Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Brain Res Bull ; 131: 117-122, 2017 May.
Article in English | MEDLINE | ID: mdl-28373151

ABSTRACT

OBJECTIVE: Neuropathies are a nerve disorders that caused by diabetes. Neuropathy affects over 50% of diabetic patients. High blood glucose and their toxic byproducts are the main causes for nerve dysfunction. In the present study, we examined the neroprotective effects of cerium oxide (CeO2) nanoparticles in diabetic rats. METHOD: Rats divided into four groups: control group, diabetic group, the diabetic group treated with CeO2 nanoparticle at a dose of 65mg/kg and diabetic group received CeO2 nanoparticle at a dose of 85mg/kg. Diabetes was induced by single intraperitoneal injection of 65mg/kg streptozotocin (STZ). 8 weeks after the induction of diabetes, body weight and pain sensitivity in all groups were measured. The blood sample was collected for biochemical analysis. The dorsal root ganglion (DRG) neurons were isolated for histopathological stain and morphometric parameters studies. RESULTS: Reduction of body weight, total thiol molecules (TTM), total antioxidant power (TAP) and ADP/ATP ratio in diabetic rat was reversed by CeO2 nanoparticles administration. We showed that lipid peroxidation (LPO) and nociception latency were significantly increased in STZ-treated rats and decreased after CeO2 nanoparticles administration. DRG neurons showed obvious vacuole and various changes in diameter, area and the count of A and B cells in STZ-diabetic rat. CeO2 nanoparticles improved the histopathology and morphological abnormalities of DRG neurons. CONCLUSION: Our study concluded the CeO2 nanoparticles have a protective effect against the development of DN.


Subject(s)
Cerium/pharmacology , Diabetic Neuropathies/drug therapy , Animals , Antioxidants/pharmacology , Blood Glucose/drug effects , Body Weight/drug effects , Cerium/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Ganglia, Spinal/drug effects , Lipid Peroxidation/drug effects , Male , Nanoparticles/therapeutic use , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Rats , Rats, Wistar , Sulfhydryl Compounds
2.
J Cell Biochem ; 118(7): 1879-1888, 2017 07.
Article in English | MEDLINE | ID: mdl-28169456

ABSTRACT

The protective effects and mechanisms of DADS on IL-1ß-mediated oxidative stress and mitochondrial apoptosis were investigated in C28I2 human chondrocytes. The effect of various concentrations of DADS (1, 5 10, 25, 50, and 100 µM) on C28I2 cell viability was evaluated in different times (2, 4, 8, 16, and 24 h) to obtain the non-cytotoxic concentrations of drug by MTT-assay. The protective effect of non-toxic concentrations of DADS on experimentally induced oxidative stress and apoptosis by IL-1ß in C28I2 was evaluated. The effects of DADS on IL-1ß-induced intracellular ROS production and lipid peroxidation were detected and the proteins expression of Nrf2, Bax, Bcl-2, caspase-3, total and phosphorylated JNK, and P38 MAPKs were analyzed by Western blotting. The mRNA expression of detoxifying phase II/antioxidant enzymes including heme oxygenase-1, NAD(P)H quinine oxidoreductase, glutathione S-transferase-P1, catalase, superoxide dismutase-1, glutathione peroxidase-1, -3, -4 were evaluated by reverse transcription-polymerase chain reaction. DADS in 1, 5, 10, and 25 µM concentrations had no cytotoxic effect after 24 h. Pretreatment with DADS remarkably increased Nrf2 nuclear translocation as well as the genes expression of detoxifying phase II/antioxidant enzymes and reduced IL-1ß-induced elevation of ROS, lipid peroxidation, Bax/Bcl-2 ratio, caspase-3 activation, and JNK and P38 phosphorylation. DADS could considerably reduce IL-1ß-induced oxidative stress and consequent mitochondrial apoptosis, as the major mechanisms of chondrocyte cell death in an experimental model of osteoarthritis. It may be considered as natural product in protecting OA-induced cartilage damage in clinical setting. J. Cell. Biochem. 118: 1879-1888, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Allyl Compounds/pharmacology , Chondrocytes/drug effects , Chondrocytes/metabolism , Disulfides/pharmacology , Interleukin-1beta/pharmacology , Oxidative Stress/drug effects , Antioxidants/metabolism , Apoptosis/drug effects , Catalase/metabolism , Cell Line , Cell Survival/drug effects , Glutathione Peroxidase/metabolism , Humans , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/drug effects , Superoxide Dismutase-1/metabolism , Glutathione Peroxidase GPX1
3.
Anticancer Drugs ; 28(1): 51-58, 2017 01.
Article in English | MEDLINE | ID: mdl-27548348

ABSTRACT

Restoration of p53 function triggers cell death and eliminates tumors in vivo. Identification of p53-reactivating small molecules such as PRIMA-1 holds promise for effective new anticancer therapies. Here, we investigated the effects of small molecule PRIMA-1 on cell viability and expression of p53-regulated genes and proteins in the acute promyelocytic leukemia-derived NB4 cell line. Our results showed that PRIMA-1 had antileukemic properties in acute promyelocytic leukemia-derived NB4 cells. PRIMA-1-triggered apoptosis in a dose-dependent and time-dependent manner as indicated by the MTT assay and annexin-V staining. Apoptosis induction by PRIMA-1 was associated with caspase-9, caspase-7 activation and PARP cleavage. p21 protein expression was increased after PRIMA-1 treatment and real-time PCR analysis of proapoptotic p53 target genes indicated upregulation of Bax and Noxa. Western blot analysis showed that IκBα phosphorylation and its degradation were inhibited by PRIMA-1. Moreover, protein expression of nuclear factor-κB-regulated antiapoptotic (Bcl-2 and XIAP) and proliferative (c-Myc) gene products was decreased. Importantly, PRIMA-1 did not show any significant apoptotic effect in normal human peripheral blood mononuclear cells. These in-vitro studies imply that p53 reactivation by small compounds may become a novel anticancer therapy in acute promyelocytic leukemia.


Subject(s)
Aza Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Leukemia, Promyelocytic, Acute/drug therapy , NF-kappa B/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Humans , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Telomerase/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics
4.
Neurol Res ; 37(7): 624-32, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25786672

ABSTRACT

OBJECTIVE: Oxidative stress has been recognized as the major factor for the development of diabetes and its complications. Cerium oxide and Yttrium oxide nanoparticles are known as free radicals scavengers. The aim of this study was to investigate the protective effect of CeO2 and Y2O3 on oxidative stress induced by high glucose in undifferentiated rat pheochromocytoma (PC12) cells. METHODS: In this study, undifferentiated PC12 cells were exposed to high glucose (25 mg/ml, 24 hours) and the protective effects of CeO2 and Y2O3 nanoparticles were evaluated. The viability of undifferentiated PC12 cells was determined by MTT assay. The levels of reactive oxygen species (ROS) were measured using 2,7-dichlorodihydrofluorescein diacetate (DCF). The expression levels of pro-apoptotic Bax, anti-apoptotic Bcl-2 and caspase3 proteins were also detected by western blotting. Total antioxidant power (TAP), total thiol molecules (TTM) and lipid peroxidation (LPO) were also evaluated. RESULTS: CeO2 and Y2O3 increased survival of undifferentiated PC12 cells exposed to high glucose-induced oxidative stress. CeO2 and Y2O3 pre-treatment decreased ROS production, LPO, Bax and caspase-3 proteins expression. Both nanoparticles have also increased the TTM and Bcl-2 protein expression. DISCUSSION: These findings suggest that CeO2 and Y2O3 protect the undifferentiated PC12 cells against the oxidative stress and apoptosis induced by high glucose.


Subject(s)
Cerium/pharmacology , Glucose/toxicity , Metal Nanoparticles , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Yttrium/pharmacology , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Cell Survival/physiology , Free Radical Scavengers/pharmacology , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Oxidative Stress/physiology , PC12 Cells , Rats , Reactive Oxygen Species/metabolism , Sulfhydryl Compounds/metabolism , bcl-2-Associated X Protein/metabolism
5.
J Diabetes Metab Disord ; 13(1): 76, 2014.
Article in English | MEDLINE | ID: mdl-25688339

ABSTRACT

BACKGROUND: Many studies suggested mesenchymal stem cells (MSCs) transplantation as a new approach to control hyperglycemia in type 1 diabetes mellitus through differentiation mechanism. In contrary others believed that therapeutic properties of MSCs is depends on paracrine mechanisms even if they were not engrafted. This study aimed to compare these two approaches in control of hyperglycemia in STZ-induced diabetic rats. METHODS: Animals were divided into five groups: normal; diabetic control; diabetic received MSCs; diabetic received supernatant of MSCs; diabetic received co-administration of MSCs with supernatant. Blood glucose, insulin levels and body weight of animals were monitored during experiment. Immunohistochemical and immunofluorescence analysis were performed to monitor functionality and migration of labeled-MSCs to pancreas. RESULTS: First administration of MSCs within the first 3 weeks could not reduce blood glucose, but second administration significantly reduced blood glucose after week four compared to diabetic controls. Daily injection of supernatant could not reduce blood glucose as efficient as MSCs. Interestingly; Co-administration of MSCs with supernatant significantly reduced blood glucose more than other treated groups. Insulin levels and body weight were significantly increased in MSCs + supernatant-treated animals compared to other groups. Immunohistological analysis showed an increase in number and size of islets per section respectively in supernatant, MSCs and MSCs + supernatant-treated groups. CONCLUSION: Present study exhibited that repeated-injection of MSCs reduced blood glucose and increased serum insulin levels in recipient rats. Injection of supernatant could not reverse hyperglycemia as efficient as MSCs. Interestingly; co-administration of MSCs with supernatant could reverse hyperglycemia more than either group alone.

6.
Neurosci Lett ; 558: 31-6, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24211690

ABSTRACT

Telmisartan is an angiotensin II type 1 receptor blocker and partial agonist of peroxisome proliferator-activated receptor gamma (PPAR-γ). Here, we investigated the protective capacity of telmisartan against high glucose (HG)-elicited oxidative damage in PC12 cells. The activity of lactate dehydrogenase (LDH), NADPH oxidase (NOX), superoxide dismutase (SOD), catalase (CAT) as well as the levels of malondialdehyde (MDA), glutathione (GSH), intracellular reactive oxygen species (ROS), cell viability and DNA fragmentation were measured in HG-treated PC12 cells with and without telmisartan co-treatment. Moreover, the direct antioxidant effect of telmisartan was determined by 2,2-azinobis-(3-ethylbenzothiazoline-6-sulphonic acid) (ABTS) assay and protein expression of Bax, Bcl-2, cleaved caspase-3 and NOX subunit p47phox by western blotting. Telmisartan exhibited antioxidant activity in the ABTS assay with the IC50 value of 37.5 µM. Pretreatment of PC12 cells with telmisartan, prior to HG exposure, was associated with a marked diminution in cleaved caspase-3 expression, DNA fragmentation, Bax/Bcl-2 ratio, intracellular ROS and MDA levels. Additionally, the cell viability, GSH level, SOD and CAT activity were notably elevated by telmisartan, whereas the activity and the protein expression of NADPH oxidase subunit p47phox were attenuated. Interestingly, co-treatment with GW9662, a PPAR-γ antagonist, partially inhibited the beneficial effects of telmisartan. These findings suggest that telmisartan has protective effects on HG-induced neurotoxicity in PC12 cells, which may be related to its antioxidant action and inhibition of NADPH oxidase. Furthermore, the results show that PPAR-γ activation is involved in the neuroprotective effects of telmisartan.


Subject(s)
Angiotensin II Type 2 Receptor Blockers/pharmacology , Antioxidants/pharmacology , Benzimidazoles/pharmacology , Benzoates/pharmacology , Glucose/metabolism , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Anilides/pharmacology , Animals , Apoptosis/drug effects , Catalase/metabolism , Cell Survival/drug effects , Glutathione/metabolism , Malondialdehyde/metabolism , NADP/metabolism , PC12 Cells , PPAR gamma/antagonists & inhibitors , Rats , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Telmisartan
7.
J. physiol. biochem ; 69(3): 451-458, sept. 2013.
Article in English | IBECS | ID: ibc-121664

ABSTRACT

Stem cells with the ability to differentiate into insulin-producing cells (IPCs) are becoming the most promising therapy for diabetes mellitus and reduce the major limitations of availability and allogeneic rejection of beta cell transplantations. Mesenchymal stem cells (MSCs) are pluripotent stromal cells with the ability to proliferate and differentiate into a variety of cell types including endocrine cells of the pancreas. This study sought to inspect the in vitro differentiation of human adipose-derived tissue stem cells into IPCs which could provide an abundant source of cells for the purpose of diabetic cell therapy in addition to avoid immunological rejection. Adipose-derived MSCs were obtained from liposuction aspirates and induced to differentiate into insulin-secreting cells under a three-stage protocol based on a combination of low-glucose DMEM medium, â-mercaptoethanol, and nicotinamide for pre-induction and high-glucose DMEM, β-mercaptoethanol, nicotinamide, and exendin-4 for induction stages of differentiation. Differentiation was evaluated by the analysis of morphology, dithizone staining, RT-PCR, and immunocytochemistry. Morphological changes including typical islet-like cell clusters were observed by phase-contrast microscope at the end of differentiation protocol. Based on dithizone staining, differentiated cells were positive and undifferentiated cells were not stained. Furthermore, RT-PCR results confirmed the expression of insulin, PDX1, Ngn3, PAX4, and GLUT2 in differentiated cells. Moreover, insulin production by the IPCs was confirmed by immunocytochemistry analysis. It is concluded that adipose-derived MSCs could differentiate into insulin-producing cells in vitro (AU)


Subject(s)
Humans , Mesenchymal Stem Cells/physiology , Adiposity/physiology , Insulin-Secreting Cells/physiology , Cell- and Tissue-Based Therapy/trends
8.
Expert Opin Biol Ther ; 13(7): 959-72, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23536977

ABSTRACT

OBJECTIVE: Today, cell therapy is considered a promising alternative in treatment of several diseases such as type 1 diabetes. Loss of transplanted stem cell and more importantly scarcity in the number of cells reaching to target tissue is a major obstacle in cell therapy. There is evidences showing that deferoxamine (DFO), an iron chelator, increases the mobilization and homing of progenitor cells through increasing the stability of hypoxia-inducible factor 1α (HIF-1α) protein. In this study, the effect of DFO on some factors involved in homing of bone marrow-derived mesenchymal stem cell was investigated, and the other objectives of this research were to determine whether DFO is able to increase migration and subsequent homing of mesenchymal stem cell (MSCs) both in vitro and in vivo in streptozotocin-diabetic rats. RESEARCH DESIGN AND METHODS: MSCs were treated by DFO in minimal essential medium α (αMEM) for 24 h. The expression and localization of HIF-1α were evaluated by western blotting and immunocytochemistry. The expression of C-X-C chemokine receptor type 4 (CXCR-4) and chemokine receptor 2 (CCR2) were assessed by western blotting and RT-PCR. The activity of matrix metalloproteinases (MMP) -2 and -9 were measured by gelatin zymography. Finally, in vitro migration of MSCs toward different concentrations of stromal cell-derived factor and monocyte chemotactic protein-1 were also evaluated. To demonstrate the homing of MSCs in vivo, DFO-treated chloromethyl-benzamidodialkylcarbocyanine-labeled MSCs were injected into the tail vein of rats, and the number of stained MSCs reaching to the pancreas were determined after 24 h. RESULTS: In DFO-treated MSCs, expression of HIF-1α (p < 0.001), CXCR4 (p < 0.001), CCR2 (p < 0.001), and the activity of MMP-2 (p < 0.01) and MMP-9 (p < 0.05) were significantly increased compared to control groups. Elevation of HIF-1α, upregulation of CXCR4/CCR2 and higher activity of MMP-2/MMP-9 in DFO-treated MSCs were reversed by 2-methoxyestradiol (2-ME; 5 µmol), a HIF-1α inhibitor. The in vitro migrations as well as in vivo homing of DFO-treated MSCs were also significantly higher than control groups (p < 0.05). CONCLUSIONS: Preconditioning of MSCs by DFO prior to transplantation could increase homing of MSCs through affecting some chemokine receptors as well as proteases involved and eventually improving the efficacy of cell therapy.


Subject(s)
Cell Movement , Cell Proliferation , Deferoxamine/pharmacology , Diabetes Mellitus, Experimental/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/drug effects , Siderophores/pharmacology , Animals , Apoptosis , Blotting, Western , Cell Adhesion , Cell Differentiation , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Flow Cytometry , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transplantation Conditioning
9.
Gynecol Endocrinol ; 29(5): 470-3, 2013 May.
Article in English | MEDLINE | ID: mdl-23461768

ABSTRACT

OBJECTIVE: It is estimated that 5-7% of women of reproductive age have polycystic ovary syndrome (PCOS). Chronic anovulation, hyperandrogenism, and insulin resistance (IR) are the main characters of this complex syndrome. IR, diabetes and obesity are all strongly correlated with PCOS; moreover, the possibility of direct androgen mediated renin-angiotensin system (RAS) stimulation in women with PCOS is also reported. The aim of the present study was to investigate the correlation between RAS, IR and PCOS. STUDY DESIGN: Thirty one women with PCOS, diagnosed by the Rotterdam criteria, were compared with thirty six control subjects. Both case and control groups were evaluated regarding to their basal hormonal profile, fasting blood sugar, IR, angiotensin converting enzyme (ACE) activity, plasma renin activity (PRA) and angiotensin II (AngІІ) levels. RESULTS: Compared to controls both ACE activity (p = 0.04) and AngΙΙ levels (p = 0.01) were significantly higher in case group. No significant differences between patients and controls were found in PRA. The results demonstrated that IR (p = 0.02) and fasting insulin (p = 0.004) were higher in case group, there was also a positive correlation between ACE activity and IR in case group (p = 0.02, r = 0.2). CONCLUSION: These results may suggest that there is an important correlation between ACE activity and IR in patients with PCOS.


Subject(s)
Angiotensin II/blood , Insulin Resistance , Peptidyl-Dipeptidase A/blood , Polycystic Ovary Syndrome/blood , Renin-Angiotensin System , Renin/blood , Adult , Case-Control Studies , Cross-Sectional Studies , Female , Humans , Up-Regulation/physiology , Young Adult
10.
J Physiol Biochem ; 69(3): 451-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23271274

ABSTRACT

Stem cells with the ability to differentiate into insulin-producing cells (IPCs) are becoming the most promising therapy for diabetes mellitus and reduce the major limitations of availability and allogeneic rejection of beta cell transplantations. Mesenchymal stem cells (MSCs) are pluripotent stromal cells with the ability to proliferate and differentiate into a variety of cell types including endocrine cells of the pancreas. This study sought to inspect the in vitro differentiation of human adipose-derived tissue stem cells into IPCs which could provide an abundant source of cells for the purpose of diabetic cell therapy in addition to avoid immunological rejection. Adipose-derived MSCs were obtained from liposuction aspirates and induced to differentiate into insulin-secreting cells under a three-stage protocol based on a combination of low-glucose DMEM medium, ß-mercaptoethanol, and nicotinamide for pre-induction and high-glucose DMEM, ß-mercaptoethanol, nicotinamide, and exendin-4 for induction stages of differentiation. Differentiation was evaluated by the analysis of morphology, dithizone staining, RT-PCR, and immunocytochemistry. Morphological changes including typical islet-like cell clusters were observed by phase-contrast microscope at the end of differentiation protocol. Based on dithizone staining, differentiated cells were positive and undifferentiated cells were not stained. Furthermore, RT-PCR results confirmed the expression of insulin, PDX1, Ngn3, PAX4, and GLUT2 in differentiated cells. Moreover, insulin production by the IPCs was confirmed by immunocytochemistry analysis. It is concluded that adipose-derived MSCs could differentiate into insulin-producing cells in vitro.


Subject(s)
Adipose Tissue/cytology , Insulin-Secreting Cells/cytology , Insulin/biosynthesis , Mesenchymal Stem Cells/cytology , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adult , Biomarkers/metabolism , Cell Differentiation , Cells, Cultured , Culture Media , Exenatide , Glucose/metabolism , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Mercaptoethanol/chemistry , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microscopy, Phase-Contrast , Niacinamide/metabolism , Peptides/metabolism , Peptides/pharmacology , Venoms/metabolism , Venoms/pharmacology
11.
Cytotherapy ; 15(3): 263-6, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23253439

ABSTRACT

Diabetic retinopathy (DR), a leading cause of vision loss and a significant source of morbidity, is the most common ocular complication of prolonged diabetes mellitus. Most therapeutic approaches address DR by preventing or destroying neovasculature; however, this fails to eliminate pathogenic causes. Mesenchymal stem cells (MSCs) are a promising candidate for cell therapy because they have unique regenerative potential and provide an option to manage retinal injuries. Transplantation of MSCs in rats with diabetes induced by streptozocin administration was shown to ameliorate DR. However, the poor viability and homing of MSCs after transplantation may reduce the efficacy of cell therapy. Intravitreal transplantation of MSCs was shown to augment vascular endothelial growth factor (VEGF). More recent studies have found a central role for VEGF in vascular lesion formation in DR and proposed blockage of VEGF as an effective approach to manage DR. Atorvastatin, a 3-hydroxy-3-methyl-glutaryl coenzyme A reductase inhibitor, has been proven to decrease VEGF production of MSCs under hypoxic conditions. It has also been demonstrated that atorvastatin increases the viability of MSCs through the adenosine monophosphate-activated protein kinase-endothelial nitric oxide synthase signaling pathway. There is also evidence that nitric oxide improves homing of MSCs by increasing chemokine-related receptor CXCR4 expression. It could be hypothesized that co-administration of MSCs with atorvastatin may be a significant step forward in development of an efficient MSC therapy of DR through preventing excess VEGF production by MSCs under hypoxic conditions as well as increasing the viability and homing of transplanted MSCs.


Subject(s)
Diabetic Retinopathy/therapy , Heptanoic Acids/administration & dosage , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Pyrroles/administration & dosage , Vascular Endothelial Growth Factor A/metabolism , Animals , Atorvastatin , Diabetic Retinopathy/pathology , Gene Expression Regulation , Genetic Therapy , Humans , Mesenchymal Stem Cells/cytology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III , Rats , Receptors, CXCR4/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors
12.
Biocell ; 36(1): 37-45, 2012 Apr.
Article in English | MEDLINE | ID: mdl-23173303

ABSTRACT

The ultimate goal of tissue engineering is to design and fabricate functional human tissues that are similar to natural cells and are capable of regeneration. Preparation of cell aggregates is one of the important steps in 3D tissue engineering technology, particularly in organ printing. Two simple methods, hanging drop (HD) and conical tube (CT) were utilized to prepare cell aggregates. The size and viability of the aggregates obtained at different initial cell densities and pre-culture duration were compared. The proliferative ability of the cell aggregates and their ability to spread in culture plates were also investigated. In both methods, the optimum average size of the aggregates was less than 500 microm. CT aggregates were smaller than HD aggregates. 5,000 cells per drop HD aggregates showed a marked ability to attach and spread on the culture surface. The proliferative ability reduced when the initial cell density was increased. Comparing these methods, we found that the HD method having better size controlling ability as well as enhanced ability to maintain higher rates of viability, spreading, and proliferation. In conclusion, smaller HD aggregates might be a suitable choice as building blocks for making bioink particles in bioprinting technique.


Subject(s)
Bioprinting/instrumentation , Cell Aggregation/physiology , Cell Culture Techniques/methods , Cell Proliferation , Tissue Engineering/instrumentation , Animals , Bioprinting/methods , CHO Cells , Cell Size , Cell Survival , Cells, Cultured , Cricetinae , Humans , Tissue Engineering/methods
13.
Med Hypotheses ; 79(6): 717-8, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22981008

ABSTRACT

Recent studies have proposed cell therapy as an alternative therapeutic strategy for many disease states such as diabetes mellitus. Among different cell types mesenchymal stem cells (MSC) have attracted a significant attention based on their intriguing potentials. However MSC therapy is limited as a large portion of transplanted cells undergo apoptosis after transplantation. Therefore, proposing a strategy to overcome this obstacle may be of great value. Recent studies have shown that hypoxia preconditioning (HPC) may improve cell viability after transplantation. Both HPC and hyperglycemia are reported to exert effects by different levels of ROS overproduction. Overdose of ROS in this case would trigger the apoptosis and thereby decreased cell viability after transplantation. Apelin; the endogenous ligand for the previously orphaned G protein-coupled receptor APJ is shown to exert anti apoptotic effects On oxidative stress-induced apoptosis in MSCs via MAPK/ERK1/2 and PI3K/AKT signaling pathways. Accordingly it has been hypothesized that pretreatment of HPC-MSC(s) with apelin 13 would be an effective approach to modify and possibly enhance the efficacy of MSCs in cell therapy of diabetes.


Subject(s)
Cell Hypoxia , Diabetes Mellitus/surgery , Intercellular Signaling Peptides and Proteins/physiology , Mesenchymal Stem Cell Transplantation , Apelin , Apoptosis , Humans , Models, Theoretical , Reactive Oxygen Species/metabolism
14.
Biocell ; 36(1): 37-45, Apr. 2012. ilus, graf, tab
Article in English | LILACS | ID: lil-657492

ABSTRACT

The ultimate goal of tissue engineering is to design and fabricate functional human tissues that are similar to natural cells and are capable of regeneration. Preparation of cell aggregates is one of the important steps in 3D tissue engineering technology, particularly in organ printing. Two simple methods, hanging drop (HD) and conical tube (CT) were utilized to prepare cell aggregates. The size and viability of the aggregates obtained at different initial cell densities and pre-culture duration were compared. The proliferative ability of the cell aggregates and their ability to spread in culture plates were also investigated. In both methods, the optimum average size of the aggregates was less than 500 µm. CT aggregates were smaller than HD aggregates. 5,000 cells per drop HD aggregates showed a marked ability to attach and spread on the culture surface. The proliferative ability reduced when the initial cell density was increased. Comparing these methods, we found that the HD method having better size controlling ability as well as enhanced ability to maintain higher rates of viability, spreading, and proliferation. In conclusion, smaller HD aggregates might be a suitable choice as building blocks for making bioink particles in bioprinting technique.


Subject(s)
Animals , Cricetinae , Humans , Bioprinting/instrumentation , Cell Proliferation , Cell Aggregation/physiology , Cell Culture Techniques/methods , Tissue Engineering/instrumentation , Bioprinting/methods , Cell Size , Cell Survival , Cells, Cultured , CHO Cells , Tissue Engineering/methods
15.
Biocell ; 36(1): 37-45, Apr. 2012. ilus, graf, tab
Article in English | BINACIS | ID: bin-129345

ABSTRACT

The ultimate goal of tissue engineering is to design and fabricate functional human tissues that are similar to natural cells and are capable of regeneration. Preparation of cell aggregates is one of the important steps in 3D tissue engineering technology, particularly in organ printing. Two simple methods, hanging drop (HD) and conical tube (CT) were utilized to prepare cell aggregates. The size and viability of the aggregates obtained at different initial cell densities and pre-culture duration were compared. The proliferative ability of the cell aggregates and their ability to spread in culture plates were also investigated. In both methods, the optimum average size of the aggregates was less than 500 Am. CT aggregates were smaller than HD aggregates. 5,000 cells per drop HD aggregates showed a marked ability to attach and spread on the culture surface. The proliferative ability reduced when the initial cell density was increased. Comparing these methods, we found that the HD method having better size controlling ability as well as enhanced ability to maintain higher rates of viability, spreading, and proliferation. In conclusion, smaller HD aggregates might be a suitable choice as building blocks for making bioink particles in bioprinting technique.(AU)


Subject(s)
Animals , Cricetinae , Humans , Bioprinting/instrumentation , Cell Aggregation/physiology , Cell Culture Techniques/methods , Cell Proliferation , Tissue Engineering/instrumentation , Bioprinting/methods , CHO Cells , Cell Size , Cell Survival , Cells, Cultured , Tissue Engineering/methods
16.
Med Hypotheses ; 78(4): 465-7, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22289340

ABSTRACT

Mesenchymal stem cells (MSCs) therapy has emerged as a potent therapeutic strategy to improve myocardial infarction. However MSCs therapy encounters a few obstacles regarding the poor viability of the transplanted cells. Therefore, it is important to explore a strategy to enhance post-transplanted MSC viability. To overcome this problem, several protocols were suggested mainly by activating PI3K/Akt pathway. The PI3K/Akt cascade regulates several cellular processes such as proliferation and apoptosis. Finasteride is a specific inhibitor of type II 5α-reductase; the enzyme converts testosterone (T) to the more potent androgen receptor agonist dihydrotestosterone (DHT). Testosterone is found to stimulate rapid phosphorylation of Akt, and thereby activate the PI3K/Akt pathway. This pathway could lead to decreased apoptosis of the MSCs via increasing the expression of Bcl-2 and reducing Bax expression. It has been also reported that DHT would confine the differentiation capacity of MSCs so that a reduction in DHT levels caused by Finasteride would be accompanied by increased facilitation in differentiation of MSCs to cardiomyocyte by means of the signals originating from the injured cardiac tissue. These mechanisms could propose the potential role for Finasteride to improve the MSCs therapy for myocardial infarction.


Subject(s)
Cell Survival/drug effects , Finasteride/pharmacology , Mesenchymal Stem Cell Transplantation/methods , Models, Biological , Myocardial Infarction/therapy , Signal Transduction/drug effects , Apoptosis/physiology , Dihydrotestosterone/metabolism , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , bcl-2-Associated X Protein/metabolism
17.
Toxicol Mech Methods ; 21(3): 225-30, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21244221

ABSTRACT

Lead exposure continues to be a significant public health problem. Osteoporosis, inhibition of fracture healing, and cartilage functional impairment have been reported from lead exposure. Mesenchymal stem cells (MSCs) are a bone marrow population of cells with the ability to differentiate into various cell types, particularly osteocytes and chondrocytes. Despite intensive investigation on the effect of lead poisoning on various cell types, there is very little if any report on the effect of lead on MSCs. The aim of this study, therefore, was to investigate the effect of lead acetate on rat bone marrow derived MSCs toxicity and its mechanism by examining the role of pro- and anti-apoptotic proteins in this process. It was revealed that lead acetate could induce cell death in a dose-dependent manner using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl tetrazolium (MTT) assay. Compared to controls, the significant over-expression of pro-apoptotic proteins, including Bax, caspases-9, -3, and p53, with no significant change in anti-apoptotic Bcl(2) protein were obtained in lead-treated cells using western blotting analysis. There was a significant increase in DNA fragmentation in treated MSCs compared to controls using flow-cytometry. Finally, it might be concluded that lead acetate could induce cell toxicity and apoptosis in MSCs, causing instability in mitochondria and in turn activation of the intrinsic pathway including over-expression of Bax, caspase 9 and caspase 3, leading to DNA damage and activation of P53.


Subject(s)
Apoptosis/drug effects , Bone Marrow/drug effects , Environmental Pollutants/toxicity , Mesenchymal Stem Cells/drug effects , Organometallic Compounds/toxicity , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Survival/drug effects , Male , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/metabolism
18.
Toxicol Mech Methods ; 20(6): 316-20, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20521854

ABSTRACT

Gram-negative bacteria and their endotoxins may be a causal or complicating factor in many serious diseases. Bacterial lipopolysaccharides (LPS) could potentially be human pathogens. Among the many disorders induced by LPS, neurodegenerative diseases such as Parkinson's are reported and are of great interest. Despite the evidence on LPS-induced neurodegeneration, the exact mechanism is unknown. The purpose of this study was to investigate the cytotoxic effect of LPS and also to examine the involvement of Bax, pro-apoptotic, Bcl-2, anti-apoptotic, and caspase-3, the executioner of apoptosis, protein expression, during LPS-induced apoptosis in neuronal PC12 cells. The cell viability was evaluated by MTT assy. The expression of pro-apoptotic Bax and anti-apoptotic Bcl2 and caspase -3 protein expressions were measured by immunoblotting. The results showed that LPS could reduce cell viability after 72 h in a dose-dependent manner which was statistically significant in concentration of 200 microg/ml. In western blotting analysis, LPS (200 microg/ml) also enhanced expression of pro-apoptotic Bax and pro-caspase-3 proteins compared to controls, while the expression of Bcl-2 protein was not changed significantly. The Bax/Bcl-2 ratio was significantly increased in LPS-treated cells compared to controls. From the present results, it might be concluded that LPS can cause PC12 cell death, in which apoptosis plays an important role, possibly by the mitochondrial pathway through higher expression of the Bax as well as caspase 3 protein.


Subject(s)
Apoptosis/drug effects , Caspase 3/biosynthesis , Lipopolysaccharides/toxicity , Neurons/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , bcl-2-Associated X Protein/biosynthesis , Animals , Blotting, Western , Cell Culture Techniques , Cell Survival/drug effects , Neurons/enzymology , Neurons/metabolism , Neurons/pathology , PC12 Cells , Rats
19.
Biocell ; 34(1): 53-5, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20506631

ABSTRACT

The effects of human versus mouse EGF on cell growth and culture duration were studied to optimize a human limbal stem cells culture method for therapeutical autologous transplantation. Limbal cells were obtained by trypsin digestion and transferred to a culture medium. The time needed to reach full confluence in culture was determined. Specific antibodies to corneal stem cell marker (P63) versus corneal epithelial differentiation marker (K3) were used for histochemical determinations. A high proportion of P63 positive cells (85 +/- 4.6%), and a correspondingly low proportion K3 positive cells (15 +/- 3.8%) indicated that most cultured cells remained undifferentiated and were considered as stem cells (mean +/- SE, n=10). Cultures reached full confluency after 17.3 +/- 1.2 days when the medium was supplemented with human EGF, while 21.7 +/- 1.5 days were needed when the medium was supplemented with mouse EGF. The results showed that limbal stem cells proliferate more easily and reach to full confluency in a shorter time if the medium is supplemented with hEGF rather than with mEGF.


Subject(s)
Adult Stem Cells/cytology , Limbus Corneae/cytology , Adult Stem Cells/drug effects , Animals , Cell Culture Techniques , Cell Proliferation/drug effects , Cell Separation , Cells, Cultured , Epidermal Growth Factor/pharmacology , Epithelium, Corneal/cytology , Humans , Mice , Species Specificity
20.
Biocell ; 34(1): 53-55, Apr. 2010. ilus
Article in English | LILACS | ID: lil-595043

ABSTRACT

The effects of human versus mouse EGF on cell growth and culture duration were studied to optimize a human limbal stem cells culture method for therapeutical autologous transplantation. Limbal cells were obtained by trypsin digestion and transferred to a culture medium. The time needed to reach full confluence in culture was determined. Specific antibodies to corneal stem cell marker (P63) versus corneal epithelial differentiation marker (K3) were used for histochemical determinations. A high proportion of P63 positive cells (85 +/- 4.6%), and a correspondingly low proportion K3 positive cells (15 +/- 3.8%) indicated that most cultured cells remained undifferentiated and were considered as stem cells (mean +/- SE, n=10). Cultures reached full confluency after 17.3 +/- 1.2 days when the medium was supplemented with human EGF, while 21.7 +/- 1.5 days were needed when the medium was supplemented with mouse EGF. The results showed that limbal stem cells proliferate more easily and reach to full confluency in a shorter time if the medium is supplemented with hEGF rather than with mEGF.


Subject(s)
Humans , Animals , Adult Stem Cells/cytology , Adult Stem Cells , Epithelium, Corneal/cytology , Epidermal Growth Factor/pharmacology , Limbus Corneae/cytology , Cell Proliferation , Cell Culture Techniques , Cell Separation , Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...