Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 6(7): e22750, 2011.
Article in English | MEDLINE | ID: mdl-21829502

ABSTRACT

BACKGROUND: Netrins have been extensively studied in the developing central nervous system as pathfinding guidance cues, and more recently in non-neural tissues where they mediate cell adhesion, migration and differentiation. Netrin-4, a distant relative of Netrins 1-3, has been proposed to affect cell fate determination in developing epithelia, though receptors mediating these functions have yet to be identified. METHODOLOGY/PRINCIPAL FINDINGS: Using human embryonic pancreatic cells as a model of developing epithelium, here we report that Netrin-4 is abundantly expressed in vascular endothelial cells and pancreatic ductal cells, and supports epithelial cell adhesion through integrins α2ß1 and α3ß1. Interestingly, we find that Netrin-4 recognition by embryonic pancreatic cells through integrins α2ß1 and α3ß1 promotes insulin and glucagon gene expression. In addition, full genome microarray analysis revealed that fetal pancreatic cell adhesion to Netrin-4 causes a prominent down-regulation of cyclins and up-regulation of negative regulators of the cell cycle. Consistent with these results, a number of other genes whose activities have been linked to developmental decisions and/or cellular differentiation are up-regulated. CONCLUSIONS/SIGNIFICANCE: Given the recognized function of blood vessels in epithelial tissue morphogenesis, our results provide a mechanism by which endothelial-derived Netrin-4 may function as a pro-differentiation cue for adjacent developing pancreatic cell populations expressing adhesion receptors α2ß1 and α3ß1 integrins.


Subject(s)
Cell Adhesion , Cell Differentiation , Endothelium, Vascular/metabolism , Epithelial Cells/metabolism , Glucagon/metabolism , Insulin/metabolism , Integrin alpha2beta1/metabolism , Integrin alpha3beta1/metabolism , Nerve Growth Factors/metabolism , Pancreatic Ducts/cytology , Biomarkers/metabolism , Blotting, Western , Cell Movement , Cell Proliferation , Endothelium, Vascular/cytology , Fetus/cytology , Fetus/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Developmental , Glucagon/genetics , Humans , Immunoenzyme Techniques , Immunoprecipitation , Insulin/genetics , Nerve Growth Factors/genetics , Netrins , Oligonucleotide Array Sequence Analysis , Pancreatic Ducts/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
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
3.
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
4.
Diabetes ; 55(10): 2723-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17003336

ABSTRACT

The impact of extracellular matrix on insulin production needs to be understood both to optimize the derivation of functional beta-cells for transplantation and to understand mechanisms controlling islet neogenesis and glucose homeostasis. In this study, we present evidence that adhesion to some common matrix constituents has a profound impact on the transcription, secretion, and storage of insulin by human beta-cells. The integrin-dependent adhesion of fetal beta-cells to both collagen IV and vitronectin induces significant glucose-independent insulin secretion and a substantial reciprocal decline in insulin content. Collagen IV, but not vitronectin, induces comparable responses in adult beta-cells. Inhibition of extracellular signal-regulated kinase activation abrogates matrix-induced insulin secretion and effectively preserves the insulin content of adherent beta-cells. Using real-time PCR, we demonstrate that adhesion of both fetal and adult beta-cells to collagen IV and vitronectin also results in the marked suppression of insulin gene transcription. Based on these findings, we contend that integrin-dependent adhesion and signaling in response to certain matrices can have a significant negative impact on insulin production by primary human beta-cells. Such responses were not found to be associated with cell death but may precede beta-cell dedifferentiation.


Subject(s)
Extracellular Matrix/physiology , Insulin-Secreting Cells/metabolism , Insulin/biosynthesis , Integrins/physiology , Butadienes/pharmacology , Cell Adhesion/physiology , Cells, Cultured , Collagen Type IV/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fetus/cytology , Humans , Insulin/metabolism , Insulin Secretion , Nitriles/pharmacology , Signal Transduction/drug effects , Transcription, Genetic/physiology , Vitronectin/physiology
5.
J Biol Chem ; 279(51): 53762-9, 2004 Dec 17.
Article in English | MEDLINE | ID: mdl-15485856

ABSTRACT

Collagens have been shown to influence the survival and function of cultured beta-cells; however, the utilization and function of individual collagen receptors in beta-cells is largely unknown. The integrin superfamily contains up to five collagen receptors, but we have determined that alpha(1)beta(1) is the primary receptor utilized by both fetal and adult beta-cells. Cultured beta-cells adhered to and migrated on collagen type IV (Col-IV), and these responses were mediated almost exclusively by alpha(1)beta(1). The migration of cultured beta-cells to Col-IV significantly exceeded that to other matrix components suggesting that this substrate is of unique importance for beta-cell motility. The interaction of alpha(1)beta(1) with Col-IV also resulted in significant insulin secretion at basal glucose concentrations. A subset of beta-cells in developing islets was confirmed to express alpha(1)beta(1), and this expression co-localized with Col-IV in the basal membranes of juxtaposed endothelial cells. Our findings indicate that alpha(1)beta(1) and Col-IV contribute to beta-cell functions known to be important for islet morphogenesis and glucose homeostasis.


Subject(s)
Collagen Type IV/physiology , Insulin/metabolism , Integrin alpha1beta1/metabolism , Islets of Langerhans/metabolism , Aged , Cell Adhesion , Cell Movement , Cell Separation , Cell Survival , Cells, Cultured , Collagen/metabolism , Flow Cytometry , Glucose/metabolism , Humans , Immunohistochemistry , Insulin Secretion , Integrins/metabolism , Microscopy, Fluorescence , Middle Aged , Pancreas/embryology , Time Factors
6.
J Biol Chem ; 279(17): 17731-7, 2004 Apr 23.
Article in English | MEDLINE | ID: mdl-14766759

ABSTRACT

The role of individual integrins in human beta-cell development and function is largely unknown. This study describes the contribution of alpha(v)-integrins to human beta-cell adhesion, spreading, and motility. Developmental differences in alpha(v)-integrin utilization are addressed by comparing the responses of adult and fetal beta-cells, and vitronectin is used as a substrate based on its unique pattern of expression in the developing pancreas. Fetal and adult beta-cells attached equally to vitronectin and integrin alpha(v)beta(5) was found to support the adhesion of both mature and immature beta-cell populations. Fetal beta-cells were also observed to spread and migrate on vitronectin, and integrin alpha(v)beta(1) was found to be essential for these responses. In contrast to their fetal counterparts, adult beta-cells failed to either spread or migrate and this deficit was associated with a marked down-regulation of alpha(v)beta(1) expression in adult islet preparations. The integrin alpha(v)beta(3) was not found to support significant beta-cell attachment or migration. Based on our findings, we conclude that integrins alpha(v)beta(5) and alpha(v)beta(1) are important mediators of human beta-cell adhesion and motility, respectively. By supporting fetal beta-cell migration, alpha(v)beta(1) could play an important role in early motile processes required for islet neogenesis.


Subject(s)
Integrin alphaV/physiology , Islets of Langerhans/cytology , Adult , Aged , Blotting, Western , Cadaver , Cell Adhesion , Cell Line , Cell Movement , Collagenases/metabolism , Down-Regulation , Flow Cytometry , Gene Expression Regulation, Developmental , Humans , Integrin alphaV/metabolism , Integrins/metabolism , Microscopy, Fluorescence , Middle Aged , Pancreas/cytology , Pancreas/embryology , Precipitin Tests , Protein Binding , Receptors, Vitronectin/metabolism , Time Factors , Vitronectin/metabolism
7.
Dev Cell ; 5(5): 695-707, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14602071

ABSTRACT

Netrins, axon guidance cues in the CNS, have also been detected in epithelial tissues. In this study, using the embryonic pancreas as a model system, we show that Netrin-1 is expressed in a discrete population of epithelial cells, localizes to basal membranes, and specifically associates with elements of the extracellular matrix. We demonstrate that alpha6beta4 integrin mediates pancreatic epithelial cell adhesion to Netrin-1, whereas recruitment of alpha6beta4 and alpha3beta1 regulate the migration of CK19+/PDX1+ putative pancreatic progenitors on Netrin-1. These results provide evidence for the activation of epithelial cell adhesion and migration by a neural chemoattractant, and identify Netrin-1/integrin interactions as adhesive/guidance cues for epithelial cells.


Subject(s)
Cell Adhesion/physiology , Cell Movement/physiology , Epithelial Cells/metabolism , Integrin alpha3beta1/metabolism , Integrin alpha6beta4/metabolism , Nerve Growth Factors/metabolism , Animals , Cell Line , Epithelial Cells/cytology , Extracellular Matrix Proteins/metabolism , Humans , Netrin-1 , Pancreas/cytology , Pancreas/embryology , Pancreas/metabolism , Peptide Fragments/metabolism , Protein Binding , Protein Subunits/metabolism , Tumor Suppressor Proteins
8.
J Refract Surg ; 18(5): 529-34, 2002.
Article in English | MEDLINE | ID: mdl-12361153

ABSTRACT

PURPOSE: To compare the cytotoxic and mutagenic effect of 213-nm and 193-nm laser radiation on cultured mammalian cells. METHODS: Chinese hamster lung (V79) cells were exposed to 193-nm radiation from an argon fluorine excimer laser or 213-nm radiation from a 5th harmonic Nd:YAG laser. The cytotoxic action of the lasers was compared by determining the number of V79 cell colonies that formed 1 week after irradiating cells with different doses of 193-nm or 213-nm laser radiation or with continuous wave 254-nm radiation. The cytotoxic action of the lasers on primary cultures of human corneal fibroblasts was also compared. The mutagenic potential of the lasers was compared by measuring the number of ouabain or 6-Thioguanine(6TG)-resistant V79 mutants that formed after exposing V79 cells to 193-nm or 213-nm radiation. RESULTS: The dose of 193-nm laser radiation that resulted in 37% survival (D37) of V79 cells was estimated to be 11.3 mJ/cm2 compared to 3.2 mJ/cm2 for 213-nm laser radiation and 1.2 mJ/cm2 for 254-nm UV radiation. The mean number of ouabain-induced mutants induced at the D37 for 193-nm, 213-nm, and 254-nm laser radiation were 28, 166, and 279 mutants/10(7) cells, respectively. Continuous wave 254-nm radiation induced 6TG-resistant colonies, but there was no significant induction of 6TG-resistant mutants by either laser. CONCLUSIONS: Although the in vitro data presented herein may or may not be meaningful to humans, the 213-nm Nd:YAG laser was more cytotoxic and mutagenic than the 193-nm excimer laser on cultured mammalian cells but was less cytotoxic and mutagenic than 254-nm radiation.


Subject(s)
Lasers/adverse effects , Lung/radiation effects , Mutagenesis , Animals , Cell Survival , Cornea/cytology , Cornea/radiation effects , Cricetinae , Cricetulus , Fibroblasts , In Vitro Techniques , Lung/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...