Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Acta Biomater ; 169: 372-386, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37597679

RESUMO

Recent studies have demonstrated the critical role of cardiac-resident macrophages (cMacs) in the maintenance of physiological homeostasis. However, recruitment of circulating monocyte-derived macrophages decreases cMac levels post-myocardial infarction (MI). Transplanting cMacs is not an ideal option due to their low survival rates and the risk of immunological rejection. However, extracellular vesicle therapy has the potential to provide a feasible and safe alternative for cardiac repair. In this study, cell membrane-modified extracellular vesicles (MmEVs) were developed for heart repair by modifying cMac-derived extracellular vesicles (mEVs) with monocyte membranes, resulting in immune evasion and sequential targeted localization to damaged regions through expression of CD47 on MmEVs and strong affinity between monocyte membrane proteins and CCL2. Additionally, to fully exploit the potential clinical application of MmEVs and achieve a better curative effect, thymosin ß4 (Tß4) was loaded into the nanoparticles, resulting in Tß4-MmEVs. In vitro experiments indicated that both the MmEVs and Tß4-MmEVs promoted cardiomyocyte proliferation and endothelial cell migration. Animal experiments suggested that MI mice treated with MmEVs and Tß4-MmEVs exhibited reduced myocardial fibrosis and increased vascular density compared to the control group. Thus, we posit that these targeted nanoparticles hold significant potential for MI adjuvant therapy and may open new avenues for cardiac repair and regeneration. STATEMENT OF SIGNIFICANCE: Extracellular vesicles (EVs) derived from bioactive parent cell sources involved in pathological and repair processes for cardiovascular disease have emerged as a compelling strategy for regenerative therapy. In this study, we constructed monocyte membrane-modified extracellular vesicles loaded with a drug (Tß4-MmEVs) for heart repair that exhibit extraordinary abilities of immune evasion and sequential localization to damaged regions owing to the presence of CD47 and the strong affinity between monocytes and damaged cardiomyocytes and endothelial cells. The bioactivities of Tß4-MmEVs on enhancing cardiomyocyte and endothelial cell proliferation were validated both in vitro and in vivo. Effective development and implementation of therapeutically membrane-modified nanoparticles from homologous origins can provide a reference for adjuvant therapy in clinical MI management.


Assuntos
Antígeno CD47 , Monócitos , Animais , Camundongos , Células Endoteliais , Macrófagos , Miócitos Cardíacos
2.
Nat Commun ; 14(1): 2094, 2023 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-37055411

RESUMO

Extracellular vesicles have shown good potential in disease treatments including ischemic injury such as myocardial infarction. However, the efficient production of highly active extracellular vesicles is one of the critical limitations for their clinical applications. Here, we demonstrate a biomaterial-based approach to prepare high amounts of extracellular vesicles with high bioactivity from endothelial progenitor cells (EPCs) by stimulation with silicate ions derived from bioactive silicate ceramics. We further show that hydrogel microspheres containing engineered extracellular vesicles are highly effective in the treatment of myocardial infarction in male mice by significantly enhancing angiogenesis. This therapeutic effect is attributed to significantly enhanced revascularization by the high content of miR-126a-3p and angiogenic factors such as VEGF and SDF-1, CXCR4 and eNOS in engineered extracellular vesicles, which not only activate endothelial cells but also recruit EPCs from the circulatory system.


Assuntos
Células Progenitoras Endoteliais , Vesículas Extracelulares , Infarto do Miocárdio , Camundongos , Masculino , Animais , Neovascularização Fisiológica , Infarto do Miocárdio/terapia , Transdução de Sinais
3.
Acta Biomater ; 149: 82-95, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35777549

RESUMO

The overproduction of reactive oxygen species (ROS) and burst of inflammation following cardiac ischemia-reperfusion (I/R) are the leading causes of cardiomyocyte injury. Monotherapeutic strategies designed to enhance anti-inflammatory or anti-ROS activity explicitly for treating I/R injury have demonstrated limited success because of the complex mechanisms of ROS production and induction of inflammation. Intense oxidative stress leads to sustained injury, necrosis, and apoptosis of cardiomyocytes. The damaged and necrotic cells can release danger-associated molecular patterns (DAMPs) that can cause the aggregation of immune cells by activating Toll-like receptor 4 (TLR4). These immune cells also promote ROS production by expressing NADPH oxidase. Finally, ROS production and inflammation form a vicious cycle, and ROS and TLR4 are critical nodes of this cycle. In the present study, we designed and prepared an injectable hydrogel system of EGCG@Rh-gel by co-assembling epigallocatechin-3-gallate (EGCG) and the rhein-peptide hydrogel (Rh-gel). The co-assembled hydrogel efficiently blocked the ROS-inflammation cycle by ROS scavenging and TLR4 inhibition. Benefited by the abundant noncovalent interactions of π-π stacking and hydrogen bonding between EGCG and Rh-gel, the co-assembled hydrogel had good mechanical strength and injectable property. Following the injection EGCG@Rh-gel into the damaged region of the mice's heart after I/R, the hydrogel enabled to achieve long-term sustained release and treatment, improve cardiac function, and significantly reduce the formation of scarring. Further studies demonstrated that these beneficial outcomes arise from the reduction of ROS production, inhibition of inflammation, and induction of anti-apoptosis in cardiomyocytes. Therefore, EGCG@Rh-gel is a promising drug delivery system to block the ROS-inflammation cycle for resisting myocardial I/R injury. STATEMENT OF SIGNIFICANCE: 1. Monotherapeutic strategies designed to enhance anti-inflammatory or anti-ROS effects for treating I/R injury have demonstrated limited success because of the complex mechanisms of ROS and inflammation. 2. ROS production and inflammation form a vicious cycle, and ROS and TLR4 are critical nodes of this cycle. 3. Here, we designed an injectable hydrogel system of EGCG@Rh-gel by co-assembling epigallocatechin-3-gallate (EGCG) and a rhein-peptide hydrogel (Rh-gel). EGCG@Rh-gel efficiently blocked the ROS-inflammation cycle by ROS scavenging and TLR4 inhibition. 4. EGCG@Rh-gel achieved long-term sustained release and treatment, improved cardiac function, and significantly reduced the formation of scarring after I/R. 5. The beneficial outcomes arise from reducing ROS production, inhibiting inflammation, and inducing anti-apoptosis in cardiomyocytes.


Assuntos
Catequina , Traumatismo por Reperfusão Miocárdica , Animais , Catequina/farmacologia , Cicatriz/tratamento farmacológico , Preparações de Ação Retardada/uso terapêutico , Hidrogéis/farmacologia , Hidrogéis/uso terapêutico , Inflamação/tratamento farmacológico , Camundongos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Espécies Reativas de Oxigênio , Receptor 4 Toll-Like
4.
Chem Commun (Camb) ; 58(62): 8690-8693, 2022 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-35833251

RESUMO

We report a peptidic dual-targeting drug delivery platform (integrins targeting and self-assembly instructed by matrix metalloproteinases) towards inflamed endothelial cells, which improved the anti-inflammatory ability of the loaded drug (i.e., puerarin) in vitro and thus improved the antiatherogenic effect of the loaded drug (i.e., puerarin) in vivo.


Assuntos
Aterosclerose , Células Endoteliais , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Apolipoproteínas E , Aterosclerose/tratamento farmacológico , Camundongos , Peptídeos/farmacologia , Peptídeos/uso terapêutico
5.
Bioact Mater ; 14: 416-429, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35386821

RESUMO

Currently, stem cell transplantations in cardiac repair are limited owing to disadvantages, such as immunological rejection and poor cell viability. Although direct injection of exosomes can have a curative effect similar to that of stem cell transplantation, high clearance hinders its application in clinical practice. Previous reports suggested that induction of coronary collateralization can be a desired method of adjunctive therapy for someone who had missed the optimal operation time to attenuate myocardial ischemia. In this study, to mimic the paracrine and biological activity of stem cells, we developed artificial stem cells that can continuously release Tß4-exosomes (Tß4-ASCs) by encapsulating specific exosomes within microspheres using microfluidics technology. The results show that Tß4-ASCs can greatly promote coronary collateralization in the periphery of the myocardial infarcted area, and its therapeutic effect is superior to that of directly injecting the exosomes. In addition, to better understand how it works, we demonstrated that the Tß4-ASC-derived exosomes can enhance the angiogenic capacity of coronary endothelial cells (CAECs) via the miR-17-5p/PHD3/Hif-1α pathway. In brief, as artificial stem cells, Tß4-ASCs can constantly release functional exosomes and stimulate the formation of collateral circulation after myocardial infarction, providing a feasible and alternative method for clinical revascularization.

6.
Biomaterials ; 279: 121231, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34739980

RESUMO

Cell therapy offers a promising paradigm for heart tissue regeneration. Human induced pluripotent stem cells (hiPS) and their cardiac derivatives are emerging as a novel treatment for post-myocardial infarction repair. However, the immature phenotype and function of hiPS-derived cardiomyocytes (hiPS-CMs), particularly poor electrical coupling, limit their potential as a therapy. Herein, we developed a hybrid gold nanoparticle (AuNP)-hyaluronic acid (HA) hydrogel matrix encapsulating hiPS-CMs to overcome this limitation. Methacrylate-modified-HA was used as the backbone and crosslinked with a matrix metalloproteinase-2 (MMP-2) degradable peptide to obtain a MMP-2-responsive hydrogel; RGD peptide was introduced as an adhesion point to enhance biocompatibility; AuNPs were incorporated to regulate the mechanical and topological properties of the matrix by significantly increasing its stiffness and surface roughness, thereby accelerating gap junction formation in hiPS-CMs and orchestrating calcium handling via the αnß1integrin-mediated ILK-1/p-AKT/GATA4 pathway. Transplanted AuNP-HA-hydrogel-encapsulated-hiPS-CMs developed more robust gap junctions in the infarcted mice heart and resynchronized electrical conduction of the ventricle post-myocardial infarction. The hiPS-CMs delivered by the hydrogels exerted stronger angiogenic effects, which also contributed to the recovery process. This study provides insight into constructing an injectable biomimetic for structural and functional renovation of the injured heart.


Assuntos
Células-Tronco Pluripotentes Induzidas , Nanopartículas Metálicas , Animais , Junções Comunicantes , Ouro , Humanos , Ácido Hialurônico , Metaloproteinase 2 da Matriz , Camundongos , Miócitos Cardíacos
7.
ACS Appl Mater Interfaces ; 13(19): 22131-22141, 2021 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-33957750

RESUMO

The reconstruction of blood perfusion is a crucial therapeutic method to save and protect cardiac function after acute myocardial infarction (AMI). The activation of the hepatocyte growth factor precursor (pro-HGF) has a significant effect on promoting angiogenesis and antiapoptosis. The oxygen/glucose deprivation (OGD) caused by AMI could induce vascular adventitia fibroblasts to differentiate into myofibroblasts and secrete the pro-HGF. Meanwhile, the specific Met receptor of the hepatocyte growth factor (HGF) is upregulated in endothelial cells during AMI. However, the poor prognosis of AMI suggests that the pro-HGF is not effectively activated. Improving the activation efficiency of the pro-HGF may play a positive role in the treatment of AMI. Herein, we designed supramolecular nanofibers self-assembled by compound 1 (Comp.1, Nap-FFEG-IVGGYPWWMDV), which can strongly activate the pro-HGF and initiate HGF-Met signaling. Studies have proven that Comp.1 possesses a better ability to activate the pro-HGF to perform antiapoptosis and pro-angiogenesis. In vivo results have confirmed that the retention time of Comp.1 and its accumulation in the infarct area of the heart are promoted. Moreover, Comp.1 plays an effective role in promoting angiogenesis in the marginal area of AMI, reducing myocardial fibrosis, and protecting cardiac function. Herein, we will optimize the structure of bioactive peptides through supramolecular self-assembly and amplify their therapeutic effect by improving their efficiency, providing a new strategy for the therapy of AMI.


Assuntos
Fator de Crescimento de Hepatócito/metabolismo , Infarto do Miocárdio/terapia , Nanofibras , Neovascularização Patológica , Precursores de Proteínas/metabolismo , Sequência de Aminoácidos , Fator de Crescimento de Hepatócito/química , Células Endoteliais da Veia Umbilical Humana , Humanos , Nanofibras/química , Precursores de Proteínas/química , Transdução de Sinais , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Biochem Genet ; 59(5): 1185-1202, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33715137

RESUMO

Hypertrophic cardiomyopathy (HCM) is one of the most common genetic heart diseases. Its features include abnormal cardiomyocyte hypertrophy, microvascular dysfunction, and increased accumulation of intercellular matrix. We aim to unravel genes associated with the pathogenesis of HCM and provide a potential target for diagnosis and treatment. Key modules were identified by weighted gene co-expression network analysis (WGCNA). A miRNA-mRNA network was constructed with the predicted miRNA and the most likely hub gene was screened out for gene set enrichment analysis (GSEA). The diagnostic capacity of hub gene was verified by receiver operating characteristic (ROC) curves. Single-cell sequencing (sc-RNA seq) data of normal adult hearts were used to further predict the specific cell types expressing the hub gene. WGCNA assigned genes into different modules and found that the genes contained in the red module had the strongest positive correlation with HCM disease. 2.5% of the genes were common between DEG and hub genes. With the miRNA-mRNA network, osteomodulin (OMD) was identified as the most potential hub gene. GSEA showed that OMD was mainly involved in the synthesis of extracellular matrix and had a certain inhibitory effect on the immune system. The expression of OMD in HCM was validated and ROC curve analysis showed that OMD could distinguish HCM from controls with the area under the curve (AUC) > 0.7. The sc-RNA seq revealed that OMD was mainly expressed in the later stages of cardiac fibroblasts, suggesting that OMD may have an effect on fibroblasts, participating in the pathogenesis of HCM. OMD may serve as a biomarker and therapeutic target for HCM in the future.


Assuntos
Biomarcadores/análise , Cardiomiopatia Hipertrófica/patologia , Proteínas da Matriz Extracelular/genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Proteoglicanas/genética , Cardiomiopatia Hipertrófica/genética , Perfilação da Expressão Gênica , Humanos , Curva ROC
9.
Theranostics ; 11(5): 2263-2277, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33500724

RESUMO

Extracellular vesicles (EVs) are nanoscale extracellular vesicles derived from endocytosis that are crucial to intercellular communication. EVs possess natural biocompatibility and stability that allow them to cross biological membranes and that protect them from degradation. Recent studies have shown that EVs-mediated crosstalk between different cell types in the heart could play important roles in the maintenance of cardiac homeostasis and the pathogenesis of heart diseases. In particular, EVs secreted by different types of stem cells exhibit cardioprotective effects. However, numerous studies have shown that intravenously injected EVs are quickly cleared by macrophages of the mononuclear phagocyte system (MPS) and preferentially accumulate in MPS organs such as the liver, spleen, and lung. In this review, we discuss exosome biogenesis, the role of EVs in heart diseases, and challenges in delivering EVs to the heart. Furthermore, we extensively discuss the targeted delivery of EVs for treating ischemic heart disease. These understandings will aid in the development of effective treatment strategies for heart diseases.


Assuntos
Vesículas Extracelulares/transplante , Traumatismos Cardíacos/terapia , Animais , Comunicação Celular , Endocitose , Humanos
10.
J Cell Mol Med ; 25(1): 170-183, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33314583

RESUMO

Yes-associated protein (YAP), a major effector of the Hippo signalling pathway, is widely implicated in vascular pathophysiology processes. Here, we identify a new role of YAP in the regulation of vascular senescence. The inhibition or deficiency and overexpression of YAP were performed in human umbilical vein endothelial cells (HUVECs) and isolated vascular tissues. Cellular and vascular senescence was assessed by analysis of the senescence-associated ß-galactosidase (SA-ß-gal) and expression of senescence markers P16, P21, P53, TERT and TRF1. We found that YAP was highly expressed in old vascular tissues, inhibition and knockdown of YAP decreased senescence, while overexpression of YAP increased the senescence in both HUVECs and vascular tissues. In addition, autophagic flux blockage and mTOR pathway activation were observed during YAP-induced HUVECs and vascular senescence, which could be relieved by the inhibition and knockdown of YAP. Moreover, YAP-promoted cellular and vascular senescence could be relieved by mTOR inhibition. Collectively, our findings indicate that YAP may serve as a potential therapeutic target for ageing-associated cardiovascular disease.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autofagia , Vasos Sanguíneos/patologia , Senescência Celular , Serina-Treonina Quinases TOR/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Ratos Sprague-Dawley , Proteínas de Sinalização YAP
11.
Diabetes Care ; 43(10): 2574-2580, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32732375

RESUMO

OBJECTIVE: To investigate whether intraindividual variability of fasting glucose (FG) in young adulthood is associated with coronary artery calcification (CAC) progression in middle age. RESEARCH DESIGN AND METHODS: We included 2,256 CARDIA (Coronary Artery Risk Development Study in Young Adults) participants with CAC assessment by computed tomography scanner at baseline (2000-2001) and 10 years later (2010-2011). CAC progression was assessed for each individual as the difference of logarithmic CAC scores at follow-up and baseline (log[CAC (follow-up) + 1] - log[CAC (baseline) + 1]). FG variability was defined by the coefficient of variation about the mean FG (FG-CV), the SD of FG (FG-SD), and the average real variability of FG (FG-ARV) during the 10-year follow-up. We investigated the association between FG variability and CAC progression with adjustment for demographics, clinical risk factors, mean FG level, change in FG level, diabetes incidence, and medication use. RESULTS: After multivariable adjustment, 1-SD increment in FG-CV was associated with worse progression of CAC as demonstrated as percent change in CAC, with incident CAC 5.9% (95% CI 1.0, 10.7) and any CAC progression 6.7% (95% CI 2.3, 11.1) during 10 years. Similar findings were also observed in FG-SD and FG-ARV. CONCLUSIONS: Higher FG variability during young adulthood was associated with greater CAC progression in middle age, suggesting its value in predicting risk for subclinical coronary artery diseases.


Assuntos
Glicemia/metabolismo , Doença da Artéria Coronariana/epidemiologia , Doença da Artéria Coronariana/patologia , Jejum/sangue , Calcificação Vascular/epidemiologia , Calcificação Vascular/patologia , Adolescente , Adulto , Idade de Início , Envelhecimento/fisiologia , Glicemia/análise , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/etiologia , Diabetes Mellitus/epidemiologia , Angiopatias Diabéticas/epidemiologia , Angiopatias Diabéticas/etiologia , Progressão da Doença , Feminino , Seguimentos , Humanos , Hiperglicemia/sangue , Hiperglicemia/complicações , Hiperglicemia/epidemiologia , Incidência , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Calcificação Vascular/sangue , Calcificação Vascular/etiologia , Adulto Jovem
12.
Chinese Journal of School Health ; (12): 543-546, 2020.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-821414

RESUMO

Objective@#To explore the effect of drawing therapy on the behavior of children with attention deficit hyperactivity disorder (ADHD), and to provide a new perspective for the treatment of children with attention deficit hyperactivity disorder (ADHD).@*Methods@#In June 2019, a random sample of three primary schools in 1-6 grade in Baishan who diagnosed with ADHD was recruited, the experimental group 10 ADHD children were given painting therapy intervention for 14 weeks, four times a week, 120 mins each time, the control group 10 ADHD children received no intervention. During the study period all the participants do not receive any other treatment. The ADHD Rating Scale Parent version,the Conners Parents Symptom Questionnaire(PSQ) and the Rey Complex Figure test was assessed before and after the intervention to evaluate the symptoms of ADHD children.@*Results@#In terms of PSQ score test, there were significant differences between the scores of four dimension factors in the experimental group after therapeutic intervention and those before intervention and those after intervention in the control group(P<0.05). The follow-up test showed that the PSQ score in the experimental group remained good (P<0.05). There were two dimension factors that were not significantly different in the post-test and follow-up post-test and before intervention. In Rey complex memory graph test score test, it was found that the score of four dimensions after intervention in the experimental group was significantly different from that before intervention and after intervention in the control group (P<0.05). After 12 weeks, the score of follow-up test remained good (P<0.05).@*Conclusion@#Painting therapy has a significant effect on ADHD children’s behavioral management ability and working memory ability, and the effect can last for at least 12 weeks.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...