Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Diabetologia ; 66(7): 1306-1321, 2023 07.
Article in English | MEDLINE | ID: mdl-36995380

ABSTRACT

AIMS/HYPOTHESIS: Wolfram syndrome is a rare autosomal recessive disorder caused by pathogenic variants in the WFS1 gene. It is characterised by insulin-dependent diabetes mellitus, optic nerve atrophy, diabetes insipidus, hearing loss and neurodegeneration. Considering the unmet treatment need for this orphan disease, this study aimed to evaluate the therapeutic potential of glucagon-like peptide 1 receptor (GLP-1R) agonists under wolframin (WFS1) deficiency with a particular focus on human beta cells and neurons. METHODS: The effect of the GLP-1R agonists dulaglutide and exenatide was examined in Wfs1 knockout mice and in an array of human preclinical models of Wolfram syndrome, including WFS1-deficient human beta cells, human induced pluripotent stem cell (iPSC)-derived beta-like cells and neurons from control individuals and individuals affected by Wolfram syndrome, and humanised mice. RESULTS: Our study shows that the long-lasting GLP-1R agonist dulaglutide reverses impaired glucose tolerance in WFS1-deficient mice, and that exenatide and dulaglutide improve beta cell function and prevent apoptosis in different human WFS1-deficient models including iPSC-derived beta cells from people with Wolfram syndrome. Exenatide improved mitochondrial function, reduced oxidative stress and prevented apoptosis in Wolfram syndrome iPSC-derived neural precursors and cerebellar neurons. CONCLUSIONS/INTERPRETATION: Our study provides novel evidence for the beneficial effect of GLP-1R agonists on WFS1-deficient human pancreatic beta cells and neurons, suggesting that these drugs may be considered as a treatment for individuals with Wolfram syndrome.


Subject(s)
Induced Pluripotent Stem Cells , Insulin-Secreting Cells , Optic Atrophy , Wolfram Syndrome , Humans , Animals , Mice , Wolfram Syndrome/drug therapy , Wolfram Syndrome/genetics , Exenatide/therapeutic use , Optic Atrophy/pathology , Insulin-Secreting Cells/pathology , Mice, Knockout
2.
Front Cell Dev Biol ; 10: 967765, 2022.
Article in English | MEDLINE | ID: mdl-36060810

ABSTRACT

In vitro differentiation of human induced pluripotent stem cells (iPSCs) into beta cells represents an important cell source for diabetes research. Here, we fully characterized iPSC-derived beta cell function in vitro and in vivo in humanized mice. Using a 7-stage protocol, human iPSCs were differentiated into islet-like aggregates with a yield of insulin-positive beta cells comparable to that of human islets. The last three stages of differentiation were conducted with two different 3D culture systems, rotating suspension or static microwells. In the latter, homogeneously small-sized islet-like aggregates were obtained, while in rotating suspension size was heterogeneous and aggregates often clumped. In vitro function was assessed by glucose-stimulated insulin secretion, NAD(P)H and calcium fluctuations. Stage 7 aggregates slightly increased insulin release in response to glucose in vitro. Aggregates were transplanted under the kidney capsule of NOD-SCID mice to allow for further in vivo beta cell maturation. In transplanted mice, grafts showed glucose-responsiveness and maintained normoglycemia after streptozotocin injection. In situ kidney perfusion assays showed modulation of human insulin secretion in response to different secretagogues. In conclusion, iPSCs differentiated with equal efficiency into beta cells in microwells compared to rotating suspension, but the former had a higher experimental success rate. In vitro differentiation generated aggregates lacking fully mature beta cell function. In vivo, beta cells acquired the functional characteristics typical of human islets. With this technology an unlimited supply of islet-like organoids can be generated from human iPSCs that will be instrumental to study beta cell biology and dysfunction in diabetes.

3.
JCI Insight ; 5(2)2020 01 30.
Article in English | MEDLINE | ID: mdl-31877117

ABSTRACT

Friedreich ataxia is an autosomal recessive neurodegenerative disease associated with a high diabetes prevalence. No treatment is available to prevent or delay disease progression. Friedreich ataxia is caused by intronic GAA trinucleotide repeat expansions in the frataxin-encoding FXN gene that reduce frataxin expression, impair iron-sulfur cluster biogenesis, cause oxidative stress, and result in mitochondrial dysfunction and apoptosis. Here we examined the metabolic, neuroprotective, and frataxin-inducing effects of glucagon-like peptide-1 (GLP-1) analogs in in vivo and in vitro models and in patients with Friedreich ataxia. The GLP-1 analog exenatide improved glucose homeostasis of frataxin-deficient mice through enhanced insulin content and secretion in pancreatic ß cells. Exenatide induced frataxin and iron-sulfur cluster-containing proteins in ß cells and brain and was protective to sensory neurons in dorsal root ganglia. GLP-1 analogs also induced frataxin expression, reduced oxidative stress, and improved mitochondrial function in Friedreich ataxia patients' induced pluripotent stem cell-derived ß cells and sensory neurons. The frataxin-inducing effect of exenatide was confirmed in a pilot trial in Friedreich ataxia patients, showing modest frataxin induction in platelets over a 5-week treatment course. Taken together, GLP-1 analogs improve mitochondrial function in frataxin-deficient cells and induce frataxin expression. Our findings identify incretin receptors as a therapeutic target in Friedreich ataxia.


Subject(s)
Exenatide/pharmacology , Friedreich Ataxia/drug therapy , Gene Expression Regulation/drug effects , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Mitochondria/metabolism , Adolescent , Adult , Aged , Animals , Brain/pathology , Cerebellum/pathology , Disease Models, Animal , Exenatide/therapeutic use , Female , Friedreich Ataxia/genetics , Friedreich Ataxia/metabolism , Ganglia, Spinal/pathology , Gene Knock-In Techniques , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Iron/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Oxidative Stress , Reactive Oxygen Species/metabolism , Trinucleotide Repeat Expansion , Young Adult , Frataxin
4.
Diabetologia ; 63(2): 395-409, 2020 02.
Article in English | MEDLINE | ID: mdl-31796987

ABSTRACT

AIMS/HYPOTHESIS: During the onset of type 2 diabetes, excessive dietary intake of saturated NEFA and fructose lead to impaired insulin production and secretion by insulin-producing pancreatic beta cells. The majority of data on the deleterious effects of lipids on functional beta cell mass were obtained either in vivo in rodent models or in vitro using rodent islets and beta cell lines. Translating data from rodent to human beta cells remains challenging. Here, we used the human beta cell line EndoC-ßH1 and analysed its sensitivity to a lipotoxic and glucolipotoxic (high palmitate with or without high glucose) insult, as a way to model human beta cells in a type 2 diabetes environment. METHODS: EndoC-ßH1 cells were exposed to palmitate after knockdown of genes related to saturated NEFA metabolism. We analysed whether and how palmitate induces apoptosis, stress and inflammation and modulates beta cell identity. RESULTS: EndoC-ßH1 cells were insensitive to the deleterious effects of saturated NEFA (palmitate and stearate) unless stearoyl CoA desaturase (SCD) was silenced. SCD was abundantly expressed in EndoC-ßH1 cells, as well as in human islets and human induced pluripotent stem cell-derived beta cells. SCD silencing induced markers of inflammation and endoplasmic reticulum stress and also IAPP mRNA. Treatment with the SCD products oleate or palmitoleate reversed inflammation and endoplasmic reticulum stress. Upon SCD knockdown, palmitate induced expression of dedifferentiation markers such as SOX9, MYC and HES1. Interestingly, SCD knockdown by itself disrupted beta cell identity with a decrease in mature beta cell markers INS, MAFA and SLC30A8 and decreased insulin content and glucose-stimulated insulin secretion. CONCLUSIONS/INTERPRETATION: The present study delineates an important role for SCD in the protection against lipotoxicity and in the maintenance of human beta cell identity. DATA AVAILABILITY: Microarray data and all experimental details that support the findings of this study have been deposited in in the GEO database with the GSE130208 accession code.


Subject(s)
Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Palmitic Acid/pharmacology , Stearoyl-CoA Desaturase/metabolism , Apoptosis/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Insulin Secretion/drug effects , Proto-Oncogene Proteins c-myc/metabolism , SOX9 Transcription Factor/metabolism , Transcription Factor HES-1/metabolism
5.
Nucleic Acids Res ; 46(19): 10302-10318, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30247717

ABSTRACT

Transfer RNAs (tRNAs) are non-coding RNA molecules essential for protein synthesis. Post-transcriptionally they are heavily modified to improve their function, folding and stability. Intronic polymorphisms in CDKAL1, a tRNA methylthiotransferase, are associated with increased type 2 diabetes risk. Loss-of-function mutations in TRMT10A, a tRNA methyltransferase, are a monogenic cause of early onset diabetes and microcephaly. Here we confirm the role of TRMT10A as a guanosine 9 tRNA methyltransferase, and identify tRNAGln and tRNAiMeth as two of its targets. Using RNA interference and induced pluripotent stem cell-derived pancreatic ß-like cells from healthy controls and TRMT10A-deficient patients we demonstrate that TRMT10A deficiency induces oxidative stress and triggers the intrinsic pathway of apoptosis in ß-cells. We show that tRNA guanosine 9 hypomethylation leads to tRNAGln fragmentation and that 5'-tRNAGln fragments mediate TRMT10A deficiency-induced ß-cell death. This study unmasks tRNA hypomethylation and fragmentation as a hitherto unknown mechanism of pancreatic ß-cell demise relevant to monogenic and polygenic forms of diabetes.


Subject(s)
DNA Methylation , Diabetes Mellitus/genetics , Insulin-Secreting Cells/metabolism , Methyltransferases/genetics , RNA, Transfer/metabolism , Aged , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Differentiation/genetics , Cells, Cultured , DNA Fragmentation , Diabetes Mellitus/metabolism , Genetic Linkage , Humans , Induced Pluripotent Stem Cells/physiology , Insulin-Secreting Cells/physiology , Methyltransferases/deficiency , Methyltransferases/metabolism , Middle Aged , Mutation , Rats
6.
Mol Metab ; 6(9): 1024-1039, 2017 09.
Article in English | MEDLINE | ID: mdl-28951826

ABSTRACT

BACKGROUND: Pancreatic ß cell dysfunction and death are central in the pathogenesis of most if not all forms of diabetes. Understanding the molecular mechanisms underlying ß cell failure is important to develop ß cell protective approaches. SCOPE OF REVIEW: Here we review the role of endoplasmic reticulum stress and dysregulated endoplasmic reticulum stress signaling in ß cell failure in monogenic and polygenic forms of diabetes. There is substantial evidence for the presence of endoplasmic reticulum stress in ß cells in type 1 and type 2 diabetes. Direct evidence for the importance of this stress response is provided by an increasing number of monogenic forms of diabetes. In particular, mutations in the PERK branch of the unfolded protein response provide insight into its importance for human ß cell function and survival. The knowledge gained from different rodent models is reviewed. More disease- and patient-relevant models, using human induced pluripotent stem cells differentiated into ß cells, will further advance our understanding of pathogenic mechanisms. Finally, we review the therapeutic modulation of endoplasmic reticulum stress and signaling in ß cells. MAJOR CONCLUSIONS: Pancreatic ß cells are sensitive to excessive endoplasmic reticulum stress and dysregulated eIF2α phosphorylation, as indicated by transcriptome data, monogenic forms of diabetes and pharmacological studies. This should be taken into consideration when devising new therapeutic approaches for diabetes.


Subject(s)
Endoplasmic Reticulum Stress/physiology , Eukaryotic Initiation Factor-2/metabolism , Insulin-Secreting Cells/metabolism , Animals , Apoptosis , Cell Death , Diabetes Mellitus/metabolism , Endoplasmic Reticulum/metabolism , Humans , Phosphorylation , Signal Transduction , Unfolded Protein Response , eIF-2 Kinase/metabolism
7.
Tissue Eng Part A ; 22(13-14): 971-84, 2016 07.
Article in English | MEDLINE | ID: mdl-27329070

ABSTRACT

Human-induced pluripotent stem cell (hiPSC)-derived hepatocytes are anticipated as important surrogates for primary human hepatocytes in applications ranging from basic research to drug discovery and regenerative medicine. Although methods for differentiating hepatocyte-like cells (HLCs) from hiPSCs have developed remarkably, the limited yield of fully functional HLCs is still a major obstacle to their utility. A three-dimensional (3D) culture environment could improve the in vitro hepatic maturation of HLCs. Here we compare 3D hydrogel models of hiPSC-derived HLCs in agarose microwells (3D Petri Dish; 3DPD), nanofibrillar cellulose hydrogels (Growdex; 3DNFC), or animal extracellular matrix-based hydrogels (3D Matrigel; 3DMG). In all the tested 3D biomaterial systems, HLCs formed aggregates. In comparison with two-dimensional monolayer culture, 3DPD and 3DMG models showed both phenotypic and functional enhancement in HLCs over 2.5 weeks of 3D culture. Specifically, we found higher hepatocyte-specific gene expression levels and enhanced cytochrome P450 functions. Our work suggests that transferring HLCs into 3D hydrogel systems can expedite the hepatic maturation of HLCs irrespective of the biochemical nature of the 3D hydrogel. Both plant-based nonembedding and animal-based embedding 3D hydrogel models enhanced the maturation.


Subject(s)
Cell Differentiation , Hepatocytes/metabolism , Hydrogels/chemistry , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Hepatocytes/cytology , Humans , Induced Pluripotent Stem Cells/cytology
8.
PLoS One ; 10(7): e0134551, 2015.
Article in English | MEDLINE | ID: mdl-26230325

ABSTRACT

Wnt/beta-catenin signaling plays a central role in guiding the differentiation of the posterior parts of the primitive gut tube into intestinal structures in vivo and some studies suggest that FGF4 is another crucial factor for intestinal development. The aim of this study was to define the effects of Wnt and FGF4 on intestinal commitment in vitro by establishing conditions for differentiation of human pluripotent stem cells (hPSC) into posterior endoderm (hindgut) and further to self-renewing intestinal-like organoids. The most prominent induction of the well-established intestinal marker gene CDX2 was achieved when hPSC-derived definitive endoderm cells were treated with Wnt agonist molecule CHIR99021 during differentiation to hindgut. FGF4 was found to be dispensable during intestinal commitment, but it had an early role in repressing development towards the hepatic lineage. When hindgut stage cells were further cultured in 3D, they formed self-renewing organoid structures containing all major intestinal cell types even without exogenous R-spondin1 (RSPO1), a crucial factor for the culture of epithelial organoids derived from adult intestine. This may be explained by the presence of a mesenchymal compartment in the hPSC-derived organoids. Addition of WNT3A increased the expression of the Paneth cell marker Lysozyme in hPSC-derived organoid cultures, whereas FGF4 inhibited both the formation and maturation of intestinal-like organoids. Similar hindgut and organoid cultures were established from human induced pluripotent stem cells, implying that this approach can be used to create patient-specific intestinal tissue models for disease modeling in vitro.


Subject(s)
Cell Differentiation , Fibroblast Growth Factor 4/administration & dosage , Intestines/cytology , Pluripotent Stem Cells/cytology , Thrombospondins/administration & dosage , CDX2 Transcription Factor , Cell Line , Gene Expression/drug effects , Homeodomain Proteins/genetics , Humans , Intestinal Mucosa/metabolism , Pluripotent Stem Cells/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology
9.
PLoS One ; 10(3): e0116668, 2015.
Article in English | MEDLINE | ID: mdl-25822230

ABSTRACT

Small RNA molecules, including microRNAs (miRNAs), play critical roles in regulating pluripotency, proliferation and differentiation of embryonic stem cells. miRNA-offset RNAs (moRNAs) are similar in length to miRNAs, align to miRNA precursor (pre-miRNA) loci and are therefore believed to derive from processing of the pre-miRNA hairpin sequence. Recent next generation sequencing (NGS) studies have reported the presence of moRNAs in human neurons and cancer cells and in several tissues in mouse, including pluripotent stem cells. In order to gain additional knowledge about human moRNAs and their putative development-related expression, we applied NGS of small RNAs in human embryonic stem cells (hESCs) and fibroblasts. We found that certain moRNA isoforms are notably expressed in hESCs from loci coding for stem cell-selective or cancer-related miRNA clusters. In contrast, we observed only sparse moRNAs in fibroblasts. Consistent with earlier findings, most of the observed moRNAs derived from conserved loci and their expression did not appear to correlate with the expression of the adjacent miRNAs. We provide here the first report of moRNAs in hESCs, and their expression profile in comparison to fibroblasts. Moreover, we expand the repertoire of hESC miRNAs. These findings provide an expansion on the known repertoire of small non-coding RNA contents in hESCs.


Subject(s)
Gene Expression , Human Embryonic Stem Cells/metabolism , MicroRNAs/genetics , RNA, Small Untranslated/genetics , Base Sequence , Binding Sites , Cell Line , Computational Biology , Gene Expression Profiling , Gene Library , High-Throughput Nucleotide Sequencing , Humans , MicroRNAs/chemistry , Molecular Sequence Annotation , Molecular Sequence Data , RNA, Small Untranslated/chemistry , Sequence Alignment
10.
Duodecim ; 130(19): 1973-80, 2014.
Article in Finnish | MEDLINE | ID: mdl-25558618

ABSTRACT

The pancreas and the liver are developmentally closely connected with each other.The development of stem cell technology has enabled the production of functional pancreatic endocrine cells and hepatocytes from pluripotent human stem cells. The differentiation of cells takes place by mimicking the events of developmental biology on a cell culture dish. The research is aiming at the development of cell replacement therapy for diabetes and hepatic insufficiency. Transplantations of islet cells have proven the possibilities of this strategy as a replacement of insulin therapy. Although there are promising initial clinical observations on hepatocyte transplantation, the limited growth capacity of these cells restricts the efficiency of the treatment.


Subject(s)
Liver Diseases/therapy , Pancreatic Diseases/therapy , Stem Cell Transplantation/methods , Cell Differentiation , Chronic Disease , Diabetes Mellitus/therapy , Hepatocytes/cytology , Humans , Islets of Langerhans/cytology
11.
PLoS One ; 8(10): e76205, 2013.
Article in English | MEDLINE | ID: mdl-24098444

ABSTRACT

Correct interactions with extracellular matrix are essential to human pluripotent stem cells (hPSC) to maintain their pluripotent self-renewal capacity during in vitro culture. hPSCs secrete laminin 511/521, one of the most important functional basement membrane components, and they can be maintained on human laminin 511 and 521 in defined culture conditions. However, large-scale production of purified or recombinant laminin 511 and 521 is difficult and expensive. Here we have tested whether a commonly available human choriocarcinoma cell line, JAR, which produces high quantities of laminins, supports the growth of undifferentiated hPSCs. We were able to maintain several human pluripotent stem cell lines on decellularized matrix produced by JAR cells using a defined culture medium. The JAR matrix also supported targeted differentiation of the cells into neuronal and hepatic directions. Importantly, we were able to derive new human induced pluripotent stem cell (hiPSC) lines on JAR matrix and show that adhesion of the early hiPSC colonies to JAR matrix is more efficient than to matrigel. In summary, JAR matrix provides a cost-effective and easy-to-prepare alternative for human pluripotent stem cell culture and differentiation. In addition, this matrix is ideal for the efficient generation of new hiPSC lines.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Cell Culture Techniques , Cell Differentiation , Cell Line , Choriocarcinoma/genetics , Choriocarcinoma/metabolism , Choriocarcinoma/pathology , Embryonic Stem Cells , Extracellular Matrix/metabolism , Feeder Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Laminin/metabolism , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/metabolism
12.
Exp Cell Res ; 319(17): 2535-44, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23954819

ABSTRACT

Activin/Nodal and Wnt signaling are known to play important roles in the regional specification of endoderm. Here we have investigated the effect of the length of stimulation with Activin A plus Wnt3a on the development of hepatic and pancreatic progenitors from the definitive endoderm (DE) cells derived from human pluripotent stem cells (hPSC). We show that DE-cells derived from hPSC with 3 days high Activin A and Wnt3a treatment were able to differentiate further into both tested endodermal lineages. When prolonging the DE-induction protocol from 3 to 5 or 7 days, almost pure DE-marker positive cell populations were obtained. However, these cells had an impaired pancreatic differentiation capacity, while they still developed into hepatocyte-like cells. Further propagation of the DE-cells in the presence of Wnt3a and Activin A led to the complete loss of differentiation capacity into hepatic or pancreatic lineages. When Wnt3a was removed after 24h from the initiation of the differentiation, the cells were able to differentiate into PDX1+/NKX6.1+ pancreatic progenitors even with longer DE induction time while efficiency of hepatic differentiation was lower. Our results suggest that both the length and the timing of Wnt3a treatment during DE induction are crucial for the final differentiation outcome. Although it is possible to derive apparently pure DE cells with prolonged Activin A/Wnt-stimulation, their progenitor capacity is restricted to a limited time window.


Subject(s)
Activins/pharmacology , Cell Differentiation/drug effects , Endoderm/cytology , Wnt3A Protein/pharmacology , Cell Lineage , Embryonic Induction , Hepatocytes/cytology , Hepatocytes/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Pancreas/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
13.
Stem Cells Transl Med ; 2(2): 83-93, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23341440

ABSTRACT

Functional hepatocytes, cardiomyocytes, neurons, and retinal pigment epithelial (RPE) cells derived from human embryonic stem cells (hESCs) or human induced pluripotent stem cells (hiPSCs) could provide a defined and renewable source of human cells relevant for cell replacement therapies, drug discovery, toxicology testing, and disease modeling. In this study, we investigated the differences between the differentiation potentials of three hESC lines, four retrovirally derived hiPSC lines, and one hiPSC line derived with the nonintegrating Sendai virus technology. Four independent protocols were used for hepatocyte, cardiomyocyte, neuronal, and RPE cell differentiation. Overall, cells differentiated from hESCs and hiPSCs showed functional similarities and similar expression of genes characteristic of specific cell types, and differences between individual cell lines were also detected. Reactivation of transgenic OCT4 was detected specifically during RPE differentiation in the retrovirally derived lines, which may have affected the outcome of differentiation with these hiPSCs. One of the hiPSC lines was inferior in all directions, and it failed to produce hepatocytes. Exogenous KLF4 was incompletely silenced in this cell line. No transgene expression was detected in the Sendai virus-derived hiPSC line. These findings highlight the problems related to transgene expression in retrovirally derived hiPSC lines.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Gene Silencing , Pluripotent Stem Cells/cytology , Transduction, Genetic/methods , Cell Line , Embryonic Stem Cells/physiology , Hepatocytes/cytology , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Myocytes, Cardiac/cytology , Nerve Net/physiology , Neurons/cytology , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/physiology , Retinal Pigment Epithelium/cytology , Sendai virus/genetics , Transgenes/genetics
14.
Stem Cells Dev ; 22(5): 707-16, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23106381

ABSTRACT

Lectins are carbohydrate-binding proteins, which occur ubiquitously in nature and are abundant in all living organisms from bacteria to mammals. They have several biological functions among which cell adhesion is well known and characterized. Based on the characterization of the glycome of human embryonic stem cells (hESCs), we have investigated the properties of glycan-binding lectins as a novel class of culture support matrices supporting hESC culture. We report that an Erythrina cristagalli lectin (agglutinin) (ECA) matrix supported the undifferentiated growth and significantly increased the plating efficiency of both hESC and human induced pluripotent stem cells when used in conjunction with pinacidil, an antihypertensive drug with ROCK inhibition activity. As a matrix, ECA maintained pluripotency, robust proliferation with a normal karyotype, and the ability to differentiate both in vitro and in vivo. Therefore, our findings indicate that lectins are potential candidates for design of culture and differentiation methods, and that ECA is a potent simple defined matrix for human pluripotent stem cells.


Subject(s)
Embryonic Stem Cells/cytology , Erythrina , Hepatocytes/cytology , Induced Pluripotent Stem Cells/cytology , Plant Lectins , Pluripotent Stem Cells/cytology , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Embryonic Stem Cells/metabolism , Hemagglutinins , Humans , Pinacidil/pharmacology , rho-Associated Kinases/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...