Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Hepatology ; 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38546278

RESUMO

BACKGROUND AND AIMS: The immunomodulatory characteristics of mesenchymal stem cells (MSCs) make them a promising therapeutic approach for liver fibrosis (LF). Here, we postulated that MSCs could potentially suppress the pro-fibrotic activity of intrahepatic B cells, thereby inhibiting LF progression. APPROACH AND RESULTS: Administration of MSCs significantly ameliorated LF as indicated by reduced myofibroblast activation, collagen deposition, and inflammation. The treatment efficacy of MSCs can be attributed to decreased infiltration, activation, and pro-inflammatory cytokine production of intrahepatic B cells. Single-cell RNA sequencing revealed a distinct intrahepatic B cell atlas, and a subtype of naive B cells (B-II) was identified, which were markedly abundant in fibrotic liver, displaying mature features with elevated expression of several proliferative and inflammatory genes. Transcriptional profiling of total B cells revealed that intrahepatic B cells displayed activation, proliferation, and pro-inflammatory gene profile during LF. Fibrosis was attenuated in mice ablated with B cells (µMT) or in vivo treatment with anti-CD20. Moreover, fibrosis was recapitulated in µMT after adoptive transfer of B cells, which in turn could be rescued by MSC injection, validating the pathogenic function of B cells and the efficacy of MSCs on B cell-promoted LF progression. Mechanistically, MSCs could inhibit the proliferation and cytokine production of intrahepatic B cells through exosomes, regulating the Mitogen-activated protein kinase and Nuclear factor kappa B signaling pathways. CONCLUSIONS: Intrahepatic B cells serve as a target of MSCs, play an important role in the process of MSC-induced amelioration of LF, and may provide new clues for revealing the novel mechanisms of MSC action.

2.
Cell Mol Life Sci ; 81(1): 124, 2024 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-38466420

RESUMO

Acute lung injury (ALI) is an inflammatory disease associated with alveolar injury, subsequent macrophage activation, inflammatory cell infiltration, and cytokine production. Mesenchymal stem cells (MSCs) are beneficial for application in the treatment of inflammatory diseases due to their immunomodulatory effects. However, the mechanisms of regulatory effects by MSCs on macrophages in ALI need more in-depth study. Lung tissues were collected from mice for mouse lung organoid construction. Alveolar macrophages (AMs) derived from bronchoalveolar lavage and interstitial macrophages (IMs) derived from lung tissue were co-cultured, with novel matrigel-spreading lung organoids to construct an in vitro model of lung organoids-immune cells. Mouse compact bone-derived MSCs were co-cultured with organoids-macrophages to confirm their therapeutic effect on acute lung injury. Changes in transcriptome expression profile were analyzed by RNA sequencing. Well-established lung organoids expressed various lung cell type-specific markers. Lung organoids grown on spreading matrigel had the property of functional cells growing outside the lumen. Lipopolysaccharide (LPS)-induced injury promoted macrophage chemotaxis toward lung organoids and enhanced the expression of inflammation-associated genes in inflammation-injured lung organoids-macrophages compared with controls. Treatment with MSCs inhibited the injury progress and reduced the levels of inflammatory components. Furthermore, through the nuclear factor-κB pathway, MSC treatment inhibited inflammatory and phenotypic transformation of AMs and modulated the antigen-presenting function of IMs, thereby affecting the inflammatory phenotype of lung organoids. Lung organoids grown by spreading matrigel facilitate the reception of external stimuli and the construction of in vitro models containing immune cells, which is a potential novel model for disease research. MSCs exert protective effects against lung injury by regulating different functions of AMs and IMs in the lung, indicating a potential mechanism for therapeutic intervention.


Assuntos
Lesão Pulmonar Aguda , Células-Tronco Mesenquimais , Pneumonia , Camundongos , Animais , Macrófagos Alveolares/metabolismo , Lipopolissacarídeos/farmacologia , Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/terapia , Pulmão/metabolismo , Macrófagos/metabolismo , Modelos Animais de Doenças , Inflamação/terapia , Inflamação/metabolismo , Organoides/metabolismo
3.
J Med Virol ; 94(6): 2714-2726, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35199373

RESUMO

Dynamic changes in metabolites may affect liver disease progression, and provide new methods for predicting liver damage. We used ultra-performance liquid chromatography-mass spectroscopy to assess serum metabolites in healthy controls (HC), and patients with acute hepatitis E (AHE) or hepatitis E virus acute liver failure (HEV-ALF). The principal component analysis, partial least squares discriminant analysis, and discriminant analysis of orthogonal projections to latent structures models illustrated significant differences in the metabolite components between AHE patients and HCs, or between HEV-ALF and AHE patients. In pathway enrichment analysis, we further identified two altered pathways, including linoleic acid metabolism and phenylalanine, tyrosine, and tryptophan biosynthesis, when comparing AHE patients with HCs. Linoleic acid metabolism and porphyrin and chlorophyll metabolism pathways were significantly different in HEV-ALF when compared with AHE patients. The discriminative performances of differential metabolites showed that taurocholic acid, glycocholic acid, glycochenodeoxycholate-3-sulfate, and docosahexaenoic acid could be used to distinguish HEV-ALF from AHE patients. The serum levels of glycocholic acid, taurocholic acid, deoxycholic acid glycine conjugate, and docosahexaenoic acid were associated with the prognosis of HEV-ALF patients. Dynamic changes in serum metabolites were associated with AHE infection and severity. The identified metabolites can be used to diagnose and predict the prognosis of HEV-ALF.


Assuntos
Vírus da Hepatite E , Hepatite E , Doença Aguda , Ácidos Docosa-Hexaenoicos , Ácido Glicocólico , Humanos , Ácido Linoleico , Ácido Taurocólico
4.
Pharmacol Res ; 179: 106229, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35470065

RESUMO

Acute liver injury (ALI) is characterized by massive hepatocyte necrosis and subsequent recruitment of myeloid cells to liver. Mesenchymal stem cells (MSCs) have therapeutic potential for ALI through their immunoregulation on macrophages, but the mechanism is not completely clear due to the heterogeneity and controversy of liver macrophages. Here, we detected the survival rate, biochemical indexes, histopathology, and inflammatory chemokine levels to assess the efficacy of MSC treatment on CCl4-induced ALI of C57BL/6 mice. Furthermore, flow cytometry and single-cell RNA sequencing (scRNA-Seq) were used to precisely distinguish macrophage populations and reveal the immunoregulation of MSCs. MSC treatment could effectively alleviate ALI and mitigate the recruitment of mononuclear phagocytes. Flow cytometry and scRNA-Seq analyses collectively indicated that there were monocytes with high Ly6C expression and heterogeneous monocyte-derived macrophages (MoMF) with low Ly6C expression in liver. Ly6Chi pro-inflammatory monocytes and Ly6Clo MoMF with powerful phagocytosis dominated during the acute injury period. MSC treatment promoted the transition from Ly6Chi to Ly6Clo population, inhibit the proinflammatory function of monocytes and promote the lysosomal function of MoMF. Furthermore, MSCs attenuated the recruitment of neutrophils by reducing the expression of CXCL2 of MoMF. MoMF with high expression of arginase 1 appeared during the recovery period, and MSCs could increase their expression of arginase 1, which may promote liver repair. To sum up, we demonstrated the characteristics of distinct MoMF during different periods of ALI and revealed their functional changes after MSC treatment, providing immunotherapeutic targets for MSC treatment of ALI.


Assuntos
Células-Tronco Mesenquimais , Análise de Célula Única , Animais , Arginase/metabolismo , Arginase/farmacologia , Homeostase , Fígado , Macrófagos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
5.
J Clin Lab Anal ; 36(6): e24461, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35476781

RESUMO

BACKGROUND: As an important non-apoptotic cell death method, oncosis has been reported to be closely associated with tumors in recent years. However, few research reported the relationship between oncosis and lung cancer. METHODS: In this study, we established an oncosis-based algorithm comprised of cluster grouping and a risk assessment model to predict the survival outcomes and related tumor immunity of patients with lung adenocarcinomas (LUAD). We selected 11 oncosis-related lncRNAs associated with the prognosis (CARD8-AS1, LINC00941, LINC01137, LINC01116, AC010980.2, LINC00324, AL365203.2, AL606489.1, AC004687.1, HLA-DQB1-AS1, and AL590226.1) to divide the LUAD patients into different clusters and different risk groups. Compared with patients in clsuter1, patients in cluster2 had a survival advantage and had a relatively more active tumor immunity. Subsequently, we constructed a risk assessment model to distinguish between patients into different risk groups, in which low-risk patients tend to have a better prognosis. GO enrichment analysis revealed that the risk assessment model was closely related to immune activities. In addition, low-risk patients tended to have a higher content of immune cells and stromal cells in tumor microenvironment, higher expression of PD-1, CTLA-4, HAVCR2, and were more sensitive to immune checkpoint inhibitors (ICIs), including PD-1/CTLA-4 inhibitors. The risk score had a significantly positive correlation with tumor mutation burden (TMB). The survival curve of the novel oncosis-based algorithm suggested that low-risk patients in cluster2 have the most obvious survival advantage. CONCLUSION: The novel oncosis-based algorithm investigated the prognosis and the related tumor immunity of patients with LUAD, which could provide theoretical support for customized individual treatment for LUAD patients.


Assuntos
Adenocarcinoma , Neoplasias Pulmonares , RNA Longo não Codificante , Algoritmos , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Humanos , Pulmão/metabolismo , Proteínas de Neoplasias/metabolismo , Prognóstico , Receptor de Morte Celular Programada 1 , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Medição de Risco , Microambiente Tumoral/genética
6.
Br J Clin Pharmacol ; 87(12): 4737-4746, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33982806

RESUMO

AIMS: Type 1 interferon (IFN) is used to treat patients with coronavirus disease-2019 (COVID-19) but robust supporting evidence is lacking. We investigated the association between IFN-α-2b and the clinical outcomes of patients with COVID-19. METHODS: A total of 1401 patients were enrolled, with 852 (60.8%) patients receiving 5 000 000 U of IFN-α-2b via aerosol inhalation twice daily. The primary outcome was a composite measure consisting of mechanical ventilation, intensive care unit (ICU) admission and death. A subgroup analysis was performed to investigate the impact of the IFN-α-2b initiation schedule on symptom onset. RESULTS: The risk probability for crude endpoints was lower in the IFN-α-2b group (3.8%) than in the non-IFN-α-2b group (9.3%, P < .001). After adjusting the confounding factors, IFN-α-2b therapy achieved a reduction of 64% in occurrence of endpoint events (hazard ratio, 0.36; 95% confidence interval [CI], 0.21-0.62). In the subgroup analysis, compared with patients who received IFN-α-2b treatment 0-2 days after symptom onset, the hazard ratio for endpoints was 2.2 (95% CI, 0.43-11.13) in patients who received the therapy 3-5 days after symptom onset, 5.89 (95% CI, 0.99-35.05) in patients who received the therapy 6-8 days after symptom onset, and remained at a high level thereafter. CONCLUSIONS: IFN-α-2b aerosol inhalation therapy may be associated with improved clinical outcomes in patients with COVID-19, and delayed IFN-α-2b intervention was associated with increased probabilities of risk events. Further randomized clinical trials are needed to validate the preliminary findings of this study.


Assuntos
COVID-19 , Aerossóis , Humanos , Interferon alfa-2 , Proteínas Recombinantes , Respiração Artificial , SARS-CoV-2
7.
J Cell Physiol ; 235(10): 6779-6793, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-31990045

RESUMO

The placenta and umbilical cord are pre-eminent candidate sources of mesenchymal stem cells (MSCs). However, placenta-derived MSCs (P-MSCs) showed greater proliferation capacity than umbilical cord-derived MSCs (UC-MSCs) in our study. We investigated the drivers of this proliferation difference and elucidated the mechanisms of proliferation regulation. Proteomic profiling and Gene Ontology (GO) functional enrichment were conducted to identify candidate proteins that may influence proliferation. Using lentiviral or small interfering RNA infection, we established overexpression and knockdown models and observed changes in cell proliferation to examine whether a relationship exists between the candidate proteins and proliferation capacity. Real-time quantitative polymerase chain reaction, western blot analysis, and immunofluorescence assays were conducted to elucidate the mechanisms underlying proliferation. Six candidate proteins were selected based on the results of proteomic profiling and GO functional enrichment. Through further validation, yes-associated protein 1 (YAP1) and ß-catenin were confirmed to affect MSCs proliferation rates. YAP1 and ß-catenin showed increased nuclear colocalization during cell expansion. YAP1 overexpression significantly enhanced proliferation capacity and upregulated the expression of both ß-catenin and the transcriptional targets of Wnt signaling, CCND1, and c-MYC, whereas silencing ß-catenin attenuated this influence. We found that YAP1 directly interacts with ß-catenin in the nucleus to form a transcriptional YAP/ß-catenin/TCF4 complex. Our study revealed that YAP1 and ß-catenin caused the different proliferation capacities of P-MSCs and UC-MSCs. Mechanism analysis showed that YAP1 stabilized the nuclear ß-catenin protein, and also triggered the Wnt/ß-catenin pathway, promoting proliferation.


Assuntos
Proliferação de Células/fisiologia , Células-Tronco Mesenquimais/fisiologia , Placenta/fisiologia , Cordão Umbilical/fisiologia , Núcleo Celular/metabolismo , Núcleo Celular/fisiologia , Células Cultivadas , Feminino , Humanos , Células-Tronco Mesenquimais/metabolismo , Placenta/metabolismo , Gravidez , Proteômica/métodos , Fatores de Transcrição/metabolismo , Cordão Umbilical/metabolismo , Regulação para Cima/fisiologia , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
8.
Cell Tissue Res ; 380(1): 79-91, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31823005

RESUMO

Microenvironmental factors such as oxygen concentration mediate key effects on the biology of mesenchymal stromal cells (MSCs). Herein, we performed an in-depth characterization of the metabolic behavior of MSCs derived from the placenta, umbilical cord, and adipose tissue (termed hPMSCs, UC-MSCs, and AD-MSCs, respectively) at physiological (hypoxic; 5% oxygen [O2]) and standardized (normoxic; 21% O2) O2 concentrations using chemical isotope labeling liquid chromatography-mass spectrometry. 12C- and 13C-isotope dansylation (Dns) labeling was used to analyze the amine/phenol submetabolome, and 2574 peak pairs or metabolites were detected and quantified, from which 52 metabolites were positively identified using a library of 275 Dns-metabolite standards; 2189 metabolites were putatively identified. Next, we identified six metabolites using the Dns library, as well as 14 hypoxic biomarkers from the human metabolome database out of 96 altered metabolites. Ultimately, metabolic pathway analyses were performed to evaluate the associated pathways. Based on pathways identified using the Kyoto Encyclopedia of Genes and Genomes, we identified significant changes in the metabolic profiles of MSCs in response to different O2 concentrations. These results collectively suggest that O2 concentration has the strongest influence on hPMSCs metabolic characteristics, and that 5% O2 promotes arginine and proline metabolism in hPMSCs and UC-MSCs but decreases gluconeogenesis (alanine-glucose) rates in hPMSCs and AD-MSCs. These changes indicate that MSCs derived from different sources exhibit distinct metabolic profiles.


Assuntos
Hipóxia Celular/fisiologia , Cromatografia Líquida/métodos , Células-Tronco Mesenquimais/metabolismo , Espectrometria de Massas em Tandem/métodos , Humanos , Marcação por Isótopo
9.
Exp Cell Res ; 372(1): 52-60, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30227120

RESUMO

Autophagy has been reported to have a pivotal role in maintaining stemness, regulating immunomodulation and enhancing the survival of mesenchymal stem cells (MSCs). However, the effect of autophagy on MSC metabolism is largely unknown. Here, we report a workflow for examining the impact of autophagy on human placenta-derived MSC (hPMSC) metabolome profiling with chemical isotope labeling (CIL) LC-MS. Rapamycin or 3-methyladenine was successfully used to induce or inhibit autophagy, respectively. Then, 12C- and 13C-dansylation labeling LC-MS were used to profile the amine/phenol submetabolome. A total of 935 peak pairs were detected and 50 metabolites were positively identified using the dansylation metabolite standards library, and 669 metabolites were putatively identified based on an accurate mass match in metabolome databases. 12C/13C-p-dimethylaminophenacyl bromide labeling LC-MS was used to analyze the carboxylic acid submetabolome; 4736 peak pairs were detected, among which 33 metabolites were positively identified in the dimethylaminophenacyl metabolite standards library, and 3007 metabolites were putatively identified. PCA/OPLS-DA analysis combined with volcano plots and Venn diagrams was used to determine the significant metabolites. Metabolites pathway analysis demonstrated that hPMSCs appeared to generate more ornithine with the arginine and proline metabolism pathway and utilized more pantothenic acid to synthesize acetyl-CoA in the beta-alanine metabolism pathway when autophagy was activated. Meanwhile, acetyl-CoA conversion to fatty acids led to accumulation in the fatty acid biosynthesis pathway. In contrast, when autophagy was suppressed, a reduction in metabolites demonstrated weakened metabolic activity in these metabolic pathways. Our research provides a more comprehensive understanding of hPMSC metabolism associated with autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Metaboloma , Placenta/metabolismo , Acetilcoenzima A/metabolismo , Adenina/análogos & derivados , Adenina/farmacologia , Adulto , Arginina/metabolismo , Isótopos de Carbono , Cromatografia Líquida , Ácidos Graxos/biossíntese , Feminino , Humanos , Marcação por Isótopo/métodos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Metabolômica/métodos , Ornitina/metabolismo , Placenta/citologia , Gravidez , Cultura Primária de Células , Análise de Componente Principal , Prolina/metabolismo , Sirolimo/farmacologia , Espectrometria de Massas em Tandem , beta-Alanina/metabolismo
10.
J Proteome Res ; 17(5): 1866-1878, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29671598

RESUMO

The placenta resides in a physiologically low oxygen microenvironment of the body. Hypoxia induces a wide range of stem cell cellular activities. Here, we report a workflow for exploring the role of physiological (hypoxic, 5% oxygen) and original cell culture (normoxic, 21% oxygen) oxygen concentrations in regulating the metabolic status of human placenta-derived mesenchymal stem cells (hPMSCs). The general biological characteristics of hPMSCs were assessed via a variety of approaches such as cell counts, flow cytometry and differentiation study. A sensitive 13C/12C-dansyl labeling liquid chromatography-mass spectrometry (LC-MS) method targeting the amine/phenol submetabolome was used for metabolic profiling of the cell and corresponding culture supernatant. Multivariate and univariate statistical analyses were used to analyze the metabolomics data. hPMSCs cultured in hypoxia display smaller size, higher proliferation, greater differentiation ability and no difference in immunophenotype. Overall, 2987 and 2860 peak pairs or metabolites were detected and quantified in hPMSCs and culture supernatant, respectively. Approximately 86.0% of cellular metabolites and 84.3% of culture supernatant peak pairs were identified using a dansyl standard library or matched to metabolite structures using accurate mass search against human metabolome libraries. The orthogonal partial least-squares discriminant analysis (OPLS-DA) showed a clear separation between the hypoxic group and the normoxic group. Ten metabolites from cells and six metabolites from culture supernatant were identified as potential biomarkers of hypoxia. This study demonstrated that chemical isotope labeling LC-MS can be used to reveal the role of oxygen in the regulation of hPMSC metabolism, whereby physiological oxygen concentrations may promote arginine and proline metabolism, pantothenate and coenzyme A (CoA) biosynthesis, and alanine, aspartate and glutamate metabolism.


Assuntos
Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Metaboloma/efeitos dos fármacos , Oxigênio/farmacologia , Placenta/citologia , Diferenciação Celular , Proliferação de Células , Tamanho Celular , Células Cultivadas , Cromatografia Líquida , Feminino , Humanos , Hipóxia , Marcação por Isótopo , Células-Tronco Mesenquimais/citologia , Metabolômica/métodos , Oxigênio/metabolismo , Gravidez , Espectrometria de Massas em Tandem
11.
Med Sci Monit ; 23: 3001-3009, 2017 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-28628604

RESUMO

BACKGROUND The complete blood count (CBC) is the most common examination used to monitor overall health in clinical practice. Whether there is a relationship between CBC indexes and alanine transaminase (ALT) and aspartate aminotransferase (AST) has been unclear. MATERIAL AND METHODS In this study, 572 normal-weight and 346 overweight Chinese subjects were recruited. The relationship between CBC indexes with ALT and AST were analyzed by Pearson and Spearman correlations according to their sex, then we conducted colinearity diagnostics and multiple linear regression (MLR) analysis. A prediction model was developed by a back-propagation artificial neural network (BP-ANN). RESULTS ALT was related to 4 CBC indexes in the male normal-weight group and 3 CBC indexes in the female group. In the overweight group, ALT had a similar relationship with the normal group, but there was only 1 index related with AST in the normal-weight group and male overweight groups. The ALT regression models were developed in normal-weight and overweight people, which had better correlation coefficient (R>0.3). After training 1000 epochs, the BP-ANN models of ALT achieved higher correlations than MLR models in normal-weight and overweight people. CONCLUSIONS ALT is a more suitable index than AST for developing a regression model. ALT can be predicted by CBC indexes in normal-weight and overweight individuals based on a BP-ANN model, which was better than MLR analysis.


Assuntos
Alanina Transaminase/sangue , Povo Asiático , Aspartato Aminotransferases/sangue , Redes Neurais de Computação , Adulto , Contagem de Células Sanguíneas , Feminino , Humanos , Masculino , Sobrepeso/sangue , Sobrepeso/enzimologia , Análise de Regressão
12.
Cell Physiol Biochem ; 35(6): 2299-308, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25895935

RESUMO

BACKGROUND: Stem cell-based therapy in liver diseases has received increasing interest over the past decade, but direct evidence of the homing and implantation of transplanted cells is conflicting. Reliable labeling and tracking techniques are essential but lacking. The purpose of this study was to establish human placenta-derived mesenchymal stem cells (hPMSCs) expressing green fluorescent protein (GFP) and to assay their hepatic functional differentiation in vitro. METHODS: The GFP gene was transduced into hPMSCs using a lentivirus to establish GFP(+) hPMSCs. GFP(+) hPMSCs were analyzed for their phenotypic profile, viability and adipogenic, osteogenic and hepatic differentiation. The derived GFP(+) hepatocyte-like cells were evaluated for their metabolic, synthetic and secretory functions, respectively. RESULTS: GFP(+) hPMSCs expressed high levels of HLA I, CD13, CD105, CD73, CD90, CD44 and CD29, but were negative for HLA II, CD45, CD31, CD34, CD133, CD271 and CD79. They possessed adipogenic, osteogenic and hepatic differentiation potential. Hepatocyte-like cells derived from GFP(+) hPMSCs showed typical hepatic phenotypes. CONCLUSIONS: GFP gene transduction has no adverse influences on the cellular or biochemical properties of hPMSCs or markers. GFP gene transduction using lentiviral vectors is a reliable labeling and tracking method. GFP(+) hPMSCs can therefore serve as a tool to investigate the mechanisms of MSC-based therapy, including hepatic disease therapy.


Assuntos
Diferenciação Celular/fisiologia , Proteínas de Fluorescência Verde/metabolismo , Hepatócitos/fisiologia , Células-Tronco Mesenquimais/fisiologia , Placenta/fisiologia , Adipogenia/fisiologia , Antígenos CD/metabolismo , Sobrevivência Celular/fisiologia , Células Cultivadas , Feminino , Hepatócitos/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/fisiologia , Placenta/metabolismo , Gravidez
13.
Hepatobiliary Pancreat Dis Int ; 14(2): 186-93, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25865692

RESUMO

BACKGROUND: Cell therapy has been promising for various diseases. We investigated whether transplantation of human umbilical cord mesenchymal stem cells (hUCMSCs) has any therapeutic effects on D-galactosamine/lipopolysaccharide (GalN/LPS)-induced fulminant hepatic failure in mice. METHODS: hUCMSCs isolated from human umbilical cord were cultured and transplanted via the tail vein into severe combined immune deficiency mice with GalN/LPS-induced fulminant hepatic failure. After transplantation, the localization and differentiation of hUCMSCs in the injured livers were investigated by immunohistochemical and genetic analyses. The recovery of the injured livers was evaluated histologically. The survival rate of experimental animals was analyzed by the Kaplan-Meier method and log-rank test. RESULTS: hUCMSCs expressed high levels of CD29, CD73, CD13, CD105 and CD90, but did not express CD31, CD79b, CD133, CD34, and CD45. Cultured hUCMSCs displayed adipogenic and osteogenic differentiation potential. Hematoxylin and eosin staining revealed that transplantation of hUCMSCs reduced hepatic necrosis and promoted liver regeneration. Transplantation of hUCMSCs prolonged the survival rate of mice with fulminant hepatic failure. Polymerase chain reaction for human alu sequences showed the presence of human cells in mouse livers. Positive staining for human albumin, human alpha-fetoprotein and human cytokeratin 18 suggested the formation of hUCMSCs-derived hepatocyte-like cells in vivo. CONCLUSIONS: hUCMSC was a potential candidate for stem cell based therapies. After transplantation, hUCMSCs partially repaired hepatic damage induced by GalN/LPS in mice. hUCMSCs engrafted into the injured liver and differentiated into hepatocyte-like cells.


Assuntos
Antígenos CD/análise , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Falência Hepática Aguda/terapia , Fígado/patologia , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/química , Albuminas/análise , Elementos Alu/genética , Animais , Diferenciação Celular , Galactosamina , Humanos , Queratina-18/análise , Lipopolissacarídeos , Lipase Lipoproteica/genética , Falência Hepática Aguda/induzido quimicamente , Falência Hepática Aguda/patologia , Masculino , Camundongos , Camundongos SCID , Necrose/etiologia , Necrose/terapia , Osteopontina/genética , RNA Mensageiro/metabolismo , Taxa de Sobrevida , Fator de Necrose Tumoral alfa/sangue , alfa-Fetoproteínas/análise
14.
Nanoscale ; 16(25): 12149-12162, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38833269

RESUMO

Together, tumor and virus-specific tissue-resident CD8+ memory T cells (TRMs) of hepatocellular carcinoma (HCC) patients with Hepatitis B virus (HBV) infection can provide rapid frontline immune surveillance. The quantity and activity of CD8+ TRMs were correlated with the relapse-free survival of patients with improved health. However, HBV-specific CD8+ TRMs have a more exhausted phenotype and respond more actively under anti-PDL1 or PD1 treatment of HBV+HCC patients. Vaccination strategies that induce a strong and sustained CD8+ TRMs response are quite promising. Herein, a biodegradable poly(D,L-lactide-co-glycolide) microsphere and nanosphere particle (PLGA N.M.P) delivery system co-assembled by anti-PD1 antibodies (aPD1) and loaded with ovalbumin (OVA-aPD1 N.M.P) was fabricated and characterized for size (200 nm and 1 µm diameter), charge (-15 mV), and loading efficiencies of OVA (238 µg mg-1 particles) and aPD1 (40 µg mg-1 particles). OVA-aPD1 N.M.P could stimulate the maturation of BMDCs and enhance the antigen uptake and presentation by 2-fold compared to free OVA. The nanoparticles also induced the activation of macrophages (RAW 264.7) to produce a high level of cytokines, including TNF-α, IL-6 and IL-10. In vivo stimulation of mice using OVA-aPD1 N.M.P robustly enhanced IFN-γ-producing-CD8+ T cell infiltration in tumor tissues and the secretion of IgG and IgG2a/IgG1 antibodies. OVA-aPD1 N.M.P delivered OVA to increase the activation and proliferation of OVA-specific CD8+ TRMs, and its combination with anti-PD1 antibodies promoted complete tumor rejection by the reversal of tumor-infiltrating CD8+ T cell exhaustion. Thus, PLGA N.M.P could induce a strong CD8+ TRMs response, further highlighting its therapeutic potential in enhancing an antitumor immune response.


Assuntos
Linfócitos T CD8-Positivos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Camundongos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Ovalbumina/química , Receptor de Morte Celular Programada 1/imunologia , Receptor de Morte Celular Programada 1/metabolismo , Nanopartículas/química , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/química , Células T de Memória/imunologia , Vacinação , Humanos , Células RAW 264.7 , Memória Imunológica
15.
Asian J Pharm Sci ; 19(1): 100889, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38419761

RESUMO

Primary sclerosing cholangitis (PSC) is an autoimmune cholangiopathy characterized by chronic inflammation of the biliary epithelium and periductal fibrosis, with no curative treatment available, and liver transplantation is inevitable for end-stage patients. Human placental mesenchymal stem cell (hpMSC)-derived exosomes have demonstrated the ability to prevent fibrosis, inhibit collagen production and possess immunomodulatory properties in autoimmune liver disease. Here, we prepared hpMSC-derived exosomes (ExoMSC) and further investigated the anti-fibrotic effects and detailed mechanism on PSC based on Mdr2-/- mice and multicellular organoids established from PSC patients. The results showed that ExoMSC ameliorated liver fibrosis in Mdr2-/- mice with significant collagen reduction in the preductal area where Th17 differentiation was inhibited as demonstrated by RNAseq analysis, and the percentage of CD4+IL-17A+T cells was reduced both in ExoMSC-treated Mdr2-/- mice (Mdr2-/--Exo) in vivo and ExoMSC-treated Th17 differentiation progressed in vitro. Furthermore, ExoMSC improved the hypersecretory phenotype and intercellular interactions in the hepatic Th17 microenvironment by regulating PERK/CHOP signaling as supported by multicellular organoids. Thus, our data demonstrate the anti-fibrosis effect of ExoMSC in PSC disease by inhibiting Th17 differentiation, and ameliorating the Th17-induced microenvironment, indicating the promising potential therapeutic role of ExoMSC in liver fibrosis of PSC or Th17-related diseases.

16.
Medicine (Baltimore) ; 102(35): e34844, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37657029

RESUMO

Currently, a reliable early prognostic marker has not been identified for lung adenocarcinoma (LUAD), the most common malignancy. Recent studies demonstrated that lysosomal rupture is involved in cancer migration, progression, and immune microenvironment formation. We performed a bioinformatics analysis of lysosomal rupture to investigate whether lysosome-related genes (LRGs) are key in LUAD. The analysis identified 23 LRGs. Cytoscape visualization identified 10 core genes (CCNA2, DLGAP5, BUB1B, KIF2C, PBK, CDC20, NCAPG, ASPM, KIF4A, ANLN). With the 23 LRGs, we established a new risk scoring rule to classify patients with LUAD into high- and low-risk groups and verified the accuracy of the risk score by receiver operating characteristic curves and established a nomogram to evaluate clinical patients. Immunotherapy effectiveness between the high- and low-risk groups was evaluated based on the tumor mutational burden and analyses of immune cell infiltration and drug sensitivity. Pathway enrichment analysis revealed that lysosomes were closely associated with glucose metabolism, amino acid metabolism, and the immune response in patients with LUAD. Lysosomes are a likely new therapeutic target and provide new directions and ideas for treating and managing patients with LUAD.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Humanos , Prognóstico , Adenocarcinoma de Pulmão/genética , Lisossomos , Biologia Computacional , Neoplasias Pulmonares/genética , Microambiente Tumoral , Cinesinas/genética
17.
Research (Wash D C) ; 6: 0207, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37600495

RESUMO

Primary sclerosing cholangitis (PSC) is a biliary disease accompanied by chronic inflammation of the liver and biliary stricture. Mesenchymal stem cells (MSCs) are used to treat liver diseases because of their immune regulation and regeneration-promoting functions. This study was performed to explore the therapeutic potential of human placental MSCs (hP-MSCs) in PSC through the Takeda G protein-coupled receptor 5 (TGR5) receptor pathway. Liver tissues were collected from patients with PSC and healthy donors (n = 4) for RNA sequencing and intrahepatic cholangiocyte organoid construction. hP-MSCs were injected via the tail vein into Mdr2-/-, bile duct ligation (BDL), and 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) mouse models or co-cultured with organoids to confirm their therapeutic effect on biliary cholangitis. Changes in bile acid metabolic profile were analyzed by liquid chromatography/tandem mass spectrometry (LC-MS/MS). Compared with healthy controls, liver tissues and intrahepatic cholangiocyte organoids from PSC patients were characterized by inflammation and cholestasis, and marked downregulation of bile acid receptor TGR5 expression. hP-MSC treatment apparently reduced the inflammation, cholestasis, and fibrosis in Mdr2-/-, BDL, and DDC model mice. By activating the phosphatidylinositol 3 kinase/extracellular signal-regulated protein kinase pathway, hP-MSC treatment promoted the proliferation of cholangiocytes, and affected the transcription of downstream nuclear factor κB through regulation of the binding of TGR5 and Pellino3, thereby affecting the cholangiocyte inflammatory phenotype.

18.
Phytomedicine ; 121: 155045, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37742526

RESUMO

BACKGROUND: Isoliquiritigenin (ISL), a natural flavonoid, has anti-tumor activity. But, the understanding of the impact and molecular mechanism of ISL on the growth of gastric cancer (GC) remains limited. PURPOSE: The study was to explore the tumor suppressive effect of ISL on GC growth both in vitro and in vivo, meanwhile, clarify its molecular mechanisms. METHODS: Cell viability was detected by cell counting kit-8 (CCK-8) assay. Apoptotic cells in vitro were monitored by Hoechst 33,342 solution. Protein expression was assessed by Western blot. Reactive oxygen species (ROS) level was evaluated by utilizing 2',7'- dichlorofluorescin diacetate (DCFH-DA). Lactic acid level was detected with L-lactate assay kit. Glucose uptake was monitored with fluorescently tagged glucose 2-[N-(7-nitrobenz-2-oxa-1,3-diaxol-4-yl)amino]-2-deoxyglucose (2-NBDG). Glycolytic proton efflux rate (GlycoPER) was evaluated by glycolytic rate assay kit. Oxygen consumption rate (OCR) was conducted by mito stress test kit. A nude mouse model of gastric cancer cell xenograft was established by subcutaneous injection with MGC803 cells. Pathological changes were evaluated by using H&E staining. Cell apoptosis in vivo was evaluated by terminal deoxy-nucleotide transferase mediated dUTP nick end labeling (TUNEL) assay. RESULTS: ISL remarkably suppressed GC growth and increased cell apoptosis. It regulated apoptosis-related and metabolism-related protein expression both in vitro and in vivo. ISL blocked glucose uptake and suppressed production and secretion of lactic acid, which was accompanied with suppressed mitochondrial oxidative phosphorylation (OXPHOS) and glycolysis but increased ROS accumulation. Overexpression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α), cellular-myelocytomatosis viral oncogene (c-Myc), hypoxia inducible factor-1α (HIF-1α), glucose transporter 4 (GLUT4) or pyruvate dehydrogenase kinase 1 (PDHK1), could abolish ISL-induced inhibition of cell viability in GC cells. CONCLUSION: These findings implicated that ISL inhibits GC growth by decreasing GLUT4 mediated glucose uptake and inducing PDHK1/PGC-1α-mediated energy metabolic collapse through depressing protein expression of c-Myc and HIF-1α in GC, suggesting its potential application for GC treatment.


Assuntos
Neoplasias Gástricas , Camundongos , Animais , Humanos , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Gástricas/tratamento farmacológico , Glucose/metabolismo , Ácido Láctico , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
19.
Stem Cell Res Ther ; 14(1): 197, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37553691

RESUMO

BACKGROUND: Mesenchymal stem cell (MSC) treatment plays a major role in the management of acute lung injury (ALI), and neutrophils are the initial line of defense against ALI. However, the effect of MSCs on neutrophils in ALI remains mostly unknown. METHODS: We investigated the characteristics of neutrophils in lung tissue of ALI mice induced by lipopolysaccharide after treatment with MSCs using single-cell RNA sequencing. Neutrophils separated from lung tissue in ALI were co-cultured with MSCs, and then samples were collected for reverse transcription-polymerase chain reaction and flow cytometry. RESULTS: During inflammation, six clusters of neutrophils were identified, annotated as activated, aged, and circulatory neutrophils. Activated neutrophils had higher chemotaxis, reactive oxygen species (ROS) production, and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase scores than aged neutrophils. Circulatory neutrophils occurred mainly in healthy tissue and were characterized by higher expression of Cxcr2 and Sell. Activated neutrophils tended to exhibit higher expression of Cxcl10 and Cd47, and lower expression of Cd24a, while aged neutrophils expressed a lower level of Cd47 and higher level of Cd24a. MSC treatment shifted activated neutrophils toward an aged neutrophil phenotype by upregulating the expression of CD24, thereby inhibiting inflammation by reducing chemotaxis, ROS production, and NADPH oxidase. CONCLUSION: We identified the immunosuppressive effects of MSCs on the subtype distribution of neutrophils and provided new insight into the therapeutic mechanism of MSC treatment in ALI.


Assuntos
Lesão Pulmonar Aguda , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Camundongos , Animais , Neutrófilos/metabolismo , Antígeno CD47/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Lesão Pulmonar Aguda/induzido quimicamente , Pulmão/metabolismo , Lipopolissacarídeos/toxicidade , Inflamação/terapia , Inflamação/metabolismo , Células-Tronco Mesenquimais/metabolismo
20.
Biochem Biophys Res Commun ; 422(4): 539-45, 2012 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-22580002

RESUMO

AIM: The aim of this study was to analyze the hepatic differentiation potential of human umbilical cord blood-derived mesenchymal stem cells (hUCBMSCs) after transplantation into severe combined immune deficiency (SCID) mice with liver injury induced by D-galactosamine/lipopolysaccharide (GalN/LPS) and to explore the possibility that cells can partially repair GalN/LPS-induced hepatic damage. METHODS: Mononuclear cells (MNCs) were isolated from fresh human umbilical cord blood, characterized by flow cytometry, and then transplanted into GalN/LPS-injured mice. Specimens were collected at 7, 14, 21, and 28 days after hUCBMSC transplantation. Histopathological changes were analyzed by hematoxylin and eosin staining. Polymerase chain reaction (PCR) for a specific marker of human cells, the human Alu sequence, was performed to locate exogenous hUCBMSCs in mouse livers. Expression of human hepatocyte-specific markers such as human albumin (hALB), human alpha-fetoprotein (hAFP), human cytokeratin 18 (hCK18), and human cytokeratin 19 (hCK19) were analyzed by reverse transcriptase (RT)-PCR and immunohistochemical staining. RESULTS: The hUCBMSCs were positive for the human MSC-specific markers CD271, CD29, CD90, CD105, and CD73, but negative for CD31, CD79b, CD133, CD34, and CD45. Histological findings showed that the hepatic damage in mice was attenuated after hMSC administration, and liver architecture was much better preserved. Human cells in the injured liver of recipient mice were detected by PCR for the human Alu sequence. In addition, expression of markers of hepatic lineage, including hALB, hAFP, hCK18, and hCK19, was detected by immunohistochemistry and RT-PCR in mouse livers after hUCBMSC transplantation, suggesting the formation of hepatocyte-like cells in vivo. CONCLUSION: MSCs from hUCB exhibit the potential to differentiate into hepatocyte-like cells in the livers of hUCB-transplanted mice as well as partially repair the liver damage induced by GalN/LPS.


Assuntos
Diferenciação Celular , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Sangue Fetal/citologia , Hepatócitos/citologia , Falência Hepática Aguda/cirurgia , Células-Tronco Mesenquimais/citologia , Albuminas/genética , Animais , Antígenos CD/análise , Antígenos CD/metabolismo , Expressão Gênica , Hepatócitos/metabolismo , Humanos , Camundongos , Camundongos SCID , alfa-Fetoproteínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA