Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Life Sci ; 356: 123014, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-39182566

RESUMO

AIMS: Parkinson's disease (PD) is a common neurodegenerative disease that has received widespread attention; however, current clinical treatments can only relieve its symptoms, and do not effectively protect dopaminergic neurons. The purpose of the present study was to investigate the therapeutic effects of human umbilical cord mesenchymal stem cell-derived exosomes loaded with brain-derived neurotrophic factor (BDNF-EXO) on PD models and to explore the underlying mechanisms of these effects. MAIN METHODS: 6-Hydroxydopamine was used to establish in vivo and in vitro PD models. Western blotting, flow cytometry, and immunofluorescence were used to detect the effects of BDNF-EXO on apoptosis and ferroptosis in SH-SY5Y cells. The in vivo biological distribution of BDNF-EXO was detected using a small animal imaging system, and dopaminergic neuron improvements in brain tissue were detected using western blotting, immunofluorescence, immunohistochemistry, and Nissl and Prussian blue staining. KEY FINDINGS: BDNF-EXO effectively suppressed 6-hydroxydopamine-induced apoptosis and ferroptosis in SH-SY5Y cells. Following intravenous administration, BDNF-EXO crossed the blood-brain barrier to reach afflicted brain regions in mice, leading to a notable enhancement in neuronal survival. Furthermore, BDNF-EXO modulated microtubule-associated protein 2 and phosphorylated tau expression, thereby promoting neuronal cytoskeletal stability. Additionally, BDNF-EXO bolstered cellular antioxidant defense mechanisms through the activation of the nuclear factor erythroid 2-related factor 2 signaling pathway, thereby conferring neuroprotection against damage. SIGNIFICANCE: The novel drug delivery system, BDNF-EXO, had substantial therapeutic effects in both in vivo and in vitro PD models, and may represent a new treatment strategy for PD.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Exossomos , Células-Tronco Mesenquimais , Doença de Parkinson , Cordão Umbilical , Exossomos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Humanos , Animais , Células-Tronco Mesenquimais/metabolismo , Cordão Umbilical/citologia , Camundongos , Doença de Parkinson/terapia , Doença de Parkinson/metabolismo , Modelos Animais de Doenças , Apoptose/efeitos dos fármacos , Oxidopamina , Masculino , Neurônios Dopaminérgicos/metabolismo , Neurônios Dopaminérgicos/patologia , Ferroptose/efeitos dos fármacos , Linhagem Celular Tumoral , Camundongos Endogâmicos C57BL
2.
ACS Biomater Sci Eng ; 10(5): 3069-3085, 2024 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-38578110

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder worldwide. Drug delivery to the brain through the blood-brain barrier (BBB) is a significant challenge in PD treatment. Exosomes, which can efficiently traverse the BBB, which many drugs cannot penetrate, are ideal natural carriers for drug delivery. In this study, the BBB shuttle peptide was modified on the exosome surfaces. Three types of exosomes were constructed, each modified with a distinct peptide (RVG29, TAT, or Ang2) and loaded with miR-133b. The safety and brain-targeting capabilities of these peptide-modified exosomes were then evaluated. Finally, the mechanism by which RVG29-Exo-133b regulates the RhoA-ROCK signaling pathway was investigated. The findings indicate that the three peptide-modified exosomes were adequately tolerated, safe, and effectively assimilated in vivo and ex vivo, with RVG29 exhibiting superior targeting to the brain. Furthermore, RVG29-Exo-133b decreased the phosphorylation level of the Tau protein by targeting the RhoA-ROCK signaling pathway. It also enhanced the motor function in mice with PD, thereby reducing the degree of depression, improving dopaminergic neuron function, and attenuating 6-OHDA-induced nerve damage. In this study, we developed a stable drug delivery mechanism that targets the intracerebral region using exosomes. Furthermore, a novel strategy was developed to manage PD and can potentially serve as a preclinical basis for utilizing exosomes in the diagnosis and treatment of neurodegenerative conditions.


Assuntos
Exossomos , MicroRNAs , Doença de Parkinson , Transdução de Sinais , Quinases Associadas a rho , Proteína rhoA de Ligação ao GTP , Exossomos/metabolismo , Animais , Quinases Associadas a rho/metabolismo , Quinases Associadas a rho/genética , MicroRNAs/metabolismo , MicroRNAs/genética , Doença de Parkinson/metabolismo , Doença de Parkinson/genética , Proteína rhoA de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Camundongos , Masculino , Camundongos Endogâmicos C57BL , Humanos , Peptídeos/metabolismo , Barreira Hematoencefálica/metabolismo
3.
Neurochem Int ; 175: 105700, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38417589

RESUMO

Currently, there is no effective treatment for Parkinson's disease (PD), and the regenerative treatment of neural stem cells (NSCs) is considered the most promising method. This study aimed to investigate the protective effect and mechanism of NSCs on neurons in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) induced cynomolgus monkey (Macaca fascicularis) model of PD. We first found that injecting NSCs into the subarachnoid space relieved motor dysfunction in PD cynomolgus monkeys, as well as reduced dopaminergic neuron loss and neuronal damage in the substantia nigra (SN) and striatum. Besides, NSCs decreased 17-estradiol (E2) level, an estrogen, in the cerebrospinal fluid (CSF) of PD cynomolgus monkeys, which shows NSCs may provide neuro-protection by controlling estrogen levels in the CSF. Furthermore, NSCs elevated proliferator-activated receptor gamma coactivator-1 alpha (PGC-1a), mitofusin 2 (MFN2), and optic atrophy 1 (OPA1) expression, three genes mediating mitochondrial biogenesis, in the SN and striatum of PD monkeys. In addition, NSCs suppress reactive oxygen species (ROS) production caused by MPTP, as well as mitochondrial autophagy, therefore preserving dopaminergic neurons. In summary, our findings show that NSCs may preserve dopaminergic and neuronal cells in an MPTP-induced PD cynomolgus monkey model. These protective benefits might be attributed to NSCs' ability of modulating estrogen balance, increasing mitochondrial biogenesis, and limiting oxidative stress and mitochondrial autophagy. These findings add to our understanding of the mechanism of NSC treatment and shed light on further clinical treatment options.


Assuntos
Intoxicação por MPTP , Células-Tronco Neurais , Doença de Parkinson , Animais , Humanos , Macaca fascicularis/metabolismo , Intoxicação por MPTP/terapia , Intoxicação por MPTP/metabolismo , Células-Tronco Neurais/metabolismo , Doença de Parkinson/metabolismo , Neurônios Dopaminérgicos , Dopamina/metabolismo , Estrogênios/farmacologia
4.
Int J Nanomedicine ; 19: 4923-4939, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38828201

RESUMO

Purpose: In recent years, exosomes have been proved to be used to treat many diseases. However, due to the lack of uniform quality control standards for exosomes, the safety of exosomes is still a problem to be solved, especially now more and more exosomes are used in clinical trials, and its non-clinical safety evaluation is particularly important. However, there is no safety evaluation standard for exosomes at present. Therefore, this study will refer to the evaluation criteria of therapeutic biological products, adopt non-human primates to evaluate the non-clinical safety of human umbilical cord mesenchymal stem cell exosomes from the general pharmacology and immunotoxicity, aiming at establishing a safety evaluation system of exosomes and providing reference for the clinical application of exosomes in the future. Methods: 3.85 × 1012 exosomes derived from human umbilical cord mesenchymal stem cells were injected into cynomolgus monkeys intravenously. The changes of general clinical conditions, hematology, immunoglobulin, Th1/Th2 cytokines, T lymphocytes and B lymphocytes, and immune organs were observed before and within 14 days after injection. Results: The results showed that exosomes did not have obvious pathological effects on the general clinical conditions, blood, coagulation function, organ coefficient, immunoglobulin, Th1/Th2 cytokines, lymphocytes, major organs, and major immune organs (spleen, thymus, bone marrow) of cynomolgus monkeys. However, the number of granulocyte-macrophage colonies in exosomes group was significantly higher than that in control group. Conclusion: To sum up, the general pharmacological results and immunotoxicity results showed that the injection of 3.85 × 1012 exosomes may have no obvious adverse reactions to cynomolgus monkeys. This dose of exosomes is relatively safe for treatment, which provides basis research for non-clinical safety evaluation of exosomes and provides reliable research basis for future clinical application of exosomes.


Assuntos
Exossomos , Macaca fascicularis , Células-Tronco Mesenquimais , Cordão Umbilical , Animais , Exossomos/química , Células-Tronco Mesenquimais/citologia , Humanos , Cordão Umbilical/citologia , Masculino , Feminino , Citocinas/metabolismo
5.
Am J Transl Res ; 15(5): 3026-3039, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37303663

RESUMO

OBJECTIVES: Ischemia-reperfusion injury is a complicated pathologic process that involves multiple factors including oxidative stress, endoplasmic reticulum stress, calcium overload, inflammatory response, disturbances in energy metabolism, apoptosis, and some newly-described forms of programmed cell death (e.g., necroptosis, autophagy, pyroptosis, patanatos, and ferroptosis). Chinese herbal monomers (CHMs) have long been applied to treat ischemia-reperfusion injury based on a solid research foundation. This paper objectively reviews in vitro and in vivo studies on the use of CHMs to protect against ischemia-reperfusion injury. METHODS: We reviewed 31 CHMs that have been shown to be effective for treating ischemia-reperfusion injury models of the heart, brain, and kidney. According to the mechanism of action, these CHMs were divided into three categories: protecting damaged histocytes, inhibiting inflammatory cells, and promoting the proliferation of damaged histocytes. Some CHMs were found to have more than one mechanism at the same time. RESULTS: Of the 31 CHMs, 28 protect damaged histocytes, 13 inhibit inflammatory cells, and three promote the proliferation of damaged histocytes. CONCLUSIONS: CHMs show promise for treating ischemia-reperfusion injury. The eexisting treatment experiences for ischemia-reperfusion injury can be used as a reference.

6.
Bioanalysis ; 14(24): 1547-1561, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36734464

RESUMO

Background: Extracellular vesicles (EVs) are important carriers of intercellular communication, used in disease diagnosis and as prognostic circulating biomarkers, and their identification and quantitative analysis are important prerequisites for their clinical application. Methods & results: A method using microchip electrophoresis with contactless conductivity detection was developed for the concentration assay of EVs. This method showed good sensitivity, reproducibility and accuracy, with good linear correlation with conventional methods (nanoparticle tracking analysis and bicinchoninic acid assay). The application to the detection of mesenchymal stem cell-derived EVs proved its applicability to clinical samples. Conclusion: This is the first study to apply this method for the detection of EVs, achieving quantitative analysis of EVs enriched in exosomes and microvesicles, and initially demonstrating the potential to separate different EV subpopulations.


Assuntos
Micropartículas Derivadas de Células , Eletroforese em Microchip , Exossomos , Vesículas Extracelulares , Reprodutibilidade dos Testes
7.
Front Bioeng Biotechnol ; 10: 949724, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36091443

RESUMO

As a new cell-free therapy, exosomes have provided new ideas for the treatment of various diseases. Human induced pluripotent stem cells (hiPSCs) cannot be used in clinical trials because of tumorigenicity, but the exosomes derived from hiPSCs may combine the advantages of iPSC pluripotency and the nanoscale size of exosomes while avoiding tumorigenicity. Currently, the safety and biodistribution of hiPSC-exosomes in vivo are unclear. Here, we investigated the effects of hiPSC-exosomes on hemolysis, DNA damage, and cytotoxicity through cell experiments. We also explored the safety of vein injection of hiPSC-exosomes in rabbits and rats. Differences in organ distribution after nasal administration were compared in normal and Parkinson's disease model mice. This study may provide support for clinical therapy and research of intravenous and nasal administration of hiPSC-exosomes.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA