Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Nat Prod Res ; : 1-10, 2024 Feb 11.
Article in English | MEDLINE | ID: mdl-38343285

ABSTRACT

Thirteen nitrogen-containing derivatives of 3,11-dioxo-olean-12-en-30-oic acid were synthesised by introducing various amino acids and nitrogen-containing heterocyclic groups at the 30-carboxyl group, starting from 18ß-glycyrrhetinic acid. Among the 13 derivatives, 10 exhibited inhibitory activity against HIV-1 PR, with IC50 values ranging from 0.19 to 0.94 mM. Notably, derivatives 2, 3 and 5 displayed relatively moderate inhibitory activity, with IC50 values below 0.24 mM. Molecular docking studies provided further insights into the interaction between derivatives (2, 3 and 5) and the active sites of HIV-1 PR. The results revealed favourable hydrophobic-hydrophobic and hydrogen bonding interactions, with docking scores ranging from -6.22 to -7.00 and glide emodel values from -62.9 to -48.6 (kcal/mol). These findings underscore the potential of derivatives 2, 3 and 5 as promising candidates for the development of HIV-1 PR inhibitors.

2.
J Am Chem Soc ; 135(5): 1669-72, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23330637

ABSTRACT

The identification of factors that promote ß cell proliferation could ultimately move type 1 diabetes treatment away from insulin injection therapy and toward a cure. We have performed high-throughput, cell-based screens using rodent ß cell lines to identify molecules that induce proliferation of ß cells. Herein we report the discovery and characterization of WS6, a novel small molecule that promotes ß cell proliferation in rodent and human primary islets. In the RIP-DTA mouse model of ß cell ablation, WS6 normalized blood glucose and induced concomitant increases in ß cell proliferation and ß cell number. Affinity pulldown and kinase profiling studies implicate Erb3 binding protein-1 and the IκB kinase pathway in the mechanism of action of WS6.


Subject(s)
High-Throughput Screening Assays , Islets of Langerhans/drug effects , Urea/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Islets of Langerhans/cytology , Mice , Molecular Structure , Molecular Weight , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
3.
RSC Adv ; 12(46): 29767-29776, 2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36321110

ABSTRACT

Traditional grass cloth has been used in China for a long time for the manufacturing of various household furnishing textiles and ladieswear. However, traditionally the grass cloth is dyed with reactive dyes in an aqueous medium, but the dyeing process is not sustainable because of high energy and water usage and the production of coloured effluent. In this work, the possibility of palm oil/water dual-phase dyeing of traditional grass cloth with a reactive dye, C.I. Reactive Blue 194 (Reactive Blue 194), was explored. The grass cloth soaked in an alkaline solution with 80-140% pick-up was dyed in a palm oil dyebath containing dye powder dispersed in a palm oil medium. The initial study confirmed that the pre-treatment of the fabric with an alkaline solution with 140% pick-up was beneficial for the uniform distribution of the dye in the fibres. The dyeing process parameters (e.g., fixation temperature, solution pH, and fixation time) for the grass cloth dyeing with the Reactive Blue 194 were optimised by using the Taguchi method. The pH of the alkali pre-treatment solution was found to be the most influential factor, as confirmed by the analysis of variance in terms of the percentage of contribution (94.41%), which was statistically significant (P < 0.05). The confirmation tests were carried out under optimal settings, and a higher K/S (24.06) was found compared with the initial condition (21.51). X-ray diffraction analysis indicated that the dyeing process did not affect the crystallinity of the grass cloth fibres. Furthermore, the recovery of palm oil from the spent dyebath was around 99%, and up to five times recycling and reuse of palm oil were studied for the dyeing of grass cloth. The colour strength of the grass cloths dyed in the palm oil recycled up to five times was similar to the cloth dyed in fresh palm oil. The results show that palm oil can be used as a dyeing medium for the sustainable dyeing of grass cloth with effluent reduction, which can be extended to the dyeing of other textile fibres.

4.
Materials (Basel) ; 15(21)2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36363411

ABSTRACT

As an edible oil, palm oil is also safe and reliable in dyeing, and the residual palm oil after dyeing can be recycled and used continuously, which is green and environmentally friendly and has great research prospects. In this research, raw ramie yarn, used for traditional grass cloth, was dyed in a palm oil medium using Reactive Blue 194. Studying the adsorption and diffusion behaviour in the dyeing process is necessary. Additionally, the kinetics and isotherm model of dyeing raw ramie yarn with Reactive Blue 194 in palm oil is studied, and the adsorption behaviour between them is discussed. For a better understanding, the raw ramie yarn dyeing adsorption behaviour was also carried out in a water medium. It was found that the dyeing rates in palm oil are distinctly faster than in water. Kinetics data suggested that the pseudo-second-order model fitted for both dyeing mediums (palm oil and water) of the adsorption of the Reactive Blue 194 dye onto raw ramie yarn. Afterward, the adsorption isotherms' results denote that the Langmuir model was suitable for palm oil dyeing medium while the Freundlich model was suited for water medium. Overall, this study has demonstrated that raw ramie yarn dyeing in a palm oil medium could be a sustainable colouration route for textile fibres with a greater dye exhaustion percentage.

5.
ChemMedChem ; 15(16): 1562-1570, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32613743

ABSTRACT

Loss of ß-cell mass and function can lead to insufficient insulin levels and ultimately to hyperglycemia and diabetes mellitus. The mainstream treatment approach involves regulation of insulin levels; however, approaches intended to increase ß-cell mass are less developed. Promoting ß-cell proliferation with low-molecular-weight inhibitors of dual-specificity tyrosine-regulated kinase 1A (DYRK1A) offers the potential to treat diabetes with oral therapies by restoring ß-cell mass, insulin content and glycemic control. GNF4877, a potent dual inhibitor of DYRK1A and glycogen synthase kinase 3ß (GSK3ß) was previously reported to induce primary human ß-cell proliferation in vitro and in vivo. Herein, we describe the lead optimization that lead to the identification of GNF4877 from an aminopyrazine hit identified in a phenotypic high-throughput screening campaign measuring ß-cell proliferation.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Structure-Activity Relationship , Dyrk Kinases
6.
J Med Chem ; 63(6): 2958-2973, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32077280

ABSTRACT

Autoimmune deficiency and destruction in either ß-cell mass or function can cause insufficient insulin levels and, as a result, hyperglycemia and diabetes. Thus, promoting ß-cell proliferation could be one approach toward diabetes intervention. In this report we describe the discovery of a potent and selective DYRK1A inhibitor GNF2133, which was identified through optimization of a 6-azaindole screening hit. In vitro, GNF2133 is able to proliferate both rodent and human ß-cells. In vivo, GNF2133 demonstrated significant dose-dependent glucose disposal capacity and insulin secretion in response to glucose-potentiated arginine-induced insulin secretion (GPAIS) challenge in rat insulin promoter and diphtheria toxin A (RIP-DTA) mice. The work described here provides new avenues to disease altering therapeutic interventions in the treatment of type 1 diabetes (T1D).


Subject(s)
Aza Compounds/chemistry , Aza Compounds/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Indoles/chemistry , Indoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Aza Compounds/pharmacokinetics , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Indoles/pharmacokinetics , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Molecular Docking Simulation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Dyrk Kinases
7.
Am J Physiol Gastrointest Liver Physiol ; 297(4): G768-80, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19643954

ABSTRACT

Several lines of studies have suggested that activins are critical mediators of inflammation and tissue repair. As activins and their receptors are expressed in the gastrointestinal tract, we tested the hypothesis that activin signaling is involved in the development of colitis by using two murine models of colitis induced by dextran sodium sulfate (DSS) or in mdr1a-/- mice. By immunohistochemistry, expression of activins was found increased in both models and correlated with the severity of inflammation. Activin expression was observed in macrophages as well as in some nonmacrophage cells. Furthermore, while activin receptors are normally expressed in colonic epithelial cells, their expression was further increased in both epithelial cells and inflammatory cells in inflamed colonic mucosa. Moreover, in vitro studies showed that activin A inhibited proliferation and induced apoptosis of intestinal epithelial cells, and this growth inhibition was largely reversed by administration of the activin inhibitor, follistatin. Because we also observed an increased number of apoptotic epithelial cells in both colitis models, the upregulation of activins occurring in colitis could be involved both in the inflammatory process and in growth inhibition of the intestinal epithelium. Importantly, in vivo administration of follistatin attenuated inflammatory cell infiltration during colitis. Rectal bleeding was reduced, and the integrity of epithelium was preserved in the DSS/follistatin-treated group compared with the group treated with DSS alone. Bromodeoxyuridine incorporation studies showed an increase in proliferative epithelial cells in the DSS/follistatin-treated group, suggesting that follistatin accelerates epithelial cell proliferation/repair during colitis. Overall, our results reveal that activin signaling may play an important role in the pathogenesis and resolution of colitis. These findings suggest new therapeutic options in inflammatory bowel diseases.


Subject(s)
Activin Receptors/metabolism , Activins/metabolism , Colitis/metabolism , Colon/metabolism , Signal Transduction , ATP Binding Cassette Transporter, Subfamily B/deficiency , ATP Binding Cassette Transporter, Subfamily B/genetics , Activin Receptors/genetics , Activins/antagonists & inhibitors , Activins/genetics , Animals , Apoptosis , Cell Line , Cell Proliferation , Colitis/chemically induced , Colitis/drug therapy , Colitis/genetics , Colitis/pathology , Colon/drug effects , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Dose-Response Relationship, Drug , Follistatin/administration & dosage , Humans , Immunohistochemistry , Injections, Intraperitoneal , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Time Factors , Up-Regulation
8.
J Cell Biol ; 163(4): 859-69, 2003 Nov 24.
Article in English | MEDLINE | ID: mdl-14638861

ABSTRACT

The SDF-1alpha/CXCR4 ligand/chemokine receptor pair is required for appropriate patterning during ontogeny and stimulates the growth and differentiation of critical cell types. Here, we demonstrate SDF-1alpha and CXCR4 expression in fetal pancreas. We have found that SDF-1alpha and its receptor CXCR4 are expressed in islets, also CXCR4 is expressed in and around the proliferating duct epithelium of the regenerating pancreas of the interferon (IFN) gamma-nonobese diabetic mouse. We show that SDF-1alpha stimulates the phosphorylation of Akt, mitogen-activated protein kinase, and Src in pancreatic duct cells. Furthermore, migration assays indicate a stimulatory effect of SDF-1alpha on ductal cell migration. Importantly, blocking the SDF-1alpha/CXCR4 axis in IFNgamma-nonobese diabetic mice resulted in diminished proliferation and increased apoptosis in the pancreatic ductal cells. Together, these data indicate that the SDF-1alpha-CXCR4 ligand receptor axis is an obligatory component in the maintenance of duct cell survival, proliferation, and migration during pancreatic regeneration.


Subject(s)
Chemokines, CXC/metabolism , Pancreas/growth & development , Protein Serine-Threonine Kinases , Receptors, CXCR4/metabolism , Stem Cells/metabolism , Animals , Apoptosis/physiology , Cell Division/physiology , Cell Movement/physiology , Cell Survival/physiology , Cells, Cultured , Chemokine CXCL12 , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Fetus , Ligands , Mice , Mice, Inbred NOD , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Pancreas/cytology , Pancreas/metabolism , Pancreatic Ducts/cytology , Pancreatic Ducts/growth & development , Pancreatic Ducts/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Regeneration/physiology , Stem Cells/cytology , src-Family Kinases/metabolism
9.
Diabetes ; 56(1): 96-106, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17192470

ABSTRACT

Fibroblast growth factors (FGFs) and their receptors (FGFRs) are key signaling molecules for pancreas development. Although FGFR3 is a crucial developmental gene, acting as a negative regulator of bone formation, its participation remains unexplored in pancreatic organogenesis. We found that FGFR3 was expressed in the epithelia in both mouse embryonic and adult regenerating pancreata but was absent in normal adult islets. In FGFR3 knockout mice, we observed an increase in the proliferation of epithelial cells in neonates, leading to a marked increase in islet areas in adults. In vitro studies showed that FGF9 is a very potent ligand for FGFR3 and activates extracellular signal-related kinases (ERKs) in pancreatic cell lines. Moreover, FGFR3 blockade or FGFR3 deficiency led to increased proliferation of pancreatic epithelial cells in vivo. This was accompanied by an increase in the proportion of potential islet progenitor cells. Thus, our results show that FGFR3 signaling inhibits the expansion of the immature pancreatic epithelium. Consequently, this study suggests that FGFR3 participates in regulating pancreatic growth during the emergence of mature islet cells.


Subject(s)
Epithelial Cells/cytology , Islets of Langerhans/physiology , Pancreas/cytology , Receptor, Fibroblast Growth Factor, Type 3/physiology , Aging , Animals , Animals, Newborn , Cell Line , Epithelial Cells/drug effects , Islets of Langerhans/cytology , Mice , Mice, Inbred NOD , Mice, Knockout , Pancreas/embryology , Receptor, Fibroblast Growth Factor, Type 3/deficiency , Receptor, Fibroblast Growth Factor, Type 3/genetics , Regeneration , Signal Transduction/physiology
10.
Diabetes ; 53(8): 2024-33, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277382

ABSTRACT

Activins regulate the growth and differentiation of a variety of cells. During pancreatic islet development, activins are required for the specialization of pancreatic precursors from the gut endoderm during midgestation. In this study, we probed the role of activin signaling during pancreatic islet cell development and regeneration. Indeed, we found that both activins and activin receptors are upregulated in duct epithelial cells during islet differentiation. Interestingly, the expression of endogenous cellular inhibitors of activin signaling, follistatin and Cripto, were also found to be augmented. Inhibition of activins significantly enhanced survival and expansion of pancreatic epithelial cells but decreased the numbers of differentiated beta-cells. Our results suggest that the homeostasis of growth and terminal differentiation requires a precise context-dependent regulation of activin signaling. Follistatin participates in this process by promoting expansion of precursor cells during pancreas growth.


Subject(s)
Activins/physiology , Epithelial Cells/cytology , Islets of Langerhans/physiology , Pancreas/cytology , Activin Receptors/physiology , Activins/antagonists & inhibitors , Activins/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Epithelial Cells/drug effects , Follistatin/pharmacology , Humans , Interferon-gamma/genetics , Islets of Langerhans/drug effects , Mice , Mice, Inbred NOD , Mice, Transgenic , Recombinant Proteins/pharmacology
11.
Nat Commun ; 6: 8372, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26496802

ABSTRACT

Insufficient pancreatic ß-cell mass or function results in diabetes mellitus. While significant progress has been made in regulating insulin secretion from ß-cells in diabetic patients, no pharmacological agents have been described that increase ß-cell replication in humans. Here we report aminopyrazine compounds that stimulate robust ß-cell proliferation in adult primary islets, most likely as a result of combined inhibition of DYRK1A and GSK3B. Aminopyrazine-treated human islets retain functionality in vitro and after transplantation into diabetic mice. Oral dosing of these compounds in diabetic mice induces ß-cell proliferation, increases ß-cell mass and insulin content, and improves glycaemic control. Biochemical, genetic and cell biology data point to Dyrk1a as the key molecular target. This study supports the feasibility of treating diabetes with an oral therapy to restore ß-cell mass, and highlights a tractable pathway for future drug discovery efforts.


Subject(s)
Cell Proliferation , Glycogen Synthase Kinase 3/genetics , Insulin-Secreting Cells/cytology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Animals , Cell Division/drug effects , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Down-Regulation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , Male , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Pyridazines/pharmacology , Dyrk Kinases
12.
Endocrinology ; 143(9): 3540-7, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12193568

ABSTRACT

Activins expressed in progenitor cells of the pancreas regulate differentiation of endocrine cells during development. Neogenesis of beta-cells takes place in adult animals under some conditions, and beta-cells are thought to arise from precursors locating in the pancreatic duct. In the present study, we investigated whether or not activins are expressed in the duct where beta-cell neogenesis is initiated. mRNA for the beta(A)- and beta(B)-subunits was expressed in isolated mouse pancreatic ducts. Immunohistochemically, the beta(A)-subunit was detected in the pancreatic duct and colocalized with cytokeratin, a marker of ductal cells. The beta(A)-subunit was also expressed in nestin-positive cells in the duct. Likewise, the beta(B)-subunit was detected in the pancreatic duct. In addition, mRNA for the type II and type IIB activin receptors was expressed in the duct. Expression of mRNA for two activin subunits was markedly increased after streptozotocin injection. Similarly, the mRNA expression was up-regulated after partial pancreatectomy. These results indicate that activins are expressed in the pancreatic duct and are up-regulated shortly after the reduction of the beta-cell mass. Induction of activins in the duct may be a critical step in the initiation of beta-cell neogenesis.


Subject(s)
Activins/genetics , Gene Expression Regulation , Islets of Langerhans/cytology , Nerve Tissue Proteins , Pancreatic Ducts/metabolism , Activin Receptors/genetics , Animals , Blood Glucose/metabolism , Cell Differentiation , Immunohistochemistry , Inhibin-beta Subunits/analysis , Inhibin-beta Subunits/genetics , Insulin/blood , Intermediate Filament Proteins/analysis , Islets of Langerhans/drug effects , Male , Mice , Nestin , Pancreatectomy , Pancreatic Ducts/chemistry , Pancreatic Ducts/cytology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/cytology , Streptozocin/pharmacology
13.
Endocrine ; 37(2): 312-21, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20960269

ABSTRACT

The growth and renewal of epithelial tissue is a highly orchestrated and tightly regulated process occurring in different tissue types under a variety of circumstances. We have been studying the process of pancreatic regeneration in mice. We have identified a cell surface protein, named EP1, which is expressed on the duct epithelium during pancreatic regeneration. Whereas it is not detected in the pancreas of normal mice, it is found in the intestinal epithelium of normal adult mice, as well as during pancreatic repair following cerulein-induced destruction of the acinar tissue. The distinctive situations in which EP1 is expressed, all of which share in common epithelial cell growth in the gastrointestinal tract, suggest that EP1 is involved in the growth and renewal of epithelial tissues in both the intestine and the pancreas.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Pancreas/cytology , Pancreas/physiology , Amino Acid Sequence , Animals , Cell Differentiation/physiology , Chemokine CXCL12/metabolism , Intestines/cytology , Intestines/physiology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Mitosis/physiology , Molecular Sequence Data , Receptors, CXCR4/metabolism , Regeneration/physiology
14.
Article in Zh | MEDLINE | ID: mdl-19670616

ABSTRACT

OBJECTIVE: To study the clinical effect of Treatment of blow-out fracture of medial orbital wall with nasoseptal cartilage under nasal endoscope. METHOD: Under a nasal endoscope, the fracture and the prolapsed orbital contents were reduced to the orbit, and then an autogenous nasoseptal cartilage was grafted into the orbital defect. The variations in the visual acuity, diplopia, enophthalmos degree and eyeball position were detected preoperatively and postoperatively. RESULT: During the follow up of three months to four years after operation, all the 28 cases showed neither loss nor distinct descent of visual acuity. The postoperative mean enophthalmos degree (1.5 +/- 0.6) mm was lower than the preoperation one(3.6 +/- 1.1) mm (P<0.05). Diplopia disappeared completely of 25 cases during 3 month after operation,while it appeared in the primary position of 2 cases. The eye movement was normal of 26 cases after operation t, and the abduction was slightly limited of 2 cases, but which was better than be for). Any displacement of filling material, infection, rejection reaction were not found of all the 28 cases. CONCLUSION: The medial orbital blow out fracture with nasal endoscope has many advantages, such as short operative route, clear surgical visual field, simple performance, light injury and no scars, and the effect of which will be really certain in the operative practice.


Subject(s)
Nasal Septum/surgery , Orbital Fractures/surgery , Adult , Endoscopy/methods , Female , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
15.
Dev Dyn ; 237(5): 1255-67, 2008 May.
Article in English | MEDLINE | ID: mdl-18393305

ABSTRACT

Nodal and its antagonist, Lefty, are important mediators specifying the laterality of the organs during embryogenesis. Nodal signals through activin receptors in the presence of its co-receptor, Cripto. In the present study, we investigated the possible roles of Nodal and Lefty signaling during islet development and regeneration. We found that both Nodal and Lefty are expressed in the pancreas during embryogenesis and islet regeneration. In vitro studies demonstrated that Nodal inhibits, whereas Lefty enhances, the proliferation of a pancreatic cell line. In addition, we showed that Lefty-1 activates MAPK and Akt phosphorylation in these cells. In vivo blockade of endogenous Lefty using neutralizing Lefty-1 monoclonal antibody results in a significantly decreased proliferation of duct epithelial cells during islet regeneration. This is the first study to decipher the expression and function of Nodal and Lefty in pancreatic growth. Importantly, our results highlight a novel function of Nodal-Lefty signaling in the regulation of expansion of pancreatic cells.


Subject(s)
Gene Expression Regulation, Developmental , Islets of Langerhans , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , Activins/metabolism , Animals , Apoptosis , Cell Line , Cell Proliferation , Enzyme Activation , Interferon-gamma/genetics , Interferon-gamma/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/embryology , Left-Right Determination Factors , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Nodal Protein , Proto-Oncogene Proteins c-akt/metabolism , Regeneration , Transforming Growth Factor beta/genetics
16.
Endocrine ; 30(1): 103-12, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17185798

ABSTRACT

Gut peptide YY (PYY) plays an important role in regulating metabolism and is expressed during the ontogeny of the pancreas. However, its biological role during endocrine cell formation is not fully understood, and its role, if any, during pancreatic regeneration in the adult has not yet been explored. The knowledge of factors involved in beta cell renewal in adult animals is clearly relevant for the design of treatment strategies for type 1 diabetes. We therefore sought to determine if observations during fetal pancreas formation also apply to pancreatic growth in adult animals. Indeed, we have found marked expansion of the PYY-expressing population during pancreatic regeneration. In addition, we demonstrate the presence of cells co-expressing PYY and the critical pancreatic transcription factor pancreatic duodenal homeobox1 (PDX-1). Interestingly, these cells also co-expressed specific islet hormones during pancreatic development and re-growth, suggesting a developmental relationship. Furthermore, we have found that PYY can act in concert with IGF-1 to stimulate cellular responsiveness in pancreatic epithelial cells in vitro. Our data suggest that PYY may be a mediator of islet cell development, as well as a cofactor for growth factor responses, not only during fetal pancreas formation but also during regeneration in adult animals.


Subject(s)
Pancreas/physiology , Peptide YY/physiology , Regeneration/physiology , Animals , Cell Differentiation/physiology , Epithelial Cells/physiology , Immunohistochemistry , Insulin-Like Growth Factor I/physiology , Interferon-gamma/genetics , Mice , Mice, Inbred NOD , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Pancreas/embryology , Pancreas/growth & development , Pancreas/metabolism , Peptide YY/genetics , Peptide YY/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Gastrointestinal Hormone/biosynthesis , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/physiology , Reverse Transcriptase Polymerase Chain Reaction
17.
J Biol Chem ; 281(19): 13574-13580, 2006 May 12.
Article in English | MEDLINE | ID: mdl-16547003

ABSTRACT

Inhibitor of DNA binding (Id) proteins bind to and inhibit the function of basic helix-loop-helix (bHLH) transcription factors including those that regulate pancreatic development. Moreover, bone morphogenetic proteins (BMPs) regulate the expression of Ids. We hypothesized that BMP4 and Id proteins play a role in the expansion and differentiation of epithelial progenitor cells. We demonstrate that BMP4 induces the expression of Id2 along with the expansion of AR42J pancreatic epithelial cells. Furthermore, neutralization of BMP4 significantly reduced duct epithelial cell expansion in a mouse model of islet regeneration. BMP4 stimulation promotes Id2 binding to the bHLH transcription factor NeuroD, which is required for the differentiation of pancreatic islet cells. Therefore, our results indicate that BMP4 stimulation blocks the differentiation of endocrine progenitor cells and instead promotes their expansion thereby revealing a novel paradigm of signaling explaining the balance between expansion and differentiation of pancreatic duct epithelial progenitors. Understanding the mechanisms of BMP and Id function elucidates a key step during pancreas embryogenesis, which is important knowledge for expanding pancreatic progenitors in vitro.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Inhibitor of Differentiation Protein 2/metabolism , Insulin-Secreting Cells/metabolism , Stem Cells/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Gene Expression Regulation, Developmental , Inhibitor of Differentiation Protein 2/genetics , Leukemia, Plasma Cell , Mice , Nerve Tissue Proteins/metabolism , Pancreas/growth & development , Pancreatic Ducts/cytology , Pancreatic Ducts/growth & development , Signal Transduction
18.
J Cell Mol Med ; 9(2): 331-44, 2005.
Article in English | MEDLINE | ID: mdl-15963253

ABSTRACT

Pancreatic islet transplantation represents an attractive approach for the treatment of diabetes. However, the limited availability of donor islets has largely hampered this approach. In this respect, the use of alternative sources of islets such as the ex vivo expansion and differentiation of functional endocrine cells for treating diabetes has become the major focus of diabetes research. Adult pancreatic stem cells /progenitor cells have yet to be recognized because limited markers exist for their identification. While the pancreas has the capacity to regenerate under certain circumstances, questions where adult pancreatic stem/progenitor cells are localized, how they are regulated, and even if the pancreas harbors a stem cell population need to be resolved. In this article, we review the recent achievements both in the identification as well as in the expansion of pancreatic stem/progenitor cells.


Subject(s)
Cell Proliferation , Pancreas/cytology , Stem Cells/cytology , Animals , Cell Differentiation/physiology , Embryo, Mammalian/cytology , Humans , Insulin/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/metabolism , Models, Biological , Pancreas/physiology , Pancreatic Ducts/cytology , Pancreatic Ducts/metabolism , Regeneration/physiology , Stem Cells/physiology
19.
Diabetes Metab Res Rev ; 19(5): 363-74, 2003.
Article in English | MEDLINE | ID: mdl-12951644

ABSTRACT

Diabetes mellitus results from the anatomical or functional loss of insulin-producing beta cells of the pancreas. Despite significant advances in current treatment, patients with diabetes still do not maintain optimal glucose levels and therefore face debilitating complications such as hypoglycemia, retinopathy or cardiovascular diseases later in life. Islet transplantation therefore holds great promise as an ultimate cure for diabetes. However, the shortage of availability of donor sources of islets for transplantation has largely hampered this therapy. In this respect, the use of alternative sources of islets such as the ex vivo culture and expansion and differentiation of functional endocrine cells for treating diabetes has been a major focus of diabetes research. The identity of the islet stem/progenitor cells has remained either elusive or at least equivocal because of the lack of cell markers for identification of these cells. Recent successes in studying the organogenesis of pancreas as well as in vitro islet progenitor cell identification studies have provided tremendous insight for the cell markers that are essential in the isolation and characterization of these cells prospectively both in vivo and in vitro. If we can identify the markers that will aid the isolation and purification of islet progenitor cells, or factors that determine pancreatic cell fate, we might be able to coerce them from turning into specific endocrine cells or pancreas in vitro. This article will focus on this subject and will review the latest achievements in the study of cell markers for islet progenitor cells.


Subject(s)
Islets of Langerhans/cytology , Stem Cells/cytology , Biomarkers , Cell Division , Diabetes Mellitus/surgery , Humans , Islets of Langerhans/physiology , Islets of Langerhans Transplantation , Regeneration , Stem Cells/physiology , Tissue Donors/supply & distribution , Tissue Expansion/methods
20.
Am J Physiol Endocrinol Metab ; 287(2): E247-54, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15039147

ABSTRACT

We assessed the function of the beta(C)-subunit of activin in hepatocytes. We studied the effect of conditioned medium of Chinese hamster ovary (CHO) cell line stably expressing the beta(C) gene (CHO-beta(C)) on growth of AML12 hepatocytes. We also examined the effect of recombinant activin C and transfection of the beta(C) gene by using adenovirus vector. CHO-beta(C) secreted activin C, a homodimer of the beta(C), as well as precursors of the beta(C). The conditioned medium of CHO-beta(C) increased both [(3)H]thymidine incorporation and the cell number in AML12 cells. It also supported survival of AML12 cells in a serum-free condition. Recombinant human activin C also increased both [(3)H]thymidine incorporation and the number of AML12 cells. Transfection of AML12 cells with the beta(C)-subunit led to the stimulation of [(3)H]thymidine incorporation. Analysis of the conditioned medium revealed that the beta(C)-subunit formed a heterodimer with the endogenous beta(A), the formation of which was dependent on the amount of beta(C) expressed. Recombinant activin C did not affect the binding of (125)I-activin A to its receptor or follistatin. These results indicate that activin C stimulates growth of AML12 cells. The beta(C)-subunit modifies the function of the beta(A)-subunit by multiple mechanisms.


Subject(s)
Hepatocytes/metabolism , Inhibin-beta Subunits/physiology , Animals , CHO Cells/metabolism , Cell Division/physiology , Cell Survival/physiology , Cells, Cultured , Cricetinae , Cricetulus , Culture Media, Conditioned/metabolism , Hepatocytes/cytology , Humans , Inhibin-beta Subunits/biosynthesis , Mice , Protein Conformation , Recombinant Proteins , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL