Open Access

Adiponectin and its receptors are involved in hypertensive vascular injury

  • Authors:
    • Ruimin Guo
    • Min Han
    • Juan Song
    • Jun Liu
    • Yanni Sun
  • View Affiliations

  • Published online on: October 25, 2017     https://doi.org/10.3892/mmr.2017.7878
  • Pages: 209-215
  • Copyright: © Guo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Adipocyte-derived adiponectin (APN) is involved in the protection against cardiovascular disease, but the endogenous APN and its receptor expression in the perivascular adipocytes and their role in hypertensive vascular injury remain unclear. The present study aimed to detect endogenous APN and its receptor expression and their protective effects against hypertensive vascular injury. APN was mainly expressed in the perivascular adipocytes, while its receptors AdipoR1 and AdipoR2 were ubiquitously expressed in the blood vessels. Angiotensin II (Ang II)‑induced hypertension resulted in a significant decrease of APN and AdipoR1 and AdipoR2 in the perivascular adipocytes and vascular cells. The migration assay used demonstrated that APN attenuated Ang II‑induced vascular smooth muscle cells migration and p38 phosphorylation Furthermore, the in vivo study demonstrated that APN receptor agonist AdipoRon attenuated Ang II‑induced hypertensive vascular hypertrophy and fibrosis. Taken together, the present study indicated that perivascular adipocytes‑derived APN attenuated hypertensive vascular injury possibly via its receptor‑mediated inhibition of p38 signaling pathway.

Introduction

Adipose tissue, as a highly active endocrine organ, produces and releases a range of bioactive substances, such as adipokines, inflammatory factors and hormones (1,2). Among these, adiponectin (APN) has been reported as a major anti-inflammatory mediator inhibiting the development of cardiovascular disease (3,4). There is enough evidence indicating that adipocyte-derived APN can exert protective effects in different animal models such as hypertension and obesity (57). However, APN attenuates cardiac inflammation and remodeling in the classical angiotensin II (Ang II)-induced hypertension model (8). However, the potential protective role of APN on the Ang II-induced vascular remodeling has not been fully elucidated.

Adiponectin, as a fat-derived hormone, fulfills a critical role as an important messenger between adipocytes and other organs through the adiponectin receptor 1 and/or receptor 2 (AdipoR1/R2). These receptors are ubiquitously expressed in the adipose tissue, skeletal muscle and cardiovascular system (9,10). Previous studies have demonstrated that APN attenuates vascular remodeling through inhibiting vascular smooth muscle cell (VSMC) proliferation and phenotype trans differentiation (11,12). However, little is known about the endogenous APN and its receptor expression and function in the vasculatures of the Ang II-induced hypertensive model. In the present study, APN and AdipoR expression in vascular cells was demonstrated and the effects of APN on VSMCs migration were investigated. The distribution of these molecules in the vasculature of Ang-II induced hypertension were detected in vivo, and whether the APN receptor agonist AdipoRon could attenuate hypertension-associated vascular injury was determined.

Materials and methods

Cell culture

Vascular cells, such as human umbilical vein endothelial cells (HUVECs; American Type Culture Collection, Manassas, VA, USA) and VSMCs (PromoCell GmbH, Heidelberg, Germany), were cultured in high glucose Dulbecco's modified Eagles medium (DMEM) with 10% fetal bovine serum (both from Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) as previously described (13). Cells at 4–6 passages were used in subsequent experiments. 3T3-L1 adipocytes were differentiated from 3T3-L1 fibroblasts (American Type Culture Collection) as previously described (14). Briefly, 3T3-L1 fibroblasts were cultured to reach confluency in a 12-well plate and were differentiated into adipocytes in the specific differentiation medium, consisting of DMEM supplemented with 10% fetal bovine serum (FBS; Gibco; Thermo Fisher Scientific, Inc.) and 3-isobutyl-1-methylxanthine (5 µM), dexamethasone (0.25 µM) (both from Sigma-Aldrich; Merck KGaA, Darmstadt, Germany) and insulin (5 µg/ml; Gibco; Thermo Fisher Scientific, Inc.) for 2 days, followed by treatment with DMEM supplemented with 10% FBS and insulin (5 µg/ml).

Animal models

Male, 8–12 week-old C57BL/6J background mice were used in the present study. To induce hypertension, mice (n=5) were subcutaneously infused with Ang II (Sigma-Aldrich; Merck KGaA) at a 1,500 ng/kg/min dose for 14 days with ALZET mini osmotic pumps (DURECT Corporation, Cupertino, CA, USA), and in the sham group mice were treated with saline (n=5). Blood pressure was measured non-invasively every day by the tail-cuff method (Softron BP-98A; Softron Co., Ltd., Tokyo, Japan). To determine the role of APN receptor in hypertensive vascular injury, Ang II-infused mice received APN receptor agonist AdipoRon (n=5, 30 mg/kg/day; Selleck Chemicals, Houston, TX, USA) via a gavage tube. The experiments were performed in adherence with the National Institutes of Health guidelines on the Use of Laboratory Animals and were approved by the Animal Care and Use Committee of Shanghai University of Traditional Chinese Medicine (Shanghai, China).

Histological analysis

Thoracic aortas of Ang II-induced hypertensive mice were obtained as previously described (15), and then were fixed in 4% paraformaldehyde at 4°C for 24 h, embedded in paraffin, and 5 µm sections were prepared for hematoxylin (15 min at room temperature) and eosin (5 min at room temperature) (H&E) and picrosirius red staining (30 min at room temperature) to assess the vascular hypertrophy and fibrosis according to the manufacturers (Wuhan Servicebio Technology Co., Ltd., Wuhan, China). Morphometric analysis was performed using Image-Pro Plus 6.0 software to assess vascular hypertrophy by measuring the medium and lumen area. Fibrotic staining was expressed as a percentage of stained areas (red) to the total areas examined. For immunofluorescence staining, paraffin sections were first deparaffinized and rehydrated in xylene (30 min), 100% ethanol (5 min), 90% ethanol (5 min) and 80% ethanol (5 min). Following boiling in 10 mM sodium citrate buffer for 10 min to unmask antigens, the slides were blocked in buffer containing 5% normal goat serum (Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature for 1 h, incubated with primary antibody at 4°C overnight, followed by incubation with the fluorochrome-conjugated secondary antibody (1:200, for 1 h at room temperature; Invitrogen; Thermo Fisher Scientific, Inc.). Primary antibodies included perilin (PLIN1, 1:100, cat no. ab3526; Abcam, Cambridge, UK), APN (1:200, cat no. AF1119; R&D Systems, Inc., Minneapolis, MN, USA), AdipoR1 (1:150, cat no. ab70362), AdipoR2 (1:150, cat no. ab77612) (both from Abcam), α smooth muscle actin (α-SMA, 1:500, cat no. A5228; Sigma-Aldrich; Merch KGaA) and p-p38 (1:100, cat no. 4511; CST Biological Reagents Co., Ltd., Shanghai, China). Images were captured by a laser scanner confocal microscopy system (LSM 710; Zeiss GmbH, Jena, Germany).

Western blot analysis

Adipocytes, VSMCs and HUVECs were lysed in a radioimmune precipitation assay buffer (0.1% sodium dodecyl sulfate, 1.0% Triton X-100, 2 µg/ml aprotonin and 2 ng/ml leupeptin) supplemented with protease inhibitor cocktail (Roche Diagnostics, Basel, Switzerland) on ice for 10 min and sonicated. Cell lysates were centrifuged (12,000 × g, 15 min at 4°C) and the supernatant was preserved at 20°C for western blot analysis. Protein concentrations were determined using the Bradford method. Aliquots of protein (20 µg) were run on a 10% sodium dodecyl sulfate-polyacrylamide gel followed by blotting onto polyvinylidene fluoride membranes by wet transfer. Membranes were blocked in blocking buffer (5% nonfat milk in TBS buffer with 0.1% Tween-20) and were incubated with primary antibody at 4°C overnight. The primary antibodies included APN (1:2,000, cat no. AF1119, R&D Systems, Inc.), AdipoR1 (1:1,000, cat no. ab70362), AdipoR2 (1:1,000, cat no. ab77612) (both from Abcam), α-SMA (1:5,000, cat no. A5228, Sigma-Aldrich; Merch KGaA), p-p38 (1:1,000, cat no. 4511), t-p38 (1:1,000, cat no. 9212) (both from CST Biological Reagents Co., Ltd.) and β-actin (1:5,000, cat no. ab8226; Abcam). The membranes were washed with TBS with Tween-20 for 3×10 min followed by incubation with the horseradish peroxidase-conjugated secondary antibodies [rabbit anti-mouse IgG (1:5,000, cat no. ab6728); goat anti-rabbit IgG (1:5,000, cat no. ab6721); rabbit anti-goat IgG (1:5,000, cat no. ab6741) (all from Abcam)] for 1 h at room temperature. Protein bands were detected by chemiluminescence (EMD Millipore, Billerica, MA, USA).

Migration assay

VSMCs migration was measured using a Transwell chamber with a polycarbonate membrane (6.5 mm diameter and 8.0 µm pore size; Corning Incorporated, Corning, NY, USA). Briefly, serum-starved VSMCs were trypsinized and resuspended in DMEM with 0.5% bovine serum albumin (Sigma Sigma-Aldrich; Merch KGaA). Cell suspension (100 µl, 1.0×106 cells/ml) was loaded in the upper compartment of the chamber. DMEM with APN (10 µg/ml) were added to lower compartment and incubated at 37°C. Following 2 h incubation, Ang II (100 nM) was added. After 6 h, the cells remaining on the upper surface of the chamber were removed with a cotton swap, migrated cells on the lower surface of the chamber were fixed in 4% paraformaldehyde at room temperature for 1 h and stained with hematoxylin (15 min at room temperature) and eosin (5 min at room temperature) and three random microscopic fields (magnification, ×200) per well were quantified by a fluorescence microscope (DM3000; Leica, Wetzlar, Germany).

Statistical analysis

Statistical analysis was performed using SPSS 19.0 (SPSS; IBM Corp., Armonk, NY, USA). Data are expressed as the mean ± standard deviation. Comparisons of experimental groups were analyzed by Student's t-test (between two groups) or one-way analysis of variance, followed by the post-hoc Dunnett's test for data with more than two groups (Levene's tests for equal variance and Welch's and Brown-Forsythe's test for unequal variances). P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of APN and AdipoRs in vascular cells

APN and AdipoR expression were detected in adipocytes, VSMCs and endothelial cells (ECs) by western blot analysis. As demonstrated in Fig. 1, APN expression was highest in the adipocytes, with the expression of APN in the VSMCs and ECs less than in the adipocytes. Additionally, AdipoR1 and AdipoR2 were expressed in the VSMCs, ECs and adipocytes.

Expression of APN and AdipoR in the vasculature

To detect APN and AdipoR expression in the blood vessels, immunostaining of cross-sections of thoracic aorta in Sham and Ang II-induced hypertensive mice was performed. Ang II infusion resulted in a dramatic increased blood pressure including systolic and diastolic blood pressure (Fig. 2A). In accordance with the in vitro results, APN was mainly distributed in the perivascular adipocytes (Fig. 2B and C, perilipin-positive cells), while AdipoR1 and AdipoR2 were expressed in both VSMCs (α smooth muscle actin-positive cells) and adipocytes (Fig. 2D and E). Ang II-induced hypertension resulted in decreased APN and AdipoR expression in the vasculature.

Effects of Ang II on APN and AdipoR expression

To detect the direct effects of Ang II on APN, APN levels in the 3T3-L1 adipocytes pretreated with Ang II were measured. The result demonstrated that APN expression was decreased in a time-dependent manner following Ang II treatment (Fig. 3A). In addition, it was also illustrated that AdipoR1 and AdipoR2 expression was decreased in a time-dependent manner following Ang II treatment in the VSMCs (Fig. 3B).

APN attenuates VSMCs migration

Vascular active substance-induced VSMCs migration serves a pivotal role in hypertension-associated vascular remodeling (16). Whether APN affects Ang II-induced VSMCs migration was examined by utilizing the Transwell chamber assay. As depicted in Fig. 4A, APN (10 µg/ml) significantly attenuated Ang II-induced VSMCs migration. The p38 signaling pathway is known to be involved in the regulation of VSMC migration (17) In the present study, the phosphorylation of p38 mitogen-activated protein kinase (MAPK) was reduced following APN treatment in Ang II-stimulated VSMCs (Fig. 4B).

AdipoRon attenuates hypertensive vascular remodeling

AdipoRon is an orally active APN receptor activator, which potentially exerts protective effects against cardiac remodeling (18,19). In order to detect the effects of AdipoRon in hypertension-associated vascular remodeling, the Ang II-infused mice were orally treated with AdipoRon (30 mg/kg/day). As demonstrated in Fig. 5A, AdipoRon treatment resulted in decreased p38 phosphorylation. In addition, AdipoRon attenuated Ang II-induced perivascular collagen deposition and vascular hypertrophy as detected by Sirious Red and H&E stainings (Fig. 5B and C, respectively).

Discussion

In the present study, the distribution of APN and AdipoR in the vasculature was detected and it was demonstrated that APN was predominantly expressed in the perivascular adipocytes, while AdipoR1 and R2 were ubiquitously expressed in the blood vessels. APN attenuated Ang II-induced VSMCs migration and p38 phosphorylation. Furthermore, APN receptor agonist, AdipoRon, appeared to attenuate Ang II-induced hypertensive vascular hypertrophy and fibrosis.

Perivascular adipocytes are an important source of adipokines and their role in regulating vascular function is well established (2022). APN, an abundant adipocyte-derived factor with a variety of biological functions, was first described in 1995 (23). Over the past two decades, numerous studies have elucidated its physiological functions, which may improve insulin sensitivity in insulin target tissues, modulate inflammatory responses, and serve a crucial role in the regulation of energy metabolism (24,25). In the cardiovascular system, APN ameliorates Ang II-induced cardiac remodeling by attenuating inflammation, it inhibits connective tissue growth factor-induced VSMCs proliferation, and promotes endothelial nitric oxide synthase expression and nitric oxide release from endothelial cells (24,26). Previous studies have demonstrated that hypertensive mice express decreased APN plasma levels compared with the controls (27). To the best of our knowledge, the present study was the first to demonstrate that hypertension induced APN downregulation in local perivascular adipocytes. VSMC dysfunction, including migration, proliferation and phenotype transdifferentiation, has an important role in hypertension-induced vascular remodeling. VSMC migration is associated with vascular remodeling when exposed to high stress, such as hypertension. It has been demonstrated that Ang II significantly increased the VSMC migration (28). APN receptors, AdipoR1 and AdipoR2, were originally identified by screening a skeletal muscle expression library for cDNAs that encoded proteins that to the globular domain of APN (29). Both receptors contain seven transmembrane domains and belong to the progestin and adipoQ receptor family, which has the opposite transmembrane topology to the G-protein coupled receptors, and have the N-terminus in the cytoplasm and the C-terminus facing the extracellular space. Although ubiquitously expressed, AdipoR1 is most abundant in skeletal muscle whereas AdipoR2 is mainly restricted to the liver (30,31). In the present study, ubiquitous expression of AdipoR1 and AdipoR2 in the vasculature was demonstrated, including ECs, VSMCs and perivascular adipocytes. In an obesity and diabetic mouse model, AdipoR1 expression was reduced in the target tissue (32,33). However, in a rat model of chronic kidney disease, AdipoR1 and AdipoR2 expression was significantly increased in renal cells (34). In the present study, AdipoR1 and AdipoR2 expression was decreased in the cultured VSMCs treated with Ang II. In addition, Ang II-induced hypertension resulted in a significant decrease of AdipoR1 and AdipoR2 expression in the perivascular adipocytes and VSMCs in vivo. AdipoR1 or AdipoR2 mediate the effects of APN by regulating multiple signaling pathways, including the peroxisome proliferator-activated receptor-α, the AMP-activated protein kinase, and the p38 MAPK (12,35,36). Previous studies have demonstrated that APN could attenuate lipid deposition and apoptosis through activating p38 MAPK pathway (37,38). In the present study, it was demonstrated that APN alone could induce p38 phosphorylation, which is in accordance with previous studies (39,40). Furthermore, Ang II promoted p38 phosphorylation. It could be speculated that the APN/p38 axis may have an inhibitory feedback effect on p38 phosphorylation, with p38 phosphorylation level is decreased following Ang II treatment. APN receptor agonist, AdipoRon, inhibited p38 phosphorylation in the vasculature of Ang II-induced hypertensive mice.

In conclusion, the present study demonstrated that Ang II-induced hypertensive mice had decreased expression of APN and AdipoR in the perivascular adipocytes and VSMCs. APN attenuated Ang II-induced VSMCs migration and p38 phosphorylation. APN receptor agonist AdipoRon attenuated Ang II-induced hypertensive vascular remodeling. These results suggest that APN and APN receptors may be potential therapeutic targets for inhibiting hypertensive vascular injury.

Acknowledgements

The present study was supported by the Putuo Hospital Level Subject (AASI index in old hypertension patients complicated with metabolism diseases; grant no. PT65487).

References

1 

Thalmann S and Meier CA: Local adipose tissue depots as cardiovascular risk factors. Cardiovasc Res. 75:690–701. 2007. View Article : Google Scholar : PubMed/NCBI

2 

Molica F, Morel S, Kwak BR, Rohner-Jeanrenaud F and Steffens S: Adipokines at the crossroad between obesity and cardiovascular disease. Thromb Haemost. 113:553–566. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Li FY, Cheng KK, Lam KS, Vanhoutte PM and Xu A: Cross-talk between adipose tissue and vasculature: Role of adiponectin. Acta Physiol (Oxf). 203:167–180. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Cai X, Li X, Li L, Huang XZ, Liu YS, Chen L, Zhang K, Wang L, Li X, Song J, et al: Adiponectin reduces carotid atherosclerotic plaque formation in ApoE−/−mice: Roles of oxidative and nitrosative stress and inducible nitric oxide synthase. Mol Med Rep. 11:1715–1721. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Withers SB, Bussey CE, Saxton SN, Melrose HM, Watkins AE and Heagerty AM: Mechanisms of adiponectin-associated perivascular function in vascular disease. Arterioscler Thromb Vasc Biol. 34:1637–1642. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Liu Y, Palanivel R, Rai E, Park M, Gabor TV, Scheid MP, Xu A and Sweeney G: Adiponectin stimulates autophagy and reduces oxidative stress to enhance insulin sensitivity during high-fat diet feeding in mice. Diabetes. 64:36–48. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Li C, Wang Z, Wang C, Ma Q and Zhao Y: Perivascular adipose tissue-derived adiponectin inhibits collar-induced carotid atherosclerosis by promoting macrophage autophagy. PLoS One. 10:e01240312015. View Article : Google Scholar : PubMed/NCBI

8 

Qi GM, Jia LX, Li YL, Li HH and Du J: Adiponectin suppresses angiotensin II-induced inflammation and cardiac fibrosis through activation of macrophage autophagy. Endocrinology. 155:2254–2265. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Yamauchi T, Kamon J, Ito Y, Tsuchida A, Yokomizo T, Kita S, Sugiyama T, Miyagishi M, Hara K, Tsunoda M, et al: Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature. 423:762–769. 2003. View Article : Google Scholar : PubMed/NCBI

10 

Wang ZV and Scherer PE: Adiponectin, the past two decades. J Mol Cell Biol. 8:93–100. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Zhan JK, Wang YJ, Wang Y, Tang ZY, Tan P, Huang W and Liu YS: Adiponectin attenuates the osteoblastic differentiation of vascular smooth muscle cells through the AMPK/mTOR pathway. Exp Cell Res. 323:352–358. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Zhang W, Shu C, Li Q, Li M and Li X: Adiponectin affects vascular smooth muscle cell proliferation and apoptosis through modulation of the mitofusin-2-mediated Ras-Raf-Erk1/2 signaling pathway. Mol Med Rep. 12:4703–4707. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Alexis JD, Wang N, Che W, Lerner-Marmarosh N, Sahni A, Korshunov VA, Zou Y, Ding B, Yan C, Berk BC and Abe J: Bcr kinase activation by angiotensin II inhibits peroxisome-proliferator-activated receptor gamma transcriptional activity in vascular smooth muscle cells. Circ Res. 104:69–78. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Yang Z, Hong LK, Follett J, Wabitsch M, Hamilton NA, Collins BM, Bugarcic A and Teasdale RD: Functional characterization of retromer in GLUT4 storage vesicle formation and adipocyte differentiation. Faseb J. 30:1037–1050. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Zhao Y, Zhang C, Wei X, Li P, Cui Y, Qin Y, Wei X, Jin M, Kohama K and Gao Y: Heat shock protein 60 stimulates the migration of vascular smooth muscle cells via Toll-like receptor 4 and ERK MAPK activation. Sci Rep. 5:153522015. View Article : Google Scholar : PubMed/NCBI

16 

Montezano AC, Cat Nguyen Dinh A, Rios FJ and Touyz RM: Angiotensin II and vascular injury. Curr Hypertens Rep. 16:4312014. View Article : Google Scholar : PubMed/NCBI

17 

Yu L, Huang X, Huang K, Gui C, Huang Q and Wei B: Ligustrazine attenuates the platelet-derived growth factor-BB-induced proliferation and migration of vascular smooth muscle cells by interrupting extracellular signal-regulated kinase and P38 mitogen-activated protein kinase pathways. Mol Med Rep. 12:705–711. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Okada-Iwabu M, Yamauchi T, Iwabu M, Honma T, Hamagami K, Matsuda K, Yamaguchi M, Tanabe H, Kimura-Someya T, Shirouzu M, et al: A small-molecule AdipoR agonist for type 2 diabetes and short life in obesity. Nature. 503:493–499. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Fisman EZ and Tenenbaum A: Adiponectin: A manifold therapeutic target for metabolic syndrome, diabetes and coronary disease? Cardiovasc Diabetol. 13:1032014. View Article : Google Scholar : PubMed/NCBI

20 

Takaoka M, Nagata D, Kihara S, Shimomura I, Kimura Y, Tabata Y, Saito Y, Nagai R and Sata M: Periadventitial adipose tissue plays a critical role in vascular remodeling. Circ Res. 105:906–911. 2009. View Article : Google Scholar : PubMed/NCBI

21 

Sheng LJ, Ruan CC, Ma Y, Chen DR, Kong LR, Zhu DL and Gao PJ: Beta3 adrenergic receptor is involved in vascular injury in deoxycorticosterone acetate-salt hypertensive mice. FEBS lett. 590:769–778. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Takaoka M, Suzuki H, Shioda S, Sekikawa K, Saito Y, Nagai R and Sata M: Endovascular injury induces rapid phenotypic changes in perivascular adipose tissue. Arterioscler Thromb Vasc Biol. 30:1576–1582. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Scherer PE, Williams S, Fogliano M, Baldini G and Lodish HF: A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem. 270:26746–26749. 1995. View Article : Google Scholar : PubMed/NCBI

24 

Lee S and Kwak HB: Role of adiponectin in metabolic and cardiovascular disease. J Exerc Rehabil. 10:54–59. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Chakraborti CK: Role of adiponectin and some other factors linking type 2 diabetes mellitus and obesity. World J Diabetes. 6:1296–1308. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Lima Freitas LC, Braga VA, do Socorro de Franca Silva M, Cruz JC, Sousa Santos SH, de Oliveira Monteiro MM and Balarini CM: Adipokines, diabetes and atherosclerosis: An inflammatory association. Front Physiol. 6:3042015.PubMed/NCBI

27 

Ndisang JF and Jadhav A: Heme arginate therapy enhanced adiponectin and atrial natriuretic peptide, but abated endothelin-1 with attenuation of kidney histopathological lesions in mineralocorticoid-induced hypertension. J Pharmacol Exp Ther. 334:87–98. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Zhang F, Ren X, Zhao M, Zhou B and Han Y: Angiotensin-(1–7) abrogates angiotensin II-induced proliferation, migration and inflammation in VSMCs through inactivation of ROS-mediated PI3 K/Akt and MAPK/ERK signaling pathways. Sci Rep. 6:346212016. View Article : Google Scholar : PubMed/NCBI

29 

Kadowaki T and Yamauchi T: Adiponectin and adiponectin receptors. Endocr Rev. 26:439–451. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Yamauchi T and Kadowaki T: Adiponectin receptor as a key player in healthy longevity and obesity-related diseases. Cell Metab. 17:185–196. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Lustig Y, Hemi R and Kanety H: Regulation and function of adiponectin receptors in skeletal muscle. Vitam Horm. 90:95–123. 2012. View Article : Google Scholar : PubMed/NCBI

32 

Wade TE, Mathur A, Lu D, Swartz-Basile DA, Pitt HA and Zyromski NJ: Adiponectin receptor-1 expression is decreased in the pancreas of obese mice. J Surg Res. 154:78–84. 2009. View Article : Google Scholar : PubMed/NCBI

33 

Lin T, Qiu Y, Liu Y, Mohan R, Li Q and Lei B: Expression of adiponectin and its receptors in type 1 diabetes mellitus in human and mouse retinas. Mol Vis. 19:1769–1778. 2013.PubMed/NCBI

34 

Yu Y, Bao BJ, Fan YP, Shi L and Li SQ: Changes of adiponectin and its receptors in rats following chronic renal failure. Ren Fail. 36:92–97. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Thundyil J, Pavlovski D, Sobey CG and Arumugam TV: Adiponectin receptor signalling in the brain. Br J Pharmacol. 165:313–327. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Almabrouk TA, Ewart MA, Salt IP and Kennedy S: Perivascular fat, AMP-activated protein kinase and vascular diseases. Br J Pharmacol. 171:595–617. 2014. View Article : Google Scholar : PubMed/NCBI

37 

Yan J, Gan L, Qi R and Sun C: Adiponectin decreases lipids deposition by p38 MAPK/ATF2 signaling pathway in muscle of broilers. Mol Biol Rep. 40:7017–7025. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Wang Y, Zhang J, Zhang L, Gao P and Wu X: Adiponectin attenuates high glucose-induced apoptosis through the AMPK/p38 MAPK signaling pathway in NRK-52E cells. PLoS One. 12:e01782152017. View Article : Google Scholar : PubMed/NCBI

39 

Pu Y, Wang M, Hong Y, Wu Y and Tang Z: Adiponectin promotes human jaw bone marrow mesenchymal stem cell chemotaxis via CXCL1 and CXCL8. J Cell Mol Med. 21:1411–1419. 2017. View Article : Google Scholar : PubMed/NCBI

40 

Huang CY, Chang AC, Chen HT, Wang SW, Lo YS and Tang CH: Adiponectin promotes VEGF-C-dependent lymphangiogenesis by inhibiting miR-27b through a CaMKII/AMPK/p38 signaling pathway in human chondrosarcoma cells. Clin Sci (Lond). 130:1523–1533. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2018
Volume 17 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo R, Han M, Song J, Liu J and Sun Y: Adiponectin and its receptors are involved in hypertensive vascular injury. Mol Med Rep 17: 209-215, 2018
APA
Guo, R., Han, M., Song, J., Liu, J., & Sun, Y. (2018). Adiponectin and its receptors are involved in hypertensive vascular injury. Molecular Medicine Reports, 17, 209-215. https://doi.org/10.3892/mmr.2017.7878
MLA
Guo, R., Han, M., Song, J., Liu, J., Sun, Y."Adiponectin and its receptors are involved in hypertensive vascular injury". Molecular Medicine Reports 17.1 (2018): 209-215.
Chicago
Guo, R., Han, M., Song, J., Liu, J., Sun, Y."Adiponectin and its receptors are involved in hypertensive vascular injury". Molecular Medicine Reports 17, no. 1 (2018): 209-215. https://doi.org/10.3892/mmr.2017.7878