1.Cancer stem-like cell-derived exosomes promotes the proliferation and invasion of human umbilical cord blood-derived mesenchymal stem cells.
Dan ZHANG ; Dawei HE ; Dian LI ; Bo TANG ; Dong HU ; Wenhao GUO ; Zhang WANG ; Lianju SHEN ; Guanghui WEI
Journal of Southern Medical University 2018;38(12):1440-1447
OBJECTIVE:
To investigate the effect of Piwil2-induced cancer stem-like cell (Piwil2-iCSC)-derived exosomes on the proliferation,migration and invasion of human umbilical cord blood-derived mesenchymal stem cells (hucMSCs).
METHODS:
Piwil2-iCSC-derived exosomes were isolated by ultracentrifugation and identified using transmission electron microscopy,nanoparticle tracking analysis and Western blotting.Exosome uptake assay was used to identify the pathway that Piwil2-iCSCderived exosomes utilized.HucMSCs were divided into control group,PBS intervention group and exosome intervention group,and CCK-8 assay,wound healing assay,Transwell assay,Western blotting and cell karyotype analysis were used to observe the proliferation,migration,invasion,expression levels of MMP2 and MMP9 proteins,and chromosome structure of hucMSCs.
RESULTS:
The diameter of Piwil2-iCSC-derived exosomes ranged from 50 nm to 100 nm,and most of them were oval or spherical capsules rich in CD9,CD63 and Piwil2 proteins.Exosomal uptake assay showed that the exosomes executed theirs functions after entering the cells.Compared with the control cells and PBS-treated cells,hucMSCs treated with the exosomes showed significantly increased number of proliferating cells (<0.05) with accelerated healing rate (<0.05 at 24 h;<0.01 at 48 h),increased invasive cells (<0.01),enhanced protein expressions of MMP2(<0.05 PBS group;<0.01 control group) and MMP9(<0.05),but their karyotype still remained 46XY without any abnormalities.
CONCLUSIONS
Piwil2-iCSC-derived exosomes can promote the proliferation,migration and invasion but does not cause cancer-like heterogeneity changes in hucMSCs.
Argonaute Proteins
;
Cell Movement
;
physiology
;
Cell Proliferation
;
physiology
;
Exosomes
;
physiology
;
Fetal Blood
;
cytology
;
Humans
;
Karyotyping
;
Mesenchymal Stem Cells
;
pathology
;
Neoplasm Invasiveness
;
Neoplastic Stem Cells
;
Umbilical Cord
;
Wound Healing
2.Effect of AB serum on human terminal erythroid differentiation ex vivo.
Jiling LIAO ; Jieying ZHANG ; Xu HAN ; Qikang HU ; Minyuan PENG ; Kunlu WU ; Jing LIU
Journal of Central South University(Medical Sciences) 2016;41(12):1245-1251
To demonstrate the effect of AB serum on terminal erythroid differentiation ex vivo.
Methods: After separation of CD34+ cells from cord blood, the cells were cultured and divided into a control group and an experimental group. The effects of AB serum were examined by the expressions of different markers (GPA, Band3 and α4-integrin) for erythroblast differentiation and enucleation by flow cytometry.
Results: The CD34+ cells were successfully differentiated to enucleated red blood cells. There were evident differences among the expressions of GPA, Band3 and α4-integrin between the 2 groups. The percentage of GPA positive cells in the experimental group was bigger than that in the control group in every time point. The expression of Band3 in the experimental group was higher than that in the control group. The expression of α4-integrin in the experimental group was lower than that in the control group. In addition, the enucleation rate in the experimental group was higher than that in the control group.
Conclusion: AB serum can promote the cell differentiation and enucleation during terminal erythroid differentiation in vitro.
ABO Blood-Group System
;
blood
;
physiology
;
Anion Exchange Protein 1, Erythrocyte
;
metabolism
;
Antigens, CD34
;
blood
;
Cell Differentiation
;
genetics
;
physiology
;
Cell Nucleus
;
Cells, Cultured
;
Erythrocytes
;
physiology
;
ultrastructure
;
Erythropoiesis
;
genetics
;
physiology
;
Fetal Blood
;
cytology
;
physiology
;
Flow Cytometry
;
Glycophorins
;
metabolism
;
Humans
;
Integrin alpha4beta1
;
metabolism
3.Transplantation of human umbilical cord-derived endothelial progenitor cells promotes re-endothelialization of the injured carotid artery after balloon injury in New Zealand white rabbits.
Cheng-heng HU ; Xiao KE ; Kui CHEN ; Da-ya YANG ; Zhi-min DU ; Gui-fu WU
Chinese Medical Journal 2013;126(8):1480-1485
BACKGROUNDCell transplantation has great potential for promoting endothelial repair and reducing the complications of percutaneous coronary intervention (PCI). The aim of this study was to investigate the effect of transplantation of human umbilical cord blood endothelial progenitor cells (EPCs) on injured arteries.
METHODSUmbilical cord blood mononuclear cells were obtained from post-partum lying-in women, and EPCs were isolated, cultured, expanded and identified by immunofluorescence. The carotid arterial endothelium of New Zealand white rabbits was injured by dilatation with a 3F balloon, and the EPCs were injected into the lumen of the injured artery in the transplanted group (n = 16), while an equal volume of phosphated buffered saline (PBS) was injected into the control group after balloon injury (n = 16). The animals were sacrificed after either 2 or 4 weeks, and the grafted cells were identified by double immunofluorescence staining with human nuclear antigen (HNA) and CD31 antibodies. Arterial cross sections were analyzed by pathology, immunohistochemistry and morphometry to evaluate the reparative effects of EPCs. Proliferating cell nuclear antigen (PCNA) and transforming growth factor (TGF)-β1 mRNA expression were detected by reverse transcription-polymerase chain reaction (RT-PCR).
RESULTSFluorescence-labeled EPCs were found in the neointima. The neointimal area and the neointimal/medial area ratio were significantly lower in the transplanted group than in the control group (P < 0.05). von Willebrand factor (vWF) immunohistostaining showed more VWF-positive cells in the transplanted animals than in the controls (8.75 ± 2.92 vs. 4.50 ± 1.77, P < 0.05). Compared with the control group, the transplanted group had lower expression of PCNA mRNA (0.67 ± 0.11 vs. 1.25 ± 0.40, P < 0.01) and higher expression of TGF-β1 mRNA (1.10 ± 0.21 vs. 0.82 ± 0.07, P < 0.05).
CONCLUSIONSEPCs derived from human umbilical cord blood were successfully transplanted into injured vessels. The transplanted EPCs inhibited neointimal hyperplasia and promoted vascular re-endothelialization.
Animals ; Carotid Artery Injuries ; immunology ; pathology ; therapy ; Cell Differentiation ; Cells, Cultured ; Cytokines ; genetics ; Endothelial Cells ; cytology ; physiology ; Fetal Blood ; cytology ; Humans ; Hyperplasia ; Male ; Neointima ; pathology ; Proliferating Cell Nuclear Antigen ; genetics ; RNA, Messenger ; analysis ; Rabbits ; Stem Cell Transplantation ; Transforming Growth Factor beta1 ; genetics
4.Comparing the osteogenic potential of canine mesenchymal stem cells derived from adipose tissues, bone marrow, umbilical cord blood, and Wharton's jelly for treating bone defects.
Byung Jae KANG ; Hak Hyun RYU ; Sung Su PARK ; Yoshihisa KOYAMA ; Masanori KIKUCHI ; Heung Myong WOO ; Wan Hee KIM ; Oh Kyeong KWEON
Journal of Veterinary Science 2012;13(3):299-310
Alternative sources of mesenchymal stem cells (MSCs) for replacing bone marrow (BM) have been extensively investigated in the field of bone tissue engineering. The purpose of this study was to compare the osteogenic potential of canine MSCs derived from adipose tissue (AT), BM, umbilical cord blood (UCB), and Wharton's jelly (WJ) using in vitro culture techniques and in vivo orthotopic implantation assays. After canine MSCs were isolated from various tissues, the proliferation and osteogenic potential along with vascular endothelial growth factor (VEGF) production were measured and compared in vitro. For the in vivo assay, MSCs derived from each type of tissue were mixed with beta-tricalcium phosphate and implanted into segmental bone defects in dogs. Among the different types of MSCs, AT-MSCs had a higher proliferation potential and BM-MSCs produced the most VEGF. AT-MSCs and UCB-MSCs showed greater in vitro osteogenic potential compared to the other cells. Radiographic and histological analyses showed that all tested MSCs had similar osteogenic capacities, and the level of new bone formation was much higher with implants containing MSCs than cell-free implants. These results indicate that AT-MSCs, UCB-MSCs, and WJ-MSCs can potentially be used in place of BM-MSCs for clinical bone engineering procedures.
Adipocytes, White/cytology/physiology
;
Alkaline Phosphatase/metabolism
;
Animals
;
Biocompatible Materials/metabolism/*therapeutic use
;
Bone Diseases/*therapy
;
Bone Marrow Cells/cytology/physiology
;
Calcification, Physiologic
;
Calcium/metabolism
;
Calcium Phosphates/metabolism/therapeutic use
;
Cell Proliferation
;
Dogs
;
Female
;
Fetal Blood/cytology/physiology
;
Flow Cytometry
;
Male
;
Mesenchymal Stromal Cells/cytology/*metabolism
;
*Osteogenesis
;
Polyesters/metabolism/therapeutic use
;
Tissue Engineering/*methods
;
Vascular Endothelial Growth Factor A/metabolism
5.Potential application of pooled human umbilical plasma in the culture of human umbilical vein endothelial cells.
Journal of Biomedical Engineering 2011;28(1):115-120
This study was aimed to look for the potential application of human umbilical plasma (HUP) in the culture of human umbilical vein endothelial cells (HUVECs). The effect of HUP concentration in cell culture medium on cell proliferation activity and cell cycle was studied. HUVECs were obtained by digesting the umbilical cord with 0.25% trypsin mixed with the equal volume of 0.1% collagenase II, then were identified by morphology and factor VIII immunohistochemistry under phase contrast microscopy. The cells cultured after 7 days showed the typical cobblestone morphology with factor VIII immunohistochemical staining positive. The study showed that the groups of 20% (HUP), 15% HUP+20% Fetal Bovine Serum (FBS) and 20% HUP+20%FBS enhanced cell proliferation activity significantly when compared with the control group (20% FBS without HUP). On the contrary, 30% HUP+20% FBS caused cell cycle arrest, which significantly hindered the proliferation of HUVECs. The study proved that although HUP might not be able to completely replace the role of vascular endothelial growth factor in cell culture, as a supplement ingredient, it was an ideal candidate to replace FBS in culture medium.
Cell Culture Techniques
;
methods
;
Cell Proliferation
;
Cells, Cultured
;
Culture Media
;
chemistry
;
Fetal Blood
;
chemistry
;
Human Umbilical Vein Endothelial Cells
;
cytology
;
Humans
;
Plasma
;
physiology
6.Notch signaling: a novel regulating differentiation mechanism of human umbilical cord blood-derived mesenchymal stem cells into insulin-producing cells in vitro.
Yan-hua HU ; De-quan WU ; Feng GAO ; Guo-dong LI ; Xin-chen ZHANG
Chinese Medical Journal 2010;123(5):606-614
BACKGROUNDHuman umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) could be induced to differentiate into insulin producing cells (IPCs) in vitro, which have good application potential in the cell replacement treatment of type-1 diabetes. However, the mechanisms regulating this differentiation have remained largely unknown. Notch signaling is critical in cell differentiation. This study investigated whether Notch signaling could regulate the IPCs differentiation of human UCB-MSCs.
METHODSUsing an interfering Notch signaling protocol in vitro, we studied the role of Notch signaling in differentiation of human UCB-MSCs into IPCs. In a control group the induction took place without interfering Notch signaling.
RESULTSHuman UCB-MSCs expressed the genes of Notch receptors (Notch 1 and Notch 2) and ligands (Jagged 1 and Deltalike 1). Human UCB-MSCs with over-expressing Notch signaling in differentiation resulted in the down-regulation of insulin gene level, proinsulin protein expression, and insulin-positive cells percentage compared with the control group. These results showed that over-expressing Notch signaling inhibited IPCs differentiation. Conversely, when Notch signaling was attenuated by receptor inhibitor, the induced cells increased on average by 3.06-fold (n = 4, P < 0.001) in insulin gene level, 2.60-fold (n = 3, P < 0.02) in proinsulin protein expression, and 1.62-fold (n = 6, P < 0.001) in the rate of IPCs compared with the control group. Notch signaling inhibition significantly promoted IPCs differentiation with about 40% of human UCB-MSCs that converted to IPCs, but these IPCs were not responsive to glucose challenge very well both in vitro and in vivo. Hence, further research has to be carried out in the future.
CONCLUSIONSNotch signaling may be an important mechanism regulating IPCs differentiation of human UCB-MSCs in vitro and Notch signaling inhibition may be an efficient way to increase the number of IPCs, which may resolve the shortage of islet of cell replacement treatment of type-1 diabetes.
Animals ; Cell Differentiation ; Fetal Blood ; cytology ; Humans ; Insulin ; biosynthesis ; Male ; Mesenchymal Stromal Cells ; cytology ; Mice ; Mice, Inbred BALB C ; Receptors, Notch ; physiology ; Signal Transduction ; physiology
7.Isolation and characterization of canine umbilical cord blood-derived mesenchymal stem cells.
Min Soo SEO ; Yun Hyeok JEONG ; Jeung Ran PARK ; Sang Bum PARK ; Kyoung Hwan RHO ; Hyung Sik KIM ; Kyung Rok YU ; Seung Hee LEE ; Ji Won JUNG ; Yong Soon LEE ; Kyung Sun KANG
Journal of Veterinary Science 2009;10(3):181-187
Human umbilical cord blood-derived mesenchymal stem cells (MSCs) are known to possess the potential for multiple differentiations abilities in vitro and in vivo. In canine system, studying stem cell therapy is important, but so far, stem cells from canine were not identified and characterized. In this study, we successfully isolated and characterized MSCs from the canine umbilical cord and its fetal blood. Canine MSCs (cMSCs) were grown in medium containing low glucose DMEM with 20% FBS. The cMSCs have stem cells expression patterns which are concerned with MSCs surface markers by fluorescence-activated cell sorter analysis. The cMSCs had multipotent abilities. In the neuronal differentiation study, the cMSCs expressed the neuronal markers glial fibrillary acidic protein (GFAP), neuronal class III beta tubulin (Tuj-1), neurofilament M (NF160) in the basal culture media. After neuronal differentiation, the cMSCs expressed the neuronal markers Nestin, GFAP, Tuj-1, microtubule-associated protein 2, NF160. In the osteogenic & chondrogenic differentiation studies, cMSCs were stained with alizarin red and toluidine blue staining, respectively. With osteogenic differentiation, the cMSCs presented osteoblastic differentiation genes by RT-PCR. This finding also suggests that cMSCs might have the ability to differentiate multipotentially. It was concluded that isolated MSCs from canine cord blood have multipotential differentiation abilities. Therefore, it is suggested that cMSCs may represent a be a good model system for stem cell biology and could be useful as a therapeutic modality for canine incurable or intractable diseases, including spinal cord injuries in future regenerative medicine studies.
Animals
;
*Cell Differentiation
;
Chondrogenesis
;
Dogs/blood/*physiology
;
Fetal Blood/*cytology
;
Mesenchymal Stem Cells/*cytology
;
Neurons/cytology
;
Osteogenesis
8.Role of sphingosine 1-phosphate receptor signaling in hematopoietic stem/progenitor cell transmigration.
Wen-chao OU ; Shi-ming LIU ; Long-geng XIONG ; Guo-qing LI ; Meng-qun TAN
Journal of Southern Medical University 2009;29(9):1862-1865
OBJECTIVETo determine the role of sphingosine 1-phosphate receptor (S1PRs ) signaling in CD34+ hematopoietic stem/progenitor cell transmigration.
METHODSCD34(+) cells were separated by Ficoll density gradient centrifugation and incubated in DMEM medium with 10% fetal calf serum. The cells were pretreated by FTY720, with or without pertussis toxin (PTX) and antiCXCR4 mAb in the medium, followed by addition of 100 ng/ml SDF-1 into the lower chamber of a Costar 24-well transwell. The migrated cells were counted using FACS and the migrating rates were determined. The expressions of sphingosine 1-phosphate receptors were analyzed in CD34(+) cells before and after the transmigration by reverse transcriptase- polymerase chain reaction (RT-PCR). Cord blood CD34(+) cells were treated with or without FTY720 (10(+) mol/L), and the expressions of CD49d (VLA-4), CD11a (LFA-1), and CD62L (L-selectin) were analyzed at 1, 8, and 16 h after the treatment.
RESULTSWhile FTY720 did not affect spontaneous migration, a substantial increase of SDF-1-induced transmigration was observed in the presence of FTY720 (15.26 2.14 to 28.64 2.37). The FTY720-enhanced transmigration was completely blocked by addition of PTX or antiCXCR4 mAb. S1p1-5 was expressed in fresh isolated cord blood CD34(+) cells. The migrating cells stimulated by FTY720 and SDF-1 only expressed S1P1, S1P3, and S1P4. The expressions of CD49d, CD11a and CD62L on CD34(+) cells treated with FTY720 remained unchanged at the selected time points as compared with the control.
CONCLUSIONSS1PRs are involved the transmigration of CD34(+) cells. The activation of S1PRs results in increased chemotactic response of CD34(+) to SDF-1. These effects are mediated through CXCR4 and PTX-sensitive Gi proteins. Only the CD34(+) cells expressing the specific receptors can rapidly transmigrate. The activation of the S1PRs does not affect the expressions of the adhesion molecules on cord blood CD34(+) cells.
Antigens, CD34 ; metabolism ; Cell Movement ; Cells, Cultured ; Chemokine CXCL12 ; pharmacology ; Fetal Blood ; cytology ; Fingolimod Hydrochloride ; Hematopoietic Stem Cell Mobilization ; Hematopoietic Stem Cells ; cytology ; drug effects ; Humans ; Propylene Glycols ; pharmacology ; Receptors, Lysosphingolipid ; metabolism ; physiology ; Signal Transduction ; Sphingosine ; analogs & derivatives ; pharmacology
9.Implantation of canine umbilical cord blood-derived mesenchymal stem cells mixed with beta-tricalcium phosphate enhances osteogenesis in bone defect model dogs.
Byung Jun JANG ; Ye Eun BYEON ; Ji Hey LIM ; Hak Hyun RYU ; Wan Hee KIM ; Yoshihisa KOYAMA ; Masanori KIKUCHI ; Kyung Sun KANG ; Oh Kyeong KWEON
Journal of Veterinary Science 2008;9(4):387-393
This study was performed to evaluate the osteogenic effect of allogenic canine umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) mixed with beta-tricalcium phosphate (beta-TCP) in orthotopic implantation. Seven hundred milligrams of beta-TCP mixed with 1 x 10(6) UCB-MSCs diluted with 0.5 ml of saline (group CM) and mixed with the same volume of saline as control (group C) were implanted into a 1.5 cm diaphyseal defect and wrapped with PLGC membrane in the radius of Beagle dogs. Radiographs of the antebrachium were made after surgery. The implants were harvested 12 weeks after implantation and specimens were stained with H&E, toluidine blue and Villanueva-Goldner stains for histological examination and histomorphometric analysis of new bone formation. Additionally, UCB-MSCs were applied to a dog with non-union fracture. Radiographically, continuity between implant and host bone was evident at only one of six interfaces in group C by 12 weeks, but in three of six interfaces in group CM. Radiolucency was found only near the bone end in group C at 12 weeks after implantation, but in the entire graft in group CM. Histologically, bone formation was observed around beta-TCP in longitudinal sections of implant in both groups. Histomorphometric analysis revealed significantly increased new bone formation in group CM at 12 weeks after implantation (p < 0.05). When applied to the non-union fracture, fracture healing was identified by 6 weeks after injection of UCB-MSCs. The present study indicates that a mixture of UCB-MSCs and beta-TCP is a promising osteogenic material for repairing bone defects.
Animals
;
Biocompatible Materials/metabolism/therapeutic use
;
Bone Substitutes/*therapeutic use
;
Calcium Phosphates/*therapeutic use
;
Dogs
;
Fetal Blood/*cytology
;
Fracture Fixation/methods/veterinary
;
Mesenchymal Stem Cells/*physiology
;
Osteogenesis/*physiology
;
Tissue Engineering/methods
;
Wound Healing/physiology
10.Vasculogenic potential of endothelial progenitor cells derived from human umbilical cord blood and their roles in neovascularization of malignant glioma.
Hua-Rong ZHANG ; Fei-Lan CHEN ; Cheng-Ping XU ; Xiu-Wu BIAN
Chinese Journal of Pathology 2008;37(1):45-50
OBJECTIVETo investigate vasculogenic potential of endothelial progenitor cells (EPCs) derived from human umbilical cord blood and their contribution to the neovascularization of malignant glioma in vivo.
METHODSEPCs were isolated from human umbilical cord blood by density gradient centrifugation. After 7-10 days of culture, EPCs were investigated for CD34 and VEGFR-2 expression by direct immunofluoresent staining. The proliferative activity, migratory capability and forming capillary-like tubules were also monitored after stimulation with VEGF(50 mg/L) in vitro. Moreover, EPCs were administered into tumor-bearing mice, and the tumor and mouse organs were examined under confocal laser scanning microscope to visualize the distribution and localization of transplanted EPCs. In order to quantity the incorporation of EPCs into tumor vessels, cryosections of the tumor tissue were double-labelled with antihuman CD31 and anti-mouse CD31.
RESULTSAfter 7 to 10 days of culture, EPCs assumed cobblestone-like monolayer growth pattern with nearly complete confluence, and expressed CD34 and VEGFR-2. Significant proliferative activity, increased migratory capability and forming capillary-like tubules were observed when stimulated with VEGF. The transplanted EPCs in vivo specifically homed to solid tumor tissue and incorporated into the tumor's endothelium. Quantitative analysis revealed that human EPCs contributed significantly to tumor neovascularization by incorporation into tumor vasculature (18.68 +/- 1.32)% of the total vessels.
CONCLUSIONEPCs possess the potential to form neovascular network in tumor and play a role in the phenotypical heterogeneity of tumor microvascular architecture.
Animals ; Antigens, CD34 ; immunology ; Endothelial Cells ; pathology ; physiology ; Endothelium, Vascular ; pathology ; physiopathology ; Fetal Blood ; cytology ; Glioma ; complications ; pathology ; Humans ; Mice ; Neovascularization, Pathologic ; etiology ; pathology ; physiopathology ; Platelet Endothelial Cell Adhesion Molecule-1 ; immunology ; Stem Cells ; pathology ; physiology ; Vascular Endothelial Growth Factor Receptor-2 ; immunology

Result Analysis
Print
Save
E-mail