Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
3.
Genes Dis ; 10(1): 239-253, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37013030

RESUMO

It is increasingly aware that gut microbiota is closely associated with atherosclerosis. However, which and how specific gut bacteria regulate the progression of atherosclerosis is still poorly understood. In this study, modified linear discriminant analysis was performed in comparing the gut microbiota structures of atherosclerotic and non-atherosclerotic mice, and Desulfovibrio desulfuricans (D. desulfuricans) was found to be associated with atherosclerosis. D. desulfuricans-treated Apoe -/- mice showed significantly aggravated atherosclerosis. The proatherogenic effect of D. desulfuricans was attributed to its ability to increase intestinal permeability and subsequent raise in the transit of lipopolysaccharide (LPS) from the intestine to the bloodstream. Excessive LPS in the blood can elicit local and systemic inflammation and activate Toll-like receptor 4 (TLR4)/nuclear factor-κB (NF-κB) signaling of endothelial cells. TAK-242, a specific inhibitor of TLR4, can ameliorate the development of D. desulfuricans-induced atherosclerosis by blocking the LPS-induced activation of TLR4/NF-κB signaling.

4.
Genes Dis ; 9(6): 1701-1715, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36157502

RESUMO

GTPase-activating SH3 domain-binding protein 2 (G3BP2) is a mediator that responds to environmental stresses through stress granule formation and is involved in the progression of chronic diseases. However, no studies have examined the contribution of G3BP2 in the oscillatory shear stress (OSS)-induced endothelial dysfunction. Here we assessed the effects of G3BP2 in endothelial cells (ECs) function and investigated the underlying mechanism. Using shear stress apparatus and partial ligation model, we identified that stress granule-related genes in ECs could be induced by OSS with RNA-seq, and then confirmed that G3BP2 was highly and specifically expressed in athero-susceptible endothelia in the OSS regions. G3bp2 -/- Apoe -/- mice had significantly decreased atherosclerotic lesions associated with deficiency of G3BP2 in protecting endothelial barrier function, decreasing monocyte adhesion to ECs and inhibiting the proinflammatory cytokine levels. Furthermore, loss of G3BP2 diminished OSS-induced inflammation in ECs by increasing YAP nucleocytoplasmic shuttling and phosphorylation. These data demonstrate that G3BP2 is a critical OSS regulated gene in regulating ECs function and that G3BP2 inhibition in ECs is a promising atheroprotective therapeutic strategy.

5.
PLoS Pathog ; 18(6): e1010596, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35666747

RESUMO

Schistosomiasis is caused by parasitic flatworms known as schistosomes and affects over 200 million people worldwide. Prevention of T cell exhaustion by blockade of PD-1 results in clinical benefits to cancer patients and clearance of viral infections, however it remains largely unknown whether loss of PD-1 could prevent or cure schistosomiasis in susceptible mice. In this study, we found that S. japonicum infection dramatically induced PD-1 expression in T cells of the liver where the parasites chronically inhabit and elicit deadly inflammation. Even in mice infected by non-egg-producing unisex parasites, we still observed potent induction of PD-1 in liver T cells of C57BL/6 mice following S. japonicum infection. To determine the function of PD-1 in schistosomiasis, we generated PD-1-deficient mice by CRISPR/Cas9 and found that loss of PD-1 markedly increased T cell count in the liver and spleen of infected mice. IL-4 secreting Th2 cells were significantly decreased in the infected PD-1-deficient mice whereas IFN-γ secreting CD4+ and CD8+ T cells were markedly increased. Surprisingly, such beneficial changes of T cell response did not result in eradication of parasites or in lowering the pathogen burden. In further experiments, we found that loss of PD-1 resulted in both beneficial T cell responses and amplification of regulatory T cells that prevented PD-1-deficient T cells from unleashing anti-parasite activity. Moreover, such PD-1-deficient Tregs exert excessive immunosuppression and express larger amounts of adenosine receptors CD39 and CD73 that are crucial for Treg-mediated immunosuppression. Our experimental results have elucidated the function of PD-1 in schistosomiasis and provide novel insights into prevention and treatment of schistosomiasis on the basis of modulating host adaptive immunity.


Assuntos
Schistosoma japonicum , Esquistossomose Japônica , Animais , Humanos , Terapia de Imunossupressão , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/genética , Linfócitos T Reguladores
6.
Int J Biol Sci ; 18(5): 2163-2180, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35342333

RESUMO

Background: TET1 has been implicated in regulating inflammation and cardiovascular disease, but a newly discovered short isoform of TET1 (termed TET1s) exhibits higher expression in adult tissues than full-length TET1. However, the precise role of TET1 in cardiovascular disease remains undefined. Methods and Results: Based on TET1-/- knockout mice (with deletion of both TET1 and TET1s ) and TET1cs/cs mice (with deletion of only TET1), we found that TET1s deletion in TET1-/- mice resulted in more serious atherosclerotic lesions in the whole aorta than TET1cs/cs in the ApoE-/- background mice fed a high-fat diet. Atherosclerotic lesions with Oil red staining were dramatically localized in the aortic arch, abdominal aorta and ligated LCA, where they were exposed to OSS. Furthermore, the OSS-induced depression of TET1s in vitro and in vivo increased inflammatory cell and red blood cell infiltration into the subendothelial layer by impairing the vascular intimal barrier. TET1s upregulation enhanced vascular endothelial barrier function by increasing gap protein connexin 40 (CX40) expression as measured by RNA-seq and was confirmed by CX40 knockdown. TET1s interaction with Sin3a increased the global and CX40 promoter histone H3K27 acetylation levels, but not DNA methylation, to induce CX40 expression. Conclusions: These data demonstrate the unexpected discovery that laminar shear stress induces TET1s expression to protect the vascular endothelial barrier by increasing CX40 expression in ECs and that TET1s overexpression may be the core step for OSS-induced atherosclerosis therapy.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Animais , Aorta/metabolismo , Aterosclerose/metabolismo , Doenças Cardiovasculares/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Estresse Mecânico
7.
Theranostics ; 11(1): 164-180, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33391468

RESUMO

Atherosclerosis (AS), the underlying cause of most cardiovascular events, is one of the most common causes of human morbidity and mortality worldwide due to the lack of an efficient strategy for targeted therapy. In this work, we aimed to develop an ideal biomimetic nanoparticle for targeted AS therapy. Methods: Based on macrophage "homing" into atherosclerotic lesions and cell membrane coating nanotechnology, biomimetic nanoparticles (MM/RAPNPs) were fabricated with a macrophage membrane (MM) coating on the surface of rapamycin-loaded poly (lactic-co-glycolic acid) copolymer (PLGA) nanoparticles (RAPNPs). Subsequently, the physical properties of the MM/RAPNPs were characterized. The biocompatibility and biological functions of MM/RAPNPs were determined in vitro. Finally, in AS mouse models, the targeting characteristics, therapeutic efficacy and safety of the MM/RAPNPs were examined. Results: The advanced MM/RAPNPs demonstrated good biocompatibility. Due to the MM coating, the nanoparticles effectively inhibited the phagocytosis by macrophages and targeted activated endothelial cells in vitro. In addition, MM-coated nanoparticles effectively targeted and accumulated in atherosclerotic lesions in vivo. After a 4-week treatment program, MM/RAPNPs were shown to significantly delay the progression of AS. Furthermore, MM/RAPNPs displayed favorable safety performance after long-term administration. Conclusion: These results demonstrate that MM/RAPNPs could efficiently and safely inhibit the progression of AS. These biomimetic nanoparticles may be potential drug delivery systems for safe and effective anti-AS applications.


Assuntos
Membrana Celular , Células Endoteliais , Macrófagos , Nanopartículas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Sirolimo/administração & dosagem , Animais , Aterosclerose , Materiais Biomiméticos , Modelos Animais de Doenças , Progressão da Doença , Portadores de Fármacos , Técnicas In Vitro , Teste de Materiais , Camundongos , Fagocitose , Placa Aterosclerótica
8.
Front Cell Dev Biol ; 9: 769673, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35118065

RESUMO

Functional genomics in a mammalian model such as mice is fundamental for understanding human biology. The CRISPR/Cas9 system dramatically changed the tempo of obtaining genetic mouse models due to high efficiency. However, experimental evidence for the establishment of sgRNA knock-in animals and analyses of their value in functional genomics are still not sufficient, particularly in mammalian models. In this study, we demonstrate that the establishment of sgRNA knock-in mice is feasible, and more importantly, crosses between sgRNA knock-in mice and the Cas9 constitutively expressing mice result in complete deletion of the target gene. Such sgRNA knock-in provides an alternative approach for in vivo genetic modification and can be useful in multiple circumstances, such as maintenance of genetically modified animals, which are difficult to breed as homozygotes, and cross of such mice to diverse genomic backgrounds to obtain genetically modified animals.

9.
Atherosclerosis ; 310: 64-74, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32919187

RESUMO

BACKGROUND AND AIMS: Atherosclerosis is mainly caused by stress in arterial microenvironments, which results in the formation of stress granules as a consequence of the stress response. As the core protein of stress granules, GTPase-activating protein (SH3 domain)-binding protein 2 (G3BP2) is known to play pivotal roles in tumour initiation, viral infection and Alzheimer's disease, but the role of G3BP2 in atherosclerosis development is poorly understood. Previous studies have shown that vaccination with epitopes from self-antigens could reduce atherosclerotic lesions. Here, we investigated the effect of immunizing ApoE-/- mice with G3BP2 peptides, and whether this immunization exerted an anti-atherogenic effect. METHODS AND RESULTS: In our study, ApoE-/- mice were fed a high-fat diet for 12 weeks from 8 to 20 weeks of age. Then, using a repetitive multiple site strategy, the mice were immunized with a Keyhole limpet haemocyanin (KLH) conjugated G3BP2 peptide for 2 weeks from weeks 16 to 18. High levels of G3BP2 antibodies were detectable before sacrifice. Histological analyses showed that the number of atherosclerotic lesions in ApoE-/- mice was significantly reduced following G3BP2 immunotherapy. The levels of pro-inflammatory cytokines and macrophages were also greatly decreased, while the collagen content of the plaques showed significant increase. Furthermore, knocking down G3BP2 in ApoE-/- mice reduced the number of lesions compared to ApoE-/- mice fed a high-fat diet for eight weeks. In vitro studies demonstrated that G3BP2 regulated ox-LDL-induced inflammation in HUVECs via controlling the localization of IκBα. CONCLUSIONS: Immunization with the G3BP2 peptide antigen or knocking down of G3BP2 significantly decreased early atherosclerotic plaques in the ApoE-/- mouse model of atherosclerosis. G3BP2 is a promising potential target for atherosclerosis therapy.


Assuntos
Aterosclerose , Placa Aterosclerótica , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/prevenção & controle , Modelos Animais de Doenças , Regulação para Baixo , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
10.
Theranostics ; 10(23): 10712-10728, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929376

RESUMO

Rationale: For intravascular stent implantation to be successful, the processes of vascular tissue repair and therapy are considered to be critical. However, the mechanisms underlying the eventual fate of vascular smooth muscle cells (VSMCs) during vascular tissue repair remains elusive. In this study, we hypothesized that M2 macrophage-derived exosomes to mediate cell-to-cell crosstalk and induce dedifferentiation phenotypes in VSMCs. Methods:In vivo, 316L bare metal stents (BMS) were implanted from the left iliac artery into the abdominal aorta of 12-week-old male Sprague-Dawley (SD) rats for 7 and 28 days. Hematoxylin and eosin (HE) were used to stain the neointimal lesions. En-face immunofluorescence staining of smooth muscle 22 alpha (SM22α) and CD68 showed the rat aorta smooth muscle cells (RASMCs) and macrophages. Immunohistochemical staining of total galactose-specific lectin 3 (MAC-2) and total chitinase 3-like 3 (YM-1) showed the total macrophages and M2 macrophages. In vitro, exosomes derived from IL-4+IL-13-treated macrophages (M2Es) were isolated by ultracentrifugation and characterized based on their specific morphology. Ki-67 staining was conducted to assess the effects of the M2Es on the proliferation of RASMCs. An atomic force microscope (AFM) was used to detect the stiffness of the VSMCs. GW4869 was used to inhibit exosome release. RNA-seq was performed to determine the mRNA profiles of the RASMCs and M2Es-treated RASMCs. Quantitative real-time PCR (qRT-PCR) analysis was conducted to detect the expression levels of the mRNAs. Western blotting was used to detect the candidate protein expression levels. T-5224 was used to inhibit the DNA binding activity of AP-1 in RASMCs. Results: M2Es promote c-KIT expression and softening of nearby VSMCs, hence accelerating the vascular tissue repair process. VSMCs co-cultured in vitro with M2 macrophages presented an increased capacity for de-differentiation and softening, which was exosome dependent. In addition, the isolated M2Es helped to promote VSMC dedifferentiation and softening. Furthermore, the M2Es enhanced vascular tissue repair potency by upregulation of VSMCs c-KIT expression via activation of the c-Jun/activator protein 1 (AP-1) signaling pathway.Conclusions: The findings of this study emphasize the prominent role of M2Es during VSMC dedifferentiation and vascular tissue repair via activation of the c-Jun/AP-1 signaling pathway, which has a profound impact on the therapeutic strategies of coronary stenting techniques.


Assuntos
Doença das Coronárias/cirurgia , Procedimentos Endovasculares/instrumentação , Macrófagos/metabolismo , Músculo Liso Vascular/imunologia , Neointima/imunologia , Animais , Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Linhagem Celular , Doença das Coronárias/imunologia , Modelos Animais de Doenças , Exossomos/imunologia , Exossomos/metabolismo , Humanos , Macrófagos/imunologia , Masculino , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , RNA-Seq , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/imunologia , Stents , Fator de Transcrição AP-1/metabolismo
11.
Mol Cell Biochem ; 474(1-2): 15-26, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32712749

RESUMO

Endothelial cells (ECs) play an important role in the pathogenesis of cardiovascular disease, especially atherosclerosis (AS). The abnormal wall shear stress (WSS) which directly contacts with ECs is the key stimulating factor leading to AS. However, the underlying mechanism of ECs responding to WSS is still incompletely understood. This study aims to explore the novel mechano-sensitive genes and its potential mechanism in response to WSS in ECs by employing bioinformatics methods based on previously available high-throughput data from zebrafish embryos, both before and after blood flow formation. Six common differentially expressed genes (DEGs) (SRGN, SLC12A3, SLC25A4, PVALB1, ITGAE.2, zgc:198419) were selected out from two high-throughput datasets (GSE126617 and GSE20707) in the GEO database. Among them, SRGN was chosen for further verification through the in vitro shear stress loading experiments with human umbilical vein endothelial cells (HUVECs) and the in vivo partial ligation of carotid artery in mice. Our data indicated that low shear stress (LSS) could enhance the expression of SRGN via the PKA/CREB-dependent signaling pathway. The proportion of Ki67+ cells and the concentration of nitric oxide (NO) were high in SRGN high expression cells, suggesting that SRGN may be involved in the proliferation of HUVECs. Furthermore, in the partial ligation of the carotid artery mice model, we observed that the expression of SRGN was significantly increased in atherosclerotic plaques induced by abnormal shear stress. Taken together, this study demonstrated that SRGN is a key gene in the response of ECs to WSS and could be involved in AS.


Assuntos
Aterosclerose/patologia , Endotélio Vascular/patologia , Mecanotransdução Celular , Neovascularização Patológica/patologia , Proteoglicanas/metabolismo , Resistência ao Cisalhamento , Estresse Mecânico , Proteínas de Transporte Vesicular/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Knockout para ApoE , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Proteoglicanas/genética , Proteínas de Transporte Vesicular/genética
12.
Appl Microbiol Biotechnol ; 104(14): 6337-6350, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32472174

RESUMO

Atherosclerosis is a chronic inflammatory disease mediated by monocyte infiltration and cholesterol deposition into the subendothelial area, resulting in foam cell development. Probiotics are live bacteria that are beneficial for health when administered orally in adequate amounts. In this study, 8-week-old atherosclerosis-prone apolipoprotein E-deficient (ApoE-/-) mice were fed with or without Lactobacillus plantarum ATCC 14917 per day for 12 weeks. Serum was collected to analyse the lipid profile, oxidative status and proinflammatory cytokines. The heart was isolated to quantify the atherosclerotic lesion size in the aortic arch. Quantitative real-time polymerase chain reaction was performed to determine the expression levels of tumour necrosis factor-alpha (TNF-α) and interleukin (IL)-1ß in the aorta. The proteins extracted from the aorta were used for Western blot analysis to assess the expression levels of nuclear factor kappa B (NF-κB) and inhibitor of NF-κB (IκBα). The composition of gut microbiota was also examined through high-throughput sequencing. Results showed that the daily consumption of L. plantarum ATCC 14917 had no effect on body weight and lipid profile. L. plantarum ATCC 14917 treatment significantly inhibited atherosclerotic lesion formation. In addition, the oxLDL, MDA, TNF-α and IL-1ß levels were significantly reduced, whereas the SOD level was induced in the bacteria + high-fat diet group. Furthermore, the administration of L. plantarum ATCC 14917 significantly attenuated IκBα protein degradation and inhibited the translocation of P65 subunits of NF-κB. L. plantarum ATCC 14917 treatment also modulated the composition of gut microbiota in ApoE-/- mice. Our findings showed that L. plantarum ATCC 14917 supplementation decreases the progression of atherosclerotic lesion formation by alleviating the inflammatory process and lowering oxidative stress.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/prevenção & controle , Citocinas/metabolismo , Lactobacillus plantarum/fisiologia , Estresse Oxidativo/efeitos dos fármacos , Probióticos/farmacologia , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Aterosclerose/patologia , Microbioma Gastrointestinal/efeitos dos fármacos , Inflamação , Camundongos , Camundongos Knockout , Inibidor de NF-kappaB alfa/metabolismo , Probióticos/administração & dosagem , Fator de Transcrição RelA/metabolismo
13.
J Nutr Biochem ; 79: 108353, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32145470

RESUMO

Probiotics are known to be beneficial in preventing different diseases in model animals, including inflammatory bowel disease. However, there are few studies on probiotics related to miRNA regulation and disease status. In this article, the beneficial role and mechanisms of the probiotic strain Bifidobacterium bifidum ATCC 29521 have been studied in ulcerative colitis using dextran sodium sulphate (DSS) model. Male C57JBL/6 mice were randomly divided into three groups (n=7): Normal group, dextran sulphate sodium (DSS) group, and Bifido group gavage with Bifidobacterium bifidum ATCC 29521 (2×108 CFU/day). Our strain restored the DSS-caused damage by regulating the expression of immune markers and tight junction proteins (TJP) in the colon; briefly by up-regulating ROS-scavenging enzymes (SOD1, SOD2, CAT, and GPX2), anti-inflammatory cytokines (IL-10, PPARγ, IL-6), TJP's (ZO-1, MUC-2, Claudin-3, and E Cadherin-1) and downregulating inflammatory genes (TNF-α, IL-1ß) in Bifido group mice. Inflammatory markers appeared to be regulated by NF-κB nuclear P65 subunit, and its translocation was inhibited in Bifido group mice colon. In addition, the expression of inflammatory genes and colonic TJP were also associated with the restoration of miRNAs (miR-150, miR-155, miR-223) in B. bifidum ATCC 29521 treated Bifido group. The dysbiosis executed by DSS was restored in the Bifido group, demonstrating that B. bifidum ATCC 29521 possessed a probiotic role in our DSS colitis mouse model. B. bifidum ATCC 29521 exhibited its probiotic role through its anti-inflammatory role by modulating miRNA-associated TJP and NF-κB regulation and by partially restoring dysbiosis.


Assuntos
Bifidobacterium bifidum , Colite Ulcerativa/tratamento farmacológico , Probióticos/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/metabolismo , Colo/metabolismo , Citocinas/metabolismo , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Disbiose/induzido quimicamente , Disbiose/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Interleucina-1beta/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , NF-kappa B/metabolismo , Probióticos/administração & dosagem , Fator de Necrose Tumoral alfa/metabolismo
14.
J Biomed Mater Res B Appl Biomater ; 108(1): 94-103, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-30974041

RESUMO

Improving the surface properties of vascular stents to accelerate endothelialization in vivo could play an important role in minimizing the risk of late thrombosis. We previously showed that mussel adhesive protein fused with VE-cadherin extracellular domain (VE-M) specifically triggered endothelial cell adhesion in vitro. In this study, using stent implants coated with VE-M, we evaluated the clinical applicability of VE-M in endothelialization recovery in vivo. First, we explored the effect of VE-M on hemocompatibility and tight junctions between endothelial cells (ECs) in vitro. VE-M significantly inhibited platelet adhesion and promoted EC proliferation. Furthermore, VE-M drastically increased the centralization of F-actin in human umbilical vein endothelial cells (HUVECs) along the cell contacts, reduced fluorescein isothiocyanate (FITC)-dextran transport across the HUVECs, and elevated expression levels of tight junction proteins (TJPs) in ECs. We then evaluated the effect of VE-M on endothelialization recovery in vivo through implantation of vascular stents. At 1 day after implantation, stents coated with VE-M recruited more endothelial progenitor cells (EPCs) than bare stents. At 7 days after implantation, VE-M stents had a greater coverage of ECs than bare stents. At 1 month after implantation, ECs on VE-M stents were appropriately elliptical in morphology and closely resembled physiological morphology. Hematoxylin-eosin (HE) staining revealed little in-stent neointima formation on VE-M stents, and SEM images revealed that smooth endothelium had formed on VE-M stents without adherent platelets. Taken together, these findings indicate that VE-M accelerates in vivo endothelialization of vascular stents via recruitment of EPCs and promotes endothelium formation and could be explored as a potential bioactive coating for vascular implant. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 108B:94-103, 2020.


Assuntos
Antígenos CD/farmacologia , Caderinas/farmacologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Neointima , Proteínas/farmacologia , Proteínas Recombinantes de Fusão/farmacologia , Stents , Junções Íntimas/metabolismo , Animais , Antígenos CD/química , Antígenos CD/genética , Caderinas/química , Caderinas/genética , Adesão Celular , Humanos , Masculino , Proteínas/química , Proteínas/genética , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética
15.
Int J Biol Macromol ; 155: 1589-1598, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-31770555

RESUMO

Currently, bioresorbable stents made with biodegradable materials are attracting more and more attentions in cardiovascular tissue engineering. Especially, poly-L-lactic acid (PLLA) stent has been regarded as the most promising one due to excellent biodegradability until serious in-stent restenosis at late stage was reported. This imply that the PLLA stent has side effect in cell function, and it is rarely reported the effect of degradation product of PLLA on endothelial function. Here we reported that lactic acid (LA) not acidic pH induced endothelial-to-mesenchymal transition (EndMT) leading to vascular fibrosis which may contribute to in-stent stenosis after PLLA stent implantation. Furthermore, we found TGF-ß1 signaling was involved in boosting EndMT by LA. These results demonstrate a mechanism of in-stent stenosis induced by PLLA and indicate its utility for the future design of polymeric vascular scaffolds.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Ácido Láctico/farmacologia , Mesoderma/patologia , Stents/efeitos adversos , Fator de Crescimento Transformador beta1/metabolismo , Animais , Constrição Patológica/induzido quimicamente , Constrição Patológica/metabolismo , Constrição Patológica/patologia , Células Endoteliais da Veia Umbilical Humana/citologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Masculino , Coelhos
16.
Adv Sci (Weinh) ; 6(12): 1900172, 2019 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-31380165

RESUMO

Cardiovascular disease is the leading cause of mortality worldwide. Atherosclerosis, one of the most common forms of the disease, is characterized by a gradual formation of atherosclerotic plaque, hardening, and narrowing of the arteries. Nanomaterials can serve as powerful delivery platforms for atherosclerosis treatment. However, their therapeutic efficacy is substantially limited in vivo due to nonspecific clearance by the mononuclear phagocytic system. In order to address this limitation, rapamycin (RAP)-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles are cloaked with the cell membrane of red blood cells (RBCs), creating superior nanocomplexes with a highly complex functionalized bio-interface. The resulting biomimetic nanocomplexes exhibit a well-defined "core-shell" structure with favorable hydrodynamic size and negative surface charge. More importantly, the biomimetic nature of the RBC interface results in less macrophage-mediated phagocytosis in the blood and enhanced accumulation of nanoparticles in the established atherosclerotic plaques, thereby achieving targeted drug release. The biomimetic nanocomplexes significantly attenuate the progression of atherosclerosis. Additionally, the biomimetic nanotherapy approach also displays favorable safety properties. Overall, this study demonstrates the therapeutic advantages of biomimetic nanotherapy for atherosclerosis treatment, which holds considerable promise as a new generation of drug delivery system for safe and efficient management of atherosclerosis.

17.
Appl Microbiol Biotechnol ; 103(15): 5993-6006, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31201452

RESUMO

Atherosclerosis is the major cause of cardiovascular diseases, which are considered the fatal ailment globally. Hypercholesterolaemia plays a critical role in the development of atherosclerosis and cardiovascular diseases. Many studies have been stated that probiotics could affect hypercholesterolemia via cholesterol metabolism. Probiotics are live bacteria which are good for our health when administered orally in high amounts. Recently, many studies have revealed the beneficial effects of the nutritional ingestion of probiotics which can decrease serum cholesterol levels. The aim of this review is, firstly, to explore the hypercholesterolemia effect of how it progresses into atherosclerosis and, secondly, to summarize the hypocholesterolaemia effect of probiotics on atherosclerosis and the up-to-date information on their basic mechanisms. The most important mechanisms responsible for the hypocholesterolemic effect of probiotics are the suppression of the reabsorption of bile acids and inhibition of the intestinal cholesterol absorption. Current studies indicate that numerous mechanisms within the cholesterol metabolism, e.g., ones involving the Niemann-Pick C1-Like 1 protein, 3-hydroxy-3-methylglutaryl-CoA reductase, and 7α- and 27α-hydroxylases, have been recommended where regulation may take place after oral intake of probiotics. However, these mechanisms are still poorly understood. Thus, further studies are required to examine the possible mechanisms, whereby probiotics can be utilized safely and considered for the treatment of hypercholesterolemia.


Assuntos
Aterosclerose/prevenção & controle , Aterosclerose/fisiopatologia , Hipercolesterolemia/complicações , Hipercolesterolemia/terapia , Probióticos/administração & dosagem , Probióticos/farmacologia , Animais , Colesterol/metabolismo , Humanos
18.
Appl Microbiol Biotechnol ; 103(7): 3181-3191, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30783721

RESUMO

Hypercholesterolemia plays a critical role in the development of atherosclerosis and cardiovascular diseases. Many works have been reported that gut microbiota could affect hypercholesterolemia through cholesterol metabolism. However, the role of gut microbiota on cholesterol transportation remains unclear. In this study, 8-week-old C57BL/6J mice were fed with high-cholesterol diet to build the hypercholesterolemic mice. Then, the hypercholesterolemic mice got the oral administration of Enterococcus faecalis ATCC19433 at a dose of 109 CFU/mL/day or PBS with high-cholesterol diet for 4 weeks. Serum was collected to detect the concentration of total cholesterol (TC). Meanwhile, pathology, histology, real-time polymerase chain reaction, Western blot, and immunofluorescence were used to evaluate the expression of ABCG5 and ABCG8 in the liver and small intestine. We also analyzed the composition of gut microbiota through high-throughput sequencing method. Oral administration of E. faecalis ATCC19433 significantly decreased the concentration of serum cholesterol in hypercholesterolemic mice. Furthermore, E. faecalis ATCC19433 reduced the concentration of liver cholesterol and improved cholesterol by increasing the expression of ABCG5 and ABCG8. Moreover, oral administration of E. faecalis ATCC19433 modulated the composition of gut microbiota and increased the counts of Lactobacillus, Bifidobacterium, and Akkermansia. Our results showed that E. faecalis ATCC19433 could exert hypocholesterolemic effect on hypercholesterolemic mice by improving transporter ABCG5 and ABCG8. E. faecalis ATCC19433 maybe contribute to the transportation of cholesterol potentially and modulate the composition of gut microbiota.


Assuntos
Colesterol/metabolismo , Enterococcus faecalis/metabolismo , Microbioma Gastrointestinal , Interações Microbianas , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/genética , Administração Oral , Animais , Bifidobacterium/isolamento & purificação , Transporte Biológico , Colesterol/sangue , Hipercolesterolemia/terapia , Lactobacillus/isolamento & purificação , Metabolismo dos Lipídeos , Lipoproteínas/genética , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
J Mater Chem B ; 6(24): 4151-4163, 2018 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-32255158

RESUMO

Endothelium is the only known completely non-thrombogenic material. In the present study, a strategy to mimic the adhesive interactions of endothelial cells (ECs) to alter the vascular microenvironment was established and applied to directing the behaviour of cells. To facilitate the regeneration of a functional endothelium in vascular lesions, we designed a recombinant mussel foot protein (Mfp-5) fused with the VE-cadherin extracellular domain EC1-2, termed VE-M. Surface coating analysis showed that recombinant VE-M successfully formed a coating on substrate materials with uniform nanorods, low roughness, and sufficient hydrophilicity. We then evaluated the effects of VE-M on the adhesion of ECs and the capture of endothelial progenitor cells (EPCs). The result demonstrated that VE-M efficiently promoted the adhesion of ECs and EPCs. The number of ECs and EPCs on VE-M was 5.5- and 1.8-fold higher, respectively, than that on bare 316L SS under static conditions, whereas there was no significant difference in the number of captured smooth muscle cells (SMCs) between VE-M and other substrates. In addition, the number of EPCs captured by VE-M was approximately four times higher than that captured by 316L SS under dynamic conditions. In particular, the result of the neutralization test indicated that VE-M specifically triggered ECs' adhesion via the interaction of VE-cadherin EC1-2. Further investigation showed that VE-M significantly increased the levels of endogenous VE-cadherin in HUVECs as well as the endothelial eNOS content, with little or no endothelial inflammation. Our results showed that VE-M could be a promising biomimetic modification for accelerating endothelialization and vascularization in tissue engineering.

20.
Cardiovasc Ther ; 35(5)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28664667

RESUMO

AIM: Pitavastatin (Pit) has been proved to efficiently inhibit the onset and progression of atherosclerosis. However, the mechanism by which Pit exerts nonlipid-related effects, such as antiinflammatory actions, is not quite clear. Our study aimed at investigating the effect of Pit on the expression of endothelial NO synthase (eNOS) and miR-155 in LPS-stimulated HUVECs to reveal the antiinflammatory mechanism of pitavastatin. METHODS: HUVECs were isolated from newborn umbilical cords and used in the experiments at passages 2-5. Cells were treated with LPS (0.05, 0.1, 1 µg/L) or LPS (0.1 µg/L)+Pit (0.01, 0.1, 1 µmol/L), untreated cells were used as control. For LPS+Pit induction, cells were firstly incubated with Pit for 1 hour before coincubation with LPS for 24 hours. eNOS mRNA and miR-155 were detected by RT-PCR, and Western blotting was used to detect protein expression of eNOS. RESULTS: Treatment of HUVECs with LPS enhanced the expression of miR-155 and reduced the expression of eNOS in mRNA and protein level in a dose-dependent manner as revealed by RT-PCR and Western blotting, respectively. Pitavastatin ameliorated LPS-induced endothelial dysfunction through upregulation of eNOS expression and downregulation of miR-155 expression. CONCLUSION: Pitavastatin increases eNOS expression and inhibits of LPS-induced miR-155 expression.


Assuntos
Anti-Inflamatórios/farmacologia , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Lipopolissacarídeos/farmacologia , MicroRNAs/metabolismo , Óxido Nítrico Sintase Tipo III/biossíntese , Quinolinas/farmacologia , Células Cultivadas , Relação Dose-Resposta a Droga , Regulação para Baixo , Indução Enzimática , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , MicroRNAs/genética , Óxido Nítrico Sintase Tipo III/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA