Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
BMC Res Notes ; 16(1): 280, 2023 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-37858260

RESUMO

OBJECTIVE: Differentiation of immortalized Mesenchymal Stromal Cells (iMSCs) into PDGFRα-positive cells under controlled growth conditions has several vital implications in functional studies concerned with the pathogenesis of Diabetic Gastroparesis (DGP). A study published previously by our research group demonstrated the importance of these cells as a novel, in-vitro model for investigating the functional role of neuronal nitric oxide synthase. The currently available methods require fresh differentiation of PDGFRα-positive cells for each round of experimentation. This leads to longer delays, higher usage of reagents, and inconsistency in reproducibility of experiments frequently. We thus aimed to establish through validation that cryopreserving and maintaining the iMSC-derived PDGFRα-positive cells for functional investigations help us to overcome these challenges. RESULTS: We demonstrated for the first time that the differentiated PDGFRα-positive cells from iMSCs can be cryopreserved and thawed to be used as per the experimental requirements with prolonged preservation of their characteristics. We assessed the viability of differentiated PDGFRα-positive cells pre- and post-freezing with the subsequent validation of their functional features using flow cytometry, qRT-PCR, and western blotting. We have been successful in demonstrating for the first time that the cryopreservation of previously differentiated PDGFRα-positive cells can be used as a feasible and cost-effective model for experimental reproducibility in functional studies of Diabetes Gastroparesis.


Assuntos
Células-Tronco Mesenquimais , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Diferenciação Celular , Criopreservação , Reprodutibilidade dos Testes
2.
J Mol Med (Berl) ; 101(11): 1379-1396, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37707557

RESUMO

Reperfusion after acute myocardial infarction further exaggerates cardiac injury and adverse remodeling. Irrespective of cardiac cell types, loss of specifically the α isoform of the protein kinase GSK-3 is protective in chronic cardiac diseases. However, the role of GSK-3α in clinically relevant ischemia/reperfusion (I/R)-induced cardiac injury is unknown. Here, we challenged cardiomyocyte-specific conditional GSK-3α knockout (cKO) and littermate control mice with I/R injury and investigated the underlying molecular mechanism using an in vitro GSK-3α gain-of-function model in AC16 cardiomyocytes post-hypoxia/reoxygenation (H/R). Analysis revealed a significantly lower percentage of infarct area in the cKO vs. control hearts post-I/R. Consistent with in vivo findings, GSK-3α overexpression promoted AC16 cardiomyocyte death post-H/R which was accompanied by an induction of reactive oxygen species (ROS) generation. Consistently, GSK-3α gain-of-function caused mitochondrial dysfunction by significantly suppressing mitochondrial membrane potential. Transcriptomic analysis of GSK-3α overexpressing cardiomyocytes challenged with hypoxia or H/R revealed that NOD-like receptor (NLR), TNF, NF-κB, IL-17, and mitogen-activated protein kinase (MAPK) signaling pathways were among the most upregulated pathways. Glutathione and fatty acid metabolism were among the top downregulated pathways post-H/R. Together, these observations suggest that loss of cardiomyocyte-GSK-3α attenuates cardiac injury post-I/R potentially through limiting the myocardial inflammation, mitochondrial dysfunction, and metabolic derangement. Therefore, selective inhibition of GSK-3α may provide beneficial effects in I/R-induced cardiac injury and remodeling. KEY MESSAGES: GSK-3α promotes cardiac injury post-ischemia/reperfusion (I/R). GSK-3α regulates inflammatory and metabolic pathways post-hypoxia/reoxygenation (H/R). GSK-3α overexpression upregulates NOD-like receptor (NLR), TNF, NF-kB, IL-17, and MAPK signaling pathways in cardiomyocytes post-H/R. GSK-3α downregulates glutathione and fatty acid metabolic pathways in cardiomyocytes post-H/R.


Assuntos
Doença da Artéria Coronariana , Infarto do Miocárdio , Traumatismo por Reperfusão , Camundongos , Animais , Quinase 3 da Glicogênio Sintase , Interleucina-17/metabolismo , Miócitos Cardíacos/metabolismo , Traumatismo por Reperfusão/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , NF-kappa B/metabolismo , Doença da Artéria Coronariana/metabolismo , Hipóxia/metabolismo , Reperfusão , Inflamação/metabolismo , Glutationa/metabolismo , Proteínas NLR/metabolismo , Ácidos Graxos/metabolismo , Ácidos Graxos/farmacologia , Apoptose
3.
Artigo em Inglês | MEDLINE | ID: mdl-37612859

RESUMO

BACKGROUND: Silver nanoparticles (AgNPs) are a focus of huge interest in biological research, including stem cell research. AgNPs synthesized using Cyperus conglomeratus root extract have been previously reported but their effects on mesenchymal stromal cells have yet to be investigated. OBJECTIVES: The aim of this study is to investigate the effects of C. conglomeratus-derived AgNPs on adipogenesis and osteogenesis of mesenchymal stromal cells. METHODS: AgNPs were synthesized using C. conglomeratus root extract, and the phytochemicals involved in AgNPs synthesis were analyzed using gas chromatography-mass spectrometry (GC-MS). The cytotoxicity of the AgNPs was tested on telomerase-transformed immortalized human bone marrow-derived MSCs-hTERT (iMSC3) and human osteosarcoma cell line (MG-63) using MTT and apoptosis assays. The uptake of AgNPs by both cells was confirmed using inductively coupled plasma-optical emission spectrometry (ICP-OES). Furthermore, the effect of AgNPs on iMSC3 adipogenesis and osteogenesis was analyzed using stain quantification and reverse transcription-quantitative polymerase chain reaction (RT-qPCR). RESULTS: The phytochemicals predominately identified in both the AgNPs and C. conglomeratus root extract were carbohydrates. The AgNP concentrations tested using MTT and apoptosis assays (0.5-64 µg/ml and 1,4 and 32 µg/ml, respectively) showed no significant cytotoxicity on iMSC3 and MG-63. The AgNPs were internalized in a concentration-dependent manner in both cell types. Additionally, the AgNPs exhibited a significant negative effect on osteogenesis but not on adipogenesis. CONCLUSION: C. conglomeratus-derived AgNPs had an impact on the differentiation capacity of iMSC3. Our results indicated that C. conglomeratus AgNPs and the associated phytochemicals could exhibit potential medical applications.

4.
Heliyon ; 8(8): e10384, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36090221

RESUMO

Bone marrow-derived mesenchymal stem cells (BMSCs) have therapeutic potential for spinal cord injury (SCI). We have shown that insulin-like growth factor 1 (IGF-1) enhances the cellular proliferation and survivability of BMSCs-derived neural progenitor cells (NPCs) by downregulating miR-22-3p. However, the functional application of BMSCs-derived NPCs has not been investigated fully. In this study, we demonstrate that knockdown of endogenous miR-22-3p in BMSCs-derived NPCs upregulates Akt1 expression, leading to enhanced cellular proliferation. RNASeq analysis reveals 3,513 differentially expressed genes in NPCs. The upregulated genes in NPCs enrich the gene ontology term associated with nervous system development. Terminally differentiated NPCs generate cells with neuronal-like morphology and phenotypes. Transplantation of NPCs in the SCI rat model results in better recovery in locomotor and sensory functions 4 weeks after transplantation. Altogether, the result of this study demonstrate that NPCs derived with IGF-1 supplementation could be differentiated into functional neural lineage cells and are optimal for stem cell therapy in SCI.

5.
BMC Genom Data ; 23(1): 17, 2022 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-35264099

RESUMO

BACKGROUND: Differentiation of Immortalized Human Bone Marrow Mesenchymal Stromal Cells - hTERT (iMSC3) into adipocytes is in vitro model of obesity. In our earlier study, rosiglitazone enhanced adipogenesis particularly the brown adipogenesis of iMSC3. In this study, the transcriptomic profiles of iMSC3 derived adipocytes with and without rosiglitazone were analyzed through mRNA sequencing. RESULTS: A total of 1508 genes were differentially expressed between iMSC3 and the derived adipocytes without rosiglitazone treatment. GO and KEGG enrichment analyses revealed that rosiglitazone regulates PPAR and PI3K-Akt pathways. The constant rosiglitazone treatment enhanced the expression of Fatty Acid Binding Protein 4 (FABP4) which enriched GO terms such as fatty acid binding, lipid droplet, as well as white and brown fat cell differentiation. Moreover, the constant treatment upregulated several lipid droplets (LDs) associated proteins such as PLIN1. Rosiglitazone also activated the receptor complex PTK2B that has essential roles in beige adipocytes thermogenic program. Several uniquely expressed novel regulators of brown adipogenesis were also expressed in adipocytes derived with rosiglitazone: PRDM16, ZBTB16, HOXA4, and KLF15 in addition to other uniquely expressed genes. CONCLUSIONS: Rosiglitazone regulated several differentially regulated genes and non-coding RNAs that warrant further investigation about their roles in adipogenesis particularly brown adipogenesis.


Assuntos
Células-Tronco Mesenquimais , Telomerase , Adipócitos , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Rosiglitazona/farmacologia , Telomerase/genética , Transcriptoma
6.
Saudi J Biol Sci ; 28(11): 6324-6331, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34764753

RESUMO

The plants extracts are widely used in traditional medicines and hence considered a potential source for drug discovery. In this study, we assessed the phytochemical composition of Asplenium dalhousiae and Asplenium polypodioides in acetone extracts and checked its antiproliferative potential in MDA-MB-231 cells. We found that both plants are rich in phenolic and flavonoid compounds and are efficient in antioxidant activities. The total phenolic compounds in A. dalhousiae were 44.15 ± 1.38 µg/mg whereas in A. polypodioides were 27.73 ± 1.35 µg/mg. Total flavonoids in A. dalhousiae were 105.39 ± 2.92 µg/mg whereas in A. polypodioides were 101.56 ± 1.75 µg/mg. The ferric reducing power assay indicates 66.38 ± 2.6% reduction by A. dalhousiae whereas 78.43 ± 0.47% reduction by A. polypodioides. Similarly, the total antioxidant capacity of A. dalhousiae was found to be 59.95 ± 1.13 whereas for A. polypodioides the recorded value was 33.03 ± 1.67%. Using GCMS analysis, we identified 25 compounds in A. dalhousiae whereas 26 in A. polypodioides. Four of these compounds are common in both plants. The morphological study and MTT assay revealed that both plants have antiproliferative potential as both plants exerted significant effects on the shape of the MDA-MB-231 cells and inhibited cellular proliferation in time and dose dependent manner. We conclude that both Asplenium plants have potential anticancer compounds.

7.
Saudi J Biol Sci ; 28(6): 3225-3234, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34121859

RESUMO

Ficus latex is rich in polyphenolic compounds and hence considered as an antioxidant and anti-proliferative. Many studies are available on Ficus carica (common fig) whereas Ficus salicifolia is less studied. F. salicifolia grows in a harsh dry environment, therefore its latex was selected in the current study along with the F. carica for their comparative anti-cancer potential and the involved molecular mechanism. Triple-negative breast cancer (TNBC) derived MDA-MB-231 cells were used in the study. MTT and morphological studies indicated that the latex of both plants has anti-proliferative effects. To know their anti-metastatic effects, a wound-healing assay was performed. Both species were able to maintain the wound size compared to the untreated cells indicating their anti-metastatic effects. Using a DNA damage assay kit, we found that both fig species have genotoxic and cytotoxic effects in MDA-MB-231 cells compared to the untreated control. To know the potential molecular mechanism involved, we used a human kinase array kit. We found that ERK2, CREB, and AKT2 were downregulated after treatment the MDA-Mb-231 cells with the latex of F. carica. We assumed that F. salicifolia will also affect the same pathways, however after confirmation through real-time (RT)-PCR, downregulations of the above mentioned pathways were confirmed in cells treated with F. carica latex, however, in cells treated with F. salicifolia the selected genes were upregulated at the transcriptional level. We conclude that latex of both species of ficus have anti-cancer effects in MDA-MB-231 cells, however differ in their level of toxicity and the mechanism of action at the molecular level.

8.
Int J Mol Sci ; 22(7)2021 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-33805311

RESUMO

It is evident that depletion of interstitial cells and dysfunction of nitric oxide (NO) pathways are key players in development of several gastrointestinal (GI) motility disorders such as diabetic gastroparesis (DGP). One of the main limitations of DGP research is the lack of isolation methods that are specific to interstitial cells, and therefore conducting functional studies is not feasible. The present study aims (i) to differentiate telomerase transformed mesenchymal stromal cells (iMSCs) into platelet-derived growth factor receptor-α-positive cells (PDGFRα-positive cells) using connective tissue growth factor (CTGF) and L-ascorbic acids; (ii) to investigate the effects of NO donor and inhibitor on the survival rate of differentiated PDGFRα-positive cells; and (iii) to evaluate the impact of increased glucose concentrations, mimicking diabetic hyperglycemia, on the gene expression of neuronal nitric oxide synthase (nNOS). A fibroblastic differentiation-induction medium supplemented with connective tissue growth factor was used to differentiate iMSCs into PDGFRα-positive cells. The medium was changed every day for 21 days to maintain the biological activity of the growth factors. Gene and protein expression, scanning electron and confocal microscopy, and flow cytometry analysis of several markers were conducted to confirm the differentiation process. Methyl tetrazolium cell viability, nitrite measurement assays, and immunostaining were used to investigate the effects of NO on PDGFRα-positive cells. The present study, for the first time, demonstrated the differentiation of iMSCs into PDGFRα-positive cells. The outcomes of the functional studies showed that SNAP (NO donor) increased the survival rate of differentiated PDGFRα-positive cells whereas LNNA (NO inhibitor) attenuated these effects. Further experimentations revealed that hyperglycemia produced a significant increase in expression of nNOS in PDGFRα-positive cells. Differentiation of iMSCs into PDGFRα-positive cells is a novel model to conduct functional studies and to investigate the involvement of NO pathways. This will help in identifying new therapeutic targets for treatment of DGP.


Assuntos
Diferenciação Celular , Células Intersticiais de Cajal/enzimologia , Células-Tronco Mesenquimais/fisiologia , Modelos Biológicos , Óxido Nítrico Sintase Tipo I , Receptor alfa de Fator de Crescimento Derivado de Plaquetas , Animais , Medula Óssea , Células Cultivadas , Complicações do Diabetes , Gastroparesia , Humanos , Células Intersticiais de Cajal/metabolismo
9.
Saudi J Biol Sci ; 27(12): 3390-3396, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33304147

RESUMO

The date palm (Phoenix dactylifera L.) is an important fruit crop with significant pharmaceutical potential. Little data are available on comparative pharmaceutical importance of the date pits. We designed this study to assess the antitumorigenic effects of date palm pits extracts from different Emiratis varieties. We used MDA-MB-231 cells derived from triple negative breasts cancer tissues as a model. We found that out of the 17 date pits extracts from 6 Emiratis varieties, three (Khalas extract in water + acetone (1:1), Abu-Maan extract in MeOH + Chloroform (1:1) and Mabroom extract in water + acetone (1:1)) were found effectively cytotoxic and changed morphology of cells in dose and time dependent manner. We found the maximum effect at 2.5 mg/mL concentration at 72 h. We calculated IC50 values for these varieties at 24 h. IC50 values for Khalas, Abu-Maan and Mabroom were 0.982 mg/mL, 1.149 mg/mL and 2.213 mg/mL respectively. We treated the cells with IC50 values of extracts and observed changes in protein profile using human kinase array kit. After analyzing the results, we suggest that EGFR/ERK/FAK pathway, eNOS and src family proteins are targets of these extracts. We conclude that date pits extracts can be a possible therapeutic agent against cancer and we suggest further studies.

10.
Cell Biosci ; 10: 126, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33133516

RESUMO

INTRODUCTION: Mesenchymal stem cells (MSCs) isolated from bone marrow have different developmental origins, including neural crest. MSCs can differentiate into neural progenitor-like cells (NPCs) under the influence of bFGF and EGF. NPCs can terminally differentiate into neurons that express beta-III-tubulin and elicit action potential. The main aim of the study was to identify key genetic markers involved in differentiation of MSCs into NPCs through transcriptomic analysis. METHOD: Total RNA was isolated from MSCs and MSCs-derived NPCs followed by cDNA library construction for transcriptomic analysis. Sample libraries that passed the quality and quantity assessments were subjected to high throughput mRNA sequencing using NextSeq®500. Differential gene expression analysis was performed using the DESeq2 R package with MSC samples being a reference group. The expression of eight differentially regulated genes was counter validated using real-time PCR. RESULTS: In total, of the 3,252 differentially regulated genes between MSCs and NPCs with two or more folds, 1,771 were upregulated genes, whereas 1,481 were downregulated in NPCs. Amongst these differential genes, 104 transcription factors were upregulated, and 45 were downregulated in NPCs. Neurogenesis related genes were upregulated in NPCs and the main non-redundant gene ontology (GO) terms enriched in NPCs were the autonomic nervous system, cell surface receptor signalling pathways), extracellular structure organisation, and programmed cell death. The main non-redundant GO terms enriched in MSCs included cytoskeleton organisation cytoskeleton structural constituent, mitotic cell cycle), and the mitotic cell cycle process Gene set enrichment analysis also confirmed cell cycle regulated pathways as well as Biocarta integrin pathway were upregulated in MSCs. Transcription factors enrichment analysis by ChEA3 revealed Foxs1 and HEYL, amongst the top five transcription factors, inhibits and enhances, respectively, the NPCs differentiation of MSCs. CONCLUSIONS: The vast differences in the transcriptomic profiles between NPCs and MSCs revealed a set of markers that can identify the differentiation stage of NPCs as well as provide new targets to enhance MSCs differentiation into NPCs.

11.
Int J Mol Sci ; 20(7)2019 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-30939750

RESUMO

Obesity is a major risk for diabetes. Brown adipose tissue (BAT) mediates production of heat while white adipose tissue (WAT) function in the storage of fat. Roles of BAT in the treatment of obesity and related disorders warrants more investigation. Peroxisome proliferator activator receptor gamma (PPAR-γ) is the master regulator of both BAT and WAT adipogenesis and has roles in glucose and fatty acid metabolism. Adipose tissue is the major expression site for PPAR-γ. In this study, the effects of rosiglitazone on the brown adipogenesis and the association of MAPK and PI3K pathways was investigated during the in vitro adipogenic differentiation of telomerase transformed mesenchymal stromal cells (iMSCs). Our data indicate that 2 µM rosiglitazone enhanced adipogenesis by over-expression of PPAR-γ and C/EBP-α. More specifically, brown adipogenesis was enhanced by the upregulation of EBF2 and UCP-1 and evidenced by multilocular fatty droplets morphology of the differentiated adipocytes. We also found that rosiglitazone significantly activated MAPK and PI3K pathways at the maturation stage of differentiation. Overall, the results indicate that rosiglitazone induced overexpression of PPAR-γ that in turn enhanced adipogenesis, particularly browning adipogenesis. This study reports the browning effects of rosiglitazone during the differentiation of iMSCs into adipocytes in association with the activation of MAPK and PI3K signaling pathways.


Assuntos
Adipócitos Marrons/efeitos dos fármacos , Adipogenia , Hipoglicemiantes/farmacologia , Sistema de Sinalização das MAP Quinases , Rosiglitazona/farmacologia , Adipócitos Marrons/citologia , Adipócitos Marrons/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Linhagem Celular , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
12.
Genom Data ; 5: 201-5, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26484256

RESUMO

Recently there has been growing interest in the differentiation of mesenchymal stem cells (MSCs) into neural lineages. Research suggests that MSCs can be differentiated into neural progenitor-like cells (NPCs) under the specific influence of paracrine factors particularly epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF). Our recent research has found that the addition of insulin-like growth factor 1 (IGF-1) with the combination of the EGF and bFGF could significantly improve the growth and survivability of MSC-derived NPCs. To unravel the molecular mechanism of the improved differentiation we compared the microRNA expression profiles of the differentiation under various combinations of growth factors. MSCs were differentiated into neural lineage in 3 groups; Group A (EGF + bFGF), Group B (EGF + bFGF + IGF-1), and Group C (without growth factor). Regulated microRNAs during the early differentiation were identified by detailed microRNA profiling using Affymetrix GeneChip version 2.0 at three time intervals (day 1, day 3 and day 5). The data were deposited in the Gene Expression Omnibus, series GSE60060.

13.
Int J Mol Sci ; 16(5): 9693-718, 2015 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-25938966

RESUMO

Insulin-like growth factor 1 (IGF-1) enhances cellular proliferation and reduces apoptosis during the early differentiation of bone marrow derived mesenchymal stem cells (BMSCs) into neural progenitor-like cells (NPCs) in the presence of epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF). BMSCs were differentiated in three groups of growth factors: (A) EGF + bFGF, (B) EGF + bFGF + IGF-1, and (C) without growth factor. To unravel the molecular mechanisms of the NPCs derivation, microarray analysis using GeneChip miRNA arrays was performed. The profiles were compared among the groups. Annotated microRNA fingerprints (GSE60060) delineated 46 microRNAs temporally up-regulated or down-regulated compared to group C. The expressions of selected microRNAs were validated by real-time PCR. Among the 46 microRNAs, 30 were consistently expressed for minimum of two consecutive time intervals. In Group B, only miR-496 was up-regulated and 12 microRNAs, including the let-7 family, miR-1224, miR-125a-3p, miR-214, miR-22, miR-320, miR-708, and miR-93, were down-regulated. Bioinformatics analysis reveals that some of these microRNAs (miR-22, miR-214, miR-125a-3p, miR-320 and let-7 family) are associated with reduction of apoptosis. Here, we summarize the roles of key microRNAs associated with IGF-1 in the differentiation of BMSCs into NPCs. These findings may provide clues to further our understanding of the mechanisms and roles of microRNAs as key regulators of BMSC-derived NPC maintenance.


Assuntos
Células da Medula Óssea/citologia , Fator de Crescimento Epidérmico/farmacologia , Fator 2 de Crescimento de Fibroblastos/farmacologia , Perfilação da Expressão Gênica , Fator de Crescimento Insulin-Like I/farmacologia , Células-Tronco Mesenquimais/citologia , MicroRNAs/genética , Células-Tronco Neurais/citologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Análise por Conglomerados , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Ontologia Genética , Redes Reguladoras de Genes/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , MicroRNAs/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Software , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
14.
BMC Neurosci ; 15: 91, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25047045

RESUMO

BACKGROUND: There has been increasing interest recently in the plasticity of mesenchymal stem cells (MSCs) and their potential to differentiate into neural lineages. To unravel the roles and effects of different growth factors in the differentiation of MSCs into neural lineages, we have differentiated MSCs into neural lineages using different combinations of growth factors. Based on previous studies of the roles of insulin-like growth factor 1 (IGF-1) in neural stem cell isolation in the laboratory, we hypothesized that IGF-1 can enhance proliferation and reduce apoptosis in neural progenitor-like cells (NPCs) during differentiation of MSCs into NCPs.We induced MSCs differentiation under four different combinations of growth factors: (A) EGF + bFGF, (B) EGF + bFGF + IGF-1, (C) EGF + bFGF + LIF, (D) EGF + bFGF + BDNF, and (E) without growth factors, as a negative control. The neurospheres formed were characterized by immunofluorescence staining against nestin, and the expression was measured by flow cytometry. Cell proliferation and apoptosis were also studied by MTS and Annexin V assay, respectively, at three different time intervals (24 hr, 3 days, and 5 days). The neurospheres formed in the four groups were then terminally differentiated into neuron and glial cells. RESULTS: The four derived NPCs showed a significantly higher expression of nestin than was shown by the negative control. Among the groups treated with growth factors, NPCs treated with IGF-1 showed the highest expression of nestin. Furthermore, NPCs derived using IGF-1 exhibited the highest cell proliferation and cell survival among the treated groups. The NPCs derived from IGF-1 treatment also resulted in a better yield after the terminal differentiation into neurons and glial cells than that of the other treated groups. CONCLUSIONS: Our results suggested that IGF-1 has a crucial role in the differentiation of MSCs into neuronal lineage by enhancing the proliferation and reducing the apoptosis in the NPCs. This information will be beneficial in the long run for improving both cell-based and cell-free therapy for neurodegenerative diseases.


Assuntos
Fator de Crescimento Insulin-Like I/metabolismo , Células-Tronco Mesenquimais/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Apoptose/fisiologia , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Células Cultivadas , Fator de Crescimento Insulin-Like I/farmacologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Células-Tronco Mesenquimais/citologia , Nestina/metabolismo , Células-Tronco Neurais/citologia , Neuroglia/citologia , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Ratos Sprague-Dawley
15.
Fertil Steril ; 95(8): 2564-7.e1-8, 2011 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-21575943

RESUMO

We report the first quantitative assessment of DNA methylation for any gene in the human preimplantation embryo to reveal that imprints exist at KvDMR1, RB1, SNRPN, and GRB10 in the human blastocyst. For comparison, in two human embryonic stem cell lines, imprints were also observed at KvDMR1, SNRPN, GRB10, and other imprinted loci, whereas RB1 and MEG3 were hypermethylated.


Assuntos
Blastocisto/fisiologia , Metilação de DNA , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Impressão Genômica , Diagnóstico Pré-Implantação/métodos , Análise de Sequência de DNA/métodos , Linhagem Celular , Técnicas de Cultura Embrionária , Proteína Adaptadora GRB10/genética , Humanos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Proteínas/genética , RNA Longo não Codificante , Proteína do Retinoblastoma/genética , Proteínas Centrais de snRNP/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA