Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Circ Res ; 135(4): 488-502, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-38979610

ABSTRACT

BACKGROUND: The long isoform of the Wnk1 (with-no-lysine [K] kinase 1) is a ubiquitous serine/threonine kinase, but its role in vascular smooth muscle cells (VSMCs) pathophysiology remains unknown. METHODS: AngII (angiotensin II) was infused in Apoe-/- to induce experimental aortic aneurysm. Mice carrying an Sm22-Cre allele were cross-bred with mice carrying a floxed Wnk1 allele to specifically investigate the functional role of Wnk1 in VSMCs. RESULTS: Single-cell RNA-sequencing of the aneurysmal abdominal aorta from AngII-infused Apoe-/- mice revealed that VSMCs that did not express Wnk1 showed lower expression of contractile phenotype markers and increased inflammatory activity. Interestingly, WNK1 gene expression in VSMCs was decreased in human abdominal aortic aneurysm. Wnk1-deficient VSMCs lost their contractile function and exhibited a proinflammatory phenotype, characterized by the production of matrix metalloproteases, as well as cytokines and chemokines, which contributed to local accumulation of inflammatory macrophages, Ly6Chi monocytes, and γδ T cells. Sm22Cre+Wnk1lox/lox mice spontaneously developed aortitis in the infrarenal abdominal aorta, which extended to the thoracic area over time without any negative effect on long-term survival. AngII infusion in Sm22Cre+Wnk1lox/lox mice aggravated the aortic disease, with the formation of lethal abdominal aortic aneurysms. Pharmacological blockade of γδ T-cell recruitment using neutralizing anti-CXCL9 (anti-CXC motif chemokine ligand 9) antibody treatment, or of monocyte/macrophage using Ki20227, a selective inhibitor of CSF1 receptor, attenuated aortitis. Wnk1 deletion in VSMCs led to aortic wall remodeling with destruction of elastin layers, increased collagen content, and enhanced local TGF-ß (transforming growth factor-beta) 1 expression. Finally, in vivo TGF-ß blockade using neutralizing anti-TGF-ß antibody promoted saccular aneurysm formation and aorta rupture in Sm22 Cre+ Wnk1lox/lox mice but not in control animals. CONCLUSION: Wnk1 is a key regulator of VSMC function. Wnk1 deletion promotes VSMC phenotype switch toward a pathogenic proinflammatory phenotype, orchestrating deleterious vascular remodeling and spontaneous severe aortitis in mice.


Subject(s)
Angiotensin II , Aortic Aneurysm, Abdominal , Aortitis , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , WNK Lysine-Deficient Protein Kinase 1 , Animals , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Humans , WNK Lysine-Deficient Protein Kinase 1/genetics , WNK Lysine-Deficient Protein Kinase 1/metabolism , Mice, Inbred C57BL , Male , Cells, Cultured , Mice, Knockout, ApoE , Disease Models, Animal , Inflammation/metabolism , Inflammation/genetics , Inflammation/pathology , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology
2.
J Cell Physiol ; 236(6): 4555-4564, 2021 06.
Article in English | MEDLINE | ID: mdl-33241567

ABSTRACT

Selenium (Se) is an essential trace element in organism. Se deficiency can cause many diseases, including vascular disease. Studies have shown that inflammation is the main inducement of vascular disease, microRNA (miRNA) can influence inflammation in various ways, and Se deficiency can affect miRNAs expression. To study the mechanism of aorta damage caused by Se deficiency, we constructed a Se deficiency porcine aorta model and found that Se deficiency can significantly inhibit miR-223, which downregulates the expression of nucleotide-binding oligomerization domain-like receptor family 3 (NLRP3). Subsequently, we found that in Se deficiency group, NLRP3, and its downstream (caspase-1, apoptosis-related spot-like protein [ASC], IL-18, IL-1ß) expression was significantly increased. In vitro, we cultured pig iliac endothelium cell lines, and constructed miR-223 knockdown and overexpression models. NLRP3 messenger RNA and protein levels were significant increased in the knockdown group, and decreased in the overexpression group. The results of this study show that Se deficiency in porcine arteries can induce inflammation through miR-223/NLRP3.


Subject(s)
Aorta/metabolism , Aortitis/metabolism , Endothelial Cells/metabolism , Inflammasomes/metabolism , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Selenium/deficiency , Animal Feed , Animal Nutritional Physiological Phenomena , Animals , Aorta/immunology , Aorta/pathology , Aortitis/genetics , Aortitis/immunology , Aortitis/pathology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/genetics , Caspase 1/metabolism , Cells, Cultured , Disease Models, Animal , Endothelial Cells/immunology , Endothelial Cells/pathology , Inflammasomes/genetics , Interleukin-18/genetics , Interleukin-18/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , MicroRNAs/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Signal Transduction , Sus scrofa
3.
Arterioscler Thromb Vasc Biol ; 40(10): 2494-2507, 2020 10.
Article in English | MEDLINE | ID: mdl-32787523

ABSTRACT

OBJECTIVE: Currently, there are no approved drugs for abdominal aortic aneurysm (AAA) treatment, likely due to limited understanding of the primary molecular mechanisms underlying AAA development and progression. BAF60a-a unique subunit of the SWI/SNF (switch/sucrose nonfermentable) chromatin remodeling complex-is a novel regulator of metabolic homeostasis, yet little is known about its function in the vasculature and pathogenesis of AAA. In this study, we sought to investigate the role and underlying mechanisms of vascular smooth muscle cell (VSMC)-specific BAF60a in AAA formation. Approach and Results: BAF60a is upregulated in human and experimental murine AAA lesions. In vivo studies revealed that VSMC-specific knockout of BAF60a protected mice from both Ang II (angiotensin II)-induced and elastase-induced AAA formation with significant suppression of vascular inflammation, monocyte infiltration, and elastin fragmentation. Through RNA sequencing and pathway analysis, we found that the expression of inflammatory response genes in cultured human aortic smooth muscle cells was significantly downregulated by small interfering RNA-mediated BAF60a knockdown while upregulated upon adenovirus-mediated BAF60a overexpression. BAF60a regulates VSMC inflammation by recruiting BRG1 (Brahma-related gene-1)-a catalytic subunit of the SWI/SNF complex-to the promoter region of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) target genes. Furthermore, loss of BAF60a in VSMCs prevented the upregulation of the proteolytic enzyme cysteine protease CTSS (cathepsin S), thus ameliorating ECM (extracellular matrix) degradation within the vascular wall in AAA. CONCLUSIONS: Our study demonstrated that BAF60a is required to recruit the SWI/SNF complex to facilitate the epigenetic regulation of VSMC inflammation, which may serve as a potential therapeutic target in preventing and treating AAA.


Subject(s)
Aortic Aneurysm, Abdominal/prevention & control , Aortitis/prevention & control , Chromosomal Proteins, Non-Histone/deficiency , Extracellular Matrix/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Vascular Remodeling , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Case-Control Studies , Cathepsins/metabolism , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , Disease Models, Animal , Extracellular Matrix/pathology , Humans , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Signal Transduction
4.
Circulation ; 139(10): 1320-1334, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30586743

ABSTRACT

BACKGROUND: The majority of the human genome comprises noncoding sequences, which are in part transcribed as long noncoding RNAs (lncRNAs). lncRNAs exhibit multiple functions, including the epigenetic control of gene expression. In this study, the effect of the lncRNA MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) on atherosclerosis was examined. METHODS: The effect of MALAT1 on atherosclerosis was determined in apolipoprotein E-deficient (Apoe-/-) MALAT1-deficient (Malat1-/-) mice that were fed with a high-fat diet and by studying the regulation of MALAT1 in human plaques. RESULTS: Apoe-/- Malat1-/- mice that were fed a high-fat diet showed increased plaque size and infiltration of inflammatory CD45+ cells compared with Apoe-/- Malat1+/+ control mice. Bone marrow transplantation of Apoe-/- Malat1-/- bone marrow cells in Apoe-/- Malat1+/+ mice enhanced atherosclerotic lesion formation, which suggests that hematopoietic cells mediate the proatherosclerotic phenotype. Indeed, bone marrow cells isolated from Malat1-/- mice showed increased adhesion to endothelial cells and elevated levels of proinflammatory mediators. Moreover, myeloid cells of Malat1-/- mice displayed enhanced adhesion to atherosclerotic arteries in vivo. The anti-inflammatory effects of MALAT1 were attributed in part to reduction of the microRNA miR-503. MALAT1 expression was further significantly decreased in human plaques compared with normal arteries and was lower in symptomatic versus asymptomatic patients. Lower levels of MALAT1 in human plaques were associated with a worse prognosis. CONCLUSIONS: Reduced levels of MALAT1 augment atherosclerotic lesion formation in mice and are associated with human atherosclerotic disease. The proatherosclerotic effects observed in Malat1-/- mice were mainly caused by enhanced accumulation of hematopoietic cells.


Subject(s)
Aorta/metabolism , Aortitis/metabolism , Atherosclerosis/metabolism , Bone Marrow Cells/metabolism , Hematopoiesis , Plaque, Atherosclerotic , RNA, Long Noncoding/metabolism , Animals , Aorta/pathology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Bone Marrow Cells/pathology , Bone Marrow Transplantation , Case-Control Studies , Disease Models, Animal , Down-Regulation , Humans , Mice, Inbred C57BL , Mice, Knockout, ApoE , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Signal Transduction
5.
Circulation ; 138(16): 1706-1719, 2018 10 16.
Article in English | MEDLINE | ID: mdl-29700120

ABSTRACT

BACKGROUND: The coagulation system is closely linked with vascular inflammation, although the underlying mechanisms are still obscure. Recent studies show that protease-activated receptor (PAR)-2, a major receptor of activated factor X, is expressed in both vascular cells and leukocytes, suggesting that PAR-2 may contribute to the pathogenesis of inflammatory diseases. Here we investigated the role of PAR-2 in vascular inflammation and atherogenesis. METHODS: We generated apolipoprotein E-deficient ( ApoE-/-) mice lacking systemic PAR-2 expression ( PAR-2-/- ApoE-/-). ApoE-/- mice, which lack or express PAR-2 only in bone marrow (BM) cells, were also generated by BM transplantation. Atherosclerotic lesions were investigated after 20 weeks on a Western-type diet by histological analyses, quantitative reverse transcription polymerase chain reaction, and Western blotting. In vitro experiments using BM-derived macrophages were performed to confirm the proinflammatory roles of PAR-2. The association between plasma activated factor X level and the severity of coronary atherosclerosis was also examined in humans who underwent coronary intervention. RESULTS: PAR-2-/- ApoE-/- mice showed reduced atherosclerotic lesions in the aortic arch ( P<0.05) along with features of stabilized atherosclerotic plaques, such as less lipid deposition ( P<0.05), collagen loss ( P<0.01), macrophage accumulation ( P<0.05), and inflammatory molecule expression ( P<0.05) compared with ApoE-/- mice. Systemic PAR2 deletion in ApoE-/-mice significantly decreased the expression of inflammatory molecules in the aorta. The results of BM transplantation experiments demonstrated that PAR-2 in hematopoietic cells contributed to atherogenesis in ApoE-/- mice. PAR-2 deletion did not alter metabolic parameters. In vitro experiments demonstrated that activated factor X or a specific peptide agonist of PAR-2 significantly increased the expression of inflammatory molecules and lipid uptake in BM-derived macrophages from wild-type mice compared with those from PAR-2-deficient mice. Activation of nuclear factor-κB signaling was involved in PAR-2-associated vascular inflammation and macrophage activation. In humans who underwent coronary intervention, plasma activated factor X level independently correlated with the severity of coronary atherosclerosis as determined by Gensini score ( P<0.05) and plaque volume ( P<0.01). CONCLUSIONS: PAR-2 signaling activates macrophages and promotes vascular inflammation, increasing atherosclerosis in ApoE-/- mice. This signaling pathway may also participate in atherogenesis in humans.


Subject(s)
Aorta, Thoracic/metabolism , Aortitis/metabolism , Atherosclerosis/metabolism , Inflammation Mediators/metabolism , Macrophage Activation , Macrophages/metabolism , Plaque, Atherosclerotic , Receptor, PAR-2/metabolism , Aged , Animals , Aorta, Thoracic/pathology , Aortitis/genetics , Aortitis/pathology , Aortitis/prevention & control , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cells, Cultured , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Diet, Western , Disease Models, Animal , Factor Xa/metabolism , Female , Humans , Lipids/blood , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Receptor, PAR-2/deficiency , Receptor, PAR-2/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
6.
Arterioscler Thromb Vasc Biol ; 38(5): 994-1006, 2018 05.
Article in English | MEDLINE | ID: mdl-29496659

ABSTRACT

OBJECTIVE: Macrophages play a central role in various stages of atherosclerotic plaque formation and progression. The local macrophages reportedly proliferate during atherosclerosis, but the pathophysiological significance of macrophage proliferation in this context remains unclear. Here, we investigated the involvement of local macrophage proliferation during atherosclerosis formation and progression using transgenic mice, in which macrophage proliferation was specifically suppressed. APPROACH AND RESULTS: Inhibition of macrophage proliferation was achieved by inducing the expression of cyclin-dependent kinase inhibitor 1B, also known as p27kip, under the regulation of a scavenger receptor promoter/enhancer. The macrophage-specific human p27kip Tg mice were subsequently crossed with apolipoprotein E-deficient mice for the atherosclerotic plaque study. Results showed that a reduced number of local macrophages resulted in marked suppression of atherosclerotic plaque formation and inflammatory response in the plaque. Moreover, fewer local macrophages in macrophage-specific human p27kip Tg mice helped stabilize the plaque, as evidenced by a reduced necrotic core area, increased collagenous extracellular matrix, and thickened fibrous cap. CONCLUSIONS: These results provide direct evidence of the involvement of local macrophage proliferation in formation and progression of atherosclerotic plaques and plaque stability. Thus, control of macrophage proliferation might represent a therapeutic target for treating atherosclerotic diseases.


Subject(s)
Aorta/pathology , Aortitis/prevention & control , Atherosclerosis/prevention & control , Cell Proliferation , Macrophage Activation , Macrophages, Peritoneal/pathology , Plaque, Atherosclerotic , Animals , Aorta/metabolism , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Collagen/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Disease Models, Animal , Fibrosis , Inflammation Mediators/metabolism , Macrophages, Peritoneal/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Mice, Transgenic , Necrosis , Signal Transduction
7.
Arterioscler Thromb Vasc Biol ; 38(10): 2295-2305, 2018 10.
Article in English | MEDLINE | ID: mdl-30354204

ABSTRACT

Objective- Signaling that activates NFκB (nuclear factor κB) in smooth muscle cells (SMCs) is integral to atherosclerosis and involves reversible ubiquitination that activates proteins downstream of proatherogenic receptors. Deubiquitination of these proteins is mediated by USP20 (ubiquitin-specific protease 20), among other deubiquitinases. We sought to determine whether USP20 activity in SMCs decreases atherosclerosis. Approach and Results- To address this question, we used male Ldlr-/- mice without (control) or with SMC-specific expression of murine USP20 (SMC-USP20-transgenic) or its dominant-negative (DN; C154S/H643Q) mutant (SMC-DN-USP20-transgenic). Before the appearance of intimal macrophages, NFκB activation in aortic medial SMCs was greater in SMC-DN-USP20-transgenic than in control mice. After 16 weeks on a Western diet, SMC-DN-USP20-transgenic mice had 46% greater brachiocephalic artery atheroma area than control mice. Congruently, aortic atherosclerosis assessed en face was 21% greater than control in SMC-DN-USP20-transgenic mice and 13% less than control in SMC-USP20-transgenic mice. In response to TNF (tumor necrosis factor), SMCs from SMC-DN-USP20-transgenic mice showed ≈3-fold greater NFκB activation than control SMCs. Silencing USP20 in SMCs with siRNA (small interfering RNA) augmented NFκB activation by ≈50% in response to either TNF or IL-1ß (interleukin-1ß). Coimmunoprecipitation experiments revealed that USP20 associates with several components of the TNFR1 (TNF receptor-1) signaling pathway, including RIPK1 (receptor-interacting protein kinase 1), a critical checkpoint in TNF-induced NFκB activation and inflammation. TNF evoked ≈2-fold more RIPK1 ubiquitination in SMC-DN-USP20-transgenic than in control SMCs, and RIPK1 was deubiquitinated by purified USP20 in vitro. Conclusions- USP20 attenuates TNF- and IL-1ß-evoked atherogenic signaling in SMCs, by deubiquitinating RIPK1, among other signaling intermediates.


Subject(s)
Aortitis/prevention & control , Atherosclerosis/prevention & control , Endopeptidases/metabolism , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Aorta/drug effects , Aorta/enzymology , Aorta/pathology , Aortitis/enzymology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cells, Cultured , Disease Models, Animal , Endopeptidases/genetics , Female , Hyperplasia , Interleukin-1beta/pharmacology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , NF-kappa B/metabolism , Neointima , Plaque, Atherosclerotic , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, LDL , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/drug effects , Ubiquitin Thiolesterase , Ubiquitination
8.
Arterioscler Thromb Vasc Biol ; 37(11): 2161-2170, 2017 11.
Article in English | MEDLINE | ID: mdl-28882868

ABSTRACT

OBJECTIVE: Abdominal aortic aneurysm (AAA) is an increasingly prevalent and ultimately fatal disease with no effective pharmacological treatment. Because matrix degradation induced by vascular inflammation is the major pathophysiology of AAA, attenuation of this inflammation may improve its outcome. Previous studies suggested that miR-33 (microRNA-33) inhibition and genetic ablation of miR-33 increased serum high-density lipoprotein cholesterol and attenuated atherosclerosis. APPROACH AND RESULTS: MiR-33a-5p expression in central zone of human AAA was higher than marginal zone. MiR-33 deletion attenuated AAA formation in both mouse models of angiotensin II- and calcium chloride-induced AAA. Reduced macrophage accumulation and monocyte chemotactic protein-1 expression were observed in calcium chloride-induced AAA walls in miR-33-/- mice. In vitro experiments revealed that peritoneal macrophages from miR-33-/- mice showed reduced matrix metalloproteinase 9 expression levels via c-Jun N-terminal kinase inactivation. Primary aortic vascular smooth muscle cells from miR-33-/- mice showed reduced monocyte chemotactic protein-1 expression by p38 mitogen-activated protein kinase attenuation. Both of the inactivation of c-Jun N-terminal kinase and p38 mitogen-activated protein kinase were possibly because of the increase of ATP-binding cassette transporter A1 that is a well-known target of miR-33. Moreover, high-density lipoprotein cholesterol derived from miR-33-/- mice reduced expression of matrix metalloproteinase 9 in macrophages and monocyte chemotactic protein-1 in vascular smooth muscle cells. Bone marrow transplantation experiments indicated that miR-33-deficient bone marrow cells ameliorated AAA formation in wild-type recipients. MiR-33 deficiency in recipient mice was also shown to contribute the inhibition of AAA formation. CONCLUSIONS: These data strongly suggest that inhibition of miR-33 will be effective as a novel strategy for treating AAA.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Aortitis/prevention & control , Inflammation Mediators/metabolism , MicroRNAs/metabolism , Angiotensin II , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/metabolism , Aortitis/chemically induced , Aortitis/genetics , Aortitis/metabolism , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Bone Marrow Transplantation , Calcium Chloride , Cell Line , Chemokine CCL2/metabolism , Cholesterol, HDL/blood , Dilatation, Pathologic , Disease Models, Animal , Female , Genetic Predisposition to Disease , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Signal Transduction , Time Factors , Transfection , Vascular Remodeling , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Arterioscler Thromb Vasc Biol ; 36(5): 886-97, 2016 05.
Article in English | MEDLINE | ID: mdl-26941015

ABSTRACT

OBJECTIVE: Kawasaki disease (KD) is the most common cause of acquired cardiac disease in US children. In addition to coronary artery abnormalities and aneurysms, it can be associated with systemic arterial aneurysms. We evaluated the development of systemic arterial dilatation and aneurysms, including abdominal aortic aneurysm (AAA) in the Lactobacillus casei cell-wall extract (LCWE)-induced KD vasculitis mouse model. METHODS AND RESULTS: We discovered that in addition to aortitis, coronary arteritis and myocarditis, the LCWE-induced KD mouse model is also associated with abdominal aorta dilatation and AAA, as well as renal and iliac artery aneurysms. AAA induced in KD mice was exclusively infrarenal, both fusiform and saccular, with intimal proliferation, myofibroblastic proliferation, break in the elastin layer, vascular smooth muscle cell loss, and inflammatory cell accumulation in the media and adventitia. Il1r(-/-), Il1a(-/-), and Il1b(-/-) mice were protected from KD associated AAA. Infiltrating CD11c(+) macrophages produced active caspase-1, and caspase-1 or NLRP3 deficiency inhibited AAA formation. Treatment with interleukin (IL)-1R antagonist (Anakinra), anti-IL-1α, or anti-IL-1ß mAb blocked LCWE-induced AAA formation. CONCLUSIONS: Similar to clinical KD, the LCWE-induced KD vasculitis mouse model can also be accompanied by AAA formation. Both IL-1α and IL-1ß play a key role, and use of an IL-1R blocking agent that inhibits both pathways may be a promising therapeutic target not only for KD coronary arteritis, but also for the other systemic arterial aneurysms including AAA that maybe seen in severe cases of KD. The LCWE-induced vasculitis model may also represent an alternative model for AAA disease.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mucocutaneous Lymph Node Syndrome/complications , Receptors, Interleukin-1 Type I/metabolism , Signal Transduction , Animals , Aorta, Abdominal/drug effects , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/prevention & control , Aortitis/genetics , Aortitis/metabolism , Aortitis/pathology , Caspase 1/deficiency , Caspase 1/genetics , Cell Proliferation , Cell Wall , Dilatation, Pathologic , Disease Models, Animal , Elastin/metabolism , Female , Gene Expression Profiling , Genotype , Humans , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1alpha/deficiency , Interleukin-1alpha/genetics , Interleukin-1beta/deficiency , Interleukin-1beta/genetics , Lacticaseibacillus casei , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Mucocutaneous Lymph Node Syndrome/chemically induced , Mucocutaneous Lymph Node Syndrome/drug therapy , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Phenotype , Receptors, Interleukin-1 Type I/deficiency , Receptors, Interleukin-1 Type I/genetics , Signal Transduction/drug effects , Time Factors
10.
Clin Sci (Lond) ; 130(15): 1363-74, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27190136

ABSTRACT

Oxidative stress and inflammation are central mediators of atherosclerosis particularly in the context of diabetes. The potential interactions between the major producers of vascular reactive oxygen species (ROS), NADPH oxidase (NOX) enzymes and immune-inflammatory processes remain to be fully elucidated. In the present study we investigated the roles of the NADPH oxidase subunit isoforms, NOX4 and NOX1, in immune cell activation and recruitment to the aortic sinus atherosclerotic plaque in diabetic ApoE(-/-) mice. Plaque area analysis showed that NOX4- and NOX1-derived ROS contribute to atherosclerosis in the aortic sinus following 10 weeks of diabetes. Immunohistochemical staining of the plaques revealed that NOX4-derived ROS regulate T-cell recruitment. In addition, NOX4-deficient mice showed a reduction in activated CD4(+) T-cells in the draining lymph nodes of the aortic sinus coupled with reduced pro-inflammatory gene expression in the aortic sinus. Conversely, NOX1-derived ROS appeared to play a more important role in macrophage accumulation. These findings demonstrate distinct roles for NOX4 and NOX1 in immune-inflammatory responses that drive atherosclerosis in the aortic sinus of diabetic mice.


Subject(s)
Aortitis/enzymology , Apolipoproteins E/deficiency , Atherosclerosis/enzymology , Diabetes Mellitus, Experimental/enzymology , Immunity, Cellular , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidases/metabolism , Sinus of Valsalva/enzymology , Animals , Aortitis/genetics , Aortitis/immunology , Aortitis/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , CD4-Positive T-Lymphocytes/enzymology , CD4-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte , Cytokines/immunology , Cytokines/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/pathology , Genetic Predisposition to Disease , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Macrophages/enzymology , Macrophages/immunology , Mice, Knockout , NADH, NADPH Oxidoreductases/deficiency , NADH, NADPH Oxidoreductases/genetics , NADPH Oxidase 1 , NADPH Oxidase 4 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Oxidative Stress , Phenotype , Plaque, Atherosclerotic , Reactive Oxygen Species/metabolism , Signal Transduction , Sinus of Valsalva/immunology , Sinus of Valsalva/pathology
11.
Arterioscler Thromb Vasc Biol ; 35(11): 2412-22, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26338301

ABSTRACT

OBJECTIVE: Thrombomodulin (TM), a glycoprotein constitutively expressed in the endothelium, is well known for its anticoagulant and anti-inflammatory properties. Paradoxically, we recently found that monocytic membrane-bound TM (ie, endogenous TM expression in monocytes) triggers lipopolysaccharide- and gram-negative bacteria-induced inflammatory responses. However, the significance of membrane-bound TM in chronic sterile vascular inflammation and the development of abdominal aortic aneurysm (AAA) remains undetermined. APPROACH AND RESULTS: Implicating a potential role for membrane-bound TM in AAA, we found that TM signals were predominantly localized to macrophages and vascular smooth muscle cells in human aneurysm specimens. Characterization of the CaCl2-induced AAA in mice revealed that during aneurysm development, TM expression was mainly localized in infiltrating macrophages and vascular smooth muscle cells. To investigate the function of membrane-bound TM in vivo, transgenic mice with myeloid- (LysMcre/TM(flox/flox)) and vascular smooth muscle cell-specific (SM22-cre(tg)/TM(flox/flox)) TM ablation and their respective wild-type controls (TM(flox/flox) and SM22-cre(tg)/TM(+/+)) were generated. In the mouse CaCl2-induced AAA model, deficiency of myeloid TM, but not vascular smooth muscle cell TM, inhibited macrophage accumulation, attenuated proinflammatory cytokine and matrix metalloproteinase-9 production, and finally mitigated elastin destruction and aortic dilatation. In vitro TM-deficient monocytes/macrophages, versus TM wild-type counterparts, exhibited attenuation of proinflammatory mediator expression, adhesion to endothelial cells, and generation of reactive oxygen species. Consistently, myeloid TM-deficient hyperlipidemic mice (ApoE(-/-)/LysMcre/TM(flox/flox)) were resistant to AAA formation induced by angiotensin II infusion, along with reduced macrophage infiltration, suppressed matrix metalloproteinase activities, and diminished oxidative stress. CONCLUSIONS: Membrane-bound TM in macrophages plays an essential role in the development of AAA by enhancing proinflammatory mediator elaboration, macrophage recruitment, and oxidative stress.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/metabolism , Aortitis/metabolism , Cell Membrane/metabolism , Macrophages, Peritoneal/metabolism , Thrombomodulin/metabolism , Angiotensin II , Animals , Aorta, Abdominal/immunology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/immunology , Aortitis/chemically induced , Aortitis/genetics , Aortitis/immunology , Calcium Chloride , Cell Membrane/immunology , Cells, Cultured , Chemotaxis , Disease Models, Animal , Elastin/metabolism , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages, Peritoneal/immunology , Matrix Metalloproteinase 9/metabolism , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism , Oxidative Stress , RNA Interference , Retrospective Studies , Signal Transduction , Thrombomodulin/deficiency , Thrombomodulin/genetics , Time Factors , Transfection
12.
Arterioscler Thromb Vasc Biol ; 35(12): 2605-16, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26515418

ABSTRACT

OBJECTIVE: Kawasaki disease (KD) is the most common cause of acute vasculitis and acquired cardiac disease among US children. We have previously shown that both TLR2/MyD88 and interleukin (IL)-1ß signaling are required for the Lactobacillus casei cell wall extract-induced KD vasculitis mouse model. The objectives of this study were to investigate the cellular origins of IL-1 production, the role of CD11c(+) dendritic cells and macrophages, and the relative contribution of hematopoietic and stromal cells for IL-1 responsive cells, as well the MyD88 signaling, in Lactobacillus casei cell wall extract-induced KD mouse model of vasculitis. APPROACH AND RESULTS: Using mouse knockout models and antibody depletion, we found that both IL-1α and IL-1ß were required for Lactobacillus casei cell wall extract-induced KD. Both dendritic cells and macrophages were necessary, and we found that MyD88 signaling was required in both hematopoietic and stromal cells. However, IL-1 response and signaling were critically required in nonendothelial stromal cells, but not in hematopoietic cells. CONCLUSIONS: Our results suggest that IL-1α and IL-1ß, as well as CD11c(+) dendritic cells and macrophages, are essential for the development of KD vasculitis and coronary arteritis in this mouse model. Bone marrow chimera experiments suggest that MyD88 signaling is important in both hematopoietic and stromal cells, whereas IL-1 signaling and response are required only in stromal cells, but not in endothelial cells. Determining the role of IL-1α and IL-1ß and of specific cell types in the KD vasculitis mouse model may have important implications for the design of more targeted therapies and understanding of the molecular mechanisms of KD immunopathologies.


Subject(s)
Aortitis/metabolism , Coronary Artery Disease/metabolism , Interleukin-1alpha/metabolism , Interleukin-1beta/metabolism , Mucocutaneous Lymph Node Syndrome/metabolism , Signal Transduction , Stromal Cells/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Aortitis/chemically induced , Aortitis/genetics , Aortitis/pathology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Carrier Proteins/metabolism , Caspase 1/metabolism , Cell Wall , Cells, Cultured , Coronary Artery Disease/chemically induced , Coronary Artery Disease/genetics , Coronary Artery Disease/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dendritic Cells/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Lacticaseibacillus casei , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mucocutaneous Lymph Node Syndrome/chemically induced , Mucocutaneous Lymph Node Syndrome/genetics , Mucocutaneous Lymph Node Syndrome/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Receptors, Interleukin-1 Type I/genetics , Receptors, Interleukin-1 Type I/metabolism , Stromal Cells/pathology , Transplantation Chimera
13.
Circ Res ; 113(8): 965-74, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-23908345

ABSTRACT

RATIONALE: Atherosclerosis is a major cause of death in patients with chronic kidney disease. Chronic inflammation of the arterial wall including invasion, proliferation, and differentiation of leukocytes is important in atherosclerotic lesion development. How atherosclerotic inflammation is altered in renal impairment is incompletely understood. OBJECTIVE: This study analyzed leukocytes of the atherosclerotic aorta in mice with impaired and normal renal function and studied a mechanism for the alteration in aortic myeloid leukocytes. METHODS AND RESULTS: Unilateral nephrectomy significantly decreased glomerular filtration rate and increased atherosclerotic lesion size and aortic leukocyte numbers in 2 murine atherosclerosis models, apolipoprotein E (Apoe(-/-)) and low-density lipoprotein (LDL) receptor-deficient (LDLr(-/-)) mice. The number of aortic myeloid cells increased significantly. They took-up less oxidized LDL, whereas CD11c expression, interaction with T cells, and aortic T cell proliferation were significantly enhanced in renal impairment. In human peripheral blood mononuclear cell cultures, chronic kidney disease serum decreased lipid uptake and increased human leukocyte antigen II (HLA II) expression. Supplementation with interleukin-17A similarly increased HLA II and CD11c expression and impaired oxidized LDL uptake. Interleukin-17A expression was increased in atherosclerotic mice with renal impairment. Ablation of interleukin-17A in LDLr(-/-) mice by lethal irradiation and reconstitution with Il17a(-/-) bone marrow abolished the effect of renal impairment on aortic CD11b(+) myeloid cell accumulation, CD11c expression, and cell proliferation. Atherosclerotic lesion size was decreased to levels observed in normal kidney function. CONCLUSIONS: Kidney function modifies arterial myeloid cell accumulation and phenotype in atherosclerosis. Our results suggest a central role for interleukin-17A in aggravation of vascular inflammation and atherosclerosis in renal impairment.


Subject(s)
Aorta/metabolism , Aortitis/metabolism , Atherosclerosis/metabolism , Interleukin-17/deficiency , Interleukin-17/metabolism , Kidney Diseases/microbiology , Leukocytes/metabolism , Plaque, Atherosclerotic , Animals , Aorta/immunology , Aorta/pathology , Aortitis/genetics , Aortitis/immunology , Aortitis/pathology , Aortitis/physiopathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/pathology , Atherosclerosis/physiopathology , CD11c Antigen/metabolism , Cells, Cultured , Disease Models, Animal , Glomerular Filtration Rate , Histocompatibility Antigens Class II/metabolism , Interleukin-17/genetics , Kidney/physiopathology , Kidney Diseases/genetics , Kidney Diseases/immunology , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Leukocytes/immunology , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, LDL/deficiency , Receptors, LDL/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
14.
Circ Res ; 112(4): 618-32, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23250987

ABSTRACT

RATIONALE: Aortic aneurysm and dissection (AAD) are major diseases of the adult aorta caused by progressive medial degeneration of the aortic wall. Although the overproduction of destructive factors promotes tissue damage and disease progression, the role of protective pathways is unknown. OBJECTIVE: In this study, we examined the role of AKT2 in protecting the aorta from developing AAD. METHODS AND RESULTS: AKT2 and phospho-AKT levels were significantly downregulated in human thoracic AAD tissues, especially within the degenerative medial layer. Akt2-deficient mice showed abnormal elastic fibers and reduced medial thickness in the aortic wall. When challenged with angiotensin II, these mice developed aortic aneurysm, dissection, and rupture with features similar to those in humans, in both thoracic and abdominal segments. Aortas from Akt2-deficient mice displayed profound tissue destruction, apoptotic cell death, and inflammatory cell infiltration that were not observed in aortas from wild-type mice. In addition, angiotensin II-infused Akt2-deficient mice showed significantly elevated expression of matrix metalloproteinase-9 (MMP-9) and reduced expression of tissue inhibitor of metalloproteinase-1 (TIMP-1). In cultured human aortic vascular smooth muscle cells, AKT2 inhibited the expression of MMP-9 and stimulated the expression of TIMP-1 by preventing the binding of transcription factor forkhead box protein O1 to the MMP-9 and TIMP-1 promoters. CONCLUSIONS: Impaired AKT2 signaling may contribute to increased susceptibility to the development of AAD. Our findings provide evidence of a mechanism that underlies the protective effects of AKT2 on the aortic wall and that may serve as a therapeutic target in the prevention of AAD.


Subject(s)
Aortic Aneurysm, Thoracic/enzymology , Aortic Dissection/enzymology , Proto-Oncogene Proteins c-akt/physiology , Aged , Aortic Dissection/etiology , Aortic Dissection/prevention & control , Angiotensin II/pharmacology , Angiotensin II/toxicity , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/etiology , Aortic Aneurysm, Thoracic/prevention & control , Aortitis/chemically induced , Aortitis/enzymology , Aortitis/genetics , Aortitis/pathology , Apoptosis/drug effects , Case-Control Studies , Cells, Cultured/drug effects , Cells, Cultured/enzymology , Elastic Tissue/pathology , Enzyme Induction , Forkhead Box Protein O1 , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/metabolism , Humans , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/deficiency , Proto-Oncogene Proteins c-akt/genetics , Tissue Inhibitor of Metalloproteinase-1/biosynthesis , Tissue Inhibitor of Metalloproteinase-1/genetics
15.
Arterioscler Thromb Vasc Biol ; 34(1): 52-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24158519

ABSTRACT

OBJECTIVE: The peroxisome proliferator-activated receptor (PPAR) δ regulates systemic lipid homeostasis and inflammation. However, the ability of PPARδ agonists to improve the pathology of pre-established lesions and whether PPARδ activation is atheroprotective in the setting of insulin resistance have not been reported. Here, we examine whether intervention with a selective PPARδ agonist corrects metabolic dysregulation and attenuates aortic inflammation and atherosclerosis. APPROACH AND RESULTS: Low-density lipoprotein receptor knockout mice were fed a chow or a high-fat, high-cholesterol (HFHC) diet (42% fat, 0.2% cholesterol) for 4 weeks. For a further 8 weeks, the HFHC group was fed either HFHC or HFHC plus GW1516 (3 mg/kg per day). GW1516 significantly attenuated pre-established fasting hyperlipidemia, hyperglycemia, and hyperinsulinemia, as well as glucose and insulin intolerance. GW1516 intervention markedly reduced aortic sinus lesions and lesion macrophages, whereas smooth muscle α-actin was unchanged and collagen deposition enhanced. In aortae, GW1516 increased the expression of the PPARδ-specific gene Adfp but not PPARα- or γ-specific genes. GW1516 intervention decreased the expression of aortic proinflammatory M1 cytokines, increased the expression of the anti-inflammatory M2 cytokine Arg1, and attenuated the iNos/Arg1 ratio. Enhanced mitogen-activated protein kinase signaling, known to induce inflammatory cytokine expression in vitro, was enhanced in aortae of HFHC-fed mice. Furthermore, the HFHC diet impaired aortic insulin signaling through Akt and forkhead box O1, which was associated with elevated endoplasmic reticulum stress markers CCAAT-enhancer-binding protein homologous protein and 78kDa glucose regulated protein. GW1516 intervention normalized mitogen-activated protein kinase activation, insulin signaling, and endoplasmic reticulum stress. CONCLUSIONS: Intervention with a PPARδ agonist inhibits aortic inflammation and attenuates the progression of pre-established atherosclerosis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Aortitis/prevention & control , Atherosclerosis/prevention & control , Insulin Resistance , PPAR delta/agonists , Receptors, LDL/deficiency , Thiazoles/pharmacology , Animals , Aortitis/blood , Aortitis/etiology , Aortitis/genetics , Aortitis/pathology , Atherosclerosis/blood , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Blood Glucose/metabolism , Cholesterol, Dietary , Diet, High-Fat , Disease Models, Animal , Dyslipidemias/blood , Dyslipidemias/drug therapy , Dyslipidemias/genetics , Dyslipidemias/metabolism , Inflammation Mediators/metabolism , Insulin/blood , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , PPAR delta/metabolism , Receptors, LDL/genetics , Signal Transduction/drug effects , Time Factors
16.
Arterioscler Thromb Vasc Biol ; 33(9): 2137-46, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23868935

ABSTRACT

OBJECTIVE: Low-density lipoprotein receptor-related protein 1 (LRP1) is a large endocytic and signaling receptor that is abundant in vascular smooth muscle cells. Mice in which the lrp1 gene is deleted in smooth muscle cells (smLRP1(-/-)) on a low-density lipoprotein receptor-deficient background display excessive platelet derived growth factor-signaling, smooth muscle cell proliferation, aneurysm formation, and increased susceptibility to atherosclerosis. The objectives of the current study were to examine the potential of LRP1 to modulate vascular physiology under nonatherogenic conditions. APPROACH AND RESULTS: We found smLRP1(-/-) mice to have extensive in vivo aortic dilatation accompanied by disorganized and degraded elastic lamina along with medial thickening of the arterial vessels resulting from excess matrix deposition. Surprisingly, this was not attributable to excessive platelet derived growth factor-signaling. Rather, quantitative differential proteomic analysis revealed that smLRP1(-/-) vessels contain a 4-fold increase in protein levels of high-temperature requirement factor A1 (HtrA1), which is a secreted serine protease that is known to degrade matrix components and to impair elastogenesis, resulting in fragmentation of elastic fibers. Importantly, our study discovered that HtrA1 is a novel LRP1 ligand. Proteomics analysis also identified excessive accumulation of connective tissue growth factor, an LRP1 ligand and a key mediator of fibrosis. CONCLUSIONS: Our findings suggest a critical role for LRP1 in maintaining the integrity of vessels by regulating protease activity as well as matrix deposition by modulating HtrA1 and connective tissue growth factor protein levels. This study highlights 2 new molecules, connective tissue growth factor and HtrA1, which contribute to detrimental changes in the vasculature and, therefore, represent new target molecules for potential therapeutic intervention to maintain vessel wall homeostasis.


Subject(s)
Aorta/enzymology , Aortitis/enzymology , Connective Tissue Growth Factor/metabolism , Myocytes, Smooth Muscle/enzymology , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Tumor Suppressor Proteins/metabolism , Age Factors , Aging , Animals , Aorta/physiopathology , Aorta/ultrastructure , Aortitis/genetics , Aortitis/pathology , Aortitis/physiopathology , Blood Pressure , Cells, Cultured , Dilatation, Pathologic , Elastic Tissue/metabolism , Endocytosis , Enzyme Activation , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , High-Temperature Requirement A Serine Peptidase 1 , Ligands , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Knockout , Proteomics/methods , Receptors, LDL/deficiency , Receptors, LDL/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
17.
Cardiovasc Res ; 120(10): 1202-1217, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38722818

ABSTRACT

AIMS: Abdominal aortic aneurysm (AAA) is a common, serious vascular disease with no effective pharmacological treatment. The nucleoside adenosine plays an important role in modulating vascular homeostasis, which prompted us to determine whether adenosine kinase (ADK), an adenosine metabolizing enzyme, modulates AAA formation via control of the intracellular adenosine level, and to investigate the underlying mechanisms. METHODS AND RESULTS: We used a combination of genetic and pharmacological approaches in murine models of AAA induced by calcium chloride (CaCl2) application or angiotensin II (Ang II) infusion to study the role of ADK in the development of AAA. In vitro functional assays were performed by knocking down ADK with adenovirus-short hairpin RNA in human vascular smooth muscle cells (VSMCs), and the molecular mechanisms underlying ADK function were investigated using RNA-sequencing, isotope tracing, and chromatin immunoprecipitation quantitative polymerase chain reaction (ChIP-qPCR). The heterozygous deficiency of ADK protected mice from CaCl2- and Ang II-induced AAA formation. Moreover, specific knockout of ADK in VSMCs prevented Ang II-induced AAA formation, as evidenced by reduced aortic extracellular elastin fragmentation, neovascularization, and aortic inflammation. Mechanistically, ADK knockdown in VSMCs markedly suppressed the expression of inflammatory genes associated with AAA formation, and these effects were independent of adenosine receptors. The metabolic flux and ChIP-qPCR results showed that ADK knockdown in VSMCs decreased S-adenosylmethionine (SAM)-dependent transmethylation, thereby reducing H3K4me3 binding to the promoter regions of the genes that are associated with inflammation, angiogenesis, and extracellular elastin fragmentation. Furthermore, the ADK inhibitor ABT702 protected mice from CaCl2-induced aortic inflammation, extracellular elastin fragmentation, and AAA formation. CONCLUSION: Our findings reveal a novel role for ADK inhibition in attenuating AAA via epigenetic modulation of key inflammatory genes linked to AAA pathogenesis.


Subject(s)
Adenosine Kinase , Aorta, Abdominal , Aortic Aneurysm, Abdominal , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Animals , Humans , Male , Mice , Adenosine/metabolism , Adenosine/analogs & derivatives , Adenosine Kinase/antagonists & inhibitors , Angiotensin II/metabolism , Aorta, Abdominal/pathology , Aorta, Abdominal/metabolism , Aorta, Abdominal/enzymology , Aortic Aneurysm, Abdominal/prevention & control , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/pathology , Aortic Aneurysm, Abdominal/enzymology , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/genetics , Aortitis/prevention & control , Aortitis/enzymology , Aortitis/pathology , Aortitis/metabolism , Aortitis/chemically induced , Aortitis/genetics , Calcium Chloride , Cells, Cultured , Disease Models, Animal , DNA Methylation , Epigenesis, Genetic , Inflammation Mediators/metabolism , Mice, Inbred C57BL , Morpholines , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines , Signal Transduction
18.
Sci Rep ; 9(1): 17067, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31745167

ABSTRACT

IL-25, a member of the IL-17 family of cytokines, is known to enhance type 2 immune responses, but suppress type 3 (IL-17A)-mediated immune responses. Mice deficient in IL-1 receptor antagonist (Il1rn-/- mice) have excessive IL-1 signaling, resulting in spontaneous development of IL-1-, TNF- and IL-17A-dependent aortitis. We found that expression of II25 mRNA was increased in the aortae of Il1rn-/- mice, suggesting that IL-25 may suppress development of IL-1-, TNF- and IL-17A-dependent aortitis in Il1rn-/- mice by inhibiting type 3-mediated immune responses. However, we unexpectedly found that Il25-/-Il1rn-/- mice showed attenuated development of aortitis, accompanied by reduced accumulation of inflammatory cells such as dendritic cells, macrophages and neutrophils and reduced mRNA expression of Il17a and Tnfa-but not Il4 or Il13-in local lesions compared with Il1rn-/- mice. Tissue-, but not immune cell-, derived IL-25 was crucial for development of aortitis. IL-25 enhanced IL-1ß and TNF production by IL-25 receptor-expressing dendritic cells and macrophages, respectively, at inflammatory sites of aortae of Il1rn-/- mice, contributing to exacerbation of development of IL-1-, TNF- and IL-17A-dependent aortitis in those mice. Our findings suggest that neutralization of IL-25 may be a potential therapeutic target for aortitis.


Subject(s)
Aortitis/immunology , Autoimmune Diseases/immunology , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukins/immunology , Animals , Aortitis/genetics , Aortitis/pathology , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , Dendritic Cells/immunology , Interleukin 1 Receptor Antagonist Protein/immunology , Interleukin-17/genetics , Interleukin-1beta/metabolism , Interleukins/genetics , Macrophages/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Neutrophils/immunology , RNA, Messenger/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
Cardiovasc Pathol ; 38: 1-6, 2019.
Article in English | MEDLINE | ID: mdl-30359839

ABSTRACT

Marfan syndrome (MFS) is a connective tissue disorder caused by mutations in the fibrillin-1 gene (FBN1), resulting in aortic aneurysm formation and dissections. Interestingly, variable aortopathy is observed even within MFS families with the same mutation. Thus, additional risk factors determine disease severity. Here, we describe a case of a 2-month-old Fbn1C1039G/+ MFS mouse with extreme aortic dilatation and increased vascular inflammation, when compared to MFS siblings, which coincided with unilateral renal cystic disease. In addition, this mouse presented with increased serum levels of creatinine, angiotensin-converting enzyme, corticosterone, macrophage chemoattractant protein-1, and interleukin-6, which may have contributed to the vascular pathology. Possibly, cystic kidney disease is associated with aneurysm progression in MFS patients. Therefore, we propose that close monitoring of the presence of renal cysts in MFS patients, during regular vascular imaging of the whole aorta trajectory, may provide insight in the frequency of cystic kidney disease and its potential as a novel indicator of aneurysm progression in MFS patients.


Subject(s)
Aorta/pathology , Aortic Aneurysm/etiology , Fibrillin-1/genetics , Kidney Diseases, Cystic/etiology , Marfan Syndrome/genetics , Animals , Aorta/metabolism , Aortic Aneurysm/blood , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Aortitis/blood , Aortitis/etiology , Aortitis/genetics , Aortitis/pathology , Biomarkers/blood , Dilatation, Pathologic , Disease Models, Animal , Fibrillin-1/metabolism , Genetic Predisposition to Disease , Kidney Diseases, Cystic/blood , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Male , Marfan Syndrome/blood , Marfan Syndrome/complications , Marfan Syndrome/diagnosis , Mice, Inbred C57BL , Mice, Transgenic , Phenotype
20.
Methods Mol Biol ; 1826: 157-182, 2018.
Article in English | MEDLINE | ID: mdl-30194600

ABSTRACT

Serpins have a wide range of functions in regulation of serine proteases in the thrombotic cascade and in immune responses, representing up to 2-10% of circulating proteins in the blood. Selected serpins also have cross-class inhibitory actions for cysteine proteases in inflammasome and apoptosis pathways. The arterial and venous systems transport blood throughout the mammalian body representing a central site for interactions between coagulation proteases and circulating blood cells (immune cells) and target tissues, a very extensive and complex interaction. While analysis of serpin functions in vitro in kinetics or gel shift assays or in tissue culture provides very necessary information on molecular mechanisms, the penultimate assessment of biological or physiological functions and efficacy for serpins as therapeutics requires study in vivo in whole animal models (some also consider cell culture to be an in vivo approach).Mouse models of arterial transplant with immune rejection as well as models of inflammatory vasculitis induced by infection have been used to study the interplay between the coagulation and immune response pathways. We describe here three in vivo vasculitis models that are used to study the roles of serpins in disease and as therapeutics. The models described include (1) mouse aortic allograft transplantation, (2) human temporal artery (TA) xenograft into immunodeficient mouse aorta, and (3) mouse herpes virus (MHV68)-induced inflammatory vasculitis in interferon-gamma receptor (IFNγR) knockout mice.


Subject(s)
Aortitis , Arteritis , Herpesviridae Infections , Rhadinovirus , Serpins , Allografts , Animals , Aorta/immunology , Aorta/pathology , Aorta/transplantation , Aortitis/genetics , Aortitis/immunology , Aortitis/pathology , Arteritis/genetics , Arteritis/immunology , Arteritis/pathology , Disease Models, Animal , Herpesviridae Infections/genetics , Herpesviridae Infections/immunology , Herpesviridae Infections/pathology , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Organ Transplantation , Rhadinovirus/genetics , Rhadinovirus/immunology , Serpins/genetics , Serpins/immunology , Temporal Arteries/immunology , Temporal Arteries/pathology , Temporal Arteries/transplantation
SELECTION OF CITATIONS
SEARCH DETAIL