Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 455
Filtrar
1.
Aesthetic Plast Surg ; 47(5): 2117-2129, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37400586

RESUMO

BACKGROUND: Cryopreserved fat has limited clinical applications due to its rapid absorption, high degree of fibrosis, and risk of complications after grafting. Many studies have verified that Adipose-derived mesenchymal stem cell-derived exosomes (ADSC-Exos) can improve fresh fat graft survival. This study assessed whether ADSC-Exos could improve the survival of cryopreserved fat grafts. METHODS: Exosomes were isolated from human ADSCs were subcutaneously engrafted with adipose tissues stored under different conditions (fresh; cryopreserved for 1 month) into the backs of BALB/c nude mice (n = 24), and exosomes or PBS were administered weekly. Grafts were harvested at 1, 2, 4, and 8 weeks, and fat retention rate, histologic, and immunohistochemical analyses were conducted. RESULTS: At 1, 2, and 4 weeks after the transfer, cryopreserved fat grafts in groups of exosome-treated showed better fat integrity, fewer oil cysts, and reduced fibrosis. Further investigations of macrophage infiltration and neovascularization revealed that those exosomes increased the number of M2 macrophages at 2 and 4 weeks (p<0.05), but had limited impact on vascularization (p>0.05). It's important to note that no significant differences (p>0.05) were observed between the two groups in both histological and immunohistochemical evaluations at 8 weeks post-transplantation. CONCLUSIONS: This study suggests that ADSC-Exos could improve the survival of cryopreserved fat grafts in the short term (within 4 weeks), but the overall improvement was poor (after 8 weeks). This suggests that the utility of using ADSC-Exos to treat cryopreserved adipose tissue grafts is limited. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each submission to which Evidence-Based Medicine rankings are applicable. This excludes Review Articles, Book Reviews, and manuscripts that concern Basic Science, Animal Studies, Cadaver Studies, and Experimental Studies. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Assuntos
Exossomos , Sobrevivência de Enxerto , Camundongos , Animais , Humanos , Exossomos/transplante , Camundongos Nus , Tecido Adiposo/transplante , Criopreservação , Células-Tronco , Fibrose
2.
Stem Cells Transl Med ; 12(3): 154-168, 2023 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-36893290

RESUMO

Intrauterine adhesions (IUA), which is characterized by endometrial fibrosis, continue to be the most common cause of uterine infertility globally. Our work revealed that 3 fibrotic progression markers (Vimentin, COL5A2, and COL1A1) were significantly increased in the endometrium of IUA patients. Mesenchymal stem cell-derived exosomes (EXOs) have been recently revealed as a cell-free therapy for fibrosis diseases. Nevertheless, the application of EXOs is restricted by the short residency duration in the target tissue. To overcome this limitation, herein, we reported an exosome-based regimen (EXOs-HP) that thermosensitive poloxamer hydrogel possessed the ability to efficiently promote the residency duration of EXOs in the uterine cavity. By downregulating fibrotic progression markers (Vimentin, COL5A2, and COL1A1), EXOs-HP could significantly restore the function and structure of the injured endometrium in the IUA model. Our work provides the theoretical and experimental foundation of EXOs-HP in treating IUA, highlighting the clinical potential of topical EXOs-HP delivery system in IUA patients.


Assuntos
Exossomos , Doenças Uterinas , Feminino , Humanos , Biomarcadores , Colágeno , Endométrio , Exossomos/transplante , Fibrose , Aderências Teciduais/tratamento farmacológico , Aderências Teciduais/patologia , Doenças Uterinas/terapia , Doenças Uterinas/patologia , Vimentina/uso terapêutico
3.
J Plast Reconstr Aesthet Surg ; 76: 219-229, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36527904

RESUMO

Recently, there has been renewed interest in autologous fat grafting both for its filler and regenerative traits. The universal application, however, has been impeded by the unstable survival rates and complications. There has been substantial research undertaken on the role of adipose-derived stem cells (ADSCs) involved in fat graft fates including angiogenesis, adipogenesis, and inflammatory regulation. As the effectors of their parental cells, ADSC-derived exosomes (ADSC-exos) encapsulating multiple bioactive cargoes mediate cell-to-cell communication in a paracrine manner. ADSC-exos have received much attention for their biocompatible and efficient therapeutic potentials as "cell-free therapy" in plastic surgery, including increasing fat grafting survival rates. In this review, we summarize the current knowledge about the biological basis of ADSC-exos, ADSC-related mechanisms of fat survival, research updates of ADSC-exos in autologous fat grafting, and discuss some challenges along with research prospects.


Assuntos
Exossomos , Procedimentos de Cirurgia Plástica , Humanos , Tecido Adiposo/transplante , Exossomos/transplante , Adipócitos , Células-Tronco
4.
J Mol Neurosci ; 72(12): 2507-2516, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36534294

RESUMO

Diabetic peripheral neuropathy (DPN) is a highly prevalent diabetic complication characterized at the molecular level by mitochondrial dysfunction and deleterious oxidative damage. No effective treatments for DPN are currently available. The present study was developed to examine the impact of exosomes derived from bone marrow mesenchymal stromal cells (BMSCs) overexpressing sirtuin 1 (SIRT1) on DPN through antioxidant activity and the preservation of mitochondrial homeostasis. A DPN model was established using 20-week-old diabetic model mice (db/db). Exosomes were prepared from control BMSCs (exo-control) and BMSCs that had been transduced with a SIRT1 lentivirus (exo-SIRT1). Sensory and motor nerve conduction velocity values were measured to assess neurological function, and mechanical and thermal sensitivity were analyzed in these animals. Exo-SIRT1 preparations exhibited a high loading capacity and readily accumulated within peripheral nerves following intravenous administration, whereupon they were able to promote improved neurological recovery relative to exo-control treatment. DPN mice exhibited significantly improved nerve conduction velocity following exo-SIRT1 treatment. Relative to exo-control-treated mice, those that underwent exo-SIRT1 treatment exhibited significantly elevated TOMM20 and Nrf2/HO-1 expression, reduced MDA levels, increased GSH and SOD activity, and increased MMP. Together, these results revealed that both exo-control and exo-SIRT1 administration was sufficient to reduce the morphological and behavioral changes observed in DPN model mice, with exo-SIRT1 treatment exhibiting superior therapeutic efficacy. These data thus provide a foundation for future efforts to explore other combinations of gene therapy and exosome treatment in an effort to alleviate DPN.


Assuntos
Exossomos , Células-Tronco Mesenquimais , Doenças do Sistema Nervoso Periférico , Animais , Camundongos , Medula Óssea/metabolismo , Exossomos/metabolismo , Exossomos/transplante , Células-Tronco Mesenquimais/metabolismo , Mitocôndrias/metabolismo , Doenças do Sistema Nervoso Periférico/metabolismo , Sirtuína 1/genética , Sirtuína 1/metabolismo , Sirtuína 1/uso terapêutico
5.
Clin Transl Med ; 12(8): e1026, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35988156

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is persistently threatening the lives of thousands of individuals globally. It triggers pulmonary oedema, driving to dyspnoea and lung failure. Viral infectivity of coronavirus disease 2019 (COVID-19) is a genuine challenge due to the mutagenic genome and mysterious immune-pathophysiology. Early reports highlighted that extracellular vesicles (exosomes, Exos) work to enhance COVID-19 progression by mediating viral transmission, replication and mutations. Furthermore, recent studies revealed that Exos derived from immune cells play an essential role in the promotion of immune cell exhaustion by transferring regulatory lncRNAs and miRNAs from exhausted cells to the active cells. Fortunately, there are great chances to modulate the immune functions of Exos towards a sustained repression of COVID-19. Engineered Exos hold promising immunotherapeutic opportunities for remodelling cytotoxic T cells' function. Immune cell-derived Exos may trigger a stable epigenetic repression of viral infectivity, restore functional cytokine-producing T cells and rebalance immune response in severe infections by inducing functional T regulatory cells (Tregs). This review introduces a view on the current outcomes of immunopathology, and immunotherapeutic applications of immune cell-derived Exos in COVID-19, besides new perspectives to develop novel patterns of engineered Exos triggering novel anti-SARS-CoV-2 immune responses.


Assuntos
COVID-19 , Exossomos , COVID-19/terapia , Citocinas , Exossomos/transplante , Humanos , Linfócitos/metabolismo , MicroRNAs/genética , MicroRNAs/uso terapêutico , RNA Longo não Codificante/genética , RNA Longo não Codificante/uso terapêutico , SARS-CoV-2
6.
Stem Cell Res Ther ; 13(1): 295, 2022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35841008

RESUMO

BACKGROUND: Recent studies have shown that bone marrow stromal cell-derived exosomes (BMSC-Exos) can be used for tissue repair. However, whether the BMSC-Exos can promote tendon-bone healing after anterior cruciate ligament reconstruction (ACLR) is still unclear. In this study, we observed in vivo and in vitro the effect of rat BMSC-Exos on tendon-bone healing after ACLR and its possible mechanism. METHODS: Highly expressed miRNAs in rat BMSC-Exos were selected by bioinformatics and verified in vitro. The effect of overexpressed miRNA in BMSC-Exos on M2 macrophage polarization was observed. A rat model of ACLR was established. The experimental components were divided into three groups: the control group, the BMSC-Exos group, and the BMSC-Exos with miR-23a-3p overexpression (BMSC-Exos mimic) group. Biomechanical tests, micro-CT, and histological staining were performed for analysis. RESULTS: Bioinformatics analysis showed that miR-23a-3p was highly expressed in rat BMSC-Exos and could target interferon regulatory factor 1 (IRF1, a crucial regulator in M1 macrophage polarization). In vitro, compared with the control group or the BMSC-Exos group, the BMSC-Exos mimic more significantly promoted the polarization of macrophages from M1 to M2. In vivo, at 2 weeks, the number of M2 macrophages in the early local stage of ACLR was significantly increased in the BMSC-Exos mimic group; at 4 and 8 weeks, compared with the control group or the BMSC-Exos group, the bone tunnels of the tibia and femur sides of the rats in the BMSC-Exos mimic group were significantly smaller, the interface between the graft and the bone was narrowed, the bone volume/total volume ratio (BV/TV) increased, the collagen type II alpha 1 level increased, and the mechanical strength increased. CONCLUSIONS: BMSC-Exos promoted M1 macrophage to M2 macrophage polarization via miR-23a-3p, reduced the early inflammatory reaction at the tendon-bone interface, and promoted early healing after ACLR.


Assuntos
Reconstrução do Ligamento Cruzado Anterior , Exossomos , Macrófagos , Células-Tronco Mesenquimais , MicroRNAs , Animais , Exossomos/transplante , Macrófagos/citologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Ratos , Tendões
7.
Cell Biochem Funct ; 40(5): 430-438, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35647674

RESUMO

The pandemic of COVID-19 caused worldwide concern. Due to the lack of appropriate medications and the inefficiency of commercially available vaccines, lots of efforts are being made to develop de novo therapeutic modalities. Besides this, the possibility of several genetic mutations in the viral genome has led to the generation of resistant strains such as Omicron against neutralizing antibodies and vaccines, leading to worsening public health status. Exosomes (Exo), nanosized vesicles, possess several therapeutic properties that participate in intercellular communication. The discovery and application of Exo in regenerative medicine have paved the way for the alleviation of several pathologies. These nanosized particles act as natural bioshuttles and transfer several biomolecules and anti-inflammatory cytokines. To date, several approaches are available for the administration of Exo into the targeted site inside the body, although the establishment of standard administration routes remains unclear. As severe acute respiratory syndrome coronavirus 2 primarily affects the respiratory system, we here tried to highlight the transplantation of Exo in the alleviation of COVID-19 pathologies.


Assuntos
COVID-19 , Exossomos , COVID-19/terapia , Citocinas , Exossomos/transplante , Humanos , SARS-CoV-2
8.
Ann Neurol ; 92(2): 230-245, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35596947

RESUMO

OBJECTIVE: Accumulation of α-synuclein (α-syn) in neurons is a prominent feature of Parkinson's disease (PD). Recently, researchers have considered that extracellular vesicles (EVs) may play an important role in protein exportation and propagation, and α-syn-containing EVs derived from the central nervous system (CNS) have been detected in peripheral blood. However, mechanistic insights into CNS-derived EVs have not been well-described. METHODS: Likely neurogenic EVs were purified from the plasma of PD patients and healthy controls using a well-established immunoprecipitation assay with anti-L1CAM-coated beads. A Prnp-SNCAA53T transgenic PD mouse model was used to evaluate the neuronal pathology induced by PD-derived L1CAM-purified EVs. EV-associated microRNA (miRNA) profiling was used to screen for altered miRNAs in PD-derived L1CAM-purified EVs. RESULTS: PD patient-derived L1CAM-purified (likely neurogenic) EVs facilitated α-syn pathology and neuron loss in Prnp-SNCAA53T transgenic PD mice. The miRNA, novel_miR_44438, was significantly increased in the PD group, which promoted α-syn accumulation and neuronal degeneration in a dose-dependent manner. Novel _miR_44438 directly targets NDST1 mRNA and inhibits the function of heparan sulfate, thus preventing exosome biogenesis and α-syn release from exosomes. INTERPRETATION: Novel_miR_44438 in PD-derived L1CAM-purified EVs inhibits the α-syn efflux from neurons thereby promoting the pathological accumulation and aggregation of α-syn. ANN NEUROL 2022;92:230-245.


Assuntos
Exossomos , MicroRNAs , Doença de Parkinson , Animais , Humanos , Camundongos , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Exossomos/metabolismo , Exossomos/transplante , Camundongos Transgênicos , MicroRNAs/genética , MicroRNAs/metabolismo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia
9.
J Diabetes Res ; 2022: 5126968, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35237694

RESUMO

The prevalence of diabetes mellitus (DM) is increasing at a staggering rate around the world. In the United States, more than 30.3 million Americans have DM. Type 2 diabetes mellitus (T2DM) accounts for 91.2% of diabetic cases and disproportionately affects African Americans and Hispanics. T2DM is a major risk factor for cardiovascular disease (CVD) and is the leading cause of morbidity and mortality among diabetic patients. While significant advances in T2DM treatment have been made, intensive glucose control has failed to reduce the development of macro and microvascular related deaths in this group. This highlights the need to further elucidate the underlying molecular mechanisms contributing to CVD in the setting of T2DM. Endothelial dysfunction (ED) plays an important role in the development of diabetes-induced vascular complications, including CVD and diabetic nephropathy (DN). Thus, the endothelium provides a lucrative means to investigate the molecular events involved in the development of vascular complications associated with T2DM. microRNAs (miRNA) participate in numerous cellular responses, including mediating messages in vascular homeostasis. Exosomes are small extracellular vesicles (40-160 nanometers) that are abundant in circulation and can deliver various molecules, including miRNAs, from donor to recipient cells to facilitate cell-to-cell communication. Endothelial cells are in constant contact with exosomes (and exosomal content) that can induce a functional response. This review discusses the modulatory role of exosomal miRNAs and proteins in diabetes-induced endothelial dysfunction, highlighting the significance of miRNAs as markers, mediators, and potential therapeutic interventions to ameliorate ED in this patient group.


Assuntos
Diabetes Mellitus Tipo 2/genética , Células Endoteliais/efeitos dos fármacos , MicroRNAs/análise , MicroRNAs/farmacologia , Biomarcadores/análise , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/fisiopatologia , Células Endoteliais/metabolismo , Exossomos/efeitos dos fármacos , Exossomos/transplante , Humanos , MicroRNAs/uso terapêutico
10.
Mol Cell Biochem ; 477(4): 1249-1260, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35119583

RESUMO

Molecular mechanisms underlying myocardial ischemia/reperfusion (MI/R) injury and effective strategies to treat MI/R injury are both in shortage. Although pyroptosis of cardiomyocytes and the protective role of cardiac fibroblasts (CFs) have been well recognized as targets to reduce MI/R injury and sudden cardiac death (SCD), the connection has not yet been established. Here, we showed that CFs protected cardiomyocytes against MI/R-induced injury through suppression of pyroptosis. A novel molecular mechanism underpinning this effect was further identified. Under hypoxia/reoxygenation condition, CFs were found to secrete exosomes, which contain increased level of microRNA-133a (miR-133a). These exosomes then delivered miR-133a into cardiomyocytes to target ELAVL1 and repressed cardiomyocyte pyroptosis. Based on this finding, we successfully developed a new strategy that used exosomes derived from CFs with overexpressed miR-133a to enhance the therapeutic outcomes for the MI/R injury. Overall, our results provide a novel molecular basis for understanding and treating MI/R injury, and our study also provides novel insight for the postmortem diagnosis of MI/R injury induced SCD by using exosome biomarker in forensic.


Assuntos
Exossomos , Fibroblastos/metabolismo , MicroRNAs/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Animais , Exossomos/metabolismo , Exossomos/transplante , Humanos , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley
11.
BMC Cardiovasc Disord ; 22(1): 61, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35172728

RESUMO

BACKGROUND: Myocardial infarction (MI) represents a severe cardiovascular disease with limited therapeutic agents. This study was aimed to elucidate the role of the exosomes derived from human placental mesenchymal stem cells (PMSCs-Exos) in MI. METHODS: PMSCs were isolated and cultured in vitro, with identification by both transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). To further investigate the effects of PMSC-Exos on MI, C57BL/6 mice were randomly divided into Sham group, MI group, and PMSC-Exos group. After 4 weeks of the intervention, cardiac function was assessed by cardiac echocardiography, electrocardiogram and masson trichrome staining; lipid indicators were determined by automatic biochemical instrument; inflammatory cytokines were measured by cytometric bead array (CBA); gut microbiota, microbial metabolites short chain fatty acids (SCFAs) as well as lipopolysaccharide (LPS) were separately investigated by 16S rRNA high throughput sequencing, gas chromatography mass spectrometry (GC-MS) and tachypleus amebocyte lysate kit; transcriptome analysis was used to test the transcriptional components (mRNA\miRNA\cirRNA\lncRNA) of PMSC-Exos. RESULTS: We found that human PMSC-Exos were obtained and identified with high purity and uniformity. MI model was successfully established. Compared to MI group, PMSC-Exos treatment ameliorated myocardial fibrosis and left ventricular (LV) remodeling (P < 0.05). Moreover, PMSC-Exos treatment obviously decreased MI molecular markers (AST/BNP/MYO/Tn-I/TC), pro-inflammatory indicators (IL-1ß, IL-6, TNF-α, MCP-1), as well as increased HDL in comparison with MI group (all P < 0.05). Intriguingly, PMSC-Exos intervention notably modulated gut microbial community via increasing the relative abundances of Bacteroidetes, Proteobacteria, Verrucomicrobia, Actinobacteria, Akkermansia, Bacteroides, Bifidobacterium, Thauera and Ruminiclostridium, as well as decreasing Firmicutes (all P < 0.05), compared with MI group. Furthermore, PMSC-Exos supplementation increased gut microbiota metabolites SCFAs (butyric acid, isobutyric acid and valeric acid) and decreased LPS in comparison with MI group (all P < 0.05). Correlation analysis indicated close correlations among gut microbiota, microbial SCFAs and inflammation in MI. CONCLUSIONS: Our study highlighted that PMSC-Exos intervention alleviated MI via modulating gut microbiota and suppressing inflammation.


Assuntos
Bactérias/crescimento & desenvolvimento , Exossomos/transplante , Microbioma Gastrointestinal , Mediadores da Inflamação/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio/cirurgia , Miocárdio/metabolismo , Animais , Bactérias/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Disbiose , Exossomos/metabolismo , Exossomos/ultraestrutura , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/microbiologia , Miocárdio/patologia , Placenta/citologia , Gravidez , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma
12.
Int J Mol Sci ; 23(3)2022 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-35163541

RESUMO

Osteoarthritis (OA) has generally been introduced as a degenerative disease; however, it has recently been understood as a low-grade chronic inflammatory process that could promote symptoms and accelerate the progression of OA. Current treatment strategies, including corticosteroid injections, have no impact on the OA disease progression. Mesenchymal stem cells (MSCs) based therapy seem to be in the spotlight as a disease-modifying treatment because this strategy provides enlarged anti-inflammatory and chondroprotective effects. Currently, bone marrow, adipose derived, synovium-derived, and Wharton's jelly-derived MSCs are the most widely used types of MSCs in the cartilage engineering. MSCs exert immunomodulatory, immunosuppressive, antiapoptotic, and chondrogenic effects mainly by paracrine effect. Because MSCs disappear from the tissue quickly after administration, recently, MSCs-derived exosomes received the focus for the next-generation treatment strategy for OA. MSCs-derived exosomes contain a variety of miRNAs. Exosomal miRNAs have a critical role in cartilage regeneration by immunomodulatory function such as promoting chondrocyte proliferation, matrix secretion, and subsiding inflammation. In the future, a personalized exosome can be packaged with ideal miRNA and proteins for chondrogenesis by enriching techniques. In addition, the target specific exosomes could be a gamechanger for OA. However, we should consider the off-target side effects due to multiple gene targets of miRNA.


Assuntos
Exossomos/transplante , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Osteoartrite/terapia , Animais , Diferenciação Celular , Condrogênese , Progressão da Doença , Exossomos/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteoartrite/imunologia , Comunicação Parácrina , Regeneração
13.
Cell Death Dis ; 13(1): 32, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013102

RESUMO

Macrophage-derived exosomes (Mφ-Exo) have multidimensional involvement in tumor initiation, progression, and metastasis, but their regulation in hepatocellular carcinoma (HCC) is not fully understood. RBPJ has been implicated in macrophage activation and plasticity. In this study we assess the role of exosomes derived from RBPJ-overexpressed macrophages (RBPJ+/+ Mφ-Exo) in HCC. The circular RNA (circRNA) profiles in RBPJ+/+ Mφ-Exo and THP-1-like macrophages (WT Mφ)-Exo was evaluated using circRNA microarray. CCK-8, Transwell, and flow cytometry analyses were used to evaluate the function of Mφ-Exo-circRNA on HCC cells. Luciferase reporter assays, RNA immunoprecipitation, and Pearson's correlation analysis were used to confirm interactions. A nude mouse xenograft model was used to further analyze the functional significance of Mφ-Exo-cirRNA in vivo. Our results shown that hsa_circ_0004658 is upregulated in RBPJ+/+ Mφ-Exo compared to WT Mφ-Exo. RBPJ+/+ Mφ-Exo and hsa_circ_0004658 inhibits proliferation and promotes apoptosis in HCC cells, whereas hsa_circ_0004658 knockdown stimulated cell proliferation and migration but restrained apoptosis in vitro and promotes tumor growth in vivo. The effects of RBPJ+/+ Mφ-Exo on HCC cells can be reversed by the hsa_circ_0004658 knockdown. Mechanistic investigations revealed that hsa_circ_0004658 acts as a ceRNA of miR-499b-5p, resulting in the de-repression of JAM3. These results indicate that exosome circRNAs secreted from RBPJ+/+ Mφ inhibits tumor progression through the hsa_circ_0004658/miR-499b-5p/JAM3 pathway and hsa_circ_0004658 may be a diagnostic biomarker and potential target for HCC therapy.


Assuntos
Carcinoma Hepatocelular/genética , Moléculas de Adesão Celular/genética , Exossomos/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Neoplasias Hepáticas/genética , MicroRNAs/genética , RNA Circular/metabolismo , Animais , Apoptose/genética , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Exossomos/transplante , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Macrófagos/metabolismo , Camundongos , Camundongos Nus , RNA Circular/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Bioengineered ; 13(2): 3194-3206, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35067167

RESUMO

Mesenchymal stem cells (MSCs)-derived exosomes (Exo) are beneficial in the use of brain damages. Restrictively, the mechanism of Exo expressing miR-124-3p in hypoxic-ischemic brain damage (HIBD) is not completely comprehended. Thereupon, this work was put forward to reveal the action of bone marrow MSCs-derived Exo (BMSCs-Exo) expressing miR-124-3p in the illness. BMSCs were isolated and transfected with miR-124-3p agomir. Then, BMSCs-Exo were extracted and identified. The newborn HIBD rats were injected with miR-124-3p-modified BMSCs-Exo or tumor necrosis factor receptor associated factor 6 (TRAF6)-related vectors. Next, neurological functions, neuron pathological and structural damages, oxidative stress and neuronal apoptosis were observed. miR-124-3p and TRAF6 expression was tested, along with their targeting relationship. miR-124-3p was down-regulated, and TRAF6 was up-regulated in newborn HIBD rats. miR-124-3p targeted TRAF6. BMSCs-Exo improved neurological functions, alleviated neuron pathological and structural damages, suppressed oxidative stress and reduced neuronal apoptosis in newborn HIBD rats, whereas BMSCs-Exo-mediated effects were enhanced by restoring miR-124-3p. Silencing TRAF6 attenuated HIBD in newborn rats, but overexpression of TRAF6 reversed the protective role of miR-124-3p-overexpressing BMSCs-Exo. This work makes it comprehensive that up-regulated exosomal miR-124-3p ameliorates HIBD in newborn rats by targeting TRAF6, which replenishes the potential agents for curing HIBD.


Assuntos
Células da Medula Óssea/metabolismo , Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Exossomos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Fator 6 Associado a Receptor de TNF/metabolismo , Animais , Lesões Encefálicas/terapia , Isquemia Encefálica/terapia , Exossomos/metabolismo , Exossomos/transplante , Masculino , Ratos , Ratos Sprague-Dawley
15.
Bioengineered ; 13(1): 1650-1665, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35001794

RESUMO

Acute kidney injury (AKI) is a susceptible factor for chronic kidney disease (CKD). There is still a lack of effective prevention methods in clinical practice. This study investigated the protective effect of the urinary exosomes from premature infants on cisplatin-induced acute kidney injury. Here we isolated exosomes from the fresh urine of premature infants. A C57BL/6 mice model of cisplatin-induced acute kidney injury was given 100 ug urinary exosomes 24 hours after model establishment. The kidneys were collected for pathological examination and the evaluation of renal tubular damage and apoptosis. In the in vitro experiment, human renal cortex/proximal tubular cells (HK-2) were induced by cisplatin to assess the effect of the urine exosomes from premature infants. Exosome microRNA (miRNA) sequencing technology was applied to investigate the miRNAs enriched in exosomes and the dual-luciferase gene reporter system to examine the targeting relationship of the miRNA with target genes. The results indicated that the urinary exosomes could decrease the serum creatinine level and the apoptosis of renal tubular cells, and reduce mice mortality. In addition, miR-30a-5p was the most abundant miRNA in the exosomes. It protected HK-2 cells from cisplatin-induced apoptosis by targeting and down-regulating the mitogen-activated protein kinase 8 (MAPK8). Together, our findings identified that the urinary exosomes derived from premature infants alleviated cisplatin-induced acute kidney injury and inhibited the apoptosis of HK-2 via miR-30a-5p, which could target MAPK8. These findings implied that urinary exosomes from premature infants riched in miR-30a-5p might become a potential treatment for AKI.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/terapia , Cisplatino/efeitos adversos , Exossomos/transplante , Recém-Nascido Prematuro/urina , Proteína Quinase 8 Ativada por Mitógeno/genética , Animais , Linhagem Celular , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Creatinina/sangue , Modelos Animais de Doenças , Regulação para Baixo , Exossomos/genética , Feminino , Células HEK293 , Humanos , Recém-Nascido , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética
16.
Neurosci Lett ; 769: 136389, 2022 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-34896256

RESUMO

BACKGROUND: Previous investigations have shown that exosome secretion from hypoxic pre-treated adipose-derived stem cells (ADSCs) affect ischemic injury treatment; however, the therapeutic effect relative to circRNA delivery is unclear. METHODS: In the present investigation inflammatory factors, nerve injury, and cognitive function were assessed using a middle cerebral artery occlusion mouse model. The isolated exosomes were identified using transmission electron microscopy and further tested by leveraging exosome particles in a nanoparticle tracking approach. Differences in circRNA expression between exosomes and hypoxic pre-treated ADSC exosomes were analyzed by high-throughput sequencing. The phenotypic transformation of microglia was detected by immunofluorescence. The circRNA and downstream target were analyzed by bioinformatics, RT-qPCR, and luciferase report. RESULTS: Exosomes from hypoxic pre-treated ADSCs improved cognitive function by reducing neuronal damage in the hippocampus after cerebral infarction. Exosomes from hypoxic pre-treated ADSCs improved cognitive function via delivery of circ-Rps5. SIRT7 and miR-124-3p were circ-Rps5 downstream targets, which was confirmed by luciferase report analysis. miR-124-3p overexpression or SIRT7 downregulation reversed the circ-Rps5-mediated M2 microglial shift under LPS conditions. Circ-Rps5-modified ADSC exosome improved cognitive function by decreasing neuronal damage and shifting microglia from an M1 to M2 phenotype in the hippocampus. CONCLUSION: The study showed that exosomes from hypoxic pre-treated ADSCs attenuated acute ischemic stroke-induced brain injury via delivery of circ-Rps5 and promoted M2 microglia/macrophage polarization.


Assuntos
Exossomos/transplante , Infarto da Artéria Cerebral Média/terapia , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Microglia/metabolismo , RNA Circular/metabolismo , Proteínas Ribossômicas/genética , Tecido Adiposo/citologia , Animais , Células Cultivadas , Cognição , Exossomos/metabolismo , Hipocampo/citologia , Hipocampo/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Circular/genética , Sirtuínas/genética , Sirtuínas/metabolismo
17.
Placenta ; 117: 161-168, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34915433

RESUMO

The emergence of COVID-19 has created a major health crisis across the globe. Invasion of SARS-CoV-2 into the lungs causes acute respiratory distress syndrome (ARDS) that result in the damage of lung alveolar epithelial cells. Currently, there is no standard treatment available to treat the disease and the resultant lung scarring is irreversible even after recovery. This has prompted researchers across the globe to focus on developing new therapeutics and vaccines for the treatment and prevention of COVID-19. Mesenchymal stem cells (MSCs) have emerged as an efficient drug screening platform and MSC-derived organoids has found applications in disease modeling and drug discovery. Perinatal tissue derived MSC based cell therapies have been explored in the treatment of various disease conditions including ARDS because of their enhanced regenerative and immunomodulatory properties. The multi-utility properties of MSCs have been described in this review wherein we discuss the potential use of MSC-derived lung organoids in screening of novel therapeutic compounds for COVID-19 and also in disease modeling to better understand the pathogenesis of the disease. This article also summarizes the rationale behind the development of MSC-based cell- and cell-free therapies and vaccines for COVID-19 with a focus on the current progress in this area. With the pandemic raging, an important necessity is to develop novel treatment strategies which will not only alleviate the disease symptoms but also avoid any off-target effects which could further increase post infection sequelae. Naturally occurring mesenchymal stem cells could be the magic bullet which fulfil these criteria.


Assuntos
Âmnio/citologia , COVID-19/terapia , Células-Tronco Mesenquimais , Placenta/citologia , SARS-CoV-2 , Cordão Umbilical/citologia , Vacinas contra COVID-19 , Terapia Baseada em Transplante de Células e Tecidos , Exossomos/transplante , Feminino , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/ultraestrutura , Gravidez , SARS-CoV-2/imunologia , Geleia de Wharton/citologia
18.
Biosci Rep ; 42(1)2022 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-34750610

RESUMO

BACKGROUND: Acute lung injury (ALI) is a respiratory disease with high morbidity and mortality rates. Currently, there is no effective treatment to complement mechanical ventilation. Exosomes and microRNAs (miRNAs) are promising agents for the management of this disease. METHODS: Exosomes were isolated from mouse bone marrow stromal stem cells (BMSCs). The levels of two miRNAs, miR-542-3P and miR-150, in exosomes were determined using RT-PCR, and miR-150 was selected for further study. ALI model was established in mice using lipopolysaccharides, and then, they were treated with saline, exosomes, miRNA agomirs, or miRNA antagomirs. The concentrations of TNF-α, IL-6, and IL-1ß and the number of neutrophils and macrophages in the bronchoalveolar lavage fluid were measured. The wet/dry weight ratio of the lung tissue was calculated, and tissue pathology and apoptosis were observed using hematoxylin and eosin and terminal deoxynucleotidyl transferase dUTP nick-end labeling staining. CD34 and VE-cadherin expression was detected using immunofluorescence. Proteins associated with apoptosis and MAPK signaling were detected using Western blotting, and miR-150 expression in lung tissue was evaluated using RT-PCR. RESULTS: We successfully isolated BMSCs and exosomes and showed that the level of miR-150 was significantly higher than that of miR-542-3p. Exosomes and miR-150 reduced inflammation and lung edema while maintaining the integrity of the alveolar structure. They also mitigated microvascular endothelial cell injury by regulating the caspase-3, Bax/Bcl-2, and MAPK signaling. CONCLUSIONS: Exosomal miR-150 attenuates lipopolysaccharide-induced ALI through the MAPK pathway.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Exossomos/transplante , Pulmão/irrigação sanguínea , Transplante de Células-Tronco Mesenquimais , MicroRNAs/metabolismo , Microvasos/enzimologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/enzimologia , Lesão Pulmonar Aguda/patologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Exossomos/genética , Exossomos/metabolismo , Lipopolissacarídeos , Pulmão/enzimologia , Pulmão/patologia , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Transdução de Sinais
19.
Bioengineered ; 13(1): 27-37, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34898374

RESUMO

Wounds are soft tissue injuries, which are difficult to heal and can easily lead to other skin diseases. Bone marrow mesenchymal stem cells (BMSCs) and the secreted exosomes play a key role in skin wound healing. This study aims to clarify the effects and mechanisms of exosomes derived from BMSCs in wound healing. Exosomes were extracted from the supernatant of the BMSCs. The expression of the micro-RNA miR-93-3p was determined by qRT-PCR analysis. HaCaT cells were exposed to hydrogen peroxide (H2O2) to establish a skin lesion model. MTT, flow cytometry, and transwell assays were conducted to determine cellular functions. The binding relationship between miR-93-3p and apoptotic peptidase activating factor 1 (APAF1) was measured using a dual luciferase reporter gene assay. The results showed that BMSC-derived exosomes or BMSC-exos promoted proliferation and migration and suppressed apoptosis in HaCaT cells damaged by H2O2. However, the depletion of miR-93-3p in BMSC-exos antagonized the effects of BMSC-exos on HaCaT cells. In addition, APAF1 was identified as a target of miR-93-3p. Overexpression of APAF1 induced the dysfunction of HaCaT cells. Collectively, the results indicate that BMSC-derived exosomes promote skin wound healing via the miR-93-3p/APAF1 axis. This finding may help establish a new therapeutic strategy for skin wound healing.


Assuntos
Fator Apoptótico 1 Ativador de Proteases/genética , Exossomos/transplante , Peróxido de Hidrogênio/efeitos adversos , Queratinócitos/citologia , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Regiões 3' não Traduzidas , Fator Apoptótico 1 Ativador de Proteases/metabolismo , Linhagem Celular , Movimento Celular , Proliferação de Células , Exossomos/genética , Células HaCaT , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/metabolismo , Células-Tronco Mesenquimais/química , Modelos Biológicos , Cicatrização
20.
J Cell Biochem ; 123(2): 183-201, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34514621

RESUMO

Exosomes are extracellular vesicles secreted by various cell types, which play important roles in physiological processes. In particular, stem cell-derived exosomes have been shown to play crucial functions in intercellular communication during the tissue healing process. This review summarizes the effects of exosomes derived from different stem cell sources on the repair of cutaneous and bone tissue, focusing on the different pathways that could be involved in the regeneration process. The biogenesis, isolation, and content of exosomes have also been discussed. The effectiveness of exosomes is broadly demonstrated for skin and bone regeneration in animal models, supporting the basis for clinical translation of exosomes as a ready-to-use cell-free therapeutic for skin and bone regeneration.


Assuntos
Regeneração Óssea , Osso e Ossos , Exossomos , Pele , Células-Tronco/metabolismo , Animais , Osso e Ossos/lesões , Osso e Ossos/metabolismo , Exossomos/metabolismo , Exossomos/transplante , Humanos , Pele/lesões , Pele/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...