Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Res Ther ; 15(1): 95, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38566259

RESUMO

BACKGROUND: Human adipose stromal cells-derived extracellular vesicles (haMSC-EVs) have been shown to alleviate inflammation in acute lung injury (ALI) animal models. However, there are few systemic studies on clinical-grade haMSC-EVs. Our study aimed to investigate the manufacturing, quality control (QC) and preclinical safety of clinical-grade haMSC-EVs. METHODS: haMSC-EVs were isolated from the conditioned medium of human adipose MSCs incubated in 2D containers. Purification was performed by PEG precipitation and differential centrifugation. Characterizations were conducted by nanoparticle tracking analysis, transmission electron microscopy (TEM), Western blotting, nanoflow cytometry analysis, and the TNF-α inhibition ratio of macrophage [after stimulated by lipopolysaccharide (LPS)]. RNA-seq and proteomic analysis with liquid chromatography tandem mass spectrometry (LC-MS/MS) were used to inspect the lot-to-lot consistency of the EV products. Repeated toxicity was evaluated in rats after administration using trace liquid endotracheal nebulizers for 28 days, and respiratory toxicity was evaluated 24 h after the first administration. In vivo therapeutic effects were assessed in an LPS-induced ALI/ acute respiratory distress syndrome (ARDS) rat model. RESULTS: The quality criteria have been standardized. In a stability study, haMSC-EVs were found to remain stable after 6 months of storage at - 80°C, 3 months at - 20 °C, and 6 h at room temperature. The microRNA profile and proteome of haMSC-EVs demonstrated suitable lot-to-lot consistency, further suggesting the stability of the production processes. Intratracheally administered 1.5 × 108 particles/rat/day for four weeks elicited no significant toxicity in rats. In LPS-induced ALI/ARDS model rats, intratracheally administered haMSC-EVs alleviated lung injury, possibly by reducing the serum level of inflammatory factors. CONCLUSION: haMSC-EVs, as an off-shelf drug, have suitable stability and lot-to-lot consistency. Intratracheally administered haMSC-EVs demonstrated excellent safety at the tested dosages in systematic preclinical toxicity studies. Intratracheally administered haMSC-EVs improved the lung function and exerted anti-inflammatory effects on LPS-induced ALI/ARDS model rats.


Assuntos
Lesão Pulmonar Aguda , Vesículas Extracelulares , Células-Tronco Mesenquimais , Síndrome do Desconforto Respiratório , Humanos , Ratos , Animais , Cromatografia Líquida , Proteômica , Lipopolissacarídeos/farmacologia , Espectrometria de Massas em Tandem , Lesão Pulmonar Aguda/terapia , Síndrome do Desconforto Respiratório/terapia , Obesidade , Controle de Qualidade , Vesículas Extracelulares/fisiologia , Células-Tronco Mesenquimais/fisiologia
2.
Acta Pharm Sin B ; 14(3): 1412-1427, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38486994

RESUMO

Mesenchymal stem cells (MSCs) experience substantial viability issues in the stroke infarct region, limiting their therapeutic efficacy and clinical translation. High levels of deadly reactive oxygen radicals (ROS) and proinflammatory cytokines (PC) in the infarct milieu kill transplanted MSCs, whereas low levels of beneficial ROS and PC stimulate and improve engrafted MSCs' viability. Based on the intrinsic hormesis effects in cellular biology, we built a microglia-inspired MSC bioengineering system to transform detrimental high-level ROS and PC into vitality enhancers for strengthening MSC therapy. This system is achieved by bioorthogonally arming metabolic glycoengineered MSCs with microglial membrane-coated nanoparticles and an antioxidative extracellular protective layer. In this system, extracellular ROS-scavenging and PC-absorbing layers effectively buffer the deleterious effects and establish a micro-livable niche at the level of a single MSC for transplantation. Meanwhile, the infarct's inanimate milieu is transformed at the tissue level into a new living niche to facilitate healing. The engineered MSCs achieved viability five times higher than natural MSCs at seven days after transplantation and exhibited a superior therapeutic effect for stroke recovery up to 28 days. This vitality-augmented system demonstrates the potential to accelerate the clinical translation of MSC treatment and boost stroke recovery.

3.
J Control Release ; 362: 210-224, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37619863

RESUMO

Mesenchymal stem cells (MSCs) exhibited remarkable therapeutic potential in ischemic stroke due to their exceptional immunomodulatory ability and paracrine effect; they have also been regarded as excellent neuroprotectant delivery vehicles with inflammatory tropism. However, the presence of high levels of reactive oxygen species (ROS) and an oxidative stress environment at the lesion site inhibits cell survival and further therapeutic effects. Using bioorthogonal click chemistry, ROS-responsive luteolin-loaded micelles were tethered to the surface of MSCs. As MSCs migrated to the ischemic brain, the micelles would achieve ROS-responsive release of luteolin to protect MSCs from excessive oxidative damage while inhibiting neuroinflammation and scavenging ROS to ameliorate ischemic stroke. This study provided an effective and prospective therapeutic strategy for ischemic stroke and a framework for a stem cell-based therapeutic system to treat inflammatory cerebral diseases.

4.
ACS Nano ; 17(9): 8646-8662, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37099675

RESUMO

Apoptotic vesicles (ApoVs) hold great promise for inflammatory regulation and tissue repair. However, little effort has been dedicated to developing ApoV-based drug delivery platforms, while the insufficient targeting capability of ApoVs also limits their clinical applications. This work presents a platform architecture that integrates apoptosis induction, drug loading, and functionalized proteome regulation, followed by targeting modification, enabling the creation of an apoptotic vesicle delivery system to treat ischemic stroke. Briefly, α-mangostin (α-M) was utilized to induce mesenchymal stem cell (MSC) apoptosis while being loaded onto MSC-derived ApoVs as an anti-oxidant and anti-inflammatory agent for cerebral ischemia/reperfusion injury. Matrix metalloproteinase activatable cell-penetrating peptide (MAP), a microenvironment-responsive targeting peptide, was modified on the surface of ApoVs to obtain the MAP-functionalized α-M-loaded ApoVs. Such engineered ApoVs targeted the injured ischemic brain after systemic injection and achieved an enhanced neuroprotective activity due to the synergistic effect of ApoVs and α-M. The internal protein payloads of ApoVs, upon α-M activation, were found engaged in regulating immunological response, angiogenesis, and cell proliferation, all of which contributed to the therapeutic effects of ApoVs. The findings provide a universal framework for creating ApoV-based therapeutic drug delivery systems for the amelioration of inflammatory diseases and demonstrate the potential of MSC-derived ApoVs to treat neural injury.


Assuntos
AVC Isquêmico , Traumatismo por Reperfusão , Acidente Vascular Cerebral , Humanos , AVC Isquêmico/tratamento farmacológico , Encéfalo , Isquemia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Traumatismo por Reperfusão/tratamento farmacológico , Acidente Vascular Cerebral/tratamento farmacológico
5.
J Extracell Vesicles ; 10(10): e12134, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34429860

RESUMO

Mesenchymal stromal cell-derived extracellular vesicles (MSC-EVs) turn out to be a promising source of cell-free therapy. Here, we investigated the biodistribution and effect of nebulized human adipose-derived MSC-EVs (haMSC-EVs) in the preclinical lung injury model and explored the safety of nebulized haMSC-EVs in healthy volunteers. DiR-labelled haMSC-EVs were used to explore the distribution of nebulized haMSC-EVs in the murine model. Pseudomonas aeruginosa-induced murine lung injury model was established, and survival rate, as well as WBC counts, histology, IL-6, TNF-α and IL-10 levels in bronchoalveolar lavage fluid (BALF) were measured to explore the optimal therapeutic dose of haMSC-EVs through the nebulized route. Twenty-four healthy volunteers were involved and received the haMSC-EVs once, ranging from 2 × 108 particles to 16 × 108 particles (MEXVT study, NCT04313647). Nebulizing haMSC-EVs improved survival rate to 80% at 96 h in P. aeruginosa-induced murine lung injury model by decreasing lung inflammation and histological severity. All volunteers tolerated the haMSC-EVs nebulization well, and no serious adverse events were observed from starting nebulization to the 7th day after nebulization. These findings suggest that nebulized haMSC-EVs could be a promising therapeutic strategy, offering preliminary evidence to promote the future clinical applications of nebulized haMSC-EVs in lung injury diseases.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Citocinas/metabolismo , Avaliação Pré-Clínica de Medicamentos , Vesículas Extracelulares/fisiologia , Lesão Pulmonar/terapia , Células-Tronco Mesenquimais/fisiologia , Adolescente , Adulto , Animais , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Feminino , Humanos , Lesão Pulmonar/microbiologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Segurança do Paciente , Infecções por Pseudomonas/microbiologia , Pseudomonas aeruginosa , Taxa de Sobrevida , Terapêutica/métodos , Adulto Jovem
6.
Adv Sci (Weinh) ; 7(19): 2000963, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33042738

RESUMO

It has been reported that Cavin1 deficiency causes lipodystrophy in both humans and mice by affecting lipid metabolism. The ablation of Cavin1 in rodents also causes a significant deviation from Mendelian ratio at weaning in a background-dependent manner, suggesting the presence of undiscovered functions of Cavin1. In the current study, the results show that Cavin1 deficiency causes neonatal death in C57BL/6J mice by dampening the storage and mobilization of glycogen in the liver, which leads to lethal neonatal hypoglycemia. Further investigation by electron microscopy reveals that Cavin1 deficiency impairs the fenestration in liver sinusoidal endothelial cells (LSECs) and impacts the permeability of endothelial barrier in the liver. Mechanistically, Cavin1 deficiency inhibits the RhoA-Rho-associated protein kinase 2-LIM domain kinase-Cofilin signaling pathway and suppresses the dynamics of the cytoskeleton, and eventually causes the reduction of fenestrae in LSECs. In addition, the defect of fenestration in LSECs caused by Cavin1 deficiency can be rescued by treatment with the F-actin depolymerization reagent latrunculin A. In summary, the current study reveals a novel function of Cavin1 on fenestrae formation in LSECs and liver glycogen metabolism, which provide an explanation for the neonatal death of Cavin1 null mice and a potential mechanism for metabolic disorders in patients with Cavin1 mutation.

7.
Acta Biochim Biophys Sin (Shanghai) ; 50(10): 968-975, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30188967

RESUMO

Fam198a is a member of four-jointed protein kinases, a secreted protein kinase family. It was identified as a caveolae-associated protein and colocalized with cavin-1 and caveolin-1 in both tissues and cells. The newly synthesized Fam198a precursor in endoplasmic reticulum (ER) was transported by caveolae biogenesis vesicles to Golgi apparatus in which it was proteolytically cleaved into the secreted mature form. The amino acid mutation analysis identified Arg 120 and 437 as the proteolytic sites in Fam198a precursor during maturation. In mouse embryo fibroblasts (MEFs) obtained from cavin-1-/- or caveolin-1-/- mice, Fam198a precursor was retained in ER and no mature Fam198a could be formed in these cells. Ectopic expression of exogenous cavin-1 in cavin-1-/- MEFs restored the blocked Fam198a post-translational process and secretion. Cavin-1 was also required for Fam198a secretion after its maturation in Golgi apparatus. Ectopic expression of cavin-1 in A549 cells restored the blocked Fam198a secretion. These results suggest that protein secretion is an important function for caveolae biogenesis pathway and the disruption of caveolae system will affect those functions played by the secreted proteins.


Assuntos
Cavéolas/metabolismo , Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Quinases/metabolismo , Células 3T3-L1 , Células A549 , Animais , Vias Biossintéticas/genética , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Embrião de Mamíferos/citologia , Fibroblastos/metabolismo , Células HeLa , Células Hep G2 , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas Quinases/genética , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transporte Proteico , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo
8.
Biochem Biophys Res Commun ; 456(3): 750-6, 2015 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-25514038

RESUMO

PTRF/cavin-1 is a protein of two lives. Its reported functions in ribosomal RNA synthesis and in caveolae formation happen in two different cellular locations: nucleus vs. plasma membrane. Here, we identified that the N-terminal leucine-zipper motif in PTRF/cavin-1 was essential for the protein to be associated with caveolae in plasma membrane. It could counteract the effect of nuclear localization sequence in the molecule (AA 235-251). Deletion of this leucine-zipper motif from PTRF/cavin-1 caused the mutant to be exclusively localized in nuclei. The fusion of this leucine-zipper motif with histone 2A, which is a nuclear protein, could induce the fusion protein to be exported from nucleus. Cell migration was greatly inhibited in PTRF/cavin-1(-/-) mouse embryonic fibroblasts (MEFs). The inhibited cell motility could only be rescued by exogenous cavin-1 but not the leucine-zipper motif deleted cavin-1 mutant. Plasma membrane dynamics is an important factor in cell motility control. Our results suggested that the membrane dynamics in cell migration is affected by caveolae associated PTRF/cavin-1.


Assuntos
Cavéolas/enzimologia , Zíper de Leucina/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Ligação a RNA/fisiologia , Células 3T3-L1 , Animais , Células CHO , Células COS , Movimento Celular , Cricetulus , Zíper de Leucina/genética , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Fosforilação , Mutação Puntual , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/genética , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...