Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Adv Sci (Weinh) ; 11(17): e2306577, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38441409

RESUMO

Spinal cord injury (SCI) leads to massive cell death, disruption, and demyelination of axons, resulting in permanent motor and sensory dysfunctions. Stem cell transplantation is a promising therapy for SCI. However, owing to the poor microenvironment that develops following SCI, the bioactivities of these grafted stem cells are limited. Cell implantation combined with biomaterial therapies is widely studied for the development of tissue engineering technology. Herein, an insulin-like growth factor-1 (IGF-1)-bioactive supramolecular nanofiber hydrogel (IGF-1 gel) is synthesized that can activate IGF-1 downstream signaling, prevent the apoptosis of neural stem cells (NSCs), improve their proliferation, and induce their differentiation into neurons and oligodendrocytes. Moreover, implantation of NSCs carried out with IGF-1 gels promotes neurite outgrowth and myelin sheath regeneration at lesion sites following SCI. In addition, IGF-1 gels can enrich extracellular vesicles (EVs) derived from NSCs or from nerve cells differentiated from these NSCs via miRNAs related to axonal regeneration and remyelination, even in an inflammatory environment. These EVs are taken up by autologous endogenous NSCs and regulate their differentiation. This study provides adequate evidence that combined treatment with NSCs and IGF-1 gels is a potential therapeutic strategy for treating SCI.


Assuntos
Hidrogéis , Fator de Crescimento Insulin-Like I , Nanofibras , Células-Tronco Neurais , Traumatismos da Medula Espinal , Animais , Ratos , Diferenciação Celular , Modelos Animais de Doenças , Hidrogéis/química , Fator de Crescimento Insulin-Like I/metabolismo , Nanofibras/química , Nanofibras/uso terapêutico , Regeneração Nervosa/efeitos dos fármacos , Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/terapia , Transplante de Células-Tronco/métodos , Feminino
2.
Tissue Eng Part C Methods ; 30(2): 73-84, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-37930732

RESUMO

Intervertebral disc degeneration (IVDD) is a major cause of low back pain, and several studies have evaluated the efficacy of extracellular vesicles (EVs) in the treatment of IVDD. The databases PubMed, Embase, and Cochrane Library were systematically searched from inception to the end of 2022 to identify studies investigating the therapeutic potential of cell-derived EVs for IVDD treatment. The following outcome measures were utilized: magnetic resonance imaging (MRI) Pfirrmann grading system, disc height index (DHI), histological grading, and apoptosis rate. A comprehensive meta-analysis was conducted, including a total of 13 articles comprising 19 studies involving 218 experimental animals. Comparative analysis between normal cell-derived EVs and placebo revealed significant reductions in MRI grade, increased DHI values, decreased nucleus pulposus cell apoptosis rates, and improved tissue grades. These findings collectively demonstrate the effective inhibition of IVDD through the application of EVs derived from cells. In conclusion, this study provides an updated synthesis of evidence supporting the efficacy of EVs as a promising therapeutic approach for IVDD treatment.


Assuntos
Vesículas Extracelulares , Degeneração do Disco Intervertebral , Disco Intervertebral , Núcleo Pulposo , Animais , Degeneração do Disco Intervertebral/terapia , Degeneração do Disco Intervertebral/patologia , Imageamento por Ressonância Magnética , Apoptose , Disco Intervertebral/diagnóstico por imagem , Disco Intervertebral/patologia
3.
Stem Cell Rev Rep ; 19(6): 2024-2037, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37256514

RESUMO

Cell‒cell communication following spinal cord injury (SCI) plays a key role in remyelination and neurological recovery. Although communication between neuron-neural stem cells (NSCs) affects remyelination, its precise mechanism remains unclear. The present study investigated the biological effects of extracellular vesicles (EVs) derived from neurons on the differentiation of NSCs and the remyelination of axons in a rat model for SCI. We found that that EVs derived from neurons promoted the differentiation of NSCs into oligodendrocytes and the remyelination of axons in SCI rats. However, neuron-derived EVs lost their biological effects after inflammatory stimulation of these neurons from which they originate. Further analysis demonstrated that the inflammatory stimulation on neurons upregulated miR-21 within EVs, which targeted SMAD 7 and upregulated the TGF-ß/SMAD2 signaling pathway, resulting in an excess of astrocytic scar boundaries and in remyelination failure. Moreover, these effects could be abolished by miR-21 inhibitors/antagomirs. Considered together, these results indicate that inflammatory stimulation of neurons prevents remyelination following SCI via the upregulation of miR-21 expression within neuron-derived EVs, and this takes place through SMAD 7-mediated activation of the TGF-ß/SMAD2 signaling pathway. Graphical Astract.


Assuntos
Vesículas Extracelulares , MicroRNAs , Remielinização , Traumatismos da Medula Espinal , Ratos , Animais , Neurônios/metabolismo , Traumatismos da Medula Espinal/genética , MicroRNAs/genética , MicroRNAs/metabolismo , Vesículas Extracelulares/metabolismo , Inflamação/genética , Inflamação/metabolismo , Fator de Crescimento Transformador beta/metabolismo
4.
Stem Cell Res Ther ; 14(1): 52, 2023 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-36959678

RESUMO

BACKGROUND: Endogenous neural stem cells (NSCs) are critical for the remyelination of axons following spinal cord injury (SCI). Cell-cell communication plays a key role in the regulation of the differentiation of NSCs. Astrocytes act as immune cells that encounter early inflammation, forming a glial barrier to prevent the spread of destructive inflammation following SCI. In addition, the cytokines released from astrocytes participate in the regulation of the differentiation of NSCs. The aim of this study was to investigate the effects of cytokines released from inflammation-stimulated astrocytes on the differentiation of NSCs following SCI and to explore the influence of these cytokines on NSC-NSC communication. RESULTS: Lipopolysaccharide stimulation of astrocytes increased bone morphogenetic protein 2 (BMP2) release, which not only promoted the differentiation of NSCs into astrocytes and inhibited axon remyelination in SCI lesions but also enriched miRNA-22-3p within extracellular vesicles derived from NSCs. These miRNA-22 molecules function as a feedback loop to promote NSC differentiation into oligodendrocytes and the remyelination of axons following SCI by targeting KDM3A. CONCLUSIONS: This study revealed that by releasing BMP2, astrocytes were able to regulate the differentiation of NSCs and NSC-NSC communication by enriching miRNA-22 within NSC-EVs, which in turn promoted the regeneration and remyelination of axons by targeting the KDM3A/TGF-beta axis and the recovery of neurological outcomes following SCI.


Assuntos
MicroRNAs , Células-Tronco Neurais , Remielinização , Traumatismos da Medula Espinal , Humanos , Astrócitos/metabolismo , Células-Tronco Neurais/metabolismo , Diferenciação Celular/fisiologia , Traumatismos da Medula Espinal/patologia , Inflamação/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Histona Desmetilases com o Domínio Jumonji/genética , Histona Desmetilases com o Domínio Jumonji/metabolismo
5.
Sci Rep ; 12(1): 8154, 2022 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-35581219

RESUMO

Ewing sarcoma (ES) is a rare disease that lacks a prognostic prediction model. This study aims to develop a nomogram and risk classification system for estimating the probability of overall survival (OS) of patients with ES. The clinicopathological data of ES were collected from the Surveillance, Epidemiology and Final Results (SEER) database from 2010 to 2018. The primary cohort was randomly assigned to the training set and the validation set. Univariate and multiple Cox proportional hazard analyses based on the training set were performed to identify independent prognostic factors. A nomogram was established to generate individualized predictions of 3- and 5-year OS and evaluated by the concordance index (C-index), the receiver operating characteristic curve (ROC), the calibration curve, the integrated discrimination improvement (IDI) and the net reclassification improvement (NRI). Based on the scores calculated with the nomogram, ES patients were divided into three risk groups to predict their survival. A total of 935 patients were identified, and a nomogram consisting of 6 variables was established. The model provided better C-indices of OS (0.788). The validity of the Cox model assumptions was evaluated through the Schönfeld test and deviance residual. The ROC, calibration curve, IDI and NRI indicated that the nomogram exhibited good performance. A risk classification system was built to classify the risk group of ES patients. The nomogram compares favourably and accurately to the traditional SEER tumour staging systems, and risk stratification provides a more convenient and effective tool for clinicians to optimize treatment options.


Assuntos
Tumores Neuroectodérmicos Primitivos Periféricos , Sarcoma de Ewing , Humanos , Nomogramas , Prognóstico , Fatores de Risco , Programa de SEER
6.
Front Mol Neurosci ; 15: 850364, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35401112

RESUMO

Spinal cord injury (SCI) often causes neuronal and axonal damage, resulting in permanent neurological impairments. Mesenchymal stem cells (MSCs) and extracellular vesicles (EVs) are promising treatments for SCI. However, the underlying mechanisms remain unclear. Herein, we demonstrated that EVs from bone marrow-derived MSCs promoted the differentiation of neural stem cells (NSCs) into the neurons and outgrowth of neurites that are extending into astrocytic scars in SCI rats. Further study found that let-7a-5p exerted a similar biological effect as MSC-EVs in regulating the differentiation of NSCs and leading to neurological improvement in SCI rats. Moreover, these MSC-EV-induced effects were attenuated by let-7a-5p inhibitors/antagomirs. When investigating the mechanism, bioinformatics predictions combined with western blot and RT-PCR analyses showed that both MSC-EVs and let-7a-5p were able to downregulate the expression of SMAD2 by inhibiting HMGA2. In conclusion, MSC-EV-secreted let-7a-5p promoted the regrowth of neurons and improved neurological recovery in SCI rats by targeting the HMGA2/SMAD2 axis.

7.
Stem Cell Rev Rep ; 18(3): 1078-1096, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34449013

RESUMO

Mesenchymal stem cells (MSCs) constitute a promising therapy for spinal cord injury (SCI) because they can provide a favorable environment for the regrowth of neurons by inhibiting receptor-regulated Smads (R-Smads) expression in endogenous neural stem cells (NSCs). However, their mechanism of action and effect on the expression of inhibitory Smads (I-Smads) remain unclear. Herein, we demonstrated that extracellular vesicles (EVs) from MSCs were able to upregulate the Smad 6 expression by carrying TGF-ß, and the Smad 6 knockdown in NSCs partially weakened the bone marrow MSC (BMSC)-EV-induced effect on neural differentiation. We found that the expression of Smad 6 did not reduced owing to the TGF-ß type I receptor kinase inhibitor, SB 431,542, treatment in the acute phase of injury in rats with SCI, thereby indicating that the Smad 6 expression was not only mediated by TGF-ß, but also by the inflammatory factors and bone morphogenetic proteins (BMPs) as well. However, in the later phase of SCI, the Smad 6 expression decreased by the addition of SB 431,542, suggesting that TGF-ß plays a key role in the mediation of Smad 6 expression in this phase. In addition, immunohistochemistry staining; hematoxylin-eosin staining; and the Basso, Beattie, and Bresnahan (BBB) scores revealed that the early inhibition of TGF-ß did not increase neuron regrowth. However, this inhibition increased the cavity and the caspase-3 expression at 24 h post-injury, leading to a worse functional outcome. Conversely, the later treatment with the TGF-ß inhibitor promoted the regrowth of neurons around the cavity, resulting in a better neurological outcome. Together, these results indicate that Smad 6 acts as a feedback regulator to prevent the over-differentiation of NSCs to astrocytes and that BMSC-EVs can upregulate Smad 6 expression by carrying TGF-ß.


Assuntos
Vesículas Extracelulares , Células-Tronco Mesenquimais , Proteína Smad6 , Traumatismos da Medula Espinal , Animais , Vesículas Extracelulares/metabolismo , Neurônios , Ratos , Proteína Smad6/genética , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/terapia , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...