Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
J Appl Toxicol ; 40(5): 600-618, 2020 05.
Article in English | MEDLINE | ID: mdl-31884710

ABSTRACT

Tris(2-ethylhexyl) phosphate (TEHP, CAS no. 78-42-2) is a plasticizer and a flame retardant, while di(2-ethylhexyl) phosphoric acid (DEHPA, CAS no. 298-07-7) is an oil additive and extraction solvent. Publicly-available information on repeated exposure to these two related organophosphate compounds is fragmentary. Hence, adult male and female Fischer rats were exposed to TEHP (300, 1000 and 3000 mg/kg body weight [BW]/day) or DEHPA (20, 60 and 180 mg/kg BW/day) by gavage for 28 consecutive days, to assess and compare their toxicities. Although significantly impaired BW gains and evidence of TEHP enzymatic hydrolysis to DEHPA were observed only in males, exposures to the highest TEHP and DEHPA doses often resulted in similar alterations of hematology, serum clinical chemistry and liver enzymatic activities in both males and females. The squamous epithelial hyperplasia and hyperkeratosis observed in the non-glandular forestomach of rats exposed to the middle and high DEHPA doses were most likely caused by the slightly corrosive nature of this chemical. Although tubular degeneration and spermatid retention were observed only in the testes of males exposed to the highest TEHP dose, numerous periodic acid-Schiff stained crystalline inclusions were observed in testis interstitial cells at all TEHP dose levels. No-observed-adverse-effect levels for TEHP and DEHPA are proposed, but the lower serum pituitary hormone levels resulting from TEHP and DEHPA exposures and the perturbations of testicular histology observed in TEHP-treated males deserve further investigation. Improved characterization of the toxicity of flame retardants will contribute to better informed substitution choices for legacy flame retardants phased-out over health concerns.


Subject(s)
Flame Retardants/toxicity , Organophosphates/toxicity , Plasticizers/toxicity , Solvents/toxicity , Administration, Oral , Animals , Biomarkers/blood , Biomarkers/urine , Female , Flame Retardants/administration & dosage , Liver/drug effects , Liver/enzymology , Liver/pathology , Male , No-Observed-Adverse-Effect Level , Organ Size , Organophosphates/administration & dosage , Plasticizers/administration & dosage , Rats, Inbred F344 , Risk Assessment , Solvents/administration & dosage , Testis/drug effects , Testis/metabolism , Testis/pathology , Time Factors , Toxicity Tests
2.
J Endocrinol ; 239(2): 181-195, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30139929

ABSTRACT

It is unknown whether there is a gene signature in pancreas which is associated with type 1 diabetes (T1D). We performed partial pancreatectomies on 30-day preinsulitic, diabetes-prone BioBreeding (BBdp) rats to prospectively identify factors involved in early prediabetes. Microarrays of the biopsies revealed downregulation of endoplasmic reticulum (ER) stress, metabolism and apoptosis. Based on these results, additional investigations compared gene expression in control (BBc) and BBdp rats age ~8, 30 and 60 days using RT-qPCR. Neonates had increased ER stress gene expression in pancreas. This was associated with decreased insulin, cleaved caspase-3 and Ins1 whereas Gcg and Pcsk2 were increased. The increase in ER stress was not sustained at 30 days and decreased by 60 days. In parallel, the liver gene profile showed a similar signature in neonates but with an early decrease of the unfolded protein response (UPR) at 30 days. This suggested that changes in the liver precede those in the pancreas. Tnf and Il1b expression was increased in BBdp pancreas in association with increased caspase-1, cleaved caspase-3 and decreased proinsulin area. Glucagon area was increased in both 30-day and 60-day BBdp rats. Increased colocalization of BIP and proinsulin was observed at 60 days in the pancreas, suggesting insulin-related ER dysfunction. We propose that dysregulated metabolism leads to ER stress in neonatal rats long before insulitis, creating a microenvironment in both pancreas and liver that promotes autoimmunity.


Subject(s)
Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 1/etiology , Endoplasmic Reticulum Stress , Pancreas/metabolism , Animals , Animals, Newborn , Apoptosis , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Gene Expression Profiling , Liver/metabolism , Oligonucleotide Array Sequence Analysis , Rats
3.
Diabetes ; 64(12): 4135-47, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26370175

ABSTRACT

Cathelicidin antimicrobial peptide (CAMP) is a naturally occurring secreted peptide that is expressed in several organs with pleiotropic roles in immunomodulation, wound healing, and cell growth. We previously demonstrated that gut Camp expression is upregulated when type 1 diabetes-prone rats are protected from diabetes development. Unexpectedly, we have also identified novel CAMP expression in the pancreatic ß-cells of rats, mice, and humans. CAMP was present even in sterile rat embryo islets, germ-free adult rat islets, and neogenic tubular complexes. Camp gene expression was downregulated in young BBdp rat islets before the onset of insulitis compared with control BBc rats. CAMP treatment of dispersed islets resulted in a significant increase in intracellular calcium mobilization, an effect that was both delayed and blunted in the absence of extracellular calcium. Additionally, CAMP treatment promoted insulin and glucagon secretion from isolated rat islets. Thus, CAMP is a promoter of islet paracrine signaling that enhances islet function and glucoregulation. Finally, daily treatment with the CAMP/LL-37 peptide in vivo in BBdp rats resulted in enhanced ß-cell neogenesis and upregulation of potentially beneficial gut microbes. In particular, CAMP/LL-37 treatment shifted the abundance of specific bacterial populations, mitigating the gut dysbiosis observed in the BBdp rat. Taken together, these findings indicate a novel functional role for CAMP/LL-37 in islet biology and modification of gut microbiota.


Subject(s)
Cathelicidins/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Dysbiosis/drug therapy , Gastrointestinal Agents/therapeutic use , Hypoglycemic Agents/therapeutic use , Islets of Langerhans/drug effects , Peptide Fragments/therapeutic use , Regeneration/drug effects , Aged , Animals , Antimicrobial Cationic Peptides/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Calcium Signaling/drug effects , Cathelicidins/pharmacology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/microbiology , Diabetes Mellitus, Type 1/pathology , Dysbiosis/metabolism , Dysbiosis/microbiology , Dysbiosis/pathology , Gastrointestinal Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Gene Expression Profiling , Germ-Free Life , Glucagon/agonists , Glucagon/metabolism , Humans , Hypoglycemic Agents/pharmacology , Insulin/agonists , Insulin/metabolism , Insulin Secretion , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans/physiology , Jejunum/drug effects , Jejunum/metabolism , Jejunum/microbiology , Male , Mice, Inbred NOD , Peptide Fragments/pharmacology , Rats, Inbred Strains , Tissue Culture Techniques
4.
Endocrinology ; 156(11): 3937-49, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26252059

ABSTRACT

Immunoregulatory and regenerative processes are activated in the pancreas during the development of type 1 diabetes (T1D) but are insufficient to prevent the disease. We hypothesized that the induction of cytoprotective heme oxygenase-1 (HO-1) by cobalt protophoryrin (CoPP) would prevent T1D by promoting anti-inflammatory and pro-repair processes. Diabetes-prone BioBreeding rats received ip CoPP or saline twice per week for 3 weeks, starting at 30 days and were monitored for T1D. Immunohistochemistry, confocal microscopy, quantitative RT-PCR, and microarrays were used to evaluate postinjection pancreatic changes at 51 days, when islet inflammation is first visible. T1D was prevented in CoPP-treated rats (29% vs 73%). Pancreatic Hmox1 was up-regulated along with islet-associated CD68(+)HO-1(+) cells, which were also observed in a striking peri-lobular interstitial infiltrate. Most interstitial cells expressed the mesenchymal marker vimentin and the hematopoietic marker CD34. Spindle-shaped, CD34(+)vimentin(+) cells coexpressed collagen V, characteristic of fibrocytes. M2 macrophage factors Krüppel-like factor 4, CD163, and CD206 were expressed by interstitial cells, consistent with pancreatic upregulation of several M2-associated genes. CoPP upregulated islet-regenerating REG genes and increased neogenic REG3ß(+) and insulin(+) clusters. Thus, short-term induction of HO-1 promoted a protective M2-like milieu in the pancreas and recruited mesenchymal cells, M2 macrophages, and fibrocytes that imparted immunoregulatory and pro-repair effects, preventing T1D.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Heme Oxygenase-1/biosynthesis , Macrophages/metabolism , Mesenchymal Stem Cells/metabolism , Pancreas/metabolism , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Collagen Type V/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/prevention & control , Enzyme Induction/drug effects , Female , Gene Expression/drug effects , Heme Oxygenase-1/genetics , Insulin/genetics , Insulin/metabolism , Kruppel-Like Factor 4 , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Macrophages/drug effects , Male , Mannose Receptor , Mannose-Binding Lectins/metabolism , Mesenchymal Stem Cells/drug effects , Microscopy, Confocal , Pancreas/drug effects , Pancreatitis-Associated Proteins , Protoporphyrins/pharmacology , Rats , Receptors, Cell Surface/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Vimentin/metabolism
5.
Toxicology ; 334: 81-93, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26066519

ABSTRACT

Rates of obesity and diabetes mellitus of Arctic populations are increasing due to multiple reasons including a departure from traditional lifestyles and alcohol consumption patterns. These populations are also exposed to a variety of anthropogenic contaminants through consumption of contaminated country foods. We have previously shown that a Northern contaminant mixture (NCM), containing 22 organic and inorganic contaminants found in the blood of Canadian Arctic populations, induces endothelial cell dysfunction and exacerbates development of non-alcoholic fatty liver disease in experimental models. In order to determine if these contaminants affect pancreas function and physiology and if obesity and alcohol can influence contaminant toxicity and the development of diabetes, lean and obese JCR rats were orally treated with NCM at 0 (vehicle), 1.6 or 16mg/kg BW for four weeks in the presence or absence of 10% (v/v) alcohol. NCM treatment altered islet morphology, increased iron deposit in pancreas, and reduced circulating and pancreatic insulin levels and circulating glucagon levels as a result of direct islet injury with ß and α cell loss with or without exposure to alcohol. Studies conducted with cultured mouse insulin-secreting (MIN6) ß cells further demonstrated that NCM inhibited insulin release and induced cell death through oxidative stress and mitochondrial dysfunction. 2,3,4,6-Tetrabromophenol, a minor component of the NCM, alone also inhibited insulin release from MIN6 cells after 10min of exposure. These results suggest that Northern contaminants may contribute to pancreatic dysfunction, and possibly development of diabetes, in some of the highly exposed Arctic populations. The implications and relevance of these findings to Northern populations remains to be confirmed through epidemiological studies.


Subject(s)
Complex Mixtures/toxicity , Diabetes Mellitus/chemically induced , Environmental Pollutants/toxicity , Insulin/metabolism , Insulinoma/metabolism , Islets of Langerhans/drug effects , Obesity/complications , Pancreatic Neoplasms/metabolism , Thinness/complications , Alcohol Drinking/adverse effects , Animals , Cell Death/drug effects , Cell Line, Tumor , Diabetes Mellitus/blood , Diabetes Mellitus/pathology , Diabetes Mellitus/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Glucagon/blood , Insulin/blood , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/physiopathology , Male , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Obesity/blood , Obesity/pathology , Obesity/physiopathology , Oxidative Stress/drug effects , Rats , Risk Assessment , Thinness/blood , Thinness/pathology , Thinness/physiopathology
6.
Immunology ; 145(3): 417-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25711680

ABSTRACT

The gut immune system and its modification by diet have been implicated in the pathogenesis of type 1 diabetes (T1D). Therefore, we investigated gut immune status in non-diabetes-prone LEW.1AR1 and diabetes-prone LEW.1AR1-iddm rats and evaluated the effect of a low antigen, hydrolysed casein (HC)-based diet on gut immunity and T1D. Rats were weaned onto a cereal-based or HC-based diet and monitored for T1D. Strain and dietary effects on immune homeostasis were assessed in non-diabetic rats (50-60 days old) and rats with recent-onset diabetes using flow cytometry and immunohistochemistry. Immune gene expression was analysed in mesenteric lymph nodes (MLN) and jejunum using quantitative RT-PCR and PCR arrays. T1D was prevented in LEW.1AR1-iddm rats by feeding an HC diet. Diabetic LEW.1AR1-iddm rats had fewer lymphoid tissue T cells compared with LEW.1AR1 rats. The percentage of CD4(+)  Foxp3(+) regulatory T (Treg) cells was decreased in pancreatic lymph nodes (PLN) of diabetic rats. The jejunum of 50-day LEW.1AR1-iddm rats contained fewer CD3(+) T cells, CD163(+) M2 macrophages and Foxp3(+) Treg cells. Ifng expression was increased in MLN and Foxp3 expression was decreased in the jejunum of LEW.1AR1-iddm rats; Ifng/Il4 was decreased in jejunum of LEW.1AR1-iddm rats fed HC. PCR arrays revealed decreased expression of M2-associated macrophage factors in 50-day LEW.1AR1-iddm rats. Wheat peptides stimulated T-cell proliferation and activation in MLN and PLN cells from diabetic LEW.1AR1-iddm rats. LEW.1AR1-iddm rats displayed gut immune cell deficits and decreased immunoregulatory capacity, which were partially corrected in animals fed a low antigen, protective HC diet consistent with other models of T1D.


Subject(s)
Diabetes Mellitus, Type 1/diet therapy , Diabetes Mellitus, Type 1/immunology , Diet, Diabetic , Digestive System/immunology , Animals , Caseins/immunology , Caseins/therapeutic use , Cytokines/genetics , Cytokines/immunology , Cytokines/metabolism , Diabetes Mellitus, Type 1/genetics , Diet , Digestive System/metabolism , Disease Models, Animal , Edible Grain/immunology , Flow Cytometry , Gene Expression/immunology , Homeostasis/genetics , Homeostasis/immunology , Humans , Immunity/genetics , Immunity/immunology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Jejunum/immunology , Jejunum/metabolism , Macrophages/immunology , Macrophages/metabolism , Rats, Inbred BB , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Weaning
7.
J Diabetes ; 7(1): 74-84, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24548670

ABSTRACT

BACKGROUND: Proprotein convertase subtilisin/kexin-type 9 (PCSK9) downregulates clearance of plasma cholesterol by liver. Its inactivation increases this clearance, reducing cardiovascular risk. However, a lack of PCSK9 could also lead to cholesterol accumulation in pancreatic islet beta cells, impairing insulin secretion. We reported earlier that 4-month-old male PCSK9-deficient (KO) C57BL/6 mice were hyperglycemic and insulin-insufficient relative to their wild-type (WT) counterparts. Here, we examined how gender and diet affect lipid and glucose homeostasis in these mice at 8 months of age. METHODS: After being fed a normal diet or a Western diet for over 6 months, KO mice were compared with same-gender WT mice for fasting plasma levels of total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), glucose, and insulin; for glucose disposal and glucose-stimulated insulin secretion (GSIS); and for pancreatic islet morphology. RESULTS: A. Females: On normal diet, KO mice showed lower plasma TC, HDL-C, and LDL-C, higher plasma glucose, and normal glucose disposal despite impaired GSIS. On Western diet, they showed comparable plasma TC and HDL-C, but lower LDL-C, higher plasma glucose, and normal glucose disposal despite impaired GSIS. B. Males: On normal and Western diets, KO mice showed lower plasma TC, HDL-C, and LDL-C, similarly elevated plasma glucose, glucose intolerance, and impaired GSIS. C. Both: KO mice on either diet showed pancreatic islet dysmorphism, with larger, possibly immature secretory granules. CONCLUSIONS: Lower LDL-C and impaired GSIS are two major phenotypes in aged PCSK9-deficient C57BL/6 mice. These phenotypes are modulated by gender and diet.


Subject(s)
Blood Glucose/metabolism , Diet, Western , Homeostasis/physiology , Proprotein Convertases/physiology , Serine Endopeptidases/physiology , Animals , Body Weight , Cholesterol/blood , Female , Immunoblotting , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lipids/blood , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Proprotein Convertase 9 , Sex Factors
8.
Diabetologia ; 57(3): 512-21, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24310561

ABSTRACT

AIMS/HYPOTHESIS: Research on the pathogenesis of type 1 diabetes relies heavily on good animal models. The aim of this work was to study the translational value of animal models of type 1 diabetes to the human situation. METHODS: We compared the four major animal models of spontaneous type 1 diabetes, namely the NOD mouse, BioBreeding (BB) rat, Komeda rat and LEW.1AR1-iddm rat, by examining the immunohistochemistry and in situ RT-PCR of immune cell infiltrate and cytokine pattern in pancreatic islets, and by comparing findings with human data. RESULTS: After type 1 diabetes manifestation CD8(+) T cells, CD68(+) macrophages and CD4(+) T cells were observed as the main immune cell types with declining frequency, in infiltrated islets of all diabetic pancreases. IL-1ß and TNF-α were the main proinflammatory cytokines in the immune cell infiltrate in NOD mice, BB rats and LEW.1AR1-iddm rats, as well as in humans. The Komeda rat was the exception, with IFN-γ and TNF-α being the main cytokines. In addition, IL-17 and IL-6 and the anti-inflammatory cytokines IL-4, IL-10 and IL-13 were found in some infiltrating immune cells. Apoptotic as well as proliferating beta cells were observed in infiltrated islets. In healthy pancreases no proinflammatory cytokine expression was observed. CONCLUSIONS/INTERPRETATION: With the exception of the Komeda rat, the animal models mirror very well the situation in humans with type 1 diabetes. Thus animal models of type 1 diabetes can provide meaningful information on the disease processes in the pancreas of patients with type 1 diabetes.


Subject(s)
Apoptosis , B-Lymphocytes/pathology , Cytokines/metabolism , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Insulin-Secreting Cells/pathology , Animals , Apoptosis/immunology , B-Lymphocytes/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/immunology , Gene Expression Regulation , Humans , Immunohistochemistry , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/metabolism , Interferon-gamma/metabolism , Mice , Mice, Inbred NOD , Rats , Rats, Inbred BB , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
9.
Diabetes ; 62(6): 2036-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23349499

ABSTRACT

We are exposed to millions of microbial and dietary antigens via the gastrointestinal tract, which likely play a key role in type 1 diabetes (T1D). We differentiated the effects of these two major environmental factors on gut immunity and T1D. Diabetes-prone BioBreeding (BBdp) rats were housed in specific pathogen-free (SPF) or germ-free (GF) conditions and weaned onto diabetes-promoting cereal diets or a protective low-antigen hydrolyzed casein (HC) diet, and T1D incidence was monitored. Fecal microbiota 16S rRNA genes, immune cell distribution, and gene expression in the jejunum were analyzed. T1D was highest in cereal-SPF (65%) and cereal-GF rats (53%) but inhibited and delayed in HC-fed counterparts. Nearly all HC-GF rats remained diabetes-free, whereas HC-fed SPF rats were less protected (7 vs. 29%). Bacterial communities differed in SPF rats fed cereal compared with HC. Cereal-SPF rats displayed increased gut CD3(+) and CD8α(+) lymphocytes, ratio of Ifng to Il4 mRNA, and Lck expression, indicating T-cell activation. The ratio of CD3(+) T cells expressing the Treg marker Foxp3(+) was highest in HC-GF and lowest in cereal-SPF rats. Resident CD163(+) M2 macrophages were increased in HC-protected rats. The cathelicidin antimicrobial peptide (Camp) gene was upregulated in the jejunum of HC diet-protected rats, and CAMP(+) cells colocalized with CD163. A cereal diet was a stronger promoter of T1D than gut microbes in association with impaired gut immune homeostasis.


Subject(s)
Cathelicidins/metabolism , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/microbiology , Edible Grain/adverse effects , Adolescent , Animals , Antimicrobial Cationic Peptides , Child , Female , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Humans , Immunohistochemistry , Jejunum/immunology , Jejunum/microbiology , Male , Rats , Rats, Inbred BB , Real-Time Polymerase Chain Reaction
10.
Ann Neurol ; 72(2): 256-68, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22926856

ABSTRACT

OBJECTIVE: Spinal muscular atrophy (SMA) is the number 1 genetic killer of young children. It is caused by mutation or deletion of the survival motor neuron 1 (SMN1) gene. Although SMA is primarily a motor neuron disease, metabolism abnormalities such as metabolic acidosis, abnormal fatty acid metabolism, hyperlipidemia, and hyperglycemia have been reported in SMA patients. We thus initiated an in-depth analysis of glucose metabolism in SMA. METHODS: Glucose metabolism and pancreas development were investigated in the Smn(2B/-) intermediate SMA mouse model and type I SMA patients. RESULTS: Here, we demonstrate in an SMA mouse model a dramatic cell fate imbalance within pancreatic islets, with a predominance of glucagon-producing α cells at the expense of insulin-producing ß cells. These SMA mice display fasting hyperglycemia, hyperglucagonemia, and glucose resistance. We demonstrate similar abnormalities in pancreatic islets from deceased children with the severe infantile form of SMA in association with supportive evidence of glucose intolerance in at least a subset of such children. INTERPRETATION: Our results indicate that defects in glucose metabolism may play an important contributory role in SMA pathogenesis.


Subject(s)
Blood Glucose/metabolism , Glucose Metabolism Disorders/etiology , Pancreatic Diseases/etiology , Spinal Muscular Atrophies of Childhood/complications , Age Factors , Animals , Animals, Newborn , Apoptosis/genetics , Blood Glucose/genetics , Cell Proliferation , Disease Models, Animal , Glucagon/blood , Humans , In Situ Nick-End Labeling , Insulin/blood , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Pancreatic Diseases/genetics , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 1 Protein/metabolism
11.
Pancreas ; 40(2): 229-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21847793

ABSTRACT

OBJECTIVES: We previously demonstrated that the expression of cellular prion protein (PrPC) in islet [beta]-cells is suppressed in hyperglycemic rats suggesting a major role for PrPC in blood glucose regulation. To further characterize the function of PrPC in glucose homeostasis, we studied glucoregulation in PrPC knockout (PrPC KO) mice. METHODS: Glucose tolerance, insulin secretion, and insulin sensitivity were analyzed to assess glucoregulation in Zrch I PrPC KO and the C57BL/6 (control) mice. Immunohistochemistry and morphometry were used to measure [beta]-cell mass. RESULTS: Male PrPC KO mice had significantly increased blood glucose concentration 60, 120, and 180 minutes after intraperitoneal injection of glucose compared with C57BL/6 mice. Female PrPC KO mice showed a less pronounced phenotype of glucose intolerance. Evaluation of [beta]-cell mass, insulin and proinsulin deficiency, and insulin resistance in male mice revealed essentially no difference between PrPC KO and control mice. The only exception was an increase in serum insulin concentration in male PrPC KO mice 5 minutes after glucose injection. CONCLUSIONS: This report is the first to show that PrPC in [beta]-cells is involved in glucoregulation. A further understanding of the role of PrPC in regulating [beta]-cell function will provide valuable insight into the mechanisms of blood glucose regulation.


Subject(s)
Blood Glucose/metabolism , Glucose Intolerance/metabolism , Glucose Intolerance/pathology , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin/metabolism , PrPC Proteins/metabolism , Animals , Female , Homeostasis , Hyperglycemia/metabolism , Hyperglycemia/pathology , Immunohistochemistry , Insulin/blood , Insulin Resistance , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , PrPC Proteins/genetics , Proinsulin/deficiency , Time Factors
12.
Can J Gastroenterol ; 25(6): 327-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21766094

ABSTRACT

Antibodies against the wheat storage globulin Glo-3A from a patient with both type 1 diabetes (T1D) and celiac disease were enriched to identify potential molecular mimicry between wheat antigens and T1D target tissues. Recombinant Glo-3A was used to enrich anti-Glo-3A immunoglobulin G antibodies from plasma by batch affinity chromatography. Rat jejunum and pancreas, as well as human duodenum and monocytes were probed, and binding was evaluated by immunohistochemistry and confocal microscopy. Glo-3A-enriched antibodies bound to a specific subset of cells in the lamina propria of rat jejunum that co-localized mostly with a marker of resident, alternatively activated CD163-positive (CD163⁺) macrophages. Blood monocytes and macrophage-like cells in human duodenum were also labelled with the enriched antibodies. Blocking studies revealed that binding to CD163⁺ macrophages was not due to cross-reactivity with anti-Glo-3A antibodies, but rather to non-Glo-3A antibodies co-purified during antibody enrichment. The novel finding of putative autoantibodies against tolerogenic intestinal CD163⁺ macrophages suggests that regulatory macrophages were targeted in this patient with celiac disease and T1D.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Celiac Disease/immunology , Diabetes Mellitus, Type 1/immunology , Macrophages/immunology , Receptors, Cell Surface/metabolism , Animals , Antibody Formation , Autoimmunity , Biomarkers , Celiac Disease/metabolism , Diabetes Mellitus, Type 1/metabolism , Extracellular Matrix Proteins/immunology , Humans , Immunoglobulin G/metabolism , Immunohistochemistry , Monocytes/immunology , Rats , Rats, Inbred BB , Seed Storage Proteins/immunology , Wheat Hypersensitivity
13.
Eur J Cell Biol ; 90(5): 414-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21277044

ABSTRACT

Several physiological processes have been purported for cellular prion protein (PrP(C)). However, the physiological function of PrP(C) is still unclear and the cellular localization of PrP(C) remains a subject of debate. PrP(C) is expressed in a wide range of tissues including islets of Langerhans. We previously demonstrated that the function of PrP(C) is associated with blood glucose regulation. Little is known of the function of PrP(C) in islet cells and specifically in ß-cells. To get first insight into the putative role of PrP(C) in ß-cells, we used far-Western immunoblotting and MS to identify candidate PrP(C)-interacting proteins. We also used Western blot, immunofluorescence (IF) and protein overlay IF to characterize the sub-cellular localization of PrP(C). Here we demonstrate in vivo that PrP(C) is abundant in the nuclear lamina of endocrine and neuronal cells and interacts with histone H1(0), histone H3 and lamin B1. The interaction of PrP(C) with histone H3 suggests that it is involved in transcriptional regulation in the nucleus. This study reveals new avenues for the elucidation of the physiological function of PrP(C) in endocrine and neuronal cells as well as the molecular mechanisms leading to prion diseases.


Subject(s)
Cell Nucleus/metabolism , Chromatin/metabolism , Endocrine System/cytology , Neurons/cytology , PrPC Proteins/metabolism , Animals , Cell Line , Chromatin/chemistry , Endocrine System/metabolism , Histones/metabolism , Lamin Type B/metabolism , Mice , Mice, Knockout , Neurons/metabolism , PrPC Proteins/genetics , Rats
14.
J Cell Physiol ; 224(2): 501-8, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20432450

ABSTRACT

Type 1 diabetes is inhibited in diabetes-prone BioBreeding (BBdp) rats fed a low-antigen hydrolyzed casein (HC) diet. In cereal-fed BBdp rats, islet expansion is defective accompanied by a futile upregulation of islet neogenesis without increased islet mass, due to a subtle blockage in islet cell cycle. We hypothesized that islet growth is enhanced before insulitis in HC-fed young BBdp rats and that islet neogenesis could be stimulated by a trophic factor, islet neogenesis-associated protein (INGAP). beta-Cell homeostasis was analyzed using immunohistochemistry, morphometry, laser capture microdissection and RT-PCR in BBdp rats fed HC or cereal diets. beta-cell proliferation in small and medium islets, and the number and area fraction of medium and large islets were increased in HC-fed animals. In situ islet cell cycle analysis revealed an increased proportion of proliferating S + G2 cells in medium and large islets of 25-45 day HC-fed rats. Expression of the cell cycle inhibitor, p16(INK4a) correlated with islet size and the percentage of p16(INK4a+) beta-cells increased in HC-fed BBdp rats, likely reflecting an increase in large islet area fraction. In HC-fed rats, extra-islet insulin(+) clusters (EIC), insulin(+) duct cells, large islet area fraction, and beta-cell mass were increased. Neurogenin-3 and Pdx-1, markers of beta-cell progenitors, were increased in EIC of weanling HC-fed rats. Daily injection of INGAP (30-45 days) increased the number of small islets, total islets, and insulin(+) cells in small ducts. Thus, in BBdp rats fed a protective HC diet, beta-cell expansion is enhanced through increased beta-cell proliferation and stimulation of islet neogenesis.


Subject(s)
Aging/pathology , Cytoprotection , Diabetes Mellitus, Experimental/pathology , Diet , Feeding Behavior , Insulin-Secreting Cells/pathology , Aging/drug effects , Animals , Apoptosis/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , Caseins/administration & dosage , Caseins/pharmacology , Cell Aggregation/drug effects , Cell Count , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Size/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cytokines/pharmacology , Cytoprotection/drug effects , Diabetes Mellitus, Experimental/metabolism , Disease Susceptibility/pathology , Homeodomain Proteins/metabolism , Homeostasis/drug effects , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Nerve Tissue Proteins/metabolism , Organ Size/drug effects , Pancreatitis-Associated Proteins , Peptide Fragments/pharmacology , Rats , Trans-Activators/metabolism
15.
J Proteome Res ; 9(3): 1203-8, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20113007

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disorder that results from the destruction of insulin-producing beta-cells in the islets of Langerhans. To date, autoimmune T-cell response and antibody reactivity to more than 20 autoantigens have been linked to this disease. Some studies have described the intermediate filament protein peripherin (PRPH) as an autoantigen associated with T1D in non-obese diabetic (NOD) mice. We evaluated immune reactivity of mouse and rabbit sera and human plasma to a 58 kDa protein expressed in RIN-m5F rat insulinoma cells. The protein was isolated using 2-DE and identified by mass spectrometry as PRPH. Antibodies from healthy humans and T1D patients, CD-1 mice, C57BL/6 mice, NOR (non-obese diabetes resistant) mice, and NOD mice reacted with PRPH on Western blots. However, antibody response to PRPH was stronger in NOD than non-autoimmune prone C57BL/6 mice. We conclude that immune reactivity to PRPH is not exclusively associated with NOD mice or human patients with T1D. Furthermore, the frequent occurrence of PRPH-reactive antibodies in mouse and human blood suggests that binding may be non-specific or could reflect the presence of natural autoantibodies against PRPH. These findings point to the need for a re-evaluation of PRPH as a T1D autoantigen in NOD mice and raise the question of the physiological relevance of such widespread immune reactivity against this peripheral nervous system protein.


Subject(s)
Autoantibodies/blood , Intermediate Filament Proteins/immunology , Membrane Glycoproteins/immunology , Nerve Tissue Proteins/immunology , Adult , Animals , Autoantibodies/metabolism , Cell Line, Tumor , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Electrophoresis, Gel, Two-Dimensional , Female , Humans , Insulinoma/immunology , Insulinoma/metabolism , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/metabolism , Male , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neuroblastoma/immunology , Neuroblastoma/metabolism , Peripherins , Rabbits , Rats , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Subcellular Fractions/chemistry , Subcellular Fractions/metabolism
16.
FEBS Lett ; 584(4): 701-6, 2010 Feb 19.
Article in English | MEDLINE | ID: mdl-20026049

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9), a liver-secreted plasma enzyme, restricts hepatic uptake of low-density lipoprotein (LDL) cholesterol by promoting the degradation of LDL receptors (LDLR). PCSK9 and LDLR are also expressed in insulin-producing pancreatic islet beta cells, possibly affecting the function of these cells. Here we show that, compared to control mice, PCSK9-null male mice over 4 months of age carried more LDLR and less insulin in their pancreas; they were hypoinsulinemic, hyperglycemic and glucose-intolerant; their islets exhibited signs of malformation, apoptosis and inflammation. Collectively, these observations suggest that PCSK9 may be necessary for the normal function of pancreatic islets.


Subject(s)
Glucose Intolerance/genetics , Islets of Langerhans/metabolism , Receptors, LDL/genetics , Serine Endopeptidases/genetics , Animals , Apoptosis , Blood Glucose/metabolism , Blotting, Western , Female , Gene Expression , Glucose Intolerance/blood , Glucose Intolerance/enzymology , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Islets of Langerhans/abnormalities , Islets of Langerhans/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Proprotein Convertase 9 , Proprotein Convertases , Receptors, LDL/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Serine Endopeptidases/deficiency , Tissue Culture Techniques
17.
Cell Metab ; 10(4): 285-95, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19808021

ABSTRACT

The Lkb1 tumor suppressor exerts its biological effects through phosphorylation and consequent activation of the AMP kinase (AMPK) family. Extensive genetic and biochemical evidence supports a role for Lkb1 in cell cycle arrest, establishment of cell polarity, and cellular energy metabolism. However, the role of Lkb1 and the AMPK family in beta cell function in vivo has not been established. We generated conditional knockout mice with a deletion of the Lkb1 gene in the beta cell compartment of pancreatic islets; these mice display improved glucose tolerance and protection against diet-induced hyperglycemia. Lkb1(-/-) beta cells are hypertrophic because of elevated mTOR activity; they also proliferate more and secrete more insulin in response to glucose. These data indicate that inhibiting Lkb1 activity in beta cells may facilitate beta cell expansion and glucose tolerance in vivo.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Cells, Cultured , Dietary Fats , Estrogen Antagonists/pharmacology , Glucose Tolerance Test , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Tamoxifen/pharmacology , Transgenes
18.
Lab Invest ; 87(12): 1240-51, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17906659

ABSTRACT

We reported previously that young BioBreeding diabetes-prone (BBdp) rats display increased neogenic extra-islet insulin+ clusters (EICs, <4 insulin+ cells) without an increase in beta-cell mass. Therefore, we investigated the possibility that abnormal islet expansion occurs in BBdp rats before the appearance of islet inflammation. Islet expansion was analyzed in pancreata from 14 to 45 day BBdp and control (BioBreeding control, BBc) rats using immunohistochemistry, morphometry, laser capture microdissection and reverse transcriptase-PCR. mRNA expression for Neurogenin-3, a developmental marker of endocrine progenitors, was three-fold greater in EIC of weanling BBdp and BBc rats compared with islet cells. With increasing age (14-30 days), Neurogenin-3 expression decreased in EIC and increased in islets. In BBdp rats, EIC number and beta-cell proliferation within EIC was greater compared with BBc animals; apoptosis did not differ. The area of small and medium islets in BBdp rats was greater than BBc rats between 14 and 30 days, but this did not result in increased total islet area or beta-cell mass. In addition, the number and area of very large islets was low at 45 days. The frequency of proliferating beta-cells decreased with increasing islet size in BBdp but was constant in BBc rats. Cell cycle analysis of islets revealed more G1 cells and fewer G2 cells in BBdp rats. The ratio of cyclinD2/Cdkn1a, genes that respectively promote or inhibit cell cycle progression, was decreased in BBdp islets. These results suggest that despite increased islet neogenesis, the capacity for islet expansion in diabetes-prone rats is compromised possibly due to decreased proliferative capacity with increasing islet size associated with a partial block at the G1/S cell cycle boundary in islet cells.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/growth & development , Islets of Langerhans/pathology , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle , Cell Proliferation , Cell Size , Cyclin D2 , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclins/metabolism , Diabetes Mellitus, Type 1/metabolism , Homeodomain Proteins/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Nerve Tissue Proteins/metabolism , Rats , Trans-Activators/metabolism
19.
Lab Invest ; 87(2): 139-49, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17146448

ABSTRACT

Cellular prion protein (PrP(C)), an N-linked glycoprotein, is expressed in a variety of tissues, but its functions remain unclear. PrP(C) is abundantly expressed in the endocrine pancreas, which regulates blood glucose homeostasis. Therefore, we investigated whether the expression of PrP(C) was altered in islets of Langerhans in a model of spontaneous type 1 diabetes, the diabetes-prone BioBreeding (BBdp) rat and a model of beta-cell adaptation to hyperglycemia, the chronic glucose-infused Sprague Dawley rat. Pancreatic sections from animals aged 7-100 days were stained immunohistochemically and evaluated using light, fluorescence and confocal microscopy. PrP(C) was ubiquitously expressed in all four major endocrine cell types within islets. Surprisingly, cytosolic inclusions containing PrP(C) were identified exclusively in a subpopulation of insulin-producing beta-cells. The inclusions exhibited different molecular characteristics from the PrP aggregates previously described in vitro in neurons. The frequency of beta-cells with PrP(C) inclusions increased with age and was threefold greater in diabetes-prone rats than in controls at 100 days. Cytosolic PrP(C) expression in beta-cells was suppressed whereas the number and size of PrP(C) inclusions markedly increased in response to hyperglycemia during the first 2 days of continuous glucose infusion in Sprague Dawley rats. In summary, this is the first report describing in vivo cytosolic PrP(C) aggregation. These unique PrP(C) inclusions were beta-cell specific, more frequent in diabetes-prone animals, and responded to hyperglycemia in glucose-infused Sprague Dawley rats. These data suggest a potential dysfunction in beta-cells of diabetes-prone rats, and point to new avenues for the study of diabetes pathogenesis.


Subject(s)
Cytosol/metabolism , Diabetes Mellitus, Type 1/metabolism , Hyperglycemia/metabolism , Insulin-Secreting Cells/metabolism , PrPC Proteins/metabolism , Age Factors , Analysis of Variance , Animals , Glucose/metabolism , Immunohistochemistry , Microscopy, Fluorescence , Rats , Rats, Inbred WF , Rats, Sprague-Dawley
20.
Lab Invest ; 85(5): 675-88, 2005 May.
Article in English | MEDLINE | ID: mdl-15765120

ABSTRACT

Tubular complexes (TC) in the pancreas contain duct-like structures with low cuboidal or flattened cells surrounding a large lumen and are thought to be a response to pancreatic injury. TC have been studied in animal models of chemical or surgically induced pancreatic damage but their occurrence has not been reported in rodent models of spontaneous autoimmune type I diabetes. We hypothesized that TC would be increased during the active phase of islet destruction in autoimmune diabetes and could contain islet progenitor cells. We analyzed TC in pancreas of Wistar Furth (WF), control (BBc) and diabetes-prone BioBreeding (BBdp) rats using immunohistochemistry and morphometry. TC were observed in all rat strains during active pancreas remodeling ( approximately 13 days). They increased between 60 and 93 days only in BBdp rats coincident with the increase in diabetes cases. Most TC were infiltrated with CD3(+) T-cells. Duct-like cells in the TC had low expression of the exocrine marker amylase, increased expression of epithelial cell markers, keratin and vimentin, and remarkably high cell proliferation and cell death. TC islets contained cells stained positive for insulin, glucagon, somatostatin, pancreatic polypeptide, as well as PDX-1, chromogranin, and hepatocyte-derived growth factor receptor, c-met. Transitional cells that were keratin(+)/insulin(+) and keratin(+)/amylase(+) cells were present in TC. The stem cell marker, nestin was upregulated in the TC region. Duct-like cells in TC of BBdp rats expressed markers of committed endocrine precursors: PDX-1, neurogenin 3 and protein gene product 9.5. This study demonstrates that TC are upregulated during beta-cell destruction and contain potential endocrine progenitors.


Subject(s)
Diabetes Mellitus, Type 1/pathology , Islets of Langerhans/pathology , Pancreas, Exocrine/pathology , Stem Cells/pathology , Amylases/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , In Situ Nick-End Labeling , Islets of Langerhans/metabolism , Keratins/metabolism , Pancreas, Exocrine/metabolism , Rats , Rats, Inbred BB , Rats, Inbred WF , Regeneration , Stem Cells/metabolism , Vimentin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL