Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Am J Physiol Cell Physiol ; 324(6): C1199-C1212, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37093037

ABSTRACT

Endothelial cell (EC) migration is critical for the repair of monolayer disruption following angioplasties, but migration is inhibited by lipid oxidation products, including lysophosphatidylcholine (lysoPC), which open canonical transient receptor potential 6 (TRPC6) channels. TRPC6 activation requires an increase in intracellular Ca2+ concentration ([Ca2+]i), the source of which is unknown. LysoPC can activate phospholipase A2 to release arachidonic acid (ArA). ArA can activate arachidonic acid-regulated calcium (ARC) channels that are formed by stromal interaction molecule 1 (STIM1) and Orai1 and Orai3 proteins. Both lysoPC and ArA can activate p38 mitogen-activated protein kinase (MAPK) that induces the phosphorylation required for STIM1-Orai3 association. This is accompanied by an increase in [Ca2+]i and TRPC6 externalization. The effect of lysoPC and ArA is not additive, suggesting activation of the same pathway. The increase in [Ca2+]i activates an Src kinase that leads to TRPC6 activation. Downregulation of Orai3 using siRNA blocks the lysoPC- or ArA-induced increase in [Ca2+]i and TRPC6 externalization and preserves EC migration. These data show that lysoPC induces activation of p38 MAPK, which leads to STIM1-Orai3 association and increased [Ca2+]i. This increase in [Ca2+]i activates an Src kinase leading to TRPC6 externalization, which initiates a cascade of events ending in cytoskeletal changes that disrupt EC migration. Blocking this pathway preserves EC migration in the presence of lipid oxidation products.NEW & NOTEWORTHY The major lysophospholipid component in oxidized LDL, lysophosphatidylcholine (lysoPC), can activate p38 MAP kinase, which in turn promotes externalization of Orai3 and STIM1-Orai3 association, suggesting involvement of arachidonic acid-regulated calcium (ARC) channels. The subsequent increase in intracellular calcium activates an Src kinase required for TRPC6 externalization. TRPC6 activation, which has been shown to inhibit endothelial cell migration, is blocked by p38 MAP kinase or Orai3 downregulation, and this partially preserves endothelial migration in lysoPC.


Subject(s)
Lysophosphatidylcholines , p38 Mitogen-Activated Protein Kinases , TRPC6 Cation Channel/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Calcium/metabolism , Stromal Interaction Molecule 1/genetics , Arachidonic Acid/pharmacology , Calcium Channels/metabolism , src-Family Kinases/metabolism , ORAI1 Protein/genetics
2.
Cell Calcium ; 112: 102734, 2023 06.
Article in English | MEDLINE | ID: mdl-37030190

ABSTRACT

Lipid oxidation products, including lysophosphatidylcholine (lysoPC), accumulate at the site of arterial injury after vascular interventions and hinder re-endothelization. LysoPC activates calcium-permeable channels, specifically canonical transient receptor potential 6 (TRPC6) channels that induce a sustained increase in intracellular calcium ion concentration [Ca2+]i and contribute to dysregulation of the endothelial cell (EC) cytoskeleton. Activation of TRPC6 leads to inhibition of EC migration in vitro and delayed re-endothelization of arterial injuries in vivo. Previously, we demonstrated the role of phospholipase A2 (PLA2), specifically calcium-independent PLA2 (iPLA2), in lysoPC-induced TRPC6 externalization and inhibition of EC migration in vitro. The ability of FKGK11, an iPLA2-specific pharmacological inhibitor, to block TRPC6 externalization and preserve EC migration was assessed in vitro and in a mouse model of carotid injury. Our data suggest that FKGK11 prevents lysoPC-induced PLA2 activity, blocks TRPC6 externalization, attenuates calcium influx, and partially preserves EC migration in vitro. Furthermore, FKGK11 promotes re-endothelization of an electrocautery carotid injury in hypercholesterolemic mice. FKGK11 has similar arterial healing effects in male and female mice on a high-fat diet. This study suggests that iPLA2 is a potential therapeutic target to attenuate calcium influx through TRPC6 channels and promote EC healing in cardiovascular patients undergoing angioplasty.


Subject(s)
Calcium , Transient Receptor Potential Channels , Male , Female , Animals , Mice , TRPC6 Cation Channel , Calcium/metabolism , Lysophosphatidylcholines/pharmacology , Phospholipases A2 , TRPC Cation Channels
3.
Cell Calcium ; 111: 102718, 2023 05.
Article in English | MEDLINE | ID: mdl-36934559

ABSTRACT

Activation of phosphatidylinositol 3-kinase (PI3K) by lipid oxidation products, including lysophosphatidylcholine (lysoPC), increases the externalization of canonical transient receptor potential 6 (TRPC6) channels leading to a subsequent increase in intracellular calcium that contributes to cytoskeletal changes which inhibit endothelial cell (EC) migration in vitro and impair EC healing of arterial injuries in vivo. The PI3K p110α and p110δ catalytic subunit isoforms regulate lysoPC-induced TRPC6 externalization in vitro, but have many other functions. The goal of the current study is to identify the PI3K regulatory subunit isoform involved in TRPC6 externalization to potentially identify a more specific treatment regimen to improve EC migration and arterial healing, while minimizing off-target effects. Decreasing the p85α regulatory subunit isoform protein levels, but not the p85ß and p55γ regulatory subunit isoforms, with small interfering RNA inhibits lysoPC-induced translocation of the PI3K catalytic subunit to the plasma membrane, dramatically decreased phosphatidylinositol (3,4,5)-trisphosphate (PIP3) production and TRPC6 externalization, and significantly improves EC migration in the presence of lysoPC. These results identify the important and specific role of p85α in controlling translocation of PI3K from the cytosol to the plasma membrane and PI3K-mediated TRPC externalization by oxidized lipids. Current PI3K inhibitors block the catalytic subunit, but our data suggest that the regulatory subunit is a novel therapeutic target to promote EC migration and healing after arterial injuries that occur with angioplasty.


Subject(s)
Phosphatidylinositol 3-Kinases , Phosphatidylinositol 3-Kinases/metabolism , TRPC6 Cation Channel , Protein Isoforms/metabolism , Cell Movement/physiology , Membranes/metabolism
4.
J Biol Chem ; 297(4): 101180, 2021 10.
Article in English | MEDLINE | ID: mdl-34509476

ABSTRACT

During vascular interventions, oxidized low-density lipoprotein and lysophosphatidylcholine (lysoPC) accumulate at the site of arterial injury, inhibiting endothelial cell (EC) migration and arterial healing. LysoPC activates canonical transient receptor potential 6 (TRPC6) channels, leading to a prolonged increase in intracellular calcium ion concentration that inhibits EC migration. However, an initial increase in intracellular calcium ion concentration is required to activate TRPC6, and this mechanism remains elusive. We hypothesized that lysoPC activates the lipid-cleaving enzyme phospholipase A2 (PLA2), which releases arachidonic acid (AA) from the cellular membrane to open arachidonate-regulated calcium channels, allowing calcium influx that promotes externalization and activation of TRPC6 channels. The focus of this study was to identify the roles of calcium-dependent and/or calcium-independent PLA2 in lysoPC-induced TRPC6 externalization. We show that lysoPC induced PLA2 enzymatic activity and caused AA release in bovine aortic ECs. To identify the specific subgroup and the isoform(s) of PLA2 involved in lysoPC-induced TRPC6 activation, transient knockdown studies were performed in the human endothelial cell line EA.hy926 using siRNA to inhibit the expression of genes encoding cPLA2α, cPLA2γ, iPLA2ß, or iPLA2γ. Downregulation of the ß isoform of iPLA2 blocked lysoPC-induced release of AA from EC membranes and TRPC6 externalization, as well as preserved EC migration in the presence of lysoPC. We propose that blocking TRPC6 activation and promoting endothelial healing could improve the outcomes for patients undergoing cardiovascular interventions.


Subject(s)
Arachidonic Acid/metabolism , Calcium Signaling , Endothelial Cells/metabolism , Phospholipases A2/metabolism , TRPC6 Cation Channel/metabolism , Animals , Cattle , Cells, Cultured , Enzyme Activation , Lipoproteins, LDL/metabolism , Lysophosphatidylcholines/metabolism
5.
Am J Physiol Cell Physiol ; 320(6): C943-C955, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33689479

ABSTRACT

Endothelial cell (EC) migration is critical for healing arterial injuries, such as those that occur with angioplasty. Impaired re-endothelialization following arterial injury contributes to vessel thrombogenicity, intimal hyperplasia, and restenosis. Oxidized lipid products, including lysophosphatidylcholine (lysoPC), induce canonical transient receptor potential 6 (TRPC6) externalization leading to increased [Ca2+]i, activation of calpains, and alterations of the EC cytoskeletal structure that inhibit migration. The p110α and p110δ catalytic subunit isoforms of phosphatidylinositol 3-kinase (PI3K) regulate lysoPC-induced TRPC6 externalization in vitro. The goal of this study was to assess the in vivo relevance of those in vitro findings to arterial healing following a denuding injury in hypercholesterolemic mice treated with pharmacologic inhibitors of the p110α and p110δ isoforms of PI3K and a general PI3K inhibitor. Pharmacologic inhibition of the p110α or the p110δ isoform of PI3K partially preserves healing in hypercholesterolemic male mice, similar to a general PI3K inhibitor. Interestingly, the p110α, p110δ, and the general PI3K inhibitor do not improve arterial healing after injury in hypercholesterolemic female mice. These results indicate a potential new role for isoform-specific PI3K inhibitors in male patients following arterial injury/intervention. The results also identify significant sex differences in the response to PI3K inhibition in the cardiovascular system, where female sex generally has a cardioprotective effect. This study provides a foundation to investigate the mechanism for the sex differences in response to PI3K inhibition to develop a more generally applicable treatment option.


Subject(s)
Catalytic Domain/physiology , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/metabolism , Hypercholesterolemia/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Wound Healing/physiology , Animals , Cattle , Cell Line , Endothelial Cells/metabolism , Female , Humans , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/metabolism , Signal Transduction/physiology
6.
Am J Physiol Cell Physiol ; 320(5): C731-C741, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33625929

ABSTRACT

Lipid oxidation products, including lysophosphatidylcholine (lysoPC) inhibit endothelial cell (EC) migration in vitro and impair EC healing of arterial injuries in vivo, in part by activating phosphatidylinositol 3-kinase (PI3K), which increases the externalization of canonical transient receptor potential 6 (TRPC6) channels and the subsequent increase in intracellular calcium. Inhibition of PI3K is a potential method to decrease TRPC6 activation and restore migration, but PI3K is involved in multiple intracellular signaling pathways and has multiple downstream effectors. The goal of this study is to identify the specific p110 catalytic subunit isoforms responsible for lysoPC-induced TRPC6 externalization to identify a target for intervention while minimizing impact on alternative signaling pathways. Down-regulation of the p110α and p110δ isoforms, but not the p110ß or p110γ isoforms, with small interfering RNA significantly decreased phosphatidylinositol (3,4,5)-trisphosphate production and TRPC6 externalization, and significantly improved EC migration in the presence of lysoPC. These results identify an additional role of p110α in EC and reveal for the first time a specific role of p110δ in EC, providing a foundation for subsequent in vivo studies to investigate the impact of p110 isoform inhibition on arterial healing after injury.


Subject(s)
Cell Movement/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Endothelial Cells/drug effects , Lysophosphatidylcholines/pharmacology , TRPC6 Cation Channel/metabolism , Animals , Calcium Signaling , Catalytic Domain , Cattle , Cell Line , Class I Phosphatidylinositol 3-Kinases/genetics , Endothelial Cells/enzymology , Humans , Isoenzymes , Kinetics , Phosphatidylinositol Phosphates/metabolism
7.
JVS Vasc Sci ; 1: 136-150, 2020.
Article in English | MEDLINE | ID: mdl-33554153

ABSTRACT

OBJECTIVE: Previous studies showed the benefit of canonical transient receptor potential 6 (TRPC6) channel deficiency in promoting endothelial healing of arterial injuries in hypercholesterolemic animals. Long-term studies utilizing a carotid wire-injury model were undertaken in wild-type (WT) and TRPC6-/- mice to determine the effects of TRPC6 on phenotypic modulation of vascular smooth muscle cells (SMC) and neointimal hyperplasia. We hypothesized that TRPC6 was essential in the maintenance or reexpression of a differentiated SMC phenotype and minimized luminal stenosis following arterial injury. METHODS: The common carotid arteries (CCA) of WT and TRPC6-/- mice were evaluated at baseline and 4 weeks after wire injury. At baseline, CCA of TRPC6-/- mice had reduced staining of MYH11 and SM22, fewer elastin lamina, luminal dilation, and wall thinning. After carotid wire injury, TRPC6-/- mice developed significantly more pronounced luminal stenosis compared with WT mice. Injured TRPC6-/- CCA demonstrated increased medial/intimal cell number and active cell proliferation when compared with WT CCA. Immunohistochemistry suggested that expression of contractile biomarkers in medial SMC were essentially at baseline levels in WT CCA at 28 days after wire injury. By contrast, at 28 days after injury medial SMC from TRPC6-/- CCA showed a significant decrease in the expression of contractile biomarkers relative to baseline levels. To assess the role of TRPC6 in systemic arterial SMC phenotype modulation, SMC were harvested from thoracic aortae of WT and TRPC6-/- mice and were characterized. TRPC6-/- SMC showed enhanced proliferation and migration in response to serum stimulation. Expression of contractile phenotype biomarkers, MYH11 and SM22, was attenuated in TRPC6-/- SMC. siRNA-mediated TRPC6 deficiency inhibited contractile biomarker expression in a mouse SMC line. CONCLUSIONS: These results suggest that TRPC6 contributes to the restoration or maintenance of arterial SMC contractile phenotype following injury. Understanding the role of TRPC6 in phenotypic modulation may lead to mechanism-based therapies for attenuation of IH.

8.
Am J Physiol Cell Physiol ; 313(5): C541-C555, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28835433

ABSTRACT

Lipid oxidation products, including lysophosphatidylcholine (lysoPC), activate canonical transient receptor potential 6 (TRPC6) channels, and the subsequent increase in intracellular Ca2+ leads to TRPC5 activation. The goal of this study is to elucidate the steps in the pathway between TRPC6 activation and TRPC5 externalization. Following TRPC6 activation by lysoPC, extracellular regulated kinase (ERK) is phosphorylated. This leads to phosphorylation of p47phox and subsequent NADPH oxidase activation with increased production of reactive oxygen species. ERK activation requires TRPC6 opening and influx of Ca2+ as evidenced by the failure of lysoPC to induce ERK phosphorylation in TRPC6-/- endothelial cells. ERK siRNA blocks the lysoPC-induced activation of NADPH oxidase, demonstrating that ERK activation is upstream of NADPH oxidase. The reactive oxygen species produced by NADPH oxidase promote myosin light chain kinase (MLCK) activation with phosphorylation of MLC and TRPC5 externalization. Downregulation of ERK, NADPH oxidase, or MLCK with the relevant siRNA prevents TRPC5 externalization. Blocking MLCK activation prevents the prolonged rise in intracellular calcium levels and preserves endothelial migration in the presence of lysoPC.


Subject(s)
Cell Movement/physiology , Endothelial Cells/metabolism , NADPH Oxidases/metabolism , TRPC Cation Channels/metabolism , Animals , Cattle , Cell Movement/drug effects , Endothelial Cells/drug effects , Enzyme Activation/physiology , Humans , Lysophosphatidylcholines/pharmacology , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , TRPC6 Cation Channel
9.
Proc Natl Acad Sci U S A ; 113(8): 2110-5, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26858457

ABSTRACT

Lipid oxidation products, including lysophosphatidylcholine (lysoPC), activate canonical transient receptor potential 6 (TRPC6) channels leading to inhibition of endothelial cell (EC) migration in vitro and delayed EC healing of arterial injuries in vivo. The precise mechanism through which lysoPC activates TRPC6 channels is not known, but calmodulin (CaM) contributes to the regulation of TRPC channels. Using site-directed mutagenesis, cDNAs were generated in which Tyr(99) or Tyr(138) of CaM was replaced with Phe, generating mutant CaM, Phe(99)-CaM, or Phe(138)-CaM, respectively. In ECs transiently transfected with pcDNA3.1-myc-His-Phe(99)-CaM, but not in ECs transfected with pcDNA3.1-myc-His-Phe(138)-CaM, the lysoPC-induced TRPC6-CaM dissociation and TRPC6 externalization was disrupted. Also, the lysoPC-induced increase in intracellular calcium concentration was inhibited in ECs transiently transfected with pcDNA3.1-myc-His-Phe(99)-CaM. Blocking phosphorylation of CaM at Tyr(99) also reduced CaM association with the p85 subunit and subsequent activation of phosphatidylinositol 3-kinase (PI3K). This prevented the increase in phosphatidylinositol (3,4,5)-trisphosphate (PIP3) and the translocation of TRPC6 to the cell membrane and reduced the inhibition of EC migration by lysoPC. These findings suggest that lysoPC induces CaM phosphorylation at Tyr(99) by a Src family kinase and that phosphorylated CaM activates PI3K to produce PIP3, which promotes TRPC6 translocation to the cell membrane.


Subject(s)
Calcium Signaling/physiology , Calmodulin/metabolism , Cell Membrane/metabolism , Cell Movement/physiology , Endothelial Cells/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TRPC Cation Channels/metabolism , Animals , Calcium/metabolism , Calmodulin/genetics , Cattle , Cell Membrane/genetics , Endothelial Cells/cytology , Enzyme Activation/physiology , Humans , Lysophosphatidylcholines/genetics , Lysophosphatidylcholines/metabolism , Phosphatidylinositol 3-Kinases/genetics , Protein Transport/physiology , TRPC Cation Channels/genetics , TRPC6 Cation Channel
10.
Atherosclerosis ; 241(2): 709-15, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26125413

ABSTRACT

OBJECTIVE: Endothelial cell (EC) migration is essential for healing of arterial injuries caused by angioplasty, but a high cholesterol diet inhibits endothelial repair. In vivo studies suggest that apolipoprotein A-I (apoA-I), the major protein constituent of HDL, is essential for normal healing of arterial injuries. ApoA-I mimetics, including 4F, have been designed to mimic the amphipathic portion of the apoA-I molecule. This study was undertaken to determine if 4F improves endothelial migration and healing. METHODS: A razor scrape assay was used to analyze the effect of 4F on EC migration in vitro. Endothelial healing in vivo was assessed following electrical injury of carotid arteries in mice. Markers of oxidative stress were also examined. RESULTS: Lipid oxidation products inhibited EC migration in vitro, but preincubation with L-4F preserved EC migration. Endothelial healing of carotid arterial injuries in mice on a high cholesterol diet was delayed compared with mice on a chow diet with 27.8% vs. 48.2% healing, respectively, at 5 days. Administration of D-4F improved endothelial healing in mice on a high cholesterol diet to 43.4%. D-4F administration had no effect on lipid levels but decreased markers of oxidation. In vivo, there was a significant inverse correlation between endothelial healing and plasma markers of oxidative stress. CONCLUSION: These studies suggested that an apoA-I mimetic can improve endothelial healing of arterial injuries by decreasing oxidative stress.


Subject(s)
Apolipoprotein A-I/metabolism , Arteries/metabolism , Oxidative Stress , Peptides/chemistry , Animals , Aorta/cytology , Apolipoprotein A-I/chemistry , Carotid Arteries/pathology , Cattle , Cell Movement , Cells, Cultured , Endothelial Cells/cytology , Hypercholesterolemia/pathology , Lipids/chemistry , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Reactive Oxygen Species/metabolism , Thrombosis , Wound Healing
11.
J Vasc Surg ; 62(4): 1040-1047.e2, 2015 Oct.
Article in English | MEDLINE | ID: mdl-24820897

ABSTRACT

OBJECTIVE: After arterial injury, endothelial cell (EC) migration is essential for healing, but lipid oxidation products activate TRPC6 and TRPC5 ion channels, leading to increased intracellular calcium and inhibition of EC migration in vitro. The objective of this study was to further evaluate the role of TRPC channels in EC migration in vitro and to validate in vitro findings in an in vivo model. METHODS: Mouse aortic ECs were cultured, and the effect of lysophosphatidylcholine, the major lysophospholipid in oxidized low-density lipoprotein, on migration was assessed in a razor-scrape assay. EC healing after a carotid injury with electrocautery was evaluated in wild-type (WT), TRPC6(-/-), and TRPC5(-/-) mice receiving either a chow or high-cholesterol (HC) diet. RESULTS: Lysophosphatidylcholine inhibited EC migration of WT ECs to 22% of baseline and of TRPC5(-/-) ECs to 53% of baseline but had minimal effect on TRPC6(-/-) EC migration. Hypercholesterolemia severely impaired EC healing in vivo, with 51.4% ± 1.8% and 24.9% ± 2.0% of the injury resurfaced with ECs at 5 days in chow-fed and HC-fed WT mice, respectively (P < .001). Hypercholesterolemia did not impair healing in TRPC6(-/-) mice, with coverage of 48.4% ± 3.4% and 46.8% ± 1.6% in chow-fed and HC-fed TRPC6(-/-) mice, respectively. Hypercholesterolemia had a reduced inhibitory effect in TRPC5(-/-) mice, with EC coverage of 51.7% ± 3.0% and 37.% ± 1.4% in chow-fed and HC-fed TRPC5(-/-) mice, respectively. CONCLUSIONS: Results suggest that activation of TRPC6 and TRPC5 channels is the key contributor to impaired endothelial healing of arterial injuries in hypercholesterolemic mice.


Subject(s)
Arteries/injuries , Endothelium, Vascular/physiology , Hypercholesterolemia/physiopathology , TRPC Cation Channels/physiology , Animals , Biomarkers/blood , Calcium/analysis , Cell Movement/physiology , Endothelial Cells/physiology , Hypercholesterolemia/blood , Hypercholesterolemia/urine , In Vitro Techniques , Inflammation/blood , Lysophosphatidylcholines/pharmacology , Mice , Oxidative Stress , Wound Healing/physiology
12.
J Vasc Surg ; 55(2): 489-96, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22047834

ABSTRACT

OBJECTIVE: Endothelial cell (EC) migration is essential for arterial healing after angioplasty. Oxidized low-density lipoproteins and oxidative stress decrease EC migration in vitro. The objective of this study was to determine the effect of hypercholesterolemia and oxidative stress on EC healing after an arterial injury. METHODS: C57BL/6 wild-type mice were placed in one of eight groups: chow diet (n = 11), high-cholesterol (HC) diet (n = 11), chow diet plus paraquat (n = 11), HC diet plus paraquat (n = 11), chow diet plus N-acetylcysteine (NAC) (n = 11), HC diet plus NAC (n = 11), chow diet plus paraquat and NAC (n = 11), and HC diet plus paraquat and NAC (n = 11). After 2 weeks on the assigned diet with or without NAC, the carotid artery was injured using electrocautery. Animals in the paraquat groups were given 1 mg/kg intraperitoneally to increase oxidative stress. After 120 hours, Evans Blue dye was infused intravenously to stain the area of the artery that remained deendothelialized. This was used to calculate the percentage of re-endothelialization. Plasma and tissue samples were analyzed for measures of oxidative stress. RESULTS: The HC diet increased oxidative stress and reduced EC healing compared with a chow diet, with EC covering 26.8% ± 2.8% and 48.1% ± 5.2% (P < .001) of the injured area, respectively. Administration of paraquat decreased healing in both chow and HC animals to 18.1% ± 3.5% (P < .001) and 9.8% ± 4.6% (P < .001), respectively. Pretreatment with NAC (120 mmol/L in drinking water) for 2 weeks prior to injury, to decrease oxidative stress, improved EC healing to 39.9% ± 5.7% (P < .001) in hypercholesterolemic mice and to 30.7% ± 3.6% (P < .001) in the paraquat group. NAC treatment improved healing to 24.6% ± 3.4% (P < .001) in hypercholesterolemic mice treated with paraquat. CONCLUSION: Re-endothelialization of arterial injuries is reduced in hypercholesterolemic mice and is inversely correlated with oxidative stress. An oral antioxidant decreases oxidative stress and improves EC healing. CLINICAL RELEVANCE: Vascular injury following cardiovascular intervention, including cardiac and peripheral arterial angioplasty and stenting, is associated with inflammation and oxidative stress. Hypercholesterolemia is also associated with increased oxidative stress. Oxidative stress, regardless of the source, induces cellular dysfunction in endothelial and smooth muscle cells that reduce healing after arterial injury. Decreasing oxidative stress with an exogenously administered antioxidant can improve endothelial cell healing, and this is important to control intimal hyperplasia and reduce the thrombogenicity of the vessel.


Subject(s)
Carotid Artery Injuries/complications , Carotid Artery, Common/pathology , Cell Proliferation , Endothelial Cells/pathology , Hypercholesterolemia/complications , Oxidative Stress , Wound Healing , Acetylcysteine/pharmacology , Animals , Antioxidants/pharmacology , Biomarkers/blood , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery, Common/drug effects , Carotid Artery, Common/metabolism , Cell Proliferation/drug effects , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Male , Mice , Mice, Inbred C57BL , Oxidants/toxicity , Oxidative Stress/drug effects , Paraquat/toxicity , Time Factors , Wound Healing/drug effects
13.
J Vasc Surg ; 51(1): 184-93, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19939614

ABSTRACT

OBJECTIVE: Limited endothelial cell (EC) coverage and anastomotic intimal hyperplasia contribute to thrombosis and failure of prosthetic grafts. Lipid accumulation and lipid oxidation are associated with decreased EC migration and intimal hyperplasia. The goal of this study was to assess the ability of antioxidants to improve graft healing in hypercholesterolemic animals. METHODS: Rabbits were placed in one of four groups: chow plus N-acetylcysteine (NAC), chow plus probucol, chow with 1% cholesterol plus NAC, or chow with 1% cholesterol plus probucol. After 2 weeks, expanded polytetrafluoroethylene grafts (12 cm long x 4-mm internal diameter) were implanted in the abdominal aorta. Grafts were removed after 6 weeks and analyzed for cholesterol content, EC coverage, anastomotic intimal thickness, and the cellular composition of the neointima. Plasma samples were obtained to assess systemic oxidative stress. The data were compared with previously reported data from animals fed diets of chow and chow with 1% cholesterol. RESULTS: Prosthetic grafts from rabbits fed chow with 1% cholesterol had significantly greater anastomotic intimal thickening and lower EC coverage than grafts from rabbits fed a regular chow diet. In hypercholesterolemic rabbits, antioxidant therapy decreased global oxidative stress as evidenced by a 40% decrease in plasma thiobarbituric acid reactive substances. In rabbits fed the chow with 1% cholesterol diet, NAC decreased intimal hyperplasia at the proximal anastomosis by 29% and significantly increased graft EC coverage from 46% to 71% (P = .03). Following a similar pattern, probucol decreased intimal hyperplasia by 43% and increased graft EC coverage to 53% in hypercholesterolemic rabbits. CONCLUSIONS: Global oxidative stress and anastomotic intimal hyperplasia are increased, and endothelialization of prosthetic grafts is significantly reduced in rabbits fed a high-cholesterol diet. Antioxidant treatment improves EC coverage and decreases intimal hyperplasia. Reducing oxidative stress may promote healing of prosthetic grafts.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Aorta, Abdominal/drug effects , Aorta, Abdominal/surgery , Blood Vessel Prosthesis Implantation , Hypercholesterolemia/therapy , Wound Healing/drug effects , Animals , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aorta, Abdominal/physiopathology , Blood Vessel Prosthesis Implantation/adverse effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cholesterol/blood , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/pathology , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Hypercholesterolemia/physiopathology , Hyperplasia , Macrophages/drug effects , Macrophages/pathology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Oxidative Stress/drug effects , Rabbits , Thiobarbituric Acid Reactive Substances/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
14.
Arch Otolaryngol Head Neck Surg ; 135(11): 1092-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19917920

ABSTRACT

OBJECTIVE: To examine the role of central neck dissection (CND) in patients with papillary thyroid cancer (PTC). DESIGN: Retrospective analysis of patients treated for PTC between 1993 and 2008. SETTING: Academic institution. PATIENTS: All patients diagnosed with PTC who underwent surgical therapy at our institution. MAIN OUTCOME MEASURES: Recurrence, hypocalcemia, hypoparathyroidism, and recurrent laryngeal nerve (RLN) injury. RESULTS: A total of 136 patients were treated for PTC, 26 of whom were excluded because their initial resection was performed at another institution. Of the 110 patients who underwent initial surgical therapy, CND was performed in 22 patients (20%), 18 with and 4 without enlarged nodes at the time of surgery. A mean (SD) of 11 (4) lymph nodes were removed, and lymph node metastases were identified in 17 patients (77%). One patient developed a recurrence in the lateral neck at 15 months' follow-up. Eighty-eight patients had no abnormal lymph nodes and did not undergo CND, 2 of whom developed a recurrence (2%) (P = .49) in the central neck at 14 months' and 11 years' follow-up. Permanent RLN injury occurred in no patient who underwent CND and in 1 patient without a CND (1%). Transient hypocalcemia occurred in 19 patients who underwent CND (86%) compared with 54 patients without a CND (61%) (P = .01). Permanent hypoparathyroidism occurred in 1 patient who underwent a CND (5%). CONCLUSION: After total thyroidectomy and CND, recurrence in the central neck is uncommon, but hypocalcemia is more common, raising questions about the use of routine CND in patients with PTC.


Subject(s)
Carcinoma, Papillary/surgery , Neck Dissection/methods , Thyroid Neoplasms/surgery , Adult , Carcinoma, Papillary/secondary , Female , Follow-Up Studies , Humans , Incidence , Lymphatic Metastasis/prevention & control , Male , Middle Aged , Neoplasm Recurrence, Local/epidemiology , Neoplasm Recurrence, Local/prevention & control , Ohio/epidemiology , Retrospective Studies , Thyroid Neoplasms/pathology , Thyroidectomy , Time Factors , Treatment Outcome
15.
Expert Rev Anticancer Ther ; 9(3): 317-29, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19275510

ABSTRACT

The incidence of thyroid cancer is increasing by 4% per year. Thyroid cancer has become the eighth most common malignancy diagnosed in women. Papillary cancer accounts for 80% of all thyroid cancer. The management of papillary thyroid cancer is challenging, primarily because there have been no prospective randomized trials to help guide therapeutic decision making. The purpose of this article is to discuss the contemporary management of papillary thyroid cancer, including the diagnosis and pre-operative evaluation, surgical management, postoperative thyroid hormone and radioiodine therapy, long-term follow-up, prognosis and management of recurrent and metastatic disease. The role of molecular markers to enhance the cytological diagnosis of papillary cancer and new molecular-based therapies will also be reviewed.


Subject(s)
Carcinoma, Papillary , Thyroid Neoplasms , Carcinoma, Papillary/diagnosis , Carcinoma, Papillary/secondary , Carcinoma, Papillary/therapy , Clinical Trials as Topic , Combined Modality Therapy , Humans , Iodine Radioisotopes/therapeutic use , Lymphatic Metastasis , Neoplasm Recurrence, Local , Prognosis , Radiopharmaceuticals/therapeutic use , Radiotherapy, Adjuvant , Thyroglobulin/blood , Thyroid Hormones/therapeutic use , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/pathology , Thyroid Neoplasms/therapy , Thyroidectomy , Treatment Outcome
16.
Am J Surg ; 195(3): 339-43; discussion 343, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18241836

ABSTRACT

BACKGROUND: Observation following thyroidectomy and parathyroidectomy has been progressively shortened. The challenge has been to reduce the duration of postoperative observation without jeopardizing patient safety. METHODS: A retrospective review of patients who underwent thyroidectomy and/or parathyroidectomy between July 1990 and March 2007 was completed to determine the frequency of life-threatening hematoma and hospital readmission and their impact on postoperative observation. RESULTS: Of 1,050 patients, life-threatening hematoma developed in 6 (.6%) patients, 5 following bilateral and 1 following unilateral thyroidectomy. Hematoma developed 10 minutes to 7 days postoperatively, four within 4 hours, one at 21 hours, and one at 7 days. Twelve patients were readmitted an average of 5 days postoperatively for hypocalcemia, hematoma, infection, or respiratory distress. CONCLUSION: Without factors contributing to bleeding, unilateral thyroidectomy and parathyroidectomy can be performed as an ambulatory procedure. To maximize safety, we recommend 4-hour and 23-hour observation following unilateral and bilateral thyroidectomy, respectively.


Subject(s)
Hematoma/epidemiology , Parathyroidectomy/adverse effects , Thyroidectomy/adverse effects , Adult , Aged , Female , Hematoma/etiology , Humans , Male , Middle Aged , Neck , Patient Readmission , Postoperative Care , Retrospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...