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2007年第27卷第1期

Leptin Regulates Neointima Formation After Arterial Injury Through Mechanisms Independent of Blood Pressure and the Leptin Receptor/STAT3 Signaling Pathways I

来源:《动脉硬化血栓血管生物学杂志》
摘要:【摘要】Background-Leptinisanadipocyte-derivedhormonecriticalforenergyhomeostasisandimplicatedinvasculardiseaseprocesses。Therelevantcellularleptinreceptorpoolsandsignalingpathwaysinvolvedinleptin-relatedvascularphenotypesinvivoareunclear。MethodsandResult......

点击显示 收起

【摘要】  Background- Leptin is an adipocyte-derived hormone critical for energy homeostasis and implicated in vascular disease processes. The relevant cellular leptin receptor pools and signaling pathways involved in leptin-related vascular phenotypes in vivo are unclear.

Methods and Results- Arterial injury was induced in wild-type ( wt ), leptin-deficient ( lep ob/ob ), and leptin receptor-deficient ( lepr db/db ) mice. Compared with wt mice, lep ob/ob and lepr db/db mice were protected from the development of neointima. Bone marrow transplantation experiments between wt and lepr db/db mice indicated that the vascular protection in lepr db/db mice was not attributable to lack of leptin receptor expression on bone marrow-derived elements. To investigate the role of the lepr- mediated signal transducer and activator of transcription 3 (STAT3) signaling pathway in the response to vascular injury, lepr s/s mice homozygous for a leptin receptor defective in STAT3 signaling underwent femoral arterial injury. Despite similar obesity and blood pressure levels, the neointimal area in lepr s/s mice was significantly increased compared with lepr db/db mice.

Conclusions- The molecular mechanism by which the leptin receptor mediates neointima formation and vascular smooth muscle cell proliferation is largely independent of the STAT3-dependent signaling pathways involved in energy balance.

Elevated levels of leptin have been associated with increased risk for cardiovascular disease. We demonstrate that mice deficient in leptin or leptin receptor are protected from neointimal formation. This effect is mediated by a non-bone marrow-derived leptin receptor pool and by signaling pathways independent of those responsible for energy balance.

【关键词】  atherosclerosis obesity remodeling restenosis


Introduction


Leptin is an adipocyte-derived hormone initially described for its profound effects on energy balance, 1 which are largely mediated via leptin receptors expressed on cells in the hypothalamus. 2 Leptin receptors have also been shown to be expressed on other cell types that may affect vascular disease processes including white blood cells, 3 platelets, 4 endothelial cells, 5 and vascular smooth muscle cells (VSMCs). 6 These receptor pools are likely to be biologically relevant as leptin has been demonstrated to promote angiogenesis, 7 thrombosis, 8-10 atherosclerosis, 9 and neointimal hyperplasia 11,12 in animal models. However, the relevant leptin receptor pools responsible for these vascular effects are unclear.


The interaction of leptin at the leptin receptor results in the phosphorylation of critical tyrosine residues 13 and subsequent activation of additional intracellular signaling. Several pathways have been implicated in post-leptin receptor signaling events toward appetite and metabolism including PI3 kinase, 14 AMP kinase, 15 and signal transducer and activator of transcription 3 (STAT3). 16 Tyr1138 of the leptin receptor mediates activation of STAT3. Substitution of Tyr1138 with serine blocks STAT3 activation in cultured cells and in mice in which the leptin receptor is replaced by this mutant ( lepr s/s ). 16 The obesity of these lepr s/s mice closely resembles that of leptin receptor-deficient ( lepr db/db ) mice. 16 However, the role of this pathway in mediating leptin receptor responses on vascular phenotypes in vivo is unknown.


This report examines the effects of leptin and leptin deficiency on neointimal hyperplasia after vascular injury. We demonstrate that the in vivo effect of leptin is not mediated by bone marrow-derived elements, is at least partially independent of leptin receptor-mediated STAT3 signaling, and is associated with the proliferation of vascular smooth muscle cells.


Methods


Mice


Wild-type ( wt ), leptin-deficient ( lep ob/ob ), and leptin receptor-deficient ( lepr db/db ) mice were purchased from the Jackson Laboratory (Bar Harbor, Me). Targeted leptin receptor mutant mice with failure to activate STAT3 signaling pathways ( lepr s/s ) were generated as previously described. 16 Lepr s/s mice had been backcrossed to the C57BL/6J strain 7 generations. Male animals, between 8 and 11 weeks of age, were fed a standard laboratory rodent diet (#5001, TestDiet, Richmond, Ind) in specific pathogen-free facilities. All procedures complied with the Principles of Laboratory and Animal Care established by the National Society for Medical Research and were approved by the University of Michigan Committee on Use and Care of Animals.


Femoral Artery Injury


Mouse femoral artery injury was performed using a model of wire-induced vascular injury as described previously. 17 This model causes endothelial damage at the site of injury followed by reproducible intimal hyperplasia. Mice were anesthetized with intraperitoneal sodium pentobarbital (120 mg/kg; Butler Co), except for mice that received a BMT, which were anesthetized with inhaled isofluorane (Baxter Healthare Corporation) using a Veterinary Anesthesia and Monitoring device (SurgiVet Inc). Mice were then secured in the supine position and placed under a dissecting microscope (Nikon SMZ-2T, Mager Scientific Inc). After a midline incision was made on the ventral left thigh region, the femoral nerve was isolated and pulled aside. The distal and proximal ends of the femoral artery were held with surgical suture for temporary control of blood flow. A small arterial branch superior to the muscular branch of the femoral artery was cauterized (DELC Cauterizer, Aaron Medical Industries Inc). One drop of 1% lidocaine was used to prevent arterial spasm. An arteriotomy of the femoral artery muscular branch was then performed. A straight guide wire (0.38 mm diameter, No. C-SF-15-15, Cook) was introduced 1 cm into the femoral artery through this muscular branch and remained in the artery for 1 minute. After the wire was removed, the muscular branch was ligated with 7.0 silk suture. Blood flow through the femoral artery was restored by releasing the surgical thread. Surgical clips were used to close the incision site.


Leptin Replacement


Recombinant murine leptin (R&D Systems) was delivered to mice via intraperitoneal (IP) injection (5.0 µg/g body weight). Adenovirus expressing murine leptin on the RSV promoter (ad-leptin; gift of Dr Savio Woo, Mount Sinai, New York) 18 was delivered to mice via tail vein injection (1.25 x 10 11 pfu/mouse). A control empty adenoviral vector was delivered to lep ob/ob via the same conditions.


Bone Marrow Transplantation


At 10 weeks of age, wt and lepr db/db mice were irradiated and bone marrow transplantation (BMT) was performed as previously described. 8 We have previously shown that this protocol leads to complete engraftment. 8 Femoral artery injury was performed six weeks following transplantation.


Histological and Morphometric Analyses


At the end of the protocol, mice were injected with bromodeoxyuridine (BrdU; Zymed Laboratories Inc) (IP, 30 µg/g body weight) 12 and 2 hours before sacrifice. The mice were perfused with saline and fixed using formalin with a 25 gauge needle, inserted into the left ventricle, at a rate of 1 mL/min. The injured femoral artery was excised, embedded in paraffin blocks, sectioned (5um), and stained for elastin using the Accustain Elastic Stain kit (Sigma-Aldrich). For quantitation, 4 cross sections taken every 2 mm were selected from each artery. The images were analyzed using Image-Pro Plus software (Media Cybernetics). Intimal (I) and medial (M) areas were measured and the ratio of I/M was calculated. Immunohistochemistry was performed as described previously. 19 Smooth muscle cells were detected by smooth muscle -actin staining using an -actin monoclonal antibody (1:100, Cedarlane Laboratories Limited). Cellular proliferation was analyzed with BrdU Staining Kit (Zymed Laboratories Inc) and the number of BrdU-positive cells were counted manually and expressed as BrdU-positive nuclei within the external elastic lamina per cross section.


Body Weight, Metabolic Parameters, and Blood Pressure


Mouse body weight was recorded weekly after arterial injury. At the time of euthanasia, blood was obtained by ventricular puncture for plasma separation. Plasma concentration of cholesterol, triglycerides, glucose (Sigma-Aldrich), and insulin (Crystal Chemical Inc) were determined at the time of sacrifice. Plasma leptin levels were determined weekly after arterial injury using a murine leptin ELISA kit (Crystal Chemical Inc). Arterial blood pressure was determined in 8- to 10-week-old wt, lepr db/db, and lepr s/s mice using a radiotelemetry system. 20 Mice of each genotype were instrumented with radiotelemetric monitoring devices, which allowed continuous nonrestrained monitoring of blood pressure. A transmitter catheter (Data Sciences) was inserted into the left carotid artery when the mouse was under isoflurane anesthesia, and the blood pressure transmitter body (Data Science, PA-C20) was routed to the abdominal cavity. After recovery, the mouse was housed in an individual cage and provided with regular mouse chow and tap water ad libitum. Mean arterial pressure was recorded at 5-minute intervals for at least 2 weeks duration. After data collection, the 5 minute sampling periods were averaged over 5 consecutive days (once diurnal rhythm was established). The 5 minute averages were then furthered condensed into 24 separate 1 hour values for each hour during the entire light-dark cycle for the purpose of graphical presentation and statistical analysis.


Cell Culture


VSMC were isolated from 7 to 14 week-old wt, lepr db/db, and lepr s/s mice (n=3 each group) as previously described. 21 Cells were grown in Dulbecco modified Eagle medium (DMEM, Gibco Inc) containing 10% fetal bovine serum (FBS, Gibco Inc) and 1% penicillin/streptomycin, and passaged as described previously. 22 For experiments, cells between passages 4 and 9 were made quiescent by incubation with serum-free DMEM for 30 hours before use. The VSMC proliferation assay 6 and quantification of cell number by crystal violet staining and optical density reading 23 were performed as previously described. Briefly, cells were seeded in 96-well plate in DMEM containing 0.5% FBS at a density of 4 x 10 3 /well and allowed to attach overnight. Medium was then replaced with DMEM containing 2% FBS and Insulin-Transferrin-Selenium premix (Gibco Inc) in the presence of either leptin (100 ng/mL; R&D Systems) or vehicle. After 3 days of incubation, cells were fixed with 10% glutaraldehyde, stained with 0.1% crystal violet solution, and dissolved with 10% acetic acid. The wells were read at 590 nm in a microplate reader (VersaMax, Molecular Devices Corp). All assays were performed in triplicate wells and 2 to 3 replications of the experiment were performed.


Statistical Analysis


Values are expressed as mean±SEM. The statistical significance of differences between groups was determined by 1-way ANOVA followed by Dunn post-hoc analysis when more than 2 experimental groups were included. The student 2-tailed t test was performed when only 2 groups were being compared. For BMT and blood pressure measurements, a 2-way analysis of variance was performed followed by the Tukey HSD test to examine significant main effects. P <0.05 was considered significant.


Results


Effect of Leptin and Leptin Receptor on Neointimal Formation


To determine the influence of leptin and the leptin receptor on neointimal formation, femoral injury was induced in wt, lep ob/ob, and lepr db/db mice. Four weeks after injury, the average femoral artery neointimal area and ratio of intima/media in lep ob/ob and lepr db/db mice was significantly reduced compared with wt mice ( Figure 1 and Table 1 ). There were no differences in the medial area between wt, lep ob/ob, and lepr db/db. The neointimal area was comprised predominantly of VSMCs ( Figure 1 ). The VSMC cellular proliferation index was significantly reduced in lep ob/ob and lepr db/db compared with wt mice ( Table 1 ).


Figure 1. Effect of Leptin on Neointimal Formation. A, Histological analyses. a: mouse model of wire-induced vascular injury; b: neointimal SMC stained with anti -actin as red, wt; c: uninjured artery, wt; d: I, injured artery stained for elastin, d: wt; e: lep ob/ob; f: lepr db/db; g: lep ob/ob receiving recombinant murine leptin; h: lep ob/ob receiving ad-leptin; I, lepr db/db receiving ad-leptin. Scale bar=20um. Arrows point to internal elastic lamina. B, Neointima area measurement. Mice lacking leptin or the leptin receptor were protected from neointimal growth: wt, n=10; lep ob/ob, n=12; lepr db/db, n=15. Leptin treatment (recombinant leptin or ad-leptin) reversed this protection in lep ob/ob but not in lepr db/db : lep ob/ob receiving leptin, n=6; lep ob/ob receiving ad-leptin, n=5; lepr db/db receiving ad-leptin, n=4. * P <0.05, **<0.001, NS=not significant.


TABLE 1. Morphometric Analyses of Cross Sections


Effect of Exogenous Leptin on Neointimal Formation


To further examine whether the reduced neointimal formation in lep ob/ob mice was attributable to the lack of leptin, recombinant murine leptin was given to lep ob/ob mice beginning 1 day before injury on a daily basis until sacrifice. Leptin levels 12 hours after injection were 45.7±5.8 ng/mL compared with basal levels of 1.0±0.1 ng/mL in wt mice. The average neointima of lep ob/ob mice receiving recombinant leptin was significantly higher than lep ob/ob mice receiving no treatment ( Figure 1 ). These results confirm that leptin plays a causal role in neointimal growth.


To test another leptin treatment strategy, lep ob/ob and lepr db/db mice were infected with ad-leptin or control adenovirus vector. Because leptin may affect thrombosis after vascular injury, 8,10 which could in turn affect neointimal formation, ad-leptin was injected 1 day after injury to reduce potential effects on acute thrombosis. Leptin levels of lep ob/ob mice treated with ad-leptin reached a peak of 230.3±58.9 ng/mL at one week after ad-leptin injection and gradually declined to 84.0±33.2 ng/mL at 2 weeks, 8.8±1.6 ng/mL at 3 weeks and 2.1±0.6 ng/mL at 4 weeks. The neointima of lep ob/ob mice receiving ad-leptin was significantly higher compared with that of lep ob/ob mice receiving no treatment ( Figure 1 ). The neointima of lep ob/ob mice receiving a control adenovirus was not different from lep ob/ob mice receiving no treatment (data not shown). This data supports the effect of leptin on neointimal formation and also demonstrates that leptin is not required at the time of injury to promote neointimal formation. The neointima of lepr db/db mice receiving ad-leptin was also examined to confirm the effects of the leptin receptor on neointimal formation and to serve as a control for nonspecific inflammatory effects of the adenovirus. The neointima of lepr db/db mice treated with ad-leptin was not different from the lepr db/db mice receiving no treatment ( Figure 1 ).


Effect of Leptin Treatment on Body Weight and Metabolic Parameters


Leptin treatment improved many metabolic parameters in lep ob/ob mice ( Table 2 ) providing evidence of an improvement in the concentration of many classic risk factors. Despite these salient changes, there was a robust leptin-mediated enhancement of neointimal growth.


TABLE 2. Effect of Leptin on Body Weight and Metabolic Profile


Effect of Bone Marrow Transplantation on Neointima Formation


To examine whether the leptin receptor-deficient phenotype was attributable to the lack of leptin receptor on circulating blood elements, BMT was performed from lepr db/db to wt, wt to wt, wt to lepr db/db, and lepr db/db to lepr db/db mice. Control mice not receiving bone marrow after irradiation died 7 days later, demonstrating effective bone marrow ablation after the irradiation. Six weeks after BMT, femoral arterial injury was performed to induce intimal hyperplasia. The neointima of wt mice receiving bone marrow from lepr db/db mice was significantly greater than that of lepr db/db mice receiving bone marrow from lepr db/db mice ( Figure 2 ). Similarly, the neointima of lepr db/db mice receiving wt bone marrow was significantly less than the neointima observed in wt mice receiving bone marrow from wt mice ( Figure 2 ).


Figure 2. Effect of bone marrow transplantation on neointimal formation. A, Histological analyses. a: wt to wt; b: lepr db/db to wt; c: wt to lepr db/db; d: lepr db/db to l epr db/db. B, Neointima area measurement. BMT did not affect neointimal phenotype of the recipient. n=7 to 8 for each group. * P <0.05, NS=not significant.


Effect of LepR-Mediated STAT3 Signaling on Neointima Formation


To determine the effect of leptin receptor-STAT3 signaling on neointimal formation, lepr s/s mice (n=9), underwent femoral arterial injury. Despite development of an obese phenotype similar to that observed in lepr db/db mice ( Table 2 ), 16 lepr s/s mice developed greater intimal hyperplasia than lepr db/db mice ( P <0.02; Figure 3a through 3 c).


Figure 3. Effect of Lepr-STAT3 signaling on neointima. Despite a similar obese phenotype (A), the lepr s/s mice developed greater intimal hyperplasia than lepr db/db mice (B and C). * P <0.02.


Effect of LepR-Mediated STAT3 Signaling on Arterial Blood Pressure


To examine whether the neointimal formation of the lepr s/s was related to arterial blood pressure we measured the mean arterial pressure of wt, lepr db/db, and lepr s/s mice. Consistent with previous studies using radiotelemetry, we detected a reduced arterial pressure in lepr db/db compared with wild-type mice. 24 Similarly, the lepr s/s were observed to have reduced blood pressure compared with wild-type ( Figure 4 ). No difference in blood pressure was observed between lepr s/s and lepr db/db mice suggesting that the leptin-mediated effects on blood pressure are mediated by the Lepr-STAT3 pathway.


Figure 4. Comparison of mean arterial pressure between wt, lepr s/s, and lepr db/db mice. Note the diurnal variation of the mean arterial pressure during the light/dark cycle. Values represent mean of 4 to 5 mice for each genotype (wt: n=5; lepr db/db : n=4; lepr s/s : n=4). Two-way analysis of variance results in significant effects of time (diurnal variation) and genotype. Post-hoc analysis reveals that wild-type mice have significantly elevated mean arterial pressure compared with both lepr s/s and lepr db/db mice ( P <0.05).


Effect of Leptin on SMC Proliferation


To further elucidate relevant signaling pathways, VSMCs were isolated from wt, lepr s/s, and lepr db/db mice. Similar to previous studies, 6 leptin induced a significant increase in cellular proliferation in wt cells compared with vehicle (leptin=0.40±0.006 versus vehicle=0.37±0.005; P <0.03). As previously shown, 6 this leptin effect was completely blocked with the phosphoinositide 3 kinase inhibitor, wortmannin (leptin + wort=0.37±0.006; P <0.02 compared with leptin + vehicle). VSMCs from lepr s/s mice also responded to leptin with an increase in proliferation compared with vehicle control (leptin=0.67±0.01 versus vehicle=0.49±0.02, P <0.00001), and this effect was blocked with wortmannin (lep+wort=0.53±0.001, P <0.00003). VSMCs from lepr db/db mice did not respond to leptin treatment.


Discussion


Leptin is a 16-kDa peptide hormone with structural features similar to those of the long-chain helical cytokine family. 1 The expression of receptors for leptin are ubiquitous, 25 indicating that leptin may affect multiple physiological processes. To determine the importance of leptin and the leptin receptor on neointimal hyperplasia, we performed femoral wire injury on wt, lep ob/ob, and lepr db/db mice. Consistent with previous studies, lep ob/ob and lepr db/db mice were protected from neointimal formation. 11,12 Similar to the findings of Schafer et al, 11 who used an iron chloride model of carotid injury, we have shown that leptin treatment (recombinant murine leptin or ad-leptin) increases neointima in the leptin deficient mice but not in mice lacking the leptin receptor, confirming a causal role for leptin and the leptin receptor in neointimal formation. In addition, cell proliferation analyses revealed that lep ob/ob and lepr db/db mice had significantly less cellular proliferation in the femoral arterial wall after injury compared with wt mice. This suggests that leptin may augment VSMC proliferation after arterial injury. Previous studies have demonstrated leptin receptor expression by VSMCs after arterial injury, supporting a direct effect of leptin on vascular smooth muscle cells. 11,12


In addition to a potential role of the VSMCs, we sought to directly examine the importance of bone marrow-derived leptin receptor in leptin-mediated effects on neointimal hyperplasia. For these studies, we performed femoral wire injury after BMT from lepr db/db to wt and from wt to lepr db/db mice. Interestingly, the recipient phenotype was unchanged in both of these experiments compared with control groups receiving marrow from genetically identical donors. This suggests that the in vivo effect of the platelet and leukocyte leptin receptor is negligible toward the response to vascular injury.


Similar to other cytokine receptors, the leptin receptor signals by activating a Jak family tyrosine kinase (Jak2) that autophosphorylates numerous tyrosine residues and phosphorylates tyrosine residues (Tyr985 and Tyr1138) on the leptin receptor during leptin stimulation. Leptin can therefore signal through activation of several different receptor-mediated pathways. 13 The relevance of these pathways toward energy balance has been established by the analysis of mice in which the leptin receptor gene has been substituted by a mutant leptin receptor ( lepr S1138) containing a Tyr1138-Ser substitution that is defective in STAT3 binding. 16 These mice develop obesity similar to that observed in lepr db/db mice, although other phenotypes associated with leptin receptor deficiency are less affected indicating the relevance of other signaling pathways such as the IRS-protein-PI3 kinase pathway. 13 The pathways responsible for the vascular effects of leptin are unknown, although both STAT3 and PI3K lepr signaling pathways have been implicated in VSMC proliferation in vitro. 26,27 To dissect these pathways in vivo, lepr s/s mice with defective STAT3 signaling were studied in the femoral arterial injury model. These mice developed an obese phenotype similar to that observed in the lepr db/db group, as previously described, 16 but they exhibited significantly greater intimal hyperplasia after arterial injury, suggesting that leptin receptor-mediated STAT3 signaling is not responsible for the leptin effect on neointimal formation after injury.


The robust effects of exogenous leptin in these experiments could be partially mediated by effects on arterial blood pressure. Leptin has been shown to influence sympathetic activation with elevations in blood pressure. 24,28,29 Furthermore, this effect on sympathetic activity appears to be centrally mediated and may not be subject to the same degree of leptin resistance as observed with leptin-receptor related feeding behavior. 30 To examine whether the in vivo effects observed in the lepr s/s mice were mediated by elevations in blood pressure, we performed radiotelemetry arterial blood pressure recordings. Consistent with previous publications we observed reduced blood pressure in lepr db/db mice. 24 In addition, the blood pressure of the lepr s/s mice was also lower than wt and was not different than lepr db/db mice. Therefore, although high doses of exogenous leptin probably elevated blood pressure at some time points in our exogenous leptin experiments, we have demonstrated that elevations in blood pressure are not mediating the increased neointimal response of the lepr s/s mice as compared with the lepr db/db mice.


To further examine the potential underlying mechanism of the increased neointimal response of the lepr s/s compared with the lepr db/db, we used a cell culture system to examine VSMC proliferation. Leptin has previously been shown to stimulate proliferation of VSMCs in culture 6 and our in vivo studies demonstrated reduced intimal VSMC proliferation in leptin deficient and leptin receptor deficient mice. Therefore, we examined whether the lepr db/db and lepr s/s mice responded differently to recombinant leptin in vitro. VSMCs isolated and cultured from lepr db/db did not respond to leptin treatment whereas leptin significantly stimulated the proliferation of VSMCs isolated from lepr s/s mice, consistent with the in vivo phenotype. Furthermore, this leptin effect on lepr s/s VSMC?s was completely blocked after incubation of the VSMCs with a PI3 kinase inhibitor, implicating this pathway in leptin receptor-mediated VSMC proliferation.


Limitations of this study include the supraphysiologic plasma concentrations of leptin obtained in the leptin replacement experiments. These experiments serve as proof of principle that leptin affects neointimal hyperplasia. The increased neointimal hyperplasia observed in the lepr s/s mice compared with the lepr db/db mice, where no exogenous leptin was added, demonstrates the potential of physiological elevations of leptin, as observed in obese states, to affect this vascular end point. An additional limitation of this study is lack of definitive proof of the relevant leptin receptor cellular pool. Although we have demonstrated that leptin effects on neointimal hyperplasia are at least partially independent of Lepr-STAT3 signaling, not dependent on blood pressure elevation, and independent of bone marrow-derived elements, it remains possible that other Lepr-mediated central nervous system pathways could influence vascular disease.


In conclusion, these data suggest that leptin enhances neointimal formation through an interaction with the leptin receptor resulting in smooth muscle cell proliferation. These effects are mediated by a leptin receptor signaling pathway that is distinct from the major pathway involved in energy balance. These findings suggest that it may be possible to design therapeutic agents capable of promoting the beneficial metabolic activities of leptin while avoiding the potential hazard of vascular complications.


Acknowledgments


Sources of Funding


This work was supported by grants HL57346 and AHA EIG (to D.T.E.), American Diabetes Association Junior Faculty Award (to P.F.B.), and DK56731 and DK57768 (to M.G.M.).


Disclosures


None.

【参考文献】
  Zhang Y, Proenca R, Maffei M, Barone M, Leopold L, Friedman JM. Positional cloning of the mouse obese gene and its human homologue. Nature. 1994; 372: 425-432.

Chua SC, Jr., Chung WK, Wu-Peng XS, Zhang Y, Liu SM, Tartaglia L, Leibel RL. Phenotypes of mouse diabetes and rat fatty due to mutations in the OB (leptin) receptor. Science. 1996; 271: 994-996.

Zarkesh-Esfahani H, Pockley G, Metcalfe RA, Bidlingmaier M, Wu Z, Ajami A, Weetman AP, Strasburger CJ, Ross RJ. High-dose leptin activates human leukocytes via receptor expression on monocytes. J Immunol. 2001; 167: 4593-4599.

Nakata M, Yada T, Soejima N, Maruyama I. Leptin promotes aggregation of human platelets via the long form of its receptor. Diabetes. 1999; 48: 426-429.

Bouloumie A, Drexler HC, Lafontan M, Busse R. Leptin, the product of Ob gene, promotes angiogenesis. Circ Res. 1998; 83: 1059-1066.

Oda A, Taniguchi T, Yokoyama M. Leptin stimulates rat aortic smooth muscle cell proliferation and migration. Kobe J Med Sci. 2001; 47: 141-150.

Ribatti D, Nico B, Belloni AS, Vacca A, Roncali L, Nussdorfer GG. Angiogenic activity of leptin in the chick embryo chorioallantoic membrane is in part mediated by endogenous fibroblast growth factor-2. Int J Mol Med. 2001; 8: 265-268.

Bodary PF, Westrick RJ, Wickenheiser KJ, Shen Y, Eitzman DT. Effect of leptin on arterial thrombosis following vascular injury in mice. J Am Med Assoc. 2002; 287: 1706-1709.

Bodary PF, Gu S, Shen Y, Hasty AH, Buckler JM, Eitzman DT. Recombinant leptin promotes atherosclerosis and thrombosis in apolipoprotein E-deficient mice. Arterioscler Thromb Vasc Biol. 2005; 25: 1634.

Konstantinides S, Schafer K, Koschnick S, Loskutoff DJ. Leptin-dependent platelet aggregation and arterial thrombosis suggests a mechanism for atherothrombotic disease in obesity. J Clin Invest. 2001; 108: 1533-1540.

Schafer K, Halle M, Goeschen C, Dellas C, Pynn M, Loskutoff DJ, Konstantinides S. Leptin promotes vascular remodeling and neointimal growth in mice. Arterioscler Thromb Vasc Biol. 2004; 24: 112-117.

Stephenson K, Tunstead J, Tsai A, Gordon R, Henderson S, Dansky HM. Neointimal formation after endovascular arterial injury is markedly attenuated in db/db mice. Arterioscler Thromb Vasc Biol. 2003; 23: 2027-2033.

Munzberg H, Myers MG, Jr. Molecular and anatomical determinants of central leptin resistance. Nat Neurosci. 2005; 8: 566-570.

Niswender KD, Morton GJ, Stearns WH, Rhodes CJ, Myers MG Jr, Schwartz MW. Intracellular signalling. Key enzyme in leptin-induced anorexia. Nature. 2001; 413: 794-795.

Minokoshi Y, Alquier T, Furukawa N, Kim YB, Lee A, Xue B, Mu J, Foufelle F, Ferre P, Birnbaum MJ, Stuck BJ, Kahn BB. AMP-kinase regulates food intake by responding to hormonal and nutrient signals in the hypothalamus. Nature. 2004; 428: 569-574.

Bates SH, Stearns WH, Dundon TA, Schubert M, Tso AW, Wang Y, Banks AS, Lavery HJ, Haq AK, Maratos-Flier E, Neel BG, Schwartz MW, Myers MG Jr. STAT3 signalling is required for leptin regulation of energy balance but not reproduction. Nature. 2003; 421: 856-859.

Sata M, Maejima Y, Adachi F, Fukino K, Saiura A, Sugiura S, Aoyagi T, Imai Y, Kurihara H, Kimura K, Omata M, Makuuchi M, Hirata Y, Nagai R. A mouse model of vascular injury that induces rapid onset of medial cell apoptosis followed by reproducible neointimal hyperplasia. J Mol Cell Cardiol. 2000; 32: 2097-2104.

Muzzin P, Eisensmith RC, Copeland KC, Woo SL. Correction of obesity and diabetes in genetically obese mice by leptin gene therapy. Proc Natl Acad Sci U S A. 1996; 93: 14804-14808.

Sjoland H, Eitzman DT, Gordon D, Westrick R, Nabel EG, Ginsburg D. Atherosclerosis progression in LDL receptor-deficient and apolipoprotein E-deficient mice is independent of genetic alterations in plasminogen activator inhibitor-1. Arterioscler Thromb Vasc Biol. 2000; 20: 846-852.

Whitesall SE, Hoff JB, Vollmer AP, D?Alecy LG. Comparison of simultaneous measurement of mouse systolic arterial blood pressure by radiotelemetry and tail-cuff methods. Am J Physiol Heart Circ Physiol. 2004; 286: H2408-H2415.

Ray JL, Leach R, Herbert JM, Benson M. Isolation of vascular smooth muscle cells from a single murine aorta. Methods Cell Sci. 2001; 23: 185-188.

Takahashi A, Taniguchi T, Ishikawa Y, Yokoyama M. Tranilast inhibits vascular smooth muscle cell growth and intimal hyperplasia by induction of p21(waf1/cip1/sdi1) and p53. Circ Res. 1999; 84: 543-550.

Kueng W, Silber E, Eppenberger U. Quantification of cells cultured on 96-well plates. Anal Biochem. 1989; 182: 16-19.

Mark AL, Shaffer RA, Correia ML, Morgan DA, Sigmund CD, Haynes WG. Contrasting blood pressure effects of obesity in leptin-deficient ob/ob mice and agouti yellow obese mice. J Hypertens. 1999; 17: 1949-1953.

Fruhbeck G. Intracellular signalling pathways activated by leptin. Biochem J. 2006; 393: 7-20.

Kakisis JD, Pradhan S, Cordova A, Liapis CD, Sumpio BE. The role of STAT-3 in the mediation of smooth muscle cell response to cyclic strain. Int J Biochem Cell Biol. 2005; 37: 1396-1406.

Liu B, Itoh H, Louie O, Kubota K, Kent KC. The role of phospholipase C and phosphatidylinositol 3-kinase in vascular smooth muscle cell migration and proliferation. J Surg Res. 2004; 120: 256-265.

Aizawa-Abe M, Ogawa Y, Masuzaki H, Ebihara K, Satoh N, Iwai H, Matsuoka N, Hayashi T, Hosoda K, Inoue G, Yoshimasa Y, Nakao K. Pathophysiological role of leptin in obesity-related hypertension. J Clin Invest. 2000; 105: 1243-1252.

Bernal-Mizrachi C, Weng S, Li B, Nolte LA, Feng C, Coleman T, Holloszy JO, Semenkovich CF. Respiratory uncoupling lowers blood pressure through a leptin-dependent mechanism in genetically obese mice. Arterioscler Thromb Vasc Biol. 2002; 22: 961-968.

Rahmouni K, Haynes G. Leptin and the central neural mechanisms of obesity hypertension. Drugs Today (Barc). 2002; 38: 807-817.


作者单位:Divisions of Cardiovascular Medicine and Endocrinology and Metabolism, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor.

作者: Peter F. Bodary; Yuechun Shen; Miina Öhman; K
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