Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2006年第26卷第10期

PAI-1 and the Metabolic Syndrome

来源:《动脉硬化血栓血管生物学杂志》
摘要:【摘要】Thelinkbetweenplasminogenactivatorinhibitor(PAI)-1andthemetabolicsyndromewithobesitywasestablishedmanyyearsago。IncreasedPAI-1levelcanbenowconsideredatruecomponentofthesyndrome。Themetabolicsyndromeisassociatedwithanincreasedriskofdeveloping......

点击显示 收起

【摘要】  The link between plasminogen activator inhibitor (PAI)-1 and the metabolic syndrome with obesity was established many years ago. Increased PAI-1 level can be now considered a true component of the syndrome. The metabolic syndrome is associated with an increased risk of developing cardiovascular disease, and PAI-1 overexpression may participate in this process. The mechanisms of PAI-1 overexpression during obesity are complex, and it is conceivable that several inducers are involved at the same time at several sites of synthesis. Interestingly, recent in vitro and in vivo studies showed that besides its role in atherothrombosis, PAI-1 is also implicated in adipose tissue development and in the control of insulin signaling in adipocytes. These findings suggest PAI-1 inhibitors serve in the control of atherothrombosis and insulin resistance.

The metabolic syndrome is associated with overexpression of PAI-1; the mechanisms involved are complex. Besides its role in atherothrombosis, PAI-1 is also implicated in obesity and insulin resistance. The development of PAI-1 inhibitors is a challenge.

【关键词】  adipose tissue atherothrombosis metabolic syndrome obesity PAI


Introduction


In blood, fibrinolysis breaks down fibrin and maintains vessel patency, and in tissues it breaks down the extracellular matrix and controls cell adhesion and migration and thus participates in tissue remodeling. Fibrinolysis is primarily regulated by plasminogen activator inhibitor type-1 (PAI-1), which prevents the escape of this potentially destructive protease system. 1-3


Increased PAI-1 levels may predispose patients to the formation of atherosclerotic plaques prone to rupture with a high lipid-to-vascular smooth muscle cells ratio as a result of decreased cell migration. 4 In humans, there is clinical evidence that increased PAI-1 levels are associated with atherothrombosis. 5,6 In large epidemiological studies, elevated plasma PAI-1 levels have been identified as a predictor of myocardial infarction. 7-11 Remarkably, the predictive ability of PAI-1 disappears after adjustment for markers of the metabolic syndrome (MetS) such as body mass index (BMI), triglycerides, and high-density lipoprotein cholesterol, 8,12-16 suggesting that the MetS is a prerequisite to high plasma PAI-1 levels in patients prone to atherothrombosis.


The purpose of this review is to update our understanding of the connections between PAI-1 and the MetS. We discuss the possible mechanisms linking increased circulating PAI-1 levels to the MetS and the recent findings implicating PAI-1 in adipose tissue development and insulin signaling, making PAI-1 more an actor rather than a simple marker of the MetS ( Figure ).


Interaction between metabolic syndrome and PAI-1 overexpression.


Contribution of MetS to PAI-1 Synthesis


Description of the MetS


The MetS consists of a cluster of metabolic abnormalities that cooccur in an individual more often than by chance. These abnormalities include obesity with a distribution of the fat in the central part of the body (visceral or android obesity), impaired glucose tolerance, hyperinsulinemia, dyslipidemia with elevated triglyceride level, low high-density lipoprotein cholesterol concentration, increased proportion of small dense lipoparticles, and hypertension, all well-documented risk factors for cardiovascular disease. 17 Individuals with the MetS are at increased risk for diabetes mellitus and cardiovascular disease. 18,19 Some investigators called into question the existence of this syndrome in the sense of its having a single underlying cause 20 and argued that the MetS does not add to global cardiovascular risk as assessed by current algorithms, which already include some of the MetS features. It is clear that the MetS has more than one cause. 21 The extent to which the MetS adds to the global risk of cardiovascular disease has to be defined, given its growing prevalence worldwide. Ectopic fat depots such as the visceral one may play a central part in linking the MetS to cardiovascular disease. Ectopic fat could be secondary to a defect of peripheral fat cell proliferation, as observed in severe insulin resistance associated with hereditary lipodystrophy, or to the failure of fat cells to increase their size and therefore to accommodate an increased energy influx, leading to a reorientation of fat storage. 22 The resulting ectopic fat depots may be a source of toxicity toward the surrounding tissues by releasing active substances such as free fatty acid 23 and deleterious adipokines such as inflammatory cytokines and PAI-1. 24


PAI-1 Is a True Component of the MetS


The link between PAI-1 and the MetS was first described by our group in 1986 and is now well-established. 25,26 Circulating PAI-1 is increased in obese subjects with the MetS, as well as in patients with type II diabetes. The more severe the MetS, the higher the plasma level of PAI-1. 27 The MetS explains a major part of plasma PAI-1 level variability, with this relationship being stronger in men than in women (45% versus 26%). 28 Interventional studies reported that if insulin resistance is improved, plasma PAI-1 levels decrease. Decreased plasma PAI-1 concentrations were observed after weight reduction by a hypocaloric diet and were associated with decreased body fat. 29 In addition, treatment with insulin-sensitizing drugs like metformin or troglitazone decrease plasma PAI-1 levels in subjects with type II diabetes and to some extent in normal obese subjects. 30,31 It has been proposed to consider increased PAI-1 levels as a true component of the MetS. 26,32


Possible Mechanisms Linking PAI-1 Overexpression to the MetS


Obviously, induction of PAI-1 overexpression is a complex process and several inducers may be involved at the same time at several sites of synthesis, including vessel walls. 33,34 Factorial analysis showed 35 that elevated circulating PAI-1 levels during obesity are not associated with the interleukin (IL)-6 driven inflammation (C-reactive protein , fibrinogen), as one would expect because PAI-1 is considered an acute phase protein whose synthesis is induced by IL6. 36,37 PAI-1 levels are also not associated with dyslipidemia but rather with the fat redistribution phenotype assessed by the measure of waist circumference and the insulin resistance state. 35,38 In this context, the role of ectopic fat depots as sites of PAI-1 synthesis may be relevant.


Adipose Tissue and Ectopic Fat Depots as Sites of PAI-1 Overexpression


Several groups have described the ability of adipocyte cell lines 39-41 and murine adipose tissue 42 to synthesize PAI-1. Subsequent reverse-transcription polymerase chain reaction (RT-PCR) and in situ hybridization studies suggested that the increased plasma PAI-1 originates primarily from the adipocyte in response to chronically elevated levels of tumor necrosis factor (TNF), insulin, and transforming growth factor (TGF)-beta. 43 PAI-1 is also produced by human adipose tissue explants, 44,45 but it is mainly localized in the stromal compartment of the adipose tissue. PAI-1 antigen was detected in purely stromal area and in small cells in direct contact with adipocytes of macrophage origin. 46,47 During human preadipocyte differentiation, PAI-1 secretion appears to be produced by contaminant macrophages. 46,48,49 These results suggest that macrophages infiltrating adipose tissues may be one of the main source of PAI-1 in patients with a MetS.


Several groups have stressed the exclusive association between high plasma PAI-1 levels and visceral obesity. 50-52 For example, changes in plasma PAI-1 levels during a weight-reducing program correlated with changes in visceral fat depot but not in subcutaneous fat depot. 53 In obese rats PAI-1 mRNA is found in both types of fat tissue but its level increased only in visceral fat during the development of obesity. 52 In obese patients, abdominal visceral fat expressed 5-fold more PAI-1 than subcutaneous tissue. 46


Ectopic fat accumulation in human liver was also associated with a strong expression of PAI-1 close to fat cells. 54 This could be related to the strong relationship between circulating PAI-1 antigen and hepatic but not peripheral insulin resistance in Pima Indians. 55 All these findings suggest that circulating PAI-1 levels are not closely dependent on fat mass but rather that they reflect fat redistribution and may be considered as a biomarker of ectopic fat storage. However, several questions remain unanswered. Is there a direct ectopic fat mass effect, an indirect connection between ectopic fat and PAI-1 through a mediator, or a common ground with a parallel evolution of PAI-1 and ectopic fat without a real connection?


Arguments for the Contribution of TNF and TGF-beta to PAI-1 Overexpression


TNF is involved in insulin resistance. 56 The group of Loskutoff was the first to emphasize the contribution of TNF in PAI-1 regulation during obesity. In ob/ob mice, deletion of both TNF receptors (TNF RI and RII) significantly reduced the plasma PAI-1 levels as well as the adipose tissue PAI-1 mRNA levels. TNF-neutralizing antibodies decreased plasma PAI-1 level, proving a direct link between TNF and PAI-1 during obesity. 57,58 Moreover, the invalidation of both TNF receptors decreased TGF-beta expression in the adipose tissue, 57 and in humans, TNF receptors, TGF-beta, and PAI-1 levels were strongly correlated within adipose tissue. 59,60 These results suggest that the TNF and TGF-beta pathways are connected within adipose tissue and may both control PAI-1 expression. The possible connection between insulin resistance, TGF-beta, and PAI-1 is further supported by the lowered expression of PAI-1 in FOXC2 +/- mice in response to TGF-beta1 treatment 61 because FOXC2 has been implicated in insulin resistance. 62,63 Thiazolidinediones decrease plasma PAI-1 levels 30,31,64 and inhibit PAI-1 synthesis through anti-TNF properties in the absence of inducible peroxisome proliferator activated receptor (PPAR ) activation, 65 indicating that the TNF pathway is a potential PAI-1 inducer during the MetS and underscoring the ability of thiazolinedione to exert anti-inflammatory properties independent of PPAR activation in several cell types. 66


In summary, the parallelism between circulating PAI-1 levels and the features of the MetS may be the reflection of an active TNF/TGF-beta pathway that modulates both insulin resistance and PAI-1 synthesis.


Possible Contribution of Local Cortisol Production


Clinical observations have highlighted the link between glucocorticoids and visceral obesity. Dexamethasone and cortisol are potent inducers of PAI-1 synthesis by cultured adipocytes and human adipose tissue, 67,68 making cortisol a possible inducer of PAI-1 during visceral obesity. Excess of circulating cortisol has failed to be demonstrated during obesity but a local cortisol production may occur within adipose tissue. 11-beta-hydroxysteroid dehydrogenase (11ß-HSD1) is expressed in most tissues. It potentiates the action of endogenous glucocorticoids by converting inactive cortisone into cortisol. Several recent experiments suggested that MetS may result from elevated 11ß-HSD1 activity. 69,70 Interestingly, in adipose tissue, 11ß-HSD1 levels paralleled those of PAI-1. Using human adipose tissue explants we observed that inactive cortisone stimulated PAI-1 secretion in an 11 ß-HSD1-dependent manner. 71 11-beta-HSD1 may be a central player in linking PAI-1, the MetS, and atherosclerosis because beside its improvement of MetS 11-beta-HSD1 inhibition prevents progression of atherosclerosis in mice. 72


The Culprit May Be Glucidolipidic Disturbances Associated With the MetS


Several groups as well as ours have suggested hyperinsulinemia and hypertriglyceridemia contribute to PAI-1 synthesis. Most cell culture experiments confirm that excesses of insulin or proinsulin, 73-75 free fatty acid, or very-low-density lipoprotein (VLDL), 76-78 directly increase PAI-1 synthesis. The apparent contradiction between the inability of insulin to induce glucose uptake and its capacity to stimulate PAI-1 synthesis led some investigators to demonstrate that some genes became insulin-resistant, whereas others, including PAI-1, continued to respond normally to insulin although insulin resistance was established. 79 Moreover the signaling pathway of PAI-1 synthesis differs in normal and insulin-resistant adipocytes. 80 This difference supports the hypothesis that the signaling pathways that remain insulin-sensitive may contribute to vascular disease associated with obesity and type II diabetes.


Such a direct effect of insulin or lipoprotein is not always supported by clinical observations. Postprandial hyperinsulinemia and hypertriglyceridemia as well as hyperinsulinemia induced during a hyperinsulinemic euglycemic clamp are not associated with increased circulating level of PAI-1 in healthy or obese subjects. 55,81-83 Even more, low-dose insulin infusion was shown to decrease Egr-1, a pro-inflammatory transcription factor, in mononuclear cells as well as some prothrombotic factors such as tissue factor and PAI-1 plasma levels in obese individuals. 84


Possible Contribution of the Renin Angiotensin System


The renin-angiotensin system mainly controls blood pressure. Angiotensin-converting enzyme inhibition significantly reduces plasma PAI-1 in obese subjects. 85 The renin angiotensin system is completely expressed in human adipose tissue and several reports suggested it plays a role on PAI-1 synthesis. Angiotensin II promotes PAI-1 production and release in cultured human adipocytes via the angiotensin II type 1 receptor (AT1 receptor), and AT1 receptor blockade reduces basal PAI-1 release and abolishes angiotensin II-stimulated PAI-1 release from adipocytes. 86 It is not known how angiotensin II acts on PAI-1 synthesis. Angiotensin II may promote PAI-1 secretion in many ways because it has been involved in local uptake, synthesis, and oxidation of lipids, inflammation, as well as cellular migration and proliferation mechanisms, but a more direct pathway cannot be ruled out. 87,88


Oxidative Stress as a Central Player


In nondiabetic humans, fat accumulation as well as PAI-1 level closely positively correlates with the markers of systemic oxidative stress. 89 Production of reactive oxygen species (ROS) increased selectively in adipose tissue of obese mice, partly because of augmented expression of NADPH oxidase and lowered expression of antioxidative enzymes induced by fat accumulation. This locally increased oxidative stress dysregulates the production of adipokines by adipose tissue such as TNF, MCP-1, and PAI-1. 90 Macrophages infiltrated in adipose tissue 91 may be involved in this process by elevating ROS production in the obese adipose tissue. Remarkably, obese mice treated with an NADPH oxidase inhibitor, adipocynin, showed reduced ROS production, improved diabetes and hyperlipidemia, and attenuated dysregulation of adipokines. 90 Thus, oxidative stress in adipose tissue and probably in other tissues may play a central role in linking most of the features that characterize the MetS and plasma PAI-1 levels.


There has been a long-lasting debate about the role of ROS in oxygen sensing. Interestingly, hypoxia and ROS increase PAI-1 expression in adipocytes via distinct signaling pathways, suggesting that both may participate in PAI-1 overexpression during obesity. 92


Place of the Circadian Rhythm of PAI-1


Finally, one could wonder whether the dysregulation of PAI-1 synthesis observed during the MetS may not be driven by dysregulation of the circadian clock, an endogenous self-sustained machinery of rhythmically acting transcriptional loops. Both clock:bmal1 and clock:bmal2 heterodimers activate the PAI-1 promoter. 93 Deletion of the Clock and Bmal1 genes results not only in circadian disturbances but also in metabolic abnormalities of lipid and glucose homeostasis, a phenotype resembling the MetS. 94,95 It could thus be suspected that PAI-1 synthesis dysregulation in obese patients is secondary to alteration of the self-regulated circadian clock, but this dysregulation needs to be elucidated.


In conclusion, the causes of PAI-1 overexpression in the MetS are complex, with much interference between biological systems. Establishment of inflammation or oxidative stress at the macrophage level as fundamental precursors is tempting and may reveal interesting avenues for a better understanding of the link between atherosclerosis and the MetS.


Contribution of PAI-1 to the Development of Adipose Tissue and Insulin Resistance


Clinical Evidence that PAI-1 Is Diabetogenic


High PAI-1 levels may help to identify a high-risk population with the potential of developing atherosclerotic disease and type II diabetes. Indeed, Festa et al showed that high plasma PAI-1 levels predict the development of diabetes. In their study the association of CRP and fibrinogen with incident diabetes was significantly attenuated after adjustment for body fat, waist circumference, or insulin sensitivity. In a logistic regression model that included age, sex, ethnicity, clinical center, smoking, BMI, insulin sensitivity, physical activity, and family history of diabetes, PAI-1 still remained significantly related to incident type II diabetes. 96 Furthermore, the same group has recently shown that progression of PAI-1 levels over time, in addition to high baseline PAI-1 levels, is associated with incident diabetes. 97 Similar findings were obtained in 2 other populations. 98,99 Based on these results it has been hypothesized that PAI-1 participates in the development of key features of the MetS. This hypothesis is also sustained by the relationship between PAI-1 gene polymorphisms, obesity, and insulin resistance in population studies. The PAI-1 gene polymorphism 4G/5G in the promoter region in position -675 has been especially studied; the 4G allele is associated with increased PAI-1 transcription compared with the 5G allele in in vitro studies and with increased plasma PAI-1 levels in vivo. 100 In some studies, 4G allele carriers were more prone to obesity and MetS but not in others. 101-104 We have recently shown there is an interaction between insulin and pro-insulin levels and the -675 4G/5G PAI-1 gene polymorphism for the risk of myocardial infarction. Patients with the highest pro-insulin levels were at risk for myocardial infarction only if they were homozygous for the 4G allele, suggesting that PAI-1 genotype may influence the vascular risk associated with hyperinsulinemia. 105 All together, these results are in favor of a role of PAI-1 gene variability in the modulation of obesity-associated phenotypes.


In Vitro Support of the Role of PAI-1 in Insulin Signaling and Adipocyte Differentiation


Studies on cultured fibroblasts showed that PAI-1 interferes with insulin signaling by preventing binding of vitronectin to integrin v ß 3 and can inhibit insulin-induced protein kinase B phosphorylation. 106,107 PAI-1 also binds IGF-5 binding protein and thus impairs insulin action. 108 Studies with adipocytes revealed interesting results. Overexpression of PAI-1 by adenovirus-mediated gene transfer inhibited differentiation. Conversely, preadipocytes from PAI-1 -/- mice showed greater differentiation than those issued from wild type mice and exhibited enhanced basal as well as insulin-stimulated glucose uptake. Inhibition of PAI-1 with a neutralizing antibody promoted 3T3 adipocyte differentiation. Remarkably, PAI-1 deficiency was able to blunt the deleterious effect induced by TNF on glucose uptake and on adipocyte differentiation marker expression levels. 109 Intriguingly, using a synthetic PAI-1 inhibitor we recently observed not an increase but a decrease in human adipocyte differentiation. 110 This effect could be attributed to the human origin of the cells or to unknown properties of the inhibitor.


In Vivo Support of the Role of PAI-1 in Obesity and the Related Glucidolipidic Disturbances


The effect of PAI-1 excess has been investigated in vivo. Mice overexpressing murine PAI-1 under the control of the aP2 promoter develop high PAI-1 expression within adipose tissue. 111 These mice exhibited adipocyte hypotrophy and a higher mRNA level of a preadipocyte marker in adipose tissue, suggesting adipocyte differentiation potential is decreased. These differences were exacerbated under high-fat diet with a significant lower body weight and smaller adipocytes associated with a lower feeding efficiency in transgenic mice. These findings suggest that PAI-1 overexpression induces impaired adipose tissue growth, which is in line with the in vitro effect of PAI-1 on murine adipocyte differentiation described earlier. 110 When looking at the metabolic parameters, it appears that old transgenic mice maintained on standard fat diet exhibit significantly higher insulinemia and a tendency to higher triglyceride levels despite lower body fat. 112 These data indicate that PAI-1 overexpression may worsen the metabolic profile; these differences, however, were not found when younger transgenic mice were subjected to high-fat diet. 111 One could wonder whether local and/or systemic PAI-1 contributes to this phenotype and whether PAI-1 is directly or indirectly involved through the modulation of TNF or TGF-beta actions. PAI-1 deficiency protects against TNF effects on adipocytes 109 and it has been recently hypothesized that PAI-1 could exert its action through inhibition of the proprotein convertase, furine, involved in TGF-beta activation and insulin receptor shedding. 113


Because of the improved insulin-stimulated glucose uptake and increased differentiation induced by PAI-1 inhibition in murine cultured adipocytes, one could expect PAI-1 deficiency to lead to higher subcutaneous fat accumulation in vivo under high-fat diet. Whereas 2 studies did not demonstrate any effect of PAI- deficiency on weight gain, 114,115 2 groups found that fat accumulation was prevented with a concomitant improvement in insulin sensitivity in mice lacking PAI-1 in 2 kinds of models, a nutritionally induced 116 and a genetic 117 murine model of obesity. The protection against obesity was linked to an increase in metabolic rate, total energy expenditure, and thermogenesis. These findings suggest the plasminogen activation system may be implicated in the control of fat accumulation in a more systemic way than that initially proposed. In the adult central nervous system, tissue-type plasminogen activator (tPA) is expressed at the mRNA and protein levels in many sites. 118,119 tPA is considered a major relay to the hypothalamic paraventricular nucleus controlling satiety and conveying both excitatory and inhibitory information to the hypothalamic-pituitary-adrenal axis. 120 It could reasonably be proposed that inhibition of tPA in the central nervous system by excess systemic or local PAI-1 may affect the control of body weight by the central nervous system; this aspect needs to be investigated.


Interestingly, we recently observed that inhibition of PAI-1 with the same synthetic inhibitor previously cited, 110 may improve insulin sensitivity in mice. This synthetic, low-molecular-weight PAI-1 inhibitor was added to the normal chow of wild-type mice for 4 weeks. After insulin injection, glycemia was lower in treated animals as insulin levels after glucose injection, suggesting higher insulin sensitivity in treated mice. 112 During high-fat diet, mice treated with the same PAI-1 inhibitor had lower body weight, glycemia, and triglyceride level than nontreated mice. 110 Overall, these data support the concept that PAI inhibition has the potential to reduce obesity and improve insulin sensitivity and may represent a new therapeutic target. This needs to be confirmed in different experimental models and the mechanisms involved should be precisely defined.


Conclusion


Several new features have been added to the MetS over time because they were frequently found associated with the metabolic syndrome. PAI-1, the main inhibitor of the fibrinolytic system, belongs to this cluster and could be considered a true component of the MetS. The mechanisms linking PAI-1 to the MetS are complex and probably interrelated, and several inducers may act jointly and at several sites of synthesis. In vitro and in vivo studies have indicated that PAI-1 might be involved in the development of obesity. Thus PAI-1 may serve as a feedback loop to limit adipose tissue expansion. Further efforts with experimental and clinical studies are needed to better understand this complex interplay. In any case, these findings support the rationale to develop PAI-1 inhibitors as they may serve to control atherothrombosis and insulin resistance.


Acknowledgments


Disclosures


None.

【参考文献】
  Lijnen HR, Collen D. Mechanisms of physiological fibrinolysis. Baillieres Clin Haematol. 1995; 8: 277-290.

Dellas C, Loskutoff DJ. Historical analysis of PAI-1 from its discovery to its potential role in cell motility and disease. Thromb Haemost. 2005; 93: 631-640.

Lijnen HR. Pleiotropic functions of plasminogen activator inhibitor-1. J Thromb Haemost. 2005; 3: 35-45.

Sobel BE. Increased plasminogen activator inhibitor-1 and vasculopathy. A reconcilable paradox. Circulation. 1999; 99: 2496-2498.Sobel reconciable paradox.

Kohler HP, Grant PJ. Plasminogen-activator inhibitor type 1 and coronary artery disease. N Engl J Med. 2000; 342: 1792-1801.

Sobel BE, Taatjes DJ, Schneider DJ. Intramural plasminogen activator inhibitor type-1 and coronary atherosclerosis. Arterioscler Thromb Vasc Biol. 2003; 23: 1979-1989.

Hamsten A, de Faire U, Walldius G, Dahlen G, Szamosi A, Landou C, Blomback M, Wiman B. Plasminogen activator inhibitor in plasma: risk factor for recurrent myocardial infarction. Lancet. 1987; 2: 3-9.

Juhan-Vague I, Pyke SDM, Alessi MC, Jespersen J, Haverkate F, Thompson SG. Fibrinolytic factors and the risk of myocardial infarction or sudden death in patients with angina pectoris. Circulation. 1996; 94: 2057-2063.

Thogersen AM, Jansson JH, Boman K, Nilsson TK, Weinehall L, Huhtasaari F, Hallmans G. High plasminogen activator inhibitor and tissue plasminogen activator levels in plasma precede a first acute myocardial infarction in both men and women: evidence for the fibrinolytic system as an independent primary risk factor. Circulation. 1998; 98: 2241-2247.

Collet JP, Montalescot G, Vicaut E, Ankri A, Walylo F, Lesty C, Choussat R, Beygui F, Borentain M, Vignolles N, Thomas D. Acute release of plasminogen activator inhibitor-1 in ST-segment elevation myocardial infarction predicts mortality. Circulation. 2003; 108: 391-394.

Smith A, Patterson C, Yarnell J, Rumley A, Ben-Shlomo Y, Lowe G. Which hemostatic markers add to the predictive value of conventional riskfactors for coronary heart disease and ischemic stroke? The Caerphilly Study. Circulation. 2005; 112: 3080-3087.

Salomaa V, Riley W, Kark JD, Nardo C, Folsom AR. Non-insulin-dependent diabetes mellitus and fasting glucose and insulin concentrations are associated with arterial stiffness indexes. The ARIC Study. Atherosclerosis Risk in Communities Study. Circulation. 1995; 91: 1432-1443.

Bavenholm P, de Faire U, Landou C, Efendic S, Nilsson J, Wiman B, Hamsten A. Progression of coronary artery disease in young male post-infarction patients is linked to disturbances of carbohydrate and lipoprotein metabolism and to impaired fibrinolytic function. Eur Heart J. 1998; 19: 402-410.

Folsom AR, Pankow JS, Williams RR, Evans GW, Province MA, Eckfeldt JH. Fibrinogen, plasminogen activator inhibitor-1, and carotid intima-media wall thickness in the NHLBI Family Heart Study. Thromb Haemost. 1998; 79: 400-404.

de Maat MP, Bladbjerg EM, Drivsholm T, Borch-Johnsen K, Moller L, Jespersen J. Inflammation, thrombosis and atherosclerosis: results of the Glostrup study. J Thromb Haemost. 2003; 1: 950-957.

Anand SS, Yi Q, Gerstein H, Lonn E, Jacobs R, Vuksan V, Teo K, Davis B, Montague P, Yusuf S. Study of Health Assessment and Risk in Ethnic Groups; Study of Health Assessment and Risk Evaluation in Aboriginal Peoples Investigators. Relationship of metabolic syndrome and fibrinolytic dysfunction to cardiovascular disease. Circulation. 2003; 108: 420-425.

Eckel RH, Grundy SM, Zimmet PZ. The metabolic syndrome. Lancet. 2005; 365: 1415-1428.

Malik S, Wong ND, Franklin SS, Kamath TV, L?Italien GJ, Pio JR, Williams GR. Impact of the metabolic syndrome on mortality from coronary heart disease, cardiovascular disease, and all causes in United States adults. Circulation. 2004; 110: 1245-1250.

Hu G, Qiao Q, Tuomilehto J, Balkau B, Borch-Johnsen K, Pyorala K, DECODE Study Group. Prevalence of the metabolic syndrome and its relation to all-cause and cardiovascular mortality in nondiabetic European men and women. Arch Intern Med. 2004; 164: 1066-1076.

Kahn R, Buse J, Ferrannini E, and Stern M. for the Am Diabetes Association and the European Association for the Study of Diabetes. The metabolic syndrome: time for a critical appraisal (ADA statement). Diabetes Care. 2005; 28: 2289-2304.

Grundy SM, Cleeman JI, Daniels SR, Donato KA, Eckel RH, Franklin BA, Gordon DJ, Krauss RM, Savage PJ, Smith SC Jr., Spertus JA, Costa F, Am Heart Association; National Heart, Lung, and Blood Institute. Diagnosis and management of the metabolic syndrome (scientific statement). Circulation. 2005; 112: 2735-2752.

Ravussin E, Smith SR. Increased fat intake, impaired fat oxidation, and failure of fat cell proliferation result in ectopic fat storage, insulin resistance, and type II diabetes mellitus. Ann N Y Acad Sci. 2002; 967: 363-378.

Boden G, Shulman GI. Free fatty acids in obesity and type II diabetes: defining their role in the development of insulin resistance and beta-cell dysfunction. Eur J Clin Invest. 2002; 32 Suppl 3: 14-23.

Hutley L, Prins JB. Fat as an endocrine organ: relationship to the metabolic syndrome. Am J Med Sci. 2005; 330: 280-289.

Vague P, Juhan-Vague I, Aillaud MF, Badier C, Viard R, Alessi MC, Collen D. Correlation between blood fibrinolytic activity, plasminogen activator inhibitor level, plasma insulin level, and relative body weight in normal and obese subjects. Metabolism. 1986; 35: 250-253.

Juhan-Vague I, Alessi MC, Vague P. Increased plasma plasminogen activator inhibitor 1 levels. A possible link between insulin resistance and atherothrombosis. Diabetologia. 1991; 34: 457-462.

Juhan-Vague I, Alessi MC, Mavri A, Morange PE. Plasminogen activator inhibitor-1, inflammation, obesity, insulin resistance and vascular risk. J Thromb Haemost. 2003; 1: 1575-1579.

Henry M, Tregouet DA, Alessi MC, Aillaud MF, Visvikis S, Siest G, Tiret L, Juhan-Vague I. Metabolic determinants are much more important than genetic polymorphisms in determining the PAI-1 activity and antigen plasma concentrations: a family study with part of the Stanislas Cohort. Arterioscler Thromb Vasc Biol. 1998; 18: 84-91.

Folsom AR, Qamhieh HT, Wing RR, Jeffery RW, Stinson VL, Kuller LH, Wu KK. Impact of weight loss on plasminogen activator inhibitor (PAI-1), factor VII, and other hemostatic factors in moderately overweight adults. Arterioscler Thromb. 1993; 13: 162-169.

Kruszynska YT, Yu JG, Olefsky JM, Sobel BE. Effects of troglitazone on blood concentrations of plasminogen activator inhibitor 1 in patients with type 2 diabetes and in lean and obese normal subjects. Diabetes. 2000; 49: 633-639.

Trost S, Pratley R, Sobel B. Impaired fibrinolysis and risk for cardiovascular disease in the metabolic syndrome and type II diabetes. Curr Diab Rep. 2006; 6: 47-54.

Mertens I, Verrijken A, Michiels JJ, Van der Planken M, Ruige JB, Van Gaal LF. Among inflammation and coagulation markers, PAI-1 is a true component of the metabolic syndrome. Int J Obes. 2006; .

Sobel BE, Woodcock-Mitchell J, Schneider DJ, Holt RE, Marutsuka K, Gold H. Increased plasminogen activator inhibitor type 1 in coronary artery atherectomy specimens from type 2 diabetic compared with nondiabetic patients: a potential factor predisposing to thrombosis and its persistence. Circulation. 1998; 97: 2213-2221.

Pandolfi A, Cetrullo D, Polishuck R, Alberta MM, Calafiore A, Pellegrini G, Vitacolonna E, Capani F, Consoli A. Plasminogen activator inhibitor type 1 is increased in the arterial wall of type II diabetic subjects. Arterioscler Thromb Vasc Biol. 2001; 21: 1378-1382.

Sakkinen PA, Wahl P, Cushman M, Lewis MR, Tracy RP. Clustering of procoagulation, inflammation, and fibrinolysis variables with metabolic factors in insulin resistance syndrome. Am J Epidemiol. 2000; 152: 897-907.

Rega G, Kaun C, Weiss TW, Demyanets S, Zorn G, Kastl SP, Steiner S, Seidinger D, Kopp CW, Frey M, Roehle R, Maurer G, Huber K, Wojta J. Inflammatory cytokines interleukin-6 and oncostatin m induce plasminogen activator inhibitor-1 in human adipose tissue. Circulation. 2005; 111: 1938-1945.

Kruithof EK, Mestries JC, Gascon MP, Ythier A. The coagulation and fibrinolytic responses of baboons after in vivo thrombin generation-effect of interleukin 6. Thromb Haemost. 1997; 77: 905-910.

Juhan-Vague I, Alessi MC, Joly P, Thirion X, Vague P, Declerck PJ, Serradimigni A, Collen D. Plasma plasminogen activator inhibitor-1 in angina pectoris. Influence of plasma insulin and acute-phase response. Arteriosclerosis. 1989; 9: 362-367.

Lundgren CH, Brown SL, Nordt TK, Sobel BE, Fujii S. Elaboration of type-1 plasminogen activator inhibitor from adipocytes. A potential pathogenetic link between obesity and cardiovascular disease. Circulation. 1996; 93: 106-110.

Ihara H, Urano T, Takada A, Loskutoff DJ. Induction of plasminogen activator inhibitor 1 gene expression in adipocytes by thiazolidinediones. FASEB J. 2001; 15: 1233-1235.

Voros G, Maquoi E, Collen D, Lijnen HR. Differential expression of plasminogen activator inhibitor-1, tumor necrosis factor-alpha, TNF-alpha converting enzyme and ADAMTS family members in murine fat territories. Biochim Biophys Acta. 2003; 1625: 36-42.

Samad F, Loskutoff DJ. The fat mouse: a powerful genetic model to study elevated plasminogen activator inhibitor 1 in obesity/NIDDM. Thromb Haemost. 1997; 78: 652-655.

Samad F, Yamamoto K, Loskutoff DJ. Distribution and regulation of plasminogen activator inhibitor-1 in murine adipose tissue in vivo. Induction by tumor necrosis factor-alpha and lipopolysaccharide. J Clin Invest. 1996; 97: 37-46.

Alessi MC, Peiretti F, Morange P, Henry M, Nalbone G, Juhan-Vague I. Production of plasminogen activator inhibitor 1 by human adipose tissue: possible link between visceral fat accumulation and vascular disease. Diabetes. 1997; 46: 860-867.

Eriksson P, Reynisdottir S, Lonnqvist F, Stemme V, Hamsten A, Arner P. Adipose tissue secretion of plasminogen activator inhibitor-1 in non-obese and obese individuals. Diabetologia. 1998; 41: 65-71.

Bastelica D, Morange P, Berthet B, Borghi H, Lacroix O, Grino M, Juhan-Vague I, Alessi MC. Stromal cells are the main plasminogen activator inhibitor-1 producing cells in human fat: evidence of differences between visceral and subcutaneous deposits. Arterioscler Thromb Vasc Biol. 2002; 22: 173-178.

Fain JN, Madan AK, Hiler ML, Cheema P, Bahouth SW. Comparison of the release of adipokines by adipose tissue, adipose tissue matrix, and adipocytes from visceral and subcutaneous abdominal adipose tissues of obese humans. Endocrinology. 2004; 145: 2273-2282.

Crandall DL, Quinet EM, Morgan GA, Busler DE, McHendry-Rinde B, Kral JG. Synthesis and secretion of plasminogen activator inhibitor-1 by human preadipocytes. J Clin Endocrinol Metab. 1999; 84: 3222-3227.

Wang B, Jenkins JR, Trayhurn P. Expression and secretion of inflammation-related adipokines by human adipocytes differentiated in culture: integrated response to TNF-alpha. Am J Physiol Endocrinol Metab. 2005; 288: E731-E740.

Vague P, Juhan-Vague I, Chabert V, Alessi MC, Atlan C. Fat distribution and plasminogen activator inhibitor activity in nondiabetic obese women. Metabolism. 1989; 38: 913-915.

Cigolini M, Targher G, Bergamo Andreis IA, Tonoli M, Agostino G, De Sandre G. Visceral fat accumulation and its relation to plasma hemostatic factors in healthy men. Arterioscler Thromb Vasc Biol. 1996; 16: 368-374.

Shimomura I, Funahashi T, Takahashi M, Maeda K, Kotani K, Nakamura T, Yamashita S, Miura M, Fukuda Y, Takemura K, Tokunaga K, Matsuzawa Y. Enhanced expression of PAI-1 in visceral fat: possible contributor to vascular disease in obesity. Nat Med. 1996; 2: 800-803.

Janand-Delenne B, Chagnaud C, Raccah D, Alessi MC, Juhan-Vague I, Vague P. Visceral fat as a main determinant of plasminogen activator inhibitor 1 level in women. Int J Obes Relat Metab Disord. 1998; 22: 312-317.

Alessi MC, Bastelica D, Mavri A, Morange P, Berthet B, Grino M, Juhan-Vague I. Plasma PAI-1 levels are more strongly related to liver steatosis than to adipose tissue accumulation. Arterioscler Thromb Vasc Biol. 2003; 23: 1262-1268.

Nagi DK, Tracy R, Pratley R. Relationship of hepatic and peripheral insulin resistance with plasminogen activator inhibitor-1 in Pima Indians. Metabolism. 1996; 45: 1243-1247.

Hotamisligil GS. The role of TNFalpha and TNF receptors in obesity and insulin resistance. J Intern Med. 1999; 245: 621-625.

Samad F, Uysal KT, Wiesbrock SM, Pandey M, Hotamisligil GS, Loskutoff DJ. Tumor necrosis factor alpha is a key component in the obesity-linked elevation of plasminogen activator inhibitor 1. Proc Natl Acad Sci U S A. 1999; 96: 6902-6907.

Pandey M, Tuncman G, Hotamisligil GS, Samad F. Divergent roles for p55 and p75 TNF-alpha receptors in the induction of plasminogen activator inhibitor-1. Am J Pathol. 2003; 162: 933-941.

Bastelica D, Mavri A, Verdierl M, Berthet B, Juhan-Vague I, Alessi MC. Relationships between fibrinolytic and inflammatory parameters in human adipose tissue: strong contribution of TNFalpha receptors to PAI-1 levels. Thromb Haemost. 2002; 88: 481-487.

Alessi MC, Bastelica D, Morange P, Berthet B, Leduc I, Verdier M, Geel O, Juhan-Vague I. Plasminogen activator inhibitor 1, transforming growth factor-beta1, and BMI are closely associated in human adipose tissue during morbid obesity. Diabetes. 2000; 49: 1374-1380.

Fujita H, Kang M, Eren M, Gleaves LA, Vaughan DE, Kume T. Foxc2 is a common mediator of insulin and transforming growth factor beta signaling to regulate plasminogen activator inhibitor type I gene expression. Circ Res. 2006; 98: 626-634.

Di Gregorio GB, Westergren R, Enerback S, Lu T, Kern PA. Expression of FOXC2 in adipose and muscle and its association with whole body insulin sensitivity. Am J Physiol Endocrinol Metab. 2004; 287: E799-E803.

Cederberg A, Gronning LM, Ahren B, Tasken K, Carlsson P, Enerback S. FOXC2 is a winged helix gene that counteracts obesity, hypertriglyceridemia and diet-induced insulin resistance. Cell. 2001; 106: 563-573.

Skurk T, Hauner H. Obesity and impaired fibrinolysis: role of adipose production of plasminogen activator inhibitor-1. Int J Obes Relat Metab Disord. 2004; 28: 1357-1364.

Liu HB, Hu YS, Medcalf RL, Simpson RW, Dear AE. Thiazolidinediones inhibit TNFalpha induction of PAI-1 independent of PPAR gamma activation. Biochem Biophys Res Commun. 2005; 19;334: 30-37.

Zhang J, Fu M, Zhao L, and Chen YE. 15-Deoxy-prostaglandin J (2) inhibits PDGF-A and -B chain expression in human vascular endothelial cells independent of PPAR gamma, Biochem Biophys Res Commun. 2002; 298: 128-132.

Morange PE, Aubert J, Peiretti F, Lijnen HR, Vague P, Verdier M, Negrel R, Juhan-Vague I, Alessi MC. Glucocorticoids and insulin promote plasminogen activator inhibitor 1 production by human adipose tissue. Diabetes. 1999; 48: 890-895.

Halleux CM, Declerck PJ, Tran SL, Detry R, Brichard SM. Hormonal control of plasminogen activator inhibitor-1 gene expression and production in human adipose tissue: stimulation by glucocorticoids and inhibition by catecholamines. J Clin Endocrinol Metab. 1999; 84: 4097-4105.

Kotelevtsev Y, Holmes MC, Burchell A, Houston PM, Schmoll D, Jamieson P, Best R, Brown R, Edwards CR, Seckl JR, Mullins JJ. 11beta-hydroxysteroid dehydrogenase type 1 knockout mice show attenuated glucocorticoid-inducible responses and resist hyperglycemia on obesity or stress. Proc Natl Acad Sci U S A. 1997; 94: 14924-14929.

Masuzaki H, Paterson J, Shinyama H, Morton NM, Mullins JJ, Seckl JR, Flier JS. A transgenic model of visceral obesity and the metabolic syndrome. Science. 2001; 294: 2166-2170.

Ayachi SE, Paulmyer-Lacroix O, Verdier M, Alessi MC, Dutour A, Grino M. 11beta-Hydroxysteroid dehydrogenase type 1-driven cortisone reactivation regulates plasminogen activator inhibitor type 1 in adipose tissue of obese women. J Thromb Haemost. 2006; 4: 621-627.

Hermanowski-Vosatka A, Balkovec JM, Cheng K, Chen HY, Hernandez M, Koo GC, Le Grand CB, Li Z, Metzger JM, Mundt SS, Noonan H, Nunes CN, Olson SH, Pikounis B, Ren N, Robertson N, Schaeffer JM, Shah K, Springer MS, Strack AM, Strowski M, Wu K, Wu T, Xiao J, Zhang BB, Wright SD, Thieringer R. 11beta-HSD1 inhibition ameliorates metabolic syndrome and prevents progression of atherosclerosis in mice. J Exp Med. 2005; 202: 517-527.

Alessi MC, Juhan-Vague I, Kooistra T, Declerck PJ, Collen D. Insulin stimulates the synthesis of plasminogen activator inhibitor 1 by the human hepatocellular cell line Hep G2. Thromb Haemost. 1988; 60: 491-494.

Kooistra T, Bosma PJ, Tons HA, van den Berg AP, Meyer P, Princen HM. Plasminogen activator inhibitor 1: biosynthesis and mRNA level are increased by insulin in cultured human hepatocytes. Thromb Haemost. 1989; 62: 723-728.

Schneider DJ, Nordt TK, Sobel BE. Stimulation by proinsulin of expression of plasminogen activator inhibitor type-I in endothelial cells. Diabetes. 1992; 41: 890-895.

Stiko-Rahm A, Wiman B, Hamsten A, Nilsson J. Secretion of plasminogen activator inhibitor-1 from cultured human umbilical vein endothelial cells is induced by very low density lipoprotein. Arteriosclerosis. 1990; 10: 1067-1073.

Nilsson L, Banfi C, Diczfalusy U, Tremoli E, Hamsten A, Eriksson P. Unsaturated fatty acids increase plasminogen activator inhibitor-1 expression in endothelial cells. Arterioscler Thromb Vasc Biol. 1998; 18: 1679-1685.

Banfi C, Mussoni L, Ris P, Cattaneo MG, Vicentini L, Battaini F, Galli C, Tremoli E. Very low density lipoprotein-mediated signal transduction and plasminogen activator inhibitor type 1 in cultured HepG2 cells. Circ Res. 1999; 85: 208-217.

Sartipy P, Loskutoff DJ. Expression profiling identifies genes that continue to respond to insulin in adipocytes made insulin-resistant by treatment with tumor necrosis factor-alpha. J Biol Chem. 2003; 278: 52298-52306.

Pandey M, Loskutoff DJ, Samad F. Molecular mechanisms of tumor necrosis factor-alpha-mediated plasminogen activator inhibitor-1 expression in adipocytes. FASEB J. 2005; 19: 1317-1319.

Potter van loon BJ, de Bart ACW, Radder JK, Frolich M, Kluft C, Meinders AE. Acute exogenous hyperinsulinemia does not result in elevation of plasma PAI-1 in humans. Fibrinolysis. 1990; 4: 93-94.

Grant PJ, Kruithof EK, Felley CP, Felber JP, Bachmann F. Short-term infusions of insulin, triacylglycerol and glucose do not cause acute increases in plasminogen activator inhibitor-1 concentrations in man. Clin Sci. 1990; 79: 513-516.

Lormeau B, Aurousseau MH, Valensi P, Paries J, Attali JR. Hyperinsulinemia and hypofibrinolysis: effects of short-term optimized glycemic control with continuous insulin infusion in type II diabetic patients. Metabolism. 1997; 46: 1074-1079.

Aljada A, Ghanim H, Mohanty P, Kapur N, Dandona P. Insulin inhibits the pro-inflammatory transcription factor early growth response gene-1 (Egr)-1 expression in mononuclear cells (MNC) and reduces plasma tissue factor (TF) and plasminogen activator inhibitor-1 (PAI-1) concentrations. J Clin Endocrinol Metab. 2002; 87: 1419-1422.

Brown NJ, Agirbasli MA, Williams GH, Litchfield WR, Vaughan DE. Effect of activation and inhibition of the renin-angiotensin system on plasma PAI-1. Hypertension. 1998; 32: 965-971.

Skurk T, Lee YM, Hauner H. Angiotensin II and its metabolites stimulate PAI-1 protein release from human adipocytes in primary culture. Hypertension. 2001; 37: 1336-1340.

Kerins DM, Hao Q, Vaughan DE. Angiotensin induction of PAI1 expression in endothelial cells is mediated by the hexapeptide angiotensin IV. J Clin Invest. 1995; 96: 2515-2520.

Vaughan DE, Lazos SA, Tong K. Angiotensin II regulates the expression of plasminogen activator inhibitor-1 in cultured endothelial cells. A potential link between the renin-angiotensin system and thrombosis. J Clin Invest. 1995; 95: 995-1001.

Ferroni P, Guagnano MT, Manigrasso MR, Ciabattoni G, Davi G. Increased plasminogen activator inhibitor-1 levels in android obesity: correlation with oxidative stress. J Thromb Haemost. 2005; 3: 1086-1087.

Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, Nakayama O, Makishima M, Matsuda M, Shimomura I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004; 114: 1752-1761.

Cancello R, Henegar C, Viguerie N, Taleb S, Poitou C, Rouault C, Coupaye M, Pelloux V, Hugol D, Bouillot JL, Bouloumie A, Barbatelli G, Cinti S, Svensson PA, Barsh GS, Zucker JD, Basdevant A, Langin D, Clement K. Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes. 2005; 54: 2277-2286.

Chen B, Lam KS, Wang Y, Wu D, Lam MC, Shen J, Wong L, Hoo RL, Zhang J, Xu A. Hypoxia dysregulates the production of adiponectin and plasminogen activatorinhibitor-1 independent of reactive oxygen species in adipocytes. Biochem Biophys Res Commun. 2006; 341: 549-556.

Schoenhard JA, Smith LH, Painter CA, Eren M, Johnson CH, Vaughan DE. Regulation of the PAI-1 promoter by circadian clock components: differential activation by BMAL1 and BMAL2. J Mol Cell Cardiol. 2003; 35: 473-481.

Turek FW, Joshu C, Kohsaka A, Lin E, Ivanova G, McDearmon E, Laposky A, Losee-Olson S, Easton A, Jensen DR, Eckel RH, Takahashi JS, Bass J. Obesity and metabolic syndrome in circadian Clock mutant mice. Science. 2005; 308: 1043-1045.

Rudic RD, McNamara P, Curtis AM, Boston RC, Panda S, Hogenesch JB, Fitzgerald GA. BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis. PLoS Biol. 2004; 2: 1893-1899.

Festa A, D?Agostino R Jr., Tracy RP, Haffner SM. Insulin Resistance Atherosclerosis Study. Elevated levels of acute-phase proteins and plasminogen activator inhibitor-1 predict the development of type II diabetes: the insulin resistance atherosclerosis study. Diabetes. 2002; 51: 1131-1137.

Festa A, Williams K, Tracy RP, Wagenknecht LE, Haffner SM. Progression of plasminogen activator inhibitor-1 and fibrinogen levels in relation to incident type II diabetes. Circulation. 2006; 113: 1753-1759.

Meigs JB, O?donnell CJ, Tofler GH, Benjamin EJ, Fox CS, Lipinska I, Nathan DM, Sullivan LM, D?Agostino RB, Wilson PW. Hemostatic markers of endothelial dysfunction and risk of incident type 2 diabetes: the Framingham Offspring Study. Diabetes. 2006; 55: 530-537.

Kanaya AM, Wassel Fyr C, Vittinghoff E, Harris TB, Park SW, Goodpaster BH, Tylavsky F, Cummings SR. Adipocytokines and incident diabetes mellitus in older adults: the independent effect of plasminogen activator inhibitor 1. Arch Intern Med. 2006; 166: 350-356.

Dawson S, Hamsten A, Wiman B, Henney A, Humphries S. Genetic variation at the plasminogen activator inhibitor-1 locus is associated with altered levels of plasma plasminogen activator inhibitor-1 activity. Arterioscler Thromb. 1991; 11: 183-190.

Hoffstedt J, Andersson IL, Persson L, Isaksson B, Arner P. The common -675 4G/5G polymorphism in the plasminogen activator inhibitor-1 gene is strongly associated with obesity. Diabetologia. 2002; 45: 584-587.

Lopes C, Dina C, Durand E, Froguel P. PAI-1 polymorphisms modulate phenotypes associated with the metabolic syndrome in obese and diabetic Caucasian population. Diabetologia. 2003; 46: 1284-1290.

Bouchard L, Mauriege P, Vohl MC, Bouchard C, Perusse L. Plasminogen activator inhibitor-1 polymorphisms are associated with obesity and fat distribution in the Quebec Family Study: evidence of interactions with menopause. Menopause. 2005; 12: 136-143.

Freeman MS, Mansfield MW. Comment on : The common -675 4G/5G polymorphism in the plasminogen activator inhibitor-1 gene is strongly associated with obesity. Diabetologia. 2002; 45: 1602-1603.

Juhan-Vague I, Morange PE, Frere C, Aillaud MF, Alessi MC, Hawe E, Boquist S, Tornvall P, Yudkin JS, Tremoli E, Margaglione M, Di Minno G, Hamsten A, Humphries SE; HIFMECH Study Group. The plasminogen activator inhibitor-1-675 4G/5G genotype influences the risk of myocardial infarction associated with elevated plasma proinsulin and insulin concentrations in men from Europe: the HIFMECH study. J Thromb Haemost. 2003; 1: 2322-2329.

Lebrun P, Baron V, Hauck CR, Schlaepfer DD, Van Obberghen E. Cell adhesion and focal adhesion kinase regulate insulin receptor substrate-1 expression. J Biol Chem. 2000; 275: 38371-38377.

Lopez-Alemany R, Redondo JM, Nagamine Y, Munoz-Canoves P. Plasminogen activator inhibitor type-1 inhibits insulin signaling by competing with alphavbeta3 integrin for vitronectin binding. Eur J Biochem. 2003; 270: 814-821.

Nam TJ, Busby W Jr, Clemmons DR. Insulin-like growth factor binding protein-5 binds to plasminogen activator inhibitor-I. Endocrinology. 1997; 138: 2972-2978.

Liang X, Kanjanabuch T, Mao SL, Hao CM, Tang YW, Declerck PJ, Hasty AH, Wasserman DH, Fogo AB, Ma LJ. Plasminogen activator inhibitor-1 modulates adipocyte differentiation. Am J Physiol Endocrinol Metab. 2006; 290: E103-E113.

Crandall DJ, Quinet EM, El Ayachi S, Hreha AL, Leik CE, SavioDA, Juhan-Vague I, Alessi MC. Modulation of adipose tissue evelopment by pharmacologic inhibition of PAI-1. Arterioscler Thromb Vasc Biol. In press

Lijnen HR, Maquoi E, Morange P, Voros G, Van Hoef B, Kopp F, Collen D, Juhan-Vague I, Alessi MC. Nutritionally induced obesity is attenuated in transgenic mice overexpressing plasminogen activator inhibitor-1. Arterioscler Thromb Vasc Biol. 2003; 23: 78-84.

Lijnen HR, Alessi MC, Van Hoef B, Collen D, Juhan-Vague I. On the role of plasminogen activator inhibitor-1 in adipose tissue development and insulin resistance in mice. J Thromb Haemost. 2005; 3: 1174-1179.

Griffiths SL, Grainger DJ. Proposal of a novel diabetogenic mechanism involving the serpin PAI-1. Bioessays. 2006; 28: 629-641.

Morange PE, Lijnen HR, Alessi MC, Kopp F, Collen D, Juhan-Vague I. Influence of PAI-1 on adipose tissue growth and metabolic parameters in a murine model of diet-induced obesity. Arterioscler Thromb Vasc Biol. 2000; 20: 1150-1154.

Lijnen HR. Effect of plasminogen activator inhibitor-1 deficiency on nutritionally-induced obesity in mice. Thromb Haemost. 2005; 93: 816-819.

Ma LJ, Mao SL, Taylor KL, Kanjanabuch T, Guan Y, Zhang Y, Brown NJ, Swift LL, McGuinness OP, Wasserman DH, Vaughan DE, Fogo AB. Prevention of obesity and insulin resistance in mice lacking plasminogen activator inhibitor 1. Diabetes. 2004; 53: 336-346.

Schafer K, Fujisawa K, Konstantinides S, Loskutoff DJ. Disruption of the plasminogen activator inhibitor 1 gene reduces the adiposity and improves the metabolic profile of genetically obese and diabetic ob/ob mice. FASEB J. 2001; 15: 1840-1842.

Teesalu T, Kulla A, Simisker A, Siren V, Lawrence DA, Asser T, Vaheri A. Tissue plasminogen activator and neuroserpin are widely expressed in the human central nervous system. Thromb Haemost. 2004; 92: 358-368.

Matys T, Pawlak R, Strickland S. Tissue plasminogen activator in the bed nucleus of stria terminalis regulates acoustic startle. Neuroscience. 2005; 135: 715-722.

Herman JP, and Cullinan WE. Neurocircuitry of stress: central control of the hypothalamo-pituitary-adrenocortical axis. Trends Neurosci. 1997; 20: 78-84.


作者单位:Marie-Christine Alessi; Irène Juhan-VagueFrom the Laboratory of Hematology, INSERM UMR 62 Faculty of Medicine, Marseilles, France.

作者: Links, Causes, and Consequences
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具