Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
In Vivo ; 31(6): 1115-1124, 2017.
Article in English | MEDLINE | ID: mdl-29102933

ABSTRACT

BACKGROUND/AIM: The kidney excretes waste materials and regulates important metabolic functions, and renal disorders constitute a significant medical problem and can result in fatalities. In the present study, mesenchymal stem cells derived from canine umbilical cord blood (cUCB-MSCs) were isolated and evaluated for their ability to improve renal function in a canine model of acute kidney injury (AKI). MATERIALS AND METHODS: The canine AKI model was developed by i.v. injection of cisplatin and gentamycin into 14 male beagle dogs. cUCB-MSCs were administered into the renal corticomedullary junction following AKI induction. Survival time, clinical signs, blood analysis and histological parameters were analyzed. RESULTS: The group treated with AKI plus cUCB-MSCs had decreased blood urea nitrogen and creatinine levels, and showed an extended life-span and improved histological manifestations. MSCs were detected around the tubules of these kidneys at the histological level. CONCLUSION: Taken together, our findings suggest that cUCB-MSCs could be an alternative therapeutic agent for canine AKI.


Subject(s)
Acute Kidney Injury/therapy , Apoptosis , Kidney/physiopathology , Mesenchymal Stem Cell Transplantation , Acute Kidney Injury/blood , Acute Kidney Injury/genetics , Acute Kidney Injury/physiopathology , Animals , Blood Urea Nitrogen , Creatinine/blood , Disease Models, Animal , Dogs , Humans , Kidney/injuries , Mesenchymal Stem Cells/metabolism
2.
ACS Appl Mater Interfaces ; 9(27): 22568-22577, 2017 Jul 12.
Article in English | MEDLINE | ID: mdl-28603967

ABSTRACT

Cellulose, which is one of the most-abundant and -renewable natural resources, has been extensively explored as an alternative substance for electrode materials such as activated carbons. Here, we demonstrate a new class of coffee-mediated green activation of cellulose as a new environmentally benign chemical-activation strategy and its potential use for all-paper flexible supercapacitors. A piece of paper towel is soaked in espresso coffee (acting as a natural activating agent) and then pyrolyzed to yield paper-derived activated carbons (denoted as "EK-ACs"). Potassium ions (K+), a core ingredient of espresso, play a viable role in facilitating pyrolysis kinetics and also in achieving a well-developed microporous structure in the EK-ACs. As a result, the EK-ACs show significant improvement in specific capacitance (131 F g-1 at a scan rate of 1.0 mV s-1) over control ACs (64 F g-1) obtained from the carbonization of a pristine paper towel. All-paper flexible supercapacitors are fabricated by assembling EK-ACs/carbon nanotube mixture-embedded paper towels (as electrodes), poly(vinyl alcohol)/KOH mixture-impregnated paper towels (as electrolytes), and polydimethylsiloxane-infiltrated paper towels (as packaging substances). The introduction of the EK-ACs (as an electrode material) and the paper towel (as a deformable and compliant substrate) enables the resulting all-paper supercapacitor to provide reliable and sustainable cell performance as well as exceptional mechanical flexibility. Notably, no appreciable loss in the cell capacitance is observed after repeated bending (over 5000 cycles) or multiple folding. The coffee-mediated green activation of cellulose and the resultant all-paper flexible supercapacitors open new material and system opportunities for eco-friendly high-performance flexible power sources.

3.
J Vet Sci ; 18(1): 59-65, 2017 Mar 30.
Article in English | MEDLINE | ID: mdl-27297412

ABSTRACT

Retinal pigment epithelium (RPE) is a major component of the eye. This highly specialized cell type facilitates maintenance of the visual system. Because RPE loss induces an irreversible visual impairment, RPE generation techniques have recently been investigated as a potential therapeutic approach to RPE degeneration. A microRNA-based technique is a new strategy for producing RPE cells from adult stem cell sources. Previously, we identified that antisense microRNA-410 (anti-miR-410) induces RPE differentiation from amniotic epithelial stem cells. In this study, we investigated RPE differentiation from umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) via anti-miR-410 treatment. We identified miR-410 as a RPE-relevant microRNA in UCB-MSCs from among 21 putative human RPE-depleted microRNAs. Inhibition of miR-410 induces overexpression of immature and mature RPE-specific factors, including MITF, LRAT, RPE65, Bestrophin, and EMMPRIN. The RPE-induced cells were able to phagocytize microbeads. Results of our microRNA-based strategy demonstrated proof-of-principle for RPE differentiation in UCB-MSCs by using anti-miR-410 treatment without the use of additional factors or exogenous transduction.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/metabolism , Otx Transcription Factors/biosynthesis , Retinal Pigment Epithelium/physiology , cis-trans-Isomerases/biosynthesis , Fetal Blood/cytology , Fetal Blood/metabolism , Gene Expression Regulation, Developmental , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Phagocytosis , Retinal Pigment Epithelium/metabolism
4.
Stem Cell Rev Rep ; 11(3): 376-86, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25351180

ABSTRACT

The retinal pigment epithelium (RPE) is a highly specialized cell type located between the choroid and neural retina of the eye. RPE degeneration causes irreversible visual impairment, extending to blindness. Cell therapy has recently emerged as a potential therapeutic approach for retinal degeneration. MicroRNA-based differentiation of stem cells is a new strategy for producing tissue-specific cell types. In this study, we developed a novel microRNA-based strategy for RPE induction from human amniotic epithelial stem cells (AESCs). We identified microRNAs involved in RPE development in AESCs. Of 29 putative human RPE-relevant microRNAs, microRNA-410 (miR-410) was predicted to target multiple RPE development-relevant genes. Inhibition of miR-410 induces overexpression of immature and mature RPE-specific factors, including OTX2, RPE65, Bestrophin and EMMPRIN. These RPE-like cells were morphologically altered toward a cobblestone-like shape and were able to phagocytize microbeads. We showed that miR-410 directly regulates predicted target genes OTX2 and RPE65. Our microRNA-based strategy demonstrated RPE differentiation in AESCs by treatment of an antisense microRNA-410 (anti-miR-410), without the use of additional factors or exogenous transduction. These findings suggest that miR-410 inhibition can be a useful tool for directed cell differentiation and an attractive method for cell therapy in human retinal degenerative diseases.


Subject(s)
Cell Differentiation/genetics , MicroRNAs/metabolism , Otx Transcription Factors/biosynthesis , cis-trans-Isomerases/biosynthesis , Amniotic Fluid/cytology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , Humans , MicroRNAs/genetics , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Stem Cells/cytology , Stem Cells/metabolism
5.
Nano Lett ; 14(10): 5677-86, 2014 Oct 08.
Article in English | MEDLINE | ID: mdl-25226349

ABSTRACT

The rapidly approaching smart/wearable energy era necessitates advanced rechargeable power sources with reliable electrochemical properties and versatile form factors. Here, as a unique and promising energy storage system to address this issue, we demonstrate a new class of heterolayered, one-dimensional (1D) nanobuilding block mat (h-nanomat) battery based on unitized separator/electrode assembly (SEA) architecture. The unitized SEAs consist of wood cellulose nanofibril (CNF) separator membranes and metallic current collector-/polymeric binder-free electrodes comprising solely single-walled carbon nanotube (SWNT)-netted electrode active materials (LiFePO4 (cathode) and Li4Ti5O12 (anode) powders are chosen as model systems to explore the proof of concept for h-nanomat batteries). The nanoporous CNF separator plays a critical role in securing the tightly interlocked electrode-separator interface. The SWNTs in the SEAs exhibit multifunctional roles as electron conductive additives, binders, current collectors and also non-Faradaic active materials. This structural/physicochemical uniqueness of the SEAs allows significant improvements in the mass loading of electrode active materials, electron transport pathways, electrolyte accessibility and misalignment-proof of separator/electrode interface. As a result, the h-nanomat batteries, which are easily fabricated by stacking anode SEA and cathode SEA, provide unprecedented advances in the electrochemical performance, shape flexibility and safety tolerance far beyond those achievable with conventional battery technologies. We anticipate that the h-nanomat batteries will open 1D nanobuilding block-driven new architectural design/opportunity for development of next-generation energy storage systems.

6.
Cell Transplant ; 23(7): 831-43, 2014.
Article in English | MEDLINE | ID: mdl-23294672

ABSTRACT

Recent studies have reported that stem cells can be isolated from various tissues such as bone marrow, fatty tissue, umbilical cord blood, Wharton's jelly, and placenta. These types of stem cell studies have also arisen in veterinary medicine. Deer antlers show a seasonal regrowth of tissue, an unusual feature in mammals. Antler tissue therefore might offer a source of stem cells. To explore the possibility of stem cell populations within deer antlers, we isolated and successfully cultured antler-derived multipotent stem cells (MSCs). Antler MSCs were maintained in a growth medium, and the proliferation potential was measured via an assay called the cumulative population doubling level. Immunophenotyping and immunostaining revealed the intrinsic characteristic stem cell markers of antler MSCs. To confirm the ability to differentiate, we conducted osteogenic, adipogenic, and chondrogenic induction under the respective differentiation conditions. We discovered that antler MSCs have the ability to differentiate into multiple lineages. In conclusion, our results show that deer antler tissue may contain MSCs and therefore may be a potential source for veterinary regenerative therapeutics.


Subject(s)
Antlers/cytology , Multipotent Stem Cells/cytology , Adipogenesis/drug effects , Animals , Cell Differentiation/drug effects , Cell Lineage , Cell Proliferation/drug effects , Cells, Cultured , Chondrogenesis/drug effects , Culture Media/pharmacology , Deer , Fibroblast Growth Factor 2/pharmacology , Immunophenotyping , Insulin-Like Growth Factor I/pharmacology , Karyotyping , Multipotent Stem Cells/metabolism , Osteogenesis/drug effects , Transcription Factors/metabolism
7.
J Vet Sci ; 14(3): 367-71, 2013.
Article in English | MEDLINE | ID: mdl-23820166

ABSTRACT

Tendinitis of the superficial digital flexor tendon (SDFT) is a significant cause of lameness in horses; however, recent studies have shown that stem cells could be useful in veterinary regenerative medicine. Therefore, we isolated and characterized equine umbilical cord blood mesenchymal stem cells (eUCB-MSCs) from equine umbilical cord blood obtained from thoroughbred mares during the foaling period. Horses that had tendinitis of the SDFT were treated with eUCB-MSCs to confirm the therapeutic effect. After eUCB-MSCs transplantation, the core lesion in the SDFT was found to decrease. These results suggest that transplantation using eUCB-MSCs could be another source of cell treatment.


Subject(s)
Cord Blood Stem Cell Transplantation , Horse Diseases/surgery , Tendinopathy/veterinary , Animals , Cord Blood Stem Cell Transplantation/veterinary , Horses , Male , Tendinopathy/surgery
8.
J Vet Sci ; 14(2): 151-9, 2013.
Article in English | MEDLINE | ID: mdl-23388430

ABSTRACT

Recent studies have shown that mesenchymal stem cells (MSCs) are able to differentiate into multi-lineage cells such as adipocytes, chondroblasts, and osteoblasts. Amniotic membrane from whole placenta is a good source of stem cells in humans. This membrane can potentially be used for wound healing and corneal surface reconstruction. Moreover, it can be easily obtained after delivery and is usually discarded as classified waste. In the present study, we successfully isolated and characterized equine amniotic membrane-derived mesenchymal stem cells (eAM-MSCs) that were cultured and maintained in low glucose Dulbecco's modified Eagle's medium. The proliferation of eAM-MSCs was measured based on the cumulative population doubling level (CPDL). Immunophenotyping of eAM-MSCs by flow cytometry showed that the major population was of mesenchymal origin. To confirm differentiation potential, a multi-lineage differentiation assay was conducted. We found that under appropriate conditions, eAM-MSCs are capable of multi-lineage differentiation. Our results indicated that eAM-MSCs may be a good source of stem cells, making them potentially useful for veterinary regenerative medicine and cell-based therapy.


Subject(s)
Amnion/cytology , Cell Differentiation , Cell Lineage , Mesenchymal Stem Cells/cytology , Adipogenesis , Amnion/physiology , Animals , Cell Proliferation , Chondrogenesis , Female , Flow Cytometry/veterinary , Horses , Immunophenotyping/veterinary , Mesenchymal Stem Cells/physiology , Osteogenesis
9.
Cell Transplant ; 22(9): 1577-90, 2013.
Article in English | MEDLINE | ID: mdl-23294734

ABSTRACT

Stem cell therapy is a potential treatment for spinal cord injury (SCI), and a variety of different stem cell types have been grafted into humans suffering from spinal cord trauma or into animal models of spinal injury. Although several studies have reported functional motor improvement after transplantation of stem cells into injured spinal cord, the benefit of these cells for treating SCI-induced neuropathic pain is not clear. In this study, we investigated the therapeutic effect of transplanting human umbilical cord blood-derived mesenchymal stem cells (hUCB-MSCs) or amniotic epithelial stem cells (hAESCs) on SCI-induced mechanical allodynia (MA) and thermal hyperalgesia (TH) in T13 spinal cord hemisected rats. Two weeks after SCI, hUCB-MSCs or hAESCs were transplanted around the spinal cord lesion site, and behavioral tests were performed to evaluate changes in SCI-induced MA and TH. Immunohistochemical and Western blot analyses were also performed to evaluate possible therapeutic effects on SCI-induced inflammation and the nociceptive-related phosphorylation of the NMDA NR1 receptor subunit. While transplantation of hUCB-MSCs showed a tendency to reduce MA, transplantation of hAESCs significantly reduced MA. Neither hUCB-MSC nor hAESC transplantation had any effect on SCI-induced TH. Transplantation of hAESCs also significantly reduced the SCI-induced increase in NMDA receptor NR1 subunit phosphorylation (pNR1) expression in the spinal cord. Both hUCB-MSCs and hAESCs reduced the SCI-induced increase in spinal cord expression of the microglial marker, F4/80, but not the increased expression of GFAP or iNOS. Taken together, these findings demonstrate that the transplantation of hAESCs into the injured spinal cord can suppress mechanical allodynia, and this effect seems to be closely associated with the modulation of spinal cord microglia activity and NR1 phosphorylation.


Subject(s)
Amnion/cytology , Amniotic Fluid/cytology , Cord Blood Stem Cell Transplantation/methods , Hyperalgesia/surgery , Mesenchymal Stem Cell Transplantation/methods , Spinal Cord Injuries/surgery , Spinal Cord/surgery , Amnion/metabolism , Animals , Disease Models, Animal , Epithelial Cells/transplantation , Humans , Hyperalgesia/etiology , Male , Mesenchymal Stem Cells/cytology , Pain Measurement , Pain Threshold/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Recovery of Function , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/complications
10.
Stem Cell Res ; 10(2): 156-65, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23276696

ABSTRACT

The human high-mobility group protein A2 (HMGA2) protein is an architectural transcription factor that transforms chromatin structure by binding to DNA. Recently, it has been reported that HMGA2 is highly expressed in fetal neural stem cells and has the capacity to promote stemness. However, there is currently no information available on the functional significance and molecular mechanisms of the cellular in vitro aging and proliferation of human umbilical cord blood-derived stromal cells (hUCBSCs). In the present study, we evaluated the direct effects of HMGA2 on the cellular aging and proliferation of hUCBSCs and investigated potential regulatory mechanisms responsible for the corresponding functions. We found that the overexpression of HMGA2 enhanced proliferation and reduced or even reversed the in vitro aging process of hUCBSCs. This effect was accompanied by the increased expression of cyclin E and CDC25A and the significantly decreased expression of cyclin-dependent kinase inhibitors. Furthermore, HMGA2 inhibition compromised cell proliferation and adipogenic differentiation in early-stage hUCBSCs. From the molecular/cellular functional analysis of microarray data, we found that HMGA2 overexpression induced a PI3K/Akt/mTOR/p70S6K cascade, which in turn suppressed the expression of p16(INK4A) and p21(CIP1/WAF1) in hUCBSCs. These results provide novel insights into the mechanism by which HMGA2 regulates the in vitro aging and proliferation of hUCBSCs.


Subject(s)
Cellular Senescence , Fetal Blood/cytology , HMGA2 Protein/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Adipogenesis/genetics , Adult , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Cellular Senescence/genetics , Down-Regulation/genetics , Enzyme Activation , Female , Gene Expression Regulation , HMGA2 Protein/antagonists & inhibitors , Humans , Models, Biological , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Stromal Cells/cytology , Stromal Cells/enzymology , Young Adult , beta-Galactosidase/metabolism
11.
Cell Transplant ; 22(1): 29-40, 2013.
Article in English | MEDLINE | ID: mdl-22963690

ABSTRACT

ZNF281 is one of the core transcription factors in embryonic stem cells (ESCs) and has activation and repression roles in the transcription of ESC genes. A known target molecule of Zfp281 (the mouse homologue of ZNF281) is Nanog. However, NANOG is not expressed in most human multipotent stem cells (hMSCs). Here, we investigated the roles of ZNF281 with a gain- and loss-of-function study. The knockdown of ZNF281 in vivo and in vitro resulted in spontaneous osteochondrogenic differentiation and reduced the proliferation of hMSCs, as determined by cell morphology and molecular markers. When ZNF281-knockdown hMSCs were subcutaneously implanted into mice along with ß-tricalcium phosphate (ß-TCP), many cells were converted into osteoblasts within 4 weeks. In contrast, the overexpression of ZNF281 in hMSCs resulted in accelerated proliferation. The expression pattern of ZNF281 correlated well with the expression of ß-CATENIN during differentiation and in the gain/loss-of-function study in hMSCs. The binding of ZNF281 to the promoter region of ß-CATENIN was observed using a chromatin immunoprecipitation (ChIP) assay. In conclusion, we propose that ZNF281 plays an important role in the maintenance and osteogenic differentiation of stem cells via the transcriptional regulation of genes including ß-CATENIN.


Subject(s)
Osteocytes/physiology , Trans-Activators/deficiency , Animals , Cell Differentiation/physiology , Cell Growth Processes/physiology , Cord Blood Stem Cell Transplantation , Fetal Blood/cytology , Gene Knockdown Techniques , Humans , Mice , Mice, Nude , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Osteocytes/cytology , Osteocytes/metabolism , Osteogenesis/genetics , Repressor Proteins , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transfection
12.
PLoS One ; 7(9): e44693, 2012.
Article in English | MEDLINE | ID: mdl-23024756

ABSTRACT

Recent studies have shown that amniotic membrane tissue is a rich source of stem cells in humans. In clinical applications, the amniotic membrane tissue had therapeutic effects on wound healing and corneal surface reconstruction. Here, we successfully isolated and identified multipotent stem cells (MSCs) from canine amniotic membrane tissue. We cultured the canine amniotic membrane-derived multipotent stem cells (cAM-MSCs) in low glucose DMEM medium. cAM-MSCs have a fibroblast-like shape and adhere to tissue culture plastic. We characterized the immunophenotype of cAM-MSCs by flow cytometry and measured cell proliferation by the cumulative population doubling level (CPDL). We performed differentiation studies for the detection of trilineage multipotent ability, under the appropriate culture conditions. Taken together, our results show that cAM-MSCs could be a rich source of stem cells in dogs. Furthermore, cAM-MSCs may be useful as a cell therapy application for veterinary regenerative medicine.


Subject(s)
Amnion/cytology , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Adipogenesis , Animals , Biomarkers/metabolism , Cell Differentiation , Chondrogenesis , Dogs , Flow Cytometry/methods , Gene Expression Profiling , Immunophenotyping , Neurogenesis , Primary Cell Culture/methods
13.
Cell Transplant ; 21(7): 1493-502, 2012.
Article in English | MEDLINE | ID: mdl-22732242

ABSTRACT

Wharton's jelly is a known stem cell source in humans. Because stem cells might provide a potential therapeutic role in canines, many stem cell sources are studied for isolation and characterization in the canine system. So far, there have been no reports identifying canine Wharton's jelly stem cells. In this study, we successfully isolated and characterized mesenchymal stem cells (MSCs) from canine Wharton's jelly. Canine Wharton's jelly-derived mesenchymal stem cells (cWJ-MSCs) that were grown in low-glucose DMEM medium have spindle-like shapes similar to human Wharton's jelly stem cells. We characterized the immunophenotypes of canine Wharton's jelly stem cells by FACS analysis and measured the cumulative population doubling level (CPDL). We investigated the differentiation of cWJ-MSCs with a trilineage differentiation assay to determine whether they were mesenchymal. Under various differentiation conditions, cWJ-MSCs presented chondrogenic, osteogenic, adipogenic, and neurogenic differentiation abilities in vitro. In conclusion, our results show that cWJ-MSCs might be a good source for stem cells. Furthermore, cWJ-MSCs might be useful as a cell therapy application for veterinary medicine.


Subject(s)
Mesenchymal Stem Cells/cytology , Wharton Jelly/cytology , Adipogenesis , Animals , Cell Differentiation , Cell Separation , Cells, Cultured , Chondrogenesis , Dogs , Immunophenotyping , Mesenchymal Stem Cells/metabolism , Neurogenesis , Osteogenesis
14.
Cytotherapy ; 14(5): 630-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22404083

ABSTRACT

BACKGROUND AIMS: Niemann-Pick disease type C1 (NPC) is an autosomal recessive cholesterol-storage disorder characterized by liver dysfunction, hepatosplenomegaly and progressive neurodegeneration. Thus far, studies of NPC mice have been performed mainly to study the brain and neurodegeneration, because degeneration in the brain was known as the primary cause of death in NPC mice. However, NPC is a systemic disease; therefore the purpose of this study was to find the possibility of a general therapeutic effect by applying and tracking transplanted human amniotic epithelial stem cells (hAESC) in NPC mice. METHODS: hAESC were administered to NPC homozygous (NPC(-/-)) mice via intravenous injection from 5 weeks of age; each recipient received 5 × 10(5) cells every other week. The body weight of each of the mice was measured every week, and the survival and state of each mouse was evaluated every day. The weight of the organs was measured, and serum chemistry, histology and the intensity of Filipin staining were evaluated. RESULTS: The effect of cell transplantation was to extend the life span and reduce the rapid loss of weight. Moreover, alleviation of tissue damage was observed more in hAESC-treated NPC(-/-) mice than in non-treated NPC(-/-) mice. Cholesterol deposition was reduced after transplantation, and the relative weight of the liver was also decreased. CONCLUSIONS: These data show that hAESC could delay the degeneration caused by fatal genetic disorders such as NPC. This study presents the prospect of relief of precipitous disease progression and the therapeutic possibility of applying hAESC to fatal genetic disorders.


Subject(s)
Amnion/cytology , Epithelial Cells/cytology , Niemann-Pick Disease, Type C/therapy , Stem Cell Transplantation , Stem Cells/cytology , Amnion/metabolism , Animals , Cholesterol/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Injections, Intravenous , Liver Diseases/therapy , Mice , Mice, Inbred BALB C , Mice, Knockout , Niemann-Pick Disease, Type C/genetics , Niemann-Pick Disease, Type C/pathology , Renal Insufficiency/therapy , Stem Cells/metabolism
15.
Stem Cells ; 30(5): 876-87, 2012 May.
Article in English | MEDLINE | ID: mdl-22311737

ABSTRACT

CD49f (integrin subunit α6) regulates signaling pathways in a variety of cellular activities. However, the role of CD49f in regulating the differentiation and pluripotency of stem cells has not been fully investigated. Therefore, in this study, human mesenchymal stem cells (hMSCs) were induced to form spheres under nonadherent culture conditions, and we found that the CD49f-positive population was enriched in MSC spheres compared with MSCs in a monolayer. The expression of CD49f regulated the ability of hMSCs to form spheres and was associated with an activation of the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway. Furthermore, the forced expression of CD49f modulated the proliferation and differentiation potentials of hMSCs through prolonged activation of PI3K/AKT and suppressed the level of p53. We showed that the pluripotency factors OCT4 and SOX2 were recruited to the putative promoter region of CD49f, indicating that OCT4 and SOX2 play positive roles in the expression of CD49f. Indeed, CD49f expression was upregulated in human embryonic stem cells (hESCs) compared with hMSCs. The elevated level of CD49f expression was significantly decreased upon embryoid body formation in hESCs. In hESCs, the knockdown of CD49f downregulated PI3K/AKT signaling and upregulated the level of p53, inducing differentiation into three germ layers. Taken together, our data suggest that the cell-surface protein CD49f has novel and dynamic roles in regulating the differentiation potential of hMSCs and maintaining pluripotency.


Subject(s)
Embryonic Stem Cells/metabolism , Integrin alpha6/biosynthesis , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Octamer Transcription Factor-3/biosynthesis , SOXB1 Transcription Factors/biosynthesis , Cell Differentiation/physiology , Cells, Cultured , Embryonic Stem Cells/cytology , Gene Expression Regulation/physiology , Humans , Mesenchymal Stem Cells/cytology , Multipotent Stem Cells/cytology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism
16.
PLoS One ; 6(11): e28068, 2011.
Article in English | MEDLINE | ID: mdl-22132214

ABSTRACT

Metformin, a Type II diabetic treatment drug, which inhibits transcription of gluconeogenesis genes, has recently been shown to lower the risk of some diabetes-related tumors, including breast cancer. Recently, "cancer stem cells" have been demonstrated to sustain the growth of tumors and are resistant to therapy. To test the hypothesis that metformin might be reducing the risk to breast cancers, the human breast carcinoma cell line, MCF-7, grown in 3-dimensional mammospheres which represent human breast cancer stem cell population, were treated with various known and suspected breast cancer chemicals with and without non-cytotoxic concentrations of metformin. Using OCT4 expression as a marker for the cancer stem cells, the number and size were measured in these cells. Results demonstrated that TCDD (100 nM) and bisphenol A (10 µM) increased the number and size of the mammospheres, as did estrogen (10 nM E2). By monitoring a cancer stem cell marker, OCT4, the stimulation by these chemicals was correlated with the increased expression of OCT4. On the other hand, metformin at 1 and 10 mM concentration dramatically reduced the size and number of mammospheres. Results also demonstrated the metformin reduced the expression of OCT4 in E2 & TCDD mammospheres but not in the bisphenol A mammospheres, suggesting different mechanisms of action of the bisphenol A on human breast carcinoma cells. In addition, these results support the use of 3-dimensional human breast cancer stem cells as a means to screen for potential human breast tumor promoters and breast chemopreventive and chemotherapeutic agents.


Subject(s)
Breast Neoplasms/pathology , Metformin/pharmacology , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/metabolism , Receptors, Estrogen/antagonists & inhibitors , Antioxidants/pharmacology , Breast Neoplasms/genetics , Cell Proliferation/drug effects , Estradiol/pharmacology , Female , Flow Cytometry , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Octamer Transcription Factor-3/genetics , Phenolsulfonphthalein/metabolism , Promoter Regions, Genetic/genetics , Receptors, Estrogen/metabolism , Response Elements/genetics , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Tumor Cells, Cultured
17.
Cytotherapy ; 13(3): 341-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20860427

ABSTRACT

BACKGROUND AIMS: Amniotic fluid (AF) is a well-known source of stem cells. However, there have been no reports regarding equine AF stem cells. We have isolated equine AF-derived multipotent stem cells (MSC) (eAF-MSC) and show that these cells exhibit self-renewal ability and multilineage differentiation. METHODS: AF was obtained from thoroughbred mares and mononuclear cells (MNC) were isolated by Ficoll-Paque density gradient. We measured the cumulative population doubling level (CPDL) and characterized the immunophenotype by flow cytometry. To investigate differentiation ability, a trilineage differentiation assay was conducted. RESULTS: eAF-MSC could be isolated and the proliferation level was high. eAF-MSC presented typical MSC phenotypic markers, as determined by flow cytometry. Moreover, eAF-MSC showed a trilineage differentiation capability. CONCLUSIONS: Equine AF is a good source of MSC. Furthermore, eAF-MSC may be useful as a cell therapy application for horses.


Subject(s)
Amniotic Fluid/cytology , Cell Separation/methods , Multipotent Stem Cells/cytology , Adipogenesis , Animals , Cell Lineage , Cell Proliferation , Cells, Cultured , Chondrogenesis , Flow Cytometry , Horses , Humans , Immunophenotyping , Multipotent Stem Cells/metabolism , Osteogenesis , Sequence Homology, Amino Acid
18.
Exp Cell Res ; 317(7): 966-75, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21182834

ABSTRACT

The signaling network of protein kinase B(PKB)/Akt has been implicated in survival of lung cancer cells. However, understanding the relative contribution of the different isoform of Akt network is nontrival. Here, we report that Akt2 is highly expressed in human lung adenocarcinoma cell line A549 cells. Suppression of Akt2 expression in A549 cells results in notable inhibition of cell poliferation, soft agar growth, and invasion, accompanying by a decrease of nucleophosmin/B23 protein. Overexpression of Akt1 restores cancerous growth of A549 cells in B23-knockdown (KD) cells while Akt2 overexpression did not restore proliferating potential in cells with downregulated B23, thus suggesting Akt2 requires B23 to drive proliferation of lung cancer cell. Loss of functional Akt2 and B23 has similar defects on cell proliferation, apoptotic resistance and cell cycle regulation, while loss of Akt1 has less defects on cell proliferation, survival and cell cycle progression in A549 cells. Moreover, overexpression of B23 rescues the proliferative block induced as a consequence of loss of Akt2. Thus our data suggest that Akt2/B23 functions as an oncogenic unit to drive tumorigenesis of A549 lung cancer cells.


Subject(s)
Cell Transformation, Neoplastic , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Nuclear Proteins/metabolism , Oncogenes , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Cell Proliferation , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Nuclear Proteins/genetics , Nucleophosmin , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism
19.
Cell Mol Life Sci ; 68(2): 325-36, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20652617

ABSTRACT

Cellular senescence involves a reduction in adult stem cell self-renewal, and epigenetic regulation of gene expression is one of the main underlying mechanisms. Here, we observed that the cellular senescence of human umbilical cord blood-derived multipotent stem cells (hUCB-MSCs) caused by inhibition of histone deacetylase (HDAC) activity leads to down-regulation of high mobility group A2 (HMGA2) and, on the contrary, to up-regulation of p16(INK)4(A), p21(CIP)¹(/WAF)¹ and p27(KIP)¹. We found that let-7a1, let-7d, let-7f1, miR-23a, miR-26a and miR-30a were increased during replicative and HDAC inhibitor-mediated senescence of hUCB-MSCs by microRNA microarray and real-time quantitative PCR. Furthermore, the configurations of chromatins beading on these miRNAs were prone to transcriptional activation during HDAC inhibitor-mediated senescence. We confirmed that miR-23a, miR-26a and miR-30a inhibit HMGA2 to accelerate the progress of senescence. These findings suggest that HDACs may play important roles in cellular senescence by regulating the expression of miRNAs that target HMGA2 through histone modification.


Subject(s)
Adult Stem Cells/metabolism , Cellular Senescence/physiology , High Mobility Group Proteins , Histone Deacetylases , MicroRNAs/metabolism , Multipotent Stem Cells/metabolism , Adipose Tissue/cytology , Adult , Adult Stem Cells/cytology , Cell Proliferation , Enzyme Activation/drug effects , Epigenomics , Female , Fetal Blood/cytology , Fetal Blood/metabolism , Gene Expression Regulation/drug effects , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , High Mobility Group Proteins/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , MAP Kinase Signaling System/drug effects , MicroRNAs/genetics , MicroRNAs/pharmacology , Microarray Analysis , Multipotent Stem Cells/cytology , Polymerase Chain Reaction , Valproic Acid/pharmacology
20.
Cell Mol Life Sci ; 67(7): 1165-76, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20049504

ABSTRACT

Aging is linked to loss of the self-renewal capacity of adult stem cells. Here, we observed that human multipotent stem cells (MSCs) underwent cellular senescence in vitro. Decreased expression of histone deacetylases (HDACs), followed by downregulation of polycomb group genes (PcGs), such as BMI1, EZH2 and SUZ12, and by upregulation of jumonji domain containing 3 (JMJD3), was observed in senescent MSCs. Similarly, HDAC inhibitors induced cellular senescence through downregulation of PcGs and upregulation of JMJD3. Regulation of PcGs was associated with HDAC inhibitor-induced hypophosphorylation of RB, which causes RB to bind to and decrease the transcriptional activity of E2F. JMJD3 expression regulation was dependant on histone acetylation status at its promoter regions. A histone acetyltransferase (HAT) inhibitor prevented replicative senescence of MSCs. These results suggest that HDAC activity might be important for MSC self-renewal by balancing PcGs and JMJD3 expression, which govern cellular senescence by p16(INK4A) regulation.


Subject(s)
Adult Stem Cells/metabolism , Cellular Senescence , Histone Deacetylases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Multipotent Stem Cells/metabolism , Repressor Proteins/metabolism , Adult Stem Cells/cytology , Cells, Cultured , Down-Regulation , Epigenesis, Genetic , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Multipotent Stem Cells/cytology , Polycomb-Group Proteins , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Repressor Proteins/genetics , Up-Regulation , Valproic Acid/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...