Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Commun Biol ; 4(1): 802, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34183774

ABSTRACT

Regeneration of the testis from pluripotent stem cells is a real challenge, reflecting the complexity of the interaction of germ cells and somatic cells. Here we report the generation of testicular somatic cell-like cells (TesLCs) including Sertoli cell-like cells (SCLCs) from mouse embryonic stem cells (ESCs) in xeno-free culture. We find that Nr5a1/SF1 is critical for interaction between SCLCs and PGCLCs. Intriguingly, co-culture of TesLCs with epiblast-like cells (EpiLCs), rather than PGCLCs, results in self-organised aggregates, or testicular organoids. In the organoid, EpiLCs differentiate into PGCLCs or gonocyte-like cells that are enclosed within a seminiferous tubule-like structure composed of SCLCs. Furthermore, conditioned medium prepared from TesLCs has a robust inducible activity to differentiate EpiLCs into PGCLCs. Our results demonstrate conditions for in vitro reconstitution of a testicular environment from ESCs and provide further insights into the generation of sperm entirely in xeno-free culture.


Subject(s)
Embryonic Stem Cells/cytology , Germ Layers/cytology , Spermatozoa/cytology , Testis/cytology , Animals , Cell Differentiation , Coculture Techniques , Male , Mice , Organoids/cytology , Transcriptome
2.
J Endocrinol ; 233(3): 217-227, 2017 06.
Article in English | MEDLINE | ID: mdl-28348116

ABSTRACT

Type 2 diabetes (T2DM) is associated with pancreatic islet dysfunction. Loss of ß-cell identity has been implicated via dedifferentiation or conversion to other pancreatic endocrine cell types. How these transitions contribute to the onset and progression of T2DM in vivo is unknown. The aims of this study were to determine the degree of epithelial-to-mesenchymal transition occurring in α and ß cells in vivo and to relate this to diabetes-associated (patho)physiological conditions. The proportion of islet cells expressing the mesenchymal marker vimentin was determined by immunohistochemistry and quantitative morphometry in specimens of pancreas from human donors with T2DM (n = 28) and without diabetes (ND, n = 38) and in non-human primates at different stages of the diabetic syndrome: normoglycaemic (ND, n = 4), obese, hyperinsulinaemic (HI, n = 4) and hyperglycaemic (DM, n = 8). Vimentin co-localised more frequently with glucagon (α-cells) than with insulin (ß-cells) in the human ND group (1.43% total α-cells, 0.98% total ß-cells, median; P < 0.05); these proportions were higher in T2DM than ND (median 4.53% α-, 2.53% ß-cells; P < 0.05). Vimentin-positive ß-cells were not apoptotic, had reduced expression of Nkx6.1 and Pdx1, and were not associated with islet amyloidosis or with bihormonal expression (insulin + glucagon). In non-human primates, vimentin-positive ß-cell proportion was larger in the diabetic than the ND group (6.85 vs 0.50%, medians respectively, P < 0.05), but was similar in ND and HI groups. In conclusion, islet cell expression of vimentin indicates a degree of plasticity and dedifferentiation with potential loss of cellular identity in diabetes. This could contribute to α- and ß-cell dysfunction in T2DM.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Secreting Cells/metabolism , Insulin-Secreting Cells/metabolism , Vimentin/metabolism , Animals , Case-Control Studies , Cells, Cultured , Humans , Hyperinsulinism/metabolism , Hyperinsulinism/veterinary , Macaca fascicularis , Macaca mulatta
3.
Stem Cell Reports ; 7(4): 764-776, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27641648

ABSTRACT

Wnt signaling is a key regulator of vertebrate heart development; however, specific roles for human cardiomyocyte development remain uncertain. Here we use human embryonic stem cells (hESCs) to analyze systematically in human cardiomyocyte development the expression of endogenous Wnt signaling components, monitor pathway activity, and dissect stage-specific requirements for canonical and noncanonical Wnt signaling mechanisms using small-molecule inhibitors. Our analysis suggests that WNT3 and WNT8A, via FZD7 and canonical signaling, regulate BRACHYURY expression and mesoderm induction; that WNT5A/5B, via ROR2 and noncanonical signaling, regulate MESP1 expression and cardiovascular development; and that later in development WNT2, WNT5A/5B, and WNT11, via FZD4 and FZD6, regulate functional cardiomyocyte differentiation via noncanonical Wnt signaling. Our findings confirm in human development previously proposed roles for canonical Wnt signaling in sequential stages of vertebrate cardiomyogenesis, and identify more precise roles for noncanonical signaling and for individual Wnt signal and Wnt receptor genes in human cardiomyocyte development.


Subject(s)
Cell Differentiation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Organogenesis , Wnt Signaling Pathway , Biomarkers , Cell Differentiation/genetics , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mesoderm/embryology , Mesoderm/metabolism , beta Catenin/metabolism
4.
PLoS One ; 11(5): e0156204, 2016.
Article in English | MEDLINE | ID: mdl-27243814

ABSTRACT

Transcription factor mediated lineage reprogramming of human pancreatic exocrine tissue could conceivably provide an unlimited supply of islets for transplantation in the treatment of diabetes. Exocrine tissue can be efficiently reprogrammed to islet-like cells using a cocktail of transcription factors: Pdx1, Ngn3, MafA and Pax4 in combination with growth factors. We show here that overexpression of exogenous Pax4 in combination with suppression of the endogenous transcription factor ARX considerably enhances the production of functional insulin-secreting ß-like cells with concomitant suppression of α-cells. The efficiency was further increased by culture on laminin-coated plates in media containing low glucose concentrations. Immunocytochemistry revealed that reprogrammed cultures were composed of ~45% islet-like clusters comprising >80% monohormonal insulin+ cells. The resultant ß-like cells expressed insulin protein levels at ~15-30% of that in adult human islets, efficiently processed proinsulin and packaged insulin into secretory granules, exhibited glucose responsive insulin secretion, and had an immediate and prolonged effect in normalising blood glucose levels upon transplantation into diabetic mice. We estimate that approximately 3 billion of these cells would have an immediate therapeutic effect following engraftment in type 1 diabetes patients and that one pancreas would provide sufficient tissue for numerous transplants.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Cellular Reprogramming Techniques/methods , Diabetes Mellitus, Experimental/therapy , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/transplantation , Paired Box Transcription Factors/metabolism , Pancreas, Exocrine/cytology , Transcription Factors/genetics , Adult , Animals , Cells, Cultured , Female , Glucose/metabolism , Humans , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, SCID , Pancreas, Exocrine/metabolism , Proinsulin/metabolism , RNA Interference , RNA, Small Interfering/genetics , Streptozocin , Young Adult
5.
PLoS One ; 10(10): e0140352, 2015.
Article in English | MEDLINE | ID: mdl-26457418

ABSTRACT

A replenishable source of insulin-producing cells has the potential to cure type 1 diabetes. Attempts to culture and expand pancreatic ß-cells in vitro have resulted in their transition from insulin-producing epithelial cells to mesenchymal stromal cells (MSCs) with high proliferative capacity but devoid of any hormone production. The aim of this study was to determine whether the transcription factor Krüppel-like factor 4 (KLF4), could induce a mesenchymal-to-epithelial transition (MET) of the cultured cells. Islet-enriched pancreatic cells, allowed to dedifferentiate and expand in adherent cell culture, were transduced with an adenovirus containing KLF4 (Ad-Klf4). Cells were subsequently analysed for changes in cell morphology by light microscopy, and for the presence of epithelial and pancreatic markers by immunocytochemistry and quantitative RT/PCR. Infection with Ad-Klf4 resulted in morphological changes, down-regulation of mesenchymal markers, and re-expression of both epithelial and pancreatic cell markers including insulin and transcription factors specific to ß-cells. This effect was further enhanced by culturing cells in suspension. However, the effects of Ad-KLf4 were transient and this was shown to be due to increased apoptosis in Klf4-expressing cells. Klf4 has been recently identified as a pioneer factor with the ability to modulate the structure of chromatin and enhance reprogramming/transdifferentiation. Our results show that Klf4 may have a role in the redifferentiation of expanded pancreatic cells in culture, but before this can be achieved the off-target effects that result in increased apoptosis would need to be overcome.


Subject(s)
Cell Transdifferentiation/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression , Kruppel-Like Transcription Factors/genetics , Pancreas/cytology , Pancreas/metabolism , Adipogenesis/genetics , Biomarkers , Cell Communication/genetics , Cell Culture Techniques , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Osteogenesis/genetics
6.
Diabetologia ; 58(6): 1300-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25810037

ABSTRACT

AIMS/HYPOTHESIS: Type 1 diabetes complicated by hypoglycaemia is prevalent in socioeconomically deprived populations. Islet transplantation is of proven efficacy in type 1 diabetes complicated by hypoglycaemia, but it is not known if nationally funded programmes reach the socioeconomically deprived. Our aim was to determine: (1) socioeconomic indices in participants referred to our nationally funded programme; and (2) if metabolic outcomes in our transplant recipients were improved. METHODS: Participants referred (n = 106) and receiving transplants (n = 18; 32 infusions) were examined with respect to socioeconomic status (deprivation category score) and their ability to work and drive. In participants followed for ≥12 months after transplantation, metabolic and anthropometric measurements (n = 14) were recorded pre- and post-transplant (assessed ~1, ~3, ~6 and ~12 months with mixed-meal tolerance tests and 6 day continuous glucose monitoring assessments). Donor data was also examined. RESULTS: There was a greater prevalence of socioeconomic deprivation in referred and transplant recipients than the general population (p < 0.05). Of the transplant recipients, 73% were socioeconomically deprived, 88% did not hold a driver's license and 94% had reduced ability to work (all p < 0.01 vs referred participants). Donors were predominantly obese and included circulatory death donors. At 12 months, 93% of participants who had received transplants had graft function, diminished frequency of hypoglycaemia (10 [4-11] vs 0 [0-2] hypoglycaemic episodes/week), improved awareness of hypoglycaemia (Gold score 7 [5-7] vs 1 [1-2]) and glycaemic control (HbA1c: 7.9% [7.2-8.5%]; 63 [55-69] mmol/mol vs 7.2% [6.8-7.5%]; 55 [51-58] mmol/mol), diminished glycaemic lability and decreased central adiposity (all p < 0.05). CONCLUSIONS/INTERPRETATION: A nationally funded islet transplant programme reaches the socioeconomically deprived and outcomes are significantly improved in this group.


Subject(s)
Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/therapy , Islets of Langerhans Transplantation/methods , Adiposity , Adult , Anthropometry , Automobile Driving , Blood Glucose/analysis , Diabetes Mellitus, Type 1/complications , Female , Financing, Government , Health Services Accessibility , Humans , Hypoglycemia/complications , Hypoglycemia/epidemiology , Hypoglycemia/therapy , Islets of Langerhans Transplantation/economics , Male , Middle Aged , Social Class , Tissue Donors , Transplant Recipients , United Kingdom , Young Adult
7.
J Mol Biol ; 425(22): 4206-22, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23871836

ABSTRACT

Using high-throughput sequencing, we have mapped sequence-directed nucleosome positioning in vitro on four plasmid DNAs containing DNA fragments derived from the genomes of sheep, drosophila, human and yeast. Chromatins were prepared by reconstitution using chicken, frog and yeast core histones. We also assembled yeast chromatin in which histone H3 was replaced by the centromere-specific histone variant, Cse4. The positions occupied by recombinant frog and native chicken histones were found to be very similar. In contrast, nucleosomes containing the canonical yeast octamer or, in particular, the Cse4 octamer were assembled at distinct populations of locations, a property that was more apparent on particular genomic DNA fragments. The factors that may contribute to this variation in nucleosome positioning and the implications of the behavior are discussed.


Subject(s)
Histones/metabolism , Nucleosomes/metabolism , Animals , Anura , Binding Sites , Chickens , DNA/chemistry , DNA/metabolism , Histones/chemistry , Nucleosomes/chemistry , Plasmids/chemistry , Plasmids/metabolism , Protein Binding , Yeasts/genetics , Yeasts/metabolism
8.
Diabetes ; 62(8): 2821-33, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23610058

ABSTRACT

Because of the lack of tissue available for islet transplantation, new sources of ß-cells have been sought for the treatment of type 1 diabetes. The aim of this study was to determine whether the human exocrine-enriched fraction from the islet isolation procedure could be reprogrammed to provide additional islet tissue for transplantation. The exocrine-enriched cells rapidly dedifferentiated in culture and grew as a mesenchymal monolayer. Genetic lineage tracing confirmed that these mesenchymal cells arose, in part, through a process of epithelial-to-mesenchymal transitioning (EMT). A protocol was developed whereby transduction of these mesenchymal cells with adenoviruses containing Pdx1, Ngn3, MafA, and Pax4 generated a population of cells that were enriched in glucagon-secreting α-like cells. Transdifferentiation or reprogramming toward insulin-secreting ß-cells was enhanced, however, when using unpassaged cells in combination with inhibition of EMT by inclusion of Rho-associated kinase (ROCK) and transforming growth factor-ß1 inhibitors. Resultant cells were able to secrete insulin in response to glucose and on transplantation were able to normalize blood glucose levels in streptozotocin diabetic NOD/SCID mice. In conclusion, reprogramming of human exocrine-enriched tissue can be best achieved using fresh material under conditions whereby EMT is inhibited, rather than allowing the culture to expand as a mesenchymal monolayer.


Subject(s)
Cell Differentiation/physiology , Epithelial-Mesenchymal Transition/physiology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Pancreas, Exocrine/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Glucose/pharmacology , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred NOD , Mice, SCID , Pancreas, Exocrine/drug effects , rho-Associated Kinases/metabolism
9.
PLoS One ; 7(5): e36481, 2012.
Article in English | MEDLINE | ID: mdl-22563503

ABSTRACT

Protein transduction domains (PTDs), such as the HIV1-TAT peptide, have been previously used to promote the uptake of proteins into a range of cell types, including stem cells. Here we generated pancreatic transcription factors containing PTD sequences and administered these to endoderm enriched mouse embryonic stem (ES) cells under conditions that were designed to mimic the pattern of expression of these factors in the developing pancreas. The ES cells were first cultured as embryoid bodies and treated with Activin A and Bone morphogenetic protein 4 (BMP4) to promote formation of definitive endoderm. Cells were subsequently plated as a monolayer and treated with different combinations of the modified recombinant transcription factors Pdx1 and MafA. The results demonstrate that each transcription factor was efficiently taken up by the cells, where they were localized in the nuclei. RT-qPCR was used to measure the expression levels of pancreatic markers. After the addition of Pdx1 alone for a period of five days, followed by the combination of Pdx1 and TAT-MafA in a second phase, up-regulation of insulin 1, insulin 2, Pdx1, Glut2, Pax4 and Nkx6.1 was observed. As assessed by immunocytochemistry, double positive insulin and Pdx1 cells were detected in the differentiated cultures. Although the pattern of pancreatic markers expression in these cultures was comparable to that of a mouse transformed ß-cell line (MIN-6) and human islets, the expression levels of insulin observed in the differentiated ES cell cultures were several orders of magnitude lower. This suggests that, although PTD-TFs may prove useful in studying the role of exogenous TFs in the differentiation of ES cells towards islets and other pancreatic lineages, the amount of insulin generated is well below that required for therapeutically useful cells.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/metabolism , Islets of Langerhans/metabolism , Transcription Factors/metabolism , Activins/pharmacology , Animals , Blotting, Western , Bone Morphogenetic Protein 4/pharmacology , Cell Line , Cell Line, Tumor , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Endoderm/cytology , Endoderm/metabolism , Gene Expression/drug effects , Glucose Transporter Type 2/genetics , Glucose Transporter Type 2/metabolism , HeLa Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Insulin/genetics , Insulin/metabolism , Islets of Langerhans/cytology , Maf Transcription Factors, Large/genetics , Maf Transcription Factors, Large/metabolism , Mice , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Pancreas/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics
10.
Opt Lett ; 37(7): 1247-9, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22466210

ABSTRACT

We present a new class of interferometer system that is capable of simultaneous measurement of absolute position and rotation in all six degrees of freedom (DOF) with nanometer precision. This novel capability is due to the employment of a system of interference fringes that is not periodic. One of the key strengths offered by this new approach is that the absolute position of the system can be determined with a single measurement, rather than by counting fringes during displacement from a known location. The availability of a simultaneous measurement of all six DOF eliminates many problems associated with conventional interferometry.

11.
Mol Cell Endocrinol ; 361(1-2): 40-50, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-22484460

ABSTRACT

The number of adipocyte progenitors is determined early in foetal and neonatal development in a process which may be altered by gender and excess nutrient intake, and which in turn determines fat mass in adulthood and the risk of developing obesity. Here we investigate the hypothesis that excess nutrients, in this case the long chain fatty acid palmitate, can program differentiating stem cells towards white fat lineages. The experiments were performed on mouse embryonic stem cells in chemically defined media (CDM) supplemented with bone morphogenetic protein 4 (BMP4) and all trans-retinoic acid (RA). Subsequent treatment for 21 days with palmitate not only promoted the expression of adipocyte markers and monolocular lipid deposition as observed by RT/QPCR and immunocytochemistry, but also stimulated a considerable enrichment in adipocytes as measured by flow cytometry and a lipolytic response to catecholamines. Palmitate increased protein levels of adiponectin that is preferentially expressed in subcutaneous fat, while inhibiting IGFBP2 and IGFBP3 that are associated with visceral fat. In keeping with this finding, palmitate also increased expression of the subcutaneous markers Shox2 and Twist1 and oestrogenising enzymes. Collectively, these results suggest that palmitate induces differentiation towards subcutaneous fat and that this could occur through its oestrogenising effects on the preadipocyte, suggesting a role for palmitate in programming fat development towards a metabolically favourable profile.


Subject(s)
Adipocytes, White/cytology , Adipocytes, White/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Palmitates/pharmacology , Adipocytes, White/metabolism , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 4/pharmacology , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation/genetics , Cell Line , Dehydroepiandrosterone/pharmacology , Embryonic Stem Cells/metabolism , Gene Expression Regulation/drug effects , Gonadal Steroid Hormones/metabolism , Methylamines/pharmacology , Mice , PPAR gamma/metabolism , Propionates/pharmacology , Time Factors , Tretinoin/pharmacology
12.
Mol Cell Endocrinol ; 358(1): 69-80, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22429991

ABSTRACT

The AR42J-B13 rat pancreatic acinar cell line was used to identify pancreatic transcription factors and exogenous growth factors (GFs) that might facilitate the reprogramming of exocrine cells into islets. Adenoviruses were used to induce exogenous expression of the pancreatic transcription factors (TFs) Pdx1, MafA, Ngn3 and Pax4. Individually Pdx1, MafA and Pax4 had no effect on the expression of endocrine markers, whilst adeno-Ngn3 on its own increased the expression of Pax4, Ngn3 and NeuroD. In combination the four TFs had a significant effect on the expression of insulin 1 and 2 that was associated with a change in cell morphology from a rounded to a spindle-like shape. Amongst a range of growth factors, Betacellulin and Nicotinamide were shown to enhance the effects of the four TFs. The presence of adeno-Pax4 in the differentiation cocktail was important in limiting the expression of glucagon and in generating glucose sensitive insulin secretion. Further experiments asked whether the adenoviral TFs could be replaced by protein transduction domain (PTD)-containing TFs. The results showed that the PTD-TFs could mimic in part the effects of the adeno-TFs, but the resultant cells did not undergo the important morphological change associated with differentiation to endocrine lineages and levels of endogenous markers were very much lower. In summary, the results describe a cocktail of four TFs and two GFs that can be used to induce formation of glucose sensitive insulin secreting cells from ARJ42 cells, and demonstrate that it would be difficult to replace adenoviral transduction with PTD-TFS.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Paired Box Transcription Factors/metabolism , Trans-Activators/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Betacellulin , Cell Differentiation , Cell Line , Glucagon/biosynthesis , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/biosynthesis , Niacinamide/metabolism , Pancreas/cytology , Pancreas/metabolism , Rats
13.
Mol Cell Endocrinol ; 328(1-2): 80-6, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20674663

ABSTRACT

In the mouse the developing pancreas is controlled by contact with, and signalling molecules secreted from, surrounding cells. These factors are best studied using explant cultures of embryonic tissue. The present study was undertaken to determine whether embryonic stem (ES) cells could be used as an alternative model in vitro system to investigate the role of cell-cell interactions in the developing pancreas. Transwell culture experiments showed that MIN-6 beta-cells secreted a factor or factors that promoted differentiation of ES cell derived definitive endoderm enriched cells towards a pancreatic fate. Further studies using MIN-6 condition medium showed that the factor(s) involved was restricted to MIN-6 cells, could be concentrated with ammonium sulphate, and was sensitive to heat treatment, suggesting that it was a protein or peptide. Further analyses showed that insulin or proinsulin failed to mimic the effects of the conditioned media. Collectively, these results suggest that beta-cells secrete a factor(s) capable of controlling their own differentiation and maturation. The culture system described here presents unique advantages in the identification and characterisation of these factors.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/drug effects , Endoderm/drug effects , Insulin-Secreting Cells/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Pancreas/physiology , Animals , Cell Culture Techniques , Cell Line , Cell Separation , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Dose-Response Relationship, Drug , Embryoid Bodies/cytology , Embryoid Bodies/drug effects , Embryoid Bodies/physiology , Embryonic Stem Cells/physiology , Endoderm/cytology , Endoderm/physiology , Gene Expression Regulation, Developmental/drug effects , Humans , Insulin-Secreting Cells/physiology , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Pancreas/cytology , Pancreas/drug effects , Pancreas/embryology , Rats
14.
FEBS Lett ; 583(19): 3181-6, 2009 Oct 06.
Article in English | MEDLINE | ID: mdl-19733567

ABSTRACT

An engineered zinc finger protein (eZFP) was isolated from a library based on its ability to activate expression of the endogenous insulin gene in HEK-293 cells. Using a panel of insulin promoter constructs, the eZFP was shown to act through the variable number of tandem repeat (VNTR) region located 365 base pairs upstream of the transcription start site. The eZFP also activated expression of the IGF2 gene that lies close to INS on chromosome 11p15. These results demonstrate that the INSVNTR controls expression of the insulin and IGF2 genes and provide a mechanistic explanation for previous studies that demonstrated an association between INSVNTR genotypes and placental levels of IGF2.


Subject(s)
Epigenesis, Genetic , Insulin-Like Growth Factor II/genetics , Insulin/genetics , Minisatellite Repeats/genetics , Zinc Fingers , Cell Line , Epigenesis, Genetic/drug effects , Gene Expression/drug effects , Humans , Minisatellite Repeats/drug effects , Protein Engineering , Proteins/chemical synthesis , Proteins/isolation & purification , Proteins/pharmacology , Transcription Initiation Site
15.
J Biol Chem ; 284(31): 20738-52, 2009 Jul 31.
Article in English | MEDLINE | ID: mdl-19491401

ABSTRACT

Cold stress in rodents increases the expression of UCP1 and PGC-1alpha in brown and white adipose tissue. We have previously reported that C/EBPbeta specifically binds to the CRE on the proximal Pgc-1alpha promoter and increases forskolin-sensitive Pgc-1alpha and Ucp1 expression in white 3T3-L1 preadipocytes. Here we show that in mice exposed to a cold environment for 24 h, Pgc-1alpha, Ucp1, and C/ebpbeta but not C/ebpalpha or C/ebpdelta expression were increased in BAT. Conversely, expression of the C/EBP dominant negative Chop10 was increased in WAT but not BAT during cold exposure. Reacclimatization of cold-exposed mice to a warm environment for 24 h completely reversed these changes in gene expression. In HIB-1B, brown preadipocytes, forskolin increased expression of Pgc-1alpha, Ucp1, and C/ebpbeta early in differentiation and inhibited Chop10 expression. Employing chromatin immunoprecipitation, we demonstrate that C/EBPbeta, CREB, ATF-2, and CHOP10 are bound to the Pgc-1alpha proximal CRE, but CHOP10 does not bind in HIB-1B cell lysates. Forskolin stimulation and C/EBPbeta overexpression in 3T3-L1 cells increased C/EBPbeta and CREB but displaced ATF-2 and CHOP10 binding to the Pgc-1alpha proximal CRE. Overexpression of ATF-2 and CHOP10 in 3T3-L1 cells decreased Pgc-1alpha transcription. Knockdown of Chop10 in 3T3-L1 cells using siRNA increased Pgc-1alpha transcription, whereas siRNA against C/ebpbeta in HIB-1B cells decreased Pgc-1alpha and Ucp1 expression. We conclude that the increased cAMP stimulation of Pgc-1alpha expression is regulated by the combinatorial effect of transcription factors acting at the CRE on the proximal Pgc-1alpha promoter.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/cytology , Adipose Tissue, White/cytology , Cyclic AMP/pharmacology , Response Elements/genetics , Trans-Activators/genetics , Transcription Factor CHOP/metabolism , Activating Transcription Factor 2/metabolism , Adipocytes/drug effects , Aging/drug effects , Aging/metabolism , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cold Temperature , Cyclic AMP Response Element-Binding Protein/metabolism , Down-Regulation/drug effects , Gene Knockdown Techniques , Humans , Ion Channels/metabolism , Mice , Mitochondrial Proteins/metabolism , Organ Specificity/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Binding/drug effects , Protein Isoforms/metabolism , RNA, Small Interfering/metabolism , Rats , Transcription Factors , Transcription, Genetic/drug effects , Uncoupling Protein 1
16.
Biochem J ; 421(2): e1-4, 2009 Jun 26.
Article in English | MEDLINE | ID: mdl-19552623

ABSTRACT

Regenerative medicine, including cell-replacement strategies, may have an important role in the treatment of Type 1 and Type 2 diabetes, both of which are associated with decreased islet cell mass. To date, significant progress has been made in deriving insulin-secreting beta-like cells from human ES (embryonic stem) cells. However, the cells are not fully differentiated, and there is a long way to go before they could be used as a replenishable supply of insulin-secreting beta-cells for transplantation. For this reason, adult pancreatic stem cells are seen as an alternative source that could be expanded and differentiated ex vivo, or induced to form new islets in situ. In this issue of the Biochemical Journal, Mato et al. used drug selection to purify a population of stellate cells from explant cultures of pancreas from lactating rats. The selected cells express some stem-cell markers and can be grown for over 2 years as a fibroblast-like monolayer. When plated on extracellular matrix, along with a cocktail of growth factors that included insulin, transferrin, selenium and the GLP-1 (glucagon-like peptide-1) analogue exendin-4, the cells differentiated into cells that expressed many of the phenotypic markers characteristic of a beta-cell, and exhibited an insulin-secretory response, albeit weak, to glucose. The ability to purify this cell population opens up the possibility of unravelling the mechanisms that control self-renewal and differentiation of pancreatic cells that share some of the properties of stem cells.


Subject(s)
Insulin-Secreting Cells/cytology , Pancreas/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Humans , Mice , Pancreas/metabolism , Rats
17.
J Biol Chem ; 284(11): 6716-24, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19129193

ABSTRACT

The PrP gene encodes the cellular isoform of the prion protein (PrP(c)) which has been shown to be crucial to the development of transmissible spongiform encephalopathies (TSEs). PrP knock-out mice, which do not express endogenous PrP(c), exhibit resistance to TSE disease. The regulation of PrP gene expression represents, therefore, a crucial factor in the development of TSEs. Two sequence motifs in the PrP promoter (positions -287 to -263 from transcriptional start) were previously reported as being highly conserved, and it was suggested that they represent binding sites for as yet unidentified transcription factors. To test this hypothesis, binding of nuclear proteins was analyzed by electrophoretic mobility shift assays using ovine or murine cells and tissues with radiolabeled DNA probes containing the conserved motif sequences. Specific binding was observed to both motifs, and polymorphic variants of these motifs exhibited differential binding. Two proteins bound to these motifs were identified as the Yin Yang 1 (YY1) (motif 1) and E4BP4 (motif 2) transcription factors. Functional promoter analysis of four different promoter variants revealed that motif 1 (YY1) was associated with inhibitory activity in the context of the PrP promoter, whereas motif 2 (E4BP4) was linked to a slight enhancing activity. This represents the first demonstration of binding of nuclear factors to two highly conserved DNA sequence motifs within mammalian PrP promoters. The action of these factors on the PrP promoter is haplotype-specific, leading us to propose that the prion protein expression pattern and, with it, the distribution of TSE infectivity may be associated with PrP promoter genotype.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , PrPC Proteins/biosynthesis , Response Elements/physiology , YY1 Transcription Factor/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/genetics , HeLa Cells , Humans , Mice , PrPC Proteins/genetics , Prion Diseases/genetics , Prion Diseases/metabolism , Sheep , YY1 Transcription Factor/genetics
18.
Stem Cells ; 27(2): 341-51, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19056911

ABSTRACT

Embryonic stem (ES) cells represent a possible source of islet tissue for the treatment of diabetes. Achieving this goal will require a detailed understanding of how the transcription factor cascade initiated by the homeodomain transcription factor Pdx1 culminates in pancreatic beta-cell development. Here we describe a genetic approach that enables fine control of Pdx1 transcriptional activity during endoderm differentiation of mouse and human ES cell. By activating an exogenous Pdx1VP16 protein in populations of cells enriched in definitive endoderm we show a distinct lineage-dependent requirement for this transcription factor's activity. Mimicking the natural biphasic pattern of Pdx1 expression was necessary to induce an endocrine pancreas-like cell phenotype, in which 30% of the cells were beta-cell-like. Cell markers consistent with the different beta-cell differentiation stages appeared in a sequential order following the natural pattern of pancreatic development. Furthermore, in mouse ES-derived cultures the differentiated beta-like cells secreted C-peptide (insulin) in response to KCl and 3-isobutyl-1-methylxanthine, suggesting that following a natural path of development in vitro represents the best approach to generate functional pancreatic cells. Together these results reveal for the first time a significant effect of the timed expression of Pdx1 on the non-beta-cells in the developing endocrine pancreas. Collectively, we show that this method of in vitro differentiation provides a template for inducing and studying ES cell differentiation into insulin-secreting cells.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Trans-Activators/metabolism , Activins/pharmacology , Amyloid/genetics , Animals , Blotting, Western , Bone Morphogenetic Protein 4/pharmacology , C-Peptide/metabolism , Cell Differentiation/drug effects , Cell Line , Embryonic Stem Cells/drug effects , Flow Cytometry , Gene Expression/drug effects , HeLa Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Humans , Immunohistochemistry , Insulin/genetics , Islet Amyloid Polypeptide , LIM-Homeodomain Proteins , Mice , Polymerase Chain Reaction , Tamoxifen/analogs & derivatives , Tamoxifen/pharmacology , Trans-Activators/genetics , Trans-Activators/physiology , Transcription Factors
19.
Mol Cell Endocrinol ; 294(1-2): 1-9, 2008 Nov 06.
Article in English | MEDLINE | ID: mdl-18687378

ABSTRACT

In recent years major progress has been made in understanding the role of transcription factors in the development of the endocrine pancreas in the mouse. Here we describe how a number of these transcription factors play a role in maintaining the differentiated phenotype of the beta cell, and in the mechanisms that allow the beta cell to adapt to changing metabolic demands that occur throughout life. Amongst these factors, Pdx1 plays a critical role in defining the region of the primitive gut that will form the pancreas, Ngn3 expression drives cells towards an endocrine lineage, and a number of additional proteins including Pdx1, in a second wave of expression, Pax4, NeuroD1/beta2, and MafA act as beta cell differentiation factors. In the mature beta cell Pdx1, MafA, beta2, and Nkx2.2 play important roles in regulating expression of insulin and to some extent other genes responsible for maintaining beta cell function. We emphasise here that data from gene expression studies in rodents seldom map on to the known structure of the corresponding human promoters. In the adult the beta cell is particularly susceptible to autoimmune-mediated attack and to the toxic metabolic milieu associated with over-eating, and utilises a number of these transcription factors in its defence. Pdx1 has anti-apoptotic and proliferative activities that help facilitate the maintenance of beta cell mass, while Ngn3 may be involved in the recruitment of progenitor cells, and Pax4 (and possibly HNF1alpha and Hnf4alpha) in the proliferation of beta cells in the adult pancreas. Other transcription factors with a more widespread pattern of expression that play a role in beta cell survival or proliferation include Foxo1, CREB family members, NFAT, FoxM1, Snail and Asc-2.


Subject(s)
Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Transcription Factors/metabolism , Animals , Cell Survival , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Nuclear Proteins , Regeneration
20.
Biochem Soc Trans ; 36(Pt 3): 363-5, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18481959

ABSTRACT

Obesity is a metabolic disorder, which has been recognized as a global epidemic. It contributes to insulin resistance, the major cause of Type 2 diabetes, as well as to the development of other related diseases. Our basic premise is that a better understanding of how adult stem cells of the pancreas contribute to the maintenance of the pancreatic beta-cell pool against the increased metabolic demands associated with obesity may provide new therapeutic targets for treating diabetes. At the same time, if we knew more about the biology of adipocyte formation, maintenance and deposition in obese individuals, perhaps some control over the adipocyte tissue mass of these individuals would be facilitated and treatment of obesity would become available. Many investigations in the field are therefore aimed at describing how adipocyte stem cells function in the various sites of fat deposition and the extent to which these stem cells contribute to both brown and white adipocytes. Studies on the differentiation of human embryonic stem cells along the pancreatic and adipocyte lineages may therefore better inform approaches to these studies.


Subject(s)
Metabolic Diseases/pathology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Diabetes Mellitus, Type 2/pathology , Humans , Obesity/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...