Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters











Database
Language
Publication year range
1.
J Cell Mol Med ; 23(4): 2890-2900, 2019 04.
Article in English | MEDLINE | ID: mdl-30710421

ABSTRACT

Both type 2 diabetes (T2D) and obesity are characterized by excessive hyperlipidaemia and subsequent lipid droplet (LD) accumulation in adipose tissue. To investigate whether LDs also accumulate in ß-cells of T2D patients, we assessed the expression of PLIN2, a LD-associated protein, in non-diabetic (ND) and T2D pancreata. We observed an up-regulation of PLIN2 mRNA and protein in ß-cells of T2D patients, along with significant changes in the expression of lipid metabolism, apoptosis and oxidative stress genes. The increased LD buildup in T2D ß-cells was accompanied by inhibition of nuclear translocation of TFEB, a master regulator of autophagy and by down-regulation of lysosomal biomarker LAMP2. To investigate whether LD accumulation and autophagy were influenced by diabetic conditions, we used rat INS-1 cells to model the effects of hyperglycaemia and hyperlipidaemia on autophagy and metabolic gene expression. Consistent with human tissue, both LD formation and PLIN2 expression were enhanced in INS-1 cells under hyperglycaemia, whereas TFEB activation and autophagy gene expression were significantly reduced. Collectively, these results suggest that lipid clearance and overall homeostasis is markedly disrupted in ß-cells under hyperglycaemic conditions and interventions ameliorating lipid clearance could be beneficial in reducing functional impairments in islets caused by glucolipotoxicity.


Subject(s)
Autophagy , Diabetes Mellitus, Type 2/physiopathology , Gene Expression Regulation , Hyperglycemia/pathology , Hyperlipidemias/pathology , Insulin-Secreting Cells/pathology , Lipids/analysis , Animals , Apoptosis , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Case-Control Studies , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Humans , Hyperglycemia/etiology , Hyperglycemia/metabolism , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Insulin-Secreting Cells/metabolism , Insulinoma/genetics , Insulinoma/metabolism , Insulinoma/pathology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Lysosomes/metabolism , Lysosomes/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Perilipin-2/genetics , Perilipin-2/metabolism , Rats , Tumor Cells, Cultured
2.
Differentiation ; 90(4-5): 77-90, 2015.
Article in English | MEDLINE | ID: mdl-26558987

ABSTRACT

Regeneration of ß-cells in diabetic patients is an important goal of diabetes research. Islet Neogenesis Associated Protein (INGAP) was discovered in the partially duct-obstructed hamster pancreas. Its bioactive fragment, pentadecapeptide 104-118 (INGAP-P), has been shown to reverse diabetes in animal models and to improve glucose homeostasis in patients with diabetes in clinical trials. Further development of INGAP as a therapy for diabetes requires identification of target cells in the pancreas and characterization of the mechanisms of action. We hypothesized that adult human pancreatic ductal cells retain morphogenetic plasticity and can be induced by INGAP to undergo endocrine differentiation. To test this hypothesis, we treated the normal human pancreatic ductal cell line (HPDE) with either INGAP-P or full-length recombinant protein (rINGAP) for short-term periods. Our data show that this single drug treatment induces both proliferation and transdifferentiation of HPDE cells, the latter being characterized by the rapid sequential activation of endocrine developmental transcription factors Pdx-1, Ngn3, NeuroD, IA-1, and MafA and subsequently the expression of insulin at both the mRNA and the protein levels. After 7 days, C-peptide was detected in the supernatant of INGAP-treated cells, reflecting their ability to secrete insulin. The magnitude of differentiation was enhanced by embedding the cells in Matrigel, which led to islet-like cluster formation. The islet-like clusters cells stained positive for nuclear Pdx-1 and Glut 2 proteins, and were expressing Insulin mRNA. These new data suggest that human adult pancreatic ductal cells retain morphogenetic plasticity and demonstrate that a short exposure to INGAP triggers their differentiation into insulin-expressing cells in vitro. In the context of the urgent search for a regenerative and/or cellular therapy for diabetes, these results make INGAP a promising therapeutic candidate.


Subject(s)
Antigens, Neoplasm/pharmacology , Biomarkers, Tumor/pharmacology , C-Peptide/metabolism , Cell Proliferation/drug effects , Cell Transdifferentiation/drug effects , Cytokines/pharmacology , Diabetes Mellitus/therapy , Insulin/metabolism , Peptide Fragments/pharmacology , Transcription Factors/physiology , Adult , Animals , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , Cell Line , Collagen/pharmacology , Cricetinae , Drug Combinations , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Laminin/pharmacology , Lectins, C-Type/genetics , Pancreatic Ducts , Pancreatitis-Associated Proteins , Proteoglycans/pharmacology , Recombinant Proteins/pharmacology
3.
Am J Physiol Endocrinol Metab ; 303(7): E917-27, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22850686

ABSTRACT

Islet neogenesis-associated protein (INGAP) was discovered in the partially duct-obstructed hamster pancreas as a factor inducing formation of new duct-associated islets. A bioactive portion of INGAP, INGAP(104-118) peptide (INGAP-P), has been shown to have neogenic and insulin-potentiating activity in numerous studies, including recent phase 2 clinical trials that demonstrated improved glucose homeostasis in both type 1 and type 2 diabetic patients. Aiming to improve INGAP-P efficacy and to understand its mechanism of action, we cloned the full-length protein (rINGAP) and compared the signaling events induced by the protein and the peptide in RIN-m5F cells that respond to INGAP with an increase in proliferation. Here, we show that, although both rINGAP and INGAP-P signal via the Ras/Raf/ERK pathway, rINGAP is at least 100 times more efficient on a molar basis than INGAP-P. For either ligand, ERK1/2 activation appears to be pertussis toxin sensitive, suggesting involvement of a G protein-coupled receptor(s). However, there are clear differences between the peptide and the protein in interactions with the cell surface and in the downstream signaling. We demonstrate that fluorescent-labeled rINGAP is characterized by clustering on the membrane and by slow internalization (≤5 h), whereas INGAP-P does not cluster and is internalized within minutes. Signaling by rINGAP appears to involve Src, in contrast to INGAP-P, which appears to activate Akt in addition to the Ras/Raf/ERK1/2 pathway. Thus our data suggest that interactions of INGAP with the cell surface are important to consider for further development of INGAP as a pharmacotherapy for diabetes.


Subject(s)
Antigens, Neoplasm/pharmacology , Biomarkers, Tumor/pharmacology , Cytokines/pharmacology , Islets of Langerhans/drug effects , Peptide Fragments/pharmacology , Recombinant Proteins/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Cloning, Molecular , Lectins, C-Type , Pancreatitis-Associated Proteins , Pertussis Toxin/pharmacology , Rats , Signal Transduction/drug effects
4.
J Endocrinol ; 211(3): 231-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21933872

ABSTRACT

Given the inherent therapeutic potential of the morphogenetic plasticity of adult human islets, the identification of factors controlling their cellular differentiation is of interest. The epidermal growth factor (EGF) family has been identified previously in the context of pancreatic organogenesis. We examined the role of EGF in an in vitro model whereby adult human islets are embedded in a collagen gel and dedifferentiated into duct-like epithelial structures (DLS). We demonstrated that DLS formation was EGF dependent, while residual DLS formation in the absence of added EGF was abrogated by EGF receptor inhibitor treatment. With respect to signaling, EGF administration led to an increase in c-Jun NH2-terminal kinase (JNK) phosphorylation early in DLS formation and in AKT and extracellular signal-regulated kinase (ERK) phosphorylation late in the process of DLS formation, concomitant with the increased proliferation of dedifferentiated cells. In the absence of EGF, these phosphorylation changes are not seen and the typical increase in DLS epithelial cell proliferation seen after 10 days in culture is attenuated. Thus, in our model, EGF is necessary for islet cell dedifferentiation, playing an important role in both the onset of DLS formation (through JNK) and in the proliferation of these dedifferentiated cells (through AKT and ERK).


Subject(s)
Cell Dedifferentiation/drug effects , Epidermal Growth Factor/pharmacology , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Adult , Base Sequence , Cell Dedifferentiation/genetics , Cell Dedifferentiation/physiology , DNA Primers/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Gene Expression , Humans , In Vitro Techniques , Islets of Langerhans/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Ligands , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
5.
Protein Expr Purif ; 69(1): 1-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19635567

ABSTRACT

Islet Neogenesis Associated Protein (INGAP) is implicated in pancreatic islet neogenesis. INGAP peptide, a pentadecapeptide comprising amino acids 104-118, reverses diabetes in rodents and improves glucose homeostasis in patients with diabetes. The mechanism of INGAP action is unknown, but such studies would benefit from the availability of the full-length recombinant protein (rINGAP). Here we report the production of rINGAP from 293-SF cells following lentiviral transduction, and its characterization by MALDI-TOF and Q-TOF Mass Spectrometry, and HPLC. Importantly, we show that rINGAP exhibits 100x the bioactivity of INGAP peptide on a molar basis in an in vitro assay of human islet regeneration.


Subject(s)
Antigens, Neoplasm/biosynthesis , Biomarkers, Tumor/biosynthesis , Lectins, C-Type/biosynthesis , Recombinant Proteins/biosynthesis , Amino Acid Sequence , Animals , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/genetics , Antigens, Neoplasm/isolation & purification , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Biomarkers, Tumor/isolation & purification , Cells, Cultured , Chromatography, High Pressure Liquid , Cricetinae , Gene Expression Regulation , Humans , Islets of Langerhans/physiology , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lectins, C-Type/isolation & purification , Lentivirus/genetics , Mass Spectrometry , Mesocricetus , Molecular Sequence Data , Molecular Weight , Pancreatitis-Associated Proteins , RNA, Messenger/genetics , RNA, Messenger/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Regeneration/physiology , Subcellular Fractions/metabolism , Transduction, Genetic
6.
J Bacteriol ; 189(16): 5976-86, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17557828

ABSTRACT

We identified a single open reading frame that is strongly similar to ArcR, a member of the Crp/Fnr family of bacterial transcriptional regulators, in all sequenced Staphylococcus aureus genomes. The arcR gene encoding ArcR forms an operon with the arginine deiminase (ADI) pathway genes arcABDC that enable the utilization of arginine as a source of energy for growth under anaerobic conditions. In this report, we show that under anaerobic conditions, S. aureus growth is subject to glucose catabolic repression and is enhanced by arginine. Likewise, glucose and arginine have reciprocal effects on the transcription of the arcABDCR genes. Furthermore, we show using a mutant deleted for arcR that the transcription of the arc operon under anaerobic conditions depends strictly on a functional ArcR. These findings are supported by proteome analyses, which showed that under anaerobic conditions the expression of the ADI catabolic proteins depends on ArcR. Bioinformatic analysis of S. aureus ArcR predicts an N-terminal nucleotide binding domain and a C-terminal helix-turn-helix DNA binding motif. ArcR binds to a conserved Crp-like sequence motif, TGTGA-N(6)-TCACA, present in the arc promoter region and thereby activates the expression of the ADI pathway genes. Crp-like sequence motifs were also found in the regulatory regions of some 30 other S. aureus genes mostly encoding anaerobic enzymatic systems, virulence factors, and regulatory systems. ArcR was tested and found to bind to the regulatory regions of four such genes, adh1, lctE, srrAB, and lukM. In one case, for lctE, encoding l-lactate dehydrogenase, ArcR was able to bind only in the presence of cyclic AMP. These observations suggest that ArcR is likely to play an important role in the expression of numerous genes required for anaerobic growth.


Subject(s)
Arginine/metabolism , Bacterial Proteins/physiology , DNA-Binding Proteins/physiology , Hydrolases/metabolism , Staphylococcus aureus/genetics , Gene Expression Regulation, Bacterial , Hydrolases/genetics , Multigene Family , Operon/genetics , Staphylococcus aureus/metabolism , Transcription, Genetic
7.
J Endocrinol ; 191(1): 65-81, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17065390

ABSTRACT

Restoration of a functional beta-cell mass in a patient with diabetes may hold the key for curing the disease. In recent years, there has been increasing interest in the development of new strategies to induce beta-cell regeneration and new islet formation in situ and a role for Reg proteins has been suggested. One such protein, islet neogenesis associated protein (INGAP), is a member of the Reg3 family of proteins and has been shown to induce islet neogenesis. Elucidation of the mechanisms and factors involved in the regulation of expression of INGAP and related proteins is, therefore, of great importance. Here, we report the establishment of the first in vitro tissue model of INGAP expression that consists of epithelial cystic structures derived from hamster pancreatic acinar tissue cultured in collagen matrix. The objective of this study was to characterize INGAP expression in this model and to investigate the role of pro-inflammatory cytokines and growth factors. Using quantitative reverse transcriptase PCR, we show that INGAP expression correlates with cyst formation and size suggesting the involvement of intra-luminal pressure associated with cyst growth. We also demonstrate for the first time that INGAP gene expression was significantly induced by treatment with interleukin (IL)-6 and further enhanced by a combination of IL-6 with dexamethazone and nicotinamide. Additionally, our data suggest that the effect of IL-6 on INGAP expression is mediated via the JAK/STAT3 signaling pathway. In summary, the in vitro model of INGAP expression described here represents an important step in the development of strategies for the use of INGAP and related proteins as islet neogenic agents in the pharmacotherapy of both type-1 and type-2 diabetes.


Subject(s)
Cytokines/pharmacology , Gene Expression Regulation , Lectins, C-Type/genetics , Models, Animal , Pancreas, Exocrine/metabolism , Pancreas, Exocrine/pathology , Regeneration , Animals , Blotting, Western/methods , Cell Differentiation , Collagen , Cricetinae , DNA Primers , Dactinomycin/pharmacology , Genetic Engineering , Interferon-gamma/pharmacology , Interleukin-1beta/pharmacology , Interleukin-6/pharmacology , Lectins, C-Type/analysis , Lectins, C-Type/metabolism , Male , Mesocricetus , Microscopy, Confocal , Pancreas, Exocrine/ultrastructure , Pancreatic Ducts/physiology , Pancreatic Ducts/ultrastructure , Pancreatitis-Associated Proteins , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Tissue Culture Techniques , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL