Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Publication year range
1.
J Cell Physiol ; 231(5): 1065-76, 2016 May.
Article in English | MEDLINE | ID: mdl-26395760

ABSTRACT

Generation of fully functional hematopoietic multipotent progenitor (MPP) cells from human pluripotent stem cells (hPSCs) has a great therapeutic potential to provide an unlimited cell source for treatment of hematological disorders. We previously demonstrated that CD34(+) CD31(+) CD144(+) population derived from hPSCs contain hemato-endothelial progenitors (HEPs) that give rise to hematopoietic and endothelial cells. Here, we report a differentiation system to generate definitive hematopoietic MPP cells from HEPs via endothelial monolayer. In the presence of angiogenic factors, HEPs formed an endothelial monolayer, from which hematopoietic clusters emerged through the process of endothelial-to-hematopoietic transition (EHT). EHT was significantly enhanced by hematopoietic growth factors. The definitive MPP cells generated from endothelial monolayer were capable of forming multilineage hematopoietic colonies, giving rise to T lymphoid cells, and differentiating into enucleated erythrocytes. Emergence of hematopoietic cells from endothelial monolayer occurred transiently. Hematopoietic potential was lost during prolonged culture of HEPs in endothelial growth conditions. Our study demonstrated that CD34(+) CD31(+) CD144(+) HEPs gave rise to hematopoietic MPP cells via hemogenic endothelial cells that exist transiently. The established differentiation system provides a platform for future investigation of regulatory factors involved in de novo generation of hematopoietic MPP cells and their applications in transplantation.


Subject(s)
Endothelial Cells/cytology , Hematopoiesis , Multipotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Antigens, CD/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , Endothelial Cells/drug effects , Erythroid Cells/cytology , Erythroid Cells/drug effects , Hematopoiesis/drug effects , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Multipotent Stem Cells/drug effects , Pluripotent Stem Cells/drug effects
2.
J Cell Biochem ; 116(12): 2735-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26012423

ABSTRACT

Patient-specific human induced-pluripotent stem cells (hiPSCs) represent important cell sources to treat patients with acquired blood disorders. To realize the therapeutic potential of hiPSCs, it is crucial to understand signals that direct hiPSC differentiation to a hematopoietic lineage fate. Our previous study demonstrated that CD34(+)CD31(+) cells derived from human pluripotent stem cells (hPSCs) contain hemato-endothelial progenitors (HEPs) that give rise to hematopoietic cells and endothelial cells. Here, we established a serum-free and feeder-free system to induce the differentiation of hPSC-derived CD34(+)CD31(+) progenitor cells to erythroid cells. We show that extracellular matrix (ECM) proteins promote the differentiation of CD34(+)CD31(+) progenitor cells into CD235a(+) erythroid cells through CD41(+)CD235a(+) megakaryocyte-erythroid progenitors (MEP). Erythropoietin (EPO) is a predominant factor for CD34(+)CD31(+) progenitor differentiation to erythroid cells, whereas transforming growth factor beta (TGF-ß) inhibits the development of CD34(+)CD31(+) progenitor cells. Apoptosis of progenitor cells is induced by TGF-ß in early erythroid differentiation. Suppression of TGF-ß signaling by SB431542 at early stage of CD34(+)CD31(+) progenitor differentiation induces the erythroid cell generation. Together, these findings suggest that TGF-ß suppression and EPO stimulation promote erythropoiesis of CD34(+)CD31(+) progenitor cells derived from hPSCs.


Subject(s)
Erythroid Cells/drug effects , Erythropoiesis/drug effects , Erythropoietin/antagonists & inhibitors , Pluripotent Stem Cells/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Antigens, CD34/metabolism , Benzamides/administration & dosage , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Dioxoles/administration & dosage , Erythroid Cells/cytology , Erythropoietin/genetics , Hematopoietic Stem Cells/cytology , Humans , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/transplantation , Transforming Growth Factor beta/genetics
3.
Stem Cells Dev ; 22(20): 2765-76, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23758278

ABSTRACT

Derived from mesoderm precursors, hemangioblasts are bipotential common progenitors of hematopoietic cells and endothelial cells. The regulatory events controlling hematopoietic and endothelial lineage specification are largely unknown, especially in humans. In this study, we establish a serum-free and feeder-free system with a high-efficient embryoid body (EB) generation to investigate the signals that direct differentiation of human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs). Consistent with previous studies, the CD34(+)CD31(+)VE-cadherin(+) (VEC(+)) cells derived from hPSCs contain hematopoietic and endothelial progenitors. In the presence of hematopoietic and endothelial growth factors, some of CD34(+)CD31(+)VEC(+) cells give rise to blast colony-forming cells (BL-CFCs), which have been used to characterize bipotential hemangioblasts. We found that the level of the transforming growth factor beta (TGF-ß) 1 protein is increased during hPSC differentiation, and that TGF-ß signaling has the double-edged effect on hematopoietic and endothelial lineage differentiation in hPSCs. An addition of TGF-ß to hPSC differentiation before mesoderm induction promotes the development of mesoderm and the generation of CD34(+)CD31(+)VEC(+) cells. An addition of TGF-ß inhibitor, SB431542, before mesoderm induction downregulates the expression of mesodermal markers and reduces the number of CD34(+)CD31(+)VEC(+) progenitor cells. However, inhibition of TGF-ß signaling after mesoderm induction increases CD34(+)CD31(+)VEC(+) progenitors and BL-CFCs. These data provide evidence that a balance of positive and negative effects of TGF-ß signaling at the appropriate timing is critical, and potential means to improve hematopoiesis and vasculogenesis from hPSCs.


Subject(s)
Embryoid Bodies/metabolism , Endothelial Cells/metabolism , Hematopoietic Stem Cells/metabolism , Induced Pluripotent Stem Cells/metabolism , Transforming Growth Factor beta1/metabolism , Benzamides/pharmacology , Cell Differentiation , Cell Lineage/drug effects , Cell Proliferation , Coculture Techniques , Dioxoles/pharmacology , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryonic Development/genetics , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Lymphocyte Activation , Mesoderm/cytology , Mesoderm/metabolism , Primary Cell Culture , Signal Transduction , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology
4.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 28(5): 524-8, 2012 May.
Article in Chinese | MEDLINE | ID: mdl-22558992

ABSTRACT

AIM: To prepare functional monoclonal antibodies(mAb)against recombinant human Flt-1(rhFlt-1). METHODS: A cell line stable secreting mAb was established by using FLT-1 extracellular domain III as antigen and hybridoma technique. Then it was purified in large-scale from mouse ascites by protein G affinity chromatography. The characteristics of mAb were then determined by ELISA, Western blotting and FACS. RESULTS: The immunoglobin subtype of mAb XA12 was IgG1 with kappa (κ) light chains, and it could recognize rhFlt-1 specifically. Furthermore, mAb XA12 could bind to rhFlt-1with high affinity (K=1.28±0.05 nmol/L). It could also be used to detect Flt-1-positive cells, such as human umbilical vascular endothelial cells (HUVECs) and K562/A02 in a dose-dependent fashion. CONCLUSION: A hybridoma cell line secreting functional anti-rhFlt-1 mAb was successfully prepared. The antibody can be used to study the function of Flt-1 and further potentially optimized for clinical purpose.


Subject(s)
Antibodies, Monoclonal/immunology , Vascular Endothelial Growth Factor Receptor-1/immunology , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hybridomas/immunology , K562 Cells , Mice , Mice, Inbred BALB C , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-1/analysis
5.
PLoS One ; 7(3): e32782, 2012.
Article in English | MEDLINE | ID: mdl-22403708

ABSTRACT

Multidrug resistance mediated by P-glycoprotein in cancer cells has been a major issue that cripples the efficacy of chemotherapy agents. Aimed for improved efficacy against resistant cancer cells, we designed and synthesized 25 oxindole derivatives based on indirubin by structure-activity relationship analysis. The most potent one was named PH II-7, which was effective against 18 cancer cell lines and 5 resistant cell lines in MTT assay. It also significantly inhibited the resistant xenograft tumor growth in mouse model. In cell cycle assay and apoptosis assay conducted with flow cytometry, PH II-7 induced S phase cell cycle arrest and apoptosis even in resistant cells. Consistently revealed by real-time PCR, it modulates the expression of genes related to the cell cycle and apoptosis in these cells, which may contributes to its efficacy against them. By side-chain modification and FITC-labeling of PH II-7, we were able to show with confocal microscopy that not only it was not pumped by P-glycoprotein, it also attenuated the efflux of Adriamycin by P-glycoprotein in MDR tumor cells. Real-time PCR and western blot analysis showed that PH II-7 down-regulated MDR1 gene via protein kinase C alpha (PKCA) pathway, with c-FOS and c-JUN as possible mediators. Taken together, PH II-7 is a dual-functional compound that features both the cytotoxicity against cancer cells and the inhibitory effect on P-gp mediated drug efflux.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Drug Design , Drug Resistance, Multiple/drug effects , Indoles/chemical synthesis , Indoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Doxorubicin/metabolism , Female , Humans , Indoles/chemistry , Intracellular Space/drug effects , Intracellular Space/metabolism , Mice , Molecular Imaging , Oxindoles , Protein Kinase C-alpha/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship , Transcriptome/drug effects , Xenograft Model Antitumor Assays
6.
Stem Cell Res ; 8(1): 26-37, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22099018

ABSTRACT

Robust expansion and genetic manipulation of human embryonic stem cells (hESCs) and induced-pluripotent stem (iPS) cells are limited by poor cell survival after enzymatic dissociation into single cells. Although inhibition of apoptosis is implicated for the single-cell survival of hESCs, the protective role of attenuation of apoptosis in hESC survival has not been elucidated. Bcl-xL is one of several anti-apoptotic proteins, which are members of the Bcl-2 family of proteins. Using an inducible system, we ectopically expressed Bcl-xL gene in hESCs, and found a significant increase of hESC colonies in the single-cell suspension cultures. Overexpression of Bcl-xL in hESCs decreased apoptotic caspase-3(+) cells, suggesting attenuation of apoptosis in hESCs. Without altering the kinetics of pluripotent gene expression, the efficiency to generate embryoid bodies (EBs) in vitro and the formation of teratoma in vivo were significantly increased in Bcl-xL-overexpressing hESCs after single-cell dissociation. Interestingly, the number and size of hESC colonies from cluster cultures were not affected by Bcl-xL overexpression. Several genes of extracellular matrix and adhesion molecules were upregulated by Bcl-xL in hESCs without single-cell dissociation, suggesting that Bcl-xL regulates adhesion molecular expression independent of cell dissociation. In addition, the gene expressions of FAS and several TNF signaling mediators were downregulated by Bcl-xL. These data support a model in which Bcl-xL promotes cell survival and increases cloning efficiency of dissociated hESCs without altering hESC self-renewal by i) attenuation of apoptosis, and ii) upregulation of adhesion molecules to facilitate cell-cell or cell-matrix interactions.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , bcl-X Protein/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cell Survival , Clone Cells , Down-Regulation , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Humans , Mice , Mice, Nude , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction , Teratoma/pathology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
7.
Eur J Pharmacol ; 669(1-3): 38-44, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21871878

ABSTRACT

Multidrug resistance (MDR) is a major impediment to the effective chemotherapy of many human malignancies, and novel MDR reversal agents are desirable for combination therapy to reduce MDR, enhance anti-tumor activity and reduce side effects. Overexpression of P-glycoprotein (P-gp) is the most prevalent cause of MDR in cancer tissues, and resistance to apoptosis is a common characteristic for the multidrug resistant cancer cells. Our group has synthesized a novel potent anti-tumor indirubin derivative, PHII-7. In this study, MCF-7/ADR cells, an adriamycin (ADR)-selected human breast tumor cell line with the MDR phenotype, were used to investigate the anticancer properties of this novel indirubin derivative. Cytotoxicity and apoptosis assays showed that PHII-7 significantly inhibited cell growth, induced apoptosis, potentiated ADR cytotoxicity and restored chemotherapy sensitivity in the MDR cancer cells. Further studies indicated that by down-regulation of P-gp expression, PHII-7 partially inhibited P-gp efflux pump function and increased intracellular accumulation of Rhodamine 123, a P-gp substrate. These results provide a biochemical basis for possible clinical application of PHII-7 alone or in combination with conventional antineoplastic agents in the treatment MDR tumors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Indoles/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans
8.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 18(2): 441-4, 2010 Apr.
Article in Chinese | MEDLINE | ID: mdl-20416184

ABSTRACT

This study was aimed to investigate the growth and multiple differentiation potential of human umbilical cord tissue derived mesenchymal stem cells (UC-MSCs) transfected by a retroviral vector with catalase (CAT) gene. The UC-MSCs cultured in vitro were transfected by using pMSCV carrying GFP (pMSCV-GFP) and pMSCV carrying CAT (pMSCV-GFP-CAT) respectively, then the MSC-GFP cell line and MSC-GFP-CAT cell line were obtained by sorting of flow cytometry. The GFP expression was observed by a fluorescent microscopy at 48 hours after CAT gene transfection. The GFP+ cells were sorted by flow cytometry. The activity of CAT in GFP+ cells was detected by catalase assay kit. The proliferative capacity of transfected UC-MSCs was determined by cell counting kit-8. The differentiation ability of gene-transfected GFP+ cells into osteogenesis and adipogenesis was observed by von Kossa and oil red O staining. The results indicated that green fluorescence in UC-MSCs was observed at 48 hours after transfection, and the fluorescence gradually enhanced to a steady level on day 3. The percentage of MSCs-GFP was (25.54+/-8.65)%, while the percentage of MSCs-GFP-CAT was (35.4+/-18.57)%. The activity of catalase in UC-MSCs, MSCs-GFP, MSCs-GFP-CAT cells were 19.5, 20.3, 67.2 U, respectively. The transfected MSCs-GFP-CAT could be induced into osteoblasts and adipocytes. After 21 days, von Kossa staining showed induced osteoblasts. Many lipid droplets with high refractivity occurred in cytoplasm of the transfected UC-MSCs, and showed red fat granules in oil red O staining cells. There were no significant differences between transfected and non-transfected UC-MSCs cells (p>0.05). It is concluded that UC-MSCs are successfully transfected by retrovirus carrying GFP or CAT gene, the activity of catalase increased by 3.4-fold. The transfected UC-MSCs maintain proliferation potential and ability of differentiation into osteoblasts and adipocytes.


Subject(s)
Catalase/genetics , Mesenchymal Stem Cells/metabolism , Retroviridae/genetics , Catalase/metabolism , Cell Differentiation , Cells, Cultured , Flow Cytometry , Humans , Mesenchymal Stem Cells/cytology , Transfection , Umbilical Cord/cytology
9.
Sheng Wu Gong Cheng Xue Bao ; 25(4): 580-6, 2009 Apr.
Article in Chinese | MEDLINE | ID: mdl-19637635

ABSTRACT

To prepare a soluble human extracellular III domain of Flt1 and analyze its biological activity. The gene encoding extracellular domain III of Flt-1 was cloned into the expression vector pAZY by RT-PCR from human umbilical vein endothelial cell (HUVEC), and induced to express in Escherichia coli by low phosphoric medium, the product was purified by E-tag affinity chromatography. SDS-PAGE and Western blotting analysis showed that Flt-1 gene domain III gene was expressed in E. coli and the yield of the soluble fusion protein was about 1.10 mg/L. Enzyme-Linked ImmunoSorbent Assay (ELISA) revealed that the Flt-1 domain III was able to bind to VEGF165 dose-dependently. Monolayer denudation assay and Transwell assay showed that the fusion protein could inhibit HUVECs migration induced by conditional medium with 50 ng/mL VEGF165 and 100 ng/mL bFGF. In conclusion, Flt-1 gene domain III gene has been successfully cloned and expressed in E. coli, which will be useful in both the research on the function of Flt-1 gene domain III and preparation of anti-Flt-1 monoclonal antibody in the future.


Subject(s)
Genetic Vectors/genetics , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor Receptor-1/metabolism , Cloning, Molecular , Endothelial Cells/cytology , Endothelial Cells/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Extracellular Space/metabolism , Humans , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/pharmacology , Solubility , Umbilical Veins/cytology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...