Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cytotherapy ; 25(5): 502-509, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36513574

RESUMO

BACKGROUND AIMS: As evidenced by ongoing clinical trials and increased activity in the commercial sector, extracellular vesicle (EV)-based therapies have begun the transition from bench to bedside. As this progression continues, one critical aspect of EV clinical translation is understanding the effects of storage and transport conditions. Several studies have assessed the impact of storage on EV characteristics such as morphology, uptake and component content, but effects of storage duration and temperature on EV functional bioactivity and, especially, loaded cargo are rarely reported. METHODS: The authors assessed EV outcomes following storage at different temperatures (room temperature, 4°C, -20°C, -80°C) for various durations as well as after lyophilization. RESULTS: Mesenchymal stromal cell (MSC) EVs were observed to retain key aspects of their bioactivity (pro-vascularization, anti-inflammation) for up to 4-6 weeks at -20°C and -80°C and after lyophilization. Furthermore, via in vitro assays and an in vivo wound healing model, these same storage conditions were also demonstrated to enable preservation of the functionality of loaded microRNA and long non-coding RNA cargo in MSC EVs. CONCLUSIONS: These findings extend the current understanding of how EV therapeutic potential is impacted by storage conditions and may inform best practices for handling and storing MSC EVs for both basic research and translational purposes.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , MicroRNAs , Cicatrização
2.
Noncoding RNA ; 8(5)2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36287123

RESUMO

Extracellular vesicles (EVs) have emerged as promising therapeutic entities in part due to their potential to regulate multiple signaling pathways in target cells. This potential is derived from the broad array of constituent and/or cargo molecules associated with EVs. Among these, microRNAs (miRNAs) are commonly implicated as important and have been associated with a wide variety of EV-induced biological phenomena. While controlled loading of single miRNAs is a well-documented approach for enhancing EV bioactivity, loading of multiple miRNAs has not been fully leveraged to maximize the potential of EV-based therapies. Here, an established approach to extrinsic nucleic acid loading of EVs, sonication, was utilized to load multiple miRNAs in HEK293T EVs. Combinations of miRNAs were compared to single miRNAs with respect to anti-inflammatory outcomes in assays of increasing stringency, with the combination of miR-146a, miR-155, and miR-223 found to have the most potential amongst the tested groups.

3.
Methods Mol Biol ; 2504: 231-239, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35467291

RESUMO

Extracellular vesicles (EVs) have emerged as key mediators of intercellular communication and consequently have the potential to be potent therapeutic vectors. Beyond their endogenous function, EVs are also being harnessed as drug delivery vehicles with possible benefits over synthetic nanoparticle systems. Despite advances in loading exogenous molecules into extracellular vesicles, efficient incorporation of nucleic acids remains a challenge due to aggregation and degradation. In this chapter, we detail a method to load EVs with negatively charged cargo, in particular nucleic acids, by modifying the internal pH of the vesicles to be acidic. This approach demonstrates that pH modification of EVs enables efficient loading of nucleic acids with functional cargo.


Assuntos
Vesículas Extracelulares , Nanopartículas , Ácidos Nucleicos , Comunicação Celular , Vesículas Extracelulares/metabolismo , Concentração de Íons de Hidrogênio , Ácidos Nucleicos/metabolismo
4.
iScience ; 24(12): 103441, 2021 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-34877498

RESUMO

Extracellular miRNAs (ex-miRNAs) mediate intercellular communication and play a role in diverse physiological and pathological processes. Using small RNA sequencing, we identify that miRNAs are the most abundant RNA species in the plasma and differentially expressed in murine and human sepsis, such as miR-146a-5p. Exogenous miR-146a-5p, but not its duplex precursor, induces a strong immunostimulatory response through a newly identified UU-containing motif and TLR7 activation, and an immunotolerance by rapid IRAK-1 protein degradation via TLR7→MyD88 signaling and proteasome activation, whereas its duplex precursor acts by targeting 3' UTR of Irak-1 gene via Ago2 binding. miR-146a knockout in mice offers protection against sepsis with attenuated interleukin-6 (IL-6) storm and organ injury, improved cardiac function, and better survival. In septic patients, the plasma miR-146a-5p concentrations are closely associated with the two sepsis outcome predictors, blood lactate and coagulopathy. These data demonstrate the importance of extracellular miR-146a-5p in innate immune regulation and sepsis pathogenesis.

5.
Immunohorizons ; 4(9): 561-572, 2020 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-32958516

RESUMO

Previous studies have demonstrated that transient myocardial ischemia leads to release of cellular nucleic acids such as RNA. Extracellular RNA reportedly plays a pivotal role in myocardial inflammation and ischemic injury in animals. RNA profiling has identified that numerous microRNA (miRNAs), such as ss-miR-146a-5p, are upregulated in plasma following myocardial ischemia, and certain uridine-rich miRNAs exhibit strong proinflammatory effects in immune cells via ssRNA-sensing mechanism. However, the effect of extracellular miRNAs on myocardial inflammation and cardiac cell function remains unknown. In this study, we treated adult mouse cardiomyocytes with miR-146a-5p loaded in extracellular vesicles and observed a dose- and TLR7-dependent production of CXCL-2, IL-6, and TNF-α. In vivo, a single dose of myocardial injection of miR-146a-5p induced both cytokine expression (CXCL2, IL-6, and TNF-α) and innate immune cell activation (CD45+ leukocytes, Ly6Cmid+ monocytes, Ly6G+ neutrophils), which was significantly attenuated in the hearts of TLR7 KO mice. We discovered that conditioned media from miR-146a-treated macrophages stimulated proinflammatory cytokine production in adult cardiomyocytes and significantly inhibited their sarcomere shortening. Finally, using an electric cell impedance-sensing assay, we found that the conditioned media from miR-146a-treated cardiac fibroblasts or cardiomyocytes impaired the barrier function of coronary artery endothelial cells. Taken together, these data demonstrate that extracellular miR-146a-5p activates multiple cardiac cells and induces myocardial inflammation and cardiomyocyte dysfunction via intercellular interaction and innate immune TLR7 nucleic acid sensing.


Assuntos
Vasos Coronários/patologia , Vesículas Extracelulares/metabolismo , Imunidade Inata , Glicoproteínas de Membrana/metabolismo , MicroRNAs/metabolismo , Miócitos Cardíacos/patologia , Receptor 7 Toll-Like/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Citocinas/metabolismo , Células Endoteliais/metabolismo , Humanos , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Ratos , Ratos Sprague-Dawley , Receptor 7 Toll-Like/genética
6.
Mol Ther ; 28(3): 975-985, 2020 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-31911034

RESUMO

Based on their identification as physiological nucleic acid carriers in humans and other organisms, extracellular vesicles (EVs) have been explored as therapeutic delivery vehicles for DNA, RNA, and other cargo. However, efficient loading and functional delivery of nucleic acids remain a challenge, largely because of potential sources of degradation and aggregation. Here, we report that protonation of EVs to generate a pH gradient across EV membranes can be utilized to enhance vesicle loading of nucleic acid cargo, specifically microRNA (miRNA), small interfering RNA (siRNA), and single-stranded DNA (ssDNA). The loading process did not impair cellular uptake of EVs, nor did it promote any significant EV-induced toxicity response in mice. Cargo functionality was verified by loading HEK293T EVs with either pro- or anti-inflammatory miRNAs and observing the effective regulation of corresponding cellular cytokine levels. Critically, this loading increase is comparable with what can be accomplished by methods such as sonication and electroporation, and is achievable without the introduction of energy associated with these methods that can potentially damage labile nucleic acid cargo.


Assuntos
Vesículas Extracelulares/metabolismo , Concentração de Íons de Hidrogênio , MicroRNAs/metabolismo , Transporte Biológico , Vesículas Extracelulares/ultraestrutura , Células HEK293 , Humanos , MicroRNAs/genética , Ácidos Nucleicos/metabolismo
7.
Biomaterials ; 222: 119423, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31442885

RESUMO

Vascularization is a crucial process during the growth and development of bone 1, yet it remains one of the main challenges in the reconstruction of large bone defects. The use of in vitro coculture of human umbilical vein endothelial cells (HUVECs) and human mesenchymal stem cells (hMSCs) has been one of the most explored options. Both cell types secrete specific growth factors that are mutually beneficial, and studies suggested that cell-cell communication and paracrine secretion could be affected by a number of factors. However, little is known about the effect of cell patterning and the distance between cell populations on their crosstalk. In the present study, we showed that the separation and distance between ECs and MSCs populations affects angiogenesis by modulating cell-cell communication. HUVECs grown farther apart from MSCs (˃400 µm) presented characteristics of an early stage of angiogenesis (migration/proliferation). Results showed an increase in the up-regulation of VEGF, FGF-2, and ITGA3 (integrins) but a smaller fold change in the expression of VE-Cadherin and Ang-1. HUVECs were also still highly proliferative. On the contrary, HUVECs incubated closer (≤200 µm) to MSCs, showed signs of stabilization, mainly an increase in Ang-1 and VE-cadherin expression, as well as tighter monolayers. Conditioned media collected from HUVECs and MSCs grown ≤200 µm apart preferentially promoted tube formation, a later stage of angiogenesis, due in part to a significant increase in Ang-1 paracrine secretion. In addition, in groups in which fibers were printed farther apart (400 µm), cells produced EVs with a significantly increase cargo. Finally, in vivo experiment results showed an increase in blood vessels density and new bone thickness after 12 weeks of implantation in rat cranial defect, further suggesting the higher efficiency of indirect ECs/MSCs contact in prompting the release of paracrine signals that stimulate the angiogenesis of local tissues, and enhanced subsequent bone regeneration.


Assuntos
Técnicas de Cocultura/métodos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/metabolismo , Animais , Regeneração Óssea/genética , Regeneração Óssea/fisiologia , Comunicação Celular/genética , Comunicação Celular/fisiologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Masculino , Células-Tronco Mesenquimais/citologia , Neovascularização Fisiológica/genética , Neovascularização Fisiológica/fisiologia , Ratos
8.
J Tissue Eng Regen Med ; 13(11): 2031-2039, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31408915

RESUMO

One of the leading causes of death worldwide is heart failure. Despite advances in the treatment and prevention of heart failure, the number of affected patients continues to increase. We have recently developed 3D-bioprinted biomaterial-free cardiac tissue that has the potential to improve cardiac function. This study aims to evaluate the in vivo regenerative potential of these 3D-bioprinted cardiac patches. The cardiac patches were generated using 3D-bioprinting technology in conjunction with cellular spheroids created from a coculture of human-induced pluripotent stem cell-derived cardiomyocytes, fibroblasts, and endothelial cells. Once printed and cultured, the cardiac patches were implanted into a rat myocardial infarction model (n = 6). A control group (n = 6) without the implantation of cardiac tissue patches was used for comparison. The potential for regeneration was measured 4 weeks after the surgery with histology and echocardiography. 4 weeks after surgery, the survival rates were 100% and 83% in the experimental and the control group, respectively. In the cardiac patch group, the average vessel counts within the infarcted area were higher than those within the control group. The scar area in the cardiac patch group was significantly smaller than that in the control group. (Figure S1) Echocardiography showed a trend of improvement of cardiac function for the experimental group, and this trend correlated with increased patch production of extracellular vesicles. 3D-bioprinted cardiac patches have the potential to improve the regeneration of cardiac tissue and promote angiogenesis in the infarcted tissues and reduce the scar tissue formation.


Assuntos
Células Imobilizadas , Insuficiência Cardíaca , Células-Tronco Pluripotentes Induzidas , Miocárdio , Impressão Tridimensional , Regeneração , Alicerces Teciduais , Animais , Células Imobilizadas/metabolismo , Células Imobilizadas/patologia , Células Imobilizadas/transplante , Feminino , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/terapia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Células-Tronco Pluripotentes Induzidas/transplante , Ratos Endogâmicos Lew , Ratos Nus
9.
Tissue Eng Part C Methods ; 25(4): 243-252, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30913987

RESUMO

Ischemic cardiomyopathy poses a significant public health burden due to the irreversible loss of functional cardiac tissue. Alternative treatment strategies include creation of three-dimensional (3D) cardiac tissues to both replace and augment injured native tissue. In this study, we utilize a net mold-based method to create a biomaterial-free 3D cardiac tissue and compare it to current methods using biomaterials. Cardiomyocytes, fibroblasts, and endothelial cells were combined using a hanging drop method to create spheroids. For the net mold patch method, spheroids were seeded into a net mold-based system to create biomaterial-free 3D cardiac patches. For the gel patch, spheroids were embedded in a collagen gel. Immunohistochemistry revealed increased alignment, vascularization, collagen I expression, cell viability, and higher density of cells in the net mold patch compared with the gel patch. Furthermore, in vivo testing in a left anterior descending artery ligation rat model found increased ejection fraction and smaller scar area following implantation of the net mold patch. We present a novel and simple reproducible method to create biomaterial-free 3D net mold patches that may potentially improve the treatment of heart failure in the future.


Assuntos
Materiais Biocompatíveis/farmacologia , Coração/fisiologia , Engenharia Tecidual/métodos , Animais , Artérias/cirurgia , Linhagem Celular , Tamanho Celular , Sobrevivência Celular/efeitos dos fármacos , Colágeno/farmacologia , Eletrocardiografia , Exossomos/metabolismo , Feminino , Coração/diagnóstico por imagem , Coração/efeitos dos fármacos , Humanos , Ligadura , Ratos , Ratos Endogâmicos Lew , Ratos Nus , Esferoides Celulares/citologia
10.
J Immunol ; 201(11): 3392-3400, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30355788

RESUMO

We have previously reported that a group of host cellular microRNAs (miRNAs; miR-34a-5p, miR-122-5p, miR-145-5p, miR-146a-5p, miR-210-3p) are released into the blood during sepsis, some of which are capable of inducing complement activation, cytokine production, and leukocyte migration. Extracellular vesicles (EVs) have been proposed as vehicles for extracellular miRNA-mediated intercellular communication. However, the biological function of plasma EVs and the associated miRNAs in sepsis are largely unknown. In this study, we tested the hypothesis that plasma EVs in sepsis are proinflammatory and EV-associated miRNAs are responsible for EV-induced cytokine production. Compared with those of sham mice, the plasma EVs from septic mice were slightly smaller (157 ± 2 versus 191 ± 6 nm, p < 0.0001), but more abundant [(1.6 ± 0.14) × 1010 versus (0.93 ± 0.14) × 1010/ml plasma, p < 0.003]. miRNA array revealed that among 65 miRNAs, 8 miRNAs exhibited >1.5-fold increase in septic EVs compared with sham EVs, including miR-126-3p, miR-122-5p, miR-146a-5p, miR-145-5p, miR-26a-5p, miR-150-5p, miR-222-3p, and miR-181a-5p. Septic but not sham EVs were proinflammatory, promoting IL-6, TNF-α, IL-1ß, and MIP-2 production. The effects of EVs were resistant to polymyxin B (an endotoxin inhibitor) but significantly inhibited by anti-miR inhibitors against miR-34a, miR-122, and miR-146a. Moreover, the septic EV-induced cytokine production was attenuated in TLR7-/- or MyD88-/- cells but remained the same in TLR3-/- or Trif-/- cells. In vivo, mice i.p. injected with septic EVs had marked peritoneal neutrophil migration, which was significantly attenuated in MyD88-/- mice. Taken together, these data demonstrate that plasma EVs of septic animals play an important role in inflammation, and EV-associated miRNAs likely mediate the cytokine production via TLR7-MyD88 signaling.


Assuntos
Vesículas Extracelulares/imunologia , Inflamação/imunologia , Macrófagos/imunologia , Glicoproteínas de Membrana/metabolismo , MicroRNAs/genética , Sepse/imunologia , Receptor 7 Toll-Like/metabolismo , Animais , Circulação Sanguínea , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Inflamação/genética , Glicoproteínas de Membrana/genética , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Plasma/metabolismo , Polimixina B/metabolismo , Sepse/genética , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Receptor 7 Toll-Like/genética
11.
AAPS J ; 20(1): 1, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-29181730

RESUMO

Recently, extracellular vesicles (EVs)-including exosomes, microvesicles, and others-have attracted interest as cell-derived biotherapeutics and drug delivery vehicles for a variety of applications. This interest stems from favorable properties of EVs, including their status as mediators of cell-cell communication via transfer of biological cargo and their reported ability to cross biological barriers that impede many delivery systems. However, there are many challenges to translation and widespread application of EV-based therapeutics. One such challenge that has yet to be extensively studied involves EV preservation and storage, which must be addressed to enable use of therapeutic EVs beyond resource-intensive settings. Studies to date suggest that the most promising mode of storage is - 80°C; however, understanding of storage-mediated effects is still limited. Additionally, the effects of storage appear to vary with sample source. The lack of knowledge about and standardization of EV storage may ultimately hinder widespread clinical translation. This mini-review reports current knowledge in the field of EV preservation and storage stability and highlights future directions in the area that could be critical to eventual development of EV therapies.


Assuntos
Terapia Biológica/métodos , Sistemas de Liberação de Medicamentos , Vesículas Extracelulares , Exossomos , Humanos
12.
Cell Mol Bioeng ; 9(3): 315-324, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27800035

RESUMO

Extracellular vesicles (EVs), including exosomes and microvesicles, have emerged as promising drug delivery vehicles for small RNAs (siRNA and miRNA) due to their natural role in intercellular RNA transport. However, the application of EVs for therapeutic RNA delivery may be limited by loading approaches that can induce cargo aggregation or degradation. Here, we report the use of sonication as a means to actively load functional small RNAs into EVs. Conditions under which EVs could be loaded with small RNAs with minimal detectable aggregation were identified, and EVs loaded with therapeutic siRNA via sonication were observed to be taken up by recipient cells and capable of target mRNA knockdown leading to reduced protein expression. This system was ultimately applied to reduce expression of HER2, an oncogenic receptor tyrosine kinase that critically mediates breast cancer development and progression, and could be extended to other therapeutic targets. These results define important parameters informing the application of sonication as a small RNA loading method for EVs and demonstrate the potential utility of this approach for versatile cancer therapy.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...