Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
2.
Exp Mol Med ; 52(4): 615-628, 2020 04.
Article in English | MEDLINE | ID: mdl-32273566

ABSTRACT

The mammalian target of rapamycin (mTOR) signaling pathway efficiently regulates the energy state of cells and maintains tissue homeostasis. Dysregulation of the mTOR pathway has been implicated in several human diseases. Rapamycin is a specific inhibitor of mTOR and pharmacological inhibition of mTOR with rapamycin promote cardiac cell generation from the differentiation of mouse and human embryonic stem cells. These studies strongly implicate a role of sustained mTOR activity in the differentiating functions of embryonic stem cells; however, they do not directly address the required effect for sustained mTOR activity in human cardiac progenitor cells. In the present study, we evaluated the effect of mTOR inhibition by rapamycin on the cellular function of human cardiac progenitor cells and discovered that treatment with rapamycin markedly attenuated replicative cell senescence in human cardiac progenitor cells (hCPCs) and promoted their cellular functions. Furthermore, rapamycin not only inhibited mTOR signaling but also influenced signaling pathways, including STAT3 and PIM1, in hCPCs. Therefore, these data reveal a crucial function for rapamycin in senescent hCPCs and provide clinical strategies based on chronic mTOR activity.


Subject(s)
Cellular Senescence/drug effects , Myoblasts, Cardiac/drug effects , Myoblasts, Cardiac/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/antagonists & inhibitors , Cell Differentiation , Cell Proliferation , Cells, Cultured , Computational Biology/methods , Gene Expression Profiling , Humans , Sirolimus/pharmacology , Stem Cells/metabolism
3.
Tissue Eng Regen Med ; 17(3): 323-333, 2020 06.
Article in English | MEDLINE | ID: mdl-32227286

ABSTRACT

BACKGROUND: Despite promising advances in stem cell-based therapy, the treatment of ischemic cardiovascular diseases remains a big challenge due to both the insufficient in vivo viability of transplanted cells and poor angiogenic potential of stem cells. The goal of this study was to develop therapeutic human cardiac progenitor cells (hCPCs) for ischemic cardiovascular diseases with a novel M13 peptide carrier. METHOD: In this study, an engineered M13 peptide carrier was successfully generated using a QuikChange Kit. The cellular function of M13 peptide carrier-treated hCPCs was assessed using a tube formation assay and scratch wound healing assay. The in vivo engraftment and cell survival bioactivities of transplanted cells were demonstrated by immunohistochemistry after hCPC transplantation into a myocardial infarction animal model. RESULTS: The engineered M13RGD+SDKP peptide carrier, which expressed RGD peptide on PIII site and SDKP peptide on PVIII site, did not affect morphologic change and proliferation ability in hCPCs. In contrast, hCPCs treated with M13RGD+SDKP showed enhanced angiogenic capacity, including tube formation and migration capacity. Moreover, transplanted hCPCs with M13RGD+SDKP were engrafted into the ischemic region and promoted in vivo cell survival. CONCLUSION: Our present data provides a promising protocol for CPC-based cell therapy via short-term cell priming of hCPCs with engineered M13RGD+SDKP before cell transplantation for treatment of cardiovascular disease.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Myocardial Infarction/therapy , Peptides/metabolism , Stem Cell Transplantation , Stem Cells/drug effects , Animals , Bacteriophage M13/genetics , Cardiovascular Diseases , Cell Survival , Endothelial Cells , Genetic Engineering , Humans , Male , Mice, Inbred BALB C , Myocytes, Cardiac/transplantation , Peptides/pharmacology , Wound Healing
4.
Mar Drugs ; 17(7)2019 Jul 03.
Article in English | MEDLINE | ID: mdl-31277207

ABSTRACT

The purpose of the present study is to improve the endothelial progenitor cells (EPC) activation, proliferation, and angiogenesis using enzyme-aided extraction of fucoidan by amyloglucosidase (EAEF-AMG). Enzyme-aided extraction of fucoidan by AMG (EAEF-AMG) significantly increased EPC proliferation by reducing the reactive oxygen species (ROS) and decreasing apoptosis. Notably, EAEF-AMG treated EPCs repressed the colocalization of TSC2/LAMP1 and promoted perinuclear localization of mTOR/LAMP1 and mTOR/Rheb. Moreover, EAEF-AMG enhanced EPC functionalities, including tube formation, cell migration, and wound healing via regulation of AKT/Rheb signaling. Our data provided cell priming protocols to enhance therapeutic applications of EPCs using bioactive compounds for the treatment of CVD.


Subject(s)
Endothelial Progenitor Cells/drug effects , Glucan 1,4-alpha-Glucosidase/metabolism , Polysaccharides/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Neovascularization, Physiologic/drug effects , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein/metabolism , Wound Healing/drug effects
5.
Biochem Biophys Res Commun ; 515(4): 600-606, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31178140

ABSTRACT

Colorectal cancer is one of the leading causes of cancer-related deaths. Due to relapse after current therapy regimens, cancer stem cells (CSCs) are being studied to target this small tumor-initiating population. Anterior gradient 2 (AGR2), a disulfide isomerase protein, is a well-known pro-oncogenic/metastatic oncogene overexpressed in various tumor tissues, including colon cancer. We found that AGR2 was a novel stem cell marker that was regulated by the canonical Wnt/ß-catenin pathway in colon CSCs. AGR2 was highly co-expressed with surface stem cell markers in spheroidal culture. Silencing of AGR2 resulted in decreased sphere-forming ability and down-regulated expression of stem cell markers, whereas the opposite effects were seen with AGR2 overexpression. Moreover, patients with high ß-catenin and AGR2 expression showed lower overall survival than those with low expression. In conclusion, our study describes a novel role for AGR2 as a stem cell marker that is highly regulated by canonical Wnt/ß-catenin signaling in colorectal cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Mucoproteins/metabolism , Neoplastic Stem Cells/metabolism , Oncogene Proteins/metabolism , Wnt Signaling Pathway , Cell Line, Tumor , Gene Expression Profiling , Gene Silencing , HCT116 Cells , HEK293 Cells , Humans , Neoplasm Metastasis , Signal Transduction , Spheroids, Cellular , Wnt Proteins/metabolism , beta Catenin/metabolism
6.
Biochem Biophys Res Commun ; 516(1): 149-156, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31202462

ABSTRACT

Anterior gradient protein 2 homolog (AGR2) belongs to the disulfide isomerase family of endoplasmic reticulum proteins. Itis overexpressed in several types of solid tumors, including tumors of the prostate, lung, and pancreas. However, the role of AGR2 in breast cancer and the regulatory mechanisms underlying AGR2 protein expressionare not fullyunderstood. We demonstrated that AGR2 levels are increased under hypoxic conditions and in breast cancer tumors. Mechanistically, Twist1 binds to, and activates the AGR2 promoter via an E-box sequence. Under hypoxic conditions, the increased expression of ARG2 is attenuated when Twist1 levels are reduced by shRNA. Conversely, Twist1 overexpression fully reverses decreased AGR2 levels upon HIF-1α knockdown. Notably, AGR2 is required for Twist1-induced proliferation, migration, and invasion of breast cancer cells. Collectively, these findings extend our understanding of AGR2 regulation in breast cancer and may contribute to development of Twist1-AGR2 targeting therapeutics for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mucoproteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Twist-Related Protein 1/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Humans , Middle Aged , Promoter Regions, Genetic
7.
Oxid Med Cell Longev ; 2019: 6492029, 2019.
Article in English | MEDLINE | ID: mdl-31223423

ABSTRACT

Cardiovascular diseases (CVDs) are a major cause of death worldwide. Due to the prevalence of many side effects and incomplete recovery from pharmacotherapies, stem cell therapy is being targeted for the treatment of CVDs. Among the different types of stem cells, endothelial progenitor cells (EPCs) have great potential. However, cellular replicative senescence decreases the proliferation, migration, and overall function of EPCs. Sirtuin 1 (SIRT1) has been mainly studied in the mammalian aging process. MHY2233 is a potent synthetic SIRT1 activator and a novel antiaging compound. We found that MHY2233 increased the expression of SIRT1, and its deacetylase activity thereby decreased expression of the cellular senescence biomarkers, p53, p16, and p21. In addition, MHY2233 decreased senescence-associated beta-galactosidase- (SA-ß-gal-) positive cells and senescence-associated secretory phenotypes (SASPs), such as the secretion of interleukin- (IL-) 6, IL-8, IL-1α, and IL-1ß. MHY2233 treatment protected senescent EPCs from oxidative stress by decreasing cellular reactive oxygen species (ROS) levels, thus enhancing cell survival and function. The angiogenesis, proliferation, and migration of senescent EPCs were enhanced by MHY2233 treatment. Thus, MHY2233 reduces replicative and oxidative stress-induced senescence in EPCs. Therefore, this novel antiaging compound MHY2233 might be considered a potent therapeutic agent for the treatment of age-associated CVDs.


Subject(s)
Benzoxazoles/pharmacology , Endothelial Progenitor Cells/drug effects , Sirtuin 1/metabolism , Cellular Senescence/drug effects , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/metabolism , Fetal Blood/cytology , Fetal Blood/diagnostic imaging , Fetal Blood/metabolism , Humans , Resveratrol/pharmacology , Signal Transduction/drug effects
8.
Mar Drugs ; 17(6)2019 Jun 21.
Article in English | MEDLINE | ID: mdl-31234277

ABSTRACT

Cardiac progenitor cells (CPCs) are resident stem cells present in a small portion of ischemic hearts and function in repairing the damaged heart tissue. Intense oxidative stress impairs cell metabolism thereby decreasing cell viability. Protecting CPCs from undergoing cellular apoptosis during oxidative stress is crucial in optimizing CPC-based therapy. Histochrome (sodium salt of echinochrome A-a common sea urchin pigment) is an antioxidant drug that has been clinically used as a pharmacologic agent for ischemia/reperfusion injury in Russia. However, the mechanistic effect of histochrome on CPCs has never been reported. We investigated the protective effect of histochrome pretreatment on human CPCs (hCPCs) against hydrogen peroxide (H2O2)-induced oxidative stress. Annexin V/7-aminoactinomycin D (7-AAD) assay revealed that histochrome-treated CPCs showed significant protective effects against H2O2-induced cell death. The anti-apoptotic proteins B-cell lymphoma 2 (Bcl-2) and Bcl-xL were significantly upregulated, whereas the pro-apoptotic proteins BCL2-associated X (Bax), H2O2-induced cleaved caspase-3, and the DNA damage marker, phosphorylated histone (γH2A.X) foci, were significantly downregulated upon histochrome treatment of hCPCs in vitro. Further, prolonged incubation with histochrome alleviated the replicative cellular senescence of hCPCs. In conclusion, we report the protective effect of histochrome against oxidative stress and present the use of a potent and bio-safe cell priming agent as a potential therapeutic strategy in patient-derived hCPCs to treat heart disease.


Subject(s)
Myocytes, Cardiac/drug effects , Naphthoquinones/pharmacology , Oxidative Stress/drug effects , Stem Cells/drug effects , Annexin A5/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Caspase 3/metabolism , Cell Survival/drug effects , Cells, Cultured , Cellular Senescence/drug effects , DNA Damage/drug effects , Humans , Hydrogen Peroxide/pharmacology , Myocytes, Cardiac/metabolism , Reperfusion Injury/chemically induced , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Russia , bcl-2-Associated X Protein/metabolism
9.
Stem Cells Int ; 2018: 7453161, 2018.
Article in English | MEDLINE | ID: mdl-30510587

ABSTRACT

Cross talks between the renin-angiotensin system (RAS), sympathetic nervous system, and vascular homeostasis are tightly coordinated in hypertension. Angiotensin II (Ang II), a key factor in RAS, when abnormally activated, affects the number and bioactivity of circulating human endothelial progenitor cells (hEPCs) in hypertensive patients. In this study, we investigated how the augmentation of Ang II regulates adrenergic receptor-mediated signaling and angiogenic bioactivities of hEPCs. Interestingly, the short-term treatment of hEPCs with Ang II drastically attenuated the expression of beta-2 adrenergic receptor (ADRB2), but did not alter the expression of beta-1 adrenergic receptor (ADRB1) and Ang II type 1 receptor (AT1R). EPC functional assay clearly demonstrated that the treatment with ADRB2 agonists significantly increased EPC bioactivities including cell proliferation, migration, and tube formation abilities. However, EPC bioactivities were decreased dramatically when treated with Ang II. Importantly, the attenuation of EPC bioactivities by Ang II was restored by treatment with an AT1R antagonist (telmisartan; TERT). We found that AT1R binds to ADRB2 in physiological conditions, but this binding is significantly decreased in the presence of Ang II. Furthermore, TERT, an Ang II-AT1R interaction blocker, restored the interaction between AT1R and ADRB2, suggesting that Ang II might induce the dysfunction of EPCs via downregulation of ADRB2, and an AT1R blocker could prevent Ang II-mediated ADRB2 depletion in EPCs. Taken together, our report provides novel insights into potential therapeutic approaches for hypertension-related cardiovascular diseases.

10.
Oxid Med Cell Longev ; 2018: 4528184, 2018.
Article in English | MEDLINE | ID: mdl-30002788

ABSTRACT

Diabetic cardiomyopathy (DCM) is tightly linked to heart disorders and dysfunction or death of the cardiomyocytes including resident cardiac progenitor cells (CPCs) in diabetic patients. In order to restore loss of function of resident or transplanted CPCs, much research has focused on novel therapeutic strategies including the discovery of novel function-modulating factors such as reactive oxygen species (ROS) scavengers. Here, we developed and defined a novel antioxidant, MHY-1684, for enhancing the angiogenic potential of CPCs against ROS-related DCM. Short-term treatment with MHY-1684 restored ROS-induced CPC cell death. Importantly, MHY-1684 decreased hyperglycemia-induced mitochondrial ROS generation and attenuated hyperglycemia-induced mitochondrial fragmentation. We observed that the activation process of both Drp1 (phosphorylation at the site of Ser616) and Fis-1 is drastically attenuated when exposed to high concentrations of D-glucose with MHY-1684. Interestingly, phosphorylation of Drp1 at the site of Ser637, which is an inhibitory signal for mitochondrial fusion, is restored by MHY-1684 treatment, suggesting that this antioxidant may affect the activation and inhibition of mitochondrial dynamics-related signaling and mitochondrial function in response to ROS stress. In conclusion, our finding of the novel compound, MHY-1684, as an ROS scavenger, might provide an effective therapeutic strategy for CPC-based therapy against diabetic cardiomyopathy.


Subject(s)
Antioxidants/pharmacology , Hyperglycemia/metabolism , Mitochondria/metabolism , Oxidative Stress/drug effects , Stem Cells/metabolism , Blotting, Western , Cell Survival/drug effects , Cells, Cultured , Dynamins , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Peroxynitrous Acid/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Stem Cells/cytology , Stem Cells/drug effects
11.
Mol Cells ; 41(6): 582-590, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29890822

ABSTRACT

Endothelial progenitor cells (EPCs) and outgrowth endothelial cells (OECs) play a pivotal role in vascular regeneration in ischemic tissues; however, their therapeutic application in clinical settings is limited due to the low quality and quantity of patient-derived circulating EPCs. To solve this problem, we evaluated whether three priming small molecules (tauroursodeoxycholic acid, fucoidan, and oleuropein) could enhance the angiogenic potential of EPCs. Such enhancement would promote the cellular bioactivities and help to develop functionally improved EPC therapeutics for ischemic diseases by accelerating the priming effect of the defined physiological molecules. We found that preconditioning of each of the three small molecules significantly induced the differentiation potential of CD34+ stem cells into EPC lineage cells. Notably, long-term priming of OECs with the three chemical cocktail (OEC-3C) increased the proliferation potential of EPCs via ERK activation. The migration, invasion, and tube-forming capacities were also significantly enhanced in OEC-3Cs compared with unprimed OECs. Further, the cell survival ratio was dramatically increased in OEC-3Cs against H2O2-induced oxidative stress via the augmented expression of Bcl-2, a prosurvival protein. In conclusion, we identified three small molecules for enhancing the bioactivities of ex vivo-expanded OECs for vascular repair. Long-term 3C priming might be a promising methodology for EPC-based therapy against ischemic diseases.


Subject(s)
Endothelial Progenitor Cells/metabolism , Cell Differentiation , Cell Proliferation , Humans
12.
Biochem Biophys Res Commun ; 499(3): 675-680, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29604275

ABSTRACT

Mesenchymal stem cells (MSCs) are multipotent progenitor cells with self-renewing properties; thus, transplanting functionally enhanced MSCs might be a promising strategy for cell therapy against ischemic diseases. However, extensive oxidative damage in ischemic tissue affects the cell fate of transplanted MSCs, eventually resulting in cell damage and autophagic cell death. Oleuropein (OLP) is a bioactive compound isolated from olives and olive oil that harbors antioxidant properties. This study aimed to investigate the potential cytoprotective effects of OLP against oxidative stress and autophagic cell death in MSCs. We found that short-term priming with OLP attenuated H2O2-induced apoptosis by regulating the pro-apoptotic marker Bax and the anti-apoptotic markers Bcl-2 and Mcl-1. Notably, OLP inhibits H2O2 -induced autophagic cell death by modulating autophagy-related death signals, including mTOR (mammalian target of rapamycin), ULK1 (unc-51 like autophagy activating kinase 1), Beclin-1, AMPK (AMP-activated protein kinase), and LC3 (microtubule-associated protein 1a/1b-light chain 3). Our data suggest that OLP might reduce H2O2-induced autophagy and cell apoptosis in MSCs by regulating both the AMPK-ULK axis and the Bcl-2-Mcl-1 axis. Consequently, short-term cell priming with OLP might enhance the therapeutic effect of MSCs against ischemic vascular diseases, which provides an important potential improvement for emerging therapeutic strategies.


Subject(s)
Adipose Tissue/pathology , Autophagy/drug effects , Hydrogen Peroxide/toxicity , Iridoids/pharmacology , Mesenchymal Stem Cells/pathology , AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , Autophagy-Related Protein-1 Homolog/metabolism , Cells, Cultured , Cytoprotection/drug effects , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Iridoid Glucosides , Iridoids/chemistry , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Signal Transduction/drug effects
13.
Korean J Physiol Pharmacol ; 22(2): 203-213, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29520173

ABSTRACT

Tumor undergo uncontrolled, excessive proliferation leads to hypoxic microenvironment. To fulfill their demand for nutrient, and oxygen, tumor angiogenesis is required. Endothelial progenitor cells (EPCs) have been known to the main source of angiogenesis because of their potential to differentiation into endothelial cells. Therefore, understanding the mechanism of EPC-mediated angiogenesis in hypoxia is critical for development of cancer therapy. Recently, mitochondrial dynamics has emerged as a critical mechanism for cellular function and differentiation under hypoxic conditions. However, the role of mitochondrial dynamics in hypoxia-induced angiogenesis remains to be elucidated. In this study, we demonstrated that hypoxia-induced mitochondrial fission accelerates EPCs bioactivities. We first investigated the effect of hypoxia on EPC-mediated angiogenesis. Cell migration, invasion, and tube formation was significantly increased under hypoxic conditions; expression of EPC surface markers was unchanged. And mitochondrial fission was induced by hypoxia time-dependent manner. We found that hypoxia-induced mitochondrial fission was triggered by dynamin-related protein Drp1, specifically, phosphorylated DRP1 at Ser637, a suppression marker for mitochondrial fission, was impaired in hypoxia time-dependent manner. To confirm the role of DRP1 in EPC-mediated angiogenesis, we analyzed cell bioactivities using Mdivi-1, a selective DRP1 inhibitor, and DRP1 siRNA. DRP1 silencing or Mdivi-1 treatment dramatically reduced cell migration, invasion, and tube formation in EPCs, but the expression of EPC surface markers was unchanged. In conclusion, we uncovered a novel role of mitochondrial fission in hypoxia-induced angiogenesis. Therefore, we suggest that specific modulation of DRP1-mediated mitochondrial dynamics may be a potential therapeutic strategy in EPC-mediated tumor angiogenesis.

14.
BMB Rep ; 51(2): 92-97, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29335071

ABSTRACT

B cell leukemia/lymphoma 3 (Bcl3) plays a pivotal role in immune homeostasis, cellular proliferation, and cell survival, as a co-activator or co-repressor of transcription of the NF-κB family. Recently, it was reported that Bcl3 positively regulates pluripotency genes, including Oct4, in mouse embryonic stem cells (mESCs). However, the role of Bcl3 in the maintenance of pluripotency and self-renewal activity is not fully established. Here, we report the dynamic regulation of the proliferation, pluripotency, and self-renewal of mESCs by Bcl3 via an influence on Nanog transcriptional activity. Bcl3 expression is predominantly observed in immature mESCs, but significantly decreased during cell differentiation by LIF depletion and in mESC-derived EBs. Importantly, the knockdown of Bcl3 resulted in the loss of self-renewal ability and decreased cell proliferation. Similarly, the ectopic expression of Bcl3 also resulted in a significant reduction of proliferation, and the self-renewal of mESCs was demonstrated by alkaline phosphatase staining and clonogenic single cell-derived colony assay. We further examined that Bcl3-mediated regulation of Nanog transcriptional activity in mESCs, which indicated that Bcl3 acts as a transcriptional repressor of Nanog expression in mESCs. In conclusion, we demonstrated that a sufficient concentration of Bcl3 in mESCs plays a critical role in the maintenance of pluripotency and the self-renewal of mESCs via the regulation of Nanog transcriptional activity. [BMB Reports 2018; 51(2): 92-97].


Subject(s)
Cell Self Renewal , Down-Regulation/genetics , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Nanog Homeobox Protein/genetics , Pluripotent Stem Cells/cytology , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Animals , B-Cell Lymphoma 3 Protein , Gene Expression Regulation , Gene Knockdown Techniques , Mice , Models, Biological , Nanog Homeobox Protein/metabolism , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic/genetics
15.
Biomol Ther (Seoul) ; 24(4): 363-70, 2016 Jul 01.
Article in English | MEDLINE | ID: mdl-27350339

ABSTRACT

Cardiovascular disease is the most common cause of death in diabetic patients. Hyperglycemia is the primary characteristic of diabetes and is associated with many complications. The role of hyperglycemia in the dysfunction of human cardiac progenitor cells that can regenerate damaged cardiac tissue has been investigated, but the exact mechanism underlying this association is not clear. Thus, we examined whether hyperglycemia could regulate mitochondrial dynamics and lead to cardiac progenitor cell dysfunction, and whether blocking glucose uptake could rescue this dysfunction. High glucose in cardiac progenitor cells results in reduced cell viability and decreased expression of cell cycle-related molecules, including CDK2 and cyclin E. A tube formation assay revealed that hyperglycemia led to a significant decrease in the tube-forming ability of cardiac progenitor cells. Fluorescent labeling of cardiac progenitor cell mitochondria revealed that hyperglycemia alters mitochondrial dynamics and increases expression of fission-related proteins, including Fis1 and Drp1. Moreover, we showed that specific blockage of GLUT1 improved cell viability, tube formation, and regulation of mitochondrial dynamics in cardiac progenitor cells. To our knowledge, this study is the first to demonstrate that high glucose leads to cardiac progenitor cell dysfunction through an increase in mitochondrial fission, and that a GLUT1 blocker can rescue cardiac progenitor cell dysfunction and downregulation of mitochondrial fission. Combined therapy with cardiac progenitor cells and a GLUT1 blocker may provide a novel strategy for cardiac progenitor cell therapy in cardiovascular disease patients with diabetes.

16.
Mol Cells ; 39(2): 111-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26674968

ABSTRACT

MiR399f plays a crucial role in maintaining phosphate homeostasis in Arabidopsis thaliana. Under phosphate starvation conditions, AtMYB2, which plays a role in plant salt and drought stress responses, directly regulates the expression of miR399f. In this study, we found that miR399f also participates in plant responses to abscisic acid (ABA), and to abiotic stresses including salt and drought. Salt and ABA treatment induced the expression of miR399f, as confirmed by histochemical analysis of promoter-GUS fusions. Transgenic Arabidopsis plants overexpressing miR399f (miR399f-OE) exhibited enhanced tolerance to salt stress and exogenous ABA, but hypersensitivity to drought. Our in silico analysis identified ABF3 and CSP41b as putative target genes of miR399f, and expression analysis revealed that mRNA levels of ABF3 and CSP41b decreased remarkably in miR399f-OE plants under salt stress and in response to treatment with ABA. Moreover, we showed that activation of stress-responsive gene expression in response to salt stress and ABA treatment was impaired in miR399f-OE plants. Thus, these results suggested that in addition to phosphate starvation signaling, miR399f might also modulates plant responses to salt, ABA, and drought, by regulating the expression of newly discovered target genes such as ABF3 and CSP41b.


Subject(s)
Abscisic Acid/pharmacology , Arabidopsis Proteins/genetics , Arabidopsis/genetics , Basic-Leucine Zipper Transcription Factors/genetics , Gene Expression Regulation, Plant , Globulins/genetics , MicroRNAs/genetics , Abscisic Acid/metabolism , Adaptation, Physiological , Arabidopsis/drug effects , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Basic-Leucine Zipper Transcription Factors/metabolism , Droughts , Genes, Reporter , Globulins/metabolism , MicroRNAs/metabolism , Phosphates/deficiency , Plants, Genetically Modified , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Salinity , Signal Transduction , Sodium Chloride/pharmacology , Stress, Physiological , Trans-Activators/genetics , Trans-Activators/metabolism , beta-Glucosidase/genetics , beta-Glucosidase/metabolism
17.
Front Plant Sci ; 6: 963, 2015.
Article in English | MEDLINE | ID: mdl-26583028

ABSTRACT

The phytohormone abscisic acid (ABA) induces accumulation of reactive oxygen species (ROS), which can disrupt seed dormancy and plant development. Here, we report the isolation and characterization of an Arabidopsis thaliana mutant called ars1 (aba and ros sensitive 1) that showed hypersensitivity to ABA during seed germination and to methyl viologen (MV) at the seedling stage. ARS1 encodes a nuclear protein with one zinc finger domain, two nuclear localization signal (NLS) domains, and one nuclear export signal (NES). The ars1 mutants showed reduced expression of a gene for superoxide dismutase (CSD3) and enhanced accumulation of ROS after ABA treatment. Transient expression of ARS1 in Arabidopsis protoplasts strongly suppressed ABA-mediated ROS production. Interestingly, nuclear-localized ARS1 translocated to the cytoplasm in response to treatment with ABA, H2O2, or MV. Taken together, these results suggest that ARS1 modulates seed germination and ROS homeostasis in response to ABA and oxidative stress in plants.

18.
Stem Cells ; 33(5): 1490-500, 2015 May.
Article in English | MEDLINE | ID: mdl-25537795

ABSTRACT

The Lnk adaptor protein is a strong negative regulator that affects self-renewal of hematopoietic stem cells and vascular repair in injured tissues. However, the signaling mechanisms through which these proteins influence the vascular regeneration function of endothelial progenitor cells (EPCs) remain unknown. In this study, we investigated the effect of Lnk-targeted small interfering RNA (si-lnk) on the clonogenic proliferative potential and vascular regenerative function of EPCs and the activation of the JAK/STAT3 signaling pathway. Treatment with stem cell factor (SCF) increased the clonogenic proliferation of si-lnk EPCs. Importantly, activation of the JAK2/STAT3 pathway was enhanced in SCF-sensitized si-lnk EPCs. In a hind limb model of ischemia, transplantation of si-lnk EPCs increased the blood flow ratio, capillary density, proliferation, and survival of transplanted cells, and the secretion of pivotal angiogenic cytokines at ischemic sites. These results provide strong evidence that si-lnk regulates the clonogenic proliferative potential of EPCs through the activation of the JAK2/STAT3 signaling pathway, thereby accelerating angiogenesis and promoting repair in injured hind limb ischemia. Stem Cells 2014;33:1490-1500.


Subject(s)
Endothelial Progenitor Cells/metabolism , Hindlimb/blood supply , Intracellular Signaling Peptides and Proteins/metabolism , Ischemia/therapy , Signal Transduction , Umbilical Cord/cytology , Wound Healing , Adaptor Proteins, Signal Transducing , Animals , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cell Survival/drug effects , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/transplantation , Gene Targeting , Hindlimb/pathology , Ischemia/pathology , Ischemia/physiopathology , Janus Kinase 2/metabolism , Male , Membrane Proteins , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Recovery of Function/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Stem Cell Factor/pharmacology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Wound Healing/drug effects
19.
PLoS One ; 9(8): e106310, 2014.
Article in English | MEDLINE | ID: mdl-25166961

ABSTRACT

INTRODUCTION: Despite the crucial role of endothelial progenitor cells (EPCs) in vascular regeneration, the specific interactions between EPCs and hematopoietic cells remain unclear. METHODS: In EPC colony forming assays, we first demonstrated that the formation of EPC colonies was drastically increased in the coculture of CD34+ and CD34- cells, and determined the optimal concentrations of CD34+ cells and CD34- cells for spindle-shaped EPC differentiation. RESULTS: Functionally, the coculture of CD34+ and CD34- cells resulted in a significant enhancement of adhesion, tube formation, and migration capacity compared with culture of CD34+ cells alone. Furthermore, blood flow recovery and capillary formation were remarkably increased by the coculture of CD34+ and CD34- cells in a murine hind-limb ischemia model. To elucidate further the role of hematopoietic cells in EPC differentiation, we isolated different populations of hematopoietic cells. T lymphocytes (CD3+) markedly accelerated the early EPC status of CD34+ cells, while macrophages (CD11b+) or megakaryocytes (CD41+) specifically promoted large EPC colonies. CONCLUSION: Our results suggest that specific populations of hematopoietic cells play a role in the EPC differentiation of CD34+ cells, a finding that may aid in the development of a novel cell therapy strategy to overcome the quantitative and qualitative limitations of EPC therapy.


Subject(s)
Antigens, CD34/metabolism , Endothelial Progenitor Cells/physiology , Fetal Blood/cytology , Hindlimb/blood supply , Ischemia/therapy , Animals , Cell Differentiation , Cells, Cultured , Coculture Techniques , Cord Blood Stem Cell Transplantation , Disease Models, Animal , Endothelial Progenitor Cells/cytology , Fetal Blood/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred BALB C
20.
Korean J Physiol Pharmacol ; 18(2): 163-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24757379

ABSTRACT

Endothelial progenitor cells (EPCs) are known to play an important role in the repair of damaged blood vessels. We used an endothelial progenitor cell colony-forming assay (EPC-CFA) to determine whether EPC numbers could be increased in healthy individuals through regular exercise training. The number of functional EPCs obtained from human peripheral blood-derived AC133 stem cells was measured after a 28-day regular exercise training program. The number of total endothelial progenitor cell colony-forming units (EPC-CFU) was significantly increased compared to that in the control group (p=0.02, n=5). In addition, we observed a significant decrease in homocysteine levels followed by an increase in the number of EPC-CFUs (p=0.04, n=5), indicating that the 28-day regular exercise training could increase the number of EPC colonies and decrease homocysteine levels. Moreover, an inverse correlation was observed between small-endothelial progenitor cell colony-forming units (small-EPC-CFUs) and plasma homocysteine levels in healthy men (r=-0.8125, p=0.047). We found that regular exercise training could increase the number of EPC-CFUs and decrease homocysteine levels, thus decreasing the cardiovascular disease risk in men.

SELECTION OF CITATIONS
SEARCH DETAIL
...