Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Heliyon ; 10(11): e31799, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38882290

ABSTRACT

Endothelial dysfunction and inflammation are clinically significant risk factors for cardiovascular diseases in hypertension. Although immune cells play a role in hypertension, the impact of plasmacytoid dendritic cells in established renovascular hypertension-induced cardiovascular complications is not fully understood. We investigated plasmacytoid dendritic cells' contribution to arterial endothelial dysfunction and inflammation in renovascular hypertension. A two-kidney one-clip (2K1C) model for four weeks in both male and female mice was used to induce renovascular hypertension. We treated mice with or without anti-PDCA-1 antibodies for one week to deplete the plasmacytoid dendritic cells. Renovascular hypertension causes cardiac hypertrophy, lung edema, and microvascular endothelial dysfunction associated with inflammation induction in mice. Moreover, renovascular hypertension affects the profile of immune cells, including dendritic cells and macrophages, with variations between male and female mice. Interestingly, the depletion of plasmacytoid dendritic cells significantly reduces blood pressure, cardiac hypertrophy, lung edema, inflammation, and oxidative stress and improves microvascular endothelial function via the endoplasmic reticulum (ER) stress, autophagy, and mTOR-dependent mechanisms. Plasmacytoid dendritic cells significantly contribute to the development of cardiovascular complications in renovascular hypertension by modulating immune cells, inflammation, oxidative stress, and ER stress.

2.
J Am Heart Assoc ; 12(14): e029668, 2023 07 18.
Article in English | MEDLINE | ID: mdl-37345828

ABSTRACT

Background Heart failure with preserved ejection fraction (HFpEF) is a significant unmet need in cardiovascular medicine and remains an untreatable cardiovascular disease. The role and mechanism of interleukin-1ß in HFpEF pathogenesis are poorly understood. Methods and Results C57/Bl6J and interleukin-1ß-/- male mice were randomly divided into 4 groups. Groups 1 and 2: C57/Bl6J and interleukin-1ß-/- mice were fed a regular diet for 4 months and considered controls. Groups 3 and 4: C57/Bl6 and interleukin-1ß-/- mice were fed a high-fat diet with N[w]-nitro-l-arginine methyl ester (endothelial nitric oxide synthase inhibitor, 0.5 g/L) in the drinking water for 4 months. We measured body weight, blood pressure, diabetes status, cardiac function/hypertrophy/inflammation, fibrosis, vascular endothelial function, and signaling. C57/Bl6 fed a high-fat diet and N[w]-nitro-l-arginine methyl ester in the drinking water for 4 months developed HFpEF pathogenesis characterized by obesity, diabetes, hypertension, cardiac hypertrophy, lung edema, low running performance, macrovascular and microvascular endothelial dysfunction, and diastolic cardiac dysfunction but no change in cardiac ejection fraction compared with control mice. Interestingly, the genetic disruption of interleukin-1ß protected mice from HFpEF pathogenesis through the modulation of the inflammation and endoplasmic reticulum stress mechanisms. Conclusions Our data suggest that interleukin-1ß is a critical driver in the development of HFpEF pathogenesis, likely through regulating inflammation and endoplasmic reticulum stress pathways. Our findings provide a potential therapeutic target for HFpEF treatment.


Subject(s)
Cardiomyopathies , Drinking Water , Heart Failure , Mice , Male , Animals , Heart Failure/genetics , Heart Failure/prevention & control , Stroke Volume/physiology , Interleukin-1beta , Cardiomyopathies/complications , Inflammation/pathology
3.
Diabetes Metab Syndr Obes ; 15: 2633-2642, 2022.
Article in English | MEDLINE | ID: mdl-36065460

ABSTRACT

Purpose: Vascular endothelial dysfunction is well established in type 2 diabetes. Interleukin-12 (IL-12) and endoplasmic reticulum (ER) stress are up-regulated in type 2 diabetic patients and animal models of type 2 diabetes. However, the role and underlying mechanisms of IL-12 and the ER stress CHOP in endothelial dysfunction are not fully understood. Methods: We generated double knockout mice between db-/db- and p40IL-12-/- mice (db-/db-p40-IL-12-/-) and endoplasmic (ER) stress-CHOP-/- mice (db-/db-CHOP-/-). We performed a glucose tolerance test (GTT) to determine the effect of IL-12 and ER stress CHOP on glucose metabolism. We assessed the endothelial function and determined the phosphorylation level of eNOS, Akt, AMPK, and the expression of ER stress (CHOP, BIP), and oxidative stress (Nox2 and Nox4 and NADPH oxidase activity). Results: The results showed that GTT was improved in db-/db-p40-IL-12-/- and db-/db-CHOP-/- suggesting IL-12 and CHOP as parts of a mechanism involved in the development of type 2 diabetes. The microvascular endothelial dysfunction in db-/db- mouse is associated with decreased phosphorylated eNOS, Akt, AMPK, and increased CHOP, BIP, Nox2, and Nox4 expressions. Interestingly, disrupting IL-12 and ER stress CHOP in db-/db- mice significantly improved endothelial function, increased survival markers expression and decreased ER and oxidative stress. Conclusion: Using a genetic approach, these findings provide evidence that IL-12 and ER stress CHOP play a significant role in microvascular endothelial dysfunction in type 2 diabetes.

4.
Inflammation ; 45(5): 1864-1874, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35386038

ABSTRACT

High Mobility Group Box 1 (HMGB1) is a ubiquitous, highly conserved nuclear and cytosolic protein that has diverse biological roles depending on its cellular location and posttranslational modifications. The HMGB1 is localized in the nucleus but can be translocated to the cytoplasm to modulate the intracellular signaling and eventually secreted outside the cells. It is widely established that HMGB1 plays a key role in inflammation; however, the role of HMGB1 in the cardiovascular diseases is not well understood. In this review, we will discuss the latest reports on the pathophysiological link between HMGB1 and cardiovascular complications, with special emphasis on the inflammation. Thus, the understanding of the role of HMGB1 may provide new insights into developing new HMGB1-based therapies.


Subject(s)
Cardiovascular Diseases , HMGB1 Protein , Cell Nucleus/metabolism , Cytoplasm/metabolism , HMGB1 Protein/metabolism , Humans , Inflammation/metabolism
7.
World J Cardiol ; 11(6): 159-170, 2019 Jun 26.
Article in English | MEDLINE | ID: mdl-31367278

ABSTRACT

Cardiovascular diseases are the number one cause of morbidity and mortality in the United States and worldwide. The induction of the endoplasmic reticulum (ER) stress, a result of a disruption in the ER homeostasis, was found to be highly associated with cardiovascular diseases such as hypertension, diabetes, ischemic heart diseases and heart failure. This review will discuss the latest literature on the different aspects of the involvement of the ER stress in cardiovascular complications and the potential of targeting the ER stress pathways as a new therapeutic approach for cardiovascular complications.

8.
Biochim Biophys Acta Mol Basis Dis ; 1865(2): 403-412, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30414897

ABSTRACT

BACKGROUND: Microvascular dysfunction is a major complication in hypertensive patients. We previously reported that CD4+CD25+ T regulatory cells (Treg) play an important preventive role in hypertension-induced vascular dysfunction. However, whether Treg cells therapy and autophagy inhibition could rescue Treg cells survival and microvascular function in established hypertension is an important question that remained unanswered. METHODS & RESULTS: Here we showed that Treg cells from mice model of established hypertension displayed an enhanced apoptotic rate, which was rescued with Treg cells transfer and autophagy inhibition. We also showed increased autophagy in mesenteric resistance artery (MRA) in mice with established hypertension. Importantly, the inhibition of autophagy or one single transfer of Treg cells into mice with established hypertension improved the microvascular function independently of high blood pressure. The protection involves the modulation of interleukin-10 (IL-10), inflammation, endoplasmic reticulum (ER) stress, oxidative stress, Akt, and eNOS. CONCLUSIONS: The present study suggests that Treg cells survival is regulated by autophagy. Also, Treg cells as a cellular therapy aimed at rescuing the microvascular function through an autophagy-dependent mechanism and independently of arterial blood pressure lowering effects. Because our mouse model of established hypertension mimics the clinical situation, our results have the potential for new therapeutic approaches that involve the manipulation of Treg cells and autophagy to overcome established hypertension-induced cardiovascular complications.


Subject(s)
Hypertension/immunology , Hypertension/physiopathology , Lymphocyte Depletion , Microvessels/physiopathology , T-Lymphocytes, Regulatory/immunology , Animals , Arterial Pressure , Biomarkers/metabolism , Lymphocyte Count , Mice, Inbred C57BL , Models, Biological , NADPH Oxidases/metabolism , Oxidative Stress , Phosphorylation , Systole , Vascular Resistance
9.
J Hypertens ; 36(2): 377-386, 2018 02.
Article in English | MEDLINE | ID: mdl-29611835

ABSTRACT

OBJECTIVES: Stromal interacting molecule-1 (STIM1) plays a role in coordinating calcium signaling in different cell types. The increase or deletion of STIM1 expression in cardiomyocyte causes cardiac complication. Moreover, the deletion of STIM1 in endothelial cell causes vascular endothelial dysfunction. However, the disruption of STIM1 in smooth muscle cells (SMC) has no effect on endothelial function but protects vascular function when mice are infused with angiotensin-II. Nevertheless, the role of SMC-STIM1 in acute and chronic myocardial infarction (MI) induced by acute ischemia-reperfusion injury and permanent coronary artery occlusion is unknown. METHODS AND RESULTS: Stim1 were generated and crossed into the SM22α-Cre backgrounds. SM22α-Cre causes deletion of STIM1 floxed genes in adult SMC (Stim1). Control and Stim1 mice were subjected to acute ischemia-reperfusion injury. Hearts were then harvested and incubated with triphenyltetrazolium chloride to determine the infarct size. In control mice which are subjected to ischemia-reperfusion, the heart developed a significant infarct associated with an increase in STIM1 expression. Interestingly, the infarct size was substantially reduced in Stim1 mice. The protection in Stim1 mice against ischemia-reperfusion injury involves the modulation of endoplasmic reticulum stress, apoptosis, oxidative stress, protein kinase B, and mitogen-activated protein (MAP) kinase (ERK1/2 and p38) signaling, and inflammation. Furthermore, in another model of chronic MI induced by permanent coronary artery occlusion, SMC-STIM1 disruption significantly reduced myocardial infarct size and improved cardiac function. CONCLUSION: Our results provide new evidence that SMC-STIM1 disruption is a novel mechanism that protects the heart from MI through reduction of endoplasmic reticulum stress, oxidative stress, MAP-Kinase, apoptosis, and inflammation.


Subject(s)
Myocardial Infarction/etiology , Myocardial Infarction/genetics , Myocytes, Smooth Muscle/metabolism , Reperfusion Injury/complications , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Animals , Apoptosis , Coronary Vessels/surgery , Endoplasmic Reticulum Stress , Ligation , MAP Kinase Signaling System , Male , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/complications , Myocytes, Cardiac , Oxidative Stress , Protective Factors , Proto-Oncogene Proteins c-akt/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Pflugers Arch ; 470(3): 471-480, 2018 03.
Article in English | MEDLINE | ID: mdl-29288332

ABSTRACT

We previously reported that EGFR tyrosine kinase (EGFRtk) activity and endoplasmic reticulum (ER) stress are enhanced in type 2 diabetic (T2D) mice and cause vascular dysfunction. In the present study, we determined the in vivo contribution of EGFRtk and ER stress in acute myocardial infarction induced by acute ischemia (40 min)-reperfusion (24 h) (I/R) injury in T2D (db-/db-) mice. We treated db-/db- mice with EGFRtk inhibitor (AG1478, 10 mg/kg/day) for 2 weeks. Mice were then subjected to myocardial I/R injury. The db-/db- mice developed a significant infarct after I/R injury. The inhibition of EGFRtk significantly reduced the infarct size and ER stress induction. We also determined that the inhibition of ER stress (tauroursodeoxycholic acid, TUDCA, 150 mg/kg per day) in db-/db- significantly decrease the infarct size indicating that ER stress is a downstream mechanism to EGFRtk. Moreover, AG1478 and TUDCA reduced myocardium p38 and ERK1/2 MAP-kinases activity, and increased the activity of the pro-survival signaling cascade Akt. Additionally, the inhibition of EGFRtk and ER stress reduced cell apoptosis and the inflammation as indicated by the reduction in macrophages and neutrophil infiltration. We determined for the first time that the inhibition of EGFRtk protects T2D heart against I/R injury through ER stress-dependent mechanism. The cardioprotective effect of EGFRtk and ER stress inhibition involves the activation of survival pathway, and inhibition of apoptosis, and inflammation. Thus, targeting EGFRtk and ER stress has the potential for therapy to overcome myocardial infarction in T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Endoplasmic Reticulum Stress , ErbB Receptors/metabolism , Myocardial Infarction/metabolism , Animals , Apoptosis , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetic Cardiomyopathies/drug therapy , ErbB Receptors/antagonists & inhibitors , MAP Kinase Signaling System , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Taurochenodeoxycholic Acid/pharmacology , Taurochenodeoxycholic Acid/therapeutic use , Tyrphostins/pharmacology , Tyrphostins/therapeutic use
11.
Am J Pathol ; 187(11): 2590-2601, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28837799

ABSTRACT

Recently, IL-12 emerged as a critical player in type 2 diabetes complications. We previously reported that ischemia-induced angiogenesis is compromised in type 2 diabetic mice. In this study, we determined that IL-12 disruption rescued angiogenesis and arteriogenesis in type 2 diabetic mice. To induce type 2 diabetes, wild-type (WT), p40IL-12-/- (p40-/-), and p35IL-12-/- (p35-/-) mice were fed a high-fat diet (HFD) for 12 weeks. Body weight, glucose test tolerance, and insulin test tolerance were assessed. After 12 weeks of an HFD, the femoral artery was ligated and blood flow recovery was measured every week for 4 weeks. WT, p40-/-, and p35-/- mice fed an HFD become obese after 12 weeks and exhibit glucose intolerance and insulin resistance. Blood flow recovery was fully restored in 2 to 3 weeks after femoral artery ligation in all groups of mice fed a normal diet. However, after 12 weeks of an HFD, blood flow recovery was compromised in WT mice, whereas it was fully recovered in p40-/- and p35-/- mice. The mechanism of blood flow recovery involves an increase in capillary/arteriole density, endothelial nitric oxide synthase/Akt/vascular endothelial growth factor receptor 2 signaling, and a reduction in oxidative stress and inflammation. The disruption of IL-12 promotes angiogenesis and increases blood flow recovery in obese type 2 diabetic mice by an endothelial nitric oxide synthase/Akt/vascular endothelial growth factor receptor 2/oxidative stress-inflammation-dependent mechanism.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelium, Vascular/metabolism , Interleukin-12/metabolism , Neovascularization, Pathologic/metabolism , Animals , Diet, High-Fat , Endothelium, Vascular/pathology , Insulin Resistance/physiology , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type III/metabolism , Obesity/complications , Obesity/metabolism , Oxidative Stress
13.
Arterioscler Thromb Vasc Biol ; 36(9): 1900-9, 2016 09.
Article in English | MEDLINE | ID: mdl-27470514

ABSTRACT

OBJECTIVES: Chronic hypertension is the most critical risk factor for cardiovascular disease, heart failure, and stroke. APPROACH AND RESULTS: Here we show that wild-type mice infused with angiotensin II develop hypertension, cardiac hypertrophy, perivascular fibrosis, and endothelial dysfunction with enhanced stromal interaction molecule 1 (STIM1) expression in heart and vessels. All these pathologies were significantly blunted in mice lacking STIM1 specifically in smooth muscle (Stim1(SMC-/-)). Mechanistically, STIM1 upregulation during angiotensin II-induced hypertension was associated with enhanced endoplasmic reticulum stress, and smooth muscle STIM1 was required for endoplasmic reticulum stress-induced vascular dysfunction through transforming growth factor-ß and nicotinamide adenine dinucleotide phosphate oxidase-dependent pathways. Accordingly, knockout mice for the endoplasmic reticulum stress proapoptotic transcriptional factor, CCAAT-enhancer-binding protein homologous protein (CHOP(-/-)), were resistant to hypertension-induced cardiovascular pathologies. Wild-type mice infused with angiotensin II, but not Stim1(SMC-/-) or CHOP(-/-) mice showed elevated vascular nicotinamide adenine dinucleotide phosphate oxidase activity and reduced phosphorylated endothelial nitric oxide synthase, cGMP, and nitrite levels. CONCLUSIONS: Thus, smooth muscle STIM1 plays a crucial role in the development of hypertension and associated cardiovascular pathologies and represents a promising target for cardiovascular therapy.


Subject(s)
Blood Pressure , Cardiomegaly/metabolism , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Stromal Interaction Molecule 1/metabolism , Vasodilation , Angiotensin II , Animals , Blood Pressure/drug effects , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Cyclic GMP/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress , Fibrosis , Genetic Predisposition to Disease , Hypertension/genetics , Hypertension/physiopathology , Hypertension/prevention & control , Male , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/physiopathology , Myocardium/metabolism , Myocardium/pathology , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitrites/metabolism , Phenotype , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction , Stromal Interaction Molecule 1/deficiency , Stromal Interaction Molecule 1/genetics , Time Factors , Transcription Factor CHOP/deficiency , Transcription Factor CHOP/genetics , Transforming Growth Factor beta/metabolism , Vasodilation/drug effects , Vasodilator Agents/pharmacology
14.
Diabetes Metab Res Rev ; 31(1): 39-49, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24652705

ABSTRACT

BACKGROUND: We previously reported that enhanced nuclear factor kappa B (NFκB) activity is responsible for resistance arteries dysfunction in type 2 diabetic mice. METHODS: In this study, we aimed to determine whether augmented NFκB activity also impairs conductance artery (thoracic aorta) function in type 2 diabetic mice. We treated type 2 diabetic (db(-) /db(-) ) and control (db(-) /db(+) ) mice with two NFκB inhibitors (dehydroxymethylepoxyquinomicin, 6 mg/kg, twice a week and IKK-NBD peptide, 500 µg/kg/day) for 4 weeks. RESULTS: As expected, the NFκB inhibition did not affect blood glucose level and body weight. Thoracic aorta vascular endothelium-dependent relaxation (EDR), determined by the wire myograph, was impaired in diabetic mice compared with control and was significantly improved after NFκB inhibition. Interestingly, thoracic EDR was also rescued in db(-) /db(-p50NFκB-/-) and db(-) /db(-PARP-1-/-) double knockout mice compared with db(-) /db(-) mice. Similarly, the acute in vitro down regulation of NFκB-p65 using p65 shRNA lentiviral particles in arteries from db(-) /db(-) mice also improved thoracic aorta EDR. Western blot analysis showed that the p65NFκB phosphorylation, cleaved PARP-1 and COX-2 expression were increased in thoracic aorta from diabetic mice, which were restored after NFκB inhibition and in db(-) /db(-p-50NFκB-/-) and db(-) /db(-PARP-1-/-) mice. CONCLUSIONS: The present results indicate that in male type 2 diabetic mice, the augmented NFκB activity also impairs conductance artery function through PARP-1 and COX-2-dependent mechanisms.


Subject(s)
Arteries/drug effects , Benzamides/pharmacology , Cyclohexanones/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Heart Conduction System/drug effects , I-kappa B Proteins/pharmacology , NF-kappa B/antagonists & inhibitors , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiopathology , Arteries/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Heart Conduction System/physiology , Male , Mice , Mice, Knockout , Vasodilation/drug effects
16.
Biochim Biophys Acta ; 1843(6): 1063-75, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24576409

ABSTRACT

BACKGROUND: We recently reported that ER stress plays a key role in vascular endothelial dysfunction during hypertension. In this study we aimed to elucidate the mechanisms by which ER stress induction and oxidative stress impair vascular endothelial function. METHODOLOGY/PRINCIPAL FINDINGS: We conducted in vitro studies with primary endothelial cells from coronary arteries stimulated with tunicamycin, 1µg/mL, in the presence or absence of two ER stress inhibitors: tauroursodeoxycholic acid (Tudca), 500µg/mL, and 4-phenylbutyric acid (PBA), 5mM. ER stress induction was assessed by enhanced phosphorylation of PERK and eIF2α, and increased expression of CHOP, ATF6 and Grp78/Bip. The ER stress induction increased p38 MAPK phosphorylation, Nox2/4 mRNA levels and NADPH oxidase activity, and decreased eNOS promoter activity, eNOS expression and phosphorylation, and nitrite levels. Interestingly, the inhibition of p38 MAPK pathway reduced CHOP and Bip expressions enhanced by tunicamycin and restored eNOS promoter activation as well as phosphorylation. To study the effects of ER stress induction in vivo, we used C57BL/6J mice and p47phox(-/-) mice injected with tunicamycin or saline. The ER stress induction in mice significantly impaired vascular endothelium-dependent and independent relaxation in C57BL/6J mice compared with p47phox(-/-) mice indicating NADPH oxidase activity as an intermediate for ER stress in vascular endothelial dysfunction. CONCLUSION/SIGNIFICANCE: We conclude that chemically induced ER stress leads to a downstream enhancement of p38 MAPK and oxidative stress causing vascular endothelial dysfunction. Our results indicate that inhibition of ER stress could be a novel therapeutic strategy to attenuate vascular dysfunction during cardiovascular diseases.


Subject(s)
Coronary Vessels/pathology , Endoplasmic Reticulum Stress/physiology , Endothelium, Vascular/pathology , Oxidative Stress , Vascular Diseases/pathology , Animals , Antiviral Agents/pharmacology , Blood Pressure/drug effects , Blotting, Western , Cells, Cultured , Comet Assay , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Luciferases/metabolism , Male , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , NADPH Oxidase 2 , NADPH Oxidase 4 , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , NADPH Oxidases/physiology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tunicamycin/pharmacology , Vascular Diseases/metabolism
17.
Diabetes ; 62(6): 2078-87, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23349490

ABSTRACT

Type 2 diabetes (T2D) is associated with vascular dysfunction. We hypothesized that increased nuclear factor-κB (NF-κB) signaling contributes to vascular dysfunction in T2D. We treated type 2 diabetic (db(-)/db(-)) and control (db(-)/db(+)) mice with two NF-κB inhibitors (6 mg/kg dehydroxymethylepoxyquinomicin twice a week and 500 µg/kg/day IKK-NBD peptide) for 4 weeks. Pressure-induced myogenic tone was significantly potentiated, while endothelium-dependent relaxation (EDR) was impaired in small coronary arterioles and mesenteric resistance artery from diabetic mice compared with controls. Interestingly, diabetic mice treated with NF-κB inhibitors had significantly reduced myogenic tone potentiation and improved EDR. Importantly, vascular function was also rescued in db(-)/db(-p50NF-κB-/-) and db(-)/db(-PARP-1-/-) double knockout mice compared with db(-)/db(-) mice. Additionally, the acute in vitro downregulation of NF-κB-p65 using p65NF-κB short hairpin RNA lentivirus in arteries from db(-)/db(-) mice also improved vascular function. The NF-κB inhibition did not affect blood glucose level or body weight. The RNA levels for Sp-1 and eNOS phosphorylation were decreased, while p65NF-κB phosphorylation, cleaved poly(ADP-ribose) polymerase (PARP)-1, and cyclooxygenase (COX)-2 expression were increased in arteries from diabetic mice, which were restored after NF-κB inhibition and in db(-)/db(-p50NF-κB-/-) and db(-)/db(-PARP-1-/-) mice. In the current study, we provided evidence that enhanced NF-κB activity impairs vascular function by PARP-1-, Sp-1-, and COX-2-dependent mechanisms in male type 2 diabetic mice. Therefore, NF-κB could be a potential target to overcome diabetes-induced vascular dysfunction.


Subject(s)
Cyclooxygenase 2/metabolism , Diabetes Mellitus, Type 2/metabolism , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Sp1 Transcription Factor/metabolism , Animals , Benzamides/pharmacology , Cells, Cultured , Cyclohexanones/pharmacology , Cyclooxygenase 2/genetics , Diabetes Mellitus, Type 2/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Male , Mice , NF-kappa B/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Real-Time Polymerase Chain Reaction , Sp1 Transcription Factor/genetics
18.
Am J Pathol ; 180(1): 410-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22067908

ABSTRACT

Type 2 diabetes is a key risk factor for ischemia-dependent pathology; therefore, a significant medical need exists to develop novel therapies that increase the formation of new vessels. We explored the therapeutic potential of epidermal growth factor receptor tyrosine kinase (EGFRtk) and extracellular signal-regulated kinase 1/2 (ERK1/2) inhibition in impaired ischemia-induced neovascularization in type 2 diabetes. Unilateral femoral artery ligation was performed in diabetic (db(-)/db(-)) and their control (db(-)/db(+)) mice for 4 weeks, followed by treatments with EGFRtk and ERK1/2 inhibitors (AG1478, 10 mg/kg/day and U0126, 400 µg/kg/day, respectively) for 3 weeks. Neovascularization, blood flow recovery, vascular and capillary density, and endothelial nitric oxide synthase activity were significantly impaired and were associated with enhanced EGFRtk and ERK1/2 activity in db(-)/db(-) mice. EGFRtk and ERK1/2 inhibitors did not have any effect in control mice, while in db(-)/db(-) mice there was a significant increase in neovascularization, blood flow recovery, vascular and capillary density, endothelial nitric oxide synthase activity, and were associated with a decrease in EGFRtk and ERK1/2 activity. Our data demonstrated that the inhibition of EGFRtk and ERK1/2 restored ischemia-induced neovascularization and blood flow recovery in type 2 diabetic mice. Thus, EGFRtk and ERK1/2 could be possible targets to protect from ischemia-induced vascular pathology in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/prevention & control , Diabetic Angiopathies/prevention & control , ErbB Receptors/antagonists & inhibitors , Hindlimb/blood supply , Ischemia/prevention & control , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Blood Flow Velocity/physiology , Blood Glucose/metabolism , Body Weight/physiology , Capillaries/physiology , Cyclic GMP/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/physiopathology , Diabetic Angiopathies/blood , Diabetic Angiopathies/physiopathology , Insulin/metabolism , Ischemia/blood , Ischemia/physiopathology , Male , Mice , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/metabolism
19.
J Pathol ; 227(2): 165-74, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22081301

ABSTRACT

Endoplasmic reticulum (ER) stress and inflammation are important mechanisms that underlie many of the serious consequences of type II diabetes. However, the role of ER stress and inflammation in impaired ischaemia-induced neovascularization in type II diabetes is unknown. We studied ischaemia-induced neovascularization in the hind-limb of 4-week-old db - /db- mice and their controls treated with or without the ER stress inhibitor (tauroursodeoxycholic acid, TUDCA, 150 mg/kg per day) and interleukin-1 receptor antagonist (anakinra, 0.5 µg/mouse per day) for 4 weeks. Blood pressure was similar in all groups of mice. Blood glucose, insulin levels, and body weight were reduced in db - /db- mice treated with TUDCA. Increased cholesterol and reduced adiponectin in db - /db- mice were restored by TUDCA and anakinra treatment. ER stress and inflammation in the ischaemic hind-limb in db - /db- mice were attenuated by TUDCA and anakinra treatment. Ischaemia-induced neovascularization and blood flow recovery were significantly reduced in db - /db- mice compared to control. Interestingly, neovascularization and blood flow recovery were restored in db - /db- mice treated with TUDCA or anakinra compared to non-treated db - /db- mice. TUDCA and anakinra enhanced eNOS-cGMP, VEGFR2, and reduced ERK1/2 MAP-kinase signalling, while endothelial progenitor cell number was similar in all groups of mice. Our findings demonstrate that the inhibition of ER stress and inflammation prevents impaired ischaemia-induced neovascularization in type II diabetic mice. Thus, ER stress and inflammation could be potential targets for a novel therapeutic approach to prevent impaired ischaemia-induced vascular pathology in type II diabetes.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Blood Vessels/drug effects , Diabetes Mellitus, Type 2/drug therapy , Diabetic Angiopathies/prevention & control , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum/drug effects , Interleukin 1 Receptor Antagonist Protein/pharmacology , Ischemia/drug therapy , Muscle, Skeletal/blood supply , Taurochenodeoxycholic Acid/pharmacology , Animals , Biomarkers/blood , Blood Vessels/immunology , Blood Vessels/metabolism , Blood Vessels/pathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Diabetes Mellitus, Type 2/pathology , Diabetic Angiopathies/blood , Diabetic Angiopathies/etiology , Diabetic Angiopathies/immunology , Diabetic Angiopathies/pathology , Disease Models, Animal , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Hindlimb , Ischemia/blood , Ischemia/complications , Ischemia/immunology , Ischemia/pathology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Recovery of Function , Regional Blood Flow/drug effects , Signal Transduction/drug effects , Time Factors
20.
Am J Pathol ; 178(1): 434-41, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224080

ABSTRACT

Coronary artery disease in patients with hypertension is increasing worldwide and leads to severe cardiovascular complications. The cellular and molecular mechanisms that underlie this pathologic condition are not well understood. Experimental and clinical research indicates that immune cells and inflammation play a central role in the pathogenesis of cardiovascular diseases. Recently, it has been reported that CD4(+)CD25(+) regulatory T cells (Tregs) regulate heart fibrosis in hypertension. In this study, we determined the role of Tregs in coronary arteriolar endothelial dysfunction in angiotensin II-dependent hypertensive mice. Mice infused with angiotensin II had significantly increased blood pressure, as determined using telemetry, and apoptotic Treg numbers, as measured using flow cytometry. The mice displayed inflammation, assessed by macrophage activation/infiltration into coronary arterioles and the heart, and increased local tumor necrosis factor-α release, which participates in reduced coronary arteriolar endothelial-dependent relaxation in response to acetylcholine using an arteriograph. Hypertensive mice injected with Tregs isolated from control mice had significantly reduced macrophage activation and infiltration, reduced tumor necrosis factor-α release, and improved coronary arteriolar endothelium-dependent relaxation. Our novel data indicate that Tregs are important in the development of coronary arteriolar endothelial dysfunction in hypertension. These results suggest a new direction in the investigation of vascular disease in hypertension and could lead to a therapeutic strategy that involves immune system modulation using Tregs.


Subject(s)
Coronary Vessels/physiopathology , Endothelium, Vascular/physiopathology , Hypertension/physiopathology , T-Lymphocytes, Regulatory/immunology , Animals , Arterioles/immunology , Arterioles/physiopathology , Coronary Vessels/immunology , Endothelium, Vascular/immunology , Hypertension/immunology , Lymphocyte Count , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...