Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Blood ; 137(16): 2152-2160, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33649757

ABSTRACT

Thrombotic, vascular, and bleeding complications are the most common causes of morbidity and mortality in the Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). In these disorders, circulating red cells, leukocytes, and platelets, as well as some vascular endothelial cells, each have abnormalities that are cell-intrinsic to the MPN driver mutations they harbor (eg, JAK2 V617F). When these cells are activated in the MPNs, their interactions with each other create a highly proadhesive and prothrombotic milieu in the circulation that predisposes patients with MPN to venous, arterial, and microvascular thrombosis and occlusive disease. Bleeding problems in the MPNs are caused by the MPN blood cell-initiated development of acquired von Willebrand disease. The inflammatory state created by MPN stem cells in their microenvironment extends systemically to amplify the clinical thrombotic tendency and, at the same time, preferentially promote further MPN stem cell clonal expansion, thereby generating a vicious cycle that favors a prothrombotic state in these diseases.


Subject(s)
Hemorrhage/pathology , Microvessels/pathology , Myeloproliferative Disorders/pathology , Thrombosis/pathology , Vascular Diseases/pathology , Animals , Hemorrhage/etiology , Humans , Inflammation/etiology , Inflammation/pathology , Myeloproliferative Disorders/complications , Neoplasms/complications , Neoplasms/pathology , Thrombosis/etiology , Tumor Microenvironment , Vascular Diseases/etiology
2.
Blood ; 124(2): 184-7, 2014 Jul 10.
Article in English | MEDLINE | ID: mdl-24868077

ABSTRACT

Paraneoplastic thrombocytosis is associated with many solid tumors and often correlates with reduced survival. Recent studies suggest that a pathogenic feed back loop may be operative between platelets and tumor cells, with reciprocal interactions between tumor growth/metastasis and thrombocytosis/platelet activation. Specific molecular pathways have been identified in which tumors can stimulate platelet production and activation; activated platelets can, in turn, promote tumor growth and metastasis. Taken together, these findings provide exciting new potential targets for therapeutic intervention.


Subject(s)
Paraneoplastic Syndromes/pathology , Animals , Antineoplastic Agents/therapeutic use , Blood Platelets/pathology , Blood Platelets/physiology , Cell Proliferation , Humans , Molecular Targeted Therapy , Paraneoplastic Syndromes/drug therapy , Paraneoplastic Syndromes/etiology , Thrombocytosis/drug therapy , Thrombocytosis/etiology , Thrombocytosis/pathology , Tumor Microenvironment/physiology
3.
Blood ; 118(7): 1998-2006, 2011 Aug 18.
Article in English | MEDLINE | ID: mdl-21653942

ABSTRACT

Hyperhomocysteinemia (HHcy) is associated with endothelial dysfunction (ED), but the mechanism is largely unknown. In this study, we investigated the role and mechanism of HHcy-induced ED in microvasculature in our newly established mouse model of severe HHcy (plasma total homocysteine, 169.5 µM). We found that severe HHcy impaired nitric oxide (NO)- and endothelium-derived hyperpolarizing factor (EDHF)-mediated, endothelium-dependent relaxations of small mesenteric arteries (SMAs). Endothelium-independent and prostacyclin-mediated endothelium-dependent relaxations were not changed. A nonselective Ca(2+)-activated potassium channel (K(Ca)) inhibitor completely blocked EDHF-mediated relaxation. Selective blockers for small-conductance K(Ca) (SK) or intermediate-conductance K(Ca) (IK) failed to inhibit EDHF-mediated relaxation in HHcy mice. HHcy increased the levels of SK3 and IK1 protein, superoxide (O(2)(-)), and 3-nitrotyrosine in the endothelium of SMAs. Preincubation with antioxidants and peroxynitrite (ONOO(-)) inhibitors improved endothelium-dependent and EDHF-mediated relaxations and decreased O(2)(-) production in SMAs from HHcy mice. Further, EDHF-mediated relaxation was inhibited by ONOO(-) and prevented by catalase in the control mice. Finally, L-homocysteine stimulated O(2)(-) production, which was reversed by antioxidants, and increased SK/IK protein levels and tyrosine nitration in cultured human cardiac microvascular endothelial cells. Our results suggest that HHcy impairs EDHF relaxation in SMAs by inhibiting SK/IK activities via oxidation- and tyrosine nitration-related mechanisms.


Subject(s)
Biological Factors/metabolism , Cystathionine beta-Synthase/genetics , Hyperhomocysteinemia/physiopathology , Mesenteric Arteries/physiopathology , Vasodilation , Animals , Cardiovascular Diseases/etiology , Cell Line , Gene Deletion , Homocysteine/blood , Humans , Hyperhomocysteinemia/genetics , Hyperhomocysteinemia/metabolism , Mesenteric Arteries/metabolism , Mice , Mice, Transgenic , Nitrogen Oxides/metabolism
4.
Article in English | MEDLINE | ID: mdl-30166693
5.
Am J Physiol Heart Circ Physiol ; 302(7): H1367-77, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22268102

ABSTRACT

Subtle alterations in cerebral blood flow can impact the health and function of brain cells and are linked to cognitive decline and dementia. To understand hemodynamics in the three-dimensional vascular network of the cerebral cortex, we applied two-photon excited fluorescence microscopy to measure the motion of red blood cells (RBCs) in individual microvessels throughout the vascular hierarchy in anesthetized mice. To resolve heartbeat- and respiration-dependent flow dynamics, we simultaneously recorded the electrocardiogram and respiratory waveform. We found that centerline RBC speed decreased with decreasing vessel diameter in arterioles, slowed further through the capillary bed, and then increased with increasing vessel diameter in venules. RBC flow was pulsatile in nearly all cortical vessels, including capillaries and venules. Heartbeat-induced speed modulation decreased through the vascular network, while the delay between heartbeat and the time of maximum speed increased. Capillary tube hematocrit was 0.21 and did not vary with centerline RBC speed or topological position. Spatial RBC flow profiles in surface vessels were blunted compared with a parabola and could be measured at vascular junctions. Finally, we observed a transient decrease in RBC speed in surface vessels before inspiration. In conclusion, we developed an approach to study detailed characteristics of RBC flow in the three-dimensional cortical vasculature, including quantification of fluctuations in centerline RBC speed due to cardiac and respiratory rhythms and flow profile measurements. These methods and the quantitative data on basal cerebral hemodynamics open the door to studies of the normal and diseased-state cerebral microcirculation.


Subject(s)
Cerebral Cortex/blood supply , Cerebrovascular Circulation/physiology , Heart/physiology , Respiratory Mechanics/physiology , Algorithms , Anesthesia , Animals , Arterioles/physiology , Blood Flow Velocity/physiology , Capillaries/physiology , Electrocardiography , Female , Hematocrit , Male , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence, Multiphoton , Myocardial Contraction/physiology , Pulsatile Flow , Venules/physiology
6.
FASEB J ; 24(8): 2804-17, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20305127

ABSTRACT

Hyperhomocysteinemia is an independent risk factor for cardiovascular disease. Homocysteine (Hcy) metabolism involves multiple enzymes; however, tissue Hcy metabolism and its relevance to methylation remain unknown. Here, we established gene expression profiles of 8 Hcy metabolic and 12 methylation enzymes in 20 human and 19 mouse tissues through bioinformatic analysis using expression sequence tag clone counts in tissue cDNA libraries. We analyzed correlations between gene expression, Hcy, S-adenosylhomocysteine (SAH), and S-adenosylmethionine (SAM) levels, and SAM/SAH ratios in mouse tissues. Hcy metabolic and methylation enzymes were classified into two types. The expression of Type 1 enzymes positively correlated with tissue Hcy and SAH levels. These include cystathionine beta-synthase, cystathionine-gamma-lyase, paraxonase 1, 5,10-methylenetetrahydrofolate reductase, betaine:homocysteine methyltransferase, methionine adenosyltransferase, phosphatidylethanolamine N-methyltransferases and glycine N-methyltransferase. Type 2 enzyme expressions correlate with neither tissue Hcy nor SAH levels. These include SAH hydrolase, methionyl-tRNA synthase, 5-methyltetrahydrofolate:Hcy methyltransferase, S-adenosylmethionine decarboxylase, DNA methyltransferase 1/3a, isoprenylcysteine carboxyl methyltransferases, and histone-lysine N-methyltransferase. SAH is the only Hcy metabolite significantly correlated with Hcy levels and methylation enzyme expression. We established equations expressing combined effects of methylation enzymes on tissue SAH, SAM, and SAM/SAH ratios. Our study is the first to provide panoramic tissue gene expression profiles and mathematical models of tissue methylation regulation.


Subject(s)
Computational Biology/methods , Gene Expression Regulation , Homocysteine/metabolism , Metabolic Networks and Pathways/genetics , Animals , Enzymes/genetics , Gene Expression Profiling , Humans , Infant, Newborn , Methylation , Mice , S-Adenosylhomocysteine/analysis , S-Adenosylmethionine/analysis
7.
Hematol Oncol Clin North Am ; 35(2): 305-324, 2021 04.
Article in English | MEDLINE | ID: mdl-33641871

ABSTRACT

Thrombotic, vascular, and bleeding complications are the most frequent causes of morbidity and mortality in myeloproliferative neoplasms (MPNs). The interplay and reciprocal amplification between two factors are considered to lead to thrombosis in MPNs: (1) circulating blood cell-intrinsic abnormalities caused by an MPN driver mutation in their hematopoietic progenitor/stem cells, interacting with vascular endothelial cells, show prothrombotic and proadhesive phenotypes; and (2) a state of usually subclinical systemic inflammation that fuels the thrombotic tendency. Prevention and treatment require maintenance of hematocrit less than 45% and cytoreductive therapy in patients with a high risk for thrombotic and vascular complications.


Subject(s)
Myeloproliferative Disorders , Neoplasms , Thrombosis , Cytoreduction Surgical Procedures , Endothelial Cells , Hematopoietic Stem Cells , Hemorrhage/etiology , Humans , Myeloproliferative Disorders/complications , Neoplasms/complications , Thrombosis/etiology
8.
Circulation ; 120(19): 1893-902, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19858416

ABSTRACT

BACKGROUND: Hyperhomocysteinemia (HHcy) is an independent risk factor for cardiovascular disease. Monocytes display inflammatory and resident subsets and commit to specific functions in atherogenesis. In this study, we examined the hypothesis that HHcy modulates monocyte heterogeneity and leads to atherosclerosis. METHODS AND RESULTS: We established a novel atherosclerosis-susceptible mouse model with both severe HHcy and hypercholesterolemia in which the mouse cystathionine beta-synthase (CBS) and apolipoprotein E (apoE) genes are deficient and an inducible human CBS transgene is introduced to circumvent the neonatal lethality of the CBS deficiency (Tg-hCBS apoE(-/-) Cbs(-/-) mice). Severe HHcy accelerated atherosclerosis and inflammatory monocyte/macrophage accumulation in lesions and increased plasma tumor necrosis factor-alpha and monocyte chemoattractant protein-1 levels in Tg-hCBS apoE(-/-) Cbs(-/-) mice fed a high-fat diet. Furthermore, we characterized monocyte heterogeneity in Tg-hCBS apoE(-/-) Cbs(-/-) mice and another severe HHcy mouse model (Tg-S466L Cbs(-/-)) with a disease-relevant mutation (Tg-S466L) that lacks hyperlipidemia. HHcy increased monocyte population and selective expansion of inflammatory Ly-6C(hi) and Ly-6C(mid) monocyte subsets in blood, spleen, and bone marrow of Tg-S466L Cbs(-/-) and Tg-hCBS apoE(-/-) Cbs(-/-) mice. These changes were exacerbated in Tg-S466L Cbs(-/-) mice with aging. Addition of l-homocysteine (100 to 500 micromol/L), but not l-cysteine, maintained the Ly-6C(hi) subset and induced the Ly-6C(mid) subset in cultured mouse primary splenocytes. Homocysteine-induced differentiation of the Ly-6C(mid) subset was prevented by catalase plus superoxide dismutase and the NAD(P)H oxidase inhibitor apocynin. CONCLUSIONS: HHcy promotes differentiation of inflammatory monocyte subsets and their accumulation in atherosclerotic lesions via NAD(P)H oxidase-mediated oxidant stress.


Subject(s)
Atherosclerosis/immunology , Cystathionine beta-Synthase/genetics , Homocystinuria/immunology , Hyperhomocysteinemia/immunology , Monocytes/immunology , Animals , Apolipoproteins E/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Body Weight , Cells, Cultured , Chemokine CCL2/blood , Cystathionine beta-Synthase/metabolism , Disease Models, Animal , Female , Homocystinuria/metabolism , Homocystinuria/pathology , Humans , Hyperhomocysteinemia/metabolism , Hyperhomocysteinemia/pathology , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/pathology , Oxidative Stress/physiology , Pregnancy , Severity of Illness Index , Spleen/cytology , Tumor Necrosis Factor-alpha/blood , Vasculitis/immunology , Vasculitis/metabolism , Vasculitis/pathology
9.
Circ Res ; 102(1): 42-50, 2008 Jan 04.
Article in English | MEDLINE | ID: mdl-17967787

ABSTRACT

Reactive oxygen species (ROS) and oxidant stress are important mediators of cardiovascular pathologies including atherosclerosis. One source of ROS in the vasculature is free heme released from hemoglobin. Because Egr-1, the regulator of cell proliferation and apoptosis, is also induced by oxidant stress and is likewise implicated in atherosclerosis, we examined the regulation of Egr-1 by heme in vascular smooth muscle cells (SMCs). Hemin increased Egr-1 expression (mRNA, protein) within 30 minutes and ERK-1/2 phosphorylation and nuclear translocation within 5 minutes. Inhibiting hemin-induced ERK-1/2 activation by U0126 (MAPK-inhibitor), the antioxidant N-acetyl cysteine, the NADPH oxidase inhibitors apocynin and diphenyleneiodonium chloride, the superoxide scavenger tiron, or tricarbonyldichlororuthenium(II)-dimer (carbon-monoxide donor; CORM-2) blocked hemin-induced Egr-1 expression. Hemin activated Elk-1, SRF, and NF-kappaB and promoted their interaction with the Egr-1 promoter. Downregulating Elk-1 (via siRNA) or blocking NF-kappaB activation (via BAY-11-7082) abolished hemin induction of Egr-1. Finally, hemin-induced Egr-1 bound the promoters of tissue factor (TF), Plasminogen Activator Inhibitor (PAI)-1, and NGF-1A Binding (NAB)-2, upregulating their expression, and increased the biochemical activity of TF and PAI-1. Upregulation of Egr-1 and its target genes by heme-induced oxidant stress may be an important event in the initiation and progression of inflammatory vascular diseases such as atherosclerosis.


Subject(s)
Early Growth Response Protein 1/genetics , Hemin/pharmacology , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Smooth Muscle/metabolism , NF-kappa B/metabolism , Up-Regulation/drug effects , ets-Domain Protein Elk-1/metabolism , Cells, Cultured , Early Growth Response Protein 1/drug effects , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/drug effects , Oxidative Stress/drug effects , Promoter Regions, Genetic , Reactive Oxygen Species , Up-Regulation/genetics
11.
J Cardiovasc Pharmacol Ther ; 14(2): 116-24, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19342499

ABSTRACT

Guanosine-specific cyclic nucleotide signaling is suggested to serve protective actions in the vasculature; however, the influence of selective pharmacologic modulation of cyclic guanosine monophosphate- synthesizing soluble guanylate cyclase or cyclic guanosine monophosphate-degrading phosphodiesterase on vessel remodeling has not been thoroughly examined. In this study, rat carotid artery balloon injury was performed and the growth-modulating effects of the soluble guanylate cyclase stimulator YC-1 or the cyclic guanosine monophosphate-dependent phosphodiesterase-V inhibitor zaprinast were examined. YC-1 or zaprinast elevated vessel cyclic guanosine monophosphate content, reduced medial wall and neointimal cell proliferation, stimulated medial and neointimal cellular apoptosis, and markedly attenuated neointimal remodeling in comparable fashion. Interestingly, soluble guanylate cyclase inhibition by 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one failed to noticeably alter neointimal growth, and concomitant zaprinast with YC-1 did not modify any parameter compared to individual treatments. These results provide novel in vivo evidence that YC-1 and zaprinast inhibit injury-induced vascular remodeling through antimitogenic and proapoptotic actions and may offer promising therapeutic approaches against vasoproliferative disorders.


Subject(s)
Carotid Artery Injuries/drug therapy , Cyclic GMP/metabolism , Indazoles/pharmacology , Purinones/pharmacology , Animals , Apoptosis/drug effects , Carotid Artery Injuries/physiopathology , Cell Proliferation/drug effects , Disease Models, Animal , Enzyme Activators/pharmacology , Guanylate Cyclase/drug effects , Guanylate Cyclase/metabolism , Male , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley
12.
J Am Soc Echocardiogr ; 32(12): 1574-1585, 2019 12.
Article in English | MEDLINE | ID: mdl-31587969

ABSTRACT

BACKGROUND: Myeloproliferative neoplasm (MPN) has been associated with pulmonary hypertension (PH) on the basis of small observational studies, but the mechanism and clinical significance of PH in MPN are not well established. The aims of this study were to expand understanding of PH in a well-characterized MPN cohort via study of PH-related symptoms, mortality risk, and cardiac remodeling sequalae of PH using quantitative echocardiographic methods. METHODS: The population comprised a retrospective cohort of patients with MPN who underwent transthoracic echocardiography: Doppler-derived pulmonary arterial systolic pressure applied established cutoffs for PH (≥35 mm Hg) and advanced PH (≥50 mm Hg); right ventricular (RV) performance was assessed via conventional indices (tricuspid annular plane systolic excursion [TAPSE], S') and global longitudinal strain. Symptoms and mortality were discerned via standardized review. RESULTS: Three hundred one patients were studied; 56% had echocardiography-demonstrated PH (20% advanced) paralleling a high prevalence (67%) among patients with invasively quantified PASP. PH was associated with adverse left ventricular (LV) remodeling indices, including increased myocardial mass and diastolic dysfunction (P ≤ .001 for all): LV mass and filling pressure (P < .01) were associated with PH independent of LV ejection fraction. RV dysfunction by strain and TAPSE and S' increased in relation to PH (P ≤ .001) and was about threefold greater among patients with advanced PH compared with those without PH. Patients with RV dysfunction were more likely to report dyspnea, as were those with advanced PH (P < .05). During median follow-up of 2.2 years, all-cause mortality was 27%. PH grade (hazard ratio, 1.9; 95% CI, 1.1-3.0; P = .012) and TAPSE- and S'-demonstrated RV dysfunction (hazard ratio, 3.3; 95% CI, 1.3-8.2; P = .01) were independently associated with mortality; substitution of global longitudinal strain for TAPSE and S' yielded similar associations of RV dysfunction with death (hazard ratio, 3.2; 95% CI, 1.5-6.7; P = .003) independent of PH. CONCLUSIONS: PH is highly prevalent in patients with MPN and is linked to LV diastolic dysfunction; echocardiography-quantified RV dysfunction augments risk for mortality independent of PH.


Subject(s)
Heart Ventricles/diagnostic imaging , Hypertension, Pulmonary/complications , Neoplasms/complications , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right/physiology , Aged , Disease Progression , Echocardiography, Doppler/methods , Female , Follow-Up Studies , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/physiopathology , Male , Middle Aged , Retrospective Studies , Ventricular Dysfunction, Right/diagnosis , Ventricular Dysfunction, Right/etiology
13.
Circ Res ; 99(6): 598-606, 2006 Sep 15.
Article in English | MEDLINE | ID: mdl-16931800

ABSTRACT

We previously reported that hyperhomocysteinemia (HHcy), an independent risk factor of coronary artery disease (CAD), is associated with increased atherosclerosis and decreased plasma high-density lipoprotein cholesterol (HDL-C) in cystathionine beta-synthase-/apolipoprotein E-deficient (CBS(-/-)/apoE(-/-)) mice. We observed that plasma homocysteine (Hcy) concentrations are negatively correlated with HDL-C and apolipoprotein A1 (apoA-I) in patients with CAD. We found the loss of large HDL particles, increased HDL-free cholesterol, and decreased HDL protein in CBS(-/-)/apoE(-/-) mice, and attenuated cholesterol efflux from cholesterol-loaded macrophages to plasma in CBS(-/-)/apoE(-/-) mice. ApoA-I protein was reduced in the plasma and liver, but hepatic apoA-I mRNA was unchanged in CBS(-/-)/apoE(-/-) mice. Moreover, Hcy (0.5 to 2 mmol/L) reduced the levels of apoA-I protein but not mRNA and inhibited apoA-1 protein synthesis in mouse primary hepatocytes. Further, plasma lecithin:cholesterol acyltransferase (LCAT) substrate reactivity was decreased, LCAT specific activity increased, and plasma LCAT protein levels unchanged in apoE(-/-)/CBS(-/-) mice. Finally, the clearance of plasma HDL cholesteryl ester, but not HDL protein, was faster in CBS(-/-)/apoE(-/-) mice, correlated with increased scavenger receptor B1, and unchanged ATP-binding cassette transporter A1 protein expression in the liver. These findings indicate that HHcy inhibits reverse cholesterol transport by reducing circulating HDL via inhibiting apoA-I protein synthesis and enhancing HDL-C clearance.


Subject(s)
Apolipoprotein A-I/biosynthesis , Cholesterol, HDL/metabolism , Hyperhomocysteinemia/metabolism , Lipoproteins, HDL/blood , Aged , Animals , Apolipoprotein A-I/antagonists & inhibitors , Apolipoproteins E/deficiency , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Coronary Artery Disease/etiology , Cystathionine/deficiency , Hepatocytes/metabolism , Homocysteine/blood , Humans , Hyperhomocysteinemia/blood , Male , Mice , Mice, Knockout , Middle Aged
14.
Arterioscler Thromb Vasc Biol ; 27(9): 1976-83, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17715404

ABSTRACT

OBJECTIVE: We previously reported that homocysteine (Hcy) inhibits endothelial cell (EC) growth and promotes vascular smooth muscle cell (VSMC) proliferation. This study characterized and directly compared Hcy transport in cultured human aortic ECs (HAECs) and smooth muscle cells (HASMCs). METHODS AND RESULTS: Hcy (10 micromol/L) was transported into both cell types in a time-dependent fashion but was approximately 4-fold greater in HASMCs, and is nonstereoenantiomer specific. Hcy transport in HAECs had a Michaelis-Menten constant (Km) of 39 micromol/L and a maximal transport velocity (Vmax) of 873 pmol/mg protein/min. In contrast, Hcy transport in HASMCs had a lower affinity (Km = 106 micromol/L) but a higher transport capacity (Vmax = 4192 pmol/mg protein/min). Competition studies revealed that the small neutral amino acids tyrosine, cysteine, glycine, serine, alanine, methionine, and leucine inhibited Hcy uptake in both cell types, but the inhibition was greater for tyrosine, serine, glycine, and alanine in HAECs. Sodium-depletion reduced Hcy transport to 16% in HAECs and 56% in HASMCs. Increases in pH from 6.5 to 8.2 or lysosomal inhibitors blocked Hcy uptake only in HAECs. In addition, Hcy shares carrier systems with cysteine, in a preferable order of alanine-serine-cysteine (ASC) > aspartate and glutamate (X(AG)) = large branched-chain neutral amino acids (L) transporter systems in HAECs and ASC > L > X(AG) in HASMCs. The sodium-dependent system ASC plays a predominant role for Hcy transport in vascular cells. CONCLUSIONS: Transport system ASC predominantly mediates Hcy transport in EC and is lysosomal dependent.


Subject(s)
Amino Acid Transport System ASC/physiology , Endothelial Cells/physiology , Homocysteine/metabolism , Myocytes, Smooth Muscle/physiology , Aorta/cytology , Biological Transport, Active/physiology , Cells, Cultured , Humans , Kinetics , Lysosomes , Muscle, Smooth, Vascular/cytology
15.
Arterioscler Thromb Vasc Biol ; 25(12): 2515-21, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16210565

ABSTRACT

OBJECTIVE: A risk factor for cardiovascular disease, hyperhomocystinemia (HHcy), is associated with endothelial dysfunction. In this study, we examined the mechanistic role of HHcy in endothelial dysfunction. METHODS AND RESULTS: Through the use of 2 functional models, aortic rings and intravital video microscopy of the cremaster, we found that arterial relaxation in response to the endothelium-dependent vessel relaxant, acetylcholine or the nitric oxide synthase (NOS) activator (A23187), was significantly impaired in cystathionine beta-synthase null (CBS(-/-)) mice. However, the vascular smooth muscle cell (VSMC) response to the nitric oxide (NO) donor (SNAP) was preserved in CBS(-/-) mice. In addition, superoxide dismutase and catalase failed to restore endothelium-dependent vasodilatation. Endothelial nitric oxide synthase (eNOS) activity was significantly reduced in mouse aortic endothelial cells (MAECs) of CBS(-/-) mice, as well as in Hcy-treated mouse and human aortic endothelial cells (HAECs). Hcy-mediated eNOS inhibition--which was not rescued by adenoviral transduction of superoxide dismutase and glutathione peroxidase, or by tetrahydrobiopterin, sepiapterin, and arginine supplementations in MAEC--was associated with decreased protein expression and increased threonine 495 phosphorylation of eNOS in HAECs. Ultimately, a protein kinase C (PKC) inhibitor, GF109203X (GFX), reversed Hcy-mediated eNOS inactivation and threonine 495 phosphorylation in HAECs. CONCLUSIONS: These data suggest that HHcy impairs endothelial function and eNOS activity, primarily through PKC activation.


Subject(s)
Endothelium, Vascular/enzymology , Hyperhomocysteinemia/metabolism , Nitric Oxide Synthase Type II/metabolism , Protein Kinase C/metabolism , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Aorta, Thoracic/cytology , Arginine/analogs & derivatives , Arginine/blood , Arginine/metabolism , Arginine/pharmacology , Biopterins/analogs & derivatives , Biopterins/metabolism , Biopterins/pharmacology , Cells, Cultured , Cystathionine beta-Synthase/genetics , Cystathionine beta-Synthase/metabolism , Endothelium, Vascular/cytology , Enzyme Activation/physiology , Female , Gene Expression Regulation, Enzymologic , Glutathione Peroxidase/genetics , Homocysteine/blood , Humans , Hyperhomocysteinemia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type III , Pterins/metabolism , Pterins/pharmacology , Superoxide Dismutase/genetics , Vasodilation/physiology , Glutathione Peroxidase GPX1
16.
Cleve Clin J Med ; 73(8): 767-74, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16913202

ABSTRACT

For most patients, elevated platelet counts are benign and require no treatment, but for some, severe complications or death may ensue. This article discusses how to classify thrombocytosis and identify which patients require treatment, describes the characteristic complications that may arise, and provides an algorithm for management.


Subject(s)
Thrombocytosis , Algorithms , Diagnosis, Differential , Hemorrhage/etiology , Humans , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/diagnosis , Thrombocytosis/diagnosis , Thrombocytosis/etiology , Thrombocytosis/therapy , Thrombopoietin/metabolism , Thrombosis/etiology
17.
FASEB J ; 18(6): 768-70, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14977877

ABSTRACT

Platelet-derived growth factor (PDGF) contributes to vascular disease by stimulating the growth of vascular smooth muscle cells (SMCs). Since amino acids are required for cell growth, the present study examined the effect of PDGF on system L amino acid transport, which is the predominant cellular pathway for the uptake of essential amino acids. System L amino acid transport was monitored by measuring the uptake of L-leucine. Treatment of SMCs with PDGF stimulated L-leucine transport in a concentration- and time-dependent manner, and this was associated with a selective increase in LAT1 mRNA and protein. PDGF failed to induce the expression of the other system L transport proteins, LAT2 and the heavy chain of the 4F2 cell surface antigen. The induction of LAT1 by PDGF was dependent on de novo RNA and protein synthesis and on mTOR activity. Serum, thrombin, and angiotensin II likewise stimulated L-leucine transport by inducing LAT1 expression. Inhibition of system L amino acid transport by the model substrate 2-aminobicyclo-(2,2,1)-heptane-2-carboxylic acid blocked growth factor-mediated SMC proliferation and induced SMC apoptosis, whereas it had no effect on quiescent cells. These results demonstrate that growth factors stimulate system L amino acid transport by inducing LAT1 gene expression and that system L amino acid transport is essential for SMC proliferation and survival. The capacity of vascular mitogens to induce LAT1 expression may represent a basic mechanism by which tho acid transport * apoptosis


Subject(s)
Large Neutral Amino Acid-Transporter 1/biosynthesis , Muscle, Smooth, Vascular/metabolism , Platelet-Derived Growth Factor/pharmacology , Amino Acid Transport System L/metabolism , Animals , Cell Division , Cell Survival , Gene Expression Regulation , Large Neutral Amino Acid-Transporter 1/genetics , Models, Biological , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Protein Kinases/metabolism , Rats , TOR Serine-Threonine Kinases
18.
Cardiovasc Res ; 55(2): 396-405, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-12123779

ABSTRACT

OBJECTIVE: Carbon monoxide (CO) is generated from vascular smooth muscle cells via the degradation of heme by the enzyme heme oxygenase-1. Since smooth muscle cell apoptosis is associated with numerous vascular disorders, we investigated whether CO regulates apoptosis in vascular smooth muscle. METHODS AND RESULTS: Treatment of cultured rat aortic smooth muscle cells with a combination of cytokines (interleukin-1beta, 5 ng/ml; tumor necrosis factor-alpha, 20 ng/ml; interferon-gamma, 200 U/ml) for 48 h stimulated apoptosis, as demonstrated by DNA laddering, annexin V binding, and caspase-3 activation. However, the exogenous administration of CO inhibited cytokine-mediated apoptosis. The antiapoptotic action of CO was partially dependent on the activation of soluble guanylate cyclase and was associated with the inhibition of mitochondrial cytochrome c release and with the suppression of p53 expression. Incubation of smooth muscle cells with the cytokines also resulted in a pronounced increase in heme oxygenase-1 protein after 24 h of stimulation. The addition of the heme oxygenase inhibitor, zinc protoporphyrin-IX, or the CO scavenger, hemoglobin, stimulated apoptosis following 24 h of cytokine exposure. CONCLUSIONS: These results demonstrate that CO, either administered exogenously or endogenously derived from heme oxygenase-1 activity, inhibits vascular smooth muscle cell apoptosis. The ability of CO to block smooth muscle cell apoptosis may play an important role in blocking lesion formation at sites of vascular injury.


Subject(s)
Apoptosis/drug effects , Carbon Monoxide/pharmacology , Muscle, Smooth, Vascular/drug effects , Animals , Carbon Monoxide/physiology , Cell Culture Techniques , Cytochrome c Group/metabolism , Cytokines/antagonists & inhibitors , Cytokines/pharmacology , Dose-Response Relationship, Drug , Guanylate Cyclase/physiology , Heme Oxygenase (Decyclizing)/metabolism , Heme Oxygenase-1 , Male , Mitochondria/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats
19.
FEBS Lett ; 541(1-3): 52-6, 2003 Apr 24.
Article in English | MEDLINE | ID: mdl-12706818

ABSTRACT

Since apoptosis of endothelial cells (ECs) plays an important role in the pathogenesis of atherosclerosis, we investigated the effect of cyclic stretch on EC apoptosis. Application of moderate, physiologic levels of cyclic stretch (6-10% at 1 Hz) inhibited EC apoptosis. This anti-apoptotic effect was dependent on the activation of phosphatidylinositol 3-kinase and associated with the activation of Akt and the phosphorylation of Bad. Interestingly, a higher potentially pathologic level of cyclic stretch (20% at 1 Hz) stimulated EC apoptosis. The ability of physiologic cyclic stretch to inhibit EC apoptosis may provide a previously unrecognized mechanism by which hemodynamic forces exert an anti-atherogenic effect.


Subject(s)
Apoptosis , Endothelium, Vascular/metabolism , Protein Serine-Threonine Kinases , Animals , Cattle , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Nitric Oxide/physiology , Periodicity , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Stress, Mechanical
20.
Am J Hypertens ; 17(8): 712-7, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15288886

ABSTRACT

BACKGROUND: The increase in vessel wall strain in hypertension contributes to arterial remodeling by stimulating vascular smooth muscle cell (SMC) proliferation and collagen synthesis. Because L-proline is essential for the synthesis of collagen and cell growth, we examined whether cyclic strain regulates the transcellular transport of L-proline by vascular SMC. METHODS: Cultured rat aortic SMCs were subjected to mechanical strain using the Flexercell 3000 Strain Unit. RESULTS: Cyclic strain increased L-proline transport in a time- and strain-degree-dependent manner that was inhibited by cycloheximide or actinomycin D. Kinetic studies indicated that cyclic strain-induced L-proline uptake was mediated by an increase in transport capacity independent of any change in the affinity for L-proline. Cyclic strain stimulated the expression of system A amino acid transporter 2 mRNA in a time-dependent fashion that paralleled the increase in L-proline transport. Cyclic strain also induced the release of transforming growth factor-beta1 in a time- and strain-dependent manner. Moreover, conditioned media from SMCs exposed to cyclic strain stimulated the transport of L-proline in control, static SMCs and this was significantly attenuated by a transforming growth factor-beta1 neutralizing antibody. CONCLUSIONS: These results demonstrate that cyclic strain stimulates L-proline transport by inducing system A amino acid transporter 2 gene expression through the autocrine release of transforming growth factor-beta1. The ability of cyclic strain to induce system A amino acid transporter 2 expression may promote arterial remodeling in hypertension by providing vascular SMCs with the necessary intracellular levels of L-proline required for collagen synthesis and cell growth.


Subject(s)
Blood Pressure/physiology , Hypertension/metabolism , Muscle, Smooth, Vascular/metabolism , Proline/pharmacokinetics , Amino Acid Transport System A/genetics , Animals , Aorta, Thoracic/cytology , Autocrine Communication/physiology , Cells, Cultured , Muscle, Smooth, Vascular/cytology , RNA, Messenger/metabolism , Rats , Stress, Mechanical , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1
SELECTION OF CITATIONS
SEARCH DETAIL