Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
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
2.
Transl Res ; 179: 183-198, 2017 01.
Article in English | MEDLINE | ID: mdl-27677687

ABSTRACT

The rise in new cases of type 1 diabetes (T1D) in genetically susceptible individuals over the past half century has been attributed to numerous environmental "triggers" or promoters such as enteroviruses, diet, and most recently, gut bacteria. No single cause has been identified in humans, likely because there are several pathways by which one can develop T1D. There is renewed attention to the role of the gut and its immune system in T1D pathogenesis based largely on recent animal studies demonstrating that altering the gut microbiota affects diabetes incidence. Although T1D patients display dysbiosis in the gut microbiome, it is unclear whether this is cause or effect. The heart of this question involves several moving parts including numerous risk genes, diet, viruses, gut microbiota, timing, and loss of immune tolerance to ß-cells. Most clinical trials have addressed only one aspect of this puzzle using some form of immune suppression, without much success. The key location where our genes meet and deal with the environment is the gastrointestinal tract. The influence of all of its major contents, including microbes, diet, and immune system, must be understood as part of the integrative biology of T1D before we can develop durable means of preventing, treating, or curing this disease. In the present review, we expand our previous gut-centric model based on recent developments in the field.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Gastrointestinal Tract/metabolism , Gene-Environment Interaction , Immunity/genetics , Pancreas/metabolism , Pancreas/pathology , Animals , Humans , Models, Biological
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.
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
6.
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
7.
PLoS One ; 9(8): e104343, 2014.
Article in English | MEDLINE | ID: mdl-25090610

ABSTRACT

Obesity has become one of the largest public health challenges worldwide. Recently, certain bacterial and viral pathogens have been implicated in the pathogenesis of obesity. In the present study, we retrospectively analyzed clinical data, plasma samples and post-mortem tissue specimens derived from a risk assessment study in bovine spongiform encephalopathy (BSE)-infected female cynomolgus monkeys (Macaca fascicularis). The original study design aimed to determine minimal infectious doses after oral or intracerebral (i.c.) infection of macaques to assess the risk for humans. High-dose exposures resulted in 100% attack rates and a median incubation time of 4.7 years as described previously. Retrospective analyses of clinical data from high-dosed macaques revealed that foodborne BSE transmission caused rapid weight gain within 1.5 years post infection (ß = 0.915; P<0.0001) which was not seen in age- and sex-matched control animals or i.c. infected animals. The rapid-onset obesity was not associated with impaired pancreatic islet function or glucose metabolism. In the early preclinical phase of oral transmission associated with body weight gain, prion accumulation was confined to the gastrointestinal tract. Intriguingly, immunohistochemical findings suggest that foodborne BSE transmission has a pathophysiological impact on gut endocrine cells which may explain rapid weight gain. To our knowledge, this is the first experimental model which clearly demonstrates that foodborne pathogens can induce obesity.


Subject(s)
Encephalopathy, Bovine Spongiform/complications , Foodborne Diseases/physiopathology , Obesity/etiology , Prions/pathogenicity , Animals , Cattle , Encephalopathy, Bovine Spongiform/metabolism , Encephalopathy, Bovine Spongiform/physiopathology , Encephalopathy, Bovine Spongiform/transmission , Female , Humans , Macaca fascicularis , Obesity/metabolism , Obesity/physiopathology , Primates , Prions/genetics , Weight Gain/physiology
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.
PLoS One ; 8(2): e57936, 2013.
Article in English | MEDLINE | ID: mdl-23469110

ABSTRACT

BACKGROUND: Infant dietary exposures have been linked to type 1 diabetes (T1D) development. IgG4 antibody responses to food antigens are associated with food intolerances but have not been explored prospectively in the period preceding T1D. METHODS: Using a case-cohort design, IgG4 antibodies to ß-lactoglobulin, gluten, and ovalbumin were measured in plasma collected annually from 260 DAISY participants. Of those, 77 developed islet autoimmunity (IA), defined as positive for either insulin, GAD65 or IA-2 autoantibodies on two consecutive visits, and 22 developed T1D. RESULTS: In mixed model analysis adjusting for HLA-DR status, T1D family history, age and ethnicity, higher ß-lactoglobulin IgG4 concentrations were associated with shorter breastfeeding duration (beta = -0.03, 95% Confidence Interval: -0.05, -0.006) and earlier first cow's milk exposure (beta = -0.04, 95% Confidence Interval: -0.08, 0.00). Higher gluten IgG4 was associated with older age at gluten introduction (beta = 0.06, 95% Confidence Interval: 0.00, 0.13). In proportional hazards analysis adjusting for HLA-DR status, T1D family history and ethnicity, IgG4 against individual or multiple dietary antigens throughout childhood were not associated with IA. In addition, mean antigen-specific IgG4 concentrations in infancy (age <2 years) were not associated with risk of IA nor progression to T1D. Higher ovalbumin IgG4 at first IA positive visit was marginally associated with progression to T1D (Hazard Ratio: 1.39, 95% Confidence Interval: 1.00, 1.92). CONCLUSION: We found no association between the IgG4 response to ß-lactoglobulin, gluten, and the development of either IA or T1D. The association between higher ovalbumin and progression to T1D in children with IA should be explored in other populations.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/immunology , Diet , Immunoglobulin G/immunology , Islets of Langerhans/immunology , Child, Preschool , Diabetes Mellitus, Type 1/diagnosis , Dietary Proteins/immunology , Disease Progression , Humans , Infant , Prognosis
10.
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
11.
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
12.
BMC Res Notes ; 5: 385, 2012 Jul 28.
Article in English | MEDLINE | ID: mdl-22838494

ABSTRACT

BACKGROUND: The 7S globulins are plant seed storage proteins that have been associated with the development of a number of human diseases, including peanut allergy. Immune reactivity to the wheat seed storage protein globulin-3 (Glo-3) has been associated with the development of the autoimmune disease type 1 diabetes in diabetes-prone rats and mice, as well as in a subset of human patients. FINDINGS: The present study characterized native wheat Glo-3 in salt-soluble wheat seed protein extracts. Glo-3-like peptides were observed primarily in the wheat embryo. Glo-3-like proteins varied significantly in their molecular masses and isoelectric points, as determined by two dimensional electrophoresis and immunoblotting with anti-Glo-3A antibodies. Five major polypeptide spots were identified by mass spectrometry and N-terminal sequencing as belonging to the Glo-3 family. CONCLUSIONS: These results in combination with our previous findings have allowed for the development of a hypothetical model of the post-translational events contributing to the wheat 7S globulin profile in mature wheat kernels.


Subject(s)
Globulins/metabolism , Plant Proteins/metabolism , Protein Processing, Post-Translational , Triticum/metabolism , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Endosperm/metabolism , Globulins/chemistry , Isoelectric Point , Models, Biological , Molecular Weight , Plant Proteins/chemistry , Sequence Analysis, Protein , Tandem Mass Spectrometry , Triticum/embryology
13.
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
14.
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
15.
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
16.
Biometals ; 24(4): 709-22, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21347851

ABSTRACT

Haemophilus ducreyi, a gram-negative and heme-dependent bacterium, is the causative agent of chancroid, a genital ulcer sexually transmitted infection. Heme acquisition in H. ducreyi proceeds via a receptor mediated process in which the initial event involves binding of hemoglobin and heme to their cognate outer membrane proteins, HgbA and TdhA, respectively. Following this specific interaction, the fate of the periplasmic deposited heme is unclear. Using protein expression profiling of the H. ducreyi periplasmic proteome, a periplasmic-binding protein, termed hHbp, was identified whose expression was enhanced under heme-limited conditions. The gene encoding this protein was situated in a locus displaying genetic characteristics of an ABC transporter. The purified protein bound heme in a dose-dependent and saturable manner and this binding was specifically competitively inhibited by heme. The hhbp gene functionally complemented an Escherichia coli heme uptake mutant. Expression of the heme periplasmic-binding protein was detected in a limited survey of H. ducreyi and H. influenzae clinical strains. These results indicate that the passage of heme into the cytoplasm of H. ducreyi involves a heme dedicated ABC transporter.


Subject(s)
Haemophilus ducreyi , Heme/metabolism , Periplasmic Binding Proteins/metabolism , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Gene Expression Profiling , Haemophilus ducreyi/genetics , Luminescence , Periplasmic Binding Proteins/chemistry , Periplasmic Binding Proteins/genetics , Proteome/genetics
17.
J Pediatr Gastroenterol Nutr ; 52(1): 21-5, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21119542

ABSTRACT

OBJECTIVE: Celiac disease (CD) is an autoimmune disease triggered by exposure to gluten-containing foods. IgA autoantibodies to tissue transglutaminase (TTG) are elevated in CD, but little is known about the gastrointestinal state before the appearance of TTG. Antibodies to wheat storage globulin Glo-3A have been studied in type 1 diabetes, and may be a marker of altered mucosal barrier and/or immune function. In the present study, we investigated antibody responses to Glo-3A in CD. PATIENTS AND METHODS: In the Diabetes Autoimmunity Study in the Young, children were studied prospectively from birth for the appearance of TTG and CD. Fifty cases of CD were frequency matched with 50 controls on age (of TTG seroconversion in the case), sex, ethnicity, presence of a first-degree relative with type 1 diabetes mellitus, and human leukocyte antigen -DR3 genotype. In cases and controls, IgG antibodies to Glo-3A were analyzed in a blinded manner in the sample collected at the time of seroconversion to TTG positivity (or the matched sample in controls) and in all of the previous samples since birth (mean 4.5 samples). The association between Glo-3A antibody levels and CD case status was explored using t tests at the TTG-positive visit and when Glo-3A levels were highest, and mixed modeling to describe Glo-3A over time. RESULTS: At the time of first elevated TTG (mean 4.9 years), patients with CD had higher Glo-3A antibody levels than controls (13.3 ± 17.2 vs 7.6 ± 11.7, P = 0.005). In both cases and controls, Glo-3A antibodies appear to peak at a mean age of 2.9 years, before mean age of initial TTG seroconversion. The peak Glo-3A antibody levels were higher in cases than controls (25.5 ± 21.8 vs 14.9 ± 18.3 P = 0.0007). Using mixed modeling to account for multiple visits per person, cases had higher levels of Glo-3A antibodies than controls at all ages from birth to TTG seroconversion (ß = 0.53, P = 0.002). CONCLUSIONS: Compared with controls, CD cases have higher Glo-3A antibody responses in the beginning years, before initial detection of TTG.


Subject(s)
Antibodies/blood , Celiac Disease/immunology , Plant Proteins/immunology , Celiac Disease/diagnosis , Child , Child, Preschool , Female , Humans , Male , Prospective Studies , Transglutaminases/immunology
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL