干细胞之家 - 中国干细胞行业门户第一站

 

 

搜索
朗日生物

免疫细胞治疗专区

欢迎关注干细胞微信公众号

  
查看: 388143|回复: 240
go

Endothelial cell dysfunction: can*t live with it, how to live without it

Rank: 1

积分
威望
0  
包包
0  
发表于 2009-4-21 13:01 |显示全部帖子
Division of Nephrology and Renal Research Institute, Departments of Medicine and Pharmacology, New York Medical College, Valhalla, New York8 \0 a  m2 u) [  e5 n( K/ L
* J+ y# I! n* U$ T# w. c
ABSTRACT, U' F% n  G7 c
: ^3 Y3 @- t& M) G. L6 X# F* K
Endothelial cell dysfunction is emerging as an ultimate culprit for diverse cardiovascular diseases and cardiovascular complications of chronic renal diseases, yet the definition of this new syndrome, its pathophysiology, and therapy remain poorly defined. Here, I summarize some molecular mechanisms leading from hyperhomocystinemia, elevated asymmetric dimethylarginine, and advanced glycolation end product-modified protein level to the proatherogenic, prothrombogenic, and proinflammatory endothelial phenotype and offer a model of endothelial dysfunction based on the interconnectedness of diverse functions. Finally, several therapeutic strategies to prevent and correct endothelial dysfunction are discussed in the light of uncertainty of their action modulated by the endothelial dysfunction per se.
3 B; }$ X& P+ y( \0 Y
8 {' G9 {+ v: ~/ _6 e. S/ @uncoupled endothelial nitric oxide synthase; oxidative stress; vascular wall; atherogenesis6 Q' ~1 Z1 z% c# h, D

) _% T$ `- b5 \* |$ ~- w+ N, H5 uTOWARD THE DEFINITION OF ENDOTHELIAL DYSFUNCTION: THE SYNDROME IN THE MAKING" m' D9 P; C9 ~7 B

& o" y  U; I" W& ~/ X; m' fA COMPUTER SEARCH OF THE INTERNATIONAL database Ovid shows that since 1983 the number of entries containing the term "endothelial dysfunction" as a keyword has been exponentially increasing. The term was first used to describe a defect in the removal of 5-hydroxytryptamine and norepinephrine in the pulmonary circulation of rabbits intratracheally injected with bleomycin, an agent causing the development of pulmonary fibrosis (20) and, a year later, in reference to the decrease in angiotensin-converting enzyme and plasminogen activator activities, with no change in prostacyclin production by the pulmonary endothelium, after monocrotaline-induced lung injury (70). These investigators concluded that "monocrotaline-induced pulmonary injury is accompanied, and in some cases preceded, by structural and functional abnormalities in the pulmonary endothelium." The modern usage of the term is associated with the study by Ludmer and co-workers (66), who observed that acetylcholine-induced vasorelaxation is impaired, even reversed, in atherosclerotic coronary arteries and this abnormality "occurs early... in the course of coronary atherosclerosis." The same group of investigators has reported a similar angiographic pattern after coronary injection of acetylcholine in recipients of cardiac transplants (38). They hypothesized that the "impaired response to acetylcholine is a common early finding in heart transplant patients and emphasizes the potential importance of endothelial dysfunction in the development of atherosclerosis." A host of studies by other investigative groups has confirmed these observations (35, 80, 81, 110). Because the term endothelial dysfunction has not only persevered during the past two decades but is also being used (and sometimes perhaps misused) with increasing frequency, despite the lack of a proper definition, an attempt to summarize the accrued knowledge and define the syndrome would be timely., R* e$ U# U! E9 {8 i( {. Z, h6 \

- U! X. e% j3 Q$ aThe last 20 years have established that the vascular endothelium, rather than being a mere barrier between intravascular and interstitial compartments, is a widely spread organ responsible for the regulation of hemodynamics, angiogenic vascular remodeling, metabolic, synthetic, anti-inflammatory, and antithrombogenic processes. Based on the diversity of endothelial functions, it is logical to expect that the definition of the syndrome of "endothelial cell dysfunction" (ECD) should be broad enough to encompass disturbances in the barrier function of the vascular endothelium; its impaired antithrombogenic properties; perturbed angiogenic competence; inappropriate regulation of vascular smooth muscle tonicity, proliferative capacity, and migratory properties; perturbed synthetic functions; and deterrence of neutrophils and monocytes from diapedesis (Fig. 1, a diagrammatic presentation of ECD as a transportation hub harboring diverse functional units). The phenotype of endothelial cells characterized by these abnormalities, expressed in various degrees, is emerging as a hallmark of several highly prevalent cardiovascular and renal diseases, including obesity and diabetes, as well as their complications. I shall discuss this broadened definition of ECD below.+ Z9 p8 F# }$ ?0 v* I% n
# \  D0 c+ M! K% q. Z
PATHOGENETIC MECHANISMS% [4 h  n$ C/ K! O; i3 }
$ E6 [8 c& Z$ I5 _5 d6 i
The convergence of traditional risk factors, genetic predisposition, and local and yet unknown factors acting on endothelial cells all contributes to the development of ECD, the "ultimate risk of the risk factors" (10). In addition to traditional cardiovascular risk factors, a host of complementary mechanisms responsible for the high prevalence of cardiovascular complications has been described. Among the nontraditional risk factors that appear to gain in importance are elevated asymmetric dimethylarginine (ADMA) levels, hyperhomocystinemia (HHCy), and protein modification by nonenzymatic advanced glycation. Several years ago, we elected to study the cellular and molecular derangements induced by the above pathogenic stimuli using nonbiased functional genomic screening. Toward this end, we performed cDNA microarray screening of "cardiovascular relevant" genes modified by two major contributors to ECD in chronic renal disease, homocysteine and the nitric oxide synthase (NOS) inhibitor NG-nitro-L-arginine methyl ester (L-NAME), partially mimicking the effect of ADMA (it is necessary to emphasize, however, that these inhibitors are not equivalent). The results of these studies into some cellular consequences of several cDNA screens have been published; thus I shall describe briefly only the essential observations (23, 59–61, 88, 92, 111).
9 S: [6 J2 V' n5 e( W$ N
/ g+ X1 {4 F6 H; m) O5 zFunctional analysis and retesting of some findings using RT-PCR and Western blotting showed that pathophysiologically relevant concentrations of homocysteine result in a profound induction of 3-hydroxy-3-methylglutaryl coenzyme A reductase, increased synthesis and accumulation of cholesterol in the endothelial cells, and uncoupling of endothelial NOS (eNOS) due to oxidative stress, all manifestations of developing endothelial lipidosis (by analogy with hepatic lipidosis) (60). Upregulation of connexin-43 expression was accompanied by the displacement of this gap junctional protein from the plasma membrane to the mitochondrion, depletion of myo-endothelial gap junctional communication, and the failure of the putative endothelium-derived hyperpolarizing factor to traverse myo-endothelial junctions and relax vascular smooth muscle cells (61). Together with the uncoupling of eNOS (111), these findings explain the complex pathogenesis of vasomotor failure. These and other abnormalities and their role in the development of ECD are schematically illustrated in Fig. 2.
% A4 S3 B; L  t5 P  e( ~4 }" R* o. N  f% v& [0 b
The functional sequelae of eNOS inhibition with L-NAME, partially confirmed using ADMA by RT-PCR and Western blot analyses, are presented in Fig. 3. Inhibition of NO synthesis leads to the derangement not only of endothelium-dependent vasorelaxation but also to the proatherogenic sequelae, including enhanced adhesion and transmigration of monocytes and enhanced platelet aggregation. Upregulation of LOX-1, a major receptor for oxidized LDL, results in increased oxidative stress and lipid accumulation in endothelial cells (88, 89). Increased synthesis of several chains of collagen and induction of integrin receptors participate in the switch of endothelial cells toward the profibrotic phenotype (O'Riordan E and Goligorsky MS, unpublished observations). Overexpression of plasminogen activator inhibitor (PAI-1) by endothelial cells presented with advanced glycolation end product (AGE)-modified collagen I is responsible for the early upregulation of PAI-1, which is causatively linked to the inappropriate formation of capillary networks during diabetic and/or end-stage renal disease (ESRD) wound healing and eventual vascular dropout (23).
4 |7 ~4 b# @. E7 _. e* Y8 a9 S3 }6 C: r
It has been debated whether eNOS gene polymorphism contributes to ECD. Several potential candidate sites have been identified, including Glu298Asp, -786T>C, and intron 4 polymorphisms and incriminating Asp298, -786C allele in the promoter and intron 4 with the increased risk of cardiovascular complications and its association with ESRD (75). A recent meta-analysis of 26 studies involving more than 23,000 subjects has demonstrated that homozygosity for Asp298 and intron 4a alleles of eNOS results in a moderately increased risk of ischemic heart disease (19). Gene polymorphism of other components of the system is beyond the scope of this review.
  z! _. U- }, `9 A6 \; z( `
: X( }/ D9 X% A2 }CLINICAL SIGNS OF ECD0 \7 s1 J0 d( }( Q

4 j8 i9 X6 _7 O9 I* p: [7 @: NManifestations of ECD are many and stem from the aberrations in individual functions of the endothelium. Figure 1 depicts the relationships between perturbed endothelial functions and the ensuing clinical manifestations. These include hypertension, macro- and microvasculopathy due to the endothelial lipidosis and atherogenesis, impaired deterrence of inflammatory cells, increased vascular permeability manifesting as microalbuminuria, and impaired angiogenic competence. Each of these manifestations has been extensively reviewed elsewhere (41, 79, 105).% a$ g" v4 A8 a

; y5 ?" p7 g" S" C3 ~FINAL COMMON PATHWAY: REACTIVE OXYGEN INTERMEDIATES AND REACTIVE NITROGEN INTERMEDIATES LEADING TO ECD, PREMATURE ENDOTHELIAL CELL SENESCENCE, AND APOPTOSIS* b" s1 {2 V3 {
  d$ e! v* I! J! r3 h; S* e
Diverse risk factors and pathological processes targeting the vascular endothelium elicit a default response, excessive generation of reactive oxygen intermediates and reactive nitrogen intermediates, the now well-established centerpiece of the response-to-injury hypothesis of atherosclerosis (46, 82). This uniformly present component of pathological reaction modifies endothelial cell functions and leads to cell demise via premature senescence and apoptosis. We have recently demonstrated an increased frequency of prematurely senescent cells in vivo in aortas of young Zucker diabetic fat rats with chronic renal insufficiency, compared with lean controls. NO production by these senescent endothelial cells was decreased in association with the tissue accumulation of nitrotyrosine-modified proteins, a footprint of oxidative and nitrosative stress (12, 24). Development of premature senescence of endothelial cells in vitro could be prevented and reversed by treatments with the peroxynitrite scavenger ebselen, the eNOS intermediate N-hydroxy-L-arginine (NOHA), or the SOD mimetic Mn(III)tetrakis(4-benzoic acid) porphyrin chloride. Concomitant with the reversal of senescence, ebselen and NOHA each restored NO production to control levels. Chronic treatment of Zucker diabetic fat rats with ebselen not only prevented, but also partially reversed, ECD (12). These findings indicate that the metabolic syndrome with chronic renal insufficiency in vivo elicits premature senescence of the vascular endothelium. Premature senescence of the vascular endothelium, a consequence of reduced NO availability and peroxynitrite and/or superoxide excess, is presumed to be an important contributor to vasculopathy. These metabolic derangements, together with the oxidized LDL, TNF-, and peroxynitrite, eventually lead to the increased incidence of apoptosis of endothelial cells, further contributing to accelerated atherosclerosis (reviewed in Ref. 83). The proposed cascade of events along the pathway of endothelial cell involution triggered by different risk factors is depicted in Fig. 4. It predicts that various traditional and nontraditional risk factors, at times accompanied by an acute event, acting on endothelial cells lead to the excessive generation of reactive oxygen intermediates, reactive nitrogen intermediates, and acquisition of a proinflammatory, prothrombogenic, and proatherogenic phenotype, eventually affecting cell cycle arrest and premature senescence. The latter is characterized by an overt vasculopathy, which is reversible in Zucker diabetic fat rats. The predominance of apoptotic death of endothelial cells heralds irreversible damage and may be accompanied by vascular rarefaction.
- g3 y+ g6 W, T
9 y% I+ [3 M6 XCLINICAL ASSESSMENT
4 K( `# |0 y! O8 `0 d
4 S5 H) ^  h) q$ F: h' |- JThe principle of uncertainty is best seen in clinical settings. By detecting the syndrome at preclinical stages, at present a particularly difficult task, there is a chance of reversing it. Detecting the syndrome with certainty through its ominous manifestations may turn out to be too late a stage for any meaningful therapy to halt its progression or reverse it. The "men born in 1914" plethysmographic study with a more than 20-year follow-up provided an early indication of the possible role of ECD in increased mortality (48). However, due to the relative novelty of the syndrome and the lack of reliable noninvasive ways of diagnosing it, clinical assessment of patients remains unsatisfactory. These problems underscore the existing lack of well-established criteria for the diagnosis of ECD. A variety of surrogate markers of ECD have been proposed, including elevated plasma levels of PAI-1, tissue plasminogen activator, and von Willebrand factor. Stehouwer (90) suggests that "estimates of different types of ECD may be obtained indirectly by measuring endothelium-dependent vasodilation, plasma levels of endothelium-derived regulatory proteins and, possibly, microalbuminuria." Plethysmographic or ultrasonographic measurements of brachial artery responses to flow or acetylcholine have been advocated as promising markers of ECD, but their sensitivity in detecting coronary artery disease (CAD) was found to be 49% (4). Several studies have already characterized impaired macrovascular blood flow responses in ESRD patients (5, 69). Shamim-Uzzaman et al. (86) have compared brachial artery flow-mediated dilatation with cutaneous microcirculation monitored using laser Doppler flowmetry in patients with CAD and demonstrated no differences in flow-mediated dilatation between CAD and control subjects, whereas there were significant abnormalities in the microcirculatory vasodilatory responses. Our studies of ESRD patients using laser Doppler flowmetry and imaging showed that several parameters characterizing reactive and thermal hyperemia are impaired in a majority of these patients, regardless of the presence of known CAD (91). By interrogating the microvasculature with changing shear stress or temperature, it is possible to unmask the states of vasodilatory failure. The available data imply that microvasculopathy is highly prevalent in this cohort of patients, even when there are no clinical manifestations of CAD.
: ~! m9 P7 S( z/ V7 m& o1 Z2 s: C" r4 x
UNCERTAINTY IN THE OUTCOMES OF THERAPEUTIC INTERVENTIONS
) x" C* O( x! U( `. _; M$ v+ s! W  ~# j! S# h- D4 z. Y! H
Attempts at correcting ECD have been mounting, but their success rate remains low to variable, as will be discussed below. Therapeutic interventions, which appear to be pathophysiologically rational, occasionally turned out to produce some paradoxical results. The main point of the forthcoming discussion will be to demonstrate these unexpected results and their possible explanation on the basis of the coincidental expression of "uncoupled" eNOS, which introduces an aberration into the seemingly rational therapy of ECD. The outcomes appear to be largely dependent on the preexisting status of the vascular endothelium.1 ~" Z4 |  L% [

/ _1 B, c7 `/ [6 m/ leNOS Gene and Protein Delivery& _2 s  n) F8 `
  i0 G  b  \% u7 z9 w) m3 `3 @
Ex vivo gene transfer of eNOS into atherosclerotic aortic rings resulted in an improved acetylcholine-induced vasodilation (73). There is recent experimental evidence for the inhibition of neointimal vascular lesions by in vivo transfer of the eNOS gene (106). Using recombinant eNOS for transduction of human saphenous veins, Cable et al. (16) showed augmented NO production in coronary artery bypass conduits. In vivo eNOS gene transfer has been shown to reduce hypertension, enhance vasomotor function of carotid arteries in hypercholesterolemic rabbits, and restore NO availability in stroke-prone spontaneously hypertensive rats (2, 3, 21, 63). However, overexpression of eNOS has been shown to accelerate development of atherosclerotic lesions in apolipoprotein E-deficient mice due to uncoupling of the enzyme and enhanced generation of superoxide (77). These data provide a cautionary note on the biological context dependence of gene therapy.; A2 y2 f4 Q; w9 }; z" F$ E% d

% E8 B' o% Q. C- J5 b5 C: ^* l' ]Local gene transfer has been accomplished by wrapping a silicone collar around the adventitial surface of arteries and using it as a reservoir for plasmid/liposome-mediated VEGF gene transfer. Overexpression of VEGF prevented intimal thickening in a NO-dependent fashion. Whether such an approach can be utilized to effectively limit neointimal vascular lesions at the sites of anastomosed vessels or balloon catheter-induced endothelial injury remains to be established. Khurana et al. (56) have recently summarized potential targets for gene therapy including VEGF, FGF, HGF, angiopoietins, eNOS, and others and expressed cautious optimism for their use as cardiovascular therapeutics. One should keep in mind, however, that as simple a procedure as treadmill exercise led to increased expression of eNOS and extracellular SOD in mice (31).6 ^3 H: [& ]! J1 W
' V2 v. t8 D9 u. F+ w
Correction of ECD by Supplying eNOS Substrate
! P/ `( I6 x7 ^  }* ^" q4 M9 i1 y$ _$ Y" _
Both an absolute L-arginine deficiency or a relative deficiency due to the retention of ADMA may result in eNOS uncoupling. Several investigative groups have demonstrated that L-arginine infusion restored the acetylcholine-induced coronary vasorelaxation in hypercholesterolemic patients who, before L-arginine, responded to acetylcholine with paradoxical vasoconstriction and that L-arginine supplementation improved endothelium-dependent vasodilatation in cholesterol-fed rabbits and patients with hypercholesterolemia (29, 35, 39). In addition, L-arginine appears to ameliorate reperfusion injury after myocardial ischemia, inhibit intimal hyperplasia after balloon catheterization-induced endothelial injury, and attenuate endothelial adhesiveness in hypercholesterolemia (64, 96, 105). On the other hand, a randomized, double-blind, placebo-controlled study of L-arginine supplementation in patients with moderate chronic renal failure was deemed ineffective in improving renal function (29). Failure to improve endothelial function was reported in pediatric and adult patients with chronic renal insufficiency (7, 30). In a group of healthy subjects, systemic infusion of L-arginine was found to induce vasodilation and inhibit platelet aggregation, effects that were in part attributable to the stimulation of insulin release (40). In hypercholesterolemic rabbits, supplementation with L-arginine results in a regression of preexisting lesions. This effect is attributed to the induction of apoptosis of macrophages in atheromas (107). Boger and Bode-Boger (9) provided an excellent review of clinical pharmacology of L-arginine, summarizing diseases in which L-arginine beneficially affected cardiovascular end points (exercise tolerance, anginal symptoms, vasospastic attacks): peripheral arterial disease, CAD, congestive heart failure, hypercholesterolemia, and Raynaud syndrome.
# X* p- h) V! A  Z& Q1 O% W% a, |: l  L% X/ w: v6 e
Adverse effects of supplemental L-arginine have been reported as well (22). Loscalzo (65) provided a plausible conceptual framework for the balance between benefits vis-角-vis drawbacks of such a therapy by indicating that only a small fraction of this amino acid is utilized as a NOS substrate, whereas a much larger portion of the L-arginine pool is used in the synthesis of creatine. L-Arginine overload may result, therefore, in the increase in homocysteine formation and methylation stress, which could counterbalance any benefits of providing NOS with the substrate. These consideration should discourage any uncontrolled use of L-arginine in clinical settings.
! @& q0 [) @6 ]4 T& p# {6 w* a
5 b# C* ~3 K: X) ^+ hAn intriguing observation that short polymers of L-arginine cross plasma membranes independently of the basic amino acid transporter (which serves to carry arginine monomers) has led to preliminary trials of L-arginine heptamers for correction of ECD (100). It was demonstrated that short polymers of arginine inhibit myointimal hyperplasia. Attempts to inhibit the production or accelerate the catabolism of ADMA have not yet produced pharmacologically useful in vivo inhibitors (101).2 u: s. t3 K( n0 t# L

2 ~1 t6 E- n2 F4 dNOHA, an intermediate in the synthesis of NO and a scavenger of superoxide (13, 93, 96), has attracted attention as a potential therapeutic agent. In the recent studies of premature senescence of endothelial cells, Chen et al. (24) showed the ability of NOHA to prevent premature senescence and reverse it with the potency superior to L-arginine.' Q+ s9 p) X  E/ ?9 K% H% k5 L* @
3 `, v; c* P3 r. p) h; S+ O
Correction of Endothelial Dysfunction Due to the Deficiency of eNOS Cofactors
% b0 i4 w0 b: S: A/ k
) {0 `% Y2 a# a7 A- T4 t! q" yThe redox state of the cell and availability of cofactors represent another therapeutic target. Efficacy of ascorbic acid in CAD, congestive heart failure, and vasospastic angina has been reported, in part mediated by regeneration of reduced forms of glutathione and/or tetrahydrobiopterin (49, 52, 53, 97). Improvement of endothelial function with tetrahydrobiopterin was documented in familial hypercholecterolemia, as well as in nitroglycerin-tolerant and insulin-resistant rats (44, 87, 92). A cautionary note has been made that an excessive amount of tetrahydrobiopterin may be deleterious as it can redox-cycle and generate superoxide (95). Even lesser certainty surrounds the efficacy of vitamin E as assessed in randomized clinical studies; except for the Cambridge Heart Antioxidant Study, which showed reduction of nonfatal myocardial infarction, trials have failed to demonstrate a beneficial effect of vitamin E (98).
  }$ P7 f9 P( K8 A/ j# `. `
' w- a' s8 L; w3 [7 N6 EThe third National Health and Nutrition Examination Survey has provided evidence that 50% of the U.S. population had folate intakes below the recommended dietary allowance, and folate deficiency showed a strong correlation with HHCy, a major risk factor for atherosclerosis (85). Supplementation with folic acid (5–10 mg/day) or its active circulating metabolite, 5-methyltetrahydrofolate, restores impaired endothelium-dependent vasodilatation in patients with HHCy or hypercholesterolemia (6, 104). Based on these and other findings (reviewed in Ref. 43), it is predicted that supplementation with folic acid would prevent 4% of deaths due to CAD, thus recommending the need to revise daily folate requirements. Based on the above findings, fortification of cereals with folate was implemented in 1998, already resulting in a 19% decreased incidence of neural tube defect in the United States, reduction of HHCy, but, so far, no detectable reduction of cardiovascular mortality (69).
; e3 G1 R; s- U( c. z6 W
. l3 H' E; b. h3 L; X) VAnother antioxidant, probucol, has been demonstrated to preserve endothelial function in hypercholesterolemic rabbits, thus giving further credibility to the therapeutic strategy of limiting oxidant stress and its consequences in patients with ECD (55). Clinical trials of probucol, however, varied from unsuccessful (13) to efficacious in preventing postangioplasty restenosis (94). Halliwell (45) has provided a comprehensive summary of clinical effects of antioxidants.
2 ?2 _& w/ j% f; W5 c) `
- s! D. p" c( M  c. cCorrection of eNOS Function
, V3 F) ?. ]( P
7 w! R6 K2 Q% i3-Hydroxy-3-methylglutaryl-CoA reductase inhibitors (statins) may exert beneficial effects through reducing the levels of cholesterol and LDL and/or increasing the level of HDL. Liao et al. (62) provided evidence that oxidized LDL decreases eNOS transcription and destabilizes its mRNA. In addition, oxidized LDL may suppress L-arginine uptake by platelets and, by inference, endothelial cells (25). The existing vast literature on atherogenic lipids and endothelial dysfunction has been comprehensively summarized (1). It is also possible that improved levels of HDL may have beneficial effects on endothelial dysfunction (109). It has been appreciated that the effects of 3-hydroxy-3-methylglutaryl-CoA reductase inhibitors on improving endothelial dysfunction transcend their lipid-lowering action (103). Lamas's group (47) has demonstrated that statins prevent the suppression of eNOS mRNA by oxidized LDL in endothelial cells. Another mechanism whereby statins stimulate eNOS involves activation of protein kinase B/Akt and consequent phosphorylation of eNOS (57). Akt-dependent, shear stress-induced activation of eNOS may also be responsible for the improvement in ECD associated with as simple a maneuver as physical exercise (8). Some of the above-mentioned beneficial effects of statins on ECD could be attributed to their ability to generate and regenerate tetrahydrobiopterin (47) and thus restore the coupled state of eNOS.% N; S- \7 K: g

# U3 l+ F: Y8 f. H1 qImprovement in ECD has been noticed in patients with CAD or congestive heart failure receiving angiotensin-converting enzyme inhibitors (reviewed in Ref. 36). Their acute effects on ECD are due to the inhibition of angiotensin II production with the concomitant reduction in oxidative stress and to the bradykinin-dependent stimulation of eNOS (reviewed in Ref. 10). Chronic angiotensin-converting enzyme inhibition has been related to the enhanced expression of eNOS (64) and suppression of PAI-1 (102).
  }/ ]! a2 V2 X9 s; Q
% X6 R! n4 X! j; f* r, e17-Estradiol treatment of hypercholesterolemic rabbits with severe ECD has been found to improve endothelium-dependent vasorelaxation concomitantly with a reduction in the size of atheromas, without altering serum cholesterol level in these animals (34). Similarly, estrogens reduced atherosclerotic lesions in apolipoprotein E-deficient mice (11). One of the potential mechanisms for the observed effects of estrogens has been linked to the estrogen receptor--induced activation of eNOS (26). Hisamoto et al. (51) demonstrated activation of Akt and phosphorylation of eNOS mediated via estrogen receptor- by a nongenomic mechanism. In addition, they showed that activation of eNOS could be mediated by the ERK-dependent cascade, independent of Akt. Clinical trials of estrogens, however, have been terminated. The efficacy of hormone replacement (estrogen and progestin) as well as estrogen replacement therapies to decrease coronary heart disease and incidence of stroke in postmenopausal women "may depend on maintenance of a healthy endothelium," as discussed in a recent excellent review (58), and their failure to achieve these end points could be attributed to uncoupled eNOS.( ]0 k3 V$ s3 b% X* E0 [

; E9 Q' k4 J1 J/ |  n+ Q7 @Peroxisome proliferator-activated receptor- (PPAR) ligands are emerging as potential therapeutics in ECD. PPAR is expressed in endothelial cells, and its ligation with 15-deoxy-prostaglandin J2 or ciglitazone has been shown to stimulate NO production (17). Several members of the thiazolidinedione family have been shown to activate eNOS (27), inhibit endothelial-leukocyte interaction (54), and reduce vascular wall inflammation (78). Recently, PPAR agonists (fibrates) have been shown to increase the half-life of eNOS mRNA and the protein abundance (42), although the precise mechanism(s) of this action remains to be established. The previously documented anti-inflammatory action of fibrates and their ability to suppress induction of inducible NOS (28) could account for some additional benefits of PPAR agonists via increased bioavailability of NO and reduced formation of peroxynitrite, a product of reaction between NO and superoxide.6 s0 X+ T( H& ^* F
" `' z! V# O" {
Peroxynitrite Scavenging and Prevention of Endothelial Cell Senescence
- x, K+ X/ N  s# m, J; K& w! M3 v, c/ o2 A8 N% `$ G
Recent studies from our laboratory showed that human umbilical vein endothelial cells (HUVEC) cultured on glycated collagen I or Matrigel lattices develop premature senescence (24). Development of premature senescence of HUVEC on glycated collagen could be prevented and reversed by treatment with the peroxynitrite scavenger ebselen. Concomitant with the reversal of senescence, a selenorganic compound, ebselen, restored NO production to levels observed in HUVEC grown on unmodified collagen. Our findings indicate that exposure to glycated collagen in vitro elicits premature senescence of the vascular endothelium, and ebselen can prevent or reverse it. In vivo testing of these findings is in progress. Chronic treatment of Zucker diabetic fat rats with ebselen prevented and partially reversed ECD (12).
- g% b8 ^+ d# |0 [: I# M( c; V/ X
; K# j$ Y2 |2 x5 |: `Supplementation with NO Donors
8 z5 S' B+ k3 [7 H
8 p3 F* @$ c( q; FThis is by far the oldest therapeutic approach, based on the early observations of the efficacy of nitroglycerin. While this strategy remains one of the cornerstones of treatment in patients with CAD, development of tolerance hinders its efficacy. Novel S-nitrosothiols have been synthesized, which in vitro are devoid of tolerance induction (68). Several new approaches to deliver NO have emerged. Synthesis of NO-aspirin is reported to combine benefits of both parental ingredients, resulting in the improved inhibition of proinflammatory and prothrombotic pathways (33). Recent data demonstrated prevention of pulmonary thromboembolisms by NO-releasing aspirin (71). The same nitroderivative of aspirin, NCX 4016, has been reported to reduce postischemic infarct size and ventricular dysfunction in rabbits and rats (84). A NO-releasing naproxen derivative has been shown to possess antihypertensive properties (74). Delivery of NO by inhalation has been investigated and found to exert vasodilatory actions not only in the pulmonary circulation but also systemically (18). cGMP-dependent effects of NO may be enhanced by slowing the degradation of this second messenger. Therefore, a rational strategy to increase the half-life of cGMP in vascular smooth muscle cells and platelets is related to the inhibition of phosphodiesterase type V. In this vein, dipyridamole has been shown to potentiate vasodilatory and antiaggregation effects of NO (14). Caution is required in administering NO donors: the presence of oxidative stress may diminish their efficacy or even enhance formation of peroxynitrite. Moreover, systemic administration of NO donors will result in generalized effects (as opposed to its endogenous generation), which may be undesirable.
4 `5 L& j; h& ]$ x% O3 B- s/ b3 [3 b! N
CONCLUSIONS AND FUTURE DIRECTIONS
. h: Z9 T, i1 j9 w3 x! s3 x) T5 m% s2 X
; r0 x& l4 i6 i" m; L. ^. qThe data and arguments presented above allow me to formulate a provisional functional definition of the syndrome: ECD is a syndrome induced by diverse intrinsic and extrinsic factors that lead to disturbances in the barrier function of the vascular endothelium; its impaired antithrombogenic properties; perturbed angiogenic competence; inappropriate regulation of vascular smooth muscle tonicity, proliferative capacity, and migratory properties; and perturbed synthetic functions and deterrence of neutrophils and monocytes from diapedesis. ECD may be reversible within a certain time frame.
/ D6 C2 G$ ]3 M  Z( u2 U/ w: c2 e. M$ a+ r
The phenotype of endothelial cells exposed to several highly prevalent risk factors in ESRD, such as ADMA, HHCy, and AGE, is definitively proatherogenic. These are cells characterized by 1) decreased production of bioavailable NO; 2) increased adhesiveness for monocytes and polymorphonuclear cells; 3) accumulation of cholesterol and oxidized LDL, both leading to endothelial lipidosis; 4) defective transmission of endothelium-derived hyperpolarizing factor to the smooth muscle cells; 5) enhanced expression of profibrotic genes; and 6) tendency toward premature senescence and apoptosis.' W- J, j5 g  d  z8 T
% _9 o; e) G* u
It is quite possible that individual manifestations of vascular dysfunction such as inadequate angiogenesis at ischemic foci, increased adhesion of leukocytes, loss of antithrombogenic properties, inappropriate proliferation of vascular smooth muscle cells, and changed patterns of matrix deposition, each just slightly perturbed, collectively are capable of triggering early preclinical forms of generalized ECD. This would imply that therapeutic intervention cannot be based on a single agent; rather their combination would have improved chances of preventing and reversing ECD. The data linking eNOS function and NO generation or availability to vascular remodeling and signaling, hyperlipidemia, AGE, and hyperglycemia, as presented above, should be viewed within the broader framework of ECD. Accordingly, it is important to learn more about noninvasive techniques to assess the functional state of eNOS, the bioavailability of NO, and expression of other surrogate markers of ECD in the clinical settings. Our pharmacopeia needs to be enriched with agents to 1) correct elevated ADMA levels, 2) elevate homocysteine levels, 3) effectively suppress oxidative stress in the vascular wall, 4) correct increased vascular permeability, 5) improve impaired angiogenesis, and 6) curtail adhesion and diapedesis of monocytes. Future research in this area is needed to define early preclinical markers of ECD. Given accumulating evidence that disturbances of NO production or availability are major determinants of ECD, an intensification of therapeutic efforts toward correction of eNOS activity and NO bioavailability in blood vessels is warranted.
( l# L" P% T2 M+ N/ i" X% Z
2 c8 D( r, N# i' s1 _  ZGRANTS
3 P: J  p7 t5 A$ x! P( }4 [* k
, r4 h( a' R+ F- l6 n7 n# R: x. B5 @Relevant studies in the author's laboratory were supported in part by National Institute of Diabetes and Digestive and Kidney Diseases Grants DK-54602 and DK-45462 and the Westchester Artificial Kidney Foundation.: S5 Y1 B/ @* ~
( M$ p  N; p4 L
ACKNOWLEDGMENTS5 g! K' I8 M* F& ^

/ S/ E& L5 W+ M6 B6 `* M" x6 CI am grateful to all former and current postdoctoral fellows and colleagues who have contributed to my present view on the subject.! c! w) B5 ?+ A7 @( a0 {  \

/ G7 @9 @2 J& d, B( fFOOTNOTES
$ W) N% J4 L" V: V  M) {# S- e% d& Z9 z
REFERENCES" P) n9 ^! O# Z, Q3 D4 p. @
$ }. w" [/ B  o
Adams M, Kinlay S, Blake G, Orford J, Ganz P, and Selwyn A. Atherogenic lipids and endothelial dysfunction: mechanisms in the genesis of ischemic syndromes. Annu Rev Med 51: 149–167, 2000./ P5 g& |4 J& G
1 K4 z( p/ [4 w: `" o& N; \( L
Alexander MY, Brosnan J, Hamilton C, Fennell J, Beattie E, Jardine E, Heistad DD, and Dominiczak AF. Gene transfer of eNOS but not Cu/Zn SOD restores NO availability in the SHRSP. Cardiovasc Res 47: 609–617, 2000.
) y7 u/ G: d, P2 M" i3 y
! X& l# g7 j0 E( bAlexander MY, Brosnan M, and Hamilton C. Gene transfer of eNOS improves NO-dependent endothelial function in a hypertensive rat model. Cardiovasc Res 43: 798–807, 1999.
5 v/ Y* j9 B2 O/ f
# J6 K* `' D& P1 mAnderson T, Uehata A, and Garhard M. Close relation of endothelial function in the human coronary and peripheral circulations. J Am Coll Cardiol 26: 1235–1241, 1995.- Z) O/ ^( \4 V, \: S
$ W2 l+ Z. q+ j8 A0 z' e! N
Annuk M, Lind L, Linde T, and Fellstrom B. Impaired endothelium-dependent vasodilation in renal failure in humans. Nephrol Dial Transplant 16: 302–306, 2001.
* }8 A- h. o- L  U, h
0 O% e% e+ w5 z6 DBellamy M, McDowell I, Ramsey M, Brownlee M, Newcombe R, and Lewis MJ. Oral folate enhances endothelial function in hyperhomocysteinemic subjects. Eur J Clin Invest 29: 659–662, 1999.
  M9 g8 k" Y. J+ y) @0 K
9 I# \! F/ M/ x* D' F* b" U( k+ X! ABennett-Richards K, Kattenhorn M, Donald A, Oakley G, Varghese Z, Bruckdorfer R, Deanfield J, and Rees L. Oral L-arginine does not improve endothelial dysfunction in children with chronic renal failure. Kidney Int 62: 1372–1378, 2002.
4 t7 a6 H# q6 M* z9 g$ t  u+ x9 U8 p9 T. g2 z# J6 a
Bernstein R, Ochoa F, and Xu X. Function and production of nitric oxide in the coronary circulation of the conscious dog during exercise. Circ Res 79: 840–848, 1996.( f  U' f4 h+ n4 O  w

2 F, G) {8 e7 F5 [: o' k, OBoger RH and Bode-Boger SM. The clinical pharmacology of L-arginine. Annu Rev Pharmacol Toxicol 41: 79–99, 2001.0 p% M- Z( z- M
! i7 Z, R0 Z2 Q/ H! e3 d
Bonetti P, Lerman L, and Lerman A. Endothelial dysfunction. A marker of atherosclerotic risk. Arterioscler Thromb Vasc Biol 23: 168–175, 2003.
3 N" Z2 N' e9 P0 W6 |" i' J1 e' \4 A- a
Bourassa PA, Milos P, Gaynor B, Breslow J, and Aiello RJ. Estrogen reduces atherosclerotic lesion development in apolipoprotein E-deficient mice. Proc Natl Acad Sci USA 93: 10022–10027, 1996.
9 }3 v$ {2 u" g& s7 J7 F7 ?
% u7 y1 T" t& ~Brodsky S, Gaelikman O, Chen J, Zhang F, Togashi N, Crabtree M, Gross S, Nasjletti A, and Goligorsky MS. Prevention and reversal of premature endothelial cell senescence and vasculopathy in obesity-induced diabetes by ebselen. Circ Res 94: 377–384, 2004.
6 Z$ H3 [$ K* `  Y9 }" V/ R/ X7 Y/ h& Q5 ~0 p6 S
Buga GM, Wei LH, Bauer PM, Fukuto JM, and Ignarro LJ. NG-hydroxy-L-arginine and nitric oxide inhibit Caco-2 tumor cell proliferation by distinct mechanisms. Am J Physiol Regul Integr Comp Physiol 275: R1256–R1264, 1998.
, i# _3 y) U3 |: d; h; `
6 Q5 i4 H4 g$ PBult H. Nitric oxide and atherosclerosis: possible implications for therapy. Mol Med Today: 510–518, 1996.3 w- @# r& Q. t0 ^8 f1 d$ i
; z$ P% X+ X3 v
Bult H, Fret H, Jordaens F, and Herman A. Dipyridamole potentiates the anti-aggregating and vasodilator activity of nitrioc oxide. Eur J Pharmacol 199: 1–8, 1991.
5 U9 R* s- ^4 a( [" b' Q& z3 X
& W3 k8 I( L9 e( kCable DG, O'Brien T, Shaff HV, and Pompili VJ. Recombinant eNOS-transduced human saphenous veins: gene therapy to augment nitric oxide production in bypass conduits. Circulation 96: II173–II178, 1997.
  S  W4 S# {0 g# k0 V: X; @8 H/ K; r& V2 B/ g
Calnek D, Mazella L, Roser S, Roman J, and Hart M. Peroxisome proliferator-activated receptor  ligands increase release of NO from endothelial cells. Arterioscl Thromb Vasc Biol 23: 52–57, 2003.  h2 ?. B0 O4 |' Y  p. G: A* \
1 F7 j% t, Q& T1 a- s
Cannon R, Schechter A, Panza J, Ognibene F, Pease-Fye M, Waclawiw M, Shelhamer J, and Gladwin M. Effects of inhaled NO on regional blood flow are consistent with intravascular NO dilevery. J Clin Invest 108: 279–287, 2001.' R3 f$ L3 T/ p$ Z6 c$ X, y

) E4 k+ h+ W$ j9 _Casas J, Bautista L, Humphries S, and Hingorani A. Endothelial nitric oxide synthase genotype and ischemic heart disease. Circulation 109: 1359–1365, 2004.
0 f& ^2 _7 F5 ~* ]& b9 C0 H  D* Y
; C& y. |3 ^7 m9 a$ W  RCatravas J, Lazo J, Dobuler K, Mills L, and Gillis C. Pulmonary endothelial dysfunction in the presence or absence of interstitial injury induced by intratracheally injected bleomycin in rabbits. Am Rev Respir Dis 128: 740–746, 1983.
$ v+ Q( M% {# M, W" F0 D5 p6 H" r, U. X
Channon KM, Hu S, and Valentina N. In vivo gene transfer of NOS enhances vasomotor function of carotid arteries from normal and cholesterol-fed rabbits. Circulation 98: 1905–1911, 1998.
9 l8 d1 \# b3 X. {/ n! x, j. S3 j  T& i3 N: ~
Chen J, Kuhlencordt P, Astern J, and Huang P. Effects of chronic treatment with L-arginine on atherosclerosis in apoE-knockout mice. Arterioscler Tromb Vasc Biol 22: 97–103, 2002.
, b4 V; u& S5 S2 _8 e! B4 U7 u; r" y% h" [+ a- ~$ f
Chen J, Brodsky S, Hampel D, Li H, Weinstein T, Gafter U, Norman J, Fine L, and Goligorsky MS. Delayed branching of endothelial capillary cords in glycated collagen I is mediated by the early induction of PAI-1. Am J Physiol Renal Physiol 281: F71–F80, 2001.
" t/ f- L7 b; f& L2 c" H, {, M/ |: h4 K$ ~6 ^
Chen J, Brodsky S, Goligorsky DM, Hampel D, Li H, Gross SS, and Goligorsky MS. Glycated collagen I induces premature senescence-like phenotypic changes in endothelial cells. Circ Res 90: 1290–1298, 2002.
; S5 W9 ?$ ?& n' P% |& S$ Q% M2 J, `& w! |- n
Chen L, Mehta P, and Mehta JL. Oxidized LDL decreases L-arginine uptake and eNOS protein expression in human platelets: relevance of the effect of oxidized LDL on platelet function. Circulation 93: 1740–1746, 1996.! o1 P0 u" W" o# B# p+ ]

/ k2 e! B: q' r5 R! xChen Z, Yuhanna I, Galcheva-Gargova Z, Karas R, Mendelsohn M, and Shaul PW. Estrogen receptor alpha mediates the nongenomic activation of endothelial nitric oxide synthase by estrogen. J Clin Invest 103: 401–406, 1999.9 @$ r) B% y% @, k# a4 Z- o

0 I& Q0 f" g. s) O5 F6 p( GCho D, Choi Y, Jo S, and Yo I. Nitric oxide production and regulation of endothelial nitric oxide synthase phosphorylation by prolonged treatment with troglitazone. J Biol Chem 279: 2499–2506, 2004.& F# c' b6 r8 {5 a0 ~9 S7 w

0 C" l4 G  P: C* m; n1 A8 u  _. |Colville-Nash P, Qureshi S, Willis D, and Willoughy D. Inhibition of inducible nitric oxide synthase by peroxisome proliferator-activated receptor agonists: correlation with induction of heme oxygenase 1. J Immunol 161: 978–984, 1998.
8 y6 G- g' w; n2 M" H/ M: n: N( @, ?% n9 v; k# X" J0 E$ l4 p4 F
Creager MA, Gallagher S, Girerd X, Coleman S, Dzau VJ, and Cooke JP. L-Arginine improves endothelium-dependent vasodilation in hypercholesterolemic human. J Clin Invest 90: 1248–1253, 1992.
# g" g" s5 N6 f. O; I' L, m% I% x/ h; ?! w
Cross J, Donald A, Kharbanda R, Deanfield J, Woolfson R, and MacAllister R. Acute administration of L-arginine does not improve arterial endothelial function in chronic renal failure. Kidney Int 60: 2318–2323, 2001.+ T4 i* s8 L* F3 }- j" S. T7 B
) H( X' R5 W6 W8 z7 \0 F9 m3 B
Davis M, Cai H, McCann L, Fukai T, and Harrison D. Role of c-Src in regulation of endothelial nitric oxide synthase expression during exercise training. Am J Physiol Heart Circ Physiol 284: H1449–H1453, 2003.& M, x2 d7 z& |9 ]' _

  y, O9 E1 g! h0 j9 sDe Nicola L, Bellizzi V, Minutolo R, Andreucci M, Capuano A, Garibotto G, Corso G, Andreucci V, and Cianciaruso B. Randomized, double-blind, placebo-controlled study of arginine supplementation in chronic renal failure. Kidney Int 56: 674–684, 1999.) g) y3 V) w0 v

" x5 `0 L+ ^" I& a' N. `Del Soldato P, Sorrentino R, and Pinto A. NO-aspirins: a class of new antiinflammatory and antithrombotic agents. Trends Pharmacol Sci 20: 319–323, 1999.
$ ^' x& ]: w- z0 C2 [1 ?$ P3 Z: S! X" M. O( l, Z0 D' T. S8 {
Nascimento CA, Kauser K, and Rubanyi GM. Effect of 17-estradiol in hypercholesterolemic rabbits with severe endothelial dysfunction. Am J Physiol Heart Circ Physiol 276: H1788–H1794, 1999.
% Z- q& ]# G$ T" e* \0 H& U
% T- z0 z& H: M- }8 W$ VDrexler H, Zeiher A, Meinzer K, and Just H. Correction of endothelial dysfunction in coronary microcirculation of hypercholesterolaemic patients by L-arginine. Lancet 338: 1546–1550, 1991.
+ [; @$ f+ d1 A: s7 I- W& }. i) ?- j9 v, H( s: ]6 @' V4 T: q
Drexler H and Hornig B. Endothelial dysfunction in human disease. J Mol Cell Cardiol 31: 51–60, 1999.: \4 d) |: {; {
! z- h, Y% C5 S
Drexler H, Zeiher A, Meinzer K, and Just H. Correction of endothelial dysfunction in coronary microcirculation of hypercholesterolaemic patients by L-arginine. Lancet 338: 1546–1550, 1991.
$ _3 w+ J& |! D: u& x) \, a3 _9 _! Z6 C
Fish R, Nabel E, Selwyn A, Ludmer P, Mudge G, Kirshenbaum J, Schoen F, Alexander R, and Ganz P. Responses of coronary arteries of cardiac transplant patients to acetylcholine. J Clin Invest 81: 21–31, 1988.+ V5 U' O& s3 f. ^- A3 I! _: t" X( M
6 s# l! ]' E6 K1 v: Y* S" v
Girerd XJ, Hirsch A, Cooke JP, Dzau VJ, and Creager MA. L-Arginine augments endothelium-dependent vasodilation in cholesterol-fed rabbits. Circ Res 67: 1301–1308, 1990.& `# M6 Q; `2 w, ~5 s

/ H( ~. t5 B8 n7 e8 S; J- tGiugliano D, Marfella R, Verazzo G, Acampora R, Coppola L, Cozzolino D, and D'Onofrio F. The vascular effects of L-arginine in humans. The role of endogenous insulin. J Clin Invest 99: 433–438, 1997.5 q8 R; K1 t" b$ ^5 l

# e/ U# o# C$ q9 P2 u7 cGokce N, Keaney J, and Vita J. Endotheliopathies: clinical manifestations of endothelial dysfunction. In: Thrombosis and Hemorrhage, edited by Loscalzo J and Shafer A. Baltimore, MD: Williams and Wilkins, 1998, p. 901–924.
. N& L$ P, u- G0 X! u. M3 v1 x
* k' _, N# Q1 K$ U, \5 QGoya K, Sumitani S, Xu X, Kitamura T, Yamamoto H, Kurebayashi S, Saito H, Kouhara H, Kasayama S, and Kawase I. Peroxisome proliferator-activated receptor alpha agonists increase nitric oxide synthase expression in vascular endothelial cells. Arterioscler Thromb Vasc Biol 24: 658–663, 2004.
8 E' G1 w/ q/ W6 b  v' j0 I" i) `/ T0 I' h* k
Graham IM and O'Callaghan P. The role of folic acid in the prevention of cardiovascular disease. Curr Opin Lipidol 11: 577–587, 2000.
3 v6 o( D  A# ~$ L# e  [5 t5 P" |6 u6 u# R- K2 {0 \7 Y
Gruhn N, Aldershvile J, and Boesgaard S. Tetrahydrobiopterin improves endothelium-dependent vasodilation in nitroglycerin-tolerant rats. Eur J Pharmacol 416: 245–249, 2001.8 s' B' i5 t$ W" Z8 f
! ?( {8 \6 ^- \; Q, |. e+ U
Halliwell B. Lipid peroxidation, antioxidants and cardiovascular disease: how should we move forward Cardiovasc Res 47: 410–418, 2000.( j0 S: P3 t6 w0 }' Y
4 x; a9 A0 Y* `9 R' c
Harrison D. Cellular and molecular mechanisms of endothelial cell dysfunction. J Clin Invest 100: 2153–2157, 1997.' q$ Y4 m' S% Q0 X( d- f2 T6 [+ k- B

+ ~0 ]8 j* T8 Q7 p1 EHattori Y, Nakanishi N, Akimoto K, Yoshida M, and Kasai K. HMG-CoA reductase inhibitor increases GTP cyclohydrolase I mRNA and tetrahydrobiopterin in vascular endothelail cells. Arterioscler Thromb Vasc Biol 23: 176–182, 2003.
( X. Q! R' {* S( r' e: z6 W
* N4 d% z7 K2 ~9 }8 \Hedblad B, Ogren M, Janzon L, Isacsson S, and Lindell S. Low pulse-wave amplitude during reactive leg hyperemia: an independent early marker for ischemic heart disease and death. Results from the 21-year follow-up of the prospective cohort study "Men born in 1914," Malmo, Sweden. J Intern Med 236: 161–168, 1994." w. A, h4 y1 _. H% E2 z
6 ]5 g2 l4 O( y0 o- \; q3 E! p# G* q
Heller R, Munscher-Paulig F, Grabner R, and Till U. L-Ascorbic acid potentiates nitric oxide synthesis in endothelial cells. J Biol Chem 274: 8254–8260, 1999.: J2 f9 G. X$ `' ?6 }, B3 e1 P
! N  r- ^( k) E" i2 f! Z- w+ l
Hernandez-Perera O, Perez-Sala D, Navarro-Antolin J, Sanchez-Pascuala R, Hernandez G, Diaz C, and Lamas S. Effects of 3-hydroxy-3-methyglutaryl-CoA reductase inhibitors, atorvastatin and simvastatin, on the expression of endothelin-1 and endothelial nitric oxide synthase in vascular endothelial cells. J Clin Invest 101: 2711–2719, 1998.
- w/ P8 C7 r& R) J! l' t# }% y0 M, f- @. x4 D, C1 g8 g
Hisamoto K, Ohmichi M, Kanda Y, Adachi K, Nishio Y, Hayakawa J, Mabuchi S, Takahashi K, Tasaka K, Miyamoto Y, Taniguchi N, and Murata Y. Induction of eNOS phosphorylation by the raloxifene analog LY117018 is differentially mediated by Akt and extracellular signal-regulated protein kinase in vascular endothelial cells. J Biol Chem 276: 47642–47649, 2001.
  P) G+ e8 G9 Q4 X: L9 g5 u  B! V! g5 _4 m( D
Hofmann H and Schmidt HW. Thiol dependence of nitric oxide synthase. Biochemistry 34: 13443–13452, 1995.
7 v: e; d0 s( P# p- P7 D1 R9 i% X4 q" w
) O- D: x& j3 CHuang A, Venema R, Vita J, and Keaney J. Ascorbic acid enhances endothelial nitric oxide production in a tetrahydrobiopterin-dependent manner (Abstract). Circulcatin 100: 1702, 1999./ s3 i* R) E- c) ~6 |- B8 Y$ c

6 I: s5 @: K* c" vJackson S, Parhami F, Xi X, Berliner J, Hsueh W, Law R, and Demer L. Peroxisome proliferator-activated receptor activators target human endothelial cells to inhibit leukocyte-endothelial cell interaction. Arterioscler Thromb Vasc Biol 19: 2094–2104, 1999., R* l! Z& p5 P+ X# p8 D2 w
4 T  k" C# Y- E9 e& I
Keaney JF, Xu A, Cunningham D, Jackson T, Frei B, and Vita JA. Dietary probucol preserves endothelial function in cholesterol-fed rabbits by limiting vascular oxidative stress and superoxide generation. J Clin Invest 95: 2520–2529, 1995.# }7 M1 i3 W% [& Z; i" O
9 Q1 m0 [: ?3 [+ g+ Y
Khurana R, Martin J, and Zachary I. Gene therapy for cardiovascular disease. Hypertension 38: 1210–1216, 2001.) @9 _" Y4 f% L' m

6 j) f( e% a: E1 x# XKureichi Y, Luo Z, Shiojima I, Bialik A, Fulton D, Lefer D, Sessa WC, and Walsh K. The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic animals. Nature Med 6: 1004–1010, 2000.
# v6 z7 H0 C+ t0 a. J1 K& `/ y" v5 \5 H6 q" R' A
Kwang K and Sakuma I. Should progestins be blamed for the failure of hormone replacement therapy to reduce cardiovascular events in randomized controlled trials Arterioscler Thromb Vasc Biol 24: 1171–1179, 2004." _+ c4 @2 R8 W6 V2 H( E& H6 h

9 A. c- M' d1 {. g5 [8 wLi H and Goligorsky MS. Endothelial gene responses to homocysteine: relation to atherosclerosis. Exp Nephrol 10: 164–169, 2002.
6 c5 [3 J* k. Y* x) F
& T* k9 S. J* }- C0 q, ?& p& bLi H, Lewis A, Brodsky S, Rieger R, Iden C, and Goligorsky MS. Homocysteine induces HMGCoA reductase in vascular endothelial cells: a mechanism for development of atherosclerosis. Circulation 105: 1037–1043, 2002.
5 G9 j( B: Z1 `' Y8 J4 ]& Z1 |5 W6 W  Z: G5 [' {* ?
Li H, Brodsky S, Brink P, Nasjletti A, and Goligorsky MS. Paradoxical overexpression and translocation of connexin-43 in homocysteine-treated endothelial cells. Am J Physiol Heart Circ Physiol 282: H2124–H2133, 2002.
, J9 j, t+ I' |! e( x& l( h" W; W3 x! a7 k+ U
Liao J, Shin W, Lee W, and Clark SL. Oxidized LDL decreases the expression of eNOS. J Biol Chem 270: 319–324, 1995.
' Z5 t" ~" y# I) b6 ^4 D
6 u7 O9 |7 Q0 b! R" Q7 ~( a% O3 N; ELin KF, Chao L, and Chao J. Prolonged reduction of high blood pressure with human NOS gene delivery. Hypertension 30: 307–313, 1997.
% T! h. }1 t# g5 m( i3 c$ t* d. |) D; x. g* `" U9 ?
Linz W, Wohlfart P, Scholkens B, Malinski T, and Wiemer G. Interactions among ACE, kinins and NO. Cardiovasc Res 43: 549–561, 1999.' g, _' v! j% D( s7 y" a/ @
/ b6 D5 |/ o2 V5 K9 O5 k
Loscalzo J. Adverse effects of supplemental L-arginine in atherosclerosis. Consequences of methylation stress in a complex catabolism Arterioscler Thromb Vasc Biol 23: 3–5, 2003.8 A+ {- U& o6 Q" ^# K% P
. [9 L) M- d3 `9 }: Q% {$ G
Ludmer P, Selwyn A, Shook T, Wayne R, Mudge G, Alexander R, and Ganz P. Paradoxical vasoconstriction induced by acetylcholine in atherosclerotic coronary arteries. New Engl J Med 315: 1046–1051, 1986.
6 J* }! [8 I, B9 ]4 G" N/ P3 l$ k: G. p; ]* G
McNamara DB, Bedi B, Aurora H, Tena L, Ignarro LJ, Kadowitz PJ, and Akers DL. L-Arginine inhibits balloon catheter-induced intimal hyperplasia. Biochem Biophys Res Commun 193: 291–296, 1993.
% r5 R/ R# X) Y* ?7 w$ P; F
8 J; Q  L5 v! K) `Miller M, Roseberry M, Mazzei F, Butler A, Webb D, and Megson I. Novel S-nitrosothiols do not engender vascular tolerance and remain effective in glyceryltrinitrate-tolerant rat femoral arteries. Eur J Pharmacol 408: 335–343, 2000.
4 r, e: o) N3 W9 W
# Q* e+ I/ S0 Q2 {Moat S, Doshi S, Lang D, McDowell I, Lewis M, and Goodfellow J. Treatment of coronary heart disease with folic acid: is there a future Am J Physiol Heart Circ Physiol 287: H1–H7, 2004.
* ?- W# I4 c  }! L' }$ B$ p; I( K) b& ]1 n- u' |" |
Molteni A, Ward W, Tsao C, Port C, and Solliday N. Monocrotaline-induced pulmonary endothelial dysfunction in rats. Proc Soc Exp Biol Med 176: 88–94, 1984.. C9 I: Z0 M9 C8 w
( ]8 }9 R7 `8 ~  P6 d) C
Momi S, Emerson M, Paul W, Leone M, Mezzasoma A, DelSoldato P, Page C, and Gresele P. Prevention of pulmonary thromboembolism by NCX 4016, a nitric oxide-releasing aspirin. Eur J Pharmacol 397: 177–185, 2000.0 P3 Z2 m5 J# D+ b$ M( `6 m3 D

0 P  A5 y3 y; @( x7 e/ y3 ZMorris ST, McMurray JJ, Rodger RS, and Jardine AG. Impaired endothelium-dependent vasodilatation in uraemia. Nephrol Dial Transplant 15: 1194–1200, 2000.
# i' F7 @2 t6 l8 \4 L, S3 A9 p& L5 d
. _; U. @9 A% m  L3 T! P+ S! kMozes G, Kullo I, and Mohacsi T. Ex vivo gene transfer of eNOS to atherosclerotic rabbit aortic rings improves relaxation to acetylcholine. Atherosclerosis 141: 265–271, 1998.
3 X1 W4 n7 [' R3 @: N- A) g4 [5 H* b" m( a! _+ N4 g. j
Muscara M, McKnight W, Lovren F, Triggle C, Cirino G, and Wallace J. Antihypertensive properties of a nitric oxide-releasing naproxen derivative in two-kidney, one-clip rats. Am J Physiol Heart Circ Physiol 279: H528–H535, 2000.: j5 h, W( D& A8 }" D: c7 `. F
; O  M- Z0 S0 @, `. e3 W& S
Noiri E, Satoh H, Taguchi J, Brodsky S, Nakao A, Ogawa Y, Nishijima S, Yokomizo T, Tokunaga K, and Fujita T. Association of eNOS Glu298Asp polymorphism with end-stage renal disease. Hypertension 40: 535–540, 2002.: J$ a, d0 J1 i

3 {1 l1 `/ h: Y$ D! HOzaki M, Kawashima S, Yamashita T, Hirase T, Namiki M, Inoue N, Hirata K, Yasui H, Sakurai H, Yoshida Y, Masada M, and Yokoyama M. Overexpression of eNOS accelerates atherosclerotic lesion formation in apoE-deficient mice. J Clin Invest 110: 331–340, 2002.  J5 N; ?: ~7 B7 a* x# u

2 F. D' E) k& {( VPasceri V, Wu H, Willerson J, and Yeh E. Modulation of vascular inflammation in vitro and in vivo by peroxisome proliferator-activated receptor-gamma activators. Circulation 101: 235–238, 2000./ E% ?  k0 u0 x. ~2 q5 O. K5 V
9 K4 S) t- H( V6 p( p
Petricone F, Ceravolo R, and Pujia A. Prognostic significance of endothelial dysfunction in hypertensive patients. Circulation 104: 2673–2678, 2001.
2 {4 w8 G6 [# ?% d/ j( c, ^& w
7 c( I1 d% m( ]" yQuyiumi AA, Dakak N, Andrews N, Husain S, Arora S, Gilligan D, and Panza JA. Nitric oxide activity in the human coronary circulation. Impact of risk factors for coronary athrosclerosis. J Clin Invest 95: 1747–1753, 1995.
" `( R4 B# U% \5 x1 m
4 E( c1 M8 j5 P7 yReddy KG, Nair R, Sheehan H, and Hodgson J. Evidence that selective endothelial dysfunction may occur in the absence of angiographic or ultrasound atherosclerosis in patients with risk factors for athrosclerosis. J Am Coll Cardiol 23: 833–843, 1994.
: M/ X% F+ X: L8 r6 F6 g  Z6 g4 A2 _/ n% B8 F  h: Z1 {4 Y
Ross R. The pathogenesis of atheroscleroisis: a perspective for 1990s. Nature 326: 801–809, 1993.! X, D% }) t: w$ o8 P" [
3 s/ k, Z1 R3 U+ d; n5 }  u
Rossig L, Dimmeler S, and Zeiher AM. Apoptosis in the vascular wall and atherosclerosis. Basic Res Cardiol 96: 11–22, 2001.
+ ?6 x' ?4 \5 M# F# T8 Q' P) J+ i/ V' ~; r; Q( b' {
Rossoni G, Manfredi B, DeGennaro CV, Bernareggi M, and Berti F. The nitroderivative of aspirin, NCX 4016, reduces infarct size caused by myocardial ischemia-reperfusion in the anesthetized rat. J Pharm Exp Ther 297: 380–387, 2001.
, B8 E1 L$ h4 E. G4 E4 \& L2 [! w% F1 D, O4 z; W2 _1 e
Selhub J, Jacques P, and Rosenberg I. Serum total homocysteine concentrations in the Third National Health and Nutrition Examination Survey (1991–1994): population reference ranges and contribution of vitamin status to high serum concentrations. Ann Intern Med 131: 331–339, 1999.
" O; @8 w4 @, a+ V' C  m$ o
9 t& M" ^/ w( K) ?Shamim-Uzzaman QA, Pfenninger D, Kehrer C, Chakrabarti A, Kacirotti N, Ruberfire M, Brook R, and Rajagopalan S. Altered cutaneous microvascular responses to reactive hyperemia in coronary artery disease: a comparative study with conduit vessel responses. Clin Sci (Colch) 103: 267–273, 2002.0 X& }  n3 N% ?1 d5 o

' b$ b+ V8 n, sShinozaki K, Kashiwagi A, Nishio Y, Okamura T, Yoshida Y, Masada M, Toda N, and Kikkawa R. Abnormal biopterin metabolism is a major cause of impaired endothelium-dependent relaxation through NO/O2- imbalance in insulin-resistant rat aorta. Diabetes 48: 2437–2445, 1999.
3 v& y! r# Q+ S3 U/ \- o
  Y1 V% l' N, p: W0 }* qSmirnova I, Kajstura M, Sawamura T, and Goligorsky MS. Asymmetric dimethylarginine upregulates LOX-1 in activated macrophages: the role in foam cell formation. Am J Physiol Heart Circ Physiol 287: H782–H790, 2004.+ M# ?9 _2 E  m- w& r  [" A

+ V0 W& m+ n# v  e# ]" |1 x5 ^. D& nSmirnova I, Sawamura T, and Goligorsky MS. Upregulation of lectin-like oxidized low-density lipoprotein receptor-1 LOX-1 in endothelial cells by nitric oxide deficiency. Am J Physiol Renal Physiol 287: F25–F32, 2004.: j/ M2 h- u9 i7 W  F$ O

* z" v* G6 g) r6 C! n) F, WStehouwer CDA. Is measurement of endothelial dysfunction clinically useful Eur J Clin Invest 29: 459–461, 1999( A2 |) P/ d& T8 x
9 V5 t' N6 h" G) ^
Stewart J, Kohen A, Brouder D, Rahim F, Adler S, Garrick R, and Goligorsky MS. Noninvasive interrogation of microvasculature for signs of endothelial dysfunction in patients with chronic renal failure. Am J Physiol Heart Circ Physiol 287: H2687–H2696, 2004. First published August 5, 2004; doi doi:10.1152/ajpheart.00287.2004.
. A. I7 d" b$ }+ }* o6 T7 H% ]3 ]! Y/ z" d6 A4 d' j
Stroes E, Kastelein J, Cosentino F, Erkelens W, Wever R, Koomans H, Luscher T, and Rabelink T. Tetrahydrobiopterin restores endothelial function in hypercholesterolemia. J Clin Invest 99: 41–46, 1997.
3 D1 @' s4 L7 x  b7 U
9 j7 o! H6 O' O# h1 _Stuehr D, Kwon N, Nathan C, Griffith O, Feldman P, and Wiseman J. N-hydroxy-L-arginine is an intermediate in the biosynthesis of nitric oxide from L-arginine. J Biol Chem 266: 6259–6263, 1991.
* I/ ~7 C7 w& h# f2 M4 v2 }) y8 T
Tardif JC, Cote G, Lesperance J, Bourassa M, Lambert J, Doucet S, Bilodeau L, Nattel S, and de Guise P. Probucol and multivitamins in the prevention of restenosis after coronary angioplasty. Multivitamins and Probucol Study Group. N Engl J Med 337: 365–372, 1997.
1 b" ~: \4 `! _& U/ U& f
; D" g! |! }# `; S  qTarpey M. Sepiapterin treatment in atherosclerosis. Arterioscler Thromb Vasc Biol 22: 1519–1521, 2002.
" i) S% T3 y3 L9 w
$ D5 U  T2 v. p; e/ M1 k8 KTierney D, Huang H, Martasek P, Masters B S, Silverman R, and Hoffman BM. ENDOR spectroscopic evidence for the position and structure of NG-hydroxy-L-arginine bound to holo-neuronal nitric oxide synthase. Biochemistry 38: 3704–3710, 1999.
6 o) _$ Y  h, l& B' d* }$ G: f: `. X9 ^$ t# b* l. N
Tomasian D, Keaney J, and Vita J. Antioxidants and the bioactivity of endothelium-derived nitric oxide. Cardiovasc Res 47: 426–435, 2000.
2 k( u5 Y4 e2 h( K" X
3 v. `/ D" s. ^- x7 Y# A# KTomasian D and Steinberg D. Is there a potential therapeutic role for vitamin E or other antioxidants in atherosclerosis Curr Opin Lipidol 11: 603–607, 2000.
7 ?" E; b/ L1 A
% r4 x3 K8 Y' I$ i- p# zTsao PS, McEvoy L, Drexler H, Butcher E, and Cooke JP. Enhanced endothelial adhesiveness in hypercholesterolemia is attenuated by L-arginine. Circulation 89: 2176–2182, 1994.
/ ]) `0 ^  }3 w9 K! j8 N& w: y
( r9 E5 [. H1 rUemura S, Fathman C G, Rothbard J, and Cooke JP. Rapid and efficient vascular transport of arginine polymers inhibits myointimal hyperplasia. Circulation 102: 2629–2635, 2000.; N' D; p2 x, x% i8 P

/ j% G% O# x* Y& BVallance P and Chan N. Endothelial function and nitric oxide: clinical relevance. Heart 85: 342–350, 2001./ y0 e& L5 j2 Z/ m
/ H+ h% u: Y( x$ S8 C( p! Q+ T
Vaughan CJ, Murphy M B, and Buckley BM. Statins do more than just lower cholesterol. Lancet 348: 1079–1082, 1996.3 u  @4 D9 n+ a& O& j  \+ F
# O* Q/ x& C, c, @9 _6 z
Vaughan D, Rouleau J, Ridker P, Arnold J, Menapace F, and Pfeffer MA. Effects of ramipril on plasma fibrinolytic balance in patients with acute anterior infarction. Circulation 96: 442–447, 1997.
# ^* w( P' ]- N5 B" i' a6 \
0 o) M9 i  a% oVerhaar MC, Wever R, Kastelein J, van Dam T, Koomans H, and Rabelink TJ. 5-Methyltetrahydrofolate, the active form of folic acid, improves endothelial function in familial hypercholesterolemia. Circulation 97: 237–241, 1998.. _/ r. @$ R* _

) g, r! X: `. ?* ?Vita J and Keaney J. Endothelial function: a barometer for cardiovascular risk Circulation 106: 640–642, 2002.9 ^$ @, x$ q, A/ @* q- S4 T

" L5 c9 y' t4 F! J5 LVon der Leyen HE, Gibbons G, Morishita R, Lewis N, Zhang L, Nakajima M, Kaneda Y, Cooke JP, and Dzau VJ. Gene therapy inhibiting neointimal vascular lesion: in vivo transfer of endothelial nitric oxide synthase gene. Proc Natl Acad Sci USA 92: 1137–1141, 1995.; x" ~* P5 Q! B8 K5 Q; N4 N) p

5 @  h: Q* l, D, dWang B-Y, Ho H, Lin P, Schwarzacher S, Pollman M, Gibbons G, Tsao P, and Cooke JP. Regression of atherosclerosis Role of nitric oxide and apoptosis. Circulation 99: 1236–1241, 1999.
( d/ G& H9 X+ i: ?# g/ Y4 r4 B) A$ v7 ^  I$ }: ~8 Y% }: U
Weyrich AS, Ma X, and Lefer AM. The role of L-arginine in ameliorating reperfusion injury after myocardial ischemia in the cat. Circulation 86: 279–288, 1992.
$ d% a' |/ m4 P0 ^2 M
, m1 @5 i1 |6 p% h9 G: BYuhanna I, Zhu Y, Cox B, Hahner L, Osborne-Lawrence S, Lu P, Marcel Y, Anderson R, Mendelson M, Hobbs H, and Shaul PW. HDL binding to scavenger receptor B1 activates eNOS. Nat Med 7: 853–857, 2001.
7 y: b: ?) q4 K! O9 u. X
; b2 u# _; f2 c+ oZeiher AM, Drexler H, Wollschlager H, and Just H. Modulation of coronary vasomotor tone in humans Progressive endothelial dysfunction with different early stages of coronary atherosclerosis. Circulation 83: 391–401, 1991.) `" g6 g5 l1 z* ]  _+ O  E8 a
/ @- w$ _* m6 q, w  M5 V9 |& B) m
Zhang X, Jin H, Li H, Ebin Z, Brodsky S, and Goligorsky MS. Effects of homocysteine on nitric oxide production by endothelial cells. Am J Physiol Renal Physiol 279: F671–F678, 2000.(Michael S. Goligorsky)

Rank: 2

积分
107 
威望
107  
包包
1889  
发表于 2015-5-29 07:18 |显示全部帖子
宁愿选择放弃,不要放弃选择。  

Rank: 2

积分
79 
威望
79  
包包
1769  
发表于 2015-7-6 16:26 |显示全部帖子
我喜欢这个贴子  

Rank: 2

积分
68 
威望
68  
包包
1752  
发表于 2015-7-14 22:09 |显示全部帖子
干细胞之家微信公众号
感觉好像在哪里看过了,汗~  

Rank: 2

积分
77 
威望
77  
包包
1730  
发表于 2015-7-22 12:54 |显示全部帖子
希望大家帮我把这个帖发给你身边的人,谢谢!  

Rank: 2

积分
79 
威望
79  
包包
1769  
发表于 2015-8-10 08:19 |显示全部帖子
干细胞治疗糖尿病  

Rank: 2

积分
68 
威望
68  
包包
1752  
发表于 2015-8-11 14:17 |显示全部帖子
看完了这么强的文章,我想说点什么,但是又不知道说什么好,想来想去只想  

Rank: 2

积分
73 
威望
73  
包包
1833  
发表于 2015-8-15 17:11 |显示全部帖子
一楼的位置好啊..  

Rank: 2

积分
68 
威望
68  
包包
1752  
发表于 2015-8-19 19:54 |显示全部帖子
干细胞产业是朝阳产业

Rank: 2

积分
162 
威望
162  
包包
1724  
发表于 2015-9-5 17:46 |显示全部帖子
原来是这样  
你需要登录后才可以回帖 登录 | 注册
验证问答 换一个

Archiver|干细胞之家 ( 吉ICP备2021004615号-3 )

GMT+8, 2024-4-16 15:06

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.