Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
JACC Basic Transl Sci ; 8(2): 189-200, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36908664

ABSTRACT

The authors determined the effect of the GLP-1 receptor agonist liraglutide on endothelial surface expression of vascular cell adhesion molecule (VCAM)-1 in murine apolipoprotein E knockout atherosclerosis. Contrast-enhanced ultrasound molecular imaging using microbubbles targeted to VCAM-1 and control microbubbles showed a 3-fold increase in endothelial surface VCAM-1 signal in vehicle-treated animals, whereas in the liraglutide-treated animals the signal ratio remained around 1 throughout the study. Liraglutide had no influence on low-density lipoprotein cholesterol or glycated hemoglobin, but reduced TNF-α, IL-1ß, MCP-1, and OPN. Aortic plaque lesion area and luminal VCAM-1 expression on immunohistology were reduced under liraglutide treatment.

2.
Nutr Diabetes ; 13(1): 1, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36604407

ABSTRACT

AIMS: To investigate the influence of the dose in the FITC-Dextran 4kDa (FD-4) permeability test in an obese mouse model, we tested the bodyweight dose regimen and a lean body mass-based dose regimen in high fat diet (HFD) mice and low fat diet (LFD) mice. We hypothesized that the FD-4 permeation result would be dose-dependent. METHODS: The two dose regimens were compared in HFD and LFD mice. Furthermore, we conducted a dose-response study to test the effect of a low or high dose of FD-4 in weight-stratified lean mice. Gene analysis of tight junctions was also carried out. RESULTS: The FD-4 intestinal permeability test was dose-dependent as we found a significant increase in plasma levels of FD-4 in obese mice with the bodyweight dose regimen. However, this difference was not detectable with the lean body mass dose regimen, even with variability-adjusted group sizes. However, the qPCR analysis revealed a decrease in tight junction gene expression in obese mice. Furthermore, we found a dose-dependent significant increase in FD-4 measured in plasma samples in lean mice. No significant difference in intestinal weight was observed between lean and obese mice. CONCLUSION: Evaluation of the intestinal permeability by FD-4 with the typical bodyweight dose regimen in obese mice will be confounded by the significant difference in dose given when compared to a lean control group. If the test dose is based on lean body mass, no significant difference in intestinal permeability is observed, even with large group sizes. Furthermore, we showed a dose-dependent difference in plasma FD-4 levels in lean mice. Therefore, we conclude that the dose should be based on lean body mass for the FD-4 permeability test if mice with considerable obesity differences are to be compared or to use another test with fixed doses.


Subject(s)
Intestines , Obesity , Mice , Animals , Mice, Obese , Obesity/metabolism , Permeability , Mice, Inbred C57BL
3.
Am J Physiol Cell Physiol ; 322(6): C1260-C1269, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35442827

ABSTRACT

Exocrine glands in the submucosa of the proximal duodenum secrete alkaline fluid containing mucus to protect the intestinal mucosa from acidic stomach contents. These glands, known as Brunner's glands, express high glucagon-like peptide 1 receptor (GLP-1R) levels. Previous studies have suggested that activation of the GLP-1R induces expression of barrier protective genes in Brunner's glands. Still, the lack of a viable in vitro culture of Brunner's glands has hampered additional studies of the functional consequences of GLP-1R activation. In this study, we established a procedure to isolate and culture cells derived from murine Brunner's glands. The isolated glandular cells retained functional GLP-1R expression in culture, making this in vitro system suitable for the study of GLP-1R activation. We found that cells derived from the Brunner's glands of mice pretreated with semaglutide contained significantly more mucus compared with Brunner's glands from vehicle-treated mice. Our data suggest a protective intestinal response upon semaglutide treatment, but further studies are required to leverage the full potential of cultured Brunner's gland cells.


Subject(s)
Brunner Glands , Glucagon-Like Peptide-1 Receptor , Animals , Brunner Glands/chemistry , Brunner Glands/metabolism , Cell Culture Techniques , Duodenum/metabolism , Glucagon-Like Peptide-1 Receptor/analysis , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Intestinal Mucosa/metabolism , Male , Mice , Mucus
4.
Atheroscler Plus ; 49: 32-41, 2022 Aug.
Article in English | MEDLINE | ID: mdl-36644202

ABSTRACT

Background and aims: Randomized clinical studies have shown a reduction in cardiovascular outcomes with glucagon-like peptide 1 receptor agonist (GLP-1RA) treatment with the hypothesized mechanisms being an underlying effect on atherosclerosis. Here, we aimed to assess the pharmacological effects of semaglutide in an atheroprone murine model that recapitulates central mechanisms related to vascular smooth muscle cell (VSMC) phenotypic switching and endothelial dysfunction known to operate within the atherosclerotic plaque. Methods: In study A, we employed an electrical current to the carotid artery in ApoE-/- mice to induce severe VSMC injury and death, after which the arteries were allowed to heal for 4 weeks. In study B, a constrictive cuff was added for 6 h at the site of the healed segment to induce a disturbance in blood flow. Results: Compared to vehicle, semaglutide treatment reduced the intimal and medial area by ∼66% (p = 0.007) and ∼11% (p = 0.0002), respectively. Following cuff placement, expression of the pro-inflammatory marker osteopontin and macrophage marker Mac-2 was reduced (p < 0.05) in the semaglutide-treated group compared to vehicle. GLP-1R were not expressed in murine carotid artery and human coronary vessels with and without atherosclerotic plaques, and semaglutide treatment did not affect proliferation of cultured primary human VSMCs. Conclusions: Semaglutide treatment reduced vessel remodelling following electrical injury and blood flow perturbation in an atheroprone mouse model. This effect appears to be driven by anti-inflammatory and -proliferative mechanisms independent of GLP-1 receptor-mediated signalling in the resident vascular cells. This mechanism of action may be important for cardiovascular protection.

5.
Sci Rep ; 11(1): 5241, 2021 03 04.
Article in English | MEDLINE | ID: mdl-33664407

ABSTRACT

Angiotensin converting enzyme inhibitors, among them captopril, improve survival following myocardial infarction (MI). The mechanisms of captopril action remain inadequately understood due to its diverse effects on multiple signalling pathways at different time periods following MI. Here we aimed to establish the role of captopril in late-stage post-MI remodelling. Left anterior descending artery (LAD) ligation or sham surgery was carried out in male C57BL/6J mice. Seven days post-surgery LAD ligated mice were allocated to daily vehicle or captopril treatment continued over four weeks. To provide comprehensive characterization of the changes in mouse heart following MI a 3D light sheet imaging method was established together with automated image analysis workflow. The combination of echocardiography and light sheet imaging enabled to assess cardiac function and the underlying morphological changes. We show that delayed captopril treatment does not affect infarct size but prevents left ventricle dilation and hypertrophy, resulting in improved ejection fraction. Quantification of lectin perfused blood vessels showed improved vascular density in the infarct border zone in captopril treated mice in comparison to vehicle dosed control mice. These results validate the applicability of combined echocardiographic and light sheet assessment of drug mode of action in preclinical cardiovascular research.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Captopril/pharmacology , Myocardial Infarction/drug therapy , Ventricular Function, Left/drug effects , Animals , Disease Models, Animal , Echocardiography , Heart Ventricles/diagnostic imaging , Heart Ventricles/drug effects , Heart Ventricles/pathology , Humans , Male , Mice , Microscopy , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/genetics , Myocardial Infarction/pathology
6.
Sci Rep ; 10(1): 21523, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33299076

ABSTRACT

Complications of atherosclerosis are the leading cause of morbidity and mortality worldwide. Various genetically modified mouse models are used to investigate disease trajectory with classical histology, currently the preferred methodology to elucidate plaque composition. Here, we show the strength of light-sheet fluorescence microscopy combined with deep learning image analysis for characterising and quantifying plaque burden and composition in whole aorta specimens. 3D imaging is a non-destructive method that requires minimal ex vivo handling and can be up-scaled to large sample sizes. Combined with deep learning, atherosclerotic plaque in mice can be identified without any ex vivo staining due to the autofluorescent nature of the tissue. The aorta and its branches can subsequently be segmented to determine how anatomical position affects plaque composition and progression. Here, we find the highest plaque accumulation in the aortic arch and brachiocephalic artery. Simultaneously, aortas can be stained for markers of interest (for example the pan immune cell marker CD45) and quantified. In ApoE-/- mice we observe that levels of CD45 reach a plateau after which increases in plaque volume no longer correlate to immune cell infiltration. All underlying code is made publicly available to ease adaption of the method.


Subject(s)
Plaque, Atherosclerotic/diagnostic imaging , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Animals , Aorta/pathology , Aortic Diseases , Apolipoproteins E/analysis , Atherosclerosis/complications , Atherosclerosis/pathology , Deep Learning , Disease Models, Animal , Female , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence/methods , Receptors, LDL/analysis
7.
Atherosclerosis ; 314: 1-9, 2020 12.
Article in English | MEDLINE | ID: mdl-33129080

ABSTRACT

BACKGROUND AND AIMS: Clinical interventions targeting nonlipid risk factors are needed given the high residual risk of atherothrombotic events despite effective control of dyslipidemia. Dickkopf-1 (DKK1) plays a lipid-independent role in vascular pathophysiology but its involvement in atherosclerosis development and its therapeutic attractiveness remain to be established. METHODS: Patient data, in vitro studies and pharmacological intervention in murine models of atherosclerosis were utilized. RESULTS: In patients' material (n = 127 late stage plaque specimens and n = 10 control vessels), DKK1 mRNA was found to be higher in atherosclerotic plaques versus control arteries. DKK1 protein was detected in the luminal intimal area and in the necrotic core of plaques. DKK1 was released from isolated primary human platelets (~12 - 21-fold) and endothelial cells (~1.4-2.5-fold) upon stimulation with different pathophysiological stimuli. In ApoE-/- and Ldlr-/- mice, plasma DKK1 concentrations were similar to those observed in humans, whereas DKK1 expression in different atheroprone arterial segments was very low/absent. Chronic treatment with a neutralizing DKK1 antibody effectively reduced plasma concentrations, however, plaque lesion area was not reduced in ApoE-/- and Ldlr-/- mice fed a western diet for 14 and 16 weeks. Anti-DKK1 treatment increased bone volume and bone mineral content. CONCLUSIONS: Functional inhibition of DKK1 with an antibody does not alter atherosclerosis progression in classical murine models. This may reflect the absence of DKK1 expression in plaques and more advanced animal disease models could be needed to evaluate the role and therapeutic attractiveness of DKK1 in late stage complications such as plaque destabilization, calcification, rupture and thrombosis.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Antibodies, Neutralizing , Atherosclerosis/prevention & control , Disease Models, Animal , Endothelial Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
8.
Basic Clin Pharmacol Toxicol ; 123(6): 704-713, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29953740

ABSTRACT

Although commonly associated with obesity, non-alcoholic fatty liver disease (NAFLD) is also present in the lean population representing a unique disease phenotype. Affecting 25% of the world's population, NAFLD is associated with increased mortality especially when progressed to non-alcoholic steatohepatitis (NASH). However, no approved pharmacological treatments exist. Current research focuses mainly on NASH associated with obesity, leaving the effectiveness of promising treatments in lean NASH virtually unknown. This study therefore aimed to evaluate the effect of liraglutide (glucagon-like peptide 1 analogue) and dietary intervention, alone and in combination, in guinea pigs with non-obese NASH. After 20 weeks of high-fat feeding (20% fat, 15% sucrose, 0.35% cholesterol), 40 female guinea pigs were block-randomized based on weight into four groups receiving one of four treatments for 4 weeks: continued high-fat diet (HF, control), high-fat diet and liraglutide treatment (HFL), chow diet (4% fat, 0% sucrose, 0% cholesterol; HFC) or chow diet and liraglutide treatment (HFCL). High-fat feeding induced NASH with severe fibrosis. Liraglutide decreased inflammation (p < 0.05) and hepatocyte ballooning (p < 0.05), while increasing hepatic α-tocopherol (p = 0.0154). Dietary intervention did not improve liver histopathology significantly, but decreased liver weight (p = 0.004), plasma total cholesterol (p = 0.0175), LDL-cholesterol (p = 0.0063), VLDL-cholesterol (p = 0.0034), hepatic cholesterol (p < 0.0001) and increased hepatic vitamin C (p = 0.0099). Combined liraglutide and dietary intervention induced a rapid weight loss, necessitating periodical liraglutide dose adjustment/discontinuation, limiting the strength of the findings from this group. Collectively, this pre-clinical study supports the beneficial effect of liraglutide on NASH and extends this notion to lean NASH.


Subject(s)
Hypoglycemic Agents/therapeutic use , Liraglutide/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Diet, High-Fat/adverse effects , Female , Guinea Pigs , Lipids/analysis , Liver/chemistry , Liver/drug effects , Liver/pathology , Liver Glycogen/analysis , Non-alcoholic Fatty Liver Disease/diet therapy , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress/drug effects
9.
Sci Rep ; 8(1): 5416, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29615808

ABSTRACT

In the apolipoprotein E-deficient mouse, the gut microbiota has an impact on the development of atherosclerosis, but whether such correlations are also present in rats requires investigation. Therefore, we studied female SD-Apoe tm1sage (Apoe-/-) rats fed either a Western diet or a low-fat control diet with or without gluten, which is known to promote gut microbiota changes, until 20 weeks of age. We hypothesized that the manifestation of atherosclerosis would be more severe in Apoe-/- rats fed the Western high-fat diet, as compared with rats fed the low-fat diet, and that atherosclerosis would be accelerated by gluten. Both Western diet-feeding and gluten resulted in significant changes in gut microbiota, but the microbiota impact of gluten was transient. Compared with Apoe-/- rats fed a low-fat diet, Western diet-fed Apoe-/- rats were heavier and became glucose intolerant with increased levels of oxidative stress. They developed early fatty streak lesions in their aortic sinus, while there was no evidence of atherosclerosis in the thoracic aorta. No conclusions could be made on the impact of gluten on atherosclerosis. Although Western diet-fed Apoe-/- rats exhibited a more human-like LDL dominated blood lipid profile, signs of obesity, type 2 diabetes and cardiovascular disease were modest.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/metabolism , Atherosclerosis/pathology , Diet, Western/adverse effects , Animals , Aorta/drug effects , Aorta/pathology , Atherosclerosis/microbiology , Body Weight/drug effects , Diet, High-Fat/adverse effects , Female , Gastrointestinal Microbiome , Insulin Resistance , Liver/pathology , Oxidative Stress/drug effects , Rats , Time Factors
10.
JACC Basic Transl Sci ; 3(6): 844-857, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30623143

ABSTRACT

The glucagon-like peptide-1 receptor agonists (GLP-1RAs) liraglutide and semaglutide reduce cardiovascular risk in type 2 diabetes patients. The mode of action is suggested to occur through modified atherosclerotic progression. In this study, both of the compounds significantly attenuated plaque lesion development in apolipoprotein E-deficient (ApoE-/-) mice and low-density lipoprotein receptor-deficient (LDLr-/-) mice. This attenuation was partly independent of weight and cholesterol lowering. In aortic tissue, exposure to a Western diet alters expression of genes in pathways relevant to the pathogenesis of atherosclerosis, including leukocyte recruitment, leukocyte rolling, adhesion/extravasation, cholesterol metabolism, lipid-mediated signaling, extracellular matrix protein turnover, and plaque hemorrhage. Treatment with semaglutide significantly reversed these changes. These data suggest GLP-1RAs affect atherosclerosis through an anti-inflammatory mechanism.

11.
PLoS One ; 11(12): e0168396, 2016.
Article in English | MEDLINE | ID: mdl-27992511

ABSTRACT

Chronic kidney disease (CKD) leads to uremia. CKD is characterized by a gradual increase in kidney fibrosis and loss of kidney function, which is associated with a progressive increase in risk of atherosclerosis and cardiovascular death. To prevent progression of both kidney fibrosis and atherosclerosis in uremic settings, insight into new treatment options with effects on both parameters is warranted. The GLP-1 analogue liraglutide improves glucose homeostasis, and is approved for treatment of type 2 diabetes. Animal studies suggest that GLP-1 also dampens inflammation and atherosclerosis. Our aim was to examine effects of liraglutide on kidney fibrosis and atherosclerosis in a mouse model of moderate uremia (5/6 nephrectomy (NX)). Uremic (n = 29) and sham-operated (n = 14) atherosclerosis-prone low density lipoprotein receptor knockout mice were treated with liraglutide (1000 µg/kg, s.c. once daily) or vehicle for 13 weeks. As expected, uremia increased aortic atherosclerosis. In the remnant kidneys from NX mice, flow cytometry revealed an increase in the number of monocyte-like cells (CD68+F4/80-), CD4+, and CD8+ T-cells, suggesting that moderate uremia induced kidney inflammation. Furthermore, markers of fibrosis (i.e. Col1a1 and Col3a1) were upregulated, and histological examinations showed increased glomerular diameter in NX mice. Importantly, liraglutide treatment attenuated atherosclerosis (~40%, p < 0.05) and reduced kidney inflammation in NX mice. There was no effect of liraglutide on expression of fibrosis markers and/or kidney histology. This study suggests that liraglutide has beneficial effects in a mouse model of moderate uremia by reducing atherosclerosis and attenuating kidney inflammation.


Subject(s)
Atherosclerosis/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/administration & dosage , Liraglutide/administration & dosage , Receptors, LDL/genetics , Uremia/drug therapy , Animals , Atherosclerosis/genetics , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Diabetes Mellitus, Type 2/genetics , Disease Models, Animal , Fibrosis , Gene Knockout Techniques , Hypoglycemic Agents/pharmacology , Kidney/drug effects , Kidney/immunology , Kidney/pathology , Liraglutide/pharmacology , Male , Mice , Up-Regulation/drug effects , Uremia/immunology
12.
Nutr Metab (Lond) ; 13: 51, 2016.
Article in English | MEDLINE | ID: mdl-27512407

ABSTRACT

BACKGROUND: Non-alcoholic fatty liver disease (NAFLD) and dyslipidemia are closely related. Diet plays an important role in the progression of these diseases, but the role of specific dietary components is not completely understood. Therefore, we investigated the role of dietary sucrose and fat/cholesterol on the development of dyslipidemia and NAFLD. METHODS: Seventy female guinea pigs were block-randomized (based on weight) into five groups and fed a normal chow diet (control: 4 % fat), a very high-sucrose diet (vHS: 4 % fat, 25 % sucrose), a high-fat diet (HF: 20 % fat, 0.35 % cholesterol), a high-fat/high-sucrose diet (HFHS: 20 % fat, 15 % sucrose, 0.35 % cholesterol) or a high-fat/very high-sucrose diet (HFvHS: 20 % fat, 25 % sucrose, 0.35 % cholesterol) for 16 and 25 weeks. RESULTS: All three high-fat diets induced dyslipidemia with increased concentrations of plasma cholesterol (p < 0.0001), LDL-C (p < 0.0001) and VLDL-C (p < 0.05) compared to control and vHS. Contrary to this, plasma triglycerides were increased in control and vHS compared to high-fat fed animals (p < 0.01), while circulating levels of free fatty acids were even between groups. Histological evaluation of liver sections revealed non-alcoholic steatohepatitis (NASH) with progressive inflammation and bridging fibrosis in high-fat fed animals. Accordingly, hepatic triglycerides (p < 0.05) and cholesterol (p < 0.0001) was increased alongside elevated levels of alanine and aspartate aminotransferase (p < 0.01) compared to control and vHS. CONCLUSION: Collectively, our results suggest that intake of fat and cholesterol, but not sucrose, are the main factors driving the development and progression of dyslipidemia and NAFLD/NASH.

13.
PLoS One ; 11(1): e0146439, 2016.
Article in English | MEDLINE | ID: mdl-26799618

ABSTRACT

The importance of the gut microbiota (GM) in disease development has recently received increased attention, and numerous approaches have been made to better understand this important interplay. For example, metabolites derived from the GM have been shown to promote atherosclerosis, the underlying cause of cardiovascular disease (CVD), and to increase CVD risk factors. Popular interest in the role of the intestine in a variety of disease states has now resulted in a significant proportion of individuals without coeliac disease switching to gluten-free diets. The effect of gluten-free diets on atherosclerosis and cardiovascular risk factors is largely unknown. We therefore investigated the effect of a gluten-free high-fat cholesterol-rich diet, as compared to the same diet in which the gluten peptide gliadin had been added back, on atherosclerosis and several cardiovascular risk factors in apolipoprotein E-deficient (Apoe-/-) mice. The gluten-free diet transiently altered GM composition in these mice, as compared to the gliadin-supplemented diet, but did not alter body weights, glucose tolerance, insulin levels, plasma lipids, or atherosclerosis. In parallel, other Apoe-/- mice fed the same diets were treated with ampicillin, a broad-spectrum antibiotic known to affect GM composition. Ampicillin-treatment had a marked and sustained effect on GM composition, as expected. Furthermore, although ampicillin-treated mice were slightly heavier than controls, ampicillin-treatment transiently improved glucose tolerance both in the absence or presence of gliadin, reduced plasma LDL and VLDL cholesterol levels, and reduced aortic atherosclerotic lesion area. These results demonstrate that a gluten-free diet does not seem to have beneficial effects on atherosclerosis or several CVD risk factors in this mouse model, but that sustained alteration of GM composition with a broad-spectrum antibiotic has beneficial effects on CVD risk factors and atherosclerosis. These findings support the concept that altering the microbiota might provide novel treatment strategies for CVD.


Subject(s)
Atherosclerosis/epidemiology , Cardiovascular Diseases/epidemiology , Diet, Gluten-Free , Diet, High-Fat , Gastrointestinal Microbiome/drug effects , Plaque, Atherosclerotic/pathology , Ampicillin/pharmacology , Animals , Apolipoproteins E/genetics , Cholesterol/blood , Gastrointestinal Microbiome/physiology , Gliadin/metabolism , Glucose/metabolism , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Mice , Mice, Inbred C57BL , Mice, Knockout , Plaque, Atherosclerotic/blood
14.
PLoS One ; 10(7): e0131306, 2015.
Article in English | MEDLINE | ID: mdl-26162095

ABSTRACT

Low grade inflammation is present in pre-clinical and human type 2 diabetes. In this process, several cytokines like IL-1ß and inflammatory cells like macrophages are activated and demonstrated to participate to the disease initiation and progression. IL-20 is a cytokine known to play non-redundant roles in progression of several inflammatory diseases. To address the therapeutic effect of inhibiting the IL-20 pathway in diabetes, diabetic db/db mice were treated with neutralizing anti-IL20 antibodies in vivo and both metabolic and inflammatory parameters were followed. Diabetic islets expressed the IL-20 cytokine and all IL-20 receptor components in elevated levels compared to resting non-diabetic islets. Islets were responsive to ex vivo IL-20 stimulation measured as SOCS induction and KC and IL-6 production. Neutralizing anti-IL20 treatment in vivo had no effect on HbA1c or weight although the slope of blood glucose increase was lowered. In contrast, anti-IL20 treatment significantly reduced the systemic low-grade inflammation and modulated the local pancreatic immunity. Significant reduction of the systemic IL-1ß and MCP-1 was demonstrated upon anti-IL20 treatment which was orchestrated with a reduced RANTES, IL-16 and IL-2 but increased TIMP-1, MCP-1 and IL-6 protein expression locally in the pancreas. Interestingly, anti-IL20 treatment induced an expansion of the myeloid suppressor CD11bGr1int macrophage while reducing the number of CD8 T cells. Taken together, anti-IL20 treatment showed moderate effects on metabolic parameters, but significantly altered the low grade local and systemic inflammation. Hence, future combination therapies with anti-IL20 may provide beneficial therapeutic effects in type 2 diabetes through a reduction of inflammation.


Subject(s)
Antibodies, Neutralizing/pharmacology , Diabetes Mellitus, Type 2/prevention & control , Glycated Hemoglobin/metabolism , Inflammation/prevention & control , Interleukins/pharmacology , Animals , Cell Line, Tumor , Cytokines/blood , Cytokines/immunology , Cytokines/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/immunology , Flow Cytometry , Gene Expression , Humans , Inflammation/genetics , Inflammation/immunology , Interleukins/genetics , Interleukins/immunology , Male , Mice, Inbred C57BL , Pancreas/drug effects , Pancreas/immunology , Pancreas/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Spleen/drug effects , Spleen/immunology , Spleen/metabolism
15.
PLoS One ; 7(10): e44270, 2012.
Article in English | MEDLINE | ID: mdl-23049674

ABSTRACT

AIMS/HYPOTHESIS: Several studies have shown that adiponectin can lower blood glucose in diabetic mice. The aim of this study was to establish an effective adiponectin production process and to evaluate the anti-diabetic potential of the different adiponectin forms in diabetic mice and sand rats. METHODS: Human high molecular weight, mouse low molecular weight and mouse plus human globular adiponectin forms were expressed and purified from mammalian cells or yeast. The purified protein was administered at 10-30 mg/kg i.p. b.i.d. to diabetic db/db mice for 2 weeks. Furthermore, high molecular weight human and globular mouse adiponectin batches were administered at 5-15 mg/kg i.p. b.i.d. to diabetic sand rats for 12 days. RESULTS: Surprisingly, none of our batches had any effect on blood glucose, HbA1c, plasma lipids or body weight in diabetic db/db mice or sand rats. In vitro biological, biochemical and biophysical data suggest that the protein was correctly folded and biologically active. CONCLUSIONS/INTERPRETATION: Recombinant adiponectin is ineffective at lowering blood glucose in diabetic db/db mice or sand rats.


Subject(s)
Adiponectin/pharmacology , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Recombinant Proteins/pharmacology , Adiponectin/genetics , Adiponectin/metabolism , Animals , Body Weight/drug effects , Chromatography, Gel , Cloning, Molecular , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel , Gerbillinae , Glycated Hemoglobin/metabolism , HEK293 Cells , Humans , Lipids/blood , Mice , Mice, Obese , Mutagenesis, Site-Directed , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , Species Specificity
16.
Diabetes ; 56(1): 8-15, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17192459

ABSTRACT

Metabolic effects of the glucagon-like peptide-1 analog liraglutide and the dipeptidyl peptidase-IV inhibitor vildagliptin were compared in rats made obese by supplementary candy feeding. Female Sprague-Dawley rats were randomized to 12-week diets of chow or chow plus candy. The latter were randomized for 12 further weeks to continue their diet while receiving 0.2 mg/kg liraglutide twice daily subcutaneously, 10 mg/kg vildagliptin twice daily orally, or vehicle or to revert to chow-only diet. Energy expenditure was measured, and oral glucose tolerance tests (OGTTs) were performed. Body composition was determined by dual-energy X-ray absorptiometry scanning, and pancreatic beta-cell mass was determined by histology. Candy feeding increased weight, fat mass, and feeding-associated energy expenditure. Liraglutide or reversal to chow diet fully reversed weight and fat gains. Liraglutide was associated with decreased calorie intake and shifted food preference (increased chow/decreased candy consumption). Despite weight loss, liraglutide-treated rats did not decrease energy expenditure compared with candy-fed controls. Vildagliptin affected neither weight, food intake, nor energy expenditure. OGTTs, histology, and blood analyses indirectly suggested that both drugs increased insulin sensitivity. Liraglutide and vildagliptin inhibited obesity-associated increases in beta-cell mass. This was associated with weight and fat mass normalization with liraglutide, but not vildagliptin, where the ratio of beta-cell to body mass was low.


Subject(s)
Adamantane/analogs & derivatives , Body Weight/drug effects , Candy , Energy Intake/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Hypoglycemic Agents/pharmacology , Adamantane/pharmacology , Animals , Body Composition/drug effects , Energy Metabolism/drug effects , Glucagon-Like Peptide 1/pharmacology , Liraglutide , Nitriles , Obesity , Pancreas/drug effects , Pancreas/pathology , Pyrrolidines , Rats , Rats, Sprague-Dawley , Vildagliptin
17.
In Vivo ; 20(2): 195-203, 2006.
Article in English | MEDLINE | ID: mdl-16634519

ABSTRACT

BACKGROUND: Subcutaneously-implanted glucose sensors for continuous glucose monitoring have the potential to replace serial blood glucose measurements. The objective of the present study was to test whether continuous glucose measurements could be obtained with glucose sensors implanted in the subcutis of pigs. Moreover, the in vivo biocompatibility of the sensors was evaluated since an inflammatory reaction may lead to drift in sensor-signaling. MATERIALS AND METHODS: Two types of glucose sensor were implanted for 3 days in the subcutis of hyperglycemic pigs. The plasma glucose concentration was correlated to the sensor outputs, and tissue was sampled for histological evaluation. RESULTS: There was a good correlation between the interstitial fluid and blood glucose levels. However, there was a statistical significantly difference in linearity from days 0 and 1 to day 2 (p<0.001) and variations in the sensitivity and background current of individual sensors were observed over time. The tissue reaction caused by the sensors was a mild focal subacute fibrinous dermatitis. CONCLUSION: Continuous glucose measurement can be achieved by glucose sensors implanted in the subcutis of pigs. The observed drift in sensor signals over time may have been caused by heterophils, macrophages and/or fibrinogen at the tissue-sensor interface.


Subject(s)
Biosensing Techniques/veterinary , Blood Glucose/analysis , Monitoring, Physiologic/veterinary , Swine/blood , Animals , Biosensing Techniques/instrumentation , Dermatitis/etiology , Dermatitis/pathology , Extracellular Fluid/chemistry , Extracellular Fluid/metabolism , Female , Models, Animal , Monitoring, Physiologic/instrumentation , Monitoring, Physiologic/methods , Prostheses and Implants , Prosthesis Design , Reproducibility of Results , Subcutaneous Tissue/pathology
18.
Am J Physiol Endocrinol Metab ; 290(4): E670-7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16278249

ABSTRACT

At present, the best available estimators of beta-cell mass in humans are those based on measurement of insulin levels or appearance rates in the circulation. In several animal models, these estimators have been validated against beta-cell mass in lean animals. However, as many diabetic humans are obese, a correlation between in vivo tests and beta-cell mass must be evaluated over a range of body weights to include different levels of insulin sensitivity. For this purpose, obese (n = 10) and lean (n = 25) Göttingen minipigs were studied. Beta-cell mass had been reduced (n = 16 lean, n = 5 obese) with a combination of nicotinamide (67 mg/kg) and streptozotocin (125 mg/kg), acute insulin response (AIR) to intravenous glucose and/or arginine was tested, pulsatile insulin secretion was evaluated by deconvolution (n = 30), and beta-cell mass was determined histologically. AIR to 0.3 (r(2) = 0.4502, P < 0.0001) or 0.6 g/kg glucose (r(2) = 0.6806, P < 0.0001), 67 mg/kg arginine (r(2) = 0.5730, P < 0.001), and maximum insulin concentration (r(2) = 0.7726, P < 0.0001) were all correlated to beta-cell mass when evaluated across study groups, and regression lines were not different between lean and obese groups except for AIR to 0.3 g/kg glucose. Baseline pulse mass was not significantly correlated to beta-cell mass across the study groups (r(2) = 0.1036, NS), whereas entrained pulse mass did show a correlation across groups (r(2) = 0.4049, P < 0.001). This study supports the use of in vivo tests of insulin responses to evaluate beta-cell mass over a range of body weights in the minipig. Extensive stimulation of insulin secretion by a combination of glucose and arginine seems to give the best correlation to beta-cell mass.


Subject(s)
Insulin-Secreting Cells/physiology , Insulin/metabolism , Obesity/metabolism , Animals , Arginine/pharmacology , Blood Glucose/metabolism , Body Weight , Diabetes Mellitus, Experimental/metabolism , Glucose Tolerance Test , Insulin/blood , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Niacinamide/pharmacology , Obesity/blood , Obesity/pathology , Predictive Value of Tests , Retrospective Studies , Streptozocin/pharmacology , Swine , Swine, Miniature
19.
ILAR J ; 45(3): 303-13, 2004.
Article in English | MEDLINE | ID: mdl-15229377

ABSTRACT

Animal models of type 1 diabetes remain essential tools for investigation of the etiology and pathogenesis of the disease and, importantly, for the development of effective new treatments. Although a range of well-characterized and widely used models of type 1 diabetes in rodents are currently available, large animal models are a valuable complement to rodent models for both physiological and practical reasons. The pig is very useful in many aspects as a model for human physiology and pathophysiology because many organ systems of this species, as well as physiological and pathophysiological responses, resemble those of the human. The Göttingen minipig is particularly suitable for long-term studies because of its inherent small size and ease of handling, even at full maturity. Of particular relevance to the field of type 1 diabetes are the many similarities evident between humans and pigs with regard to pharmacokinetics of compounds after subcutaneous administration, structure and function of the gastrointestinal tract, morphology of the pancreas, and the overall metabolic status of the two species. Because spontaneous type 1-like diabetes is very rare in pigs, a model of the condition must be induced experimentally, either surgically or chemically. This process is discussed, and the use of the pig as a model in islet transplantation and diabetic complications is briefly summarized.


Subject(s)
Diabetes Mellitus, Type 1/etiology , Disease Models, Animal , Pancreatectomy , Streptozocin , Swine, Miniature , Animals , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Swine , Swine, Miniature/physiology
20.
Eur J Pharmacol ; 494(2-3): 283-8, 2004 Jun 28.
Article in English | MEDLINE | ID: mdl-15212985

ABSTRACT

Glucagon-like peptide-1 (GLP-1), a future treatment for type 2 diabetes, is efficiently degraded by the enzyme dipeptidyl peptidase IV (DPP IV), yielding the major metabolite GLP-1-(9-36)-amide. In this study, we examined the potential glucose lowering effect of GLP-1-(9-36)-amide in mice and found that GLP-1-(9-36)-amide (3 and 10 nmol/kg) did not affect insulin secretion or glucose elimination when administered intravenously together with glucose (1 g/kg). This was observed both in normal mice and in transgenic mice having a complete disruption of the signalling from the GLP-1 receptor. Furthermore, after blocking insulin secretion, using diazoxide (25 mg/kg), no effect on insulin-independent glucose disposal of GLP-1-(9-36)-amide was observed. Therefore, GLP-1-(9-36)-amide does not affect glucose disposal in mice either in the presence or absence of intact GLP-1-receptors or in the presence or absence of stimulated insulin levels. This suggests that the GLP-1 metabolite is not involved in the regulation of glucose homeostasis.


Subject(s)
Blood Glucose/metabolism , Hypoglycemic Agents/pharmacology , Insulin/blood , Peptides/pharmacology , Animals , Diazoxide/pharmacology , Female , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/administration & dosage , Injections, Intravenous , Insulin/physiology , Islets of Langerhans/drug effects , Mice , Mice, Transgenic , Peptides/administration & dosage , Potassium Channels/agonists , Receptors, Glucagon/drug effects , Receptors, Glucagon/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...