Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 827
Filtrar
1.
Clin. transl. oncol. (Print) ; 24(3): 517-531, marzo 2022.
Artigo em Inglês | IBECS | ID: ibc-203546

RESUMO

BackgroundPancreatic ductal adenocarcinoma (PDAC) is one of the most fatal malignant tumors of the digestive system. Many patients are diagnosed at an advanced stage and lose eligibility for surgery. Moreover, there are few effective methods for treating pancreatic ductal cell carcinoma. Increasing attention has been given to microRNAs (miRNAs) and their regulatory roles in tumor progression. In this study, we investigated the effects of exosomes extracted from human umbilical cord mesenchymal stem cells (HUCMSCs) carrying hsa-miRNA-128-3p on pancreatic cancer cells.MethodsBased on existing experimental and database information, we selected Galectin-3, which is associated with pancreatic cancer, and the corresponding upstream hsa-miRNA-128-3p. We extracted HUCMSCs from a fresh umbilical cord, hsa-miRNA-128-3p was transfected into HUCMSCs, and exosomes containing hsa-miRNA-128-3p were extracted and collected. The effect of exosomes rich in hsa-miRNA-128-3p on pancreatic cancer cells was analyzed.ResultsThe expression of Galectin-3 in normal pancreatic duct epithelial cells was significantly lower than that in PDAC cell lines. We successfully extracted HUCMSCs from the umbilical cord and transfected hsa-miRNA-128-3p into HUCMSCs. Then we demonstrated that HUCMSC-derived exosomes with hsa-miRNA-128-3p could suppress the proliferation, invasion, and migration of PANC-1 cells in vitro by targeting Galectin-3.ConclusionHsa-miRNA-128-3p could be considered as a potential therapy for pancreatic cancer. We provided a new idea for targeted therapy of PDAC.


Assuntos
Carcinoma Ductal Pancreático/patologia , Exossomos/fisiologia , Galectina 3/fisiologia , Células-Tronco Mesenquimais/ultraestrutura , Cordão Umbilical/citologia , Neoplasias Pancreáticas/patologia , Células Tumorais Cultivadas
2.
BMC Cardiovasc Disord ; 22(1): 61, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-35172728

RESUMO

BACKGROUND: Myocardial infarction (MI) represents a severe cardiovascular disease with limited therapeutic agents. This study was aimed to elucidate the role of the exosomes derived from human placental mesenchymal stem cells (PMSCs-Exos) in MI. METHODS: PMSCs were isolated and cultured in vitro, with identification by both transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). To further investigate the effects of PMSC-Exos on MI, C57BL/6 mice were randomly divided into Sham group, MI group, and PMSC-Exos group. After 4 weeks of the intervention, cardiac function was assessed by cardiac echocardiography, electrocardiogram and masson trichrome staining; lipid indicators were determined by automatic biochemical instrument; inflammatory cytokines were measured by cytometric bead array (CBA); gut microbiota, microbial metabolites short chain fatty acids (SCFAs) as well as lipopolysaccharide (LPS) were separately investigated by 16S rRNA high throughput sequencing, gas chromatography mass spectrometry (GC-MS) and tachypleus amebocyte lysate kit; transcriptome analysis was used to test the transcriptional components (mRNA\miRNA\cirRNA\lncRNA) of PMSC-Exos. RESULTS: We found that human PMSC-Exos were obtained and identified with high purity and uniformity. MI model was successfully established. Compared to MI group, PMSC-Exos treatment ameliorated myocardial fibrosis and left ventricular (LV) remodeling (P < 0.05). Moreover, PMSC-Exos treatment obviously decreased MI molecular markers (AST/BNP/MYO/Tn-I/TC), pro-inflammatory indicators (IL-1ß, IL-6, TNF-α, MCP-1), as well as increased HDL in comparison with MI group (all P < 0.05). Intriguingly, PMSC-Exos intervention notably modulated gut microbial community via increasing the relative abundances of Bacteroidetes, Proteobacteria, Verrucomicrobia, Actinobacteria, Akkermansia, Bacteroides, Bifidobacterium, Thauera and Ruminiclostridium, as well as decreasing Firmicutes (all P < 0.05), compared with MI group. Furthermore, PMSC-Exos supplementation increased gut microbiota metabolites SCFAs (butyric acid, isobutyric acid and valeric acid) and decreased LPS in comparison with MI group (all P < 0.05). Correlation analysis indicated close correlations among gut microbiota, microbial SCFAs and inflammation in MI. CONCLUSIONS: Our study highlighted that PMSC-Exos intervention alleviated MI via modulating gut microbiota and suppressing inflammation.


Assuntos
Bactérias/crescimento & desenvolvimento , Exossomos/transplante , Microbioma Gastrointestinal , Mediadores da Inflamação/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio/cirurgia , Miocárdio/metabolismo , Animais , Bactérias/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Disbiose , Exossomos/metabolismo , Exossomos/ultraestrutura , Feminino , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/microbiologia , Miocárdio/patologia , Placenta/citologia , Gravidez , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma
3.
Placenta ; 117: 161-168, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34915433

RESUMO

The emergence of COVID-19 has created a major health crisis across the globe. Invasion of SARS-CoV-2 into the lungs causes acute respiratory distress syndrome (ARDS) that result in the damage of lung alveolar epithelial cells. Currently, there is no standard treatment available to treat the disease and the resultant lung scarring is irreversible even after recovery. This has prompted researchers across the globe to focus on developing new therapeutics and vaccines for the treatment and prevention of COVID-19. Mesenchymal stem cells (MSCs) have emerged as an efficient drug screening platform and MSC-derived organoids has found applications in disease modeling and drug discovery. Perinatal tissue derived MSC based cell therapies have been explored in the treatment of various disease conditions including ARDS because of their enhanced regenerative and immunomodulatory properties. The multi-utility properties of MSCs have been described in this review wherein we discuss the potential use of MSC-derived lung organoids in screening of novel therapeutic compounds for COVID-19 and also in disease modeling to better understand the pathogenesis of the disease. This article also summarizes the rationale behind the development of MSC-based cell- and cell-free therapies and vaccines for COVID-19 with a focus on the current progress in this area. With the pandemic raging, an important necessity is to develop novel treatment strategies which will not only alleviate the disease symptoms but also avoid any off-target effects which could further increase post infection sequelae. Naturally occurring mesenchymal stem cells could be the magic bullet which fulfil these criteria.


Assuntos
Âmnio/citologia , COVID-19/terapia , Células-Tronco Mesenquimais , Placenta/citologia , SARS-CoV-2 , Cordão Umbilical/citologia , Vacinas contra COVID-19 , Terapia Baseada em Transplante de Células e Tecidos , Exossomos/transplante , Feminino , Humanos , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/ultraestrutura , Gravidez , SARS-CoV-2/imunologia , Geleia de Wharton/citologia
4.
Shock ; 57(2): 281-290, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34798633

RESUMO

BACKGROUND: Cell-based therapies using mesenchymal stem cell derived extracellular vesicles (EVs) improve neurologic outcomes in animal models of traumatic brain injury (TBI), stroke, and hemorrhage. Using a porcine 7-day survival model of TBI and hemorrhagic shock (HS), we previously demonstrated that EV-treatment was associated with reduced brain lesion size, neurologic severity score, and cerebral inflammation. However, the underlying cellular and genomic mechanisms remain poorly defined. We hypothesize that EV treatment modulates the brain transcriptome to enhance neuroprotection and neurorestoration following TBI + HS. METHODS: Swine were subjected to severe TBI (8-mm cortical impact) and HS (40% blood volume). After 1 h of shock, animals were randomized (n = 4/group) to treatment with either lactated Ringer's (LR) or LR + EV. Both groups received fluid resuscitation after 2 h of shock, and autologous packed red blood cells 5 h later.After 7-days, brains were harvested and RNA-sequencing was performed. The transcriptomic data were imported into the iPathway pipeline for bioinformatics analyses. RESULTS: 5,273 genes were differentially expressed in the LR + EV group versus LR alone (total 9,588 measured genes). Genes with the greatest upregulation were involved in synaptic transmission and neuronal development and differentiation, while downregulated genes were involved in inflammation. GO-terms experiencing the greatest modulation were involved in inflammation, brain development, and cell adhesion. Pathway analysis revealed significant modulation in the glutamatergic and GABAergic systems. Network analysis revealed downregulation of inflammation, and upregulation of neurogenesis, and neuron survival and differentiation. CONCLUSIONS: In a porcine model of TBI + HS, EV treatment was associated with an attenuation of cerebral inflammatory networks and a promotion of neurogenesis and neuroplasticity. These transcriptomic changes could explain the observed neuroprotective and neurorestorative properties associated with EV treatment.


Assuntos
Lesões Encefálicas Traumáticas/terapia , Vesículas Extracelulares/transplante , Células-Tronco Mesenquimais/ultraestrutura , Choque Hemorrágico/terapia , Animais , Encéfalo , Modelos Animais de Doenças , Intervenção Médica Precoce , Neuroproteção/genética , Suínos , Transcriptoma
5.
Clin Transl Oncol ; 24(3): 517-531, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34811696

RESUMO

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most fatal malignant tumors of the digestive system. Many patients are diagnosed at an advanced stage and lose eligibility for surgery. Moreover, there are few effective methods for treating pancreatic ductal cell carcinoma. Increasing attention has been given to microRNAs (miRNAs) and their regulatory roles in tumor progression. In this study, we investigated the effects of exosomes extracted from human umbilical cord mesenchymal stem cells (HUCMSCs) carrying hsa-miRNA-128-3p on pancreatic cancer cells. METHODS: Based on existing experimental and database information, we selected Galectin-3, which is associated with pancreatic cancer, and the corresponding upstream hsa-miRNA-128-3p. We extracted HUCMSCs from a fresh umbilical cord, hsa-miRNA-128-3p was transfected into HUCMSCs, and exosomes containing hsa-miRNA-128-3p were extracted and collected. The effect of exosomes rich in hsa-miRNA-128-3p on pancreatic cancer cells was analyzed. RESULTS: The expression of Galectin-3 in normal pancreatic duct epithelial cells was significantly lower than that in PDAC cell lines. We successfully extracted HUCMSCs from the umbilical cord and transfected hsa-miRNA-128-3p into HUCMSCs. Then we demonstrated that HUCMSC-derived exosomes with hsa-miRNA-128-3p could suppress the proliferation, invasion, and migration of PANC-1 cells in vitro by targeting Galectin-3. CONCLUSION: Hsa-miRNA-128-3p could be considered as a potential therapy for pancreatic cancer. We provided a new idea for targeted therapy of PDAC.


Assuntos
Carcinoma Ductal Pancreático/patologia , Exossomos/fisiologia , Galectina 3/fisiologia , Células-Tronco Mesenquimais/ultraestrutura , Neoplasias Pancreáticas/patologia , Cordão Umbilical/citologia , Humanos , Células Tumorais Cultivadas
6.
Development ; 148(24)2021 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-34927678

RESUMO

Lung organogenesis requires precise timing and coordination to effect spatial organization and function of the parenchymal cells. To provide a systematic broad-based view of the mechanisms governing the dynamic alterations in parenchymal cells over crucial periods of development, we performed a single-cell RNA-sequencing time-series yielding 102,571 epithelial, endothelial and mesenchymal cells across nine time points from embryonic day 12 to postnatal day 14 in mice. Combining computational fate-likelihood prediction with RNA in situ hybridization and immunofluorescence, we explore lineage relationships during the saccular to alveolar stage transition. The utility of this publicly searchable atlas resource (www.sucrelab.org/lungcells) is exemplified by discoveries of the complexity of type 1 pneumocyte function and characterization of mesenchymal Wnt expression patterns during the saccular and alveolar stages - wherein major expansion of the gas-exchange surface occurs. We provide an integrated view of cellular dynamics in epithelial, endothelial and mesenchymal cell populations during lung organogenesis.


Assuntos
Desenvolvimento Embrionário/genética , Pulmão/crescimento & desenvolvimento , Células-Tronco Mesenquimais/citologia , Organogênese/genética , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Embrião de Mamíferos/ultraestrutura , Células Epiteliais/citologia , Células Epiteliais/ultraestrutura , Regulação da Expressão Gênica no Desenvolvimento/genética , Pulmão/ultraestrutura , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , RNA-Seq , Análise de Célula Única , Transcriptoma/genética
7.
Tissue Cell ; 73: 101665, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34695652

RESUMO

OBJECTIVE(S): To investigate and test the hypotheses that FGF-2 enhanced myocardial differentiation with rat bone marrow mesenchymal stem cells (BMSCs). MATERIALS AND METHODS: Lentiviral vectors carrying the FGF-2 gene were transfected into rat BMSCs firstly. According to the different inducing agents, they were divided into the following four groups: group A (BMSCs blank control group), group B (FGF-2 induction group), group C (Lenti-FGF-2-GFP lentivirus transfection group), and the group D (Lenti-control-GFP lentiviral transfer). Then several kinds of experimental methods such as real-time PCR, immunocytochemical staining, immunofluorescence staining, Western blot, and transmission electron microscopy were used to elucidate the effects by which FGF-2 adjusts myocardial differentiation in rat BMSCs. RESULTS: The results of real-time PCR showed that GATA-4 and Nkx2.5 were expressed in all groups of cells. Compared with the experimental control group, the expression of GATA-4 and Nkx2.5 genes was the strongest after induction of 2 weeks in each induction group, and gradually decreased after induction of 4 weeks. Among them, the relative expression levels of GATA-4 and Nkx2.5 genes in Lenti-FGF-2-GFP were highest at all time points. The expressions of cTnI, cTnT, Cx43, and Desmin were detected by immunocytochemical staining and immunofluorescence staining. After 4 weeks of induction, cTnI, cTnT, Cx43, and Desmin were positively expressed in the cytoplasm of cells. Statistical analysis showed that the integrated optical density (IOD) values of the markers in the Lenti-FGF-2-GFP were the strongest. Cx43 and cTnI were weakly positive or negative in the experimental control group. There was a significant difference in the positive expression of each marker in each induction group and the experimental control group. Western blot analysis showed that Tromyosin (Tm) and Desmin were expressed in the blank group, FGF-2 drug-induced group, Lenti-FGF-2-GFP, and empty virus control transfection group after 4 weeks of induction, among which FGF-2 lentivirus transfected. The expression levels of Tm and Desmin were the highest in the staining induction group. Statistical analysis showed that the positive expressions of Tm and Desmin in each experimental group were statistically significant. Transmission electron microscopy showed that the nucleus of the cells transfected and induced by FGF-2 was located at the center of the cells. Myofilaments, rough endoplasmic reticulum, and mitochondria, and ribosomes were seen in the cytoplasm. CONCLUSION: These results indicate that FGF-2 can transfect and induce differentiation of BMSCs into cardiomyocyte-like cells. Lentivirus-mediated FGF-2 transfection induces the differentiation of bone marrow mesenchymal stem cells into cardiomyocyte-like cells better than FGF-2 direct induction.


Assuntos
Diferenciação Celular , Fator 2 de Crescimento de Fibroblastos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Animais , Forma Celular , Desmina/metabolismo , Regulação da Expressão Gênica , Células-Tronco Mesenquimais/ultraestrutura , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção
8.
Front Endocrinol (Lausanne) ; 12: 693977, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34484115

RESUMO

Since the potential roles of extracellular vesicles secreted by adipose-derived mesenchymal stem cells (ADSCs) are not well understood in collagen metabolism, the purpose of this research was to evaluate the effects of ADSCs-extracellular vesicles in stress urinary incontinence and the regulatory mechanism of delivered microRNA-93 (miR-93). ADSCs were isolated and cultured, and ADSCs-extracellular vesicles were extracted and identified. Stress urinary incontinence primary fibroblasts or satellite cells were treated with ADSCs-extracellular vesicles to detect the expression of Elastin, Collagen I, and Collagen III in fibroblasts and Pax7 and MyoD in satellite cells. After transfecting ADSCs with miR-93 mimics or inhibitors, extracellular vesicles were isolated and treated with stress urinary incontinence primary fibroblasts or satellite cells to observe cell function changes. The online prediction and luciferase activity assay confirmed the targeting relationship between miR-93 and coagulation factor III (F3). The rescue experiment verified the role of ADSCs-extracellular vesicles carrying miR-93 in stress urinary incontinence primary fibroblasts and satellite cells by targeting F3. ADSCs-extracellular vesicles treatment upregulated expression of Elastin, Collagen I, and Collagen III in stress urinary incontinence primary fibroblasts and expression of Pax7 and MyoD in stress urinary incontinence primary satellite cells. miR-93 expression was increased in stress urinary incontinence primary fibroblasts or satellite cells treated with ADSCs-extracellular vesicles. Extracellular vesicles secreted by ADSCs could deliver miR-93 to fibroblasts and then negatively regulate F3 expression; ADSCs-extracellular vesicles could reverse the effect of F3 on extracellular matrix remodeling in stress urinary incontinence fibroblasts. miR-93 expression was also increased in stress urinary incontinence primary satellite cells treated by ADSCs-extracellular vesicles. Extracellular vesicles secreted by ADSCs were delivered to satellite cells through miR-93, which directly targets F3 expression and upregulates Pax7 and MyoD expression in satellite cells. Our study indicates that miR-93 delivered by ADSCs-extracellular vesicles could regulate extracellular matrix remodeling of stress urinary incontinence fibroblasts and promote activation of stress urinary incontinence satellite cells through targeting F3.


Assuntos
Vesículas Extracelulares/fisiologia , Células-Tronco Mesenquimais/ultraestrutura , Incontinência Urinária por Estresse/prevenção & controle , Tecido Adiposo/citologia , Animais , Células Cultivadas , Contactina 1/genética , Contactina 1/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patologia , Regulação da Expressão Gênica , Masculino , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , MicroRNAs/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Incontinência Urinária por Estresse/genética , Incontinência Urinária por Estresse/metabolismo , Incontinência Urinária por Estresse/patologia
9.
Front Endocrinol (Lausanne) ; 12: 711902, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34456868

RESUMO

Background: Premature ovarian insufficiency (POI) is associated with severe physical damage and psychological burden on women. Transplantation of exosomes is an encouraging regenerative medicine method, which has the potential for restoring ovarian functions on POI with high efficiency. This study aims at evaluating the therapeutic efficacy of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-Exos) on ovarian dysfunction of POI and the role of Hippo pathway in this exosome-mediated treatment. Methods: POI mice models were established through intraperitoneal injection of cyclophosphamide. Subsequently, transplantation of hUCMSC-Exos was conducted to administer POI mice. Ovaries and plasma of these mice models were harvested after two weeks of treatment. Ovarian morphology and follicle number were assessed by hematoxylin and eosin staining. Moreover, ELISA was used to detect hormone levels, which are related to ovarian function in serum. To assess the recovery of reproductive ability, we recorded the rate of pregnancy, the amount of offspring, and the time of birth in different groups. To explore the underlying mechanisms of exosome-mediated treatment for ovarian function recovery, the proliferation of ovarian cells in vivo was detected by immunohistochemistry and immunofluorescence staining. Additionally, we conducted EdU and CCK-8 assays to assess the proliferative ability of ovarian granulosa cells (GCs) that were cultured in vitro. Western blot analysis was conducted to estimate the proteins levels of Hippo- and proliferation-associated molecules in vivo and in vitro. Results: After transplantation of hUCMSC-Exos, the ovarian function-related hormone levels and the number of ovarian follicles returned to nearly normal degrees. Meanwhile, there was a significant improvement in reproductive outcomes after exosomal treatment. Furthermore, the improvement of ovarian function and proliferation was associated with the regulation of Hippo pathway. In vitro, co-culture with exosomes significantly elevated the proliferation of ovarian GCs by regulating Hippo pathway. However, the positive effects on the proliferation of GCs were significantly depressed when key Hippo pathway molecule was inhibited. Conclusion: This study suggested that hUCMSC-Exos promoted ovarian functions and proliferation by regulating the Hippo pathway. Therefore, exosomal transplantation could be a promising and efficient clinical therapy for POI in the near future.


Assuntos
Exossomos/transplante , Via de Sinalização Hippo/fisiologia , Células-Tronco Mesenquimais/ultraestrutura , Insuficiência Ovariana Primária/terapia , Cordão Umbilical/citologia , Animais , Proliferação de Células , Exossomos/fisiologia , Feminino , Células da Granulosa/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ovário/crescimento & desenvolvimento , Ovário/fisiopatologia , Insuficiência Ovariana Primária/fisiopatologia
10.
Neurosci Lett ; 763: 136179, 2021 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-34416344

RESUMO

Neural differentiation of mesenchymal stromal cells has been widely studied. However, a comparative characterization of ultrastructural changes during neural differentiation has not been performed. In this study, we conducted scanning electron microscopy and transmission electron microscopy analysis to show the morphological changes in mesenchymal stromal cells upon induction of neural differentiation. In addition, transmission electron microscopy results demonstrated ultrastructural differences between human cranial bone marrow mesenchymal stromal cells and iliac crest bone marrow mesenchymal stromal cells. We propose that enriched microvesicles in cranial bone marrow mesenchymal stromal cells may be responsible for the increased efficiency of neural differentiation.


Assuntos
Células-Tronco Mesenquimais/ultraestrutura , Neurogênese , Crânio/citologia , Medula Óssea/irrigação sanguínea , Células Cultivadas , Humanos , Ílio/citologia , Células-Tronco Mesenquimais/fisiologia , Microscopia Eletrônica de Transmissão , Microvasos/anatomia & histologia , Cultura Primária de Células
11.
Exp Hematol ; 100: 41-51, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34228982

RESUMO

Bone marrow stromal cells (BMSCs) are a key part of the hematopoietic niche. Mouse and human BMSCs are recognized by different markers (LepR and NGFR/CD271, respectively). However, there has not been a detailed in situ comparison of both populations within the hematopoietic microenvironment. Moreover, dog BMSCs have not been characterized in situ by any of those markers. We conducted a systematic histopathological comparison of mouse, human, and dog BMSCs within their bone marrow architecture and microenvironment. Human and dog CD271+ BMSCs had a morphology, frequency, and distribution within trabecular bone marrow similar to those of mouse LepR+ BMSCs. However, mouse bone marrow had higher cellularity and megakaryocyte content. In conclusion, highly comparable bone marrow mesenchymal stromal cell distribution among the three species establishes the validity of using mouse and dog as a surrogate experimental model of hematopoietic stem cell-BMSC interactions. However, the distinct differences in adipocyte and megakaryocyte microenvironment content of mouse bone marrow and how they might influence hematopoietic stem cell interactions as compared with humans require further study.


Assuntos
Células-Tronco Mesenquimais/citologia , Animais , Células Cultivadas , Criança , Cães , Humanos , Células-Tronco Mesenquimais/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Proteínas do Tecido Nervoso/análise , Receptores para Leptina/análise , Receptores de Fator de Crescimento Neural/análise , Especificidade da Espécie , Nicho de Células-Tronco
12.
Cell Rep ; 36(2): 109352, 2021 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-34260921

RESUMO

Bone stroma contributes to the regulation of osteogenesis and hematopoiesis but also to fracture healing and disease processes. Mesenchymal stromal cells from bone (BMSCs) represent a heterogenous mixture of different subpopulations with distinct molecular and functional properties. The lineage relationship between BMSC subsets and their regulation by intrinsic and extrinsic factors are not well understood. Here, we show with mouse genetics, ex vivo cell differentiation assays, and transcriptional profiling that BMSCs from metaphysis (mpMSCs) and diaphysis (dpMSCs) are fundamentally distinct. Fate-tracking experiments and single-cell RNA sequencing indicate that bone-forming osteoblast lineage cells and dpMSCs, including leptin receptor-positive (LepR+) reticular cells in bone marrow, emerge from mpMSCs in the postnatal metaphysis. Finally, we show that BMSC fate is controlled by platelet-derived growth factor receptor ß (PDGFRß) signaling and the transcription factor Jun-B. The sum of our findings improves our understanding of BMSC development, lineage relationships, and differentiation.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/citologia , Linhagem da Célula , Animais , Animais Recém-Nascidos , Osso e Ossos/ultraestrutura , Diferenciação Celular , Células Endoteliais/citologia , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/ultraestrutura , Camundongos Endogâmicos C57BL , Especificidade de Órgãos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Transdução de Sinais , Análise de Célula Única , Células Estromais/citologia , Células Estromais/ultraestrutura , Transcrição Gênica
13.
Int J Mol Sci ; 22(13)2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34202056

RESUMO

Mesenchymal stem cells (MSC) are known for their vascular regeneration capacity by neoangiogenesis. Even though, several delivery approaches exist, particularly in the case of intravascular delivery, only limited number of cells reach the targeted tissue and are not able to remain on site. Applicated cells exhibit poor survival accompanied with a loss of functionality. Moreover, cell application techniques lead to cell death and impede the overall MSC function and survival. 3D cell spheroids mimic the physiological microenvironment, thus, overcoming these limitations. Therefore, in this study we aimed to evaluate and assess the feasibility of 3D MSCs spheroids for endovascular application, for treatment of ischemic peripheral vascular pathologies. Multicellular 3D MSC spheroids were generated at different cell seeding densities, labelled with ultra-small particles of iron oxide (USPIO) and investigated in vitro in terms of morphology, size distribution, mechanical stability as well as ex vivo with magnetic resonance imaging (MRI) to assess their trackability and distribution. Generated 3D spheroids were stable, viable, maintained stem cell phenotype and were easily trackable and visualized via MRI. MSC 3D spheroids are suitable candidates for endovascular delivery approaches in the context of ischemic peripheral vascular pathologies.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Esferoides Celulares , Animais , Técnicas de Cultura de Células , Diferenciação Celular , Humanos , Isquemia/diagnóstico , Isquemia/etiologia , Isquemia/metabolismo , Isquemia/terapia , Imageamento por Ressonância Magnética , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Doença Arterial Periférica/diagnóstico , Doença Arterial Periférica/etiologia , Doença Arterial Periférica/metabolismo , Doença Arterial Periférica/terapia , Esferoides Celulares/citologia , Esferoides Celulares/ultraestrutura , Coloração e Rotulagem
14.
Int J Mol Sci ; 22(9)2021 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-34066542

RESUMO

Biomineralization is the process by which living organisms generate organized mineral crystals. In human cells, this phenomenon culminates with the formation of hydroxyapatite, which is a naturally occurring mineral form of calcium apatite. The mechanism that explains the genesis within the cell and the propagation of the mineral in the extracellular matrix still remains largely unexplained, and its characterization is highly controversial, especially in humans. In fact, up to now, biomineralization core knowledge has been provided by investigations on the advanced phases of this process. In this study, we characterize the contents of calcium depositions in human bone mesenchymal stem cells exposed to an osteogenic cocktail for 4 and 10 days using synchrotron-based cryo-soft-X-ray tomography and cryo-XANES microscopy. The reported results suggest crystalline calcite as a precursor of hydroxyapatite depositions within the cells in the biomineralization process. In particular, both calcite and hydroxyapatite were detected within the cell during the early phase of osteogenic differentiation. This striking finding may redefine most of the biomineralization models published so far, taking into account that they have been formulated using murine samples while studies in human cell lines are still scarce.


Assuntos
Biomineralização/efeitos dos fármacos , Carbonato de Cálcio/farmacologia , Diferenciação Celular/efeitos dos fármacos , Durapatita/farmacologia , Células-Tronco Mesenquimais/citologia , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/ultraestrutura , Distribuição Normal
15.
J Endocrinol Invest ; 44(12): 2685-2698, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34024028

RESUMO

OBJECTIVES: Intracerebral hemorrhage (ICH) induced by diabetes results in further brain injury and nerve cell death. Bone marrow mesenchymal stem cell (BMSC) transplantation contributes to attenuating neurological deficits after ICH. This study investigated the mechanism of extracellular vesicles (EVs) derived from BMSCs in reducing neuroinflammation after diabetic ICH. METHODS: BMSC-EVs were isolated and identified. The rat model of db/db-ICH was established and the model rats were administered with EVs. miR-183-5p expression in brain tissues of db/db-ICH rats was detected. The brain injury of db/db-ICH rats was evaluated by measuring neurobehavioral score, brain water content and inflammatory factors. BV2 cells were cultured in vitro to establish high-glucose (HG)-Hemin-BV2 cell model. The levels of reactive oxygen species (ROS) and inflammatory factors in BV2 cells were measured, and BV2 cell viability and apoptosis were assessed. The targeting relationship between miR-183-5p and PDCD4 was predicted and verified. The activation of PDCD4/NLRP3 pathway in rat brain tissues and BV2 cells was detected. RESULTS: miR-183-5p expression was reduced in db/db-ICH rats brain tissues. BMSC-EVs ameliorated cranial nerve function, decreased brain water content and repressed inflammatory response by carrying miR-183-5p. BMSC-EVs mitigated HG-Hemin-BV2 cell injury, reduced ROS level and suppressed inflammatory response. miR-183-5p targeted PDCD4. PDCD4 promoted BV2 cell inflammation by activating the NLRP3 pathway. BMSC-EVs inhibited HG-Hemin-BV2 cell inflammation through the miR-183-5p/PDCD4/NLRP3 pathway, and inhibition of miR-183-5p reversed the protective effect of EVs. CONCLUSION: BMSC-EVs carried miR-183-5p into db/db-ICH rat brain tissues and repressed the NLRP3 pathway by targeting PDCD4, thus alleviating neuroinflammation after diabetic ICH.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Hemorragia Cerebral , Complicações do Diabetes , Vesículas Extracelulares/transplante , MicroRNAs/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Animais , Encéfalo/imunologia , Encéfalo/metabolismo , Células Cultivadas , Hemorragia Cerebral/etiologia , Hemorragia Cerebral/imunologia , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/terapia , Complicações do Diabetes/imunologia , Complicações do Diabetes/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/ultraestrutura , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/terapia , Fatores de Proteção , Ratos , Transdução de Sinais
16.
Cells Dev ; 167: 203687, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34058434

RESUMO

Bone marrow mesenchymal stem cells (BMSCs) have strong proliferative ability and multi-directional differentiation potential. Osteoarthritis is a degenerative joint disease that is closely related to the loss of osteogenic differentiation function of BMSCs. Autophagy, plays a crucial role in the maintenance of cellular functions, but its regulatory mechanism during the osteogenic differentiation of BMSCs remains unclear. In this study, we analyzed the differential gene networks and pathways during BMSC osteogenesis using bioinformatics, and further validated the regulatory roles of autophagy during the osteogenic differentiation of BMSCs in inflammatory condition in vitro. We found that Tumor necrosis factor alpha (TNF-α) treatment led to actin cytoskeleton rearrangements and inhibited osteogenic differentiation in BMSCs. In addition, TNF-α enhanced Rho-associated protein kinase 1 (ROCK1) expression and decreased autophagy activation. ROCK1 knockdown reduced Endoplasmic Reticulum stress (ER stress) and promoted autophagy, resulting reversion of osteogenic differentiation in BMSCs under inflammatory condition. Rapamycin reversed the TNF-α-induced decrease in osteogenesis of BMSCs, assessed by alkaline phosphatase (ALP) activity and Alizarin staining. Autophagy treated with inhibitor 3-Methyladenine (3-MA) further increased TNF-α-induced osteogenesis inhibition of BMSCs. Collectively, these results indicate that ER stress and dysfunction of autophagy promote inflammation-induced bone loss through the activation of ROCK1 signaling in BMSCs.


Assuntos
Autofagia , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Inflamação/patologia , Células-Tronco Mesenquimais/metabolismo , Transdução de Sinais , Quinases Associadas a rho/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/ultraestrutura , Camundongos Endogâmicos C57BL , Modelos Biológicos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Osteoporose/genética , Mapas de Interação de Proteínas/efeitos dos fármacos , Mapas de Interação de Proteínas/genética , Fator de Necrose Tumoral alfa/farmacologia
17.
Stem Cells Dev ; 30(8): 441-457, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33798004

RESUMO

Dental stem/progenitor cells are a promising cell sources for alveolar bone (AB) regeneration because of their same embryonic origin and superior osteogenic potential. However, their molecular processes during osteogenic differentiation remain unclear. The objective of this study was to identify the responsiveness of dental follicle cells (DFCs) and AB marrow-derived mesenchymal stem cells (ABM-MSCs) to recombinant human bone morphogenetic protein-2 (rhBMP-2). These cells expressed vimentin and MSC markers and did not express cytokeratin and hematopoietic stem cell markers and showed multilineage differentiation potential under specific culture conditions. DFCs exhibited higher proliferation and colony-forming unit-fibroblast efficiency than ABM-MSCs; rhBMP-2 induced DFCs to differentiate toward a cementoblast/osteoblast phenotype and ABM-MSCs to differentiate only toward a osteoblast phenotype; and rhBMP-2-induced DFCs exhibited higher osteogenic differentiation potential than ABM-MSCs. These cells adhered, grew, and produced extracellular matrix on nanohydroxyapatite/collagen/poly(l-lactide) (nHAC/PLA). During a 14-day culture on nHAC/PLA, the extracellular alkaline phosphatase (ALP) activity of DFCs decreased gradually and that of ABM-MSCs increased gradually; rhBMP-2 enhanced their extracellular ALP activity, intracellular osteocalcin (OCN), and osteopontin (OPN) protein expression; and DFCs exhibited higher extracellular ALP activity and intracellular OCN protein expression than ABM-MSCs. When implanted subcutaneously in severe combined immunodeficient mice for 3 months, DFCs+nHAC/PLA+rhBMP-2 obtained higher percentage of bone formation area, OCN, and cementum attachment protein expression and lower OPN expression than ABM-MSCs+nHAC/PLA+rhBMP-2. These results showed that DFCs possessed superior proliferation and osteogenic differentiation potential in vitro, and formed higher quantity and quality bones in vivo. It suggested that DFCs might exhibit a more sensitive responsiveness to rhBMP-2, so that DFCs enter a relatively mature stage of osteogenic differentiation earlier than ABM-MSCs after rhBMP-2 induction. The findings imply that these dental stem/progenitor cells are alternative sources for AB engineering in regenerative medicine, and developing dental tissue may provide better source for stem/progenitor cells.


Assuntos
Proteína Morfogenética Óssea 2/farmacologia , Diferenciação Celular/efeitos dos fármacos , Saco Dentário/citologia , Células-Tronco Mesenquimais/citologia , Osteogênese/efeitos dos fármacos , Células-Tronco/citologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Diferenciação Celular/genética , Células Cultivadas , Colágeno/metabolismo , Durapatita/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/ultraestrutura , Microscopia Eletrônica de Varredura , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/genética , Osteopontina/genética , Osteopontina/metabolismo , Poliésteres/metabolismo , Ratos Sprague-Dawley , Proteínas Recombinantes/farmacologia , Células-Tronco/metabolismo , Células-Tronco/ultraestrutura
18.
Int J Mol Sci ; 22(8)2021 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-33924614

RESUMO

The physiological O2 microenvironment of mesenchymal stem cells (MSCs) and osteoblasts and the dimensionality of a substrate are known to be important in regulating cell phenotype and function. By providing the physiologically normoxic environments of bone marrow (5%) and matrix (12%), we assessed their potential to maintain stemness, induce osteogenic differentiation, and enhance the material properties in the micropatterned collagen/silk fibroin scaffolds that were produced in 2D or 3D. Expression of osterix (OSX) and vascular endothelial growth factor A (VEGFA) was significantly enhanced in the 3D scaffold in all oxygen environments. At 21% O2, OSX and VEGFA expressions in the 3D scaffold were respectively 13,200 and 270 times higher than those of the 2D scaffold. Markers for assessing stemness were significantly more pronounced on tissue culture polystyrene and 2D scaffold incubated at 5% O2. At 21% O2, we measured significant increases in ultimate tensile strength (p < 0.0001) and Young's modulus (p = 0.003) of the 3D scaffold compared to the 2D scaffold, whilst 5% O2 hindered the positive effect of cell seeding on tensile strength. In conclusion, we demonstrated that the 3D culture of MSCs in collagen/silk fibroin scaffolds provided biomimetic cues for bone progenitor cells toward differentiation and enhanced the tensile mechanical properties.


Assuntos
Materiais Biomiméticos/farmacologia , Medula Óssea/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Oxigênio/metabolismo , Tecidos Suporte/química , Animais , Biomarcadores/metabolismo , Bombyx , Medula Óssea/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/ultraestrutura , Neovascularização Fisiológica/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Pressão Parcial , Ratos Sprague-Dawley , Resistência à Tração
19.
Biochem Biophys Res Commun ; 557: 151-158, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-33865223

RESUMO

Towards repairing bone defects, calcium sulfate and calcium phosphate cement have been recognized as promising bone grafts. However, the current bone cements are generally lack of proper porosity for cell migration and new tissue formation. On the other hand, porous scaffold cannot be delivered by injection, which limits its use its clinical use. Herein, we develop a novel tricalcium phosphate/calcium sulfate granule to overcome the limitations of injectable cements and traditional scaffolds. The biocompatible granule underwent in situ self-setting to form scaffold with porous structure after injection. It contributes to calcium deposition and upregulation of osteogenic genes of mesenchymal stem cells in a time-dependent manner. Within three months, cavitary bone defects of distal rabbit femurs implanted the granules exhibited better bone formation than those with those implanted with autologous bone.


Assuntos
Cimentos Ósseos/química , Regeneração Óssea/efeitos dos fármacos , Fosfatos de Cálcio/farmacologia , Sulfato de Cálcio/farmacologia , Osteogênese/efeitos dos fármacos , Engenharia Tecidual/métodos , Tecidos Suporte/química , Animais , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/ultraestrutura , Microscopia Eletrônica de Varredura , Osteoblastos/efeitos dos fármacos , Osteoblastos/ultraestrutura , Porosidade , Coelhos , Difração de Raios X
20.
J Vis Exp ; (168)2021 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-33720122

RESUMO

Magnetic nanoparticles, made of iron oxide, present a peculiar interest for a wide range of biomedical applications for which they are often internalized in cells and then left within. One challenge is to assess their fate in the intracellular environment with reliable and precise methodologies. Herein, we introduce the use of the vibrating sample magnetometer (VSM) to precisely quantify the integrity of magnetic nanoparticles within cells by measuring their magnetic moment. Stem cells are first labeled with two types of magnetic nanoparticles; the nanoparticles have the same core produced via a fast and efficient microwave-based nonaqueous sol gel synthesis and differ in their coating: the commonly used citric acid molecule is compared to polyacrylic acid. The formation of 3D cell-spheroids is then achieved via centrifugation and the magnetic moment of these spheroids is measured at different times with the VSM. The obtained moment is a direct fingerprint of the nanoparticles' integrity, with decreasing values indicative of a nanoparticle degradation. For both nanoparticles, the magnetic moment decreases over culture time revealing their biodegradation. A protective effect of the polyacrylic acid coating is also shown, when compared to citric acid.


Assuntos
Nanopartículas Magnéticas de Óxido de Ferro/química , Magnetometria , Células-Tronco Mesenquimais/metabolismo , Endocitose , Humanos , Nanopartículas Magnéticas de Óxido de Ferro/ultraestrutura , Células-Tronco Mesenquimais/ultraestrutura , Micro-Ondas , Soluções , Esferoides Celulares/metabolismo , Esferoides Celulares/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...