Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Biol Chem ; 300(5): 107224, 2024 May.
Article in English | MEDLINE | ID: mdl-38537695

ABSTRACT

Impaired cholesterol efflux and/or uptake can influence arterial lipid accumulation leading to atherosclerosis. Here, we report that tripartite motif-containing protein 13 (TRIM13), a RING-type E3 ubiquitin ligase, plays a role in arterial lipid accumulation leading to atherosclerosis. Using molecular approaches and KO mouse model, we found that TRIM13 expression was induced both in the aorta and peritoneal macrophages (pMφ) of ApoE-/- mice in response to Western diet (WD) in vivo. Furthermore, proatherogenic cytokine interleukin-1ß also induced TRIM13 expression both in pMφ and vascular smooth muscle cells. Furthermore, we found that TRIM13 via ubiquitination and degradation of liver X receptor (LXR)α/ß downregulates the expression of their target genes ABCA1/G1 and thereby inhibits cholesterol efflux. In addition, TRIM13 by ubiquitinating and degrading suppressor of cytokine signaling 1/3 (SOCS1/3) mediates signal transducer and activator of transcription 1 (STAT1) activation, CD36 expression, and foam cell formation. In line with these observations, genetic deletion of TRIM13 by rescuing cholesterol efflux and inhibiting foam cell formation protects against diet-induced atherosclerosis. We also found that while TRIM13 and CD36 levels were increased, LXRα/ß, ABCA1/G1, and SOCS3 levels were decreased both in Mφ and smooth muscle cells of stenotic human coronary arteries as compared to nonstenotic arteries. More intriguingly, the expression levels of TRIM13 and its downstream signaling molecules were correlated with the severity of stenotic lesions. Together, these observations reveal for the first time that TRIM13 plays a crucial role in diet-induced atherosclerosis, and that it could be a potential drug target against this vascular lesion.


Subject(s)
Atherosclerosis , Cholesterol , Foam Cells , Lipoproteins, LDL , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Animals , Humans , Male , Mice , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Cholesterol/metabolism , Diet, Western/adverse effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Foam Cells/metabolism , Foam Cells/pathology , Lipoproteins, LDL/metabolism , Liver X Receptors/metabolism , Liver X Receptors/genetics , Mice, Knockout, ApoE , RAW 264.7 Cells , STAT1 Transcription Factor/metabolism , STAT1 Transcription Factor/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
2.
Arterioscler Thromb Vasc Biol ; 39(9): 1739-1746, 2019 09.
Article in English | MEDLINE | ID: mdl-31433698

ABSTRACT

While the opioid epidemic has garnered significant attention, the use of methamphetamines is growing worldwide independent of wealth or region. Following overdose and accidents, the leading cause of death in methamphetamine users is cardiovascular disease, because of significant effects of methamphetamine on vasoconstriction, pulmonary hypertension, atherosclerotic plaque formation, cardiac arrhythmias, and cardiomyopathy. In this review, we examine the current literature on methamphetamine-induced changes in cardiovascular health, discuss the potential mechanisms regulating these varied effects, and highlight our deficiencies in understanding how to treat methamphetamine-associated cardiovascular dysfunction.


Subject(s)
Cardiovascular Diseases/chemically induced , Methamphetamine/toxicity , Arrhythmias, Cardiac/chemically induced , Atherosclerosis/chemically induced , Cardiomyopathies/chemically induced , Humans , Hypertension, Pulmonary/chemically induced , Vasoconstriction/drug effects
3.
Arterioscler Thromb Vasc Biol ; 39(6): 1212-1226, 2019 06.
Article in English | MEDLINE | ID: mdl-31043075

ABSTRACT

Objective- IL (interleukin)-33 has been shown to play a role in endothelial dysfunction, but its role in atherosclerosis is controversial. Therefore, the purpose of this study is to examine its role in vascular wall remodeling following injury. Approach and Results- Thrombin induced IL-33 expression in a time-dependent manner in human aortic smooth muscle cells and inhibition of its activity by its neutralizing antibody suppressed thrombin induced human aortic smooth muscle cell migration but not DNA synthesis. In exploring the mechanisms, we found that Par1 (protease-activated receptor 1), Gαq/11 (Gα protein q/11), PLCß3 (phospholipase Cß3), NFATc1 (nuclear factor of activated T cells), E2F1 (E2F transcription factor 1), and LMCD1 (LIM and cysteine-rich domains protein 1) are involved in thrombin-induced IL-33 expression and migration. Furthermore, we identified an NFAT-binding site at -100 nt that mediates thrombin-induced IL-33 promoter activity. Interestingly, we observed that NFATc1, E2F1, and LMCD1 bind to NFAT site in response to thrombin and found that LMCD1, while alone has no significant effect, enhanced either NFATc1 or E2F1-dependent IL-33 promoter activity. In addition, we found that guidewire injury induces IL-33 expression in SMC and its neutralizing antibodies substantially reduce SMC migration and neointimal growth in vivo. Increased expression of IL-33 was also observed in human atherosclerotic lesions as compared to arteries without any lesions. Conclusions- The above findings reveal for the first time that thrombin-induced human aortic smooth muscle cell migration and injury-induced neointimal growth require IL-33 expression. In addition, thrombin-induced IL-33 expression requires LMCD1 enhanced combinatorial activation of NFATc1 and E2F1.


Subject(s)
Co-Repressor Proteins/metabolism , E2F1 Transcription Factor/metabolism , Interleukin-33/metabolism , LIM Domain Proteins/metabolism , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , NFATC Transcription Factors/metabolism , Neointima , Vascular System Injuries/metabolism , Animals , Binding Sites , Cell Movement/drug effects , Cell Proliferation/drug effects , Co-Repressor Proteins/genetics , Disease Models, Animal , E2F1 Transcription Factor/genetics , Female , Femoral Artery/drug effects , Femoral Artery/injuries , Femoral Artery/metabolism , Femoral Artery/pathology , HEK293 Cells , Humans , Interleukin-33/genetics , LIM Domain Proteins/genetics , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , NFATC Transcription Factors/genetics , Promoter Regions, Genetic , Signal Transduction , Up-Regulation , Vascular System Injuries/genetics , Vascular System Injuries/pathology
4.
J Biol Chem ; 293(9): 3088-3103, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29326163

ABSTRACT

Restenosis arises after vascular injury and is characterized by arterial wall thickening and decreased arterial lumen space. Vascular injury induces the production of thrombin, which in addition to its role in blood clotting acts as a mitogenic and chemotactic factor. In exploring the molecular mechanisms underlying restenosis, here we identified LMCD1 (LIM and cysteine-rich domains 1) as a gene highly responsive to thrombin in human aortic smooth muscle cells (HASMCs). Of note, LMCD1 depletion inhibited proliferation of human but not murine vascular smooth muscle cells. We also found that by physically interacting with E2F transcription factor 1, LMCD1 mediates thrombin-induced expression of the CDC6 (cell division cycle 6) gene in the stimulation of HASMC proliferation. Thrombin-induced LMCD1 and CDC6 expression exhibited a requirement for protease-activated receptor 1-mediated Gαq/11-dependent activation of phospholipase C ß3. Moreover, the expression of LMCD1 was highly induced in smooth muscle cells located at human atherosclerotic lesions and correlated with CDC6 expression and that of the proliferation marker Ki67. Furthermore, the LMCD1- and SMCαactin-positive cells had higher cholesterol levels in the atherosclerotic lesions. In conclusion, these findings indicate that by acting as a co-activator with E2F transcription factor 1 in CDC6 expression, LMCD1 stimulates HASMC proliferation and thereby promotes human atherogenesis, suggesting an involvement of LMCD1 in restenosis.


Subject(s)
Atherosclerosis/metabolism , Co-Repressor Proteins/metabolism , LIM Domain Proteins/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Thrombin/pharmacology , Adult , Aged , Animals , Female , Humans , Male , Mice , Middle Aged , Rats , Young Adult
5.
Circulation ; 136(6): 566-582, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28487392

ABSTRACT

BACKGROUND: Atherosclerotic plaque formation results from chronic inflammation and fibroproliferative remodeling in the vascular wall. We previously demonstrated that both human and mouse atherosclerotic plaques show elevated expression of EphA2, a guidance molecule involved in cell-cell interactions and tumorigenesis. METHODS: Here, we assessed the role of EphA2 in atherosclerosis by deleting EphA2 in a mouse model of atherosclerosis (Apoe-/-) and by assessing EphA2 function in multiple vascular cell culture models. After 8 to 16 weeks on a Western diet, male and female mice were assessed for atherosclerotic burden in the large vessels, and plasma lipid levels were analyzed. RESULTS: Despite enhanced weight gain and plasma lipid levels compared with Apoe-/- controls, EphA2-/-Apoe-/- knockout mice show diminished atherosclerotic plaque formation, characterized by reduced proinflammatory gene expression and plaque macrophage content. Although plaque macrophages express EphA2, EphA2 deletion does not affect macrophage phenotype, inflammatory responses, and lipid uptake, and bone marrow chimeras suggest that hematopoietic EphA2 deletion does not affect plaque formation. In contrast, endothelial EphA2 knockdown significantly reduces monocyte firm adhesion under flow. In addition, EphA2-/-Apoe-/- mice show reduced progression to advanced atherosclerotic plaques with diminished smooth muscle and collagen content. Consistent with this phenotype, EphA2 shows enhanced expression after smooth muscle transition to a synthetic phenotype, and EphA2 depletion reduces smooth muscle proliferation, mitogenic signaling, and extracellular matrix deposition both in atherosclerotic plaques and in vascular smooth muscle cells in culture. CONCLUSIONS: Together, these data identify a novel role for EphA2 in atherosclerosis, regulating both plaque inflammation and progression to advanced atherosclerotic lesions. Cell culture studies suggest that endothelial EphA2 contributes to atherosclerotic inflammation by promoting monocyte firm adhesion, whereas smooth muscle EphA2 expression may regulate the progression to advanced atherosclerosis by regulating smooth muscle proliferation and extracellular matrix deposition.


Subject(s)
Atherosclerosis/pathology , Receptor, EphA2/genetics , Animals , Aorta/metabolism , Aorta/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Cell Lineage , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Inflammation , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phenotype , Plaque, Atherosclerotic/pathology , Receptor, EphA2/deficiency , Receptor, EphA2/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
6.
J Vasc Res ; 55(2): 98-110, 2018.
Article in English | MEDLINE | ID: mdl-29455203

ABSTRACT

BACKGROUND: Mitochondrial reactive oxygen species (ROS) contribute to inflammation and vascular remodeling during atherosclerotic plaque formation. C57BL/6N (6N) and C57BL/6J (6J) mice display distinct mitochondrial redox balance due to the absence of nicotinamide nucleotide transhydrogenase (NNT) in 6J mice. We hypothesize that differential NNT expression between these animals alters plaque development. METHODS: 6N and 6J mice were treated with AAV8-PCSK9 (adeno-associated virus serotype 8/proprotein convertase subtilisin/kexin type 9) virus leading to hypercholesterolemia, increased low-density lipoprotein, and atherosclerosis in mice fed a high-fat diet (HFD). Mice were co-treated with the mitochondria-targeted superoxide dismutase mimetic MitoTEMPO to assess the contribution of mitochondrial ROS to atherosclerosis. RESULTS: Baseline and HFD-induced vascular superoxide is increased in 6J compared to 6N mice. MitoTEMPO diminished superoxide in both groups demonstrating differential production of mitochondrial ROS among these strains. PCSK9 treatment and HFD led to similar increases in plasma lipids in both 6N and 6J mice. However, 6J animals displayed significantly higher levels of plaque formation. MitoTEMPO reduced plasma lipids but did not affect plaque formation in 6N mice. In contrast, MitoTEMPO surprisingly increased plaque formation in 6J mice. CONCLUSION: These data indicate that loss of NNT increases vascular ROS production and exacerbates atherosclerotic plaque development.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , NADP Transhydrogenase, AB-Specific/deficiency , Animals , Antioxidants/pharmacology , Aorta/drug effects , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cholesterol/blood , Disease Models, Animal , Genetic Predisposition to Disease , Hypercholesterolemia/enzymology , Hypercholesterolemia/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , NADP Transhydrogenase, AB-Specific/genetics , Organophosphorus Compounds/pharmacology , Phenotype , Piperidines/pharmacology , Plaque, Atherosclerotic , Proprotein Convertase 9/genetics , Proprotein Convertase 9/metabolism , Superoxides/metabolism , Time Factors
7.
Arterioscler Thromb Vasc Biol ; 33(10): 2316-24, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23950143

ABSTRACT

OBJECTIVE: Interleukin-19 (IL-19) is a putative Th2, anti-inflammatory interleukin. Its expression and potential role in atherogenesis are unknown. IL-19 is not detected in normal artery and is expressed to a greater degree in plaque from symptomatic versus asymptomatic patients, suggesting a compensatory counter-regulatory function. We tested whether IL-19 could reduce atherosclerosis in susceptible mice and identified plausible mechanisms. APPROACH AND RESULTS: LDLR(-/-) mice fed an atherogenic diet and injected with either 1.0 or 10.0 ng/g per day recombinant mouse IL-19 had significantly less plaque area in the aortic arch compared with controls (P<0.0001). Weight gain, cholesterol, and triglyceride levels were not significantly different. Gene expression in splenocytes from IL-19-treated mice demonstrated immune cell Th2 polarization, with decreased expression of T-bet, interferon-γ, interleukin-1ß, and interleukin-12ß and increased expression of GATA3 and FoxP3 mRNA. A greater percentage of lymphocytes were Th2 polarized in IL-19-treated mice. Cellular characterization of plaque by immunohistochemistry demonstrated that IL-19-treated mice have significantly less macrophage infiltrate compared with controls (P<0.001). Intravital microscopy revealed significantly less leukocyte adhesion in wild-type mice injected with IL-19 and fed an atherogenic diet compared with controls. Treatment of cultured endothelial cells, vascular smooth muscle cells, and bone marrow-derived macrophages with IL-19 resulted in a significant decrease in chemokine mRNA and mRNA stability protein human antigen R. CONCLUSIONS: These data suggest that IL-19 is a potent inhibitor of experimental atherosclerosis, with diverse mechanisms including immune cell polarization, decrease in macrophage adhesion, and decrease in gene expression. This may identify IL-19 as a novel therapeutic to limit vascular inflammation.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Interleukin-10/pharmacology , Aged , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/immunology , Aortic Diseases/metabolism , Aortic Diseases/pathology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/immunology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biomarkers/blood , Carotid Artery Diseases/immunology , Carotid Artery Diseases/pathology , Cells, Cultured , Cholesterol/blood , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Gene Expression Regulation/drug effects , Humans , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Interleukins/metabolism , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/immunology , Plaque, Atherosclerotic , Receptors, LDL/deficiency , Receptors, LDL/genetics , Recombinant Proteins/pharmacology , Spleen/drug effects , Spleen/immunology , Th2 Cells/drug effects , Th2 Cells/immunology , Time Factors , Triglycerides/blood
8.
bioRxiv ; 2024 Jan 21.
Article in English | MEDLINE | ID: mdl-38293157

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality worldwide1. Laminar shear stress (LSS) from blood flow in straight regions of arteries protects against ASCVD by upregulating the Klf2/4 anti-inflammatory program in endothelial cells (ECs)2-8. Conversely, disturbed shear stress (DSS) at curves or branches predisposes these regions to plaque formation9,10. We previously reported a whole genome CRISPR knockout screen11 that identified novel inducers of Klf2/4. Here we report suppressors of Klf2/4 and characterize one candidate, protocadherin gamma A9 (Pcdhga9), a member of the clustered protocadherin gene family12. Pcdhg deletion increases Klf2/4 levels in vitro and in vivo and suppresses inflammatory activation of ECs. Pcdhg suppresses Klf2/4 by inhibiting the Notch pathway via physical interaction of cleaved Notch1 intracellular domain (NICD Val1744) with nuclear Pcdhg C-terminal constant domain (CCD). Pcdhg inhibition by EC knockout (KO) or blocking antibody protects from atherosclerosis. Pcdhg is elevated in the arteries of human atherosclerosis. This study identifies a novel fundamental mechanism of EC resilience and therapeutic target for treating inflammatory vascular disease.

9.
Nat Cardiovasc Res ; 3(9): 1035-1048, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39232138

ABSTRACT

Atherosclerotic cardiovascular disease (ASCVD) is the leading cause of mortality worldwide. Laminar shear stress from blood flow, sensed by vascular endothelial cells, protects from ASCVD by upregulating the transcription factors KLF2 and KLF4, which induces an anti-inflammatory program that promotes vascular resilience. Here we identify clustered γ-protocadherins as therapeutically targetable, potent KLF2 and KLF4 suppressors whose upregulation contributes to ASCVD. Mechanistic studies show that γ-protocadherin cleavage results in translocation of the conserved intracellular domain to the nucleus where it physically associates with and suppresses signaling by the Notch intracellular domain. γ-Protocadherins are elevated in human ASCVD endothelium; their genetic deletion or antibody blockade protects from ASCVD in mice without detectably compromising host defense against bacterial or viral infection. These results elucidate a fundamental mechanism of vascular inflammation and reveal a method to target the endothelium rather than the immune system as a protective strategy in ASCVD.


Subject(s)
Atherosclerosis , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors , Atherosclerosis/metabolism , Atherosclerosis/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Animals , Humans , Disease Models, Animal , Signal Transduction , Cadherins/metabolism , Cadherins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Male , Receptors, Notch/metabolism , Receptors, Notch/genetics , Cadherin Related Proteins , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology
10.
bioRxiv ; 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39071358

ABSTRACT

Macrophage efferocytosis, the process by which phagocytes engulf and remove apoptotic cells (ACs), plays a critical role in maintaining tissue homeostasis. Efficient efferocytosis prevents secondary necrosis, mitigates chronic inflammation, and impedes atherosclerosis progression. However, the regulatory mechanisms of efferocytosis under atherogenic conditions remain poorly understood. We previously demonstrated that oxidized LDL (oxLDL), an atherogenic lipoprotein, induces mitochondrial reactive oxygen species (mtROS) in macrophages via CD36. In this study, we demonstrate that macrophage mtROS facilitate continual efferocytosis through a positive feedback mechanism. However, oxLDL disrupts continual efferocytosis by dysregulating the internalization of ACs. This disruption is mediated by an overproduction of mtROS. Mechanistically, oxLDL/CD36 signaling promotes the translocation of cytosolic PKM2 to mitochondria, facilitated by the chaperone GRP75. Mitochondrial PKM2 then binds to Complex III of the electron transport chain, inducing mtROS production. This study elucidates a novel regulatory mechanism of efferocytosis in atherosclerosis, providing potential therapeutic targets for intervention. SUMMARY: Macrophages clear apoptotic cells through a process called efferocytosis, which involves mitochondrial ROS. However, the atherogenic oxidized LDL overstimulates mitochondrial ROS via the CD36-PKM2 pathway, disrupting continual efferocytosis. This finding elucidates a novel molecular mechanism that explains defects in efferocytosis, driving atherosclerosis progression.

11.
Arterioscler Thromb Vasc Biol ; 32(3): 686-95, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22247258

ABSTRACT

OBJECTIVE: Endothelial cell activation results in altered cell-cell interactions with adjacent endothelial cells and with infiltrating leukocytes. Eph receptors and their ephrin ligands regulate cell-cell interactions during tissue remodeling, and multiple proinflammatory mediators induce endothelial EphA receptor and ephrinA ligand expression. Therefore, we sought to elucidate the role of EphA receptors and ephrinA ligands in endothelial cell activation and atherosclerosis. METHODS AND RESULTS: Quantitative reverse transcription-polymerase chain reaction screening for EphA/ephrinA expression in atherosclerosis-prone macrovascular endothelium identified EphA2, EphA4, and ephrinA1 as the dominant isoforms. Endothelial activation with oxidized low-density lipoprotein and proinflammatory cytokines induced EphA2 and ephrinA1 expression and sustained EphA2 activation, whereas EphA4 expression was unaffected. Atherosclerotic plaques from mice and humans showed enhanced EphA2 and ephrinA1 expression colocalizing in the endothelial cell layer. EphA2 activation with recombinant Fc-ephrinA1 induced proinflammatory gene expression (eg vascular cell adhesion molecule-1, E-selectin) and stimulated monocyte adhesion, whereas inhibiting EphA2 (small interfering RNA, pharmacological inhibitors) abrogated both ephrinA1-induced and oxidized low-density lipoprotein-induced vascular cell adhesion molecule-1 expression. CONCLUSION: The current data suggest that enhanced EphA2 signaling during endothelial cell activation perpetuates proinflammatory gene expression. Coupled with EphA2 expression in mouse and human atherosclerotic plaques, these data implicate EphA2 as a novel proinflammatory mediator and potential regulator of atherosclerotic plaque development.


Subject(s)
Atherosclerosis/immunology , Endothelial Cells/immunology , Inflammation Mediators/metabolism , Inflammation/immunology , Receptor, EphA2/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cell Adhesion , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/antagonists & inhibitors , Lipoproteins, LDL/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Phenotype , RNA Interference , Receptor, EphA2/antagonists & inhibitors , Receptor, EphA2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transfection
12.
Arterioscler Thromb Vasc Biol ; 32(8): e51-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22723438

ABSTRACT

OBJECTIVE: Upregulated expression of endothelial adhesion molecules and subsequent binding to cognate monocytic receptors are established paradigms in atherosclerosis. However, these proteins are the scaffolds, with their posttranslational modification with sugars providing the actual ligands. We recently showed that tumor necrosis factor-α increased hypoglycosylated (mannose-rich) N-glycans on the endothelial surface. In the present study, our aim was to determine whether (1) hypoglycosylated N-glycans are upregulated by proatherogenic stimuli (oscillatory flow) in vitro and in vivo, and (2) mannose residues on hypoglycosylated endothelial N-glycans mediate monocyte rolling and adhesion. METHODS AND RESULTS: Staining with the mannose-specific lectins concanavalin A and lens culinaris agglutinin was increased in human aortic endothelial cells exposed to oscillatory shear or tumor necrosis factor-α and at sites of plaque development and progression in both mice and human vessels. Increasing surface N-linked mannose by inhibiting N-glycan processing potentiated monocyte adhesion under flow during tumor necrosis factor-α stimulation. Conversely, enzymatic removal of high-mannose N-glycans, or masking mannose residues with lectins, significantly decreased monocyte adhesion under flow. These effects occurred without altering induced expression of adhesion molecule proteins. CONCLUSIONS: Hypoglycosylated (high mannose) N-glycans are present on the endothelial cell surface at sites of early human lesion development and are novel effectors of monocyte adhesion during atherogenesis.


Subject(s)
Atherosclerosis/pathology , Endothelial Cells/physiology , Mannose/physiology , Monocytes/physiology , Polysaccharides/chemistry , Atherosclerosis/metabolism , Cell Adhesion , Cell Communication , Cell Movement , Glycosylation , Humans , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/physiology , alpha-Mannosidase/antagonists & inhibitors
13.
Redox Biol ; 57: 102480, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36167027

ABSTRACT

Methamphetamine (METH) is an addictive illicit drug used worldwide that causes significant damage to blood vessels resulting in cardiovascular dysfunction. Recent studies highlight increased prevalence of cardiovascular disease (CVD) and associated complications including hypertension, vasospasm, left ventricular hypertrophy, and coronary artery disease in younger populations due to METH use. Here we report that METH administration in a mouse model of 'binge and crash' decreases cardiovascular function via cystathionine gamma lyase (CSE), hydrogen sulfide (H2S), nitric oxide (NO) (CSE/H2S/NO) dependent pathway. METH significantly reduced H2S and NO bioavailability in plasma and skeletal muscle tissues co-incident with a significant reduction in flow-mediated vasodilation (FMD) and blood flow velocity revealing endothelial dysfunction. METH administration also reduced cardiac ejection fraction (EF) and fractional shortening (FS) associated with increased tissue and perivascular fibrosis. Importantly, METH treatment selectively decreased CSE expression and sulfide bioavailability along with reduced eNOS phosphorylation and NO levels. Exogenous sulfide therapy or endothelial CSE transgenic overexpression corrected cardiovascular and associated pathological responses due to METH implicating a central molecular regulatory pathway for tissue pathology. These findings reveal that therapeutic intervention targeting CSE/H2S bioavailability may be useful in attenuating METH mediated cardiovascular disease.

14.
Forensic Sci Med Pathol ; 7(3): 283-6, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21305390

ABSTRACT

We report a case of acute bronchopneumonia and esophagotracheal fistula caused by a swallowed button battery in a 3-year-old girl. It was unclear exactly how long the battery had been trapped in the esophagus. The patient had undergone a tonsillectomy and adenoidectomy 3 weeks before the battery was finally exposed on an X-ray film. She refused to eat solid food after the surgery and stopped eating completely 10 days later. Three weeks after surgery, she presented to the Emergency Department with vomiting and acute respiratory distress, experienced cardiopulmonary arrest in the intensive care unit and could not be resuscitated. Postmortem examination revealed severe acute bronchopneumonia and massive blood aspiration due to an esophagotracheal fistula secondary to a button battery lodged in the esophagus. This case highlights the importance of including a swallowed button battery in the differential diagnosis of a toddler with dysphagia and anorexia.


Subject(s)
Foreign Bodies/complications , Pneumonia, Aspiration/etiology , Tracheoesophageal Fistula/etiology , Anorexia/etiology , Child, Preschool , Deglutition Disorders/etiology , Female , Forensic Pathology , Heart Arrest/etiology , Humans , Lung/pathology , Vomiting/etiology
15.
Cell Death Differ ; 28(2): 780-798, 2021 02.
Article in English | MEDLINE | ID: mdl-32968199

ABSTRACT

ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1) play a vital role in promoting cholesterol efflux. Although, the dysregulation of these transporters was attributed as one of the mechanisms of atherogenesis, what renders their dysfunction is not well explored. Previously, we have reported that thrombin without having any effect on ABCG1 levels depletes ABCA1 levels affecting cholesterol efflux. In this study, we explored the mechanisms underlying thrombin-induced depletion of ABCA1 levels both in macrophages and smooth muscle cells. Under normal physiological conditions, COP9 signalosome subunit 3 (CSN3) was found to exist in complex with ABCA1 and in the presence of proatherogenic stimulants such as thrombin, ABCA1 was phosphorylated and dissociated from CSN3, leading to its degradation. Forced expression of CSN3 inhibited thrombin-induced ABCA1 ubiquitination and degradation, restored cholesterol efflux and suppressed foam cell formation. In Western diet (WD)-fed ApoE-/- mice, CSN3 was also disassociated from ABCA1 otherwise remained as a complex in Chow diet (CD)-fed ApoE-/- mice. Interestingly, depletion of CSN3 levels in WD-fed ApoE-/- mice significantly lowered ABCA1 levels, inhibited cholesterol efflux and intensified foam cell formation exacerbating the lipid laden atherosclerotic plaque formation. Mechanistic studies have revealed the involvement of Par1-Gα12-Pyk2-Gab1-PKCθ signaling in triggering phosphorylation of ABCA1 and its disassociation from CSN3 curtailing cholesterol efflux and amplifying foam cell formation. In addition, although both CSN3 and ABCA1 were found to be colocalized in human non-lesion coronary arteries, their levels were decreased as well as dissociated from each other in advanced atherosclerotic lesions. Together, these observations reveal for the first time an anti-atherogenic role of CSN3 and hence, designing therapeutic drugs protecting its interactions with ABCA1 could be beneficial against atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/metabolism , Apolipoproteins E/physiology , Atherosclerosis/pathology , COP9 Signalosome Complex/metabolism , Macrophages, Peritoneal/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, PAR-1/physiology , ATP Binding Cassette Transporter 1/genetics , Animals , Atherosclerosis/etiology , Atherosclerosis/metabolism , COP9 Signalosome Complex/genetics , Cholesterol/metabolism , Diet, Western/adverse effects , Female , Foam Cells/metabolism , Foam Cells/pathology , Humans , Macrophages, Peritoneal/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Proto-Oncogene Proteins/genetics , RAW 264.7 Cells , Signal Transduction , Thrombin/metabolism
16.
J Clin Invest ; 130(8): 4331-4347, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32427580

ABSTRACT

Although the Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS) established the role of treating inflammation in atherosclerosis, our understanding of endothelial activation at atherosclerosis-prone sites remains limited. Disturbed flow at atheroprone regions primes plaque inflammation by enhancing endothelial NF-κB signaling. Herein, we demonstrate a role for the Nck adaptor proteins in disturbed flow-induced endothelial activation. Although highly similar, only Nck1 deletion, but not Nck2 deletion, limited flow-induced NF-κB activation and proinflammatory gene expression. Nck1-knockout mice showed reduced endothelial activation and inflammation in both models, disturbed flow- and high fat diet-induced atherosclerosis, whereas Nck2 deletion did not. Bone marrow chimeras confirmed that vascular Nck1, but not hematopoietic Nck1, mediated this effect. Domain-swap experiments and point mutations identified the Nck1 SH2 domain and the first SH3 domain as critical for flow-induced endothelial activation. We further characterized Nck1's proinflammatory role by identifying interleukin 1 type I receptor kinase-1 (IRAK-1) as a Nck1-selective binding partner, demonstrating that IRAK-1 activation by disturbed flow required Nck1 in vitro and in vivo, showing endothelial Nck1 and IRAK-1 staining in early human atherosclerosis, and demonstrating that disturbed flow-induced endothelial activation required IRAK-1. Taken together, our data reveal a hitherto unknown link between Nck1 and IRAK-1 in atherogenic inflammation.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/metabolism , Oncogene Proteins/metabolism , Plaque, Atherosclerotic/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Cell Line , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Knockout , Oncogene Proteins/genetics , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/pathology , Point Mutation , src Homology Domains
18.
Nat Commun ; 6: 7450, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26104863

ABSTRACT

Pak1 plays an important role in various cellular processes, including cell motility, polarity, survival and proliferation. To date, its role in atherogenesis has not been explored. Here we report the effect of Pak1 on atherogenesis using atherosclerosis-prone apolipoprotein E-deficient (ApoE(-/-)) mice as a model. Disruption of Pak1 in ApoE(-/-) mice results in reduced plaque burden, significantly attenuates circulating IL-6 and MCP-1 levels, limits the expression of adhesion molecules and diminishes the macrophage content in the aortic root of ApoE(-/-) mice. We also observed reduced oxidized LDL uptake and increased cholesterol efflux by macrophages and smooth muscle cells of ApoE(-/-):Pak1(-/-) mice as compared with ApoE(-/-) mice. In addition, we detect increased Pak1 phosphorylation in human atherosclerotic arteries, suggesting its role in human atherogenesis. Altogether, these results identify Pak1 as an important factor in the initiation and progression of atherogenesis.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/genetics , Macrophages, Peritoneal/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/genetics , p21-Activated Kinases/genetics , p21-Activated Kinases/metabolism , Adult , Aged , Animals , Arteries/metabolism , Atherosclerosis/metabolism , Blotting, Western , Cell Adhesion , Cell Movement , Chemokine CCL2/metabolism , Cholesterol/metabolism , Female , Humans , Interleukin-6/metabolism , Lipoproteins, LDL/metabolism , Macrophages/metabolism , Male , Mice , Mice, Knockout , Middle Aged , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Plaque, Atherosclerotic/metabolism , Young Adult
19.
Free Radic Biol Med ; 76: 147-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25152235

ABSTRACT

15(S)-Hydroxyeicosatetraenoic acid (15(S)-HETE), the major 15-lipoxygenase 1/2 (15-LO1/2) metabolite of arachidonic acid (AA), induces CD36 expression through xanthine oxidase and NADPH oxidase-dependent ROS production and Syk and Pyk2-dependent STAT1 activation. In line with these observations, 15(S)-HETE also induced foam cell formation involving ROS, Syk, Pyk2, and STAT1-mediated CD36 expression. In addition, peritoneal macrophages from Western diet-fed ApoE(-/-) mice exhibited elevated levels of xanthine oxidase and NADPH oxidase activities, ROS production, Syk, Pyk2, and STAT1 phosphorylation, and CD36 expression compared to those from ApoE(-/-):12/15-LO(-/-) mice and these events correlated with increased lipid deposits, macrophage content, and lesion progression in the aortic roots. Human atherosclerotic arteries also showed increased 15-LO1 expression, STAT1 phosphorylation, and CD36 levels as compared to normal arteries. Together, these findings suggest that 12/15-LO metabolites of AA, particularly 12/15(S)-HETE, might play a crucial role in atherogenesis by enhancing foam cell formation.


Subject(s)
CD36 Antigens/metabolism , Foam Cells/cytology , Focal Adhesion Kinase 2/metabolism , Hydroxyeicosatetraenoic Acids/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/metabolism , Animals , Apolipoproteins E/physiology , Arachidonate 12-Lipoxygenase/chemistry , Arachidonate 12-Lipoxygenase/physiology , Arachidonate 15-Lipoxygenase/chemistry , Arachidonate 15-Lipoxygenase/physiology , Blotting, Western , CD36 Antigens/genetics , Cells, Cultured , Chromatin Immunoprecipitation , Coronary Artery Disease/drug therapy , Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Electrophoretic Mobility Shift Assay , Foam Cells/drug effects , Foam Cells/metabolism , Focal Adhesion Kinase 2/genetics , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Lipids/analysis , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Knockout , NADPH Oxidases/metabolism , Phosphorylation/drug effects , Promoter Regions, Genetic/genetics , Protein-Tyrosine Kinases/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , Signal Transduction/drug effects , Syk Kinase
SELECTION OF CITATIONS
SEARCH DETAIL