Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Nat Commun ; 12(1): 3330, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099664

ABSTRACT

Human pluripotent stem cell (hPSC)-derived pancreatic ß cells are an attractive cell source for treating diabetes. However, current derivation methods remain inefficient, heterogeneous, and cell line dependent. To address these issues, we first devised a strategy to efficiently cluster hPSC-derived pancreatic progenitors into 3D structures. Through a systematic study, we discovered 10 chemicals that not only retain the pancreatic progenitors in 3D clusters but also enhance their potentiality towards NKX6.1+/INS+ ß cells. We further systematically screened signaling pathway modulators in the three steps from pancreatic progenitors toward ß cells. The implementation of all these strategies and chemical combinations resulted in generating ß cells from different sources of hPSCs with high efficiency. The derived ß cells are functional and can reverse hyperglycemia in mice within two weeks. Our protocol provides a robust platform for studying human ß cells and developing hPSC-derived ß cells for cell replacement therapy.


Subject(s)
Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Pancreas/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Cell- and Tissue-Based Therapy , Diabetes Mellitus/metabolism , Diabetes Mellitus, Experimental , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction
2.
Perm J ; 21: 16-176, 2017.
Article in English | MEDLINE | ID: mdl-28633730
3.
Article in English | MEDLINE | ID: mdl-28702240

ABSTRACT

Human embryonic stem cells (hESCs) and induced pluripotent cells (iPSCs) have the potential to differentiate into any somatic cell, making them ideal candidates for cell replacement therapies to treat a number of human diseases and regenerate damaged or non-functional tissues and organs. Key to the promise of regenerative medicine is developing standardized protocols that can safely be applied in patients. Progress towards this goal has occurred in a number of fields, including type 1 diabetes mellitus (T1D). During the past 10 years, significant technological advances in hESC/iPSC biochemistry have provided a roadmap to generate sufficient quantities of glucose-responsive, insulin-producing cells capable of eliminating diabetes in rodents. Although many of the molecular mechanisms underlying the genesis of these cells remain to be elucidated, the field of cell-based therapeutics to treat T1D has advanced to the point where the first Phase I/II trials in humans have begun. Here, we provide a concise review of the history of cell replacement therapies to treat T1D from islet transplantations and xenotranplantation, to current work in hESC/iPSC. We also highlight the latest advances in efforts to employ insulin-producing, glucose-responsive ß-like cells derived from hESC as therapeutics.

4.
J Vis Exp ; (87)2014 May 18.
Article in English | MEDLINE | ID: mdl-24895054

ABSTRACT

For almost 30 years, scientists have demonstrated that human fetal ICCs transplanted under the kidney capsule of nude mice matured into functioning endocrine cells, as evidenced by a significant increase in circulating human C-peptide following glucose stimulation(1-9). However in vitro, genesis of insulin producing cells from human fetal ICCs is low(10); results reminiscent of recent experiments performed with human embryonic stem cells (hESC), a renewable source of cells that hold great promise as a potential therapeutic treatment for type 1 diabetes. Like ICCs, transplantation of partially differentiated hESC generate glucose responsive, insulin producing cells, but in vitro genesis of insulin producing cells from hESC is much less robust(11-17). A complete understanding of the factors that influence the growth and differentiation of endocrine precursor cells will likely require data generated from both ICCs and hESC. While a number of protocols exist to generate insulin producing cells from hESC in vitro(11-22), far fewer exist for ICCs(10,23,24). Part of that discrepancy likely comes from the difficulty of working with human fetal pancreas. Towards that end, we have continued to build upon existing methods to isolate fetal islets from human pancreases with gestational ages ranging from 12 to 23 weeks, grow the cells as a monolayer or in suspension, and image for cell proliferation, pancreatic markers and human hormones including glucagon and C-peptide. ICCs generated by the protocol described below result in C-peptide release after transplantation under the kidney capsule of nude mice that are similar to C-peptide levels obtained by transplantation of fresh tissue(6). Although the examples presented here focus upon the pancreatic endoderm proliferation and ß cell genesis, the protocol can be employed to study other aspects of pancreatic development, including exocrine, ductal, and other hormone producing cells.


Subject(s)
Cytological Techniques/methods , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Pancreas/cytology , Pancreas/embryology , Animals , Cell Aggregation/physiology , Cell Growth Processes/physiology , Fetus/cytology , Humans , Male , Mice
5.
Stem Cells ; 32(9): 2360-72, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24805944

ABSTRACT

MicroRNAs (miRNAs) are noncoding, regulatory RNAs expressed dynamically during differentiation of human embryonic stem cells (hESCs) into defined lineages. Mapping developmental expression of miRNAs during transition from pluripotency to definitive endoderm (DE) should help to elucidate the mechanisms underlying lineage specification and ultimately enhance differentiation protocols. In this report, next generation sequencing was used to build upon our previous analysis of miRNA expression in human hESCs and DE. From millions of sequencing reads, 747 and 734 annotated miRNAs were identified in pluripotent and DE cells, respectively, including 77 differentially expressed miRNAs. Among these, four of the top five upregulated miRNAs were previously undetected in DE. Furthermore, the stem-loop for miR-302a, an important miRNA for both hESCs self-renewal and endoderm specification, produced several highly expressed miRNA species (isomiRs). Overall, isomiRs represented >10% of sequencing reads in >40% of all detected stem-loop arms, suggesting that the impact of these abundant miRNA species may have been overlooked in previous studies. Because of their relative abundance, the role of differential isomiR targeting was studied using the miR-302 cluster as a model system. A miRNA mimetic for miR-302a-5p, but not miR-302a-5p(+3), decreased expression of orthodenticle homeobox 2 (OTX2). Conversely, isomiR 302a-5p(+3) selectively decreased expression of tuberous sclerosis protein 1, but not OTX2, indicating nonoverlapping specificity of miRNA processing variants. Taken together, our characterization of miRNA expression, which includes novel miRNAs and isomiRs, helps establish a foundation for understanding the role of miRNAs in DE formation and selective targeting by isomiRs.


Subject(s)
Embryonic Stem Cells/physiology , Endoderm/physiology , MicroRNAs/chemistry , RNA, Small Interfering/genetics , Cell Culture Techniques , Cell Differentiation/genetics , Embryonic Stem Cells/chemistry , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endoderm/chemistry , Endoderm/cytology , Endoderm/metabolism , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Pluripotent Stem Cells/chemistry , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/physiology , RNA, Small Interfering/metabolism , Sequence Analysis, RNA , Transfection
6.
Regen Med ; 7(4): 474, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22993884
10.
PLoS One ; 7(6): e38721, 2012.
Article in English | MEDLINE | ID: mdl-22761699

ABSTRACT

The chemokine receptor CXCR4 and ligand SDF-1α are expressed in fetal and adult mouse islets. Neutralization of CXCR4 has previously been shown to diminish ductal cell proliferation and increase apoptosis in the IFNγ transgenic mouse model in which the adult mouse pancreas displays islet regeneration. Here, we demonstrate that CXCR4 and SDF-1α are expressed in the human fetal pancreas and that during early gestation, CXCR4 colocalizes with neurogenin 3 (ngn3), a key transcription factor for endocrine specification in the pancreas. Treatment of islet like clusters (ICCs) derived from human fetal pancreas with SDF-1α resulted in increased proliferation of epithelial cells in ICCs without a concomitant increase in total insulin expression. Exposure of ICCs in vitro to AMD3100, a pharmacological inhibitor of CXCR4, did not alter expression of endocrine hormones insulin and glucagon, or the pancreatic endocrine transcription factors PDX1, Nkx6.1, Ngn3 and PAX4. However, a strong inhibition of ß cell genesis was observed when in vitro AMD3100 treatment of ICCs was followed by two weeks of in vivo treatment with AMD3100 after ICC transplantation into mice. Analysis of the grafts for human C-peptide found that inhibition of CXCR4 activity profoundly inhibits islet development. Subsequently, a model pancreatic epithelial cell system (CFPAC-1) was employed to study the signals that regulate proliferation and apoptosis by the SDF-1α/CXCR4 axis. From a selected panel of inhibitors tested, both the PI 3-kinase and MAPK pathways were identified as critical regulators of CFPAC-1 proliferation. SDF-1α stimulated Akt phosphorylation, but failed to increase phosphorylation of Erk above the high basal levels observed. Taken together, these results indicate that SDF-1α/CXCR4 axis plays a critical regulatory role in the genesis of human islets.


Subject(s)
Cell Proliferation , Chemokine CXCL12/metabolism , Endocrine Cells/cytology , Fetus/cytology , Islets of Langerhans/cytology , Receptors, CXCR4/metabolism , Stem Cells/cytology , Adult , Animals , Apoptosis , Benzylamines , Blotting, Western , C-Peptide/genetics , C-Peptide/metabolism , Cell Differentiation , Cyclams , Endocrine Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fetus/metabolism , Fluorescent Antibody Technique , Heterocyclic Compounds/pharmacology , Humans , Islets of Langerhans/metabolism , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, CXCR4/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Stem Cells/metabolism
11.
Biores Open Access ; 1(4): 184-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23515456

ABSTRACT

The presence of pancreatic stem cells (PnSCs) has not been firmly demonstrated in the human or animal pancreas. Previous reports have suggested that ductal and acinar structures in the exocrine pancreas can be a potential source of progenitor cells. More recently, immature insulin precursors in the periphery of human islets have been found to self-replicate and differentiate to endocrine cells in vitro. Transplantation of these cells under the kidney capsule improves the diabetic state in mice. The controversy surrounding where PnSCs reside could be resolved if a specific marker were to be found that allowed their identification, purification, and directed differentiation to endocrine cells. We have identified in human pancreas cells positive for the stage-specific embryonic antigen 4 (SSEA4), a stem cell marker. These cells also express ductal, pancreatic progenitor, and stem cell protein markers. Interestingly, some of the SSEA4(+) cells scattered in the ducts do not show a ductal cell phenotype. SSEA4(+)-sorted cells formed aggregate-like spheres in culture and robustly differentiated to pancreatic hormone-expressing cells in conditions of high glucose concentration and B27 supplementation. We hypothesize that SSEA4(+) cells or a subpopulation of those cells residing in the pancreatic ducts may be the elusive PnSCs, and in this case, SSEA4 may represent a potential surface antigen marker for human PnSCs. The discovery of specific markers for the identification and purification of human PnSCs would greatly facilitate studies aimed at the expansion of these cells and the development of targeting tools for their potential induction to insulin-producing cells.

12.
J Biol Chem ; 286(41): 36042-36052, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21852242

ABSTRACT

Stepwise approaches for the derivation of ß-cells from human embryonic stem cells have been described. However, low levels of endocrine specification limit the final yield of insulin-producing ß-cells. In this study, we show that the pyrrolo-pyrimidine Src family kinase (SFK) inhibitor PP2 effectively promotes the endocrine specification of human embryonic stem cell derivatives based on its capacity to induce the expression of proendocrine transcription factors (NGN3, NEUROD1, NKX2.2, and PAX4) and to significantly increase the final yield of insulin-positive cells. We further demonstrate that PP2 inhibits the activation of focal adhesion kinase (FAK), and selective inhibition of this kinase is also sufficient to induce early endocrine commitment based on increased expression of NGN3, NEUROD1, and NKX2.2. Additional studies using dominant negative constructs and isolated human fetal pancreata suggest that c-Src is at least partially responsible for inhibiting early endocrine specification. Mechanistically, we propose that inhibition of SFK/FAK signaling can promote endocrine specification by limiting activation of the TGFßR/Smad2/3 pathway. Moreover, we show that inhibition of SFK/FAK signaling suppresses cell growth, increases the expression of the ß-cell-associated cyclin-dependent kinase inhibitor p57kip2, and simultaneously suppresses the expression of Id1 and Id2. This study has important implications for the derivation of ß-cells for the cell-based therapy of diabetes and sheds new light on the signaling events that regulate early endocrine specification.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/metabolism , Focal Adhesion Kinase 1/antagonists & inhibitors , Insulin-Secreting Cells/metabolism , Pluripotent Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , src-Family Kinases/antagonists & inhibitors , Antigens, Differentiation/biosynthesis , Cell Line , Cell- and Tissue-Based Therapy , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Diabetes Mellitus/metabolism , Diabetes Mellitus/therapy , Embryonic Stem Cells/cytology , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation/drug effects , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Inhibitor of Differentiation Protein 1/biosynthesis , Inhibitor of Differentiation Protein 2/biosynthesis , Insulin-Secreting Cells/cytology , Nuclear Proteins , Pluripotent Stem Cells/cytology , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcription Factors , src-Family Kinases/metabolism
13.
Proc Natl Acad Sci U S A ; 107(18): 8129-34, 2010 May 04.
Article in English | MEDLINE | ID: mdl-20406903

ABSTRACT

Using a high-throughput chemical screen, we identified two small molecules that enhance the survival of human embryonic stem cells (hESCs). By characterizing their mechanisms of action, we discovered an essential role of E-cadherin signaling for ESC survival. Specifically, we showed that the primary cause of hESC death following enzymatic dissociation comes from an irreparable disruption of E-cadherin signaling, which then leads to a fatal perturbation of integrin signaling. Furthermore, we found that stability of E-cadherin and the resulting survival of ESCs were controlled by specific growth factor signaling. Finally, we generated mESC-like hESCs by culturing them in mESC conditions. And these converted hESCs rely more on E-cadherin signaling and significantly less on integrin signaling. Our data suggest that differential usage of cell adhesion systems by ESCs to maintain self-renewal may explain their profound differences in terms of morphology, growth factor requirement, and sensitivity to enzymatic cell dissociation.


Subject(s)
Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Signal Transduction , Animals , Cadherins/metabolism , Cell Adhesion , Cell Communication , Cell Shape , Cell Survival , Cells, Cultured , Embryonic Stem Cells/cytology , Extracellular Matrix/metabolism , Humans , Integrins/metabolism , Mice , Pluripotent Stem Cells/cytology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism
14.
J Cell Physiol ; 224(1): 101-11, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20232313

ABSTRACT

A critical shortage of donor pancreata currently prevents the development of a universal cell-based therapy for type I diabetes. The ex vivo expansion of insulin-producing beta-cells offers a potential solution but is problematic due to the inherent tendency of these cells to transition into mesenchymal-like cells that are devoid of function. Here, we demonstrate for the first time that exposure to elements of the extracellular matrix (ECM) directly potentiates the mesenchymal transition of cultured fetal beta-cells and causes associated declines in insulin gene expression. Individual ECM constituents varied in their ability to induce such responses, with collagen-IV (C-IV) and fibronectin inducing strong responses, whereas laminin-1 had no significant effect. Mesenchymal transition and concomitant losses in insulin gene expression observed on C-IV were found to be dependent on beta(1)-integrin ligation and were augmented in the presence of hepatocyte growth factor. Importantly, selective inhibition of c-Src, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinase (ERK) prior to exposure to C-IV prevented mesenchymal transition and effectively preserved insulin expression. Fetal beta-cells undergoing mesenchymal transition were found to acquire alpha(1)beta(1) expression, and ligation of this integrin then promotes declines in insulin gene expression and a marked increase in beta-cell motility. Inhibition of Src-, ERK-, or JNK-dependent signaling combined with the selective regulation of matrix exposure may ultimately facilitate the development of more effective beta-cell expansion protocols.


Subject(s)
Cell Transdifferentiation , Extracellular Matrix Proteins/metabolism , Insulin-Secreting Cells/metabolism , Insulin/genetics , Integrin alpha1beta1/metabolism , Mesoderm/metabolism , Signal Transduction , Aged , CSK Tyrosine-Protein Kinase , Cell Adhesion , Cell Movement , Cell Transdifferentiation/drug effects , Cells, Cultured , Collagen Type IV/metabolism , Down-Regulation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/metabolism , Gestational Age , Hepatocyte Growth Factor/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Laminin/metabolism , Middle Aged , Pancreas/embryology , Pancreas/metabolism , Phenotype , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection , Vimentin/metabolism , src-Family Kinases
15.
Stem Cells Dev ; 19(6): 797-807, 2010 Jun.
Article in English | MEDLINE | ID: mdl-19807270

ABSTRACT

Human embryonic stem cells (hESCs) have the potential to differentiate into many adult cell types, and they are being explored as a resource for cell replacement therapies for multiple diseases. In order to optimize in vitro differentiation protocols, it will be necessary to elucidate regulatory mechanisms that contribute to lineage specification. MicroRNAs (miRNAs) are emerging as key regulators of hESC differentiation and embryonic development. In this study, we compare miRNA expression profiles between pluripotent hESCs and definitive endoderm (DE), an early step in the pathway toward the pancreatic lineage. Results from microarray analysis showed that DE can be distinguished by its unique miRNA profile, which consists of 37 significantly down-regulated and 17 up-regulated miRNAs in 2 different cell lines and in the presence/absence of feeder layers. Comparison to other hESC-derived lineages showed that most of the highly up-regulated miRNAs are specific to endoderm in early development. Notably, miR-375, which was previously implicated in regulating development and function of later stages of pancreatic development, is highly and specifically up-regulated during DE formation, suggesting that it may have a distinct role very early in development. Examination of potential mRNA targets showed that TIMM8A is repressed by ectopic miR-375 expression in pluripotent hESCs.


Subject(s)
Embryonic Stem Cells/metabolism , Endoderm/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , MicroRNAs/genetics , Animals , Base Sequence , Cell Differentiation/genetics , Cell Line , Cell Lineage/genetics , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Endoderm/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , MicroRNAs/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Time Factors , Up-Regulation/genetics
16.
Stem Cells ; 27(12): 2992-3000, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19839055

ABSTRACT

Induced pluripotent stem cell technology has attracted enormous interest for potential application in regenerative medicine. Here, we report that a specific glycogen synthase kinase 3 (GSK-3) inhibitor, CHIR99021, can induce the reprogramming of mouse embryonic fibroblasts transduced by only two factors, Oct4 and Klf4. When combined with Parnate (also named tranylcypromine), an inhibitor of lysine-specific demethylase 1, CHIR99021 can cause the reprogramming of human primary keratinocyte transduced with the two factors, Oct4 and Klf4. To our knowledge, this is the first time that human iPS cells have been generated from somatic cells without exogenous Sox2 expression. Our studies suggest that the GSK-3 inhibitor might have a general application to replace transcription factors in both mouse and human reprogramming.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming , Pluripotent Stem Cells/chemistry , SOXB1 Transcription Factors/metabolism , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/drug effects , Coculture Techniques , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3/antagonists & inhibitors , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , SOXB1 Transcription Factors/genetics
17.
Nat Methods ; 6(11): 805-8, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19838168

ABSTRACT

The slow kinetics and low efficiency of reprogramming methods to generate human induced pluripotent stem cells (iPSCs) impose major limitations on their utility in biomedical applications. Here we describe a chemical approach that dramatically improves (200-fold) the efficiency of iPSC generation from human fibroblasts, within seven days of treatment. This will provide a basis for developing safer, more efficient, nonviral methods for reprogramming human somatic cells.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/cytology , Benzamides/pharmacology , Dioxoles/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Fibroblasts/physiology , Humans , Induced Pluripotent Stem Cells/physiology , MAP Kinase Kinase 1/antagonists & inhibitors , Pyrimidines/pharmacology , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Thiazoles/pharmacology , Transduction, Genetic
19.
Stem Cells ; 27(1): 68-77, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18845765

ABSTRACT

Using human embryonic stem cells (hESCs), we describe a novel method for the rapid derivation and enrichment of cells that are comparable to primordial germ cells (PGCs) and Sertoli cells. The methodology described is based on modest changes to the growth conditions commonly used to expand hESCs and does not require genetic manipulation or complex three-dimensional culture. Remarkably, we have determined that simply reducing the size of cultured ESC colonies and manipulating the number of feeding cycles, results in the rapid emergence of cells that are comparable to migratory PGCs. Importantly, these cells can be monitored and purified on the basis of the expression of the chemokine receptor CXCR4. Under more stringent differentiating conditions these cells mature and upregulate the expression of specific germ cell markers. Importantly, this process is accompanied by the development of Sertoli-like support cells. Such cells normally provide trophic support and immunoprotection to developing germ cells and may have significant clinical utility in the prevention of graft rejection. The putative Sertoli-germ cell cocultures generated in this study may ultimately be developed to study and manipulate interactions and processes involved in human gametogenesis.


Subject(s)
Cell Culture Techniques/methods , Embryonic Stem Cells/cytology , Germ Cells/cytology , Sertoli Cells/cytology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Line , Cell Movement , Cell Shape , Cell Survival , Coculture Techniques , Colony-Forming Units Assay , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/ultrastructure , Gene Expression Regulation, Developmental , Germ Cells/metabolism , Germ Cells/ultrastructure , Humans , Male , Mice , Phenotype , Receptors, CXCR4/metabolism , Sertoli Cells/metabolism , Sertoli Cells/ultrastructure
20.
Regen Med ; 3(2): 175-80, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18307401

ABSTRACT

PURPOSE: The purpose of these studies was twofold: to reduce the level of nonhuman, potentially immunogenic sialic acid N-glycolylneuraminic acid (Neu5Gc) in human embryonic stem cells (hESCs) through culture of the cells in the absence of feeder layers; and to determine whether directed differentiation was preserved under these conditions, that is, using exclusively human-derived products. METHODS: Using a technique developed in our laboratory to culture hESCs in the absence of feeder layers, all nonhuman cell culture reagents were replaced with recombinant or human-derived reagents. The level of the nonhuman sialic acid (Neu5Gc) was measured by high-performance liquid chromatography and monitored over many passages. Subsequently, the cells were subjected to in vitro differentiation into definitive endoderm by lowering the serum concentrations and elevating the amount of activin A. RESULTS: Under standard tissue culture conditions using mouse and other animal products, the basal levels of Neu5Gc were measured between 7 and 10%. After the cell culture reagents were changed to all human products, Neu5Gc levels decreased steadily before leveling below 2%. Upon initiation of the differentiation protocol under these cell culture conditions, we observed robust endoderm formation, as measured by fluorescence-activated cell sorting analysis and the appearance of mRNA for markers of definitive endoderm (Sox17, CXCR4, Goosecoid and FoxA2). CONCLUSION: Consistent with other findings, elimination of nonhuman products in cell culture of hESCs decreases the levels of nonhuman and potentially immunogenic sialic acid levels. Furthermore, our studies demonstrate that in this feeder layer-free system, hESCs undergo directed differentiation into definitive endoderm.


Subject(s)
Embryonic Stem Cells/cytology , Endoderm/cytology , Animals , Base Sequence , Cell Differentiation , Cell Separation , Cells, Cultured , Chromatography, High Pressure Liquid , DNA Primers , Humans , Mice , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...