Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 299(6): 104777, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37142222

RESUMO

Mycobacterium tuberculosis (Mtb) WhiB3 is an iron-sulfur cluster-containing transcription factor belonging to a subclass of the WhiB-Like (Wbl) family that is widely distributed in the phylum Actinobacteria. WhiB3 plays a crucial role in the survival and pathogenesis of Mtb. It binds to the conserved region 4 of the principal sigma factor (σA4) in the RNA polymerase holoenzyme to regulate gene expression like other known Wbl proteins in Mtb. However, the structural basis of how WhiB3 coordinates with σA4 to bind DNA and regulate transcription is unclear. Here we determined crystal structures of the WhiB3:σA4 complex without and with DNA at 1.5 Å and 2.45 Å, respectively, to elucidate how WhiB3 interacts with DNA to regulate gene expression. These structures reveal that the WhiB3:σA4 complex shares a molecular interface similar to other structurally characterized Wbl proteins and also possesses a subclass-specific Arg-rich DNA-binding motif. We demonstrate that this newly defined Arg-rich motif is required for WhiB3 binding to DNA in vitro and transcriptional regulation in Mycobacterium smegmatis. Together, our study provides empirical evidence of how WhiB3 regulates gene expression in Mtb by partnering with σA4 and engaging with DNA via the subclass-specific structural motif, distinct from the modes of DNA interaction by WhiB1 and WhiB7.


Assuntos
Proteínas de Bactérias , Modelos Moleculares , Mycobacterium tuberculosis , Fatores de Transcrição , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Cristalografia por Raios X , RNA Polimerases Dirigidas por DNA/química , RNA Polimerases Dirigidas por DNA/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Mycobacterium tuberculosis/genética , Mycobacterium tuberculosis/metabolismo , Estrutura Quaternária de Proteína , Fator sigma/química , Fator sigma/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo
2.
J Biochem Mol Toxicol ; 38(1): e23556, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37867445

RESUMO

Fraxetin, a natural compound extracted from the Chinese herb Cortex Fraxini, is reported to boast extensive antitumor properties in various cancers. However, whether fraxetin exhibited an anticancer effect on bladder cancer remains unknown. In this study, cell counting kit-8 was utilized to detect cell viability. Flow cytometry analysis was performed for cell apoptosis analysis. Western blot analysis and real-time PCR were used to ascertain gene expression analysis. A mouse bladder cancer xenograft model was established and subjected to fraxetin treatment. Fraxetin reduced the viability of bladder cancer cells, induced apoptosis in vitro, and inhibited the growth of bladder cancer in vivo. Fraxetin inhibited the Akt pathway in J82 cells. In conclusion, the growth inhibitory properties of fraxetin against bladder cancer may be mediated via an Akt inhibitory effect and cell apoptosis promotion.


Assuntos
Cumarínicos , Proteínas Proto-Oncogênicas c-akt , Neoplasias da Bexiga Urinária , Camundongos , Animais , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proliferação de Células , Apoptose , Neoplasias da Bexiga Urinária/metabolismo , Linhagem Celular Tumoral
3.
J Nanobiotechnology ; 21(1): 352, 2023 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-37770932

RESUMO

BACKGROUND: Macrophages are highly plastic innate immune cells that play key roles in host defense, tissue repair, and homeostasis maintenance. In response to divergent stimuli, macrophages rapidly alter their functions and manifest a wide polarization spectrum with two extremes: M1 or classical activation and M2 or alternative activation. Extracellular vesicles (EVs) secreted from differentially activated macrophages have been shown to have diverse functions, which are primarily attributed to their microRNA cargos. The role of protein cargos in these EVs remains largely unexplored. Therefore, in this study, we focused on the protein cargos in macrophage-derived EVs. RESULTS: Naïve murine bone marrow-derived macrophages were treated with lipopolysaccharide or interlukin-4 to induce M1 or M2 macrophages, respectively. The proteins of EVs and their parental macrophages were subjected to quantitative proteomics analyses, followed by bioinformatic analyses. The enriched proteins of M1-EVs were involved in proinflammatory pathways and those of M2-EVs were associated with immunomodulation and tissue remodeling. The signature proteins of EVs shared a limited subset of the proteins of their respective progenitor macrophages, but they covered many of the typical pathways and functions of their parental cells, suggesting their respective M1-like and M2-like phenotypes and functions. Experimental examination validated that protein cargos in M1- or M2-EVs induced M1 or M2 polarization, respectively. More importantly, proteins in M1-EVs promoted viability, proliferation, and activation of T lymphocytes, whereas proteins in M2-EVs potently protected the tight junction structure and barrier integrity of epithelial cells from disruption. Intravenous administration of M2-EVs in colitis mice led to their accumulation in the colon, alleviation of colonic inflammation, promotion of M2 macrophage polarization, and improvement of gut barrier functions. Protein cargos in M2-EVs played a key role in their protective function in colitis. CONCLUSION: This study has yielded a comprehensive unbiased dataset of protein cargos in macrophage-derived EVs, provided a systemic view of their potential functions, and highlighted the important engagement of protein cargos in the pathophysiological functions of these EVs.


Assuntos
Colite , Vesículas Extracelulares , Animais , Camundongos , Macrófagos/metabolismo , Fagocitose , Vesículas Extracelulares/metabolismo , Colite/metabolismo , Inflamação/metabolismo
4.
Arterioscler Thromb Vasc Biol ; 41(4): 1487-1503, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33567868
5.
Acta Pharmacol Sin ; 42(12): 2046-2057, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33623121

RESUMO

Nuclear factor kappa B (NF-κB) activation contributes to many vascular inflammatory diseases. The present study tested the hypothesis that microRNA-17-3p (miR-17-3p) suppresses the pro-inflammatory responses via NF-κB signaling in vascular endothelium. Human umbilical vein endothelial cells (HUVECs), transfected with or without miR-17-3p agomir/antagomir, were exposed to lipopolysaccharide (LPS), and the inflammatory responses were determined. The cellular target of miR-17-3p was examined with dual-luciferase reporter assay. Mice were treated with miR-17-3p agomir and the degree of LPS-induced inflammation was determined. In HUVECs, LPS caused upregulation of miR-17-3p. Overexpression of miR-17-3p in HUVECs inhibited NIK and IKKß binding protein (NIBP) protein expression and suppressed LPS-induced phosphorylation of inhibitor of kappa Bα (IκBα) and NF-κB-p65. The reduced NF-κB activity was paralleled by decreased protein levels of NF-κB-target gene products including pro-inflammatory cytokine [interleukin 6], chemokines [interleukin 8 and monocyte chemoattractant protein-1] and adhesion molecules [vascular cell adhesion molecule-1, intercellular adhesion molecule-1 and E-selectin]. Immunostaining revealed that overexpression of miR-17-3p reduced monocyte adhesion to LPS-stimulated endothelial cells. Inhibition of miR-17-3p with antagomir has the opposite effect on LPS-induced inflammatory responses in HUVECs. The anti-inflammatory effect of miR-17-3p was mimicked by NIBP knockdown. In mice treated with LPS, miR-17-3p expression was significantly increased. Systemic administration of miR-17-3p for 3 days suppressed LPS-induced NF-κB activation and monocyte adhesion to endothelium in lung tissues of the mice. In conclusion, miR-17-3p inhibits LPS-induced NF-κB activation in HUVECs by targeting NIBP. The findings therefore suggest that miR-17-3p is a potential therapeutic target/agent in the management of vascular inflammatory diseases.


Assuntos
Endotélio Vascular/metabolismo , Quinase I-kappa B/metabolismo , Inflamação/metabolismo , MicroRNAs/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição RelA/metabolismo , Animais , Antagomirs/farmacologia , Moléculas de Adesão Celular/metabolismo , Citocinas/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/induzido quimicamente , Lipopolissacarídeos , Masculino , Camundongos , Inibidor de NF-kappaB alfa/metabolismo , Transdução de Sinais/fisiologia , Regulação para Cima/fisiologia , Quinase Induzida por NF-kappaB
6.
Nucleic Acids Res ; 47(3): 1505-1522, 2019 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-30476192

RESUMO

The role of long non-coding RNAs (lncRNAs) in regulating endothelial function through the DNA damage response (DDR) remains poorly understood. In this study, we demonstrate that lncRNA maternally expressed gene 3 (Meg3) interacts with the RNA binding protein polypyrimidine tract binding protein 3 (PTBP3) to regulate gene expression and endothelial function through p53 signaling ─ a major coordinator of apoptosis and cell proliferation triggered by the DDR. Meg3 expression is induced in endothelial cells (ECs) upon p53 activation. Meg3 silencing induces DNA damage, activates p53 signaling, increases the expression of p53 target genes, promotes EC apoptosis, and inhibits EC proliferation. Mechanistically, Meg3 silencing reduces the interaction of p53 with Mdm2, induces p53 expression, and promotes the association of p53 with the promoters of a subset of p53 target genes. PTBP3 silencing recapitulates the effects of Meg3 deficiency on the expression of p53 target genes, EC apoptosis and proliferation. The Meg3-dependent association of PTBP3 with the promoters of p53 target genes suggests that Meg3 and PTBP3 restrain p53 activation. Our studies reveal a novel role of Meg3 and PTBP3 in regulating p53 signaling and endothelial function, which may serve as novel targets for therapies to restore endothelial homeostasis.


Assuntos
Neoplasias/genética , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , RNA Longo não Codificante/genética , Proteína Supressora de Tumor p53/genética , Apoptose/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Dano ao DNA/genética , Metilação de DNA/genética , Reparo do DNA/genética , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/patologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Transdução de Sinais
7.
Am J Physiol Cell Physiol ; 318(6): C1200-C1213, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32374676

RESUMO

The liver is the central metabolic hub for carbohydrate, lipid, and protein metabolism. It is composed of four major types of cells, including hepatocytes, endothelial cells (ECs), Kupffer cells, and stellate cells. Hepatic ECs are highly heterogeneous in both mice and humans, representing the second largest population of cells in liver. The majority of them line hepatic sinusoids known as liver sinusoidal ECs (LSECs). The structure and biology of LSECs and their roles in physiology and liver disease were reviewed recently. Here, we do not give a comprehensive review of LSEC structure, function, or pathophysiology. Instead, we focus on the recent progress in LSEC research and other hepatic ECs in physiology and nonalcoholic fatty liver disease and other hepatic fibrosis-related conditions. We discuss several current areas of interest, including capillarization, scavenger function, autophagy, cellular senescence, paracrine effects, and mechanotransduction. In addition, we summarize the strengths and weaknesses of evidence for the potential role of endothelial-to-mesenchymal transition in liver fibrosis.


Assuntos
Capilares/metabolismo , Células Endoteliais/metabolismo , Cirrose Hepática/metabolismo , Fígado/irrigação sanguínea , Hepatopatia Gordurosa não Alcoólica/metabolismo , Animais , Autofagia , Capilares/patologia , Diferenciação Celular , Proliferação de Células , Senescência Celular , Células Endoteliais/patologia , Transição Epitelial-Mesenquimal , Humanos , Mediadores da Inflamação/metabolismo , Cirrose Hepática/patologia , Mecanotransdução Celular , Hepatopatia Gordurosa não Alcoólica/patologia , Comunicação Parácrina , Espécies Reativas de Oxigênio/metabolismo
8.
Int J Mol Sci ; 21(17)2020 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-32854424

RESUMO

Selective autolysosomal degradation of damaged mitochondria, also called mitophagy, is an indispensable process for maintaining integrity and homeostasis of mitochondria. One well-established mechanism mediating selective removal of mitochondria under relatively mild mitochondria-depolarizing stress is PINK1-Parkin-mediated or ubiquitin-dependent mitophagy. However, additional mechanisms such as LC3-mediated or ubiquitin-independent mitophagy induction by heavy environmental stress exist and remain poorly understood. The present study unravels a novel role of stress-inducible protein Sestrin2 in degradation of mitochondria damaged by transition metal stress. By utilizing proteomic methods and studies in cell culture and rodent models, we identify autophagy kinase ULK1-mediated phosphorylation sites of Sestrin2 and demonstrate Sestrin2 association with mitochondria adaptor proteins in HEK293 cells. We show that Ser-73 and Ser-254 residues of Sestrin2 are phosphorylated by ULK1, and a pool of Sestrin2 is strongly associated with mitochondrial ATP5A in response to Cu-induced oxidative stress. Subsequently, this interaction promotes association with LC3-coated autolysosomes to induce degradation of mitochondria damaged by Cu-induced ROS. Treatment of cells with antioxidants or a Cu chelator significantly reduces Sestrin2 association with mitochondria. These results highlight the ULK1-Sestrin2 pathway as a novel stress-sensing mechanism that can rapidly induce autophagic degradation of mitochondria under severe heavy metal stress.


Assuntos
Proteína Homóloga à Proteína-1 Relacionada à Autofagia/metabolismo , Cobre/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/fisiologia , Proteínas Nucleares/metabolismo , Proteômica/métodos , Autofagia , Sítios de Ligação , Células HEK293 , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Mitofagia , Proteínas Nucleares/química , Estresse Oxidativo , Fosforilação , Transdução de Sinais/efeitos dos fármacos
9.
Biochem Biophys Res Commun ; 503(2): 1070-1074, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-29936178

RESUMO

We previously reported that an orally administered dipeptide, Arg-Phe (RF), which causes enteroendocrine cell responses, lowered blood pressure in spontaneously hypertensive rats (SHRs). In this study, we found that Phe-Trp (FW), induced the most potent enteroendocrine cell responses out of total 338 dipeptides. An FW analogue, Phe-Trp-Gly-Lys (FWGK), which was effectively produced by tryptic digestion of bovine serum albumin, decreased blood pressure after oral administration. The minimum effective dose of FWGK (50 µg/kg) was 1/300 of that of RF (15 mg/kg). FWGK stimulated cholecystokinin (CCK) secretion in the enteroendocrine cells and exhibited vasorelaxing and antihypertensive effects via the CCK1 system.


Assuntos
Anti-Hipertensivos/farmacologia , Dipeptídeos/farmacologia , Células Enteroendócrinas/efeitos dos fármacos , Vasodilatadores/farmacologia , Administração Oral , Animais , Anti-Hipertensivos/administração & dosagem , Anti-Hipertensivos/química , Pressão Sanguínea/efeitos dos fármacos , Linhagem Celular , Colecistocinina/metabolismo , Dipeptídeos/administração & dosagem , Dipeptídeos/química , Células Enteroendócrinas/metabolismo , Masculino , Camundongos , Ratos Endogâmicos SHR , Vasodilatadores/administração & dosagem , Vasodilatadores/química
10.
Circ Res ; 118(5): 810-21, 2016 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-26830849

RESUMO

RATIONALE: The pathogenesis of insulin resistance involves dysregulated gene expression and function in multiple cell types, including endothelial cells (ECs). Post-transcriptional mechanisms such as microRNA-mediated regulation of gene expression could affect insulin action by modulating EC function. OBJECTIVE: To determine whether microRNA-181b (miR-181b) affects the pathogenesis of insulin resistance by regulating EC function in white adipose tissue during obesity. METHODS AND RESULTS: MiR-181b expression was reduced in adipose tissue ECs of obese mice, and rescue of miR-181b expression improved glucose homeostasis and insulin sensitivity. Systemic intravenous delivery of miR-181b robustly accumulated in adipose tissue ECs, enhanced insulin-mediated Akt phosphorylation at Ser473, and reduced endothelial dysfunction, an effect that shifted macrophage polarization toward an M2 anti-inflammatory phenotype in epididymal white adipose tissue. These effects were associated with increased endothelial nitric oxide synthase and FoxO1 phosphorylation as well as nitric oxide activity in epididymal white adipose tissue. In contrast, miR-181b did not affect insulin-stimulated Akt phosphorylation in liver and skeletal muscle. Bioinformatics and gene profiling approaches revealed that Pleckstrin homology domain leucine-rich repeat protein phosphatase, a phosphatase that dephosphorylates Akt at Ser473, is a novel target of miR-181b. Knockdown of Pleckstrin homology domain leucine-rich repeat protein phosphatase increased Akt phosphorylation at Ser473 in ECs, and phenocopied miR-181b's effects on glucose homeostasis, insulin sensitivity, and inflammation of epididymal white adipose tissue in vivo. Finally, ECs from diabetic subjects exhibited increased Pleckstrin homology domain leucine-rich repeat protein phosphatase expression. CONCLUSIONS: Our data underscore the importance of adipose tissue EC function in controlling the development of insulin resistance. Delivery of miR-181b or Pleckstrin homology domain leucine-rich repeat protein phosphatase inhibitors may represent a new therapeutic approach to ameliorate insulin resistance by improving adipose tissue endothelial Akt-endothelial nitric oxide synthase-nitric oxide signaling.


Assuntos
Tecido Adiposo Branco/metabolismo , Glicemia/metabolismo , Células Endoteliais/metabolismo , Homeostase/fisiologia , Resistência à Insulina/fisiologia , MicroRNAs/biossíntese , Animais , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos
11.
J Nanosci Nanotechnol ; 18(5): 3427-3432, 2018 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-29442848

RESUMO

In this work, partially exfoliated multi-walled carbon nanotubes (Px-CNTs) were prepared by oxidizing multi-walled carbon nanotubes (MWCNTs) and applied into water-based lubricant as a kind of new additives, resulting in an outstanding anti-friction effect. The Px-CNTs have the structures of both MWCNTs and graphene oxide nanoribbons (GONRs). The special structure could prevent aggregation in water-based lubricant and reduce friction effectively. At the same time, Px-CNTs generate both sliding and rolling friction like MWCNTs and GONRs simultaneously. The friction force of Px-CNTs tended to go up after declining with increasing its loading, suggesting the existence of optimum additive amount of additions. Compared with water, water with 0.5 wt% Px-CNTs further reduced the friction force up to 66.4%. Compared with CNTs-COOH and GONRs dispersed in water via a similar method, Px-CNTs in water displays remarkable friction characteristic, suggesting that the friction force of water with 0.5 wt% Px-CNTs is decreased by 19.82% and 13.82% compared with water with 0.3 wt% MWCNTs and GONRs.

12.
Biochem J ; 474(17): 2925-2935, 2017 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-28801479

RESUMO

Long non-coding RNAs (lncRNAs) are emerging as critical regulators of various biological processes and human diseases. The mechanisms of action involve their interactions with proteins, RNA and genomic DNA. Most lncRNAs display strong nuclear localization. Heterogeneous nuclear ribonucleoproteins (hnRNPs) are a large family of RNA-binding proteins that are important for multiple aspects of nucleic acid metabolism. hnRNPs are also predominantly expressed in the nucleus. This review discusses the interactions of lncRNAs and hnRNPs in regulating gene expression at transcriptional and post-transcriptional levels or by changing genomic structure, highlighting their involvements in glucose and lipid metabolism, immune response, DNA damage response, and other cellular functions. Toward the end, several techniques that are used to identify lncRNA binding partners are summarized. There are still many questions that need to be answered in this relatively new research area, which might provide novel targets to control the biological outputs of cells in response to different stimuli.


Assuntos
Núcleo Celular/metabolismo , Regulação da Expressão Gênica/fisiologia , Ribonucleoproteínas Nucleares Heterogêneas/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Núcleo Celular/genética , Dano ao DNA , Glucose/genética , Glucose/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas/genética , Humanos , Metabolismo dos Lipídeos/fisiologia , RNA Longo não Codificante/genética
13.
FASEB J ; 30(9): 3216-26, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27297585

RESUMO

Thrombogenic and inflammatory mediators, such as thrombin, induce NF-κB-mediated endothelial cell (EC) activation and dysfunction, which contribute to pathogenesis of arterial thrombosis. The role of anti-inflammatory microRNA-181b (miR-181b) on thrombosis remains unknown. Our previous study demonstrated that miR-181b inhibits downstream NF-κB signaling in response to TNF-α. Here, we demonstrate that miR-181b uniquely inhibits upstream NF-κB signaling in response to thrombin. Overexpression of miR-181b inhibited thrombin-induced activation of NF-κB signaling, demonstrated by reduction of phospho-IKK-ß, -IκB-α, and p65 nuclear translocation in ECs. MiR-181b also reduced expression of NF-κB target genes VCAM-1, intercellular adhesion molecule-1, E-selectin, and tissue factor. Mechanistically, miR-181b targets caspase recruitment domain family member 10 (Card10), an adaptor protein that participates in activation of the IKK complex in response to signals transduced from protease-activated receptor-1. miR-181b reduced expression of Card10 mRNA and protein, but not protease-activated receptor-1. 3'-Untranslated region reporter assays, argonaute-2 microribonucleoprotein immunoprecipitation studies, and Card10 rescue studies revealed that Card10 is a bona fide direct miR-181b target. Small interfering RNA-mediated knockdown of Card10 expression phenocopied effects of miR-181b on NF-κB signaling and targets. Card10 deficiency did not affect TNF-α-induced activation of NF-κB signaling, which suggested stimulus-specific regulation of NF-κB signaling and endothelial responses by miR-181b in ECs. Finally, in response to photochemical injury-induced arterial thrombosis, systemic delivery of miR-181b reduced thrombus formation by 73% in carotid arteries and prolonged time to occlusion by 1.6-fold, effects recapitulated by Card10 small interfering RNA. These data demonstrate that miR-181b and Card10 are important regulators of thrombin-induced EC activation and arterial thrombosis. These studies highlight the relevance of microRNA-dependent targets in response to ligand-specific signaling in ECs.-Lin, J., He, S., Sun, X., Franck, G., Deng, Y., Yang, D., Haemmig, S., Wara, A. K. M., Icli, B., Li, D., Feinberg, M. W. MicroRNA-181b inhibits thrombin-mediated endothelial activation and arterial thrombosis by targeting caspase recruitment domain family member 10.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , MicroRNAs/metabolismo , Trombina/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Células Endoteliais , Endotélio Vascular , Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Inflamação/metabolismo , Camundongos , MicroRNAs/genética , NF-kappa B/genética , NF-kappa B/metabolismo , Fosforilação , Interferência de RNA , Transdução de Sinais/fisiologia , Síndrome do Desfiladeiro Torácico , Trombina/genética , Trombose/etiologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
14.
J Mol Cell Cardiol ; 91: 151-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26776318

RESUMO

Wound healing is a physiological reparative response to injury and a well-orchestrated process that involves hemostasis, cellular migration, proliferation, angiogenesis, extracellular matrix deposition, and wound contraction and re-epithelialization. However, patients with type 2 diabetes mellitus (T2D) are frequently afflicted with impaired wound healing that progresses into chronic wounds or diabetic ulcers, and may lead to complications including limb amputation. Herein, we investigate the potential role of microRNA-26a (miR-26a) in a diabetic model of wound healing. Expression of miR-26a is rapidly induced in response to high glucose in endothelial cells (ECs). Punch skin biopsy wounding of db/db mice revealed increased expression of miR-26a (~3.5-fold) four days post-wounding compared to that of WT mice. Local administration of a miR-26a inhibitor, LNA-anti-miR-26a, induced angiogenesis (up to ~80%), increased granulation tissue thickness (by 2.5-fold) and accelerated wound closure (53% after nine days) compared to scrambled anti-miR controls in db/db mice. These effects were independent of altered M1/M2 macrophage ratios. Mechanistically, inhibition of miR-26a increased its target gene SMAD1 in ECs nine days post-wounding of diabetic mice. In addition, high glucose reduced activity of the SMAD1-3'-UTR. Diabetic dermal wounds treated with LNA-anti-miR-26a had increased expression of ID1, a downstream modulator or SMAD1, and decreased expression of the cell cycle inhibitor p27. These findings establish miR-26a as an important regulator on the progression of skin wounds of diabetic mice by specifically regulating the angiogenic response after injury, and demonstrate that neutralization of miR-26a may serve as a novel approach for therapy.


Assuntos
Diabetes Mellitus Experimental/genética , MicroRNAs/genética , Neovascularização Patológica/genética , Oligonucleotídeos Antissenso/genética , Proteína Smad1/genética , Ferimentos não Penetrantes/genética , Animais , Movimento Celular , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Derme/efeitos dos fármacos , Derme/metabolismo , Derme/patologia , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica , Glucose/farmacologia , Humanos , Proteína 1 Inibidora de Diferenciação/genética , Proteína 1 Inibidora de Diferenciação/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Mastócitos/metabolismo , Mastócitos/patologia , Camundongos , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Neovascularização Patológica/terapia , Oligonucleotídeos Antissenso/metabolismo , Reepitelização , Transdução de Sinais , Proteína Smad1/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ferimentos não Penetrantes/metabolismo , Ferimentos não Penetrantes/patologia , Ferimentos não Penetrantes/terapia
15.
Circ Res ; 114(1): 32-40, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24084690

RESUMO

RATIONALE: Activated nuclear factor (NF)-κB signaling in the vascular endothelium promotes the initiation and progression of atherosclerosis. Targeting endothelial NF-κB may provide a novel strategy to limit chronic inflammation. OBJECTIVE: To examine the role of microRNA-181b (miR-181b) in endothelial NF-κB signaling and effects on atherosclerosis. METHODS AND RESULTS: MiR-181b expression was reduced in the aortic intima and plasma in apolipoprotein E-deficient mice fed a high-fat diet. Correspondingly, circulating miR-181b in the plasma was markedly reduced in human subjects with coronary artery disease. Systemic delivery of miR-181b resulted in a 2.3-fold overexpression of miR-181b in the aortic intima of apolipoprotein E-deficient mice and suppressed NF-κB signaling revealed by bioluminescence imaging and reduced target gene expression in the aortic arch in apolipoprotein E-deficient/NF-κB-luciferase transgenic mice. MiR-181b significantly inhibited atherosclerotic lesion formation, proinflammatory gene expression and the influx of lesional macrophages and CD4+ T cells in the vessel wall. Mechanistically, miR-181b inhibited the expression of the target gene importin-α3, an effect that reduced NF-κB nuclear translocation specifically in the vascular endothelium of lesions, whereas surprisingly leukocyte NF-κB signaling was unaffected despite a 7-fold overexpression of miR-181b. Our findings uncover that NF-κB nuclear translocation in leukocytes does not involve importin-α3, but rather importin-α5, which miR-181b does not target, highlighting that inhibition of NF-κB signaling in the endothelium is sufficient to mediate miR-181b's protective effects. CONCLUSIONS: Systemic delivery of miR-181b inhibits the activation of NF-κB and atherosclerosis through cell-specific mechanisms in the vascular endothelium. These findings support the rationale that delivery of miR-181b may provide a novel therapeutic approach to treat chronic inflammatory diseases such as atherosclerosis.


Assuntos
Aorta/metabolismo , Aterosclerose/terapia , MicroRNAs/uso terapêutico , NF-kappa B/metabolismo , Túnica Íntima/metabolismo , Animais , Aorta/patologia , Apolipoproteínas E/genética , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Linfócitos T CD4-Positivos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Inflamação/metabolismo , Carioferinas/genética , Carioferinas/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/administração & dosagem , MicroRNAs/sangue , MicroRNAs/metabolismo , NF-kappa B/antagonistas & inibidores , Túnica Íntima/patologia
16.
Circ Res ; 113(11): 1231-41, 2013 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-24047927

RESUMO

RATIONALE: The rapid induction and orchestration of new blood vessels are critical for tissue repair in response to injury, such as myocardial infarction, and for physiological angiogenic responses, such as embryonic development and exercise. OBJECTIVE: We aimed to identify and characterize microRNAs (miR) that regulate pathological and physiological angiogenesis. METHODS AND RESULTS: We show that miR-26a regulates pathological and physiological angiogenesis by targeting endothelial cell (EC) bone morphogenic protein/SMAD1 signaling in vitro and in vivo. MiR-26a expression is increased in a model of acute myocardial infarction in mice and in human subjects with acute coronary syndromes. Ectopic expression of miR-26a markedly induced EC cycle arrest and inhibited EC migration, sprouting angiogenesis, and network tube formation in matrigel, whereas blockade of miR-26a had the opposite effects. Mechanistic studies demonstrate that miR-26a inhibits the bone morphogenic protein/SMAD1 signaling pathway in ECs by binding to the SMAD1 3'-untranslated region, an effect that decreased expression of Id1 and increased p21(WAF/CIP) and p27. In zebrafish, miR-26a overexpression inhibited formation of the caudal vein plexus, a bone morphogenic protein-responsive process, an effect rescued by ectopic SMAD1 expression. In mice, miR-26a overexpression inhibited EC SMAD1 expression and exercise-induced angiogenesis. Furthermore, systemic intravenous administration of an miR-26a inhibitor, locked nucleic acid-anti-miR-26a, increased SMAD1 expression and rapidly induced robust angiogenesis within 2 days, an effect associated with reduced myocardial infarct size and improved heart function. CONCLUSIONS: These findings establish miR-26a as a regulator of bone morphogenic protein/SMAD1-mediated EC angiogenic responses, and that manipulating miR-26a expression could provide a new target for rapid angiogenic therapy in ischemic disease states.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , MicroRNAs/fisiologia , Neovascularização Patológica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Proteína Smad1/fisiologia , Síndrome Coronariana Aguda/sangue , Síndrome Coronariana Aguda/patologia , Síndrome Coronariana Aguda/fisiopatologia , Animais , Biomarcadores/sangue , Proliferação de Células , Modelos Animais de Doenças , Desenvolvimento Embrionário/fisiologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiologia , Endotélio Vascular/fisiopatologia , Humanos , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/sangue , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Disfunção Ventricular Esquerda/fisiopatologia , Função Ventricular Esquerda/fisiologia , Peixe-Zebra
17.
J BUON ; 20(2): 505-13, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26011343

RESUMO

PURPOSE: To observe the efficacy and side effects of adjuvant dendritic cells' (DCs) vaccine combined with cytokine-induced killer cell (CIK) therapy after renal cell carcinoma (RCC) surgery (RCCS). METHODS: DCs vaccine and CIK that loaded the autologous tumor cell lysate were prepared in vitro. Four hundred and ten RCC patients were recruited, and the study group was given DCs-CIK immunotherapy, while the control group was given IFN-α therapy. RESULTS: Disease progression (recurrence, metastasis or death) showed significant differences between the two groups in clinical stage I and II patients, as well as in highly and moderately differentiated disease (p<0.05), while there was no significant difference between the two groups in patients with poorly differentiated disease (p>0.05). The 3- and 5-year overall survival rates of the DCs-CIK group (96% and 96%, respectively) exhibited significant difference compared to the IFN-α group (83% and 74%, respectively (p<0.01). Progression-free survival (PFS) between the two groups was significantly different (p<0.01). Tumor stage and DCs-CIK treatment were independent factors concerning prognosis of RCC (p<0.05). There was no severe toxicity observed in the DCs-CIK treatment group. CONCLUSIONS: Adjuvant post-RCCS DCs-CIK treatment prolonged PFS and reduced mortality, showing better overall activity compared to interferon treatment.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma de Células Renais/terapia , Células Matadoras Induzidas por Citocinas/imunologia , Células Dendríticas/imunologia , Neoplasias Renais/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Vacinas Anticâncer/efeitos adversos , Carcinoma de Células Renais/mortalidade , Terapia Combinada , Progressão da Doença , Feminino , Humanos , Interferon-alfa/efeitos adversos , Interferon-alfa/uso terapêutico , Neoplasias Renais/mortalidade , Masculino , Pessoa de Meia-Idade
18.
Arterioscler Thromb Vasc Biol ; 33(7): 1552-60, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23685559

RESUMO

OBJECTIVE: The objective of this study was to investigate the role of Kruppel-like factor (KLF) 10, a zinc-finger transcription factor, in bone marrow (BM)-derived cell responses to arterial endothelial injury. Accumulating evidence indicates that BM-derived progenitors are recruited to sites of vascular injury and contribute to endothelial repair. APPROACH AND RESULTS: In response to carotid artery endothelial denudation, KLF10 mRNA expression was markedly increased in both BM and circulating lin(-) progenitor cells. To examine the specific role of KLF10 in arterial reendothelialization, we used 2 models of endothelial denudation (wire- and thermal-induced injury) of the carotid artery in wild-type (WT) and KLF10(-/-) mice. WT mice displayed higher areas of reendothelialization compared with KLF10(-/-) mice after endothelial injury using either method. BM transplant studies revealed that reconstitution of KLF10(-/-) mice with WT BM fully rescued the defect in reendothelialization and increased lin(-)CD34(+)kinase insert domain receptor(+) progenitors in the blood and injured carotid arteries. Conversely, reconstitution of WT mice with KLF10(-/-) BM recapitulated the defects in reendothelialization and peripheral cell progenitors. The media from cultured KLF10(-)/(-) BM progenitors was markedly inefficient in promoting endothelial cell growth and migration compared with the media from WT progenitors, indicative of defective paracrine trophic effects from KLF10(-)/(-) BM progenitors. Finally, BM-derived KLF10(-/-) lin(-) progenitors from reconstituted mice had reduced CXC-chemokine receptor 4 expression and impaired migratory responses. CONCLUSIONS: Collectively, these observations demonstrate a protective role for BM-derived KLF10 in paracrine and homing responses important for arterial endothelial injury and highlight KLF10 as a possible therapeutic target to promote endothelial repair in vascular disease states.


Assuntos
Células da Medula Óssea/metabolismo , Lesões das Artérias Carótidas/metabolismo , Proliferação de Células , Fatores de Transcrição de Resposta de Crescimento Precoce/metabolismo , Células Endoteliais/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Comunicação Parácrina , Células-Tronco/metabolismo , Lesões do Sistema Vascular/metabolismo , Animais , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Transplante de Medula Óssea , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Quimiotaxia , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Fatores de Transcrição de Resposta de Crescimento Precoce/deficiência , Fatores de Transcrição de Resposta de Crescimento Precoce/genética , Células Endoteliais/patologia , Feminino , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptores CCR4/metabolismo , Transdução de Sinais , Transplante de Células-Tronco , Fatores de Tempo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia
19.
Cells ; 13(3)2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38334627

RESUMO

Heart development is a spatiotemporally regulated process that extends from the embryonic phase to postnatal stages. Disruption of this highly orchestrated process can lead to congenital heart disease or predispose the heart to cardiomyopathy or heart failure. Consequently, gaining an in-depth understanding of the molecular mechanisms governing cardiac development holds considerable promise for the development of innovative therapies for various cardiac ailments. While significant progress in uncovering novel transcriptional and epigenetic regulators of heart development has been made, the exploration of post-translational mechanisms that influence this process has lagged. Culling-RING E3 ubiquitin ligases (CRLs), the largest family of ubiquitin ligases, control the ubiquitination and degradation of ~20% of intracellular proteins. Emerging evidence has uncovered the critical roles of CRLs in the regulation of a wide range of cellular, physiological, and pathological processes. In this review, we summarize current findings on the versatile regulation of cardiac morphogenesis and maturation by CRLs and present future perspectives to advance our comprehensive understanding of how CRLs govern cardiac developmental processes.


Assuntos
Proteínas Culina , Ubiquitina , Ubiquitina/metabolismo , Proteínas Culina/metabolismo , Ubiquitinação , Ubiquitina-Proteína Ligases/metabolismo
20.
Atherosclerosis ; 392: 117506, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38518516

RESUMO

BACKGROUND AND AIMS: Long noncoding RNAs are involved in the pathogenesis of atherosclerosis. As long noncoding RNAs maternally expressed gene 3 (Meg3) prevents cellular senescence of hepatic vascular endothelium and obesity-induced insulin resistance, we decided to examine its role in cellular senescence and atherosclerosis. METHODS AND RESULTS: By analyzing our data and human and mouse data from the Gene Expression Omnibus database, we found that Meg3 expression was reduced in humans and mice with cardiovascular disease, indicating its potential role in atherosclerosis. In Ldlr-/- mice fed a Western diet for 12 weeks, Meg3 silencing by chemically modified antisense oligonucleotides attenuated the formation of atherosclerotic lesions by 34.9% and 20.1% in male and female mice, respectively, revealed by en-face Oil Red O staining, which did not correlate with changes in plasma lipid profiles. Real-time quantitative PCR analysis of cellular senescence markers p21 and p16 revealed that Meg3 deficiency aggravates hepatic cellular senescence but not cellular senescence at aortic roots. Human Meg3 transgenic mice were generated to examine the role of Meg3 gain-of-function in the development of atherosclerosis induced by PCSK9 overexpression. Meg3 overexpression promotes atherosclerotic lesion formation by 29.2% in Meg3 knock-in mice independent of its effects on lipid profiles. Meg3 overexpression inhibits hepatic cellular senescence, while it promotes aortic cellular senescence likely by impairing mitochondrial function and delaying cell cycle progression. CONCLUSIONS: Our data demonstrate that Meg3 promotes the formation of atherosclerotic lesions independent of its effects on plasma lipid profiles. In addition, Meg3 regulates cellular senescence in a tissue-specific manner during atherosclerosis. Thus, we demonstrated that Meg3 has multifaceted roles in cellular senescence and atherosclerosis.


Assuntos
Aterosclerose , Senescência Celular , Camundongos Knockout , Pró-Proteína Convertase 9 , RNA Longo não Codificante , Receptores de LDL , Animais , RNA Longo não Codificante/metabolismo , RNA Longo não Codificante/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/genética , Humanos , Masculino , Feminino , Receptores de LDL/genética , Receptores de LDL/metabolismo , Pró-Proteína Convertase 9/metabolismo , Pró-Proteína Convertase 9/genética , Modelos Animais de Doenças , Fígado/metabolismo , Fígado/patologia , Camundongos , Placa Aterosclerótica , Camundongos Endogâmicos C57BL , Doenças da Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor p16 de Quinase Dependente de Ciclina/genética , Mitocôndrias/metabolismo , Transdução de Sinais , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA