resisting resistin; it's good for the heart

3
Editorial Resisting resistin; it's good for the heart 1. Introduction During the last two decades, the prevalence of obesity has reached epidemic proportions worldwide, leading to the development of a multitude of comorbidities, including hypertension, diabetes mellitus and cardiovascular diseases [1,2]. Adipose tissue produces and secretes a wide array of adipokines, which have key roles in situ (within the adipose tissue), and as circulating bioactive factors, suggesting that adipose tissue is an essential endocrine organ, in addition to an energy storage depot that modulates fat mass and nutrient homeostasis [3,4]. Numerous hormones, growth factors and cytokines belong to the adipokine group, including tumor necrosis factor-α (TNF-α), plasminogen activator inhibitor type 1 (PAI-1), leptin, adiponectin and the recently identied resistin [5]. Secreted adipokines signal to organs of metabolic importance including the liver, the skeletal and cardiac muscles and the immune system inuencing, directly or indirectly, fuel storage and mobilization, as well as energy homeostasis, by regulating glucose, lipid and protein metabolism, cellular inammation and atherosclerosis [3]. 2. Resistin is an essential link between obesity, diabetes and inammation Resistin, the product of the RSTN gene, was discovered in 2001 by the group of Mitchell Lazar as a target gene of the anti-diabetic drug thiozolidinedione (TZD), which was down-regulated in mouse adipocytes upon treatment [6]. It was baptizedresistin because of the acquired insulin resistance that mice injected with resistin demonstrated [68]. Resistin is a ~ 12.5 kDa peptide hormone that belongs to the Resistin Like Molecules (RELM) family (also known as Adipose Tissue Specic Secretory Factor, ADSF, or Found in Inam- matory Zone, FIZZ, family) of cystein-rich secreted proteins [9]. Accordingly, the RELM/ADSF/FIZZ class of proteins is characterized by the presence of a unique pattern of 11 cysteine residues and the fact that the proteins naturally form multimeric complexes. Human and rodent resistins consist of 108 and 114 amino acids, as pre-peptides, however, their hydrophobic signal sequences are cleaved before secretion, yielding active peptides of 92 and 94 amino acids, respectively [1012]. Resistin circulates in blood as a dimeric protein consisting of two active (cleaved) polypeptides, which are disulphide linked [13]. In rodents, resistin is derived almost exclusively from fat cells [6,12,14], whereas in humans resistin is produced by inamma- tory cells, primarily macrophages [15]. Several groups have studied the roles of resistin in the pathogenesis of obesity and diabetes. Early on, Steppan and colleagues demonstrated that administration of recombinant resistin in normal mice impaired glucose tolerance, and prevented adipogenesis in murine 3T3-L1 cells, whereas blocking of resistin with inhibitory antibodies improved insulin action in diet-induced obese mice [6,8,9]. In concert with these observations, overexpression of resistin in healthy mice led to insulin resistance, causing dyslipidemia [1618], while mice decient in resistin were protected from obesity, due to reduced hepatic glucose production [19]. Moreover, the levels of resistin were found signicantly elevated in the blood plasma of genetic animal models of diabetes (db/db) and obesity (ob/ob) as well as diet-induced models of diabetic obesity [6,8,14,20]. Collectively, these lines of evidence suggested that resistin might be an important link between obesity, glucose intolerance and insulin resistance, and thus contribute to the development of diabetes. The pathophysiological roles of resistin have been investigated in humans, too, although there has been considerable controversy. Some studies reported increased levels of resistin in the plasma of patients that had suffered myocardial infarction as well as in obese and type 2 diabetic patients, compared to lean healthy control subjects [21,22]. Consistent with this, TZD treatment resulted in decreased levels of plasma resistin in type 2 diabetic patients [23,24]. Others, however, failed to show any differences in resistin expression between normal and diabetic samples or an association between the levels of resistin and TZD treatment [2528]. While the effects of resistin in glucose metabolism are still unclear, an alternate hypothesis postulated that in humans resistin might play a role in inammatory processes, given that it is primarily derived from macrophages. In agreement with this, increased plasma resistin levels were associated with the develop- ment and extent of severity of inammatory and metabolic diseases, including sepsis and septic shock, inammatory bowel syndrome, coronary artery disease and rheumatoid arthritis [2932]. Given the direct or indirect association of resistin with these diverse patholog- ical conditions, it has been suggested that resistin might represent an essential link between insulin resistance, metabolic syndromes, inammation and atherosclerosis [3]. 3. The effects of resistin in the cardiovascular systemobservations from animal models and cultures of isolated cells Recently, several groups have directed their efforts towards the elucidation of the roles of resistin in the development and potential exacerbation of cardiovascular diseases. Rothwell and colleagues were the rst to report that pre-conditioning of an isolated rat heart preparation with human recombinant resistin, prior to ischemia, signicantly impaired reperfusion recovery and contractile activity. This effect was mediated by an NFκB stimulated cascade, which promoted the release of the pro-inammatory cytokine TNFα, and the secretion of hypertrophic cardiac markers [33]. In line with this model, Wang and colleagues demonstrated that resistin expression was markedly up-regulated in a cardiac volume overload murine Journal of Molecular and Cellular Cardiology 51 (2011) 141143 0022-2828/$ see front matter © 2011 Elsevier Ltd. All rights reserved. doi:10.1016/j.yjmcc.2011.05.011 Contents lists available at ScienceDirect Journal of Molecular and Cellular Cardiology journal homepage: www.elsevier.com/locate/yjmcc

Upload: aikaterini-kontrogianni-konstantopoulos

Post on 31-Oct-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Resisting resistin; it's good for the heart

Journal of Molecular and Cellular Cardiology 51 (2011) 141–143

Contents lists available at ScienceDirect

Journal of Molecular and Cellular Cardiology

j ourna l homepage: www.e lsev ie r.com/ locate /y jmcc

Editorial

Resisting resistin; it's good for the heart

1. Introduction

During the last two decades, the prevalence of obesity has reachedepidemic proportions worldwide, leading to the development of amultitude of comorbidities, including hypertension, diabetes mellitusand cardiovascular diseases [1,2]. Adipose tissue produces andsecretes a wide array of adipokines, which have key roles in situ(within the adipose tissue), and as circulating bioactive factors,suggesting that adipose tissue is an essential endocrine organ, inaddition to an energy storage depot that modulates fat mass andnutrient homeostasis [3,4]. Numerous hormones, growth factors andcytokines belong to the adipokine group, including tumor necrosisfactor-α (TNF-α), plasminogen activator inhibitor type 1 (PAI-1),leptin, adiponectin and the recently identified resistin [5]. Secretedadipokines signal to organs of metabolic importance including theliver, the skeletal and cardiac muscles and the immune systeminfluencing, directly or indirectly, fuel storage and mobilization, aswell as energy homeostasis, by regulating glucose, lipid and proteinmetabolism, cellular inflammation and atherosclerosis [3].

2. Resistin is an essential link between obesity, diabetesand inflammation

Resistin, the product of the RSTN gene, was discovered in 2001 bythe group of Mitchell Lazar as a target gene of the anti-diabetic drugthiozolidinedione (TZD), which was down-regulated in mouseadipocytes upon treatment [6]. It was “baptized” resistin because ofthe acquired insulin resistance that mice injected with resistindemonstrated [6–8]. Resistin is a ~12.5 kDa peptide hormone thatbelongs to the Resistin Like Molecules (RELM) family (also known asAdipose Tissue Specific Secretory Factor, ADSF, or Found in Inflam-matory Zone, FIZZ, family) of cystein-rich secreted proteins [9].Accordingly, the RELM/ADSF/FIZZ class of proteins is characterized bythe presence of a unique pattern of 11 cysteine residues and the factthat the proteins naturally form multimeric complexes. Human androdent resistins consist of 108 and 114 amino acids, as pre-peptides,however, their hydrophobic signal sequences are cleaved beforesecretion, yielding active peptides of 92 and 94 amino acids,respectively [10–12]. Resistin circulates in blood as a dimeric proteinconsisting of two active (cleaved) polypeptides, which are disulphidelinked [13]. In rodents, resistin is derived almost exclusively from fatcells [6,12,14], whereas in humans resistin is produced by inflamma-tory cells, primarily macrophages [15].

Several groups have studied the roles of resistin in the pathogenesisof obesity and diabetes. Early on, Steppan and colleagues demonstratedthat administration of recombinant resistin in normal mice impairedglucose tolerance, and prevented adipogenesis in murine 3T3-L1 cells,

0022-2828/$ – see front matter © 2011 Elsevier Ltd. All rights reserved.doi:10.1016/j.yjmcc.2011.05.011

whereasblockingof resistinwith inhibitory antibodies improved insulinaction in diet-induced obese mice [6,8,9]. In concert with theseobservations, overexpression of resistin in healthy mice led to insulinresistance, causingdyslipidemia [16–18],whilemicedeficient in resistinwere protected fromobesity, due to reduced hepatic glucose production[19].Moreover, the levels of resistinwere found significantly elevated inthe blood plasma of genetic animal models of diabetes (db/db) andobesity (ob/ob) as well as diet-induced models of diabetic obesity[6,8,14,20]. Collectively, these lines of evidence suggested that resistinmight be an important link between obesity, glucose intolerance andinsulin resistance, and thus contribute to the development of diabetes.

The pathophysiological roles of resistin have been investigated inhumans, too, although there has been considerable controversy. Somestudies reported increased levels of resistin in the plasma of patientsthat had suffered myocardial infarction as well as in obese and type 2diabetic patients, compared to lean healthy control subjects [21,22].Consistent with this, TZD treatment resulted in decreased levels ofplasma resistin in type 2 diabetic patients [23,24]. Others, however,failed to show any differences in resistin expression between normaland diabetic samples or an association between the levels of resistinand TZD treatment [25–28]. While the effects of resistin in glucosemetabolism are still unclear, an alternate hypothesis postulated thatin humans resistin might play a role in inflammatory processes, giventhat it is primarily derived frommacrophages. In agreement with this,increased plasma resistin levels were associated with the develop-ment and extent of severity of inflammatory and metabolic diseases,including sepsis and septic shock, inflammatory bowel syndrome,coronary artery disease and rheumatoid arthritis [29–32]. Given thedirect or indirect association of resistin with these diverse patholog-ical conditions, it has been suggested that resistin might represent anessential link between insulin resistance, metabolic syndromes,inflammation and atherosclerosis [3].

3. The effects of resistin in the cardiovascular system—observationsfrom animal models and cultures of isolated cells

Recently, several groups have directed their efforts towards theelucidation of the roles of resistin in the development and potentialexacerbation of cardiovascular diseases. Rothwell and colleagueswere the first to report that pre-conditioning of an isolated rat heartpreparation with human recombinant resistin, prior to ischemia,significantly impaired reperfusion recovery and contractile activity.This effect was mediated by an NFκB stimulated cascade, whichpromoted the release of the pro-inflammatory cytokine TNFα, and thesecretion of hypertrophic cardiac markers [33]. In line with thismodel, Wang and colleagues demonstrated that resistin expressionwas markedly up-regulated in a cardiac volume overload murine

Page 2: Resisting resistin; it's good for the heart

142 Editorial

model subjected to aorta-caval shunt [34]. In contrast, however, Gaoand colleagues reported that pre-treatment of mouse hearts withresistin hada cardioprotective effect against either ischemia/reperfusioninjury or left anterior descending (LAD) coronary artery ligation, bydramatically reducing cardiocyte apoptosis and infarct size, throughactivation of PI3K/Akt/PKCε/KATP-dependent pathways [35].

Conversely, neonatal rat ventricular myocytes (NRVM) subjected tocyclic stretch exhibited significantly increased expression of resistin atboth the mRNA and protein levels, mediated by TNFα, ERK kinase andNFκB-regulated pathways, while treatment of NRVMwith recombinantor secreted resistin impairedglucose transport [34]. Consistentwith this,adenoviral overexpression of resistin in cultures of NRVM resulted inenhanced protein synthesis and cell size, induced the expression of fetalgenes and activated known hypertrophic signal responses [36].Moreover, adenoviral overexpression of resistin in cultures of adult ratventricularmyocytes (ARVM)depressed contractility by impairingCa2+

homeostasis and altering myofilament activation [36].Although the aforementioned studies provided important insights

about the effects of acute manipulation of the expression of resistin inthe myocardium, it was only recently that Chemaly and colleaguesexamined the consequences of long-term resistin overexpression inthe cardiovascular system in vivo[37]. To achieve long-term expres-sion of resistin in the heart, Chemaly et al used adeno associated virus(AAV) serotype 9, which was introduced to rats through tail veininjection. AAV mediated gene transfer is a promising new method forgene therapy, as AAV exhibits high infection capability of non-dividing and dividing cells, lacks pathogenicity and stably integratesinto the host cell's genome at a specific site [38,39]. Importantly, selectserotypes, with AAV9 being the most potent, have been shown totransduce cardiac (and skeletal) muscles with high efficiency [40–42].Consistent with this, AAV9 has been used to deliver cytoskeletal,membrane and signaling proteins, and more recently to down-regulate Ca2+ cycling proteins in animal models of heart failure,resulting in considerable improvement of cardiac function [43,44].

Ten weeks following overexpression of resistin, animals displayedincreased ratio of left ventricular (LV) weight/body weight andincreased end systolic LV volume, indicative of developing cardiachypertrophy. These changes were accompanied by dramatic increasesin the expression levels of at least twomolecular markers of stress andhypertrophy, atrial natriuretic factor (ANF) and β-myosin heavy chain(β-MHC). In vivo hemodynamic analysis of the treated animalsdemonstrated impaired contractile activity, mediated at least in partby increased levels of phospholamban (PLN), the sarco endoplasmicreticulum Ca2+ ATPase 2a (SERCA2a) inhibitor. Histological evalua-tion further revealed that the resistin overexpressing myocardiumappeared largely fibrotic, displaying elevated expression of essentialprofibrotic targets, including collagen 1 and 3. The fibrotic myocar-dium exhibited increased levels of apoptosis, too. Thus, the amountsof the anti-apoptotic Bcl-2 gene were considerably diminished,whereas the levels of the pro-apoptotic Bax and death caspase-3were notably increased, indicating a clear shift towards the release ofpro-apoptotic molecules, promoting cell death. Most notably, resistinoverexpressing hearts displayed higher levels of intracellular reactiveoxygen species (ROS). This increase was attributed to enhancedexpression and activation of NADPH oxidase, a superoxide-producingenzyme. A number of inflammatory cytokines, including TNFα and itscognate receptor TNFα receptor 1, were also upregulated, leading tothe activation of downstream signaling cascades, including NFκBactivation, which is a critical event in cytokine-induced inflammation.Taken together, these findings strongly suggested that long-termexpression of resistin in vivo significantly impaired LV structure andcontractility by eliciting apoptotic and inflammatory responses.

Elevated levels of circulating resistin (hyperresistinemia) havebeen correlated with myocardial dysfunction in humans and inrodents [3]. The study by Chemaly and colleagues is the first oneto investigate the effects of long-term resistin expression in the

cardiovascular system in vivo[37]. Cardiac remodeling associated withan overt hypertrophic phenotype, abnormal Ca2+ handling, wide-spread fibrosis, increased apoptosis and prominent inflammationwere the manifestations of resistin's overexpression, faithfullyrecapitulating the major features of diabetic cardiomyopathy. Conse-quently, Chemaly and colleagues proposed that resistin might be “amolecular determinant of diabetic cardiomyopathy” [37]. Undoubt-edly, their findings are convincing and exciting, as they point to anovel therapeutic target with manifold and interlinked activities invarious disease conditions. Since diabetic cardiomyopathy is amultifactorial disease, a number of additional parameters need to beconsidered, however. Does the hypertrophic phenotype developed bythe treated animals progressively worsen (i.e. beyond the 10-weektime point)? Does age or gender affect the onset and severity ofmyocardial remodeling? Do nutritional and exercise regimes factor inthe observed cardiac dysfunction? Conversely, does long-term down-regulation of resistin improve the cardiac pathology of diabetic andobese animals?What major signaling cascades does resistin regulate?More importantly, does human and rodent resistin share the samepathophysiological properties and activities? How safe is it totranslate knowledge gained from in vitro culture settings and animalmodels to human subjects, especially given the relatively lowsequence homology between rodent and human resistin (~59%),and the fact that they are produced by different tissue sources [45]?Notwithstanding the numerous questions that still need to beaddressed regarding the biology of resistin, using a series of eleganttechnologies Chemaly and colleagues were able to show that long-term expression of resistin in rat hearts induced cardiac remodelingand dysfunction.

Sources of funding

The author is supported by grants from the National Institutes ofHealth (R01 AR52768 and R21HL106197), the American HeartAssociation (GRNT3780035) and a Pilot Grant from Johns HopkinsPhysical Sciences-Oncology Center (U54CA143868), The Johns HopkinsInstitute for NanoBioTechnology.

Disclosures

None.

References

[1] Lavie CJ, Milani RV, Ventura HO. Obesity and cardiovascular disease: risk factor,paradox, and impact of weight loss. J Am Coll Cardiol May 26 2009;53(21):1925–32.

[2] James PT. Obesity: the worldwide epidemic. Clin Dermatol Jul-Aug 2004;22(4):276–80.

[3] Rabe K, Lehrke M, Parhofer KG, Broedl UC. Adipokines and insulin resistance. MolMed Nov-Dec 2008;14(11–12):741–51.

[4] Rajala MW, Scherer PE. Minireview: the adipocyte—at the crossroads of energyhomeostasis, inflammation, and atherosclerosis. Endocrinology Sep 2003;144(9):3765–73.

[5] Steppan CM, LazarMA. The current biology of resistin. J InternMedApr 2004;255(4):439–47.

[6] Steppan CM, Bailey ST, Bhat S, Brown EJ, Banerjee RR, Wright CM, et al. Thehormone resistin links obesity to diabetes. Nature Jan 18 2001;409(6818):307–12.

[7] Graveleau C, Zaha VG, Mohajer A, Banerjee RR, Dudley-Rucker N, Steppan CM,et al. Mouse and human resistins impair glucose transport in primary mousecardiomyocytes, and oligomerization is required for this biological action. J BiolChem Sep 9 2005;280(36):31679–85.

[8] Steppan CM, Lazar MA. Resistin and obesity-associated insulin resistance. TrendsEndocrinol Metab Jan-Feb 2002;13(1):18–23.

[9] Steppan CM, Brown EJ, Wright CM, Bhat S, Banerjee RR, Dai CY, et al. A family oftissue-specific resistin-like molecules. Proc Natl Acad Sci U S A Jan 16 2001;98(2):502–6.

[10] Adeghate E. An update on the biology and physiology of resistin. Cell Mol Life SciOct 2004;61(19–20):2485–96.

[11] Holcomb IN, Kabakoff RC, Chan B, Baker TW, Gurney A, Henzel W, et al. FIZZ1, anovel cysteine-rich secreted protein associated with pulmonary inflammation,defines a new gene family. EMBO J Aug 1 2000;19(15):4046–55.

Page 3: Resisting resistin; it's good for the heart

143Editorial

[12] Kim KH, Lee K, Moon YS, Sul HS. A cysteine-rich adipose tissue-specific secretoryfactor inhibits adipocyte differentiation. J Biol Chem Apr 6 2001;276(14):11252–6.

[13] Aruna B, Ghosh S, Singh AK, Mande SC, Srinivas V, Chauhan R, et al. Humanrecombinant resistin protein displays a tendency to aggregate by formingintermolecular disulfide linkages. Biochemistry Sep 16 2003;42(36):10554–9.

[14] Rajala MW, Qi Y, Patel HR, Takahashi N, Banerjee R, Pajvani UB, et al. Regulation ofresistin expression and circulating levels in obesity, diabetes, and fasting. DiabetesJul 2004;53(7):1671–9.

[15] Patel L, Buckels AC, Kinghorn IJ, Murdock PR, Holbrook JD, Plumpton C, et al.Resistin is expressed in human macrophages and directly regulated by PPARgamma activators. Biochem Biophys Res Commun Jan 10 2003;300(2):472–6.

[16] Kitagawa Y, Bujo H, Takahashi K, Shibasaki M, Ishikawa K, Yagui K, et al. Impairedglucose tolerance is accompanied by decreased insulin sensitivity in tissues of miceimplanted with cells that overexpress resistin. Diabetologia Oct 2004;47(10):1847–53.

[17] Sato N, Kobayashi K, Inoguchi T, Sonoda N, Imamura M, Sekiguchi N, et al.Adenovirus-mediated high expression of resistin causes dyslipidemia in mice.Endocrinology Jan 2005;146(1):273–9.

[18] Satoh H, Nguyen MT, Miles PD, Imamura T, Usui I, Olefsky JM. Adenovirus-mediated chronic “hyper-resistinemia” leads to in vivo insulin resistance innormal rats. J Clin Invest Jul 2004;114(2):224–31.

[19] Banerjee RR, Rangwala SM, Shapiro JS, Rich AS, Rhoades B, Qi Y, et al. Regulation offasted blood glucose by resistin. Science Feb 20 2004;303(5661):1195–8.

[20] Rajala MW, Lin Y, Ranalletta M, Yang XM, Qian H, Gingerich R, et al. Cell type-specific expression and coregulation of murine resistin and resistin-like molecule-alpha in adipose tissue. Mol Endocrinol Aug 2002;16(8):1920–30.

[21] Burnett MS, Devaney JM, Adenika RJ, Lindsay R, Howard BV. Cross-sectionalassociations of resistin, coronary heart disease, and insulin resistance. J ClinEndocrinol Metab Jan 2006;91(1):64–8.

[22] McTernan PG, Fisher FM, Valsamakis G, Chetty R, Harte A, McTernan CL, et al.Resistin and type 2 diabetes: regulation of resistin expression by insulin androsiglitazone and the effects of recombinant resistin on lipid and glucosemetabolism in human differentiated adipocytes. J Clin Endocrinol Metab Dec2003;88(12):6098–106.

[23] Bajaj M, Suraamornkul S, Hardies LJ, Pratipanawatr T, DeFronzo RA. Plasmaresistin concentration, hepatic fat content, and hepatic and peripheral insulinresistance in pioglitazone-treated type II diabetic patients. Int J Obes Relat MetabDisord Jun 2004;28(6):783–9.

[24] Heilbronn LK, Rood J, Janderova L, Albu JB, Kelley DE, Ravussin E, et al. Relationshipbetween serum resistin concentrations and insulin resistance in nonobese, obese,and obese diabetic subjects. J Clin Endocrinol Metab Apr 2004;89(4):1844–8.

[25] Iqbal N, Seshadri P, Stern L, Loh J, Kundu S, Jafar T, et al. Serum resistin is notassociated with obesity or insulin resistance in humans. Eur Rev Med PharmacolSci May-Jun 2005;9(3):161–5.

[26] Lee JH, Chan JL, Yiannakouris N, Kontogianni M, Estrada E, Seip R, et al. Circulatingresistin levels are not associated with obesity or insulin resistance in humans andare not regulated by fasting or leptin administration: cross-sectional andinterventional studies in normal, insulin-resistant, and diabetic subjects. J ClinEndocrinol Metab Oct 2003;88(10):4848–56.

[27] Kielstein JT, Becker B, Graf S, Brabant G, Haller H, Fliser D. Increased resistin bloodlevels are not associated with insulin resistance in patients with renal disease. AmJ Kidney Dis Jul 2003;42(1):62–6.

[28] Beckers S, Peeters AV, Freitas F, Mertens IL, Hendrickx JJ, Van Gaal LF, et al. Analysisof genetic variations in the resistin gene shows no associations with obesity inwomen. Obesity (Silver Spring) Apr 2008;16(4):905–7.

[29] Bokarewa M, Nagaev I, Dahlberg L, Smith U, Tarkowski A. Resistin, an adipokinewith potent proinflammatory properties. J Immunol May 1 2005;174(9):5789–95.

[30] Reilly MP, Lehrke M, Wolfe ML, Rohatgi A, Lazar MA, Rader DJ. Resistin is aninflammatory marker of atherosclerosis in humans. Circulation Feb 22 2005;111(7):932–9.

[31] Konrad A, LehrkeM, Schachinger V, Seibold F, Stark R, Ochsenkuhn T, et al. Resistinis an inflammatory marker of inflammatory bowel disease in humans. Eur JGastroenterol Hepatol Dec 2007;19(12):1070–4.

[32] Sunden-Cullberg J, Nystrom T, Lee ML, Mullins GE, Tokics L, Andersson J, et al.Pronounced elevation of resistin correlates with severity of disease in severesepsis and septic shock. Crit Care Med Jun 2007;35(6):1536–42.

[33] Rothwell SE, Richards AM, Pemberton CJ. Resistin worsens cardiac ischaemia–reperfusion injury. Biochem Biophys Res Commun Oct 13 2006;349(1):400–7.

[34] Wang BW, Hung HF, Chang H, Kuan P, Shyu KG. Mechanical stretch enhances theexpression of resistin gene in cultured cardiomyocytes via tumor necrosis factor-alpha. Am J Physiol Heart Circ Physiol Oct 2007;293(4):H2305–12.

[35] Gao J, Chang Chua C, Chen Z, Wang H, Xu X, CH R, et al. Resistin, an adipocytokine,offers protection against acute myocardial infarction. J Mol Cell Cardiol Nov2007;43(5):601–9.

[36] Kim M, Oh JK, Sakata S, Liang I, Park W, Hajjar RJ, et al. Role of resistin in cardiaccontractility and hypertrophy. J Mol Cell Cardiol Aug 2008;45(2):270–80.

[37] Chemaly ER, Hadri L, Zhang S, Kim M, Kohlbrenner E, Sheng J, et al. Long-term invivo resistin overexpression induces myocardial dysfunction and remodeling inrats. J Mol Cell Cardiol Apr 2011;23.

[38] Muller OJ, Schinkel S, Kleinschmidt JA, Katus HA, Bekeredjian R. Augmentation ofAAV-mediated cardiac gene transfer after systemic administration in adult rats.Gene Ther Dec 2008;15(23):1558–65.

[39] Hajjar RJ, Zsebo K. AAV vectors and cardiovascular disease: targeting TNF receptorin the heart: clue to way forward with AAV? Gene Ther Dec 2007;14(23):1611–2.

[40] Fechner H, Sipo I, Westermann D, Pinkert S, Wang X, Suckau L, et al. Cardiac-targeted RNA interference mediated by an AAV9 vector improves cardiac functionin coxsackievirus B3 cardiomyopathy. J Mol Med Sep 2008;86(9):987–97.

[41] Goehringer C, Rutschow D, Bauer R, Schinkel S, Weichenhan D, Bekeredjian R, et al.Prevention of cardiomyopathy in delta-sarcoglycan knockout mice after systemictransfer of targeted adeno-associated viral vectors. Cardiovasc Res Jun 1 2009;82(3):404–10.

[42] Pacak CA, Mah CS, Thattaliyath BD, Conlon TJ, Lewis MA, Cloutier DE, et al.Recombinant adeno-associated virus serotype 9 leads to preferential cardiactransduction in vivo. Circ Res Aug 18 2006;99(4):e3–9.

[43] Lipskaia L, ChemalyER, Hadri L, LompreAM,Hajjar RJ. Sarcoplasmic reticulumCa(2+)ATPase as a therapeutic target for heart failure. Expert Opin Biol Ther Jan 2010;10(1):29–41.

[44] Periasamy M, Rafael-Fortney JA. Somatic gene therapy to treat heart failure is onestep closer to reality. Cardiovasc Res Jun 1 2009;82(3):383–4.

[45] Ghosh S, Singh AK, Aruna B, Mukhopadhyay S, Ehtesham NZ. The genomicorganization of mouse resistin reveals major differences from the human resistin:functional implications. Gene Feb 13 2003;305(1):27–34.

Aikaterini Kontrogianni-KonstantopoulosUniversity of Maryland, School of Medicine,

Department of Biochemistry and Molecular Biology,Baltimore, MD 21201, USA

Tel.: +1 410 706 5788; fax: +1 410 706 8297.E-mail address: [email protected].

11 May 2011