• Non ci sono risultati.

9 Adiponectin and the Cardiovascular System

N/A
N/A
Protected

Academic year: 2022

Condividi "9 Adiponectin and the Cardiovascular System "

Copied!
10
0
0

Testo completo

(1)

191

From: Contemporary Cardiology: Diabetes and Cardiovascular Disease, Second Edition Edited by: M. T. Johnstone and A. Veves © Humana Press Inc., Totowa, NJ

9 Adiponectin and the Cardiovascular System

Suketu Shah, MD , Alina Gavrila, MD , and Christos S. Mantzoros, MD

C

ONTENTS

I

NTRODUCTION

S

TRUCTURE OF

A

DIPONECTIN

R

EGULATION OF

A

DIPONECTIN

A

DIPONECTIN AND

C

ARDIOVASCULAR

D

ISEASE

R

ISK

F

ACTORS

A

DIPONECTIN

S

D

IRECT

V

ASCULAR

E

FFECTS

F

UTURE

T

HERAPEUTIC

D

IRECTIONS

C

ONCLUSION

R

EFERENCES

INTRODUCTION

Adiponectin, a recently discovered protein produced exclusively by adipocytes, is thought to be a possible mediator between obesity, insulin resistance, and cardiovascular disease (CVD). Although its function is not entirely known, body fat distribution, insulin, sex hormones, tumor necrosis factor (TNF)-F, and peroxisome proliferator-activated receptor (PPAR)-F may contribute to its regulation. Along with being associated with cardiovascular risk factors such as diabetes and dyslipidemia, deficiency in adiponectin may also directly compromise endothelial action and promote atherosclerosis.

Our understanding of the function of fat cells has changed dramatically with the

realization of the endocrine function of adipose tissue. Initially thought to serve only as

a repository for energy via storage of triglycerides, adipocytes are now known to secrete

a variety of proteins with diverse metabolic functions. These proteins include leptin,

TNF-F, plasminogen activator inhibitor-1, acylation-stimulating protein, resistin, and

adiponectin (1,2). Adiponectin has received much attention for its putative role in diabetes

and CVD. Besides being associated with the development of diabetes, it may also have

a direct role in modulating inflammation and atherosclerosis and thereby be one of the

factors that links obesity to CVD.

(2)

STRUCTURE OF ADIPONECTIN

In the mid-1990s, four different research groups, using either human or mouse-derived samples, concurrently discovered adiponectin, alternatively termed Acrp30, apM1 pro- tein, adipoQ, and GBP28 (3–6). Produced exclusively by differentiated adipocytes, adiponectin is a 30 kDa protein composed of 244 amino acids (3,4). One of the most abundantly produced fat hormones, it comprises approx 0.01% of the plasma proteins in humans, with plasma levels ranging from 2 to 20 μg/mL (7). Slightly increasing with age, adiponectin levels have a diurnal variation with nadir at night and peak in the morning (8,9).

The adiponectin molecule has four distinct parts, with the amino terminal having two short regions consisting of a secretory signal sequence and a domain unlike any other known protein (3–6). The next two regions, a collagen-like fibrous structure followed by a globular domain at the carboxy terminal, share homology with complement factor C1q and collagen VIII and X (10). Despite different amino acid sequences, the overall tertiary structure of the globular domain has similarity to TNF- F, another protein secreted by adipocytes but having opposing actions (11,12).

REGULATION OF ADIPONECTIN

Although its structure and source are known, the regulation of adiponectin remains to be determined. The various factors thought to be involved in controlling adiponectin production and secretion include obesity, nutritional status, hormones such as insulin, leptin, glucocorticoids, sex hormones, and catecholamines, TNF-F, and PPAR-F.

Obesity and Nutritional Intake

Obesity, in general, is associated with decreased adiponectin expression in adipose tissue and plasma levels (7,13). In both men and women, overall obesity, assessed by parameters such as body mass index (BMI) and fat mass, is negatively correlated to adiponectin, although prolonged weight reduction leads to increased adiponectin levels (7,14–17). Nutritional intake does not seem to explain this relationship. Although fasting decreases adiponectin messenger ribonucleic acid (mRNA) levels in mice, serum levels remain unchanged (18). In humans, short-term fasting also does not change plasma levels of adiponectin, although prolonged caloric restriction does result in weight loss and increased adiponectin levels (14,19). Additionally, daily caloric intake, macronutrient intake, or a high-fat meal is not related to any immediate change in circulating adiponectin levels in humans except possibly in obese individuals (20–22).

Instead of food intake, the distribution of adipose tissue may be more closely associated with adiponectin. There is a strong inverse correlation between adiponectin levels and visceral or central fat, compared to subcutaneous fat (9,19). In contrast to subcutaneous adipocytes, human omental adipose tissue had a significant negative correlation with BMI, and only it responded to insulin and PPAR- F agonist administration with increased adiponectin production (23). These findings suggest that adipose tissue, particularly in the visceral distribution, may have an inhibitory mechanism for its own production of adiponectin, perhaps mediated by other factors produced by fat cells such as TNF-F (13).

Hormone Regulation

Hormones have also been suggested to regulate adiponectin. Insulin likely has a role

in regulating adiponectin, but its exact role remains controversial. In vitro studies have

shown conflicting results on whether insulin has an inhibitory or stimulatory effect on

adiponectin production and secretion (23–25), whereas in an in vivo study involving

(3)

humans, hyperinsulinemic euglycemic dosing for at least 2 hours led to a decrease in adiponectin levels (26). Therefore, further studies would be helpful to resolve the rela- tionship.

Sex hormones may also affect secretion of adiponectin, because women have higher plasma levels of adiponectin than men, independent of body composition (14). Of the sex hormones, estrogen does not seem to account for the gender-related difference in adiponectin level, because premenopausal women have higher estrogen levels and lower adiponectin concentrations than postmenopausal females and estradiol levels actually have a strong negative correlation with serum adiponectin levels, females would be expected to have lower adiponectin concentrations than men (19). Testosterone may lower adiponectin levels by possibly inhibiting its secretion, however. In mice, removal of the testes led to an increase in adiponectin, although administration of testosterone reduced adiponectin levels (27). Although one study has demonstrated no association between adiponectin and free testosterone concentrations in women, this relationship remains to be explored in men (19).

Leptin and glucocorticoids have also been thought to be involved in adiponectin regulation, because leptin is also secreted by adipose tissue and both hormones affect insulin sensitivity (28,29). Although a cross-sectional study reported a strong inverse relationship between serum adiponectin and leptin levels (30), leptin given exogenously to rodents or humans had no significant effect on the plasma concentration of adiponectin (18,19). In vitro studies show that dexamethasone suppresses adiponectin gene expres- sion (24,25), but in human studies, cortisol was found to have no correlation with circulat- ing levels of adiponectin (19). Further studies are necessary to evaluate if glucocorticoids have a local effect on adiponectin production not reflected by their serum concentrations.

Catecholamines may also suppress expression of adiponectin, because G-adrenergic agonists reduced adiponectin gene expression in cultured mouse fat cells and human adipose tissue and decreased plasma levels in mice (31). Stimulation of cultured adipocytes by isoproteronolol, a G1 and G2 agonist, leads to reduced expression of adiponectin, an effect that propranolol, a nonselective G-antagonist, can inhibit (32).

Another study in animals confirmed that peripheral injection of a G3-adrenergic agonist suppressed adiponectin mRNA expression in adipose tissue (18).

TNF-F

As another factor produced by adipocytes, TNF- F may also be involved in the regu- lation of adiponectin. TNF- F and adiponectin inhibit each other’s production in adipose tissue, in addition to having opposing actions. TNF- F decreases expression and secretion of adiponectin in mouse and human adipocytes (25,33,34) and adiponectin-knockout mice have elevated serum TNF-F levels that decrease with adiponectin administration (35). Because these two molecules share, in part, similar tertiary structure, they may exert opposite actions by acting on the same cellular receptors (11,13).

PPAR-F

PPAR- F is a transcription factor that enhances insulin sensitivity in adipose and other

tissues (36). In a randomized, double blind, placebo-controlled trial, patients with type

2 diabetes given 6 months of rosiglitazone, a PPAR-F agonist, had increased levels of

adiponectin (37), with a similar change being seen even in humans without insulin resis-

tance (38). PPAR-F agonists may mediate their effect by directly promoting adiponectin

transcription or by inhibiting the actions of TNF-F (34,39).

(4)

ADIPONECTIN AND CARDIOVASCULAR DISEASE RISK FACTORS Although the role of adiponectin has not been definitively established, evidence is mounting that it is involved in insulin resistance, diabetes, inflammation, and atheroscle- rosis (40). Because, among the various adipocytokines, it decreases with increasing body fat (7), its low levels may lead to the development of pathological states associated with obesity such as insulin resistance and CVD.

Diabetes and Insulin Resistance

Adiponectin’s involvement in CVD is likely multifactorial, but one of its main roles is likely in affecting traditional risk factors associated with coronary artery disease (CAD), particularly diabetes. As one of the diabetes susceptibility genes and the adiponectin gene both localize to 3q27, mutation at this locus has been associated with both type 2 diabetes and decreased adiponectin (41).

The majority of data for animal studies thus far suggest that adiponectin acts as an insulin-sensitizing hormone. Adiponectin-knockout mice develop insulin resistance either independently of diet or only after high-fat and high-sucrose diet, and treating these mice with adiponectin ameliorates their insulin resistance (35,42). The insulin resistance in adiponectin-deficient lipoatrophic and obese mice can partially be reversed via adiponectin administration and fully restored with both leptin and adiponectin supple- mentation (29). Furthermore, in a longitudinal study analyzing the progression of type 2 diabetes in obese monkeys, decrease in adiponectin closely parallels the observed reduc- tion in insulin sensitivity, and the obese monkeys with greater plasma levels of adiponectin had less severe insulin resistance (43).

In humans, type 2 diabetes has been associated with decreased levels of adiponectin (14). In several studies, adiponectin has a negative correlation with fasting glucose, insulin, and insulin resistance and a positive association with insulin sensitivity, indepen- dent of BMI (9,14,44). One study demonstrated that adjusting for central obesity renders the negative correlation between adiponectin and insulin resistance no longer significant, suggesting that adiponectin may mediate the relationship between central obesity and insulin resistance (19). In studies involving Pima Indians, Japanese people, and Europe- ans, subjects with lower adiponectin were more likely to develop type 2 diabetes, inde- pendent of adiposity parameters (45–47). In contrast, type 1 diabetic patients have elevated adiponectin levels compared to nondiabetic individuals, and chronically admin- istered insulin does not have an effect on adiponectin levels (48).

Although not entirely known, the cellular and molecular mechanisms linking adiponectin to improved insulin sensitivity are also likely multifactorial. In rodents, adiponectin administration enhances insulin-stimulated glucose uptake into fat and skel- etal muscle cells (49–51). By increasing fatty acid oxidation, adiponectin can also lower circulating free fatty acids (FFAs), which may improve insulin action (51,52). Another important function of adiponectin is enhancement of insulin-induced suppression of hepatic glucose production (53,54). By generating nitric oxide (NO) formation, adiponectin may also augment vascular blood flow to promote glucose uptake (55).

Taken together, all these effects could explain why giving adiponectin to mice on a high- fat and high-sucrose diet will induce weight loss and reduction in FFA, triglycerides, and glucose levels (56).

Adiponectin may also improve insulin sensitivity by promoting activation of the

insulin-signaling system (58). The main enzyme implicated in adiponectin’s action is

(5)

adenosine monophosphate-activated protein kinase (AMPK) (49–51). A recent study demonstrates that binding of adiponectin to two distinct adiponectin receptors increases the levels of this enzyme (57). AMPK prevents activation of other enzymes involved in gluconeogenesis and may stimulate enzymes contributing to fatty acid oxidation (49,50,54,56).

Dyslipidemia

Besides diabetes and insulin resistance, adiponectin is also related to dyslipidemia, another risk factor for CVD. Adiponectin is a strong independent positive predictor of high-density lipoprotein levels and is negatively associated with serum triglycerides (14,59,60). In contrast, low-density lipoprotein and total cholesterol do not have signifi- cant independent relationships to adiponectin levels (19).

Adiponectin may lead to favorable lipid profiles by stimulating fatty acid oxidation.

The administration of adiponectin to rodents has been associated with increased fatty acid oxidation in skeletal muscle, both in vitro and in vivo, an effect probably mediated by AMPK (49,50,56). However, in one study, fatty acid oxidation in muscle cells was found to be increased in adiponectin-knockout mice (61), and in a single cross-sectional study in humans, plasma levels of adiponectin did not have any correlation with lipid oxidation, as measured by energy expenditure and respiratory quotient (62). Thus, further studies are needed to clarify adiponectin’s effects on fatty acid oxidation in humans.

Hypertension

Adiponectin has also been associated with hypertension. In adiponectin-deficient mice, a high-fat and -sucrose diet led to increased blood pressure (BP) (63). Although an initial study in humans reported that hypertensive males had increased plasma levels of adiponectin (64), subsequent studies reported that BP has a negative correlation to adiponectin (65–67). However, more recent data adjusting for insulin sensitivity did not show any significant correlation with hypertension and adiponectin, indicating that insulin resistance may mediate the potential association between adiponectin and BP (68). However, adiponectin has been associated with a vasodilatory response (63), with recent evidence suggesting that adiponectin increases NO formation through AMPK (55). Further studies are needed to elucidate more completely adiponectin’s role in regu- lating BP levels.

Cigarette Smoking

Smoking and even a history of smoking have been associated with decreased levels of circulating adiponectin. Among patients with heart disease, current and former smokers had lower adiponectin levels than nonsmokers, after adjusting for BMI and insulin resis- tance (69). Possible explanations for this decrease include smoking inducing an increase in catecholamines that suppress adiponectin or consumption of adiponectin by endothe- lium injured by cigarette toxins (69,70).

ADIPONECTIN’S DIRECT VASCULAR EFFECTS

Although development of insulin resistance and alterations in lipid profile may

account for part of adiponectin’s role in CVD, low adiponectin has also been associated

with CAD independent of these risk factors, suggesting that it may have its own direct

effect on the vascular system (14,71,72).

(6)

Adiponectin might have a protective role against atherosclerosis, because increasing adiponectin levels of mice deficient in apolipoprotein (apo)-E slows the rate of athero- sclerosis and reduces lipid accumulation in arterial plaques (73). In adiponectin-knock- out mice, injury induced to a femoral artery resulted in greater neointimal thickening compared to control, independent of degree of glucose intolerance (42), but overexpres- sion of adiponectin attenuated neointimal proliferation in these mice (74).

Adiponectin’s reduction of atherosclerosis may occur through its actions on inflam- matory mediators, macrophages, smooth muscle cells, and endothelium (40). With its association with inflammatory markers, lack of adiponectin may foster an inflammatory milieu related to developing atherosclerosis and diabetes (75). In patients with or without CAD, serum C-reactive protein, an inflammatory marker, was inversely related to adiponectin (75–77). Other inflammatory markers such as phospholipase A2, interleukin- 6, and soluble E (SE)-selectin, are also negatively correlated with adiponectin in one study (75).

As an anti-inflammatory agent, adiponectin may inhibit inflammatory mediators involved in atherosclerosis, particularly TNF-F. An in vivo study in mice demonstrated that administration of adiponectin decreased serum TNF-F levels (35). Although no significant correlation occurred between adiponectin and serum TNF-F receptors 1 (sTNFR1) and 2 (sTNFR2), which are markers of activation of the TNF-F system, a study in humans found lower sTNFR2 in the highest quartile of circulating adiponectin, sug- gesting a threshold effect instead of a dose-dependent relationship (78). A different study, however, found a significant negative correlation between plasma adiponectin and TNF- F mRNA expression (13). Besides inhibiting the production of TNF-F, adiponectin may impede TNF-F’s involvement in atherosclerosis by reducing TNF-F-induced expression of endothelial cell adhesion molecules such as vascular cell adhesion molecule-1, SE-selectin, and intracellular adhesion molecule-1, that otherwise recruit monocytes and macrophages involved in atherosclerosis development (79,80). Through a cyclic adenosine monophosphate (cAMP)-dependent pathway, adiponectin may prevent TNF- F from inducing stimulation of nuclear factor PB, a transcriptional factor that promotes gene expression of endothelial adhesion molecules (80).

The anti-inflammatory effects of adiponectin may also directly involve macrophages and monocytes, an integral aspect of atherosclerotic lesions. By decreasing the level of expression of class A scavenger receptors on macrophages, adiponectin suppressed macrophage-to-foam cell transformation (81). It also attenuated the phagocytic action of macrophages and inhibited expression and secretion of TNF-F from macrophages (82).

Additionally, adiponectin may decrease proliferation of precursors of monocytes and macrophages by suppressing bone marrow production of these cells (82).

In the pathogenesis of atherosclerosis, adiponectin may also affect the proliferation of smooth muscle cells in the vascular wall by inhibiting growth factors that promote hyperplasia. Adiponectin binds to and inhibits a subtype of platelet-derived growth factor produced by platelets and foam cells (83) and blocks the proliferative action of heparin- binding epidermal growth factor (HB-EGF)-like growth factor (74). It also prevents TNF-F from inducing increase in HB-EGF mRNA production (74).

Finally, adiponectin may mediate endothelial vasodilatation, because plasma

adiponectin was independently correlated with peak forearm blood flow and vasodilator

response to reactive hyperemia (36,84), and stimulate production of the vasodilatory

agent NO in vascular endothelial cells (55).

(7)

FUTURE THERAPEUTIC DIRECTIONS

Because the adverse cardiovascular events associated with obesity may be related to the relative decrease in adiponectin, supplementing this protein exogenously, increasing endogenous production, or designing agonists for its receptor need to be tested in relation to cardiovascular outcomes. PPAR-F agonists have been shown to increase adiponectin levels in lean, obese, and diabetic humans (38). Because their ability to improve insulin resistance may be mediated by adiponectin, they may also prove to have an added indication for CVD. Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers also may increase adiponectin (68), which may mediate in part their beneficial effects in insulin-resistant states including CVD. G-Adrenergic antagonists may also have similar use, as they prevent catecholamine-induced suppression of adiponectin production (32). Finally, with the discovery of adiponectin receptors, ago- nists at these sites may allow for targeted augmentation of adiponectin’s effects in meta- bolically active tissues.

CONCLUSION

Obesity has long been associated with insulin resistance, hypertension, and CAD, but the mechanism has remained largely unknown. Adiponectin may be one of the factors that explains these associations. Because deficiencies in adiponectin may result in the development of these processes, increased endogenous production or exogenously administered adiponectin or its agonists may contribute to restoring insulin sensitivity and preventing atherosclerosis by increasing fatty acid oxidation and insulin-mediated glucose uptake, and decreasing the endothelial inflammatory process associated with atherosclerotic plaque development. Although animal studies have demonstrated ben- efits, clinical trials are needed to determine whether the beneficial effects of adiponectin can also be observed in humans and whether either adiponectin or adiponectin receptor agonists represent a novel treatment option for type II diabetes and CAD.

ACKNOWLEDGMENT

This chapter was supported by NIDDK grant DK 58785 and NIH grant K30 HL04095.

REFERENCES

1. Rajala MW, Scherer PE. Minireview: The adipocytes—at the crossroads of energy homeostasis, inflam- mation, and atherosclerosis. Endocrinology 2003;144:3765–3773.

2. Havel PJ. Control of energy homeostasis and insulin action by adipocyte hormones: leptin, acylation stimulating protein, and adiponectin. Curr Opin Lipidol 2002;13:51–59.

3. Scherer PE, Williams S, Fogliano M, Baldini G, Lodish HF. A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 1995;270:26,746–26,749.

4. Maeda K, Okubo K, Shimomura I, Funahashi T, Matsuzawa Y, Matsubara K. cDNA cloning and expression of a novel adipose specific collagen-like factor, apM1 (AdiPose Most abundant Gene tran- script 1). Biochem Biophys Res Commun 1996;221:286–289.

5. Hu E, Liang P, Spiegelman BM. AdipoQ is a novel adipose-specific gene dysregulated in obesity. J Biol Chem 1996;271:10,697–10,703.

6. Nakano Y, Tobe T, Choi-Miura NH, Mazda T, Tomita M. Isolation and characterization of GBP28, a novel gelatin-binding protein purified from human plasma. J Biochem (Tokyo) 1996;120:803–812.

7. Arita Y, Kihara S, Ouchi N, et al. Paradoxical decrease of an adipose-specific protein, adiponectin, in obesity. Biochem Biophys Res Commun 1999;257:79–83.

(8)

8. Gavrila A, Peng CK, Chan JL, Mietus JE, Goldberger AL, Mantzoros CS. Diurnal and ultradian dynam- ics of serum adiponectin in healthy men: comparison with leptin, circulating soluble leptin receptor, and cortisol patterns. J Clin Endocrinol Metab 2003;88:2838–2843.

9. Cnop M, Havel PJ, Utzschneider KM, et al. Relationship of adiponectin to body fat distribution, insulin sensitivity and plasma lipoproteins: evidence for independent roles of age and sex. Diabetologia 2003;46:459–469.

10. Pajvani UB, Du X, Combs TP, et al. Structure-function studies of the adipocyte-secreted hormone Acrp30/

adiponectin. Implications fpr metabolic regulation and bioactivity. J Biol Chem 2003;278:9073–9085.

11. Shapiro L, Scherer PE. The crystal structure of a complement-1q family protein suggests an evolutionary link to tumor necrosis factor. Curr Biol 1998;8:335–338.

12. Ruan H, Lodish HF. Insulin resistance in adipose tissue: direct and indirect effects of tumor necrosis factor-alpha. Cytokine Growth Factor Rev 2003;14:447–455.

13. Kern PA, Di Gregorio GB, Lu T, Rassouli N, Ranganathan G. Adiponectin expression from human adipose tissue: relation to obesity, insulin resistance, and tumor necrosis factor-alpha expression. Dia- betes 2003;52:1779–1785.

14. Hotta K, Funahashi T, Arita Y, et al. Plasma concentrations of a novel, adipose-specific protein, adiponectin, in type 2 diabetic patients. Arterioscler Thromb Vasc Biol 2000;20:1595–1599.

15. Yang WS, Lee WJ, Funahashi T, et al. Weight reduction increases plasma levels of an adipose-derived anti-inflammatory protein, adiponectin. J Clin Endocrinol Metab 2001;86:3815–3819.

16. Faraj M, Havel PJ, Phelis S, Blank D, Sniderman AD, Cianflone K. Plasma acylation-stimulating protein, adiponectin, leptin, and ghrelin before and after weight loss induced by gastric bypass surgery in morbidly obese subjects. J Clin Endocrinol Metab 2003;88:1594–1602.

17. Esposito K, Pontillo A, Di Palo C, et al. Effect of weight loss and lifestyle changes on vascular inflam- matory markers in obese women: a randomized trial. JAMA 2003;289:1799–1804.

18. Zhang Y, Matheny M, Zolotukhin S, Tumer N, Scarpace PJ. Regulation of adiponectin and leptin gene expression in white and brown adipose tissues: influence of beta3-adrenergic agonists, retinoic acid, leptin and fasting. Biochim Biophys Acta 2002;1584:115–122.

19. Gavrila A, Chan JL, Yiannakouris N, et al. Serum adiponectin levels are inversely associated with central fat distribution and estrogen levels but are not directly regulated by acute fasting or leptin administration in humans: cross-sectional and interventional studies. J Clin Endocrinol Metab 2003;88:4823–4831.

20. Yannakoulia M, Yiannakouris N, Bluher S, Matalas AL, Klimis-Zacas D, Mantzoros CS. Body fat mass and macronutrient intake in relation to circulating soluble leptin receptor, free leptin index, adiponectin, and resistin concentrations in healthy humans. J Clin Endocrinol Metab 2003;88:1730–1736.

21. Peake PW, Kriketos AD, Denyer GS, Campbell LV, Charlesworth JA. The postprandial response of adiponectin to a high-fat meal in normal and insulin-resistant subjects. Int J Obes Relat Metab Disord 2003;27:657–662.

22. English PJ, Coughlin SR, Hayden K, Malik IA, Wilding JP. Plasma adiponectin increases postprandially in obese, but not in lean, subjects. Obes Res 2003;11:839–844.

23. Motoshima H, Wu X, Sinha MK, et al. Differential regulation of adiponectin secretion from cultured human omental and subcutaneous adipocytes: effects of insulin and rosiglitazone. J Clin Endocrinol Metab 2002;87:5662–5667.

24. Halleux CM, Takahashi M, Delporte ML, et al. Secretion of adiponectin and regulation of apM1 gene expression in human visceral adipose tissue. Biochem Biophys Res Commun 2001;288:1102–1107.

25. Fasshauer M, Klein J, Neumann S, Eszlinger M, Paschke R. Hormonal regulation of adiponectin gene expression in 3T3-L1 adipocytes. Biochem Biophys Res Commun 2002;290:1084–1089.

26. Mohlig M, Wegewitz U, Osterhoff M, et al. Insulin decreases human adiponectin plasma levels. Horm Metab Res 2002;34:655–658.

27. Nishizawa H, Shimomura I, Kishida K, et al. Androgens decrease plasma adiponectin, an insulin- sensitizing adipocyte-derived protein. Diabetes 2002;51:2734–2741.

28. Olefsky JM, Kruszynska YT. Causes of insulin resistance. In: De Groot, L. (ed). De Groot’s Textbook of Endocrinology, 4th ed. WB Saunders: Philadelphia, PA, 2001, pp. 780–782.

29. Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone adiponectin reverses insulin resistance associated with both lipoatrophy and obesity. Nat Med 2001;7:941–946.

30. Matsubara M, Maruoka S, Katayose S. Inverse relationship between plasma adiponectin and leptin concentrations in normal-weight and obese women. Eur J Endocrinol 2002;147:173–180.

31. Delporte ML, Funahashi T, Takahashi M, Matsuzawa Y, Brichard SM. Pre- and post-translational negative effect of beta-adrenoceptor agonists on adiponectin secretion: in vitro and in vivo studies.

Biochem J 2002;367:677–685.

32. Fasshauer M, Klein J, Neumann S, Eszlinger M, Paschke R Adiponectin gene expression is inhibited by beta-adrenergic stimulation via protein kinase A in 3T3-L1 adipocytes. FEBS Lett 2001;507:142–146.

(9)

33. Bruun JM, Lihn AS, Verdich C, et al. Regulation of adiponectin by adipose tissue-derived cytokines:

in vivo and in vitro investigations in humans. Am J Physiol Endocrinol Metab 2003;285:E527–E33.

34. Maeda N, Takahashi M, Funahashi T, et al. PPARgamma ligands increase expression and plasma concentrations of adiponectin, an adipose-derived protein. Diabetes 2001;50:2094–2099.

35. Maeda N, Shimomura I, Kishida K, et al. Diet-induced insulin resistance in mice lacking adiponectin/

ACRP30. Nat Med 2002;8:731–737.

36. Hsueh WA, Law R. The central role of fat and effect of peroxisome proliferator-activated receptor- gamma on progression of insulin resistance and cardiovascular disease. Am J Cardiol 2003;92:3J–9J.

37. Yang WS, Jeng CY, Wu TJ, et al. Synthetic peroxisome proliferator-activated receptor-gamma agonist, rosiglitazone, increases plasma levels of adiponectin in type 2 diabetic patients. Diabetes Care 2002;25:376–380.

38. Yu JG, Javorschi S, Hevener AL, et al. The effect of thiazolidinediones on plasma adiponectin levels in normal, obese, and type 2 diabetic subjects. Diabetes 2002;51:2968–2974.

39. Iwaki M, Matsuda M, Maeda N, et al. Induction of adiponectin, a fat-derived antidiabetic and antiatherogenic factor, by nuclear receptors. Diabetes 2003;52:1655–1663.

40. Diez JJ, Iglesias P. The role of the novel adipocyte-derived hormone adiponectin in human disease. Eur J Endocrinol 2003;148:293–300.

41. Kondo H, Shimomura I, Matsukawa Y, et al. Association of adiponectin mutation with type 2 diabetes:

a candidate gene for the insulin resistance syndrome. Diabetes 2002;51:2325–2328.

42. Kubota N, Terauchi Y, Yamauchi T, et al. Disruption of adiponectin causes insulin resistance and neointimal formation. J Biol Chem 2002;277:25,863–25,866.

43. Hotta K, Funahashi T, Bodkin NL, et al. Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes 2001;50:1126–1133.

44. Weyer C, Funahashi T, Tanaka S, et al. Hypoadiponectinemia in obesity and type 2 diabetes: close association with insulin resistance and hyperinsulinemia. J Clin Endocrinol Metab 2001;86:1930–1935.

45. Lindsay RS, Funahashi T, Hanson RL, et al. Adiponectin and development of type 2 diabetes in the Pima Indian population. Lancet 2002;360:57–58.

46. Spranger J, Kroke A, Mohlig M, et al. Adiponectin and protection against type 2 diabetes mellitus.

Lancet 2003;361:226–228.

47. Daimon M, Oizumi T, Saitoh T, et al. Decreased serum levels of adiponectin are a risk factor for the progression to type 2 diabetes in the Japanese Population: the Funagata study. Diabetes Care 2003;26:2015–2020.

48. Imagawa A, Funahashi T, Nakamura T, et al. Elevated serum concentration of adipose-derived factor, adiponectin, in patients with type 1 diabetes. Diabetes Care 2002;25:1665–1666.

49. Tomas E, Tsao TS, Saha AK, et al. Enhanced muscle fat oxidation and glucose transport by ACRP30 globular domain: acetyl-CoA carboxylase inhibition and AMP-activated protein kinase activation. Proc Natl Acad Sci USA 2002;99:16,309–16,313.

50. Yamauchi T, Kamon J, Minokoshi Y, et al. Adiponectin stimulates glucose utilization and fatty-acid oxidation by activating AMP-activated protein kinase. Nat Med 2002;8:1288–1295.

51. Wu X, Motoshima H, Mahadev K, Stalker TJ, Scalia R, Goldstein BJ. Involvement of AMP-activated protein kinase in glucose uptake stimulated by the globular domain of adiponectin in primary rat adipocytes. Diabetes 2003;52:1355–1363.

52. Boden G. Role of fatty acids in the pathogenesis of insulin resistance and NIDDM. Diabetes 1997; 46:

3–10.

53. Berg AH, Combs TP, Du X, Brownlee M, Scherer PE. The adipocyte-secreted protein Acrp30 enhances hepatic insulin action. Nat Med 2001;7:947–953.

54. Combs TP, Berg AH, Obici S, Scherer PE, Rossetti L. Endogenous glucose production is inhibited by the adipose-derived protein Acrp30. J Clin Invest 2001;108:1875–1881.

55. Chen H, Montagnani M, Funahashi T, Shimomura I, Quon MJ. Adiponectin stimulates production of nitric oxide in vascular endothelial cells. J Biol Chem 2003;278:45,021–45,026.

56. Fruebis J, Tsao TS, Javorschi S, et al. Proteolytic cleavage product of 30-kDa adipocyte complement- related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc Natl Acad Sci USA 2001;98:2005–2010.

57. Yamauchi T, Kamon J, Ito Y, et al. Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 2003;423:762–769.

58. Stefan N, Vozarova B, Funahashi T, et al. Plasma adiponectin concentration is associated with skeletal muscle insulin receptor tyrosine phosphorylation, and low plasma concentration precedes a decrease in whole-body insulin sensitivity in humans. Diabetes 2002;51:1884–1888.

(10)

59. Zietz B, Herfarth H, Paul G, et al. Adiponectin represents an independent cardiovascular risk factor predicting serum HDL-cholesterol levels in type 2 diabetes. FEBS Lett 2003;545:103–104.

60. Matsubara M, Maruoka S, Katayose S. Decreased plasma adiponectin concentrations in women with dyslipidemia. J Clin Endocrinol Metab 2002;87:2764–2769.

61. Ma K, Cabrero A, Saha PK, et al. Increased beta-oxidation but no insulin resistance or glucose intoler- ance in mice lacking adiponectin. J Biol Chem 2002;277:34658–34661.

62. Stefan N, Vozarova B, Funahashi T, et al. Plasma adiponectin levels are not associated with fat oxidation in humans. Obes Res 2002;10:1016–1020.

63. Ouchi N, Ohishi M, Kihara S, et al. Association of hypoadiponectinemia with impaired vasoreactivity.

Hypertension 2003;42:231–234.

64. Mallamaci F, Zoccali C, Cuzzola F, et al. Adiponectin in essential hypertension. J Nephrol 2002;15:

507–511.

65. Adamczak M, Wiecek A, Funahashi T, Chudek J, Kokot F, Matsuzawa Y. Decreased plasma adiponectin concentration in patients with essential hypertension. Am J Hypertens 2003;16:72–75.

66. Kazumi T, Kawaguchi A, Sakai K, Hirano T, Yoshino G. Young men with high-normal blood pressure have lower serum adiponectin, smaller LDL size, and higher elevated heart rate than those with optimal blood pressure. Diabetes Care 2002;25:971–976.

67. Huang KC, Chen CL, Chuang LM, Ho SR, Tai TY, Yang WS. Plasma adiponectin levels and blood pressures in nondiabetic adolescent females. J Clin Endocrinol Metab 2003;88:4130–4134.

68. Furuhashi M, Ura N, Higashiura K, et al. Blockade of the renin-angiotensin system increases adiponectin concentrations in patients with essential hypertension. Hypertension 2003;42:76–81.

69. Miyazaki T, Shimada K, Mokuno H, Daida H. Adipocyte derived plasma protein, adiponectin, is asso- ciated with smoking status in patients with coronary artery disease. Heart 2003;89:663.

70. Okamoto Y, Arita Y, Nishida M, et al. An adipocyte-derived plasma protein, adiponectin, adheres to injured vascular walls. Horm Metab Res 2000;32:47–50.

71. Zoccali C, Mallamaci F, Tripepi G, et al. Adiponectin, metabolic risk factors, and cardiovascular events among patients with end-stage renal disease. J Am Soc Nephrol 2002;13:134–141.

72. Kumada M, Kihara S, Sumitsuji S, et al., and the Osaka CAD Study Group. Association of hypoadiponectinemia with coronary artery disease in men. Arterioscler Thromb Vasc Biol 2003;23:

85–89.

73. Okamoto Y, Kihara S, Ouchi N, et al. Adiponectin reduces atherosclerosis in apolipoprotein E-deficient mice. Circulation 2002;106:2767–2770.

74. Matsuda M, Shimomura I, Sata M, et al. Role of adiponectin in preventing vascular stenosis. The missing link of adipo-vascular axis. J Biol Chem 2002;277:37487–91.

75. Krakoff J, Funahashi T, Stehouwer CD, et al. Inflammatory markers, adiponectin, and risk of type 2 diabetes in the Pima Indian. Diabetes Care 2003;26:1745–1751.

76. Ouchi N, Kihara S, Funahashi T, et al. Reciprocal association of C-reactive protein with adiponectin in blood stream and adipose tissue. Circulation 2003;107:671–674.

77. Engeli S, Feldpausch M, Gorzelniak K, et al. Association between adiponectin and mediators of inflam- mation in obese women. Diabetes 2003;52:942–947.

78. Fernandez-Real JM, Lopez-Bermejo A, Casamitjana R, Ricart W. Novel interactions of adiponectin with the endocrine system and inflammatory parameters. J Clin Endocrinol Metab 2003;88:2714–2718.

79. Ouchi N, Kihara S, Arita Y, et al. Novel modulator for endothelial adhesion molecules: adipocyte- derived plasma protein adiponectin. Circulation 1999;100:2473–2476.

80. Ouchi N, Kihara S, Arita Y, et al. Adiponectin, an adipocyte-derived plasma protein, inhibits endothelial NF-kappaB signaling through a cAMP-dependent pathway. Circulation 2000;102:1296–1301.

81. Ouchi N, Kihara S, Arita Y, et al. Adipocyte-derived plasma protein, adiponectin, suppresses lipid accumulation and class A scavenger receptor expression in human monocyte-derived macrophages.

Circulation 2001;103:1057–1063.

82. Yokota T, Oritani K, Takahashi I, et al. Adiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of mac- rophages. Blood 2000;96:1723–1732.

83. Arita Y, Kihara S, Ouchi N, et al. Adipocyte-derived plasma protein adiponectin acts as a platelet- derived growth factor-BB-binding protein and regulates growth factor-induced common postreceptor signal in vascular smooth muscle cell. Circulation 2002;105:2893–2898.

84. Shimabukuro M, Higa N, Asahi T, et al. Hypoadiponectinemia is closely linked to endothelial dysfunc- tion in man. J Clin Endocrinol Metab 2003;88:3236–3240.

Riferimenti

Documenti correlati

The aim of this work is to provide design guidelines to develop a sensorized catheter to guide the laser tool to the fenestration site and conduct preliminary testing of the

Per comprendere l’apporto di queste nuove genera- zioni, nonché le differenze territoriali che sono in qual- che modo collegate alle caratteristiche del capitale socia- le locale e

The quality of the policy learning offer that each regional government can grant at an international level depends on the existence of intelligent (i.e. evidence- based)

Raffone A, Travaglino A, Saccone G, Viggiani M, Giampaolino P, Insabato L, Mollo A, de Placido G, Zullo F (2019) PTEN expres- sion in endometrial hyperplasia and risk of cancer:

1) the viscous strength of the U.U.C. controls the strength of the upper plate and the efficiency of the propagation of L.U.C. material through it. A relatively weak upper

Twenty Second European Conference on Information Systems, Tel Aviv 2014 6 relationships were shaped more by the mechanisms of the market approach, though some

Data were extracted from Bankscope, which is a database containing comprehensive information on financial companies (banks and insurance companies) in Italy. We used the

The first results of the chapter completely characterize the time optimal synthesis in the case α > π/4 and determine the optimal trajectories connecting the north pole to the