Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Chemosphere ; 346: 140480, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37879369

ABSTRACT

Maternal exposure to particulate matter derived from diesel exhaust has been shown to cause metabolic dysregulation, neurological problems, and increased susceptibility to diabetes in the offspring. Diesel exhaust is a major source of air pollution and the use of biodiesel (BD) and its blends have been progressively increasing throughout the world; however, studies on the health impact of BD vs. petrodiesel combustion-generated exhaust have been controversial in part, due to differences in the chemical and physical nature of the associated particulate matter (PM). To explore the long-term impact of prenatal exposure, pregnant mice were exposed to PM generated by combustion of petrodiesel (B0) and a 20% soy BD blend (B20) by intratracheal instillation during embryonic days 9-17 and allowed to deliver. Offspring were then followed for 52 weeks. We found that mother's exposure to B0 and B20 PM manifested in striking sex-specific phenotypes with respect to metabolic adaptation, maintenance of glucose homeostasis, and medial hypothalamic glial cell makeup in the offspring. The data suggest PM exposure limited to a narrower critical developmental window may be compensated for by the mother and/or the fetus by altered metabolic programming in a marked sex-specific and fuel-derived PM-specific manner, leading to sex-specific risk for diseases related to environmental exposure later in life.


Subject(s)
Air Pollutants , Air Pollution , Male , Female , Mice , Animals , Particulate Matter/toxicity , Particulate Matter/analysis , Vehicle Emissions/toxicity , Vehicle Emissions/analysis , Biofuels/toxicity , Biofuels/analysis , Environmental Exposure , Gasoline/analysis , Air Pollutants/toxicity
2.
J Biol Chem ; 292(30): 12449-12459, 2017 07 28.
Article in English | MEDLINE | ID: mdl-28487366

ABSTRACT

The onset of type 2 diabetes is characterized by transition from successful to failed insulin secretory compensation to obesity-related insulin resistance and dysmetabolism. Energy-rich diets in rodents are commonly studied models of compensatory increases in both insulin secretion and ß cell mass. However, the mechanisms of these adaptive responses are incompletely understood, and it is also unclear why these responses eventually fail. We measured the temporal trends of glucose homeostasis, insulin secretion, ß cell morphometry, and islet gene expression in C57BL/6NTac mice fed a 60% high-fat diet (HFD) or control diet for up to 16 weeks. A 2-fold increased hyperinsulinemia was maintained for the first 4 weeks of HFD feeding and then further increased through 16 weeks. ß cell mass increased progressively starting at 4 weeks, principally through nonproliferative growth. Insulin sensitivity was not significantly perturbed until 11 weeks of HFD feeding. Over the first 8 weeks, we observed two distinct waves of increased expression of ß cell functional and prodifferentiation genes. This was followed by activation of the unfolded protein response at 8 weeks and overt ß cell endoplasmic reticulum stress at 12-16 weeks. In summary, ß cell adaptation to an HFD in C57BL/6NTac mice entails early insulin hypersecretion and a robust growth phase along with hyperexpression of related genes that begin well before the onset of observed insulin resistance. However, continued HFD exposure results in cessation of gene hyperexpression, ß cell functional failure, and endoplasmic reticulum stress. These data point to a complex but not sustainable integration of ß cell-adaptive responses to nutrient overabundance, obesity development, and insulin resistance.


Subject(s)
Diet, High-Fat/adverse effects , Hyperinsulinism/metabolism , Insulin-Secreting Cells/metabolism , Animals , Endoplasmic Reticulum Stress , Hyperinsulinism/pathology , Insulin/metabolism , Insulin-Secreting Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Obese , Time Factors
3.
J Biol Chem ; 288(35): 25440-25449, 2013 Aug 30.
Article in English | MEDLINE | ID: mdl-23788637

ABSTRACT

The molecular mechanisms and signaling pathways that drive islet ß-cell compensation and failure are not fully resolved. We have used in vitro and in vivo systems to show that FoxO1, an integrator of metabolic stimuli, inhibits PPARγ expression in ß-cells, thus transcription of its target genes (Pdx1, glucose-dependent insulinotropic polypeptide (GIP) receptor, and pyruvate carboxylase) that are important regulators of ß-cell function, survival, and compensation. FoxO1 inhibition of target gene transcription is normally relieved when upstream activation induces its translocation from the nucleus to the cytoplasm. Attesting to the central importance of this pathway, islet expression of PPARγ and its target genes was enhanced in nondiabetic insulin-resistant rats and markedly reduced with diabetes induction. Insight into the impaired PPARγ signaling with hyperglycemia was obtained with confocal microscopy of pancreas sections that showed an intense nuclear FoxO1 immunostaining pattern in the ß-cells of diabetic rats in contrast to the nuclear and cytoplasmic FoxO1 in nondiabetic rats. These findings suggest a FoxO1/PPARγ-mediated network acting as a core component of ß-cell adaptation to metabolic stress, with failure of this response from impaired FoxO1 activation causing or exacerbating diabetes.


Subject(s)
Cell Nucleus/metabolism , Diabetes Mellitus, Experimental/metabolism , Forkhead Transcription Factors/metabolism , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , PPAR gamma/metabolism , Transcription, Genetic , Active Transport, Cell Nucleus , Animals , Cell Line , Cell Nucleus/genetics , Cell Nucleus/pathology , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , PPAR gamma/genetics , Rats , Rats, Zucker
4.
Endocrinology ; 152(4): 1300-13, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21285317

ABSTRACT

Circulating levels of matrix metalloproteinases (MMPs) and their endogenous inhibitors, tissue inhibitor of metalloproteinases (TIMPs), are altered in human obesity and may contribute to its pathology. TIMP-2 exerts MMP-dependent (MMP inhibition and pro-MMP-2 activation) and MMP-independent functions. To assess the role of TIMP-2 in a murine model of nutritionally induced obesity, weight gain in wild-type and TIMP-2 deficient [knockout (KO)] mice fed a chow or high-fat diet (HFD) was determined. The effects of diet on glucose tolerance and insulin sensitivity, as well as pancreatic ß-cell and adipocyte physiology, were assessed. Chow-fed TIMP-2 KO mice of both sexes became obese but maintained relatively normal glucose tolerance and insulin sensitivity. Obesity was exacerbated on the HFD. However, HFD-fed male, but not female, TIMP-2 KO mice developed insulin resistance with reduced glucose transporter 2 and pancreatic and duodenal homeobox 1 levels, despite increased ß-cell mass and hyperplasia. Thus, although ß-cell mass was increased, HFD-fed male TIMP-2 KO mice develop diabetes likely due to ß-cell exhaustion and failure. TIMP-2 mRNA, whose expression was greatest in sc adipose tissue, was down-regulated in HFD-fed wild-type males, but not females. Furthermore, HFD increased membrane type 1-MMP (MMP-14) expression and activity in male, but not female, sc adipose tissue. Strikingly, MMP-14 expression increased to a greater extent in TIMP-2 KO males and was associated with decreased adipocyte collagen. Taken together, these findings demonstrate a role for TIMP-2 in maintaining extracellular matrix integrity necessary for normal ß-cell and adipocyte physiology and that loss of extracellular matrix integrity may underlie diabetic and obesogenic phenotypes.


Subject(s)
Obesity/metabolism , Tissue Inhibitor of Metalloproteinase-2/deficiency , Tissue Inhibitor of Metalloproteinase-2/metabolism , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Blotting, Western , Dietary Fats/adverse effects , Female , Immunoenzyme Techniques , Insulin Resistance/genetics , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Leptin/metabolism , Male , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Obesity/chemically induced , Polymerase Chain Reaction , Sex Factors , Tissue Inhibitor of Metalloproteinase-2/genetics , Weight Gain/genetics
5.
Am J Physiol Endocrinol Metab ; 299(5): E786-93, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20716695

ABSTRACT

The physiological mechanisms that preserve pancreatic ß-cell mass (BCM) are not fully understood. Although the regulation of islet function by the autonomic nervous system (ANS) is well established, its potential roles in BCM homeostasis and compensatory growth have not been adequately explored. The parasympathetic vagal branch of the ANS serves to facilitate gastrointestinal function, metabolism, and pancreatic islet regulation of glucose homeostasis, including insulin secretion. Given the functional importance of the vagus nerve and its branches to the liver, gut, and pancreas in control of digestion, motility, feeding behavior, and glucose metabolism, it may also play a role in BCM regulation. We have begun to examine the potential roles of the parasympathetic nervous system in short-term BCM maintenance by performing a selective bilateral celiac branch-vagus nerve transection (CVX) in normal Sprague-Dawley rats. CVX resulted in no detectable effects on basic metabolic parameters or food intake through 1 wk postsurgery. Although there were no differences in BCM or apoptosis in this 1-wk time frame, ß-cell proliferation was reduced 50% in the CVX rats, correlating with a marked reduction in activated protein kinase B/Akt. Unexpectedly, acinar proliferation was increased 50% in these rats. These data suggest that the ANS, via the vagus nerve, contributes to the regulation of BCM maintenance at the level of cell proliferation and may also mediate the drive for enhanced growth under physiological conditions when insulin requirements have increased. Furthermore, the disparate effects of CVX on ß-cell and acinar cells suggest that the endocrine and exocrine pancreas respond to different neural signals in regard to mass homeostasis.


Subject(s)
Insulin-Secreting Cells/physiology , Vagus Nerve/physiology , Animals , Apoptosis/physiology , Blood Glucose/analysis , Body Weight/physiology , Cell Growth Processes/physiology , Drinking/physiology , Eating/physiology , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test , Insulin/blood , Male , Microscopy, Confocal , Rats , Rats, Sprague-Dawley , Vagus Nerve/surgery , Vagus Nerve/ultrastructure
6.
Diabetes ; 59(6): 1445-50, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20332343

ABSTRACT

OBJECTIVE: We previously showed that peroxisome proliferator-activated receptor (PPAR)-gamma in beta-cells regulates pdx-1 transcription through a functional PPAR response element (PPRE). Gene Bank blast for a homologous nucleotide sequence revealed the same PPRE within the rat glucose-dependent insulinotropic polypeptide receptor (GIP-R) promoter sequence. We investigated the role of PPARgamma in GIP-R transcription. RESEARCH DESIGN AND METHODS: Chromatin immunoprecipitation assay, siRNA, and luciferase gene transcription assay in INS-1 cells were performed. Islet GIP-R expression and immunohistochemistry studies were performed in pancreas-specific PPARgamma knockout mice (PANC PPARgamma(-/-)), normoglycemic 60% pancreatectomy rats (Px), normoglycemic and hyperglycemic Zucker fatty (ZF) rats, and mouse islets incubated with troglitazone. RESULTS: In vitro studies of INS-1 cells confirmed that PPAR-gamma binds to the putative PPRE sequence and regulates GIP-R transcription. In vivo verification was shown by a 70% reduction in GIP-R protein expression in islets from PANC PPARgamma(-/-) mice and a twofold increase in islets of 14-day post-60% Px Sprague-Dawley rats that hyperexpress beta-cell PPARgamma. Thiazolidinedione activation (72 h) of this pathway in normal mouse islets caused a threefold increase of GIP-R protein and a doubling of insulin secretion to 16.7 mmol/l glucose/10 nmol/l GIP. Islets from obese normoglycemic ZF rats had twofold increased PPARgamma and GIP-R protein levels versus lean rats, with both lowered by two-thirds in ZF rats made hyperglycemic by 60% Px. CONCLUSIONS: Our studies have shown physiologic and pharmacologic regulation of GIP-R expression in beta-cells by PPARgamma signaling. Also disruption of this signaling pathway may account for the lowered beta-cell GIP-R expression and resulting GIP resistance in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/physiology , PPAR gamma/physiology , Receptors, Gastrointestinal Hormone/genetics , Animals , Chromans/pharmacology , Drug Resistance , Gastric Inhibitory Polypeptide/physiology , Glucose/physiology , Male , Mice , PPAR gamma/deficiency , PPAR gamma/genetics , Pancreatectomy , Rats , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Gastrointestinal Hormone/physiology , Thiazolidinediones/pharmacology , Troglitazone
7.
J Biol Chem ; 283(47): 32462-70, 2008 Nov 21.
Article in English | MEDLINE | ID: mdl-18718916

ABSTRACT

We reported that peroxisome proliferator-activated receptor gamma (PPARgamma) transcriptionally regulates the beta-cell differentiation factor pancreatic duodenal homeobox (PDX)-1 based on in vitro RNA interference studies. We have now studied mice depleted of PPARgamma within the pancreas (PANC PPARgamma(-/-)) created by a Cre/loxP recombinase system, with Cre driven by the pdx-1 promoter. Male PANC PPARgamma(-/-) mice were hyperglycemic at 8 weeks of age (8.1+/-0.2 mM versus 6.4+/-0.3 mM, p=0.009) with islet cytoarchitecture and pancreatic mass of islet beta-cells that were indistinguishable from the controls. Islet PDX-1 mRNA (p=0.001) and protein levels (p=0.003) were lowered 60 and 40%, respectively, in tandem with impaired glucose-induced insulin secretion and loss of thiazolidinedione-induced increase in PDX-1 expression. We next identified a putative PPAR-response element (PPRE) in the mouse pdx-1 promoter with substantial homology to the corresponding region of the human PDX-1 promoter. Electrophoretic mobility supershift assays with nuclear extracts from beta-cell lines and mouse islets, also in vitro translated PPARgamma and retinoid X receptor, and chromatin immunoprecipitation analysis demonstrated specific binding of PPARgamma and retinoid X receptor to the human and mouse pdx-1 x PPREs. Transient transfection assays of beta-cells with reporter constructs of mutated PPREs showed dramatically reduced pdx-1 promoter activity. In summary, we have presented in vivo and in vitro evidence showing PPARgamma regulation of pdx-1 transcription in beta-cells, plus our results support an important regulatory role for PPARgamma in beta-cell physiology and thiazolidinedione pharmacology of type 2 diabetes.


Subject(s)
Gene Expression Regulation , Homeodomain Proteins/genetics , PPAR gamma/metabolism , Promoter Regions, Genetic , Trans-Activators/genetics , Animals , Base Sequence , Humans , Hyperglycemia/genetics , In Vitro Techniques , Insulin-Secreting Cells , Islets of Langerhans/metabolism , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Response Elements , Sequence Homology, Nucleic Acid
8.
Am J Physiol Endocrinol Metab ; 294(4): E679-87, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18230696

ABSTRACT

The physiological mechanisms underlying pancreatic beta-cell mass (BCM) homeostasis are complex and not fully resolved. Here we examined the factors contributing to the increased BCM following a mild glucose infusion (GI) whereby normoglycemia was maintained through 96 h. We used morphometric and immunochemical methods to investigate enhanced beta-cell growth and survival in Sprague-Dawley rats. BCM was elevated >2.5-fold over saline-infused control rats by 48 h and increased modestly thereafter. Unexpectedly, increases in beta-cell proliferation were not observed at any time point through 4 days. Instead, enhanced numbers of insulin(+) cell clusters and small islets (400-12,000 microm(2); approximately 23- to 124-microm diameter), mostly scattered among the acini, were observed in the GI rats by 48 h despite no difference in the numbers of medium to large islets. We previously showed that increased beta-cell growth in rodent models of insulin resistance and pancreatic regeneration involves increased activated Akt/PKB, a key beta-cell signaling intermediate, in both islets and endocrine cell clusters. GI in normal rats also leads to increased Akt activation in islet beta-cells, as well as in insulin(+) and insulin(-) cells in the common duct epithelium and endocrine clusters. This correlated with strong Pdx1 expression in these same cells. These results suggest that mechanisms other than proliferation underlie the rapid beta-cell growth response following a mild GI in the normal rat and involve Akt-regulated enhanced beta-cell survival potential and neogenesis from epithelial precursors.


Subject(s)
Glucose/pharmacology , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Regeneration/physiology , Animals , Cell Division/drug effects , Cell Division/physiology , Cell Survival/drug effects , Cell Survival/physiology , Insulin/blood , Insulin-Secreting Cells/physiology , Islets of Langerhans/cytology , Islets of Langerhans/growth & development , Islets of Langerhans/physiology , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Regeneration/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sodium Chloride/pharmacology
9.
Diabetes ; 56(1): 88-95, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17192469

ABSTRACT

In the 60% pancreatectomy (Px) rat model of beta-cell adaptation, normoglycemia is maintained by an initial week of beta-cell hyperplasia that ceases and is followed by enhanced beta-cell function. It is unknown how this complex series of events is regulated. We studied isolated islets and pancreas sections from 14-day post-Px versus sham-operated rats and observed a doubling of beta-cell nuclear peroxisome proliferator-activated receptor (PPAR)-gamma protein, along with a 2-fold increase in nuclear pancreatic duodenal homeobox (Pdx)-1 protein and a 1.4-fold increase in beta-cell nuclear Nkx6.1 immunostaining. As PPAR-gamma activation is known to both lower proliferation and have prodifferentiation effects in many tissues, we studied PPAR-gamma actions in INS-1 cells. A 3-day incubation with the PPAR-gamma agonist troglitazone reduced proliferation and increased Pdx-1 and Nkx6.1 immunostaining, along with glucokinase and GLUT2. Also, a 75% knockdown of PPAR-gamma using RNA interference lowered the mRNA levels of Pdx-1, glucokinase, GLUT2, and proinsulin II by more than half. Our results show a dual effect of PPAR-gamma in INS-1 cells: to curtail proliferation and promote maturation, the latter via enhanced expression of Pdx-1 and Nkx6.1. Additional studies are needed to determine whether there is a regulatory role for PPAR-gamma signaling in the beta-cell adaptation following a 60% Px in rats.


Subject(s)
Homeodomain Proteins/genetics , PPAR gamma/physiology , Trans-Activators/genetics , Animals , Cell Nucleus/physiology , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/physiology , Male , Microscopy, Confocal , PPAR alpha/genetics , Pancreatectomy , RNA/genetics , RNA/isolation & purification , Rats , Rats, Sprague-Dawley , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
10.
Diabetes ; 55(12): 3289-98, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17130472

ABSTRACT

beta-Cell mass is determined by a dynamic balance of proliferation, neogenesis, and apoptosis. The precise mechanisms underlying compensatory beta-cell mass (BCM) homeostasis are not fully understood. To evaluate the processes that maintain normoglycemia and regulate BCM during pancreatic regeneration, C57BL/6 mice were analyzed for 15 days following 60% partial pancreatectomy (Px). BCM increased in Px mice from 2 days onwards and was approximately 68% of the shams by 15 days, partly due to enhanced beta-cell proliferation. A transient approximately 2.8-fold increase in the prevalence of beta-cell clusters/small islets at 2 days post-Px contributed substantially to BCM augmentation, followed by an increase in the number of larger islets at 15 days. To evaluate the signaling mechanisms that may regulate this compensatory growth, we examined key intermediates of the insulin signaling pathway. We found insulin receptor substrate (IRS)2 and enhanced-activated Akt immunoreactivity in islets and ducts that correlated with increased pancreatic duodenal homeobox (PDX)1 expression. In contrast, forkhead box O1 expression was decreased in islets but increased in ducts, suggesting distinct PDX1 regulatory mechanisms in these tissues. Px animals acutely administered insulin exhibited further enhancement in insulin signaling activity. These data suggest that the IRS2-Akt pathway mediates compensatory beta-cell growth by activating beta-cell proliferation with an increase in the number of beta-cell clusters/small islets.


Subject(s)
Insulin-Secreting Cells/physiology , Pancreatectomy , Actins/metabolism , Animals , Blood Glucose , Cell Division , Cyclin D2 , Cyclins/metabolism , Immunoblotting , Insulin-Secreting Cells/cytology , Islets of Langerhans/anatomy & histology , Islets of Langerhans/cytology , Islets of Langerhans/physiology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Regeneration
11.
Gene Expr Patterns ; 6(8): 971-7, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16750430

ABSTRACT

Multipotent progenitor cells self renew throughout an animal's lifetime and can differentiate to give rise to different cell types. Before we can fully understand the developmental potential of progenitor cells and control their differentiation both in vivo and in vitro as stem cells, identification and characterization of the genes that control stem cell fate must first be obtained. Foxd3, a member of the forkhead family of transcriptional regulators, is required for the maintenance of embryonic stem cells and trophoblast stem cells of the early mouse embryo. We describe here the expression of this protein in the developing pancreas. Foxd3 is expressed in most beta cells and infrequently in alpha and PP cells but is not expressed in somatostatin cells. The subcellular localization of Foxd3 varies with fat content in the diet; with a high fat diet the protein is found primarily in the cytoplasm while a low fat diet results in nuclear localization. Foxd3 is differentially localized in a rat model of diabetes: it is nuclear in ZDF rats but cytoplasmic in their lean counterparts. Foxd3 is nuclear in Lep(Ob/Ob) mice.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Developmental , Pancreas/embryology , Pancreas/growth & development , Pancreas/metabolism , Repressor Proteins/metabolism , Animals , Cell Lineage , Cell Nucleus/metabolism , Cells, Cultured , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Embryo, Mammalian/metabolism , Embryonic Development/physiology , Female , Humans , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Organ Specificity , Rats , Rats, Zucker , Tissue Distribution
12.
Diabetes ; 54(8): 2294-304, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16046294

ABSTRACT

The physiological mechanisms underlying the compensatory growth of beta-cell mass in insulin-resistant states are poorly understood. Using the insulin-resistant Zucker fatty (fa/fa) (ZF) rat and the corresponding Zucker lean control (ZLC) rat, we investigated the factors contributing to the age-/obesity-related enhancement of beta-cell mass. A 3.8-fold beta-cell mass increase was observed in ZF rats as early as 5 weeks of age, an age that precedes severe insulin resistance by several weeks. Closer investigation showed that ZF rat pups were not born with heightened beta-cell mass but developed a modest increase over ZLC rats by 20 days that preceded weight gain or hyperinsulinemia that first developed at 24 days of age. In these ZF pups, an augmented survival potential of beta-cells of ZF pups was observed by enhanced activated (phospho-) Akt, phospho-BAD, and Bcl-2 immunoreactivity in the postweaning period. However, increased beta-cell proliferation in the ZF rats was only detected at 31 days of age, a period preceding massive beta-cell growth. During this phase, we also detected an increase in the numbers of small beta-cell clusters among ducts and acini, increased duct pancreatic/duodenal homeobox-1 (PDX-1) immunoreactivity, and an increase in islet number in the ZF rats suggesting duct- and acini-mediated heightened beta-cell neogenesis. Interestingly, in young ZF rats, specific cells associated with ducts, acini, and islets exhibited an increased frequency of PDX-1+/phospho-Akt+ staining, indicating a potential role for Akt in beta-cell differentiation. Thus, several adaptive mechanisms account for the compensatory growth of beta-cells in ZF rats, a combination of enhanced survival and neogenesis with a transient rise in proliferation before 5 weeks of age, with Akt serving as a potential mediator in these processes.


Subject(s)
Aging , Insulin Resistance , Islets of Langerhans/pathology , Obesity/pathology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Animals , Cell Differentiation , Cell Division , Cell Survival , Homeodomain Proteins/analysis , In Situ Nick-End Labeling , Islets of Langerhans/chemistry , Male , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Zucker , Signal Transduction , Trans-Activators/analysis
SELECTION OF CITATIONS
SEARCH DETAIL