Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Pharmacol Ther ; 195: 100-110, 2019 03.
Article in English | MEDLINE | ID: mdl-30347209

ABSTRACT

12-lipoxygenase (12-LOX) is one of several enzyme isoforms responsible for the metabolism of arachidonic acid and other poly-unsaturated fatty acids to both pro- and anti-inflammatory lipid mediators. Mounting evidence has shown that 12-LOX plays a critical role in the modulation of inflammation at multiple checkpoints during diabetes development. Due to this, interventions to limit pro-inflammatory 12-LOX metabolites either by isoform-specific 12-LOX inhibition, or by providing specific fatty acid substrates via dietary intervention, has the potential to significantly and positively impact health outcomes of patients living with both type 1 and type 2 diabetes. To date, the development of truly specific and efficacious inhibitors has been hampered by homology of LOX family members; however, improvements in high throughput screening have improved the inhibitor landscape. Here, we describe the function and role of human 12-LOX, and mouse 12-LOX and 12/15-LOX, in the development of diabetes and diabetes-related complications, and describe promise in the development of strategies to limit pro-inflammatory metabolites, primarily via new small molecule 12-LOX inhibitors.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Diabetes Complications/enzymology , Diabetes Mellitus, Type 1/enzymology , Diabetes Mellitus, Type 2/enzymology , Animals , Arachidonate 15-Lipoxygenase/metabolism , Humans , Insulin-Secreting Cells/enzymology , Lipoxygenase Inhibitors/pharmacology , Signal Transduction
2.
J Endocrinol ; 227(1): 13-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26285907

ABSTRACT

Adipose tissue (AT) inflammation is an emerging factor contributing to cardiovascular disease. STAT4 is a transcription factor expressed in adipocytes and in immune cells and contributes to AT inflammation and insulin resistance in obesity. The objective of this study was to determine the effect of STAT4 deficiency on visceral and peri-aortic AT inflammation in a model of atherosclerosis without obesity. Stat4(-/-)Apoe(-/-) mice and Apoe(-/-) controls were kept either on chow or Western diet for 12 weeks. Visceral and peri-aortic AT were collected and analyzed for immune composition by flow cytometry and for cytokine/chemokine expression by real-time PCR. Stat4(-/-)Apoe(-/-) and Apoe(-/-) mice had similar body weight, plasma glucose, and lipids. Western diet significantly increased macrophage, CD4+, CD8+, and NK cells in peri-aortic and visceral fat in Apoe(-/-) mice. In contrast, in Stat4(-/-)Apoe(-/-) mice, a Western diet failed to increase the percentage of immune cells infiltrating the AT. Also, IL12p40, TNFa, CCL5, CXCL10, and CX3CL1 were significantly reduced in the peri-aortic fat in Stat4(-/-)Apoe(-/-) mice. Importantly, Stat4(-/-)Apoe(-/-) mice on a Western diet had significantly reduced plaque burden vs Apoe(-/-) controls. In conclusion, STAT4 deletion reduces inflammation in peri-vascular and visceral AT and this may contribute via direct or indirect effects to reduced atheroma formation.


Subject(s)
Atherosclerosis/metabolism , Intra-Abdominal Fat/metabolism , Macrophages/metabolism , Panniculitis/metabolism , STAT4 Transcription Factor/metabolism , Animals , Aorta , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Atherosclerosis/etiology , Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Polarity , Cells, Cultured , Chemokines/metabolism , Cytokines/metabolism , Diet, Western/adverse effects , Female , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/pathology , Macrophages/immunology , Macrophages/pathology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Mice, Knockout , Ovary , Panniculitis/etiology , Panniculitis/immunology , Panniculitis/pathology , Random Allocation , STAT4 Transcription Factor/genetics , Specific Pathogen-Free Organisms
3.
Diabetes Obes Metab ; 15 Suppl 3: 117-29, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24003928

ABSTRACT

Inflammation is an established pathogenic player in insulin resistance, islet demise and atherosclerosis. The complex interactions between cytokines, immune cells and affected tissues result in sustained inflammation in diabetes and atherosclerosis. 12- and 15-lipoxygenase (LO), such as 12/15-LO, produces a variety of metabolites through peroxidation of fatty acids and potentially contributes to the complex molecular crosstalk at the site of inflammation. 12- and 15-LO pathways are frequently activated in tissues affected by diabetes and atherosclerosis including adipose tissue (AT), islets and the vasculature. Moreover, mice with whole body and tissue-specific knockout of 12/15-LO are protected against insulin resistance, hyperglycaemia and atherosclerosis supporting functional contribution of 12- and 15-LO pathways in diabetes and atherosclerosis. Recently, it has emerged that there is a temporal regulation of the particular isoforms of 12- and 15-LO in human AT and islets during the development of type 1 and type 2 diabetes and obesity. Analyses of tissues affected by diabetes and atherosclerosis also implied the roles of interleukin (IL)-12 and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase-1 (NOX-1) in islets and IL-17A in atherosclerosis. Future studies should aim to test the efficacy of inhibitions of these mediators for treatment of diabetes and atherosclerosis.


Subject(s)
Cytokines/physiology , Inflammation/physiopathology , Insulin Resistance/physiology , Islets of Langerhans/physiopathology , Vascular Diseases/physiopathology , Adipose Tissue/physiology , Animals , Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/physiology , Humans , Inflammation Mediators/physiology , Mice
4.
Am J Physiol Endocrinol Metab ; 300(2): E410-21, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21081706

ABSTRACT

Adipose tissue inflammation and reduced pancreatic ß-cell function are key issues in the development of cardiovascular disease and progressive metabolic dysfunction in type 2 diabetes mellitus. The aim of this study was to determine the effect of the DPP IV inhibitor sitagliptin on adipose tissue and pancreatic islet inflammation in a diet-induced obesity model. C57Bl/6J mice were placed on a high-fat (60% kcal fat) diet for 12 wk, with or without sitagliptin (4 g/kg) as a food admix. Sitagliptin significantly reduced fasting blood glucose by 21% as well as insulin by ∼25%. Sitagliptin treatment reduced body weight without changes in overall body mass index or in the epididymal and retroperitoneal fat mass. However, sitagliptin treatment led to triple the number of small adipocytes despite reducing the number of the very large adipocytes. Sitagliptin significantly reduced inflammation in the adipose tissue and pancreatic islet. Macrophage infiltration in adipose tissue evaluated by immunostaining for Mac2 was reduced by sitagliptin (P < 0.01), as was the percentage of CD11b+/F4/80+ cells in the stromal vascular fraction (P < 0.02). Sitagliptin also reduced adipocyte mRNA expression of inflammatory genes, including IL-6, TNFα, IL-12(p35), and IL-12(p40), 2.5- to fivefold as well as 12-lipoxygenase protein expression. Pancreatic islets were isolated from animals after treatments. Sitagliptin significantly reduced mRNA expression of the following inflammatory cytokines: MCP-1 (3.3-fold), IL-6 (2-fold), IL-12(p40) (2.2-fold), IL-12(p35) (5-fold, P < 0.01), and IP-10 (2-fold). Collectively, the results indicate that sitagliptin has anti-inflammatory effects in adipose tissue and in pancreatic islets that accompany the insulinotropic effect.


Subject(s)
Adipose Tissue/pathology , Anti-Inflammatory Agents , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Inflammation/pathology , Islets of Langerhans/pathology , Pyrazines/pharmacology , Triazoles/pharmacology , Adipocytes/pathology , Adipocytes/ultrastructure , Adiposity/drug effects , Animals , Body Weight/drug effects , Cytokines/metabolism , Flow Cytometry , Glucose/metabolism , Glucose/pharmacology , Glucose Intolerance/metabolism , Glucose Tolerance Test , Immunohistochemistry , Insulin/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Neutrophil Infiltration/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Sitagliptin Phosphate
5.
Hypertension ; 38(3): 361-6, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11566905

ABSTRACT

One-kidney, 1-clip rats (1K1C) or uninephrectomized controls were treated with either the superoxide dismutase mimetic tempol (0.5 mmol. kg(-1). d(-1)), angiotension type 1 receptor inhibitor losartan (50 mmol. L(-1). kg(-1). d(-1)), or both (n=6 per group) for 2 weeks. At the end of the study, systolic blood pressure (BP) decreased on average by 21% in tempol-treated and 29% in losartan-treated versus untreated 1K1C (217+/-4.4 mm Hg) and was normalized in the losartan plus tempol group. Mean BP also decreased from 159+/-3.7 mm Hg in 1K1C to 93+/-2.8 mm Hg in the losartan plus tempol group. Also, aortic wall area was reduced by 18% in losartan- or tempol-treated 1K1C and by 30% in losartan plus tempol rats compared with untreated 1K1C. Plasma renin activity was increased from 4.8+/-0.3 in untreated 1K1C to 15.9+/-0.9 ng. mL(-1). h(-1) in losartan-treated but not tempol-treated 1K1C. Superoxide generation by the isolated aortic rings assessed by lucigenin chemiluminescence was significantly decreased (by approximately 40%) in all losartan, tempol, and losartan plus tempol groups compared with untreated 1K1C. Nitrotyrosine ELISA in the kidney displayed a significant reduction, from 59+/-13 ng/mg of protein in 1K1C to 12.5+/-5 ng/mg of protein in the losartan plus tempol 1K1C. Western blotting for nNOS in kidney cortex and medulla showed a protein increase in both fractions of 1K1C versus controls and was normalized by losartan plus tempol treatment. Collectively, data show a synergistic effect of losartan and tempol on BP reduction in 1K1C rats. The mechanism may involve reduced superoxide production and nitrotyrosine formation in kidney and decreased kidney neuronal-type NO synthase expression in treated animals. This status in the oxidative balance seems to affect BP in the renal hypertensive rats.


Subject(s)
Angiotensin II/physiology , Blood Pressure/physiology , Free Radicals/metabolism , Hypertension, Renovascular/physiopathology , Tyrosine/analogs & derivatives , Angiotensin Receptor Antagonists , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Blood Pressure/drug effects , Blotting, Western , Cyclic N-Oxides/pharmacology , Disease Models, Animal , Free Radical Scavengers/pharmacology , Free Radicals/antagonists & inhibitors , Hypertension, Renovascular/metabolism , Losartan/pharmacology , Male , Nephrectomy , Nitric Oxide Synthase/drug effects , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type I , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Renal Artery/drug effects , Renal Artery/pathology , Renin/blood , Renin/drug effects , Spin Labels , Superoxides/metabolism , Systole , Time Factors , Tyrosine/drug effects , Tyrosine/metabolism
6.
Hypertension ; 37(2 Pt 2): 554-60, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230334

ABSTRACT

The mechanisms underlying the development of hypertension in obesity are not yet fully understood. We recently reported the development of hypertension in a rat model of diet-induced obesity. When Sprague-Dawley rats (n=60) are fed a moderately high fat diet (32 kcal% fat) for 10 to 16 weeks, approximately half of them develop obesity (obesity-prone [OP] group) and mild hypertension (158+/-3.4 mm Hg systolic pressure), whereas the other half (obesity-resistant [OR] group) maintains a body weight equivalent to that of a low fat control group and is normotensive (135.8+/-3.8 mm Hg). We examined the potential role of oxidative stress in the development of hypertension in this model. Lipid peroxides measured as thiobarbituric acid-reactive substances showed a significant increase in the LDL fraction of OP rats (2.8+/-0.32 nmol malondialdehyde/mg protein) compared with OR and control rats (0.9+/-0.3 nmol malondialdehyde/mg protein). Also, aortic and kidney thiobarbituric acid-reactive substances showed a significant (3- and 5- fold) increase in OP rats after 16 weeks of diet. In addition, superoxide generation by aortic rings, measured by lucigenin luminescence, showed a 2-fold increase in the OP group compared with both the OR and control groups. In addition, free isoprostane excretion and nitrotyrosine in the kidney showed an increase in OP rats only. The urine and plasma nitrate/nitrite measured by the LDH method showed a 1.8-fold decrease in OP rats compared with OR rats. However, endothelial NO synthase expression in the kidney cortex and medulla assessed by reverse transcriptase-polymerase chain reaction showed a strong increase in the OP rats versus OR and control rats (endothelial NO synthase/beta-actin ratio 1.3+/-0.04 in OP rats versus 0.44+/-0.02 in OR rats), suggesting a possible shift toward superoxide production by the enzyme. Collectively, the data show a decreased NO bioavailability in OP animals that is due in part to the increased oxidative stress.


Subject(s)
Hypertension/etiology , Obesity/complications , Oxidative Stress , Animals , Aorta, Thoracic/metabolism , Blood Pressure , Body Weight , Dietary Fats/administration & dosage , Dinoprost/analogs & derivatives , Dinoprost/urine , Disease Models, Animal , F2-Isoprostanes , Hyperlipidemias/etiology , Hypertension/blood , Hypertension/urine , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Lipid Peroxides/analysis , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase/analysis , Nitric Oxide Synthase Type III , Obesity/blood , Obesity/urine , Rats , Rats, Sprague-Dawley , Renin/blood , Thiobarbituric Acid Reactive Substances/analysis
7.
Hypertension ; 37(3): 955-60, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11244024

ABSTRACT

Chronic hypertension is associated with remodeling of small arteries. There is evidence that the high pressure itself may cause these structural changes, but the responsible mechanisms are not clearly defined. Previously we showed that pressure-induced c-fos expression in intact cannulated rat mesenteric small arteries was inhibited by genistein, a general tyrosine kinase inhibitor. The purpose of this study was to further unravel the underlying signal transduction mechanisms, and we particularly tested the involvement of src tyrosine kinases and extracellular signal-regulated kinase (ERK). Rat mesenteric small arteries were cannulated in a dual-vessel chamber. After a 60-minute equilibration period, the pressure in 1 artery was increased to 140 mm Hg, while the other artery remained at 90 mm Hg. Semiquantitative reverse transcriptase-polymerase chain reaction was used to determine c-fos expression, and Western blotting was used to examine levels of ERK phosphorylation. The involvement of src and ERK was tested with the inhibitors herbimycin A (1 micromol/L), PP1 (10 micromol/L), PP2 (10 micromol/L), and PD98059 (30 micromol/L). One-hour exposure to 140 mm Hg increased the c-fos/cyclophilin ratio 3.6-fold, from 0.29+/-0.07 to 1.06+/-0.25. All the tested inhibitors suppressed the pressure-induced increase of c-fos expression. A 5-minute exposure period to 140 mm Hg increased ERK phosphorylation, and this was abolished in the presence of PP1. The results suggest that pressure-induced c-fos expression in intact cannulated rat mesenteric small arteries may be mediated, at least in part, by src tyrosine kinases and ERK.


Subject(s)
Mesenteric Arteries/metabolism , Mitogen-Activated Protein Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Animals , Arterioles/anatomy & histology , Arterioles/metabolism , Benzoquinones , CSK Tyrosine-Protein Kinase , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Lactams, Macrocyclic , Male , Mesenteric Arteries/anatomy & histology , Mesenteric Arteries/drug effects , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Phosphorylation , Pressure , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinones/pharmacology , Rats , Rats, Wistar , Rifabutin/analogs & derivatives , src Homology Domains , src-Family Kinases
8.
Atherosclerosis ; 153(1): 191-201, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11058715

ABSTRACT

Non-insulin-dependent diabetes mellitus (NIDDM) is frequently associated with macroangiopathies and coronary heart diseases. Lipoprotein lipase (LPL), an enzyme known to undergo significant functional alterations in diabetic state, is also a potential atherogenic protein. Since, to the best of our knowledge, there are no data concerning LPL secreted by macrophages of NIDDM patients we conducted a study to assess the expression and activity of LPL secreted by monocyte-derived macrophages from NIDDM patients with cardiovascular complications versus cardiovascular patients without diabetes (controls). Isolated cells from NIDDM patients, after 7 days in culture in the presence of 20% autologous serum, readily exhibit a foam cell phenotype, in contrast to the cells from controls. Macrophages were mainly loaded with triglycerides, whose cellular amount was well correlated to triglyceridemia of NIDDM subjects. Concomitantly, macrophages from NIDDM patients displayed a approximately six-fold decrease of mRNA expression and a approximately two-fold reduction of the activity of secreted LPL, as compared to control cells. These data suggest that in complicated diabetic state, macrophage loading leading to foam cell formation is accelerated, at least in part, due to a diminished expression and activity of LPL. These observations add and extend the data that may explain the occurrence of accelerated atherogenesis and of the atherosclerotic complications associated with diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Lipid Metabolism , Lipoprotein Lipase/metabolism , Macrophages/metabolism , Adult , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell Line , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/metabolism , Diabetic Angiopathies/pathology , Female , Foam Cells/pathology , Humans , Lipoprotein Lipase/genetics , Macrophages/pathology , Male , Microscopy, Electron , Monocytes/pathology , RNA, Messenger/metabolism
9.
Hypertension ; 35(4): 1009-15, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10775577

ABSTRACT

Although obesity is a risk factor for hypertension, the relationship between these 2 conditions is not well understood. Therefore, we examined some parameters of hypertension and cardiovascular disease in a dietary model of obesity. Male Sprague-Dawley rats were provided either a control diet (C) or a diet containing 32% kcal as fat (similar to a Western diet) for 1, 3, or 10 weeks. Rats in the latter group diverged based on body weight gain into obesity-prone (OP) and obesity-resistant (OR) groups. Systolic blood pressure in OP rats was significantly higher after 10 weeks of the diet (149+/-4. 8 mm Hg) compared with both OR and C groups (131+/-3.7 and 129+/-4.5 mm Hg, respectively). The aortic wall area of OP rats was significantly increased, indicating arterial hypertrophy, and a 2-fold increase in plasma renin activity was found in OP rats compared with OR and C rats. The lipid profile showed a significant increase in plasma and VLDL triglycerides of OP versus OR and C groups as early as 3 weeks on the diet. Plasma and LDL-cholesterol levels were increased in the OP group versus the OR and C groups after 3 weeks of the diet, but the difference was blunted after 10 weeks. Lipid peroxidation (thiobarbituric acid-reactive substances) in OP rats was increased 2-fold in LDL and 1.5-fold in aortic wall compared with OR rats, suggesting an increased oxidative stress in these animals. Periodic acid-Schiff staining of the kidney showed mesangial expansion and focal sclerosis that were more prominent in OP rats than in OR rats. The results suggest that hypercholesterolemia, but not hypertriglyceridemia, is linked to the diet; that hypertension and renin-angiotensin system activation are associated with obesity; and that lipid peroxidation and renal damage are the results of both factors.


Subject(s)
Blood Pressure , Dietary Fats/adverse effects , Hypertension/etiology , Obesity/complications , Animals , Cholesterol, LDL/blood , Dietary Fats/administration & dosage , Hypertension/blood , Male , Rats , Rats, Sprague-Dawley , Triglycerides/blood
10.
Am J Physiol Heart Circ Physiol ; 278(2): H613-22, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10666094

ABSTRACT

To separate the role of ANG II from pressure in hypertrophy of the vascular wall in one-kidney, one-clip (1K1C) hypertension, experimental and sham-operated rats were given the AT(1)-receptor antagonist losartan (20 mg x kg(-1) x day(-1)) or tap water for 14 days. Mean arterial pressure was elevated in both experimental groups compared with controls. Rats were anesthetized with pentobarbital sodium, and the thoracic aorta and carotid, small mesenteric, and external spermatic arteries were harvested and embedded in paraffin. Tissue sections were used for morphological analysis, immunohistochemistry for 5-bromo-2'-deoxyuridine (BrdU) and platelet-derived growth factor (PDGF)-AA, stereological measurements, and in situ hybridization with a (35)S-labeled riboprobe for PDGF-A mRNA. Elevated cross-sectional areas of thoracic, carotid, and small mesenteric artery in 1K1C rats were not reduced by losartan. The internal diameter of the external spermatic artery and microvascular density of the cremaster muscle were reduced in 1K1C rats. The number of BrdU-positive nuclei per cross section did not differ between 1K1C and control arteries. PDGF-A mRNA was elevated in the arterial walls of 1K1C rats compared with controls and was hardly changed by losartan. PDGF-A protein stained strongly in the media of 1K1C arteries and was not inhibited by losartan; it appeared in the adventitia of all aortas and carotid arteries. These observations demonstrate that effects of ANG II mediated through the AT(1) receptor are not necessary for hypertrophy of the vascular wall during 1K1C hypertension or expression of PDGF-A.


Subject(s)
Angiotensin Receptor Antagonists , Arteries/metabolism , Arteries/pathology , Hypertension, Renal/metabolism , Hypertension, Renal/pathology , Platelet-Derived Growth Factor/metabolism , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Blood Pressure , Bromodeoxyuridine/pharmacokinetics , Capillaries/pathology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Hypertension, Renal/physiopathology , Hypertrophy , Losartan/pharmacology , Male , Mesenteric Arteries/metabolism , Mesenteric Arteries/pathology , Platelet-Derived Growth Factor/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1 , Receptor, Angiotensin, Type 2 , Reference Values
SELECTION OF CITATIONS
SEARCH DETAIL
...