Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 13(1): 12213, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37500705

ABSTRACT

Current management of HCV infection is based on Direct-Acting Antiviral Drugs (DAAs). However, resistance-associated mutations, especially in the NS3 and NS5B regions are gradually decreasing the efficacy of DAAs. The aim of the current study was to identify such mutations in the NS3, and NS5B genes in DAAs treatment-naïve Pakistani chronic HCV 3a patients. Peripheral blood samples were collected from 233 chronic HCV 3a patients at different tertiary care hospitals in Karachi, Pakistan, between August 2020 to September 2021. PCR-amplified target regions of the NS3/NS5B gene were subjected to Sanger sequencing to identify resistance-associated mutations. Phylogenetic analysis of the identified amino acid sequences was performed using HCV3a sequences of the global population in the virus pathogen resource (VIPR) database. Sequence analysis identified five amino acid mutations, Leu36Pro, Gln41His, Gln80Lys/Arg, Ala156Tyr, and Gln168Arg in the NS3 region, and two mutations Leu159Phe and Cys316Arg in the NS5B region. Phylogenetic analysis revealed a high genetic diversity in the studied isolates. Overall, the prevalence of resistance-associated substitutions was almost similar to other geographic regions worldwide. This data could be helpful in selecting the most effective treatment regimen for HCV chronically infected people in Pakistan.


Subject(s)
Antiviral Agents , Hepatitis C, Chronic , Humans , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Pakistan/epidemiology , Phylogeny , Hepacivirus , Genotype , Drug Resistance, Viral/genetics , Viral Nonstructural Proteins/genetics , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/epidemiology , Mutation
2.
Mol Biol Rep ; 50(5): 4119-4131, 2023 May.
Article in English | MEDLINE | ID: mdl-36877347

ABSTRACT

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.


Subject(s)
Antioxidants , Mesenchymal Stem Cells , Mice , Rats , Animals , Antioxidants/metabolism , Reactive Oxygen Species/metabolism , Sulfasalazine/pharmacology , Sulfasalazine/metabolism , Superoxide Dismutase-1/metabolism , Bone Marrow/metabolism , Dual Oxidases , Oxidative Stress , Mesenchymal Stem Cells/metabolism , Thioredoxin Reductase 2/metabolism
3.
Mol Cell Biochem ; 478(8): 1759-1770, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36566485

ABSTRACT

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.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Myocardial Infarction , Rats , Animals , Antioxidants/pharmacology , Antioxidants/metabolism , Bone Marrow/metabolism , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/pharmacology , Myocardium/metabolism , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Mesenchymal Stem Cells/metabolism , Aging , Mesenchymal Stem Cell Transplantation/methods
4.
Cardiovasc Ther ; 35(2)2017 Apr.
Article in English | MEDLINE | ID: mdl-28039940

ABSTRACT

INTRODUCTION: Rap1, a member of Ras superfamily of small GTP-binding proteins, is involved in cardiovascular biology in numerous ways. It is an evolutionary conserved regulator of adhesion, polarity, differentiation and growth. AIMS: Our aim was to analyze Rap1-activated rat bone marrow mesenchymal stem cells (MSCs) for their potential role in adhesion and cardiac differentiation. METHODS: Myocardial infarction (MI) was produced in Sprague Dawley (SD) rats through occlusion of the left anterior descending coronary artery. MSCs were treated with 8-pCPT-2'-O-Me-cAMP (CPT) to activate Rap1. Normal (untreated) and CPT-treated MSCs were transplanted through intramyocardial injection in respective groups. Cardiac function was assessed by echocardiography at 2 and 4 weeks after cell transplantation. Histological analysis was performed to observe changes at tissue level. RESULTS: Homing of CPT-treated MSCs was significantly (***P<.001) higher as compared to normal MSCs in the infarcted hearts. This may be due to increase in the gene expression of some of the cell adhesion molecules as evident by qRT-PCR analysis. Significant (***P<.001) improvement in the restoration of heart function in terms of left ventricular diastolic and systolic internal diameters (LVIDd, LVIDs), % ejection fraction, % fraction shortening and end-systolic and end-diastolic volumes were observed in CPT-treated MSCs as compared to the MI model. Histological analyses showed significant (***P<.001) reduction in scar formation in the CPT-treated group. Differentiation of treated MSCs into functional cardiomyocytes was evident through immunohistochemical staining. LV wall thickness was also preserved significantly (***P<.001). Blood vessel formation was more pronounced in CPT-treated group although both cell therapy groups showed significant increase as compared to MI model. CONCLUSION: Our findings showed that pharmacological activation of Epac-Rap1 improves cardiac function through better survival, adhesion and differentiation of transplanted cells. Transplantation of these MSCs in the infarct area restored functional myocardium.


Subject(s)
Cyclic AMP/analogs & derivatives , Enzyme Activators/pharmacology , Guanine Nucleotide Exchange Factors/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Myocardial Infarction/surgery , Myocardium/enzymology , Regeneration , rap1 GTP-Binding Proteins/metabolism , Animals , Cell Adhesion , Cell Differentiation , Cells, Cultured , Cyclic AMP/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Echocardiography , Enzyme Activation , Genotype , Male , Mesenchymal Stem Cells/enzymology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Phenotype , Rats, Sprague-Dawley , Recovery of Function , Time Factors , Ventricular Function, Left
5.
Life Sci ; 164: 52-59, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27593573

ABSTRACT

AIM: The study was carried out to evaluate the role of preconditioning strategies on the trans-differentiation of mature fibroblasts (NIH3T3 cells) into insulin producing ß-cells. METHODS: The NIH3T3 cells were treated with dexamethasone (5µM) and pancreatic extract (0.05 and 0.4mg/mL) separately or in combination. The treated cells were analyzed for the morphological changes, and expression of pancreatic genes and proteins by phase contrast microscopy, RT-PCR and flow cytometry/immunocytochemistry, respectively. RESULTS: Treatment of mature fibroblasts with different combinations of dexamethasone and pancreatic extract in the form of conditioned media resulted in comparable morphological changes and expression of certain pancreatic genes and proteins; however, their expression varied with each treatment. Most prominent effect was observed in case of combined treatment which resulted in significant increase (p<0.001) in gene expression levels of insulin, MafA, and Ngn3. Variable pattern was observed in insulin, MafA, Ngn3 and Sca1 expressions at the protein level. CONCLUSION: It is concluded from this study that preconditioning of NIH3T3 cells with conditioned media containing different combinations of dexamethasone and pancreatic extract can induce trans-differentiation of these cells into pancreatic ß-like cells. The conditioned media however, need to be optimized. The study may offer the possibility of improved regeneration of mature cell type that could serve as a future therapeutic option for diabetes.


Subject(s)
Cell Differentiation/drug effects , Culture Media, Conditioned/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Insulin-Secreting Cells/cytology , Animals , Dexamethasone/pharmacology , Humans , Insulin-Secreting Cells/drug effects , Mice , NIH 3T3 Cells , Pancreas/cytology , Pancreatic Extracts/genetics , Pancreatic Extracts/pharmacology , Polymerase Chain Reaction
6.
Cell Reprogram ; 18(5): 344-355, 2016 10.
Article in English | MEDLINE | ID: mdl-27500307

ABSTRACT

Insulin replacement is the current therapeutic option for type-1 diabetes. However, exogenous insulin cannot precisely represent the normal pattern of insulin secretion. Another therapeutic strategy is transplantation of pancreatic islets, but this is limited by immune rejection, intrinsic complications, and lack of donor availability. Stem cell therapy that results in the regeneration of insulin-producing cells represents an attractive choice. However, with advancing age, stem cells also undergo senescence, which leads to changes in the function of various cellular processes that result in a decrease in the regeneration potential of these aging stem cells. In this study, the effect of young and aging mesenchymal stem cells (MSCs) on the regeneration of pancreatic beta cells in streptozotocin (STZ)-induced type-1 diabetic mice was observed after hypoxic preconditioning. Hypoxia was chemically induced by 2, 4-dinitrophenol (DNP). Plasma insulin and glucose levels were measured at various time intervals, and pancreatic sections were analyzed histochemically. The effect of DNP was also analyzed on apoptosis of MSCs by flow cytometry and on gene expression of certain growth factors by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). We observed that hypoxic preconditioning caused changes in the gene expression levels of growth factors in both young and aging MSCs. Young MSCs showed significant regeneration potential compared with the aging cells in vivo. However, hypoxic preconditioning was able to improve the regeneration potential of aging MSCs. It is concluded from the present study that the regeneration potential of aging MSCs into pancreatic ß-cells can be enhanced by hypoxic preconditioning, which causes changes in the gene expression of certain growth factors.


Subject(s)
Bone Marrow Cells/cytology , Cellular Senescence , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/therapy , Hypoxia , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Gene Expression Profiling , Gene Expression Regulation , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Regeneration
7.
Mol Biol Rep ; 41(5): 3099-112, 2014 May.
Article in English | MEDLINE | ID: mdl-24469729

ABSTRACT

Mesenchymal stem cells (MSCs) show accelerated regeneration potential when these cells experience hypoxic stress. This "preconditioning" has shown promising results with respect to cardio-protection as it stimulates endogenous mechanisms resulting in multiple cellular responses. The current study was carried out to analyze the effect of hypoxia on the expression of certain growth factors in rat MSCs and cardiomyocytes (CMs). Both cell types were cultured and assessed separately for their responsiveness to hypoxia by an optimized dose of 2,4,-dinitrophenol (DNP). These cells were allowed to propagate under normal condition for either 2 or 24 h and then analyzed for the expression of growth factors by RT-PCR. Variable patterns of expression were observed which indicate that their expression depends on the time of re-oxygenation and extent of hypoxia. To see whether the growth factors released during hypoxia affect the fusion of MSCs with CMs, we performed co-culture studies in normal and conditioned medium. The conditioned medium is defined as the medium in which CMs were grown for re-oxygenation till the specified time period of either 2 or 24 h after hypoxia induction. The results showed that the fusion efficiency of cells was increased when the conditioned medium was used as compared to that in the normal medium. This may be due to the presence of certain growth factors released by the cells under hypoxic condition that promote cell survival and enhance their fusion or regenerating ability. This study would serve as another attempt in designing a therapeutic strategy in which conditioned MSCs can be used for ischemic diseases and provide more specific therapy for cardiac regeneration.


Subject(s)
Culture Media, Conditioned/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Animals , Antigens, Surface/metabolism , Cell Fusion , Cell Hypoxia , Coculture Techniques , Gene Expression , Immunohistochemistry , Immunophenotyping , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Rats
8.
Cardiovasc Ther ; 31(4): 201-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22954287

ABSTRACT

BACKGROUND: Mesenchymal stem cells (MSCs) have immense self-renewal capability. They can be differentiated into many cell types and therefore hold great potential in the field of regenerative medicine. MSCs can be converted into beating cardiomyocytes by treating them with DNA-demethylating agents. Some of these compounds are nucleoside analogs that are widely used for studying the role of DNA methylation in biological processes as well as for the clinical treatment of leukemia and other carcinomas. AIMS: To achieve a better therapeutic option for cardiovascular regeneration, this study was carried out using MSCs treated with two synthetic compounds, zebularine and 5-azacytidine. It can be expected that treated MSCs prior to transplantation may increase the likelihood of successful regeneration of damaged myocardium. METHODS: The optimized concentrations of these compounds were added separately into the culture medium and the treated cells were analyzed for the expression of cardiac-specific genes by RT-PCR and cardiac-specific proteins by immunocytochemistry and flow cytometry. Treated MSCs were cocultured with cardiomyocytes to see the fusion capability of these cells. RESULTS: mRNA and protein expressions of GATA4, Nkx2.5, and cardiac troponin T were observed in the treated MSCs. Coculture studies of MSCs and cardiomyocytes have shown improved fusion with zebularine-treated MSCs as compared to untreated and 5-azacytidine-treated MSCs. CONCLUSION: The study is expected to put forth another valuable aspect of certain compounds, that is, induction of transdifferentiation of MSCs into cardiomyocytes. This would serve as a tool for modified cellular therapy and may increase the probability of better myocardial regeneration.


Subject(s)
Azacitidine/pharmacology , Bone Marrow Cells/drug effects , Cell Transdifferentiation/drug effects , Cytidine/analogs & derivatives , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Mesenchymal Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Animals , Bone Marrow Cells/metabolism , Cell Fusion , Cells, Cultured , Coculture Techniques , Cytidine/pharmacology , DNA Modification Methylases/metabolism , Dose-Response Relationship, Drug , Female , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , Gene Expression Regulation , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Male , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factors/genetics , Transcription Factors/metabolism , Troponin T/genetics , Troponin T/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...