Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Arterioscler Thromb Vasc Biol ; 38(2): 324-334, 2018 02.
Article in English | MEDLINE | ID: mdl-29217509

ABSTRACT

OBJECTIVE: Macrophage proinflammatory responses induced by modified low-density lipoproteins (modLDL) contribute to atherosclerotic progression. How modLDL causes macrophages to become proinflammatory is still enigmatic. Macrophage foam cell formation induced by modLDL requires glycerolipid synthesis. Lipin-1, a key enzyme in the glycerolipid synthesis pathway, contributes to modLDL-elicited macrophage proinflammatory responses in vitro. The objective of this study was to determine whether macrophage-associated lipin-1 contributes to atherogenesis and to assess its role in modLDL-mediated signaling in macrophages. APPROACH AND RESULTS: We developed mice lacking lipin-1 in myeloid-derived cells and used adeno-associated viral vector 8 expressing the gain-of-function mutation of mouse proprotein convertase subtilisin/kexin type 9 (adeno-associated viral vector 8-proprotein convertase subtilisin/kexin type 9) to induce hypercholesterolemia and plaque formation. Mice lacking myeloid-associated lipin-1 had reduced atherosclerotic burden compared with control mice despite similar plasma lipid levels. Stimulation of bone marrow-derived macrophages with modLDL activated a persistent protein kinase Cα/ßII-extracellular receptor kinase1/2-jun proto-oncogene signaling cascade that contributed to macrophage proinflammatory responses that was dependent on lipin-1 enzymatic activity. CONCLUSIONS: Our data demonstrate that macrophage-associated lipin-1 is atherogenic, likely through persistent activation of a protein kinase Cα/ßII-extracellular receptor kinase1/2-jun proto-oncogene signaling cascade that contributes to foam cell proinflammatory responses. Taken together, these results suggest that modLDL-induced foam cell formation and modLDL-induced macrophage proinflammatory responses are not independent consequences of modLDL stimulation but rather are both directly influenced by enhanced lipid synthesis.


Subject(s)
Aorta/enzymology , Aortic Diseases/enzymology , Atherosclerosis/enzymology , Inflammation Mediators/metabolism , Inflammation/enzymology , Lipoproteins, LDL/blood , Macrophages/enzymology , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/metabolism , Animals , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Foam Cells/enzymology , Foam Cells/pathology , Inflammation/genetics , Inflammation/pathology , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Phosphatidate Phosphatase/deficiency , Phosphatidate Phosphatase/genetics , Plaque, Atherosclerotic , Protein Kinase C beta/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RAW 264.7 Cells , Signal Transduction
3.
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
4.
Am J Physiol Renal Physiol ; 310(10): F1123-35, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26936875

ABSTRACT

Previous research has shown that podocytes unable to assemble heparan sulfate on cell surface proteoglycan core proteins have compromised cell-matrix interactions. This report further explores the role of N-sulfation of intact heparan chains in podocyte-matrix interactions. For the purposes of this study, a murine model in which the enzyme N-deacetylase/N-sulfotransferase 1 (NDST1) was specifically deleted in podocytes and immortalized podocyte cell lines lacking NDST1 were developed and used to explore the effects of such a mutation on podocyte behavior in vitro. NDST1 is a bifunctional enzyme, ultimately responsible for N-sulfation of heparan glycosaminoglycans produced by cells. Immunostaining of glomeruli from mice whose podocytes were null for Ndst1 (Ndst1(-/-)) showed a disrupted pattern of localization for the cell surface proteoglycan, syndecan-4, and for α-actinin-4 compared with controls. The pattern of immunostaining for synaptopodin and nephrin did not show as significant alterations. In vitro studies showed that Ndst1(-/-) podocytes attached, spread, and migrated less efficiently than Ndst1(+/+) podocytes. Immunostaining in vitro for several markers for molecules involved in cell-matrix interactions showed that Ndst1(-/-) cells had decreased clustering of syndecan-4 and decreased recruitment of protein kinase-Cα, α-actinin-4, vinculin, and phospho-focal adhesion kinase to focal adhesions. Total intracellular phospho-focal adhesion kinase was decreased in Ndst1(-/-) compared with Ndst1(+/+) cells. A significant decrease in the abundance of activated integrin α5ß1 on the cell surface of Ndst1(-/-) cells compared with Ndst1(+/+) cells was observed. These results serve to highlight the critical role of heparan sulfate N-sulfation in facilitating normal podocyte-matrix interactions.


Subject(s)
Extracellular Matrix/metabolism , Heparitin Sulfate/metabolism , Podocytes/metabolism , Sulfotransferases/genetics , Syndecan-4/metabolism , Actinin/metabolism , Animals , Cell Adhesion , Cell Movement , Cells, Cultured , Cytoskeleton/metabolism , Disease Models, Animal , Focal Adhesions/metabolism , Glomerular Basement Membrane/metabolism , Integrin alpha5beta1/metabolism , Mice , Mice, Transgenic
5.
Atherosclerosis ; 242(2): 424-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26288136

ABSTRACT

Atherosclerosis is a chronic inflammatory disease of large and medium-sized arteries and the underlying cause of cardiovascular disease, a major cause of mortality worldwide. The over-accumulation of modified cholesterol-containing low-density lipoproteins (e.g. oxLDL) in the artery wall and the subsequent recruitment and activation of macrophages contributes to the development of atherosclerosis. The excessive uptake of modified-LDL by macrophages leads to a lipid-laden "foamy" phenotype and pro-inflammatory cytokine production. Modified-LDLs promote foam cell formation in part by stimulating de novo lipid biosynthesis. However, it is unknown if lipid biosynthesis directly regulates foam cell pro-inflammatory mediator production. Lipin-1, a phosphatidate phosphohydrolase required for the generation of diacylglycerol during glycerolipid synthesis has recently been demonstrated to contribute to bacterial-induced pro-inflammatory responses by macrophages. In this study we present evidence demonstrating the presence of lipin-1 within macrophages in human atherosclerotic plaques. Additionally, reducing lipin-1 levels in macrophages significantly inhibits both modified-LDL-induced foam cell formation in vitro, as observed by smaller/fewer intracellular lipid inclusions, and ablates modified-LDL-elicited production of the pro-atherogenic mediators tumor necrosis factor-α, interleukin-6, and prostaglandin E2. These findings demonstrate a critical role for lipin-1 in the regulation of macrophage inflammatory responses to modified-LDL. These data begin to link the processes of foam cell formation and pro-inflammatory cytokine production within macrophages.


Subject(s)
Lipoproteins, LDL/metabolism , Macrophages/cytology , Nuclear Proteins/metabolism , Phosphatidate Phosphatase/metabolism , Plaque, Atherosclerotic/metabolism , Animals , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/pathology , Cell Line , Dinoprostone/metabolism , Flow Cytometry , Foam Cells/cytology , Gene Expression Regulation , Humans , Immunoenzyme Techniques , Inflammation , Interleukin-6/metabolism , Lipids/chemistry , Lipoproteins, LDL/chemistry , Macrophages/metabolism , Male , Mice , Mice, Knockout , Microscopy, Fluorescence , Tumor Necrosis Factor-alpha/metabolism
6.
Am J Pathol ; 185(9): 2575-89, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26212910

ABSTRACT

Endothelial cell interactions with transitional matrix proteins, such as fibronectin, occur early during atherogenesis and regulate shear stress-induced endothelial cell activation. Multiple endothelial cell integrins bind transitional matrix proteins, including α5ß1, αvß3, and αvß5. However, the role these integrins play in mediating shear stress-induced endothelial cell activation remains unclear. Therefore, we sought to elucidate which integrin heterodimers mediate shear stress-induced endothelial cell activation and early atherogenesis. We now show that inhibiting αvß3 integrins (S247, siRNA), but not α5ß1 or αvß5, blunts shear stress-induced proinflammatory signaling (NF-κB, p21-activated kinase) and gene expression (ICAM1, VCAM1). Importantly, inhibiting αvß3 did not affect cytokine-induced proinflammatory responses or inhibit all shear stress-induced signaling, because Akt, endothelial nitric oxide synthase, and extracellular regulated kinase activation remained intact. Furthermore, inhibiting αv integrins (S247), but not α5 (ATN-161), in atherosclerosis-prone apolipoprotein E knockout mice significantly reduced vascular remodeling after acute induction of disturbed flow. S247 treatment similarly reduced early diet-induced atherosclerotic plaque formation associated with both diminished inflammation (expression of vascular cell adhesion molecule 1, plaque macrophage content) and reduced smooth muscle incorporation. Inducible, endothelial cell-specific αv integrin deletion similarly blunted inflammation in models of disturbed flow and diet-induced atherogenesis but did not affect smooth muscle incorporation. Our studies identify αvß3 as the primary integrin heterodimer mediating shear stress-induced proinflammatory responses and as a key contributor to early atherogenic inflammation.


Subject(s)
Atherosclerosis/metabolism , Gene Expression/physiology , Integrin alphaVbeta3/metabolism , NF-kappa B/metabolism , Animals , Cells, Cultured , Endothelial Cells/metabolism , Inflammation/metabolism , Male , Mice, Knockout , Signal Transduction/physiology , Stress, Mechanical , Vascular Cell Adhesion Molecule-1/metabolism
7.
Arterioscler Thromb Vasc Biol ; 34(7): 1362-73, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24833794

ABSTRACT

OBJECTIVE: Endothelial cell activation drives early atherosclerotic plaque formation. Both fibronectin deposition and accumulation of oxidized low-density lipoprotein (oxLDL) occur early during atherogenesis, and both are implicated in enhanced endothelial cell activation. However, interplay between these responses has not been established. The objective of our study was to determine whether endothelial matrix composition modulates the inflammatory properties of oxLDL. APPROACH AND RESULTS: We now show that oxLDL-induced nuclear factor-κB activation, proinflammatory gene expression, and monocyte binding are significantly enhanced when endothelial cells are attached to fibronectin compared with basement membrane proteins. This enhanced response does not result from altered oxLDL receptor expression, oxLDL uptake, or reactive oxygen species production, but results from oxLDL-induced activation of the fibronectin-binding integrin α5ß1. Preventing α5ß1 signaling (blocking antibodies, knockout cells) inhibits oxLDL-induced nuclear factor-κB activation and vascular cell adhesion molecule-1 expression. Furthermore, oxLDL drives α5ß1-dependent integrin signaling through the focal adhesion kinase pathway, and focal adhesion kinase inhibition (PF-573228, small interfering RNA) blunts oxLDL-induced nuclear factor-κB activation, vascular cell adhesion molecule-1 expression, and monocyte adhesion. Last, treatment with the α5ß1 signaling inhibitor, ATN-161, significantly blunts atherosclerotic plaque development in apolipoprotein E-deficient mice, characterized by reduced vascular cell adhesion molecule-1 expression and macrophage accumulation without affecting fibrous cap size. CONCLUSIONS: Our data suggest that α5ß1-mediated cross-talk between fibronectin and oxLDL regulates inflammation in early atherogenesis and that therapeutics that inhibit α5 integrins may reduce inflammation without adversely affecting plaque structure.


Subject(s)
Atherosclerosis/metabolism , Endothelial Cells/metabolism , Inflammation/metabolism , Integrin alpha5beta1/metabolism , Lipoproteins, LDL/metabolism , Signal Transduction , Animals , Anti-Inflammatory Agents/pharmacology , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Cell Adhesion , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Endothelial Cells/drug effects , Extracellular Matrix/metabolism , Fibronectins/metabolism , Fibrosis , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Integrin alpha5beta1/antagonists & inhibitors , Male , Mice , Mice, Knockout , Monocytes/metabolism , NF-kappa B/metabolism , Plaque, Atherosclerotic , RNA Interference , Signal Transduction/drug effects , Time Factors , Transfection , Vascular Cell Adhesion Molecule-1/metabolism
8.
Atherosclerosis ; 232(2): 277-84, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24468139

ABSTRACT

OBJECTIVE: Altered subendothelial matrix composition regulates endothelial dysfunction and early atherosclerotic plaque formation. Hyperglycemia promotes endothelial matrix remodeling associated with multiple microvascular complications of diabetes, but a role for altered matrix composition in diabetic atherogenesis has not been described. Therefore, we sought to characterize the alterations in matrix composition during diabetic atherogenesis using both in vitro and in vivo model systems. METHODS AND RESULTS: Streptozotocin-induced diabetes in atherosclerosis-prone ApoE knockout mice promoted transitional matrix expression (fibronectin, thrombospondin-1) and deposition in intima of the aortic arch as determined by qRT-PCR array and immunohistochemistry. Early plaque formation occurs at discrete vascular sites exposed to disturbed blood flow patterns, whereas regions exposed to laminar flow are protected. Consistent with this pattern, hyperglycemia-induced transitional matrix deposition was restricted to regions of disturbed blood flow. Laminar flow significantly blunted high glucose-induced fibronectin expression (mRNA and protein) and fibronectin fibrillogenesis in endothelial cell culture models, whereas high glucose-induced fibronectin deposition was similar between disturbed flow and static conditions. CONCLUSIONS: Taken together, these data demonstrate that flow patterns and hyperglycemia coordinately regulate subendothelial fibronectin deposition during early atherogenesis.


Subject(s)
Atherosclerosis/genetics , Atherosclerosis/pathology , Diabetes Mellitus/physiopathology , Hyperglycemia/pathology , Animals , Aorta/pathology , Aorta, Thoracic/pathology , Apolipoproteins E/genetics , Brachiocephalic Trunk/pathology , Carotid Arteries/pathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Fibronectins/chemistry , Fibronectins/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation , Regional Blood Flow , Thrombospondin 1/metabolism
9.
Circ Res ; 106(8): 1394-403, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20224042

ABSTRACT

RATIONALE: Atherosclerosis is initiated by blood flow patterns that activate inflammatory pathways in endothelial cells. Activation of inflammatory signaling by fluid shear stress is highly dependent on the composition of the subendothelial extracellular matrix. The basement membrane proteins laminin and collagen found in normal vessels suppress flow-induced p21 activated kinase (PAK) and nuclear factor (NF)-kappaB activation. By contrast, the provisional matrix proteins fibronectin and fibrinogen found in wounded or inflamed vessels support flow-induced PAK and NF-kappaB activation. PAK mediates both flow-induced permeability and matrix-specific activation of NF-kappaB. OBJECTIVE: To elucidate the mechanisms regulating matrix-specific PAK activation. METHODS AND RESULTS: We now show that matrix composition does not affect the upstream pathway by which flow activates PAK (integrin activation, Rac). Instead, basement membrane proteins enhance flow-induced protein kinase (PK)A activation, which suppresses PAK. Inhibiting PKA restored flow-induced PAK and NF-kappaB activation in cells on basement membrane proteins, whereas stimulating PKA inhibited flow-induced activation of inflammatory signaling in cells on fibronectin. PKA suppressed inflammatory signaling through PAK inhibition. Activating PKA by injection of the prostacyclin analog iloprost reduced PAK activation and inflammatory gene expression at sites of disturbed flow in vivo, whereas inhibiting PKA by PKA inhibitor (PKI) injection enhanced PAK activation and inflammatory gene expression. Inhibiting PAK prevented the enhancement of inflammatory gene expression by PKI. CONCLUSIONS: Basement membrane proteins inhibit inflammatory signaling in endothelial cells via PKA-dependent inhibition of PAK.


Subject(s)
Basement Membrane/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Endothelial Cells/enzymology , Inflammation/enzymology , Mechanotransduction, Cellular , p21-Activated Kinases/metabolism , Animals , Anti-Inflammatory Agents/administration & dosage , Cattle , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endothelial Cells/drug effects , Enzyme Activation , Enzyme Activators/administration & dosage , Humans , Iloprost/administration & dosage , Inflammation/drug therapy , Inflammation/physiopathology , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Integrins/metabolism , Male , Mechanotransduction, Cellular/drug effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Pulsatile Flow , Regional Blood Flow , Stress, Mechanical , Time Factors , Transfection , cdc42 GTP-Binding Protein/metabolism , p21-Activated Kinases/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...