Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
World J Stem Cells ; 16(4): 410-433, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38690517

RESUMO

BACKGROUND: Stem cells are undifferentiated cells that possess the potential for self-renewal with the capacity to differentiate into multiple lineages. In humans, their limited numbers pose a challenge in fulfilling the necessary demands for the regeneration and repair of damaged tissues or organs. Studies suggested that mesenchymal stem cells (MSCs), necessary for repair and regeneration via transplantation, require doses ranging from 10 to 400 million cells. Furthermore, the limited expansion of MSCs restricts their therapeutic application. AIM: To optimize a novel protocol to achieve qualitative and quantitative expansion of MSCs to reach the targeted number of cells for cellular transplantation and minimize the limitations in stem cell therapy protocols. METHODS: Human umbilical cord (hUC) tissue derived MSCs were obtained and re-cultured. These cultured cells were subjected to the following evaluation procedures: Immunophenotyping, immunocytochemical staining, trilineage differentiation, population doubling time and number, gene expression markers for proliferation, cell cycle progression, senescence-associated ß-galactosidase assay, human telomerase reverse transcriptase (hTERT) expression, mycoplasma, cytomegalovirus and endotoxin detection. RESULTS: Analysis of pluripotent gene markers Oct4, Sox2, and Nanog in recultured hUC-MSC revealed no significant differences. The immunophenotypic markers CD90, CD73, CD105, CD44, vimentin, CD29, Stro-1, and Lin28 were positively expressed by these recultured expanded MSCs, and were found negative for CD34, CD11b, CD19, CD45, and HLA-DR. The recultured hUC-MSC population continued to expand through passage 15. Proliferative gene expression of Pax6, BMP2, and TGFb1 showed no significant variation between recultured hUC-MSC groups. Nevertheless, a significant increase (P < 0.001) in the mitotic phase of the cell cycle was observed in recultured hUC-MSCs. Cellular senescence markers (hTERT expression and ß-galactosidase activity) did not show any negative effect on recultured hUC-MSCs. Additionally, quality control assessments consistently confirmed the absence of mycoplasma, cytomegalovirus, and endotoxin contamination. CONCLUSION: This study proposes the development of a novel protocol for efficiently expanding stem cell population. This would address the growing demand for larger stem cell doses needed for cellular transplantation and will significantly improve the feasibility of stem cell based therapies.

2.
Cell Biochem Funct ; 42(3): e4008, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38613198

RESUMO

Temporal phases of wound healing and their corresponding healing factors are essential in wound regeneration. Mesenchymal stem cells (MSCs) accelerate wound healing via their paracrine secretions by enhancing cell migration, angiogenesis, and reducing inflammation. This study evaluated the local therapeutic effect of human umbilical cord MSCs (hUCMSCs) in the healing of cold-induced burn wounds. An in vitro wound (scratch) was developed in rat skin fibroblasts. The culture was maintained in the conditioned medium (CM) which was prepared by inducing an artificial wound in hUCMSCs in a separate experiment. Treated fibroblasts were analyzed for the gene expression profile of healing mediators involved in wound closure. Findings revealed enhanced cell migration and increased levels of healing mediators in the treated fibroblasts relative to the untreated group. Cold-induced burn wounds were developed in Wistar rats, followed by a single injection of hUCMSCs. Wound healing pattern was examined based on the healing phases: hemostasis/inflammation (Days 1, 3), cell proliferation (Day 7), and remodeling (Day 14). Findings exhibited enhanced wound closure in the treated wound. Gene expression, histological, and immunohistochemical analyses further confirmed enhanced wound regeneration after hUCMSC transplantation. Temporal gene expression profile revealed that the level of corresponding cytokines was substantially increased in the treated wound as compared with the control, indicating improvement in the processes of angiogenesis and remodeling, and a substantial reduction in inflammation. Histology revealed significant collagen formation along with regenerated skin layers and appendages, whereas immunohistochemistry exhibited increased neovascularization during remodeling. Leukocyte infiltration was also suppressed in the treated group. Overall findings demonstrate that a single dose of hUCMSCs enhances wound healing in vivo, and their secreted growth factors accelerate cell migration in vitro.


Assuntos
Queimaduras , Células-Tronco , Animais , Feminino , Humanos , Ratos , Queimaduras/terapia , Inflamação , Ratos Wistar , Cicatrização
3.
Cell Biol Int ; 48(5): 594-609, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38321826

RESUMO

The regeneration of osteochondral lesions by tissue engineering techniques is challenging due to the lack of physicochemical characteristics and dual-lineage (osteogenesis and chondrogenesis). A scaffold with better mechanical properties and dual lineage capability is required for the regeneration of osteochondral defects. In this study, a hydrogel prepared from decellularized human umbilical cord tissue was developed and evaluated for osteochondral regeneration. Mesenchymal stem cells (MSCs) isolated from the umbilical cord were seeded with hydrogel for 28 days, and cell-hydrogel composites were cultured in basal and osteogenic media. Alizarin red staining, quantitative polymerase chain reaction, and immunofluorescent staining were used to confirm that the hydrogel was biocompatible and capable of inducing osteogenic differentiation in umbilical cord-derived MSCs. The findings demonstrate that human MSCs differentiated into an osteogenic lineage following 28 days of cultivation in basal and osteoinductive media. The expression was higher in the cell-hydrogel composites cultured in osteoinductive media, as evidenced by increased levels of messenger RNA and protein expression of osteogenic markers as compared to basal media cultured cell-hydrogel composites. Additionally, calcium deposits were also observed, which provide additional evidence of osteogenic differentiation. The findings demonstrate that the hydrogel is biocompatible with MSCs and possesses osteoinductive capability in vitro. It may be potentially useful for osteochondral regeneration.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Osteogênese/genética , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Células Cultivadas , Engenharia Tecidual/métodos , Hidrogéis/química , Alicerces Teciduais
4.
Cell Biochem Funct ; 42(2): e3946, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38379227

RESUMO

The underlying pathophysiology of nonhealing chronic wounds is poorly understood due to the changes occurring at the gene level and the complexity arising in their proteomic profile. Here, we elucidated the temporal and differential profile of the normal and diabetic wound-healing mediators along with their interactions and associated pathways. Skin tissues corresponding to normal and diabetic wounds were isolated at Days 0, 3, 6, and 9 representing different healing phases. Temporal gene expression was analyzed by quantitative real-time PCR. Concurrently, differential protein patterns in the wound tissues were identified by Nano LC-ESI-TOF mass spectrometry and later confirmed by Western blot analysis. Gene ontology annotation, protein-protein interaction, and protein pathway analysis were performed using DAVID, PANTHER, and STRING bioinformatics resources. Uniquely identified proteins (complement C3, amyloid beta precursor protein, and cytoplasmic linker associated protein 2) in the diabetic wound tissue implied that these proteins are involved in the pathogenesis of diabetic wound. They exhibit enhanced catalytic activity, trigger pathways linked with inflammation, and negatively regulate wound healing. However, in the normal wound tissue, axin 1, chondroitin sulfate proteoglycan 4, and sphingosine-1-phosphate receptor were identified, which are involved in proliferation, angiogenesis, and remodeling. Our findings demonstrate the correlation between elevated gene expression of tumor necrosis factor-α, interleukin (IL)-1ß, and identified mediators: aryl hydrocarbon receptor nuclear translocator, 5'-aminolevulinate synthase 2, and CXC-family, that inflicted an inflammatory response by activating downstream MAPK, JAK-STAT, and NF-κB pathways. Similarly, in normal wound tissue, the upregulated IL-4 and hepatocyte growth factor levels in conjunction with the identified proteins, serine/threonine-protein kinase mTOR and peroxisome proliferator-activated receptor gamma, played a significant role in the cellular response to platelet-derived growth factor stimulus, dermal epithelialization, and cell proliferation, processes associated with the repair mechanism. Furthermore, Western blot analysis indicated elevated levels of inflammatory markers and reduced levels of proliferative and angiogenic factors in the diabetic wound.


Assuntos
Diabetes Mellitus , Cicatrização , Humanos , Peptídeos beta-Amiloides/metabolismo , Proteômica , Pele/patologia , Diabetes Mellitus/metabolismo
5.
J Biomed Mater Res A ; 112(7): 1041-1056, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38380793

RESUMO

Extracellular matrix-based bio-scaffolds are useful for tissue engineering as they retain the unique structural, mechanical, and physiological microenvironment of the tissue thus facilitating cellular attachment and matrix activities. However, considering its potential, a comprehensive understanding of the protein profile remains elusive. Herein, we evaluate the impact of decellularization on the human amniotic membrane (hAM) based on its proteome profile, physicochemical features, as well as the attachment, viability, and proliferation of umbilical cord-derived mesenchymal stem cells (hUC-MSC). Proteome profiles of decellularized hAM (D-hAM) were compared with hAM, and gene ontology (GO) enrichment analysis was performed. Proteomic data revealed that D-hAM retained a total of 249 proteins, predominantly comprised of extracellular matrix proteins including collagens (collagen I, collagen IV, collagen VI, collagen VII, and collagen XII), proteoglycans (biglycan, decorin, lumican, mimecan, and versican), glycoproteins (dermatopontin, fibrinogen, fibrillin, laminin, and vitronectin), and growth factors including transforming growth factor beta (TGF-ß) and fibroblast growth factor (FGF) while eliminated most of the intracellular proteins. Scanning electron microscopy was used to analyze the epithelial and basal surfaces of D-hAM. The D-hAM displayed variability in fibril morphology and porosity as compared with hAM, showing loosely packed collagen fibers and prominent large pore areas on the basal side of D-hAM. Both sides of D-hAM supported the growth and proliferation of hUC-MSC. Comparative investigations, however, demonstrated that the basal side of D-hAM displayed higher hUC-MSC proliferation than the epithelial side. These findings highlight the importance of understanding the micro-environmental differences between the two sides of D-hAM while optimizing cell-based therapeutic applications.


Assuntos
Âmnio , Células-Tronco Mesenquimais , Proteoma , Cordão Umbilical , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Âmnio/citologia , Âmnio/química , Âmnio/metabolismo , Cordão Umbilical/citologia , Proteoma/análise , Proliferação de Células , Matriz Extracelular Descelularizada/química , Materiais Biocompatíveis/química
6.
Cell Biochem Funct ; 41(7): 833-844, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37814478

RESUMO

Diabetes is one of the major health issues globally. Type 1 diabetes mellitus develops due to the destruction of pancreatic ß cells. Mesenchymal stem cells (MSCs) having remarkable self-renewal and differentiation potential, can regenerate ß cells. MSCs preconditioned with bioactive small molecules possess enhanced biological features and therapeutic potential under in vivo environment. Interestingly, compounds of naphthoquinone class possess antidiabetic and anti-inflammatory properties, and can be explored as potential candidates for preconditioning MSCs. This study analyzed the effect of lawsone-preconditioned human umbilical cord MSCs (hUMSCs) on the regeneration of ß cells in the streptozotocin (STZ)-induced Type 1 diabetes (T1D) rats. hUMSCs were isolated and characterized for the presence of surface markers. MSCs were preconditioned with optimized concentration of lawsone. T1D rat model was established by injecting 50 mg/kg of STZ intraperitoneally. Untreated and lawsone-preconditioned hUMSCs were transplanted into the diabetic rats via tail vein. Fasting blood sugar and body weight were monitored regularly for 4 weeks. Pancreas was harvested and ß cell regeneration was evaluated by hematoxylin and eosin staining, and gene expression analysis. Immunohistochemistry was also done to assess the insulin expression. Lawsone-preconditioned hUMSCs showed better anti-hyperglycemic effect in comparison with untreated hUMSCs. Histological analysis presented the regeneration of islets of Langerhans with upregulated expression of ßcell genes and reduced expression of inflammatory markers. Immunohistochemistry revealed strong insulin expression in the preconditioned hUMSCs compared with the untreated hUMSCs. It is concluded from the present study that lawsone-preconditioned hMSCs were able to exhibit pronounced anti-hyperglycemic effect in vivo compared with hUMSCs alone.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Naftoquinonas , Ratos , Humanos , Animais , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Experimental/metabolismo , Naftoquinonas/farmacologia , Naftoquinonas/metabolismo , Células-Tronco Mesenquimais/metabolismo , Insulina/metabolismo , Hipoglicemiantes/farmacologia
7.
World J Stem Cells ; 15(8): 821-841, 2023 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-37700819

RESUMO

BACKGROUND: Cardiovascular diseases particularly myocardial infarction (MI) are the leading cause of mortality and morbidity around the globe. As cardiac tissue possesses very limited regeneration potential, therefore use of a potent small molecule, inhibitor Wnt production-4 (IWP-4) for stem cell differentiation into cardiomyocytes could be a promising approach for cardiac regeneration. Wnt pathway inhibitors may help stem cells in their fate determination towards cardiomyogenic lineage and provide better homing and survival of cells in vivo. Mesenchymal stem cells (MSCs) derived from the human umbilical cord have the potential to regenerate cardiac tissue, as they are easy to isolate and possess multilineage differentiation capability. IWP-4 may promote the differentiation of MSCs into the cardiac lineage. AIM: To evaluate the cardiac differentiation ability of IWP-4 and its subsequent in vivo effects. METHODS: Umbilical cord tissue of human origin was utilized to isolate the MSCs which were characterized by their morphology, immunophenotyping of surface markers specific to MSCs, as well as by tri-lineage differentiation capability. Cytotoxicity analysis was performed to identify the optimal concentration of IWP-4. MSCs were treated with 5 µM IWP-4 at two different time intervals. Differentiation of MSCs into cardiomyocytes was evaluated at DNA and protein levels. The MI rat model was developed. IWP-4 treated as well as untreated MSCs were implanted in the MI model, then the cardiac function was analyzed via echocardiography. MSCs were labeled with 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI) dye for tracking, while the regeneration of infarcted myocardium was examined by histology and immunohistochemistry. RESULTS: MSCs were isolated and characterized. Cytotoxicity analysis showed that IWP-4 was non-cytotoxic at 5 µM concentration. Cardiac specific gene and protein expression analyses exhibited more remarkable results in fourteen days treated group that was eventually selected for in vivo transplantation. Cardiac function was restored in the IWP-4 treated group in comparison to the MI group. Immunohistochemical analysis confirmed the homing of pre-differentiated MSCs that were labeled with DiI cell labeling dye. Histological analysis confirmed the significant reduction in fibrotic area, and improved left ventricular wall thickness in IWP-4 treated MSC group. CONCLUSION: Treatment of MSCs with IWP-4 inhibits Wnt pathway and promotes cardiac differentiation. These pre-conditioned MSCs transplanted in vivo improved cardiac function by cell homing, survival, and differentiation at the infarcted region, increased left ventricular wall thickness, and reduced infarct size.

8.
Mol Biol Rep ; 50(9): 7371-7380, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37450078

RESUMO

BACKGROUND: Cardiovascular diseases remain a major cause of death globally. Cardiac cells once damaged, cannot resume the normal functioning of the heart. Bone marrow derived mesenchymal stem cells (BM-MSCs) have shown the potential to differentiate into cardiac cells. Epigenetic modifications determine cell identity during embryo development via regulation of tissue specific gene expression. The major epigenetic mechanisms that control cell fate and biological functions are DNA methylation and histone modifications. However, epigenetic modifiers alone are not sufficient to generate mature cardiac cells. Various small molecules such as ascorbic acid (AA) and salvianolic acid B (SA) are known for their cardiomyogenic potential. Therefore, this study is aimed to examine the synergistic effects of epigenetic modifiers, valproic acid (VPA) and 5-azacytidine (5-aza) with cardiomyogenic molecules, AA and SA in the cardiac differentiation of MSCs. METHODS AND RESULTS: BM-MSCs were isolated, propagated, characterized, and then treated with an optimized dose of VPA or 5-aza for 24 h. MSCs were maintained in a medium containing AA and SA for 21 days. All groups were assessed for the expression of cardiac genes and proteins through q-PCR and immunocytochemistry, respectively. Results show that epigenetic modifiers VPA or 5-aza in combination with AA and SA significantly upregulate the expression of cardiac genes MEF2C, Nkx2.5, cMHC, Tbx20, and GATA-4. In addition, VPA or 5-aza pretreatment along with AA and SA enhanced the expression of the cardiac proteins connexin-43, GATA-4, cTnI, and Nkx2.5. CONCLUSION: These findings suggest that epigenetic modifiers valproic acid and 5-azacytidine in combination with ascorbic acid and salvianolic acid B promote cardiac differentiation of MSCs. This pretreatment strategy can be exploited for designing future stem cell based therapeutic strategies for cardiovascular diseases.


Assuntos
Doenças Cardiovasculares , Células-Tronco Mesenquimais , Humanos , Ácido Valproico/farmacologia , Ácido Valproico/metabolismo , Ácido Ascórbico/farmacologia , Ácido Ascórbico/metabolismo , Doenças Cardiovasculares/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Azacitidina/farmacologia , Azacitidina/metabolismo , Miócitos Cardíacos/metabolismo , Células Cultivadas
9.
Mol Biol Rep ; 50(5): 4119-4131, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36877347

RESUMO

BACKGROUND: With advancing age of stem cells, dysregulation of various processes at the cellular level occurs, thereby decreasing their regeneration potential. One of the changes that occurs during the aging process is the accumulation of reactive oxygen species (ROS), which accelerates the processes of cellular senescence and cell death. The aim of this study is to evaluate two antioxidant compounds; Chromotrope 2B and Sulfasalazine, for their antioxidant effects on young and old rat bone marrow mesenchymal stem cells (MSCs). METHODS AND RESULTS: Oxidative stress was induced in MSCs by 5 µM dexamethasone for 96 h and the cells were treated with Chromotrope 2B or Sulfasalazine, 50 µM each. The effects of antioxidant treatment following oxidative stress induction was evaluated by transcriptional profiling of genes involved in the oxidative stress and telomere maintenance. Expression levels of Cat, Gpx7, Sod1, Dhcr24, Idh1, and Txnrd2 were found to be increased in young MSCs (yMSCs) as a result of oxidative stress, while Duox2, Parp1, and Tert1 expression were found to be decreased as compared to the control. In old MSCs (oMSCs), the expressions of Dhcr24, Txnrd2, and Parp1 increased, while that of Duox2, Gpx7, Idh1, and Sod1 decreased following oxidative stress. In both MSC groups, Chromotrope 2B prompted decrease in the ROS generation before and after the induction of oxidative stress. In oMSCs, ROS content was significantly reduced in the Sulfasalazine treated group. CONCLUSION: Our findings suggest that both Chromotrope 2B and Sulfasalazine possess the potential to reduce the ROS content in both age groups, though the latter was found to be more potent. These compounds can be used to precondition MSCs to enhance their regenerative potential for future cell-based therapeutics.


Assuntos
Antioxidantes , Células-Tronco Mesenquimais , Camundongos , Ratos , Animais , Antioxidantes/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Sulfassalazina/farmacologia , Sulfassalazina/metabolismo , Superóxido Dismutase-1/metabolismo , Medula Óssea/metabolismo , Oxidases Duais , Estresse Oxidativo , Células-Tronco Mesenquimais/metabolismo , Tiorredoxina Redutase 2/metabolismo
10.
J Pak Med Assoc ; 73(Suppl 1)(2): S3-S8, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36788384

RESUMO

Objectives: To determine the effect of the pre-treatment of mesenchymal stem cells (MSCs) with minocycline on the expression of antioxidant genes and cardiac repair post myocardial infarction (MI) in rats. METHODS: Rat bone marrow derived MSCs were used in the study. Cytotoxicity of minocycline in MSCs was determined using JC1 assay to identify a safe drug dose for further experiments. The MSCs were pre-treated with 1.0 µM minocycline for 24 hours and then treated with hydrogen peroxide (H2O2), after that mRNA was isolated and the expression levels of antioxidant genes including peroxiredoxin, glutathione peroxidase, and superoxide dismutase were determined. Finally, minocycline pre-treated MSCs were used to treat rats induced with MI by the ligation of left anterior descending coronary artery. The cardiac function was evaluated at two and four weeks post MI using echocardiography. RESULTS: At 1.0 µM concentration, minocycline was found to be safe for MSCs and used for subsequent experiments. Minocycline pre-treatment was found to up regulate several antioxidant genes in oxidatively stressed MSCs. Furthermore, minocycline pre-treated MSCs displayed greater improvement in cardiac left ventricular function at two and four-weeks post MI as compared to untreated rats. CONCLUSIONS: Pre-treatment of MSCs with minocycline enhances the expression of antioxidant genes and promotes their capability to repair cardiac function after MI.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio , Ratos , Animais , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Minociclina/farmacologia , Minociclina/metabolismo , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Modelos Animais de Doenças
11.
Cell Biochem Funct ; 41(2): 223-233, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36651266

RESUMO

Central nervous system anomalies give rise to neuropathological consequences with immense damage to the neuronal tissues. Cell based therapeutics have the potential to manage several neuropathologies whereby the differentiated cells are explored for neuronal regeneration. The current study analyzes the effect of a bioactive compound, alpha terpineol (AT) on the differentiation of rat bone marrow derived mesenchymal stem cells (BM-MSCs) toward neuronal lineage, and explores regulation of differentiation process through the study of Wnt pathway mediators. BM-MSCs were cultured and characterized based on their surface markers and tri-lineage differentiation. Safe dose of AT as optimized by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium bromide assay, was used for the treatment of MSCs. Treated cells were analyzed for the neuronal, astroglial and germ layer transition markers at the gene and protein levels, by quantitative polymerase chain reaction and immunocytochemistry, respectively. Temporal expression of Wnt pathway genes was assessed during the course of neuronal differentiation. AT treated group showed significant upregulation of neuron specific (NSE, MAP2, Tau, Nestin, and NefL) and astroglial (GFAP) genes with positive expression of late neuronal markers. Germ layer transition analysis showed the overexpression of ectodermal markers (NCAM, Nestin, and Pax6), whereas endodermal (AFP, MixL1, and Sox17), and mesodermal (Mesp1 and T Brachyury) markers were also found to be upregulated. Wnt signaling pathway was activated during the initial phase (30 min) of differentiation, which later was downregulated at 1, 3, and 5 h. AT efficiently induces neuronal differentiation of BM-MSCs by regulating Wnt signaling. Overexpression of both early and late neuronal markers indicate their neuro-progenitor state and thus can be utilized as a promising approach in cellular therapeutics to treat various neurodegenerative ailments. In addition, exploration of the molecular pathways may be helpful to understand the mechanism of cell-based neuronal regeneration.


Assuntos
Células-Tronco Mesenquimais , Via de Sinalização Wnt , Ratos , Animais , Nestina/metabolismo , Nestina/farmacologia , Neurônios/metabolismo , Diferenciação Celular , Células-Tronco Mesenquimais/metabolismo , Células da Medula Óssea , Células Cultivadas
12.
Mol Cell Biochem ; 478(8): 1759-1770, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-36566485

RESUMO

Myocardial infarction (MI) damages cardiomyocytes permanently and compromises cardiac function. Mesenchymal stem cells (MSCs) with the potential to differentiate into multiple lineages are considered as one of the best options for the treatment of MI. However, aging affects their regeneration capability. With age, reactive oxygen species (ROS) accumulate in cells ultimately causing cell death. To successfully utilize these stem cells in clinic, novel strategies to improve their functional capability should be explored. In this study, we aimed to enhance the cardiac regeneration potential of bone marrow MSCs derived from aging rats by treating them with antioxidants, rutin or quercetagetin in separate in vivo experiments. Oxidative stress was induced by treating MSCs of young and aging rats with different concentrations of H2O2 which resulted in an increase in the ROS level. MSCs were treated with rutin or quercetagetin at varying concentrations and exposed to H2O2. It was observed that both antioxidants significantly (P < 0.001) suppressed H2O2-induced intracellular ROS accumulation in a dose-dependent manner. An optimized concentration of 10 µM rutin or quercetagetin was used for the in vivo experiments. MI models were developed in aging rats by ligation of left anterior descending artery and treated MSCs were transplanted in the MI models. Echocardiography was performed after 2 and 4 weeks of cell transplantation to evaluate the functional status of the infarcted heart and histological analysis was performed after 4 weeks to assess cardiac regeneration. Significant improvement was observed in cardiac parameters including LVEF% (P < 0.001), LVFS% (P < 0.01 and P < 0.001), LVIDd (P < 0.01 and P < 0.001), LVIDs (P < 0.001), LVEDV (P < 0.001) and LVESV (P < 0.001) in the treated young as well as aging MSCs. It is concluded from these findings that rutin and quercetagetin treatment enhance the regeneration efficiency of young and aging MSCs in vivo. These antioxidants can be effectively utilized to improve cellular therapy for myocardial infarction by suppressing ROS production.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Infarto do Miocárdio , Ratos , Animais , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Medula Óssea/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Miocárdio/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/metabolismo , Células-Tronco Mesenquimais/metabolismo , Envelhecimento , Transplante de Células-Tronco Mesenquimais/métodos
13.
Altern Lab Anim ; 51(1): 12-29, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36484201

RESUMO

Stem cell-based therapy presents an attractive alternative to conventional therapies for degenerative diseases. Numerous studies have investigated the capability of human umbilical cord-derived mesenchymal stem cells (hUC-MSCs) to contribute to the regeneration of cardiomyocytes, and the results have encouraged further basic and clinical studies on the MSC-based treatment of cardiomyopathies. This study aimed to determine the potential of cardiomyogenic transcription factors in differentiating hUC-MSCs into cardiac-like cells in vitro. MSCs were isolated from umbilical cord tissue and were transduced with the transcription factor genes, GATA-4 and Nkx 2.5, via infection with lentiviruses, to promote differentiation into the cardiomyogenic lineage. Gene and protein expression were analysed with qPCR and immunocytochemical staining. After transduction, differentiated cardiac-like cells showed significant expression of cardiac genes and proteins, namely GATA-4, Nkx-2.5, cardiac troponin I (cTnI) and myosin heavy chain (MHC). The cardiomyogenic-induced group significantly overexpressed cardiac-specific genes (GATA-4, Nkx-2.5, cTnI, MHC, α-actinin and Wnt2). Expression of the calcium channel gene was also significantly increased, while the sodium channel gene was downregulated in the transduced hUC-MSCs, as compared to non-transduced cells. The results suggest that GATA-4 and Nkx-2.5 interact synergistically in the activation of downstream cardiac transcription factors, demonstrating the functional convergence of hUC-MSC differentiation into cardiac-like cells. These findings could potentially be utilised in the efficient production of cardiac-like cells from stem cells; these cardiac-like cells could then be used in various applications, such as for in vivo implantation in infarcted myocardium, and for drug screening in toxicity testing.


Assuntos
Células-Tronco Mesenquimais , Miocárdio , Humanos , Diferenciação Celular/fisiologia , Miocárdio/metabolismo , Fatores de Transcrição/metabolismo , Troponina I/metabolismo , Células-Tronco Mesenquimais/metabolismo , Cordão Umbilical/metabolismo
14.
Biol Futur ; 73(4): 495-502, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36512201

RESUMO

Human umbilical cord (hUC) derived mesenchymal stem cells (MSCs) can be progressively differentiated into multiple lineages including hepatic lineages, and thus provide an excellent in vitro model system for the study of hepatic differentiation. At present, hepatic differentiation protocols are based on the use of soluble chemicals in the culture medium and provide immature hepatic like cells. Histone deacetylase inhibitors (HDACi) and DNA methyltransferase inhibitors (DNMTi) are two important epigenetic modifiers that regulate stem cell differentiation. Therefore, this study aimed to investigate the role of HDACi, valproic acid (VPA) and DNMTi,5-azacytidine (5-aza) along with a hepatic inducer in the hepatic differentiation of hUC-MSCs. hUC-MSCs were characterized via immunocytochemistry and flow cytometry. The final concentrations of VPA and 5-aza were optimized via MTT cytotoxicity assay. All treated groups were assessed for the presence of hepatic genes and proteins through qPCR and immunocytochemistry, respectively. The results showed that the pretreatment of epigenetic modifiers not only increased the hepatic genes but also increased the expression of the hepatic proteins. VPA induces hepatic differentiation in hUC-MSCs with significant gene expression of hepatic markers i.e., FOXA2 and CK8. Moreover, VPA pretreatment enhanced the expression of hepatic proteins AFP and TAT. The pretreatment of 5-aza shows significant gene expression of hepatic marker LDL-R. However, 5-aza treatment failed to induce hepatic protein expression. The results of the current study highlighted the effectiveness of epigenetic modifiers in the hepatic differentiation of hUC-MSCs. These differentiated cells can be employed in cell-based therapeutics for hepatic diseases in future.


Assuntos
Células-Tronco Mesenquimais , Ácido Valproico , Humanos , Diferenciação Celular/genética , Ácido Valproico/farmacologia , Ácido Valproico/metabolismo , Azacitidina/metabolismo , Epigênese Genética , Células-Tronco Mesenquimais/metabolismo , Cordão Umbilical
15.
World J Stem Cells ; 14(8): 633-657, 2022 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-36157910

RESUMO

BACKGROUND: Cardiovascular diseases are the major cause of mortality worldwide. Regeneration of the damaged myocardium remains a challenge due to mechanical constraints and limited healing ability of the adult heart tissue. Cardiac tissue engineering using biomaterial scaffolds combined with stem cells and bioactive molecules could be a highly promising approach for cardiac repair. Use of biomaterials can provide suitable microenvironment to the cells and can solve cell engraftment problems associated with cell transplantation alone. Mesenchymal stem cells (MSCs) are potential candidates in cardiac tissue engineering because of their multilineage differentiation potential and ease of isolation. Use of DNA methyl transferase inhibitor, such as zebularine, in combination with three-dimensional (3D) scaffold can promote efficient MSC differentiation into cardiac lineage, as epigenetic modifications play a fundamental role in determining cell fate and lineage specific gene expression. AIM: To investigate the role of collagen scaffold and zebularine in the differentiation of rat bone marrow (BM)-MSCs and their subsequent in vivo effects. METHODS: MSCs were isolated from rat BM and characterized morphologically, immunophenotypically and by multilineage differentiation potential. MSCs were seeded in collagen scaffold and treated with 3 µmol/L zebularine in three different ways. Cytotoxicity analysis was done and cardiac differentiation was analyzed at the gene and protein levels. Treated and untreated MSC-seeded scaffolds were transplanted in the rat myocardial infarction (MI) model and cardiac function was assessed by echocardiography. Cell tracking was performed by DiI dye labeling, while regeneration and neovascularization were evaluated by histological and immunohistochemical analysis, res pectively. RESULTS: MSCs were successfully isolated and seeded in collagen scaffold. Cytotoxicity analysis revealed that zebularine was not cytotoxic in any of the treatment groups. Cardiac differentiation analysis showed more pronounced results in the type 3 treatment group which was subsequently chosen for the transplantation in the in vivo MI model. Significant improvement in cardiac function was observed in the zebularine treated MSC-seeded scaffold group as compared to the MI control. Histological analysis also showed reduction in fibrotic scar, improvement in left ventricular wall thickness and preservation of ventricular remodeling in the zebularine treated MSC-seeded scaffold group. Immunohistochemical analysis revealed significant expression of cardiac proteins in DiI labeled transplanted cells and a significant increase in the number of blood vessels in the zebularine treated MSC-seeded collagen scaffold transplanted group. CONCLUSION: Combination of 3D collagen scaffold and zebularine treatment enhances cardiac differentiation potential of MSCs, improves cell engraftment at the infarcted region, reduces infarct size and improves cardiac function.

16.
Tissue Cell ; 77: 101850, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35679684

RESUMO

Wnt/ß-catenin, a highly conserved signaling pathway, is involved in determining cell fate. During heart development, Wnt signaling controls specification, proliferation and differentiation of cardiac cells. This study is aimed to investigate the role of Wnt/ß-catenin signaling in cardiac lineage commitment of human umbilical cord mesenchymal stem cells (hUCMSCs) after treatment with demethylating agents, zebularine and 2'-deoxycytidine (2-DC). hUCMSCs were treated with 20 µM zebularine or 2-DC for 24 h and cultured for 14 days. Control and treated MSCs were analyzed for cardiac lineage commitment at gene and protein levels. Significant upregulation of early and late cardiac markers, GATA4, Nkx2.5, cardiac myosin heavy chain (cMHC), α-actinin, cardiac troponin T (cTnT) and cardiac troponin I (cTnI) was observed in treated MSCs as compared to the untreated control. We also analyzed gene expression of key Wnt/ß-catenin signaling molecules in cultures of treated and untreated hUCMSCs at 24 h, and days 3, 7 and 14. The pattern of mRNA gene expression showed that Wnt/ß-catenin signaling is regulated during cardiac lineage commitment of hUCMSCs in a time-dependent manner, with the pathway being activated early but inhibited later in cardiac development. Findings of this study can lead us to identify more specific and effective strategies for cardiac lineage commitment.


Assuntos
Células-Tronco Mesenquimais , beta Catenina , Diferenciação Celular , Citidina/análogos & derivados , Desoxicitidina/farmacologia , Humanos , Miócitos Cardíacos/metabolismo , Cordão Umbilical , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo
17.
Appl Biochem Biotechnol ; 194(8): 3721-3732, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35499693

RESUMO

Stem cell-based therapy is considered an attractive tool to overcome the burden of liver diseases. However, efficient hepatic differentiation is still a big challenge for the research community. In this study, we explored a novel method for differentiation of bone marrow-derived mesenchymal stem cells (MSCs) into hepatic-like cells using 3D culture conditions and histone deacetylase inhibitor, sodium butyrate (NaBu). MSCs were characterized by the presence of cell surface markers via immunocytochemistry, flow cytometry, and by their potential for osteogenic, adipogenic, and chondrogenic differentiation. MSCs were treated with 1mM NaBu in 2D and 3D environments for 21 days. The hepatic differentiation was confirmed by qPCR and immunostaining. According to qPCR data, the 3D culture of NaBu-treated MSCs has shown significant upregulation of hepatic gene, CK-18 (P < 0.01), and hepatic proteins, AFP (P < 0.01) and ALB (P < 0.01). In addition, immunocytochemistry analysis showed significant increase (P < 0.05) in the acetylation of histones (H3 and H4) in NaBu-pretreated cells. It can be concluded from the study that NaBu-treated MSCs in 3D culture conditions can induce hepatic differentiation without the use of additional cytokines and growth factors. The method shown in this study represents an improved protocol for hepatic differentiation and could contribute to improvement in future cell-based therapeutics.


Assuntos
Células-Tronco Mesenquimais , Ácido Butírico/metabolismo , Ácido Butírico/farmacologia , Diferenciação Celular , Colágeno/metabolismo , Osteogênese
18.
Mol Cell Biochem ; 477(12): 2735-2749, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35610401

RESUMO

The therapeutic use of bone marrow mesenchymal stem cells (BM-MSCs) requires a large number of cells (1-100 × 106 cells/kg of body weight). Extensive in vitro growth is limited due to the aging of cultured BM-MSCs which leads to abnormal morphology and senescence. Hypoxia increases BM-MSC proliferation, but the question of whether hypoxia preconditioning is safe for clinical application of BM-MSCs remains to be answered. Zinc is essential for cell proliferation and differentiation, especially for the regulation of DNA synthesis and mitosis. It is a structural constituent of numerous proteins on a molecular level, including transcription factors and enzymes of cellular signaling machinery. All the tissues, fluids, and organs of the human body contain zinc. More than 95% of zinc is intracellular, of which 44% is involved in the transcription of DNA. We investigated the effects of ZnCl2 on proliferation, morphology, migration, population doubling time (PDT), and gene expression of BM-MSCs under hypoxic (1% O2) and normoxic (21% O2) environments. BM-MSCs were preconditioned with optimized concentrations of ZnCl2 under normoxic and hypoxic environments and further examined for morphology by the phase-contrast inverted microscope, cell proliferation by MTT assay, PDT, cell migration ability, and gene expression analysis. Zinc significantly enhanced the proliferation of BM-MSCs, and it decreases PDT under hypoxic and normoxic environments as compared to control cells. Migration of BM-MSCs toward the site of injury increased and expression of HIF1-α significantly decreased under hypoxic conditions as compared to non-treated hypoxic cells and control. At late passages (P9), the morphology of normoxic BM-MSCs was transformed into large, wide, and flat cells, and they became polygonal and lost their communication with other cells. Conversely, zinc-preconditioned BM-MSCs retained their spindle-shaped, fibroblast-like morphology at P9. The expression of proliferative genes was found significantly upregulated, while downregulation of genes OCT4 and CCNA2 was observed in zinc-treated BM-MSCs under both normoxic and hypoxic conditions. ZnCl2 treatment can be used for extensive expansion of BM-MSCs in aged populations to obtain a large number of cells required for systemic administration to produce therapeutic efficacy.


Assuntos
Células da Medula Óssea , Células-Tronco Mesenquimais , Humanos , Idoso , Células da Medula Óssea/metabolismo , Zinco/farmacologia , Zinco/metabolismo , Medula Óssea , Hipóxia Celular , Células Cultivadas , Células-Tronco Mesenquimais/metabolismo , Proliferação de Células , Hipóxia/metabolismo
19.
Bioimpacts ; 12(2): 147-154, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35411294

RESUMO

Introduction: Neurodegenerative diseases are accompanied by loss of neuronal function and integrity. Stem cell therapy is utilized to regenerate neurons to repair the damaged area. Regeneration potential of stem cells can be enhanced by using chemicals with known bioactive properties. In the current study, two bioactive compounds, α-pinene (AP) and thymoquinone (TQ) were explored for their neuronal differentiation potential of rat bone marrow mesenchymal stem cells (MSCs). Methods: MSCs were isolated, cultured and characterized immunocytochemically for the presence of specific surface markers. Optimized concentrations of both compounds (20 µM AP and 12 µM TQ) as determined by MTT assay, were used to treat MSCs in separate and combined groups. All groups were assessed for the presence of neuronal, astroglial, and germ layer markers through qPCR. Neuronal and glial protein expression were analyzed by immunocytochemistry. Results: Both compounds alone and in combination induced differentiation in MSCs with significant gene expression of neuronal markers i.e. neuron specific enolase (NSE), nestin, microtubule-associated protein 2 (MAP2), neurofilament light chain (Nefl) and Tau, and astroglial marker i.e. glial fibrillary acidic protein (GFAP). AP treated group also showed significant upregulation of endodermal and mesodermal markers indicating transition of ectoderm towards the other two germ layers. Conclusion: This study concludes that AP and TQ potentially differentiate MSCs into neuronal and astroglial lineages. However, AP treated group followed germ layer transition. Expression of neuronal as well as glial markers indicate that the differentiated neurons are at the neuroprogenitor stage and can be potential candidates for cellular therapeutics against neurodegenerative disorders.

20.
Mol Cell Biochem ; 476(8): 3191-3205, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33864569

RESUMO

Intervertebral disc (IVD) degeneration is an asymptomatic pathophysiological condition and a strong causative factor of low back pain. There is no cure available except spinal fusion and pain management. Stem cell-based regenerative medicine is being considered as an alternative approach to treat disc diseases. The current study aimed to differentiate human umbilical cord-mesenchymal stem cells (hUC-MSCs) into chondrocyte-like cells and to elucidate their feasibility and efficacy in the degenerated IVD rat model. Chondrogenic induction medium was used to differentiate hUC-MSCs into chondroprogenitors. Rat tail IVD model was established with three consecutive coccygeal discs. qPCR was performed to quantify the molecular markers of pain and inflammation. Histological staining was performed to evaluate the degree of regeneration. Induced chondroprogenitors showed the expression of chondrogenic genes, SOX9, TGF-ß1, ACAN, BMP2, and GDF5. Immunocytochemical staining showed positive expression of chondrogenic proteins SOX9, TGF-ß1, TGF-ß2, and Collagen 2. In in vivo study, transplanted chondroprogenitors showed better survival, homing, and distribution in IVD as compared to normal MSCs. Expression of pain and inflammatory genes at day 5 of cell transplantation modulated immune response significantly. The transplanted labeled MSCs and induced chondroprogenitors differentiated into functional nucleus pulposus (NP) cells as evident from co-localization of red (DiI) and green fluorescence for SOX9, TGF-ß1, and TGF-ß2. Alcian blue and H & E staining showed standard histological features, indicating better preservation of the NP structure and cellularity than degenerated discs. hUC-MSCs-derived chondroprogenitors showed better regeneration potential as compared to normal MSCs. The pain and inflammation genes were downregulated in the treated group as compared to the degenerated IVD.


Assuntos
Condrogênese , Inflamação/prevenção & controle , Degeneração do Disco Intervertebral/terapia , Disco Intervertebral/citologia , Células-Tronco Mesenquimais/citologia , Dor/prevenção & controle , Regeneração , Animais , Diferenciação Celular , Humanos , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/patologia , Masculino , Transplante de Células-Tronco Mesenquimais/métodos , Dor/etiologia , Dor/metabolismo , Dor/patologia , Ratos , Ratos Wistar , Transdução de Sinais , Cordão Umbilical/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA