Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2010 Dec;120(12):4342-52.
doi: 10.1172/JCI43464.

T-cadherin is critical for adiponectin-mediated cardioprotection in mice

Affiliations

T-cadherin is critical for adiponectin-mediated cardioprotection in mice

Martin S Denzel et al. J Clin Invest. 2010 Dec.

Abstract

The circulating, adipocyte-secreted hormone adiponectin (APN) exerts protective effects on the heart under stress conditions. The receptors binding APN to cardiac tissue, however, have remained elusive. Here, we report that the glycosyl phosphatidylinositol–anchored cell surface glycoprotein T-cadherin (encoded by Cdh13) protects against cardiac stress through its association with APN in mice. We observed extensive colocalization of T-cadherin and APN on cardiomyocytes in vivo. In T-cadherin-deficient mice, APN failed to associate with cardiac tissue, and its levels dramatically increased in the circulation. Pressure overload stress resulted in exacerbated cardiac hypertrophy in T-cadherin-null mice and paralleled corresponding defects in mice lacking APN. During ischemia-reperfusion injury, the absence of T-cadherin increased infarct size similar to that in APN-null mice. Myocardial AMPK is a major downstream protective signaling target of APN. In both cardiac hypertrophy and ischemia-reperfusion models, T-cadherin was necessary for APN-dependent AMPK phosphorylation. In APN-null mice, recombinant adenovirus-expressed APN reduced exaggerated hypertrophy and infarct size and restored AMPK phosphorylation as previously reported. In contrast, rescue was ineffective in mice lacking T-cadherin in addition to APN. These data suggest that T-cadherin protects from stress-induced pathological cardiac remodeling by binding APN and activating its cardioprotective functions.

PubMed Disclaimer

Figures

Figure 1
Figure 1. T-cadherin and APN in the heart.
(A) Immunostaining of WT, Tcad-KO, and APN-KO hearts using T-cadherin and APN antibodies. Scale bars: 5 μm. (B) Immunoblot of myocardial lysates of the indicated genotypes. T-cadherin was detected as a doublet in the 130-kDa pro-protein and the proteolytically cleaved 105-kDa mature isoforms. Denatured APN appeared as a single 30-kDa band. (C) Expression levels of T-cadherin mRNA in the myocardium of WT and APN-KO mice and in isolated cardiomyocytes (1°CM). (D) Serum APN ELISA from WT, Tcad-KO, and APN-KO mice. P < 0.0001, WT versus Tcad-KO.
Figure 2
Figure 2. T-cadherin directly binds APN.
(A) Differentiated C2C12 myotubes were treated with 25 μg/ml HMW APN for 30 minutes, washed with PBS, and cross-linked with dimethyl 3,3′-dithiopropionimidate dihydrochloride. Lysates were subjected to immunoprecipitation with unspecific IgGs or APN antibodies. (B) HEK293 cells stably transfected with pL-N1 control (N) or pL–T-cadherin were transiently transfected with HA-AdipoR1 or Flag-AdipoR2 coding constructs using the calcium phosphate method. Cells were incubated with 1:10 diluted serum from Tcad-KO (+ APN) or DKO mice (– APN) and subsequently washed 3 times with PBS. Immunoblotting using the indicated antibodies revealed presence of the respective proteins; AdipoR1 and -R2 were detected using the respective tags.
Figure 3
Figure 3. Exaggerated cardiac hypertrophy and reduced AMPK phosphorylation in Tcad-KO and APN-KO mice after 7 days of TAC.
(A) Representative images of trichrome-stained WT, Tcad-KO, and APN-KO hearts 7 days after sham or TAC surgery. Scale bar: 2 mm. (B) HW/BW ratio. n = 8 (WT); 7 (Tcad-KO); 11 (APN-KO). (C) M-mode echocardiograms from WT, Tcad-KO, and APN-KO mice 7 days after TAC surgery. White bars indicate diastolic LV wall thickness after TAC. (D) Quantification of LVPWd before and 7 days after TAC surgery. n = 10 (WT); 14 (Tcad-KO); 5 (APN-KO). (E) Fluorescent wheat germ agglutinin–stained cell surfaces in hearts from WT, Tcad-KO, and APN-KO mice 7 days after sham or TAC surgery. Scale bar: 10 μm. (F) Quantification of myocyte cross-sectional areas in E. n = 3 (sham) or 4 (TAC) per group. (G) Representative immunoblots from WT, Tcad-KO, and APNKO myocardial lysates 7 days after TAC using phospho-specific (Thr-172) and total anti-AMPK antibodies with reference to β-tubulin. (H) Quantification of phospho-AMPK/total AMPK ratio in G. n = 5 (WT and APN-KO); 4 (Tcad-KO). *P < 0.05, **P < 0.01, ***P < 0.001 versus WT or as indicated by brackets.
Figure 4
Figure 4. Long-term TAC leads to dilation and reduced contractility in Tcad-KO and APN-KO hearts.
(A) Representative images of trichrome-stained WT, Tcad-KO, and APN-KO hearts 28 days after TAC surgery. Scale bar: 2 mm. (B) HW/BW ratios. (C) Representative images from TUNEL-stained heart sections in WT, Tcad-KO, and APN-KO hearts, with quantification of TUNEL-positive area of whole heart sections. Scale bar: 20 μm. (D) Representative images of CD31-stained heart sections. Quantification was carried out in the free wall of the LV by counting number of capillaries per high-power field. Scale bar: 5 μm. (E) Fractional shortening (FS) was significantly reduced in Tcad-KO and APN-KO animals. (F) LV inner diameter in diastole (LVIDd) was significantly increased in Tcad-KO and APN-KO mice. (BF) n = 4 (WT and Tcad-KO); 3 (APN-KO). *P < 0.05, **P < 0.01 versus WT.
Figure 5
Figure 5. T-cadherin reexpression in APN-KO hearts after adAPN treatment.
(A) Immunostaining of heart sections from adGFP- or adAPN-treated APN-KO mice with T-cadherin and APN antibodies. Control staining confirmed absence of GFP signals in the heart. Scale bar: 5 μm. (B) Cardiac T-cadherin mRNA levels measured by qPCR in adGFP- (n = 5) or adAPN-treated (n = 3) APN-KO mice.
Figure 6
Figure 6. Reversion of exaggerated cardiac hypertrophy phenotype in APN-KO depends on T-cadherin.
(A) Representative trichrome-stained heart sections 7 days after TAC. Scale bar is 2 mm. (B) HW/BW ratios in adGFP- or adAPN-treated APN-KO and DKO mice after sham or TAC surgery. (C) Representative images of wheat germ agglutinin–stained myocardium to reveal cardiomyocyte cross-sectional area, quantified in D. Scale bar: 5 μm. (E) Representative immunoblots showing cardiac AMPK and ACC phosphorylation in adGFP- or adAPN-treated APN-KO and DKO animals 7 days after TAC. (F and G) Quantification of phospho-AMPK/total AMPK and phospho-ACC/total ACC ratios. (B, D, F, and G) n = 3 (adGFP) or 4 (adAPN) per group. *P < 0.05, **P < 0.01, ***P < 0.001.
Figure 7
Figure 7. T-cadherin is required for APN-mediated protection against ischemia-reperfusion.
(A) Representative images of myocardial tissue sections 48 hours after ischemia-reperfusion. Outlined white areas indicate IA, red color indicates AAR. (B) Quantification of infarct size by AAR/LV area, IA/AAR, and IA/LV ratios. n = 4 (adGFP-treated WT, Tcad-KO, and DKO and adAPN-treated DKO); 6 (adGFP-treated APN-KO); 3 (adAPN-treated APN-KO). *P < 0.05 versus adGFP-treated Tcad-KO, APN-KO, and DKO and adAPN-treated DKO. (C) Representative immunoblots from WT, Tcad-KO, and APN-KO myocardial lysates of ischemic areas 48 hours after ischemia-reperfusion using phospho-specific (Thr-172) and total anti-AMPK antibodies. Quantification of phospho-AMPK/total AMPK ratios (n = 3 per group). (D) Representative images of TUNEL-stained heart sections. Scale bar: 20 μm. TUNEL-positive area was analyzed in regions adjacent to IAs (n = 3 per group).*P < 0.05, **P < 0.01 versus WT.

Similar articles

Cited by

References

    1. Prasad A, Stone GW, Holmes DR, Gersh B. Reperfusion injury, microvascular dysfunction, and cardioprotection: the “dark side” of reperfusion. Circulation. 2009;120(21):2105–2112. doi: 10.1161/CIRCULATIONAHA.108.814640. - DOI - PubMed
    1. Lorell BH, Carabello BA. Left ventricular hypertrophy: pathogenesis, detection, and prognosis. Circulation. 2000;102(4):470–479. - PubMed
    1. Artham SM, Lavie CJ, Milani RV, Patel DA, Verma A, Ventura HO. Clinical impact of left ventricular hypertrophy and implications for regression. Prog Cardiovasc Dis. 2009;52(2):153–167. doi: 10.1016/j.pcad.2009.05.002. - DOI - PubMed
    1. Eisenberg MS, Mengert TJ. Cardiac resuscitation. . N Engl J Med. 2001;344(17):1304–1313. doi: 10.1056/NEJM200104263441707. - DOI - PubMed
    1. Rogers WJ, et al. Temporal trends in the treatment of over 1.5 million patients with myocardial infarction in the US from 1990 through 1999: the National Registry of Myocardial Infarction 1, 2, and 3. J Am Coll Cardiol. 2000;36(7):2056–2063. doi: 10.1016/S0735-1097(00)00996-7. - DOI - PubMed

Publication types

MeSH terms