unknown. In this study, we report that the histone methyltransferase enhancer of zeste homolog 2 (EZH2) inhibits RIG-I signal pathway in an methyltransferase-independent manner. Knockdown EZH2 expression enhances VISA-induced activation of IFN-ß promoter and NF-κB signaling. Cytosolic distributed EZH2 colocalizes with VISA and binds to its caspase recruitment domain (CARD), thus blocking its association with RIG-I. During the infection of influenza A virus (IAV) strain A/WSN/33 (WSN), EZH2 translocates to RIG-I and continuously interferes the interaction between RIG-I and VISA. Both N and C termini of EZH2 interact with VISA and attenuate its downstream signaling. WSN virus infection-induced expression of TNF-α, IFN-ß, and IL-8 is inhibited by EZH2 and its catalytic dead form ΔSET. EZH2 overexpression facilitates the replications of IAV strains WSN and A/Puerto Rico/8/34 influenza virus. Knockdown EZH2 expression activates infection-induced IFN-ß transcription and inhibits virus replication. We further provided evidence to show that pharmacological disruption of EZH2 expression by its inhibitor 3-deazaneplanocin A activates innate immune responses and attenuates the replication of WSN virus in HeLa, MDCK, and mouse primary bone marrow-derived macrophages, but not in IFN-deficient Vero cells. Collectively, these results revealed that EZH2 binds to VISA and interferes with the interaction between VISA and RIG-I. Targeting EZH2 activates mitochondria-mediated antiviral innate immune responses, and thus represses the replication of IAV in cells.


Assuntos
Imunidade Inata/imunologia , Mitocôndrias/imunologia , Complexo Repressor Polycomb 2/imunologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Chlorocebus aethiops , Proteína Potenciadora do Homólogo 2 de Zeste , Citometria de Fluxo , Células HEK293 , Células HeLa , Humanos , Immunoblotting , Imunoprecipitação , Vírus da Influenza A , Proteínas de Membrana/imunologia , Camundongos , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/imunologia , Infecções por Orthomyxoviridae/imunologia , Interferência de RNA , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Superfície Celular , Células Vero
12.
Biochem Biophys Res Commun ; 446(2): 523-8, 2014 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-24613846

RESUMO

Mitochondrial antiviral-signaling protein (MAVS), as a critical adaptor of RIG-I signaling, bridges viral RNA recognition and downstream signal activation. However, the regulating mechanisms of MAVS are not well understood. In this study, we demonstrated that eukaryotic elongation factor 1B gamma (eEF1Bγ) activates NF-кB signaling pathway through targeting MAVS. GST-pull down and mass spectrometric analysis suggested that eEF1Bγ binds to the CARD domain of MAVS. The interaction and mitochondrial colocalization of eEF1Bγ and MAVS were further verified by co-immunoprecipitation (co-IP) and immunofluorescence microscopy assays. The dual-luciferase assays showed that ectopic expression of eEF1Bγ significantly promotes the activities of transcription factor NF-кB and promoters of downstream proinflammatory cytokines Interleukin-8 (IL-8) and Interleukin-6 (IL-6). eEF1Bγ increases the abundance of MAVS by promoting its K63-linked polyubiquitination and attenuating its K48-linked polyubiquitination. Besides, proline-rich (Pro) region and CARD domain of MAVS are indispensable for the process of eEF1Bγ mediated ubiquitination. Collectively, these results demonstrated that eEF1Bγ functions as a positive regulator of NF-кB signal by targeting MAVS for activation, which provides a new regulating mechanism of antiviral responses.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , NF-kappa B/metabolismo , Fator 1 de Elongação de Peptídeos/metabolismo , Transdução de Sinais/fisiologia , Células HEK293 , Células HeLa , Humanos , Regulação para Cima/fisiologia
13.
J Virol ; 87(6): 3039-52, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23283961

RESUMO

Influenza A virus is an important pathogenic virus known to induce host cell cycle arrest in G(0)/G(1) phase and create beneficial conditions for viral replication. However, how the virus achieves arrest remains unclear. We investigated the mechanisms underlying this process and found that the nonstructural protein 1 (NS1) is required. Based on this finding, we generated a viable influenza A virus (H1N1) lacking the entire NS1 gene to study the function of this protein in cell cycle regulation. In addition to some cell cycle regulators that were changed, the concentration and activity of RhoA protein, which is thought to be pivotal for G(1)/S phase transition, were also decreased with overexpressing NS1. And in the meantime, the phosphorylation level of cell cycle regulator pRb, downstream of RhoA kinase, was decreased in an NS1-dependent manner. These findings indicate that the NS1 protein induces G(0)/G(1) cell cycle arrest mainly through interfering with the RhoA/pRb signaling cascade, thus providing favorable conditions for viral protein accumulation and replication. We further investigated the NS1 protein of avian influenza virus (H5N1) and found that it can also decrease the expression and activity of RhoA, suggesting that the H5N1 virus may affect the cell cycle through the same mechanism. The NS1/RhoA/pRb cascade, which can induce the G(0)/G(1) cell cycle arrest identified here, provides a unified explanation for the seemingly different NS1 functions involved in viral replication events. Our findings shed light on the mechanism of influenza virus replication and open new avenues for understanding the interaction between pathogens and hosts.


Assuntos
Pontos de Checagem da Fase G1 do Ciclo Celular , Vírus da Influenza A Subtipo H1N1/patogenicidade , Virus da Influenza A Subtipo H5N1/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores , Animais , Linhagem Celular , Células Epiteliais/virologia , Humanos
14.
Int J Mol Sci ; 15(9): 16226-45, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25226533

RESUMO

Many dietary compounds are known to have health benefits owing to their antioxidative and anti-inflammatory properties. To determine the molecular mechanism of these food-derived compounds, we analyzed their effect on various genes related to cell apoptosis, DNA damage and repair, oxidation and inflammation using in vitro cell culture assays. This review further tests the hypothesis proposed previously that downstream products of COX-2 (cyclooxygenase-2) called electrophilic oxo-derivatives induce antioxidant responsive elements (ARE), which leads to cell proliferation under antioxidative conditions. Our findings support this hypothesis and show that cell proliferation was inhibited when COX-2 was down-regulated by polyphenols and polysaccharides. Flattened macrophage morphology was also observed following the induction of cytokine production by polysaccharides extracted from viili, a traditional Nordic fermented dairy product. Coix lacryma-jobi (coix) polysaccharides were found to reduce mitochondrial membrane potential and induce caspase-3- and 9-mediated apoptosis. In contrast, polyphenols from blueberries were involved in the ultraviolet-activated p53/Gadd45/MDM2 DNA repair system by restoring the cell membrane potential. Inhibition of hypoxia-inducible factor-1 by saponin extracts of ginsenoside (Ginsen) and Gynostemma and inhibition of S100A4 by coix polysaccharides inhibited cancer cell migration and invasion. These observations suggest that antioxidants and changes in cell membrane potential are the major driving forces that transfer signals through the cell membrane into the cytosol and nucleus, triggering gene expression, changes in cell proliferation and the induction of apoptosis or DNA repair.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Dieta , Movimento Celular/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Humanos
15.
J Clin Invest ; 134(12)2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38950288

RESUMO

Research advances over the past 30 years have confirmed a critical role for genetics in the etiology of dilated cardiomyopathies (DCMs). However, full knowledge of the genetic architecture of DCM remains incomplete. We identified candidate DCM causal gene, C10orf71, in a large family with 8 patients with DCM by whole-exome sequencing. Four loss-of-function variants of C10orf71 were subsequently identified in an additional group of492 patients with sporadic DCM from 2 independent cohorts. C10orf71 was found to be an intrinsically disordered protein specifically expressed in cardiomyocytes. C10orf71-KO mice had abnormal heart morphogenesis during embryonic development and cardiac dysfunction as adults with altered expression and splicing of contractile cardiac genes. C10orf71-null cardiomyocytes exhibited impaired contractile function with unaffected sarcomere structure. Cardiomyocytes and heart organoids derived from human induced pluripotent stem cells with C10orf71 frameshift variants also had contractile defects with normal electrophysiological activity. A rescue study using a cardiac myosin activator, omecamtiv mecarbil, restored contractile function in C10orf71-KO mice. These data support C10orf71 as a causal gene for DCM by contributing to the contractile function of cardiomyocytes. Mutation-specific pathophysiology may suggest therapeutic targets and more individualized therapy.


Assuntos
Cardiomiopatia Dilatada , Mutação da Fase de Leitura , Camundongos Knockout , Miócitos Cardíacos , Organoides , Humanos , Animais , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Organoides/metabolismo , Organoides/patologia , Masculino , Feminino , Contração Miocárdica/genética , Adulto , Modelos Animais de Doenças
16.
Cell Microbiol ; 14(12): 1849-66, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22891964

RESUMO

The IKK/NF-κB pathway is an essential signalling process initiated by the cell as a defence against viral infection like influenza virus. This pathway is therefore a prime target for viruses attempting to counteract the host response to infection. Here, we report that the influenza A virus NS1 protein specifically inhibits IKK-mediated NF-κB activation and production of the NF-κB induced antiviral genes by physically interacting with IKK through the C-terminal effector domain. The interaction between NS1 and IKKα/IKKß affects their phosphorylation function in both the cytoplasm and nucleus. In the cytoplasm, NS1 not only blocks IKKß-mediated phosphorylation and degradation of IκBα in the classical pathway but also suppresses IKKα-mediated processing of p100 to p52 in the alternative pathway, which leads to the inhibition of nuclear translocation of NF-κB and the subsequent expression of downstream NF-κB target genes. In the nucleus, NS1 impairs IKK-mediated phosphorylation of histone H3 Ser 10 that is critical to induce rapid expression of NF-κB target genes. These results reveal a new mechanism by which influenza A virus NS1 protein counteracts host NF-κB-mediated antiviral response through the disruption of IKK function. In this way, NS1 diminishes antiviral responses to infection and, in turn, enhances viral pathogenesis.


Assuntos
Quinase I-kappa B/metabolismo , Evasão da Resposta Imune , Imunidade Inata , Vírus da Influenza A/patogenicidade , Proteínas não Estruturais Virais/metabolismo , Fatores de Virulência/metabolismo , Animais , Linhagem Celular , Humanos , Quinase I-kappa B/antagonistas & inibidores , Vírus da Influenza A/imunologia , NF-kappa B/metabolismo , Fosforilação , Mapeamento de Interação de Proteínas , Processamento de Proteína Pós-Traducional , Transporte Proteico , Proteínas não Estruturais Virais/imunologia , Fatores de Virulência/imunologia
17.
Sheng Wu Gong Cheng Xue Bao ; 39(4): 1644-1654, 2023 Apr 25.
Artigo em Zh | MEDLINE | ID: mdl-37154329

RESUMO

To explore the effect of Mlk3 (mixed lineage kinase 3) deficiency on blood pressure, Mlk3 gene knockout (Mlk3KO) mice were generated. Activities of sgRNAs targeted Mlk3 gene were evaluated by T7 endonuclease I (T7E1) assay. CRISPR/Cas9 mRNA and sgRNA were obtained by in vitro transcription, microinjected into zygote, followed by transferring into a foster mother. Genotyping and DNA sequencing confirmed the deletion of Mlk3 gene. Real- time PCR (RT-PCR), Western blotting or immunofluorescence analysis showed that Mlk3KO mice had an undetectable expression of Mlk3 mRNA or Mlk3 protein. Mlk3KO mice exhibited an elevated systolic blood pressure compared with wild-type mice as measured by tail-cuff system. Immunohistochemistry and Western blotting analysis showed that the phosphorylation of MLC (myosin light chain) was significantly increased in aorta isolated from Mlk3KO mice. Together, Mlk3KO mice was successfully generated by CRISPR/Cas9 system. MLK3 functions in maintaining blood pressure homeostasis by regulating MLC phosphorylation. This study provides an animal model for exploring the mechanism by which Mlk3 protects against the development of hypertension and hypertensive cardiovascular remodeling.


Assuntos
Sistemas CRISPR-Cas , Zigoto , Animais , Camundongos , Camundongos Knockout , Pressão Sanguínea , Técnicas de Inativação de Genes
18.
Gene ; 878: 147582, 2023 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-37353041

RESUMO

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a life-threatening condition with an unfavorable prognosis. As the pathogenesis of ARDS remains unclear, we aimed to identify the core genes associated with ARDS and the mechanisms by which competing endogenous RNAs (ceRNAs) regulate the disease's progression. METHODS: Three mRNA microarray datasets (GSE17355, GSE48787, and GSE130936), derived from the Gene Expression Omnibus (GEO) database, were selected. Common differentially expressed genes (DEGs) related to acute lung injury (ALI) were identified and subjected to enrichment analysis. Then, hub genes were figured out through the protein-protein interaction (PPI) network and functional analysis, and targeted miRNAs and lncRNAs were predicted. Finally, the ceRNA networks associated with ALI were constructed and validated experimentally. RESULTS: A total of 155 upregulated and 93 downregulated DEGs were identified in the three datasets. The TNF signaling pathway and IL-17 signaling pathway were the most enriched pathways. Then, eleven DEGs enriched in the IL-17 signaling pathway were selected as the hub genes. Three miRNAs (mmu-mir-155-5p, mmu-mir-21a-5p, and mmu-mir-122-5p), which were located in the lung tissue and predicted to bind the hub genes at the same time, and two lncRNAs (Neat1 and Tug1), which have binding sites for the aforementioned miRNAs, were filtered. With qPCR verification, we identified a ceRNA network composed of NEAT1, miR-21-5p, MMP9, and CXCL5. NEAT1 knockdown promoted the migration and reduced the expression of pro-inflammatory factor and reactive oxygen species (ROS) in lung epithelial cells. We eventually confirmed that NEAT1/miR-21-5p/CXCL5/MMP9 played a pivotal role in regulating the inflammatory response in ALI. CONCLUSION: The IL-17 signaling pathway is of great importance in the pathogenesis of ARDS. NEAT1/miR-21-5p is involved in the inflammation of ALI by regulating CXCL5 and MMP9.


Assuntos
MicroRNAs , RNA Longo não Codificante , Síndrome do Desconforto Respiratório , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Metaloproteinase 9 da Matriz/genética , Interleucina-17/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Síndrome do Desconforto Respiratório/genética , Redes Reguladoras de Genes
19.
JACC Basic Transl Sci ; 8(11): 1457-1472, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-38093741

RESUMO

Palmd-deficient mice of advanced age manifest increased aortic valve peak velocity, thickened aortic valve leaflets, and excessive extracellular matrix deposition, which are key features of calcific aortic valve disease. PALMD is predominantly expressed in endothelial cells of aortic valves, and PALMD-silenced valvular endothelial cells are prone to oscillatory shear stress-induced endothelial-to-mesenchymal transition. Mechanistically, PALMD is associated with TNFAIP3 interaction protein 1, a binding protein of TNFAIP3 and IKBKG in NF-κB signaling. Loss of PALMD impairs TNFAIP3-dependent deubiquitinating activity and promotes the ubiquitination of IKBKG and subsequent NF-κB activation. Adeno-associated virus-mediated PALMD overexpression ameliorates aortic valvular remodeling in mice with calcific aortic valve disease, indicating protection.

20.
Signal Transduct Target Ther ; 8(1): 394, 2023 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-37828006

RESUMO

Immune cell infiltration in response to myocyte death regulates extracellular matrix remodeling and scar formation after myocardial infarction (MI). Caspase-recruitment domain family member 9 (CARD9) acts as an adapter that mediates the transduction of pro-inflammatory signaling cascades in innate immunity; however, its role in cardiac injury and repair post-MI remains unclear. We found that Card9 was one of the most upregulated Card genes in the ischemic myocardium of mice. CARD9 expression increased considerably 1 day post-MI and declined by day 7 post-MI. Moreover, CARD9 was mainly expressed in F4/80-positive macrophages. Card9 knockout (KO) led to left ventricular function improvement and infarct scar size reduction in mice 28 days post-MI. Additionally, Card9 KO suppressed cardiomyocyte apoptosis in the border region and attenuated matrix metalloproteinase (MMP) expression. RNA sequencing revealed that Card9 KO significantly suppressed lipocalin 2 (Lcn2) expression post-MI. Both LCN2 and the receptor solute carrier family 22 member 17 (SL22A17) were detected in macrophages. Subsequently, we demonstrated that Card9 overexpression increased LCN2 expression, while Card9 KO inhibited necrotic cell-induced LCN2 upregulation in macrophages, likely through NF-κB. Lcn2 KO showed beneficial effects post-MI, and recombinant LCN2 diminished the protective effects of Card9 KO in vivo. Lcn2 KO reduced MMP9 post-MI, and Lcn2 overexpression increased Mmp9 expression in macrophages. Slc22a17 knockdown in macrophages reduced MMP9 release with recombinant LCN2 treatment. In conclusion, our results demonstrate that macrophage CARD9 mediates the deterioration of cardiac function and adverse remodeling post-MI via LCN2.


Assuntos
Traumatismos Cardíacos , Infarto do Miocárdio , Animais , Camundongos , Proteínas Adaptadoras de Sinalização CARD , Lipocalina-2/genética , Macrófagos/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Infarto do Miocárdio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA