Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
PLoS One ; 14(2): e0210828, 2019.
Article in English | MEDLINE | ID: mdl-30735525

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is one of the main causes of chronic liver disease. NAFLD begins with excessive lipid accumulation in the liver and progresses to nonalcoholic steatohepatitis (NASH) and cirrhosis. NAFLD is closely linked to dysregulated hepatic lipid metabolism. Although recent studies have reported that epidermal growth factor receptor (EGFR) signaling regulates lipid metabolism, the roles of EGFR and EGFR inhibitors as modulators of lipid metabolism are largely unknown. Here, we investigated whether inhibiting EGFR using the EGFR tyrosine kinase inhibitor (TKI) PD153035 improves NAFLD. Our results demonstrate that EGFR was activated in liver tissues from high fat diet (HFD)-induced NAFLD mice. Inhibiting EGFR using PD153035 significantly reduced phosphatidylinositol-3-kinase/protein kinase B signaling and sterol responsive elementary binding protein 1 and 2 expression, which prevented HFD-induced hepatic steatosis and hypercholesterolemia by reducing de novo lipogenesis and cholesterol synthesis and enhancing fatty acid oxidation. Additionally, inhibiting EGFR improved HFD-induced glucose intolerance. In conclusion, these results indicate that EGFR plays an important role in NAFLD and is a potential therapeutic target.


Subject(s)
Diabetes Mellitus, Experimental , Dietary Fats/adverse effects , ErbB Receptors/antagonists & inhibitors , Non-alcoholic Fatty Liver Disease , Quinazolines/pharmacology , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/pathology , Dietary Fats/pharmacology , ErbB Receptors/metabolism , Humans , Lipogenesis/drug effects , Male , Mice , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/pathology
2.
F1000Res ; 7: 318, 2018.
Article in English | MEDLINE | ID: mdl-29707204

ABSTRACT

Background: We previously identified the transcriptional regulator Zbtb32 as a factor that can promote T cell tolerance in the Non-Obese Diabetic (NOD) mouse, a model of Type 1 diabetes. Antigen targeted to DCIR2 + dendritic cells (DCs) in vivo inhibited both diabetes and effector T cell expansion in NOD mice. Furthermore, Zbtb32 was preferentially induced in autoreactive CD4 T cells stimulated by these tolerogenic DCIR2 + DCs, and overexpression of Zbtb32 in islet-specific T cells inhibited the diabetes development by limiting T cell proliferation and cytokine production. Methods: To further understand the role of Zbtb32 in T cell tolerance induction, we have now used CRISPR to target the Zbtb32 gene for deletion directly in NOD mice and characterized the mutant mice. We hypothesized that the systemic loss of Zbtb32 in NOD mice would lead to increased T cell activation and increased diabetes pathogenesis. Results: Although NOD.Zbtb32 -/- male NOD mice showed a trend towards increased diabetes incidence compared to littermate controls, the difference was not significant. Furthermore, no significant alteration in lymphocyte number or function was observed. Importantly, in vitro stimulation of lymphocytes from NOD.Zbtb32 -/- mice did not produce the expected hypersensitive phenotype observed in other genetic strains, potentially due to compensation by homologous genes. Conclusions: The loss of Zbtb32 in the NOD background does not result in the expected T cell activation phenotype.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Diabetes Mellitus, Experimental/epidemiology , Immune Tolerance/immunology , Lymphocyte Activation/immunology , Repressor Proteins/physiology , Animals , CRISPR-Cas Systems , Cells, Cultured , Cytokines/metabolism , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Female , Incidence , Male , Mice , Mice, Inbred NOD , Repressor Proteins/antagonists & inhibitors
3.
PLoS One ; 12(8): e0181964, 2017.
Article in English | MEDLINE | ID: mdl-28771521

ABSTRACT

Microbes, including viruses, influence type 1 diabetes (T1D) development, but many such influences remain undefined. Previous work on underlying immune mechanisms has focussed on cytokines and T cells. Here, we compared two nonobese diabetic (NOD) mouse colonies, NODlow and NODhigh, differing markedly in their cumulative T1D incidence (22% vs. 90% by 30 weeks in females). NODhigh mice harbored more complex intestinal microbiota, including several pathobionts; both colonies harbored segmented filamentous bacteria (SFB), thought to suppress T1D. Young NODhigh females had increased B-cell activation in their mesenteric lymph nodes. These phenotypes were transmissible. Co-housing of NODlow with NODhigh mice after weaning did not change T1D development, but T1D incidence was increased in female offspring of co-housed NODlow mice, which were exposed to the NODhigh environment both before and after weaning. These offspring also acquired microbiota and B-cell activation approaching those of NODhigh mice. In NODlow females, the low rate of T1D was unaffected by cyclophosphamide but increased by PD-L1 blockade. Thus, environmental exposures that are innocuous later in life may promote T1D progression if acquired early during immune development, possibly by altering B-cell activation and/or PD-L1 function. Moreover, T1D suppression in NOD mice by SFB may depend on the presence of other microbial influences. The complexity of microbial immune regulation revealed in this murine model may also be relevant to the environmental regulation of human T1D.


Subject(s)
B-Lymphocytes/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 1/immunology , Environment , T-Lymphocytes/immunology , Animals , B-Lymphocytes/pathology , B7-H1 Antigen/metabolism , Cytokines/metabolism , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/pathology , Female , Incidence , Lymphocyte Activation , Male , Mice , Mice, Inbred NOD , T-Lymphocytes/pathology
4.
J Am Soc Nephrol ; 23(3): 405-11, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22193383

ABSTRACT

Rodent models exhibit only the earliest features of human diabetic nephropathy, which limits our ability to investigate new therapies. Hypertension is a prerequisite for advanced diabetic nephropathy in humans, so its rarity in typical rodent models may partly explain their resistance to nephropathy. Here, we used the Cyp1a1mRen2 rat, in which the murine renin-2 gene is incorporated under the Cytochrome P4501a1 promoter. In this transgenic strain, administration of low-dose dietary indole-3-carbinol induces moderate hypertension. In the absence of hypertension, streptozotocin-induced diabetes resulted in a 14-fold increase in albuminuria but only mild changes in histology and gene expression despite 28 weeks of marked hyperglycemia. In the presence of induced hypertension, hyperglycemia resulted in a 500-fold increase in albuminuria, marked glomerulosclerosis and tubulointerstitial fibrosis, and induction of many of the same pathways that are upregulated in the tubulointerstitium in human diabetic nephropathy. In conclusion, although induction of diabetes alone in rodents has limited utility to model human diabetic nephropathy, renin-dependent hypertension and hyperglycemia synergize to recapitulate many of the clinical, histological, and gene expression changes observed in humans.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/etiology , Disease Models, Animal , Hyperglycemia/complications , Hypertension/complications , Renin , Albuminuria/etiology , Albuminuria/urine , Animals , Comorbidity , Cytochrome P-450 CYP1A1/genetics , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/epidemiology , Diabetic Nephropathies/pathology , Diabetic Nephropathies/urine , Fibrosis , Humans , Hyperglycemia/chemically induced , Hyperglycemia/epidemiology , Hypertension/chemically induced , Hypertension/epidemiology , Indoles/adverse effects , Kidney Tubules/pathology , Mice , Rats , Rats, Transgenic , Renin/genetics , Streptozocin/adverse effects
5.
Ann Hepatol ; 11(1): 118-27, 2012.
Article in English | MEDLINE | ID: mdl-22166570

ABSTRACT

PURPOSE: To explore the possible intermediary pathways through which diabetes mellitus (DM) adversely worsens hepatocellular carcinoma (HCC), focusing on cell life controllers as some transcription factors and inflammatory mediators. MATERIAL AND METHODS: Forty male albino rats were divided into four groups, control, cancer [given single intra-peritoneal (IP) dose of diethyl nitrosamine, NDEA, 125 mg/kg body weight], diabetic (given single dose of streptozotocin, STZ, 65 mg/kg) and cancer diabetic. HCC was initiated with NDEA, 3 weeks later, DM was induced with STZ. At 14th week, animals were sacrificed. Serum ALT, AST, GGT activities, AFP, IL-6, TNF-α levels and liver tissue Bax and Bcl2 proteins were measured. Liver sections were stained for histological examination. Both histological and AFP variations were chosen to prove cancer development. RESULTS: NDEA group showed significant increase in liver weight, serum ALT, AST, GGT, AFP, TNF-α, IL-6 and liver Bcl2 protein with decrease in total body weight, liver Bax protein and Bax/Bcl2 ratio. These effects were more pronounced in DENA plus STZ group. IL-6, TNF-α and Bcl2 were positively correlated while Bax and Bax/Bcl2 ratio were negatively correlated to AFP levels reflecting potential diagnostic value. CONCLUSION: Co-induction of DM in the course of hepatocarcinogenesis can dramatically influence disease progression through inflammation and retarded apoptosis. The suggested apoptotic and inflammatory markers seem to be beneficial diagnostic tools for HCC and improve the diagnostic performance of AFP.


Subject(s)
Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/metabolism , Cytokines/metabolism , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/metabolism , Liver Neoplasms, Experimental/epidemiology , Liver Neoplasms, Experimental/metabolism , Animals , Apoptosis/physiology , Biopsy , Carcinoma, Hepatocellular/chemically induced , Comorbidity , Diabetes Mellitus, Experimental/chemically induced , Diethylnitrosamine/adverse effects , Disease Models, Animal , Disease Progression , Interleukin-6/blood , Liver/metabolism , Liver/pathology , Liver Neoplasms, Experimental/chemically induced , Male , Rats , Rats, Mutant Strains , Streptozocin/adverse effects , Tumor Necrosis Factor-alpha/blood , bcl-2-Associated X Protein/metabolism , bcl-Associated Death Protein/metabolism
6.
Int J Exp Pathol ; 92(6): 392-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21950537

ABSTRACT

As pulmonary emphysema and diabetes mellitus are common diseases, concomitance of both is correspondingly expected to occur frequently. To examine whether insulin influences the development of inflammation in the alveolar septa, diabetic male Wistar rats (alloxan, 42 mg/kg, i.v., n = 37) and matching controls (n = 31) were used. Ten days after alloxan injection, diabetic and control rats were instilled with physiologic saline solution containing porcine pancreatic elastase (PPE, 0.25 IU/0.2 ml, right lung) or saline only (left lung). The following analyses were performed: (i) number of leucocytes in the bronchoalveolar lavage (BAL) fluid of the animals, 6 h after PPE/saline instillation (early time point); and (ii) mean alveolar diameter (µm) and quantification of elastic and collagen fibres (%) 50 days after PPE/saline instillation (late time point). Relative to controls, alloxan-induced diabetic rats showed a 42% reduction in the number of neutrophils in BAL fluid, a 20% increase in the mean alveolar diameter and a 33% decrease in elastic fibre density in the alveolar septa. Treatment of diabetic rats with 4 IU neutral protamine Hagedorn (NPH) insulin, 2 h before elastase instillation, restored the number of neutrophils in the BAL fluid. The mean alveolar diameter and elastic fibre content in alveolar septa matched the values observed in control rats if diabetic rats were treated with 4 IU NPH insulin 2 h before instillation followed by 2 IU/day for the next 50 days. Density of collagen fibres did not differ between the various groups. Thus, the data presented suggest that insulin modulates the inflammatory and repair responses in elastase-induced emphysema, and assures normal repair and tissue remodelling.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Inflammation/metabolism , Inflammation/pathology , Insulin/pharmacology , Pancreatic Elastase/adverse effects , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/metabolism , Alloxan/adverse effects , Animals , Blood Glucose/metabolism , Cell Movement/drug effects , Cell Movement/physiology , Collagen/metabolism , Comorbidity , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/epidemiology , Disease Models, Animal , Dose-Response Relationship, Drug , Elastic Tissue/metabolism , Insulin, Isophane/pharmacology , Leukocytes/pathology , Male , Pulmonary Emphysema/epidemiology , Rats , Rats, Wistar
7.
J Am Soc Hypertens ; 5(1): 7-11, 2011.
Article in English | MEDLINE | ID: mdl-21269910

ABSTRACT

Type 2 diabetes can impair the outcome of stroke as well as increase stroke risk; however, the sex difference in ischemic brain damage is not well known, and even less is known about the difference in diabetes. We therefore investigated the possible gender difference in brain damage after stroke associated with type 2 diabetes using a mouse model, KKAy. Female KKAy showed a much larger ischemic area compared with male KKAy. NADPH oxidase activity in the brain was also increased more in female than in male mice. Ovariectomy enhanced the ischemic area, and treatment with estradiol markedly attenuated the ischemic area to that in female KKAy, with a reduction of NADPH oxidase activity. Female and OVX mice showed improvement of cerebral blood flow (CBF) at 1 hour after middle cerebral artery (MCA) occlusion, but no significant difference in CBF of the ipsilateral penumbra and ipsilateral core 24 hours after MCA occlusion was observed among each group. Severe ischemic brain damage was observed in female KKAy compared with male KKAy. Estrogen showed a protective effect on the brain, at least partly from attenuation of oxidative stress in the female brain. These findings suggest that brain damage in diabetes mellitus might be more marked in women than in men.


Subject(s)
Brain Ischemia/epidemiology , Diabetes Mellitus, Experimental/epidemiology , Diabetic Angiopathies/epidemiology , Diabetic Neuropathies/epidemiology , Stroke/complications , Animals , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , NADPH Oxidases/metabolism , Ovariectomy , Sex Factors
9.
J Surg Res ; 168(1): 111-8, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-19959191

ABSTRACT

The incidence and prevalence of diabetes have recently increased. Hyperglycemia, which is commonly seen in intensive care medicine, is associated with increased morbidity and mortality. For instance, diabetes is associated with altered immune and hemostatic responses. High mobility group box 1 (HMGB1) protein plays a key role in various inflammatory diseases. This study investigated the increase in lung damage due to diabetes and the rise in HMGB1 levels in a lipopolysaccharide (LPS)-induced systemic inflammation rat model. Diabetes was induced by streptozotocin infusion 4 wk prior to LPS administration, followed by measurements of blood glucose and serum cytokine levels. Separate cohorts were sacrificed 12h post-LPS administration and analyzed for lung damage. Diabetic animals had significantly higher blood glucose and enhanced lung damage. In addition, levels of serum HMGB1, tumor necrosis factor-α, and interleukin-6 were increased in diabetic rats. Diabetes may exacerbate systemic inflammation as evidenced by higher serum HMGB1 and cytokine levels and enhanced lung damage in the rat systemic inflammation model.


Subject(s)
Acute Lung Injury/blood , Acute Lung Injury/epidemiology , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/epidemiology , HMGB1 Protein/blood , Acute Lung Injury/chemically induced , Animals , Comorbidity , Diabetes Mellitus, Experimental/chemically induced , Disease Models, Animal , Endotoxins/adverse effects , Insulin/blood , Interleukin-6/blood , Male , Rats , Rats, Wistar , Streptozocin/adverse effects , Tumor Necrosis Factor-alpha/blood
10.
Circulation ; 121(19): 2092-101, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20439786

ABSTRACT

BACKGROUND: The natural history of heterogeneous atherosclerotic plaques and the role of local hemodynamic factors throughout their development are unknown. We performed a serial study to assess the role of endothelial shear stress (ESS) and vascular remodeling in the natural history of coronary atherosclerosis. METHODS AND RESULTS: Intravascular ultrasound-based 3-dimensional reconstruction of all major coronary arteries (n=15) was performed serially in vivo in 5 swine 4, 11, 16, 23, and 36 weeks after induction of diabetes mellitus and hyperlipidemia. The reconstructed arteries were divided into 3-mm-long segments (n=304). ESS was calculated in all segments at all time points through the use of computational fluid dynamics. Vascular remodeling was assessed at each time point in all segments containing significant plaque, defined as maximal intima-media thickness >/=0.5 mm, at week 36 (n=220). Plaque started to develop at week 11 and progressively advanced toward heterogeneous, multifocal lesions at all subsequent time points. Low ESS promoted the initiation and subsequent progression of plaques. The local remodeling response changed substantially over time and determined future plaque evolution. Excessive expansive remodeling developed in regions of very low ESS, further exacerbated the low ESS, and was associated with the most marked plaque progression. The combined assessment of ESS, remodeling, and plaque severity enabled the early identification of plaques that evolved to high-risk lesions at week 36. CONCLUSIONS: The synergistic effect of local ESS and the remodeling response to plaque formation determine the natural history of individual lesions. Combined in vivo assessment of ESS and remodeling may predict the focal formation of high-risk coronary plaque.


Subject(s)
Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/physiopathology , Coronary Circulation/physiology , Endothelium, Vascular/physiology , Ventricular Remodeling/physiology , Animals , Coronary Artery Disease/epidemiology , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/diagnostic imaging , Diabetic Angiopathies/epidemiology , Diabetic Angiopathies/physiopathology , Disease Models, Animal , Male , Predictive Value of Tests , Risk Factors , Stress, Mechanical , Swine , Ultrasonography, Interventional
11.
J Cereb Blood Flow Metab ; 30(4): 729-33, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20068574

ABSTRACT

Animal models of ischemic stroke often neglect comorbidities common in patients. This study shows the feasibility of inducing stroke by 2 h of thread occlusion of the middle cerebral artery in aged (56 week old) spontaneously hypertensive rats (SHRs) with both acute (2 weeks) and chronic (36 weeks) diabetes. After modifying the streptozotocin dosing regimen to ensure that old SHRs survived the induction of diabetes, few died after induction of stroke. Induction of stroke is feasible in rats with multiple comorbidities. Inclusion of such comorbid animals may improve translation from the research laboratory to the clinic.


Subject(s)
Diabetes Mellitus, Experimental/physiopathology , Hypertension/physiopathology , Stroke/physiopathology , Animals , Comorbidity , Diabetes Mellitus, Experimental/epidemiology , Disease Models, Animal , Humans , Hypertension/epidemiology , Rats , Rats, Inbred SHR , Stroke/epidemiology , Stroke/mortality
12.
J Immunol ; 183(9): 5728-37, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19812207

ABSTRACT

Inflammation following tissue damage promotes lymphocyte recruitment, tissue remodeling, and wound healing while maintaining self tolerance. Endogenous signals associated with tissue damage and cell death have been proposed to initiate and instruct immune responses following injury. In this study, we have examined the effects of elevated levels of a candidate endogenous danger signal, heat shock cognate protein 70 (hsc70), on stimulation of inflammation and autoimmunity following cell damage. We find that damage to pancreatic beta cells expressing additional cytosolic hsc70 leads to an increased incidence of diabetes in a transgenic mouse model. Steady-state levels of activated APC and T cell populations in the draining lymph node were enhanced, which further increased following streptozotocin-induced beta cell death. In addition, proinflammatory serum cytokines, and lymphocyte recruitment were increased in hsc70 transgenic mice. Islet Ag-specific T cells underwent a greater extent of proliferation in the lymph nodes of mice expressing hsc70 following beta cell damage, suggesting elevated Ag presentation following release of Ag in the presence of hsc70. These findings suggest that an elevated content of hsc70 in cells undergoing necrotic or apoptotic cell death can increase the extent of sterile inflammation and increase the susceptibility to autoimmunity.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , HSC70 Heat-Shock Proteins/biosynthesis , HSC70 Heat-Shock Proteins/genetics , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Animals , Autoimmune Diseases/epidemiology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cattle , Cell Death/genetics , Cell Death/immunology , Cell Movement/genetics , Cell Movement/immunology , Cytosol/immunology , Cytosol/metabolism , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Incidence , Islets of Langerhans/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Rats
14.
J Sex Med ; 6 Suppl 3: 269-78, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19267849

ABSTRACT

INTRODUCTION: The combination of independent risk factors for erectile dysfunction, obesity, hypertension, and diabetes are collectively manifested in a condition known as metabolic syndrome X (MSX). However, the regulatory mechanisms responsible for the erectile dysfunction (ED) are not fully understood. Clinical studies suggest that a pleiotropic effect of statin's ability to enhance vascular relaxation might be through an impact on nitric oxide signaling or through a regulation of RhoA activation. AIM: We hypothesized that regulatory aspects of short-term statin therapy involve the alteration of the RhoA/Rho-kinase signaling cascade and will reverse the ED seen in a rat model of MSX. MAIN OUTCOME MEASURES: The magnitude and sensitivity of the voltage-dependent maintenance of intracavernosal blood pressure and mean arterial blood pressure. These responses were correlated with tissue protein and mRNA expression levels of RhoA and Rho kinases. METHODS: Erectile function was evaluated by assessing voltage-dependent stimulation of the cavernosal nerve in 16-20 weeks old lean and obese-diabetic Zucker rats treated with 5 mg/kg/day of rosuvastatin intraperitoneally for 3 days. Cavernosal tissue RhoA and Rho-kinases expression levels were evaluated by real-time reverse transcriptase-polymerase chain reaction, Western blot. RESULTS: The voltage-dependent erectile responses were suppressed by >30% in the obese-diabetic Zucker rat. The 3-day treatment with rosuvastatin partially restored the erectile response. The Rho-kinase inhibitor, H-1152, dose dependently increased the erectile responses and shifted the voltage sensitivity with statin treatment. Analysis of protein expression levels suggested elevation of RhoA and Rho kinases in obese-diabetics and statin treatment lowering Rho-kinase II. The RhoA and Rho-kinase II mRNA levels were significantly reduced in the rosuvastatin-treated obese-diabetic animals. CONCLUSIONS: These results support a hypothesis that short-term statin therapy may lower RhoA/Rho-kinase expression levels and improve cavernosal blood pressure response to Rho-kinase inhibition and voltage-stimulation, and reversing an augmented vasoconstricted state associated with diabetes and/or hypertension in MSX.


Subject(s)
Anticholesteremic Agents/pharmacology , Anticholesteremic Agents/therapeutic use , Diabetes Mellitus, Experimental/epidemiology , Electric Stimulation/methods , Enzyme Inhibitors/pharmacology , Erectile Dysfunction , Fluorobenzenes/pharmacology , Fluorobenzenes/therapeutic use , Impotence, Vasculogenic/etiology , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Metabolic Syndrome/epidemiology , Nitric Oxide/metabolism , Obesity/epidemiology , Phosphodiesterase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , rho-Associated Kinases/metabolism , Animals , Anticholesteremic Agents/administration & dosage , Blotting, Western , Enzyme Inhibitors/administration & dosage , Erectile Dysfunction/drug therapy , Erectile Dysfunction/epidemiology , Erectile Dysfunction/physiopathology , Fluorobenzenes/administration & dosage , Impotence, Vasculogenic/enzymology , Male , Phosphodiesterase Inhibitors/administration & dosage , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/administration & dosage , Rats , Rats, Zucker , Rosuvastatin Calcium , Sulfonamides/administration & dosage
15.
Diabetes ; 56(6): 1731-6, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17389329

ABSTRACT

OBJECTIVE: The biobreeding diabetes-prone (BBDP) rat spontaneously develops type 1 diabetes. Two of the genetic factors contributing to this syndrome are the major histocompatibility complex (Iddm1) and a Gimap5 mutation (Iddm2) responsible for a T-lymphopenia. Susceptibility to experimentally induced type 1 diabetes is widespread among nonlymphopenic (wild-type Iddm2) rat strains provided they share the BBDP Iddm1 allele. The question follows as to whether spontaneous and experimentally induced type 1 diabetes share susceptibility loci besides Iddm1. Our objectives were to map a novel, serendipitously discovered Iddm locus, confirm its effects by developing congenic sublines, and assess its differential contribution to spontaneous and experimentally induced type 1 diabetes. RESEARCH DESIGN AND METHODS: An unexpected reduction in spontaneous type 1 diabetes incidence (86 to 31%, P < 0.0001) was observed in a BBDP line congenic for a Wistar Furth-derived allotypic marker, RT7 (chromosome 13). Genome-wide analysis revealed that, besides the RT7 locus, a Wistar Furth chromosome 8 fragment had also been introduced. The contribution of these intervals to diabetes resistance was assessed through linkage analysis using 134 F2 (BBDP x double congenic line) animals and a panel of congenic sublines. One of these sublines, resistant to spontaneous type 1 diabetes, was tested for susceptibility to experimentally induced type 1 diabetes. RESULTS: Both linkage analysis and congenic sublines mapped a novel locus (Iddm24) to the telomeric 10.34 Mb of chromosome 8, influencing cumulative incidence and age of onset of spontaneous type 1 diabetes but not insulitis nor experimentally induced type 1 diabetes. CONCLUSIONS: This study has identified a type 1 diabetes susceptibility locus that appears to act after the development of insulitis and that regulates spontaneous type 1 diabetes exclusively.


Subject(s)
Chromosome Mapping , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Animals , Diabetes Mellitus, Experimental/genetics , Disease Models, Animal , Rats , Rats, Inbred BB , Telomere/genetics
16.
Hypertens Res ; 21(1): 1-6, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9582101

ABSTRACT

The present study was designed to develop an animal model of multiple risk factors, including obesity, hypertension, non-insulin-dependent diabetes mellitus, and hyperlipidemia. Hypothalamic obesity was induced by neonatal monosodium glutamate (MSG) treatment in spontaneously hypertensive rats (SHR). Female newborn SHR were treated intraperitoneally with 2 or 4 mg/kg body weight of MSG for 5 days. Obesity developed in SHR treated with 4 mg/kg of MSG but not in SHR treated with 2 mg/kg of MSG. Obese SHR had impaired glucose tolerance, hyperinsulinemia, and hypertriglyceridemia. However, the severity of hypertension was attenuated in obese SHR as compared with control SHR. The degree of obesity was closely related to the metabolic abnormalities, but inversely correlated with the blood pressure level. Macrovascular changes were investigated in obese SHR at 14 months of age. Intimal thickening was accelerated in the carotid artery of obese SHR as compared with that of nonobese SHR. Aortic contents of DNA and total cholesterol were significantly increased in obese SHR. SHR associated with MSG-induced obesity showed major manifestations of metabolic syndrome X. This animal model may be useful to study the clustering of risk factors for the development of macrovascular diseases.


Subject(s)
Arteriosclerosis/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Disease Models, Animal , Hypertension/physiopathology , Obesity/physiopathology , Rats, Inbred SHR , Animals , Animals, Newborn , Aorta/chemistry , Arteriosclerosis/epidemiology , Arteriosclerosis/pathology , Carotid Arteries/pathology , Cholesterol/analysis , DNA/analysis , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/epidemiology , Female , Food Additives , Glycated Hemoglobin/analysis , Hyperinsulinism/epidemiology , Hyperinsulinism/physiopathology , Hyperlipidemias/epidemiology , Hyperlipidemias/physiopathology , Hypertension/epidemiology , Hypertriglyceridemia/epidemiology , Hypertriglyceridemia/physiopathology , Obesity/chemically induced , Obesity/epidemiology , Rats , Risk Factors , Sodium Glutamate , Tunica Intima/pathology
17.
Diabetes ; 47(1): 32-8, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9421371

ABSTRACT

Previous studies have shown that anti-gamma-interferon (IFN-gamma) antibody reduces the frequency of autoimmune IDDM in the DP-BB rat. We tested the effects of systemically administered recombinant rat IFN-gamma in both DP-BB and DR-BB rats. Unexpectedly, IFN-gamma markedly reduced the incidence of IDDM as compared with control rats when administered six times per week at a dosage of 280,000 U between ages 30-35 to 105 days or ages 60-64 to 105 days. A lower dosage (28,000 U on alternate days) was also protective when administered to DP-BB rats between birth and age 60 days. However, long-lasting protection against IDDM development over the 1-year study period was achieved only by the highest dosage of IFN-gamma administered from age 30 to 105 days. Ex vivo production of tumor necrosis factor-alpha from splenic lymphoid cells (SLCs) and peritoneal macrophages of the rats treated with IFN-gamma was comparable with that of controls; however, SLCs from the IFN-gamma-treated animals secreted lower amounts of IFN-gamma after stimulation with concanavalin A. IFN-gamma treatment also markedly reduced the frequency of phenotypically activated SLC-expressing class II antigens and interleukin-2 receptor. Finally, in agreement with the observed antidiabetogenic effects, exogenously administered IFN-gamma induced neither insulitis nor IDDM development in DR-BB rats, a subline of DP-BB rats in which autoimmune diabetes rarely occurs spontaneously but can be induced by administration of polyinosinic-polycytidilic acid.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/prevention & control , Hypoglycemic Agents/pharmacology , Interferon-gamma/pharmacology , Aging/physiology , Animals , Concanavalin A/pharmacology , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/physiopathology , Dose-Response Relationship, Drug , Female , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/metabolism , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Immunosuppressive Agents/pharmacology , Incidence , Injections, Intraperitoneal , Interferon-gamma/administration & dosage , Interferon-gamma/metabolism , Interferon-gamma/therapeutic use , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/metabolism , Male , Phenotype , Random Allocation , Rats , Rats, Inbred BB , Receptors, Interleukin-2/analysis , Receptors, Interleukin-2/metabolism , Recombinant Proteins , Spleen/cytology , Spleen/metabolism , Tacrolimus/pharmacology , Tumor Necrosis Factor-alpha/metabolism
18.
Diabetes ; 46(7): 1133-40, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9200647

ABSTRACT

One theory of the pathogenesis of IDDM proposes that exposure to cow's milk proteins triggers the disease in genetically susceptible individuals. We tested this hypothesis in the BB/Wor rat model of human IDDM. Diabetes-prone (DP) BB/Wor rats spontaneously develop IDDM. Coisogenic diabetes-resistant (DR) BB/Wor rats do not develop diabetes spontaneously, but IDDM can readily be induced by treatment with polyinosinic:polycytidylic acid and depletion of RT6+ T-cells. Pregnant BB/Wor rats were fed one of four experimental diets or a standard Purina commercial rat chow (5010) that was certified to be free of cow's milk protein. Offspring were maintained on the maternal diet after weaning. DP-BB/Wor rats, fed either of two experimental diets based on hydrolyzed casein and free of intact milk protein (Nutramigen or D11236), developed diabetes at only half the rate of animals fed Purina 5010 chow. Neither the addition of bovine serum albumin (BSA) to Nutramigen nor the substitution of total milk protein for the hydrolyzed casein in the D11236 diet increased the frequency of spontaneous diabetes. In contrast, there was no relationship between diet and susceptibility of DR-BB/Wor rats to IDDM induction. However, the methods used to induce IDDM in DR-BB/Wor animals were found to induce antibodies against BSA. We conclude the following: 1) Dietary modification can reduce spontaneous IDDM expression in DP-BB/Wor rats, but the agent of protection is not elimination of cow's milk protein. 2) The addition of BSA or intact milk protein does not abrogate the effectiveness of a protective diet. 3) The genetic susceptibility of the DR-BB/Wor rat to autoimmune diabetes is unaffected by any of the tested diets, but a role of anti-BSA-like autoreactivity in IDDM expression cannot be excluded.


Subject(s)
Caseins/adverse effects , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Type 1/epidemiology , Diet/adverse effects , Milk Proteins/adverse effects , Milk/adverse effects , Animals , Caseins/administration & dosage , Cattle , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Type 1/etiology , Disease Models, Animal , Female , Incidence , Male , Milk/chemistry , Milk Proteins/administration & dosage , Rats , Rats, Inbred BB , Serum Albumin/administration & dosage , Serum Albumin/adverse effects , Serum Albumin/immunology
19.
Diabetes ; 46(1): 34-9, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8971078

ABSTRACT

IDDM in humans and in nonobese diabetic (NOD) mice is a T-cell-dependent autoimmune disease in which the beta-cells of the pancreatic islets are destroyed. Several putative beta-cell autoantigens have been identified, but insulin and its precursor, proinsulin, are the only ones that are beta-cell specific. (Pro)insulin may be a key autoantigen in IDDM. To address the role of proinsulin in the development of IDDM, we generated NOD mice transgenic for the mouse proinsulin II gene driven off a major histocompatibility complex (MHC) class II promoter to direct expression of the transgene to MHC class II bearing cells, including those in the thymus, with the aim of deleting proinsulin-reactive T-cells. The mononuclear cell infiltration of the islets (insulitis) is almost completely absent, and diabetes is prevented in these transgenic NOD mice. The mononuclear cell infiltration of the salivary glands (sialitis) and immune responses to ovalbumin (OVA) are not altered, indicating that the protective effect of the transgene is specific for islet pathology and not due to general immunosuppression. We conclude that autoimmunity to proinsulin plays a pivotal role in the development of IDDM.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Proinsulin/biosynthesis , Protein Precursors/biosynthesis , Aging , Animals , Autoantigens/immunology , Cyclophosphamide , DNA Primers , Diabetes Mellitus, Experimental/epidemiology , Diabetes Mellitus, Experimental/prevention & control , Diabetes Mellitus, Type 1/epidemiology , Diabetes Mellitus, Type 1/genetics , Female , Genes, MHC Class II , Genetic Therapy , Glutamate Decarboxylase/immunology , Humans , Insulin , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Proinsulin/genetics , Promoter Regions, Genetic , Protein Precursors/genetics , RNA, Messenger/biosynthesis , Transcription, Genetic
20.
Diabetologia ; 39(4): 383-90, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8777987

ABSTRACT

The diabetogenic action of the beta-cell toxin, alloxan, is transient when administered to mice at a dosage of 50 mg/kg. We examined whether increased cholinergic activity is involved in the compensatory mechanisms. Therefore, following administration of alloxan, methylatropine (32 mumol/kg) was given intraperitoneally once daily for 5 consecutive days. Methyl atropine worsened the degree of hyperglycaemia during the first week after alloxan administration. Recovery from the diabetes mellitus was observed in a substantial number of animals given alloxan without methyl atropine, whereas the risk of developing manifest diabetes was markedly enhanced by methyl atropine. At 35 days after alloxan administration, 33% of the animals, which were given alloxan alone and were diabetic after 4 days, still had diabetes. In contrast, of the animals rendered diabetic by alloxan with concomitant atropinization, 92% remained diabetic throughout the study (p = 0.0145 vs alloxan alone). Glucose-stimulated insulin secretion and pancreatic insulin content were markedly reduced in animals with diabetes while being less reduced in alloxan-injected animals without diabetes. Moreover, in situ hybridization and immunocytochemistry revealed markedly decreased levels of insulin mRNA and number of insulin cells in alloxan-treated animals. With regard to insulin secretion, pancreatic insulin content, insulin mRNA and insulin cell number, the reduction was the same irrespective of whether methyl atropine had been given. Thus, 5 days of atropinization increases the incidence of diabetes following alloxan at 50 mg/kg in mice. We suggest that cholinergic activity protects insulin cells from glucotoxicity during the first week after alloxan administration and therefore, reduces the frequency of diabetes.


Subject(s)
Alloxan , Atropine Derivatives/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Insulin/biosynthesis , Islets of Langerhans/metabolism , Parasympatholytics/pharmacology , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/epidemiology , Dose-Response Relationship, Drug , Drug Synergism , Female , Incidence , Insulin/blood , Islets of Langerhans/pathology , Mice , Mice, Inbred Strains , RNA, Messenger/analysis , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...