Gene transfer into pluripotent stem cells via lentiviral transduction
Recombinant lentiviral vectors are powerful tools to stably manipulate human pluripotent stem cells. They can be used to deliver ectopic genes,shRNAs,miRNAs,or any possible genetic DNA sequence into diving and nondividing cells. Here we describe a general protocol for the production of self-inactivating lentiviral vector particles and their purification to high titers by either ultracentrifugation or ultrafiltration. Next we provide a basic procedure to transduce human pluripotent stem cells and propagate clonal cell lines.
View Publication
产品类型:
产品号#:
05850
05857
05870
05875
07923
85850
85857
85870
85875
产品名:
Dispase (1 U/mL)
mTeSR™1
mTeSR™1
Keller KC et al. (MAR 2016)
Stem Cells and Development 25 13 scd.2015.0367
Wnt5a Supports Osteogenic Lineage Decisions in Embryonic Stem Cells
The specification of pluripotent stem cells into the bone-forming osteoblasts has been explored in a number of studies. However,the current body of literature has yet to adequately address the role of Wnt glycoproteins in the differentiation of pluripotent stem cells along the osteogenic lineage. During mouse embryonic stem cell (ESC) in vitro osteogenesis,the non-canonical WNT5a is expressed early on. Cells either sorted by their positive WNT5a expression or when supplemented with recombinant WNT5a (rWNT5a) during a two-day window showed significantly enhanced osteogenic yield. Mechanistically,rWNT5a supplementation up-regulated PKC,CamKII and JNK activity while antagonizing the key effector of canonical Wnt signaling: beta-catenin. Conversely,when recombinant WNT3a (rWNT3a) or other positive regulators of �?�-catenin were employed during this same time-window there was a decrease in osteogenic marker expression. However,if rWNT3a was supplemented during a time-window following rWNT5a treatment,osteogenic differentiation was enhanced both in murine and human ESCs. Elucidating the role of these WNT ligands in directing the early stages of osteogenesis has the potential to considerably improve tissue engineering protocols and applications for regenerative medicine.
View Publication
Hussain I et al. (JUL 2012)
Cell biology international 36 7 595--600
New approach to isolate mesenchymal stem cell (MSC) from human umbilical cord blood.
HUCB (human umbilical cord blood) has been frequently used in clinical allogeneic HSC (haemopoietic stem cell) transplant. However,HUCB is poorly recognized as a rich source of MSC (mesenchymal stem cell). The aim of this study has been to establish a new method for isolating large number of MSC from HUCB to recognize it as a good source of MSC. HUCB samples were collected from women following their elective caesarean section. The new method (Clot Spot method) was carried out by explanting HUCB samples in mesencult complete medium and maintained in 37°C,in a 5% CO2 and air incubator. MSC presence was established by quantitative and qualitative immunophenotyping of cells and using FITC attached to MSC phenotypic markers (CD29,CD73,CD44 and CD105). Haematopoietic antibodies (CD34 and CD45) were used as negative control. MSC differentiation was examined in neurogenic and adipogenic media. Immunocytochemistry was carried out for the embryonic markers: SOX2 (sex determining region Y-box 2),OLIG-4 (oligodendrocyte-4) and FABP-4 (fatty acid binding protein-4). The new method was compared with the conventional Rosset Sep method. MSC cultures using the Clot Spot method showed 3-fold increase in proliferation rate compared with conventional method. Also,the cells showed high expression of MSC markers CD29,CD73,CD44 and CD105,but lacked the expression of specific HSC markers (CD34 and CD45). The isolated MSC showed some differentiation by expressing the neurogenic (SOX2 and Olig4) and adipogenic (FABP-4) markers respectively. In conclusion,HUCB is a good source of MSC using this new technique.
View Publication
产品类型:
产品号#:
05401
05402
05411
15128
15168
产品名:
MesenCult™ MSC基础培养基 (人)
MesenCult™ MSC 刺激补充剂(人)
MesenCult™ 增殖试剂盒(人)
RosetteSep™人间充质干细胞富集抗体混合物
RosetteSep™人间充质干细胞富集抗体混合物
Hansen A et al. (JUN 2014)
Advanced Healthcare Materials 3 6 848--853
High-Density Polymer Microarrays: Identifying Synthetic Polymers that Control Human Embryonic Stem Cell Growth
The fabrication of high-density polymer microarray is described,allowing the simultaneous and efficient evaluation of more than 7000 different polymers in a single-cellular-based screen. These high-density polymer arrays are applied in the search for synthetic substrates for hESCs culture. Up-scaling of the identified hit polymers enables long-term cellular cultivation and promoted successful stem-cell maintenance.
View Publication
产品类型:
产品号#:
05850
05857
05870
05875
85850
85857
85870
85875
27845
27945
27840
27865
27940
27965
产品名:
mTeSR™1
mTeSR™1
Leberbauer C et al. (JAN 2005)
Blood 105 1 85--94
Different steroids co-regulate long-term expansion versus terminal differentiation in primary human erythroid progenitors.
Outgrowth,long-term self-renewal,and terminal maturation of human erythroid progenitors derived from umbilical cord blood in serum-free medium can be modulated by steroid hormones. Homogeneous erythroid cultures,as characterized by flow cytometry and dependence on a specific mixture of physiologic proliferation factors,were obtained within 8 days from a starting population of mature and immature mononuclear cells. Due to previous results in mouse and chicken erythroblasts,the proliferation-promoting effect of glucocorticoids was not unexpected. Surprisingly,however,androgen had a positive effect on the sustained expansion of human female but not male erythroid progenitors. Under optimal conditions,sustained proliferation of erythroid progenitors resulted in a more than 10(9)-fold expansion within 60 days. Terminal erythroid maturation was significantly improved by adding human serum and thyroid hormone (3,5,3'-triiodothyronine [T3]) to the differentiation medium. This resulted in highly synchronous differentiation of the cells toward enucleated erythrocytes within 6 days,accompanied by massive size decrease and hemoglobin accumulation to levels comparable to those in peripheral blood erythrocytes. Thus,obviously,different ligand-activated nuclear hormone receptors massively influence the decision between self-renewal and terminal maturation in the human erythroid compartment.
View Publication
Production of de novo cardiomyocytes: human pluripotent stem cell differentiation and direct reprogramming.
Cardiovascular disease is a leading cause of death worldwide. The limited capability of heart tissue to regenerate has prompted methodological developments for creating de novo cardiomyocytes,both in vitro and in vivo. Beyond uses in cell replacement therapy,patient-specific cardiomyocytes may find applications in drug testing,drug discovery,and disease modeling. Recently,approaches for generating cardiomyocytes have expanded to encompass three major sources of starting cells: human pluripotent stem cells (hPSCs),adult heart-derived cardiac progenitor cells (CPCs),and reprogrammed fibroblasts. We discuss state-of-the-art methods for generating de novo cardiomyocytes from hPSCs and reprogrammed fibroblasts,highlighting potential applications and future challenges.
View Publication
产品类型:
产品号#:
05850
05857
05870
05875
85850
85857
85870
85875
产品名:
mTeSR™1
mTeSR™1
Verma R et al. (AUG 2014)
The Journal of experimental medicine 211 9 1715--22
RHEX, a novel regulator of human erythroid progenitor cell expansion and erythroblast development.
Ligation of erythropoietin (EPO) receptor (EPOR) JAK2 kinase complexes propagates signals within erythroid progenitor cells (EPCs) that are essential for red blood cell production. To reveal hypothesized novel EPOR/JAK2 targets,a phosphotyrosine (PY) phosphoproteomics approach was applied. Beyond known signal transduction factors,32 new targets of EPO-modulated tyrosine phosphorylation were defined. Molecular adaptors comprised one major set including growth factor receptor-bound protein 2 (GRB2)-associated binding proteins 1-3 (GAB1-3),insulin receptor substrate 2 (IRS2),docking protein 1 (DOK1),Src homology 2 domain containing transforming protein 1 (SHC1),and sprouty homologue 1 (SPRY1) as validating targets,and SPRY2,SH2 domain containing 2A (SH2D2A),and signal transducing adaptor molecule 2 (STAM2) as novel candidate adaptors together with an ORF factor designated as regulator of human erythroid cell expansion (RHEX). RHEX is well conserved in Homo sapiens and primates but absent from mouse,rat,and lower vertebrate genomes. Among tissues and lineages,RHEX was elevated in EPCs,occurred as a plasma membrane protein,was rapidly PY-phosphorylated textgreater20-fold upon EPO exposure,and coimmunoprecipitated with the EPOR. In UT7epo cells,knockdown of RHEX inhibited EPO-dependent growth. This was associated with extracellular signal-regulated kinase 1,2 (ERK1,2) modulation,and RHEX coupling to GRB2. In primary human EPCs,shRNA knockdown studies confirmed RHEX regulation of erythroid progenitor expansion and further revealed roles in promoting the formation of hemoglobinizing erythroblasts. RHEX therefore comprises a new EPO/EPOR target and regulator of human erythroid cell expansion that additionally acts to support late-stage erythroblast development.
View Publication
产品类型:
产品号#:
04434
04444
22001
22005
22006
22007
22008
22009
22011
22012
22013
产品名:
MethoCult™H4434经典
MethoCult™H4434经典
STEMvision™ 人脐带血7-天CFU分析包
STEMvision™ 彩色人脐带血14-天CFU分析包
STEMvision™ 彩色人骨髓14-天CFU分析包
STEMvision™ 彩色人动员外周血14-天CFU分析包
STEMvision™ 小鼠总CFU分析包
STEMvision™ 小鼠髓系CFU分析包
STEMvision™ 小鼠红系CFU分析包
STEMvision™ 小鼠CFU分析包(髓系和红系)
Freedman BS et al. (OCT 2015)
Nature communications 6 May 8715
Modelling kidney disease with CRISPR-mutant kidney organoids derived from human pluripotent epiblast spheroids.
Human-pluripotent-stem-cell-derived kidney cells (hPSC-KCs) have important potential for disease modelling and regeneration. Whether the hPSC-KCs can reconstitute tissue-specific phenotypes is currently unknown. Here we show that hPSC-KCs self-organize into kidney organoids that functionally recapitulate tissue-specific epithelial physiology,including disease phenotypes after genome editing. In three-dimensional cultures,epiblast-stage hPSCs form spheroids surrounding hollow,amniotic-like cavities. GSK3β inhibition differentiates spheroids into segmented,nephron-like kidney organoids containing cell populations with characteristics of proximal tubules,podocytes and endothelium. Tubules accumulate dextran and methotrexate transport cargoes,and express kidney injury molecule-1 after nephrotoxic chemical injury. CRISPR/Cas9 knockout of podocalyxin causes junctional organization defects in podocyte-like cells. Knockout of the polycystic kidney disease genes PKD1 or PKD2 induces cyst formation from kidney tubules. All of these functional phenotypes are distinct from effects in epiblast spheroids,indicating that they are tissue specific. Our findings establish a reproducible,versatile three-dimensional framework for human epithelial disease modelling and regenerative medicine applications.
View Publication
产品类型:
产品号#:
05850
05857
05870
05875
07920
85850
85857
85870
85875
产品名:
ACCUTASE™
mTeSR™1
mTeSR™1
Mora-Bermú et al. (SEP 2016)
eLife 5
Differences and similarities between human and chimpanzee neural progenitors during cerebral cortex development.
Human neocortex expansion likely contributed to the remarkable cognitive abilities of humans. This expansion is thought to primarily reflect differences in proliferation versus differentiation of neural progenitors during cortical development. Here,we have searched for such differences by analysing cerebral organoids from human and chimpanzees using immunohistochemistry,live imaging,and single-cell transcriptomics. We find that the cytoarchitecture,cell type composition,and neurogenic gene expression programs of humans and chimpanzees are remarkably similar. Notably,however,live imaging of apical progenitor mitosis uncovered a lengthening of prometaphase-metaphase in humans compared to chimpanzees that is specific to proliferating progenitors and not observed in non-neural cells. Consistent with this,the small set of genes more highly expressed in human apical progenitors points to increased proliferative capacity,and the proportion of neurogenic basal progenitors is lower in humans. These subtle differences in cortical progenitors between humans and chimpanzees may have consequences for human neocortex evolution.
View Publication
产品类型:
产品号#:
05850
05857
05870
05875
85850
85857
85870
85875
产品名:
mTeSR™1
mTeSR™1
Smith GH (JAN 1996)
Breast cancer research and treatment 39 1 21--31
Experimental mammary epithelial morphogenesis in an in vivo model: evidence for distinct cellular progenitors of the ductal and lobular phenotype.
An in vivo transplantation system has been used to evaluate the developmental capacities of specific mouse mammary epithelial cell populations. Specifically,mouse mammary epithelial cells with distinctly limited developmental potentials have been identified using this procedure. Two distinct epithelial cell progenitors have been identified by experiments designed to determine whether basal lobular and ductal phenotypes could develop independently under conditions imposed by a limiting dilution. The prediction that these separate epithelial progenitors must exist was based upon the results from transplantation experiments carried out in epithelium-divested mammary fat pads of syngeneic mice with mammary epithelium from two different transgenic mouse models. The results presented here demonstrate the following points: 1) lobular,i.e. secretory,progenitor cells are present as distinct entities among the mammary epithelial cells found in immature virgin female mice; 2) similarly,ductal epithelial progenitors are present within the same population; 3) lobular progenitors are present in greater numbers,although both cell populations are extremely small; 4) as expected,some inocula produce outgrowths with simultaneous development of both lobular and ductal phenotypes--it is not known whether this indicates cooperative interaction between the two epithelial progenitors or signals the presence of a third progenitor type capable of producing both ductular and lobular committed daughters; 5) these findings have important consequences in the design of experiments aimed at testing the effects of known and putative mammary oncogenes and tumor suppressor genes,using techniques which include cellular transformation in vitro followed by in vivo cultivation and evaluation.
View Publication
产品类型:
产品号#:
01700
01705
05601
05610
05620
产品名:
ALDEFLUOR™ 试剂盒
ALDEFLUOR™ DEAB试剂
EpiCult™-B 人培养基
EpiCult™-B 小鼠培养基试剂盒
MammoCult™人培养基试剂盒
Jiang T et al. (FEB 2009)
Cancer research 69 3 845--54
Achaete-scute complex homologue 1 regulates tumor-initiating capacity in human small cell lung cancer.
The basic helix-loop-helix transcription factor achaete-scute complex homologue 1 (ASCL1) is essential for the development of normal lung neuroendocrine cells as well as other endocrine and neural tissues. Small cell lung cancer (SCLC) and non-SCLC with neuroendocrine features express ASCL1,where the factor may play a role in the virulence and primitive neuroendocrine phenotype of these tumors. In this study,RNA interference knockdown of ASCL1 in cultured SCLC resulted in inhibition of soft agar clonogenic capacity and induction of apoptosis. cDNA microarray analyses bolstered by expression studies,flow cytometry,and chromatin immunoprecipitation identified two candidate stem cell marker genes,CD133 and aldehyde dehydrogenase 1A1 (ALDH1A1),to be directly regulated by ASCL1 in SCLC. In SCLC direct xenograft tumors,we detected a relatively abundant CD133(high)-ASCL1(high)-ALDH1(high) subpopulation with markedly enhanced tumorigenicity compared with cells with weak CD133 expression. Tumorigenicity in the CD133(high) subpopulation depended on continued ASCL1 expression. Whereas CD133(high) cells readily reconstituted the range of CD133 expression seen in the original xenograft tumor,CD133(low) cells could not. Our findings suggest that a broad range of SCLC cells has tumorigenic capacity rather than a small discrete population. Intrinsic tumor cell heterogeneity,including variation in key regulatory factors such as ASCL1,can modulate tumorigenicity in SCLC.
View Publication