Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Metab ; 16(4): 449-61, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23000401

ABSTRACT

Reactive oxygen species (ROS) contribute to target-cell damage in inflammatory and iron-overload diseases. Little is known about iron transport regulation during inflammatory attack. Through a combination of in vitro and in vivo studies, we show that the proinflammatory cytokine IL-1ß induces divalent metal transporter 1 (DMT1) expression correlating with increased ß cell iron content and ROS production. Iron chelation and siRNA and genetic knockdown of DMT1 expression reduce cytokine-induced ROS formation and cell death. Glucose-stimulated insulin secretion in the absence of cytokines in Dmt1 knockout islets is defective, highlighting a physiological role of iron and ROS in the regulation of insulin secretion. Dmt1 knockout mice are protected against multiple low-dose streptozotocin and high-fat diet-induced glucose intolerance, models of type 1 and type 2 diabetes, respectively. Thus, ß cells become prone to ROS-mediated inflammatory damage via aberrant cellular iron metabolism, a finding with potential general cellular implications.


Subject(s)
Apoptosis/drug effects , Cation Transport Proteins/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-1beta/pharmacology , Iron/metabolism , Reactive Oxygen Species/metabolism , Animals , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Diabetes Mellitus, Experimental , Diet, High-Fat , Glucose Intolerance , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Insulin-Secreting Cells/cytology , Mice , Mice, Knockout , Models, Biological , RNA Interference , RNA, Small Interfering/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
2.
J Biol Chem ; 287(21): 17269-17280, 2012 May 18.
Article in English | MEDLINE | ID: mdl-22457355

ABSTRACT

In vitro expansion of ß-cells from adult human pancreatic islets would overcome donor ß-cell shortage for cell replacement therapy for diabetes. Using a ß-cell-specific labeling system we have shown that ß-cell expansion is accompanied by dedifferentiation resembling epithelial-mesenchymal transition and loss of insulin expression. Epigenetic analyses indicate that key ß-cell genes maintain open chromatin structure in expanded ß-cell-derived (BCD) cells, although they are not transcribed. In the developing pancreas important cell-fate decisions are regulated by NOTCH receptors, which signal through the Hairy and Enhancer of Split 1 (HES1) transcription regulator. We have reported that BCD cell dedifferentiation and proliferation in vitro correlate with reactivation of the NOTCH pathway. Inhibition of HES1 expression using shRNA during culture initiation results in reduced ß-cell replication and dedifferentiation, suggesting that HES1 inhibition may also affect BCD cell redifferentiation following expansion. Here, we used HES1 shRNA to down-regulate HES1 expression in expanded human BCD cells, showing that HES1 inhibition is sufficient to induce BCD cell redifferentiation, as manifested by a significant increase in insulin expression. Combined treatment with HES1 shRNA, cell aggregation in serum-free medium, and a mixture of soluble factors further stimulated the redifferentiation of BCD cells. In vivo analyses demonstrated the ability of the redifferentiated cells to replace ß-cell function in hyperglycemic immunodeficient mice. These findings demonstrate the redifferentiation potential of ex vivo expanded BCD cells and the reproducible differentiating effect of HES1 inhibition in these cells.


Subject(s)
Cell Dedifferentiation , Insulin-Secreting Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction , Adolescent , Adult , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Proliferation , Cells, Cultured , Epigenesis, Genetic/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Humans , Insulin/biosynthesis , Insulin-Secreting Cells/cytology , Male , Middle Aged , Transcription Factor HES-1
3.
PLoS One ; 6(9): e25566, 2011.
Article in English | MEDLINE | ID: mdl-21984932

ABSTRACT

BACKGROUND: Expansion of beta cells from the limited number of adult human islet donors is an attractive prospect for increasing cell availability for cell therapy of diabetes. However, attempts at expanding human islet cells in tissue culture result in loss of beta-cell phenotype. Using a lineage-tracing approach we provided evidence for massive proliferation of beta-cell-derived (BCD) cells within these cultures. Expansion involves dedifferentiation resembling epithelial-mesenchymal transition (EMT). Epigenetic analyses indicate that key beta-cell genes maintain open chromatin structure in expanded BCD cells, although they are not transcribed. Here we investigated whether BCD cells can be redifferentiated into beta-like cells. METHODOLOGY/PRINCIPAL FINDING: Redifferentiation conditions were screened by following activation of an insulin-DsRed2 reporter gene. Redifferentiated cells were characterized for gene expression, insulin content and secretion assays, and presence of secretory vesicles by electron microscopy. BCD cells were induced to redifferentiate by a combination of soluble factors. The redifferentiated cells expressed beta-cell genes, stored insulin in typical secretory vesicles, and released it in response to glucose. The redifferentiation process involved mesenchymal-epithelial transition, as judged by changes in gene expression. Moreover, inhibition of the EMT effector SLUG (SNAI2) using shRNA resulted in stimulation of redifferentiation. Lineage-traced cells also gave rise at a low rate to cells expressing other islet hormones, suggesting transition of BCD cells through an islet progenitor-like stage during redifferentiation. CONCLUSIONS/SIGNIFICANCE: These findings demonstrate for the first time that expanded dedifferentiated beta cells can be induced to redifferentiate in culture. The findings suggest that ex-vivo expansion of adult human islet cells is a promising approach for generation of insulin-producing cells for transplantation, as well as basic research, toxicology studies, and drug screening.


Subject(s)
Cell Differentiation/physiology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Humans , RNA, Small Interfering , Snail Family Transcription Factors , Tissue Culture Techniques , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Biochem Biophys Res Commun ; 341(2): 291-8, 2006 Mar 10.
Article in English | MEDLINE | ID: mdl-16446152

ABSTRACT

Beta-cell replacement represents the ultimate cure for type 1 diabetes, however it is limited by availability of organ donors. Adult human islets are difficult to propagate in culture, and efforts to expand them result in dedifferentiation. Here we describe conditions for expansion of adult human islet cells, as well as a way for their redifferentiation. Most cells in islets isolated from human pancreata were induced to replicate within the first week of culture in expansion medium. Cells were propagated for 16 population doublings, without a change in replication rate or noticeable cell mortality, representing an expansion of over 65,000-fold. Replication was accompanied by a decrease in expression of key beta-cell genes. Shift of the cells to differentiation medium containing betacellulin resulted in redifferentiation, as manifested by restoration of beta-cell gene expression and insulin content. These methods may allow transplantation of functional islet cells from single donors into multiple recipients.


Subject(s)
Gene Expression Regulation , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Adult , Aged , Cell Culture Techniques , Cell Differentiation , Cells, Cultured , Dose-Response Relationship, Drug , Female , Glucose/pharmacology , Humans , Insulin/metabolism , Ischemia , Male , Microscopy, Confocal , Microscopy, Fluorescence , Middle Aged , Peptides/chemistry , Phenotype , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
5.
Diabetes ; 54(9): 2568-75, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16123344

ABSTRACT

beta-Cell transplantation is viewed as a cure for type 1 diabetes; however, it is limited by the number of pancreas donors. Human stem cells offer the promise of an abundant source of insulin-producing cells, given the existence of methods for manipulating their differentiation. We have previously demonstrated that the expression of the beta-cell transcription factor pancreatic duodenal homeobox 1 (PDX-1) in human fetal liver cells activates multiple aspects of the beta-cell phenotype. These cells, termed FH-B-TPN cells, produce insulin, release insulin in response to physiological glucose levels, and replace beta-cell function in diabetic immunodeficient mice. However, they deviate from the normal beta-cell phenotype by the lack of expression of a number of beta-cell genes, the expression of non-beta-cell genes, and a lower insulin content. Here we aimed to promote differentiation of FH-B-TPN cells toward the beta-cell phenotype using soluble factors. Cells cultured with activin A in serum-free medium upregulated expression of NeuroD and Nkx2.2 and downregulated paired box homeotic gene 6 (PAX-6). Glucokinase and prohormone convertase 1/3 were also upregulated, whereas pancreatic polypeptide and glucagon as well as liver markers were downregulated. Insulin content was increased by up to 33-fold, to approximately 60% of the insulin content of normal beta-cells. The cells were shown to contain human C-peptide and release insulin in response to physiological glucose levels. Cell transplantation into immunodeficient diabetic mice resulted in the restoration of stable euglycemia. The cells continued to express insulin in vivo, and no cell replication was detected. Thus, the manipulation of culture conditions induced a significant and stable differentiation of FH-B-TPN cells toward the beta-cell phenotype, making them excellent candidates for beta-cell replacement in type 1 diabetes.


Subject(s)
Cell Differentiation/physiology , Hepatocytes/metabolism , Homeodomain Proteins/metabolism , Insulin/metabolism , Stem Cells/physiology , Trans-Activators/metabolism , Activins/pharmacology , Betacellulin , Exenatide , Gene Expression Regulation , Homeobox Protein Nkx-2.2 , Humans , Inhibin-beta Subunits/pharmacology , Intercellular Signaling Peptides and Proteins/pharmacology , Niacinamide/pharmacology , Nuclear Proteins , Peptides/pharmacology , Phenotype , Transcription Factors , Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...