Review Topical Sections

The functional roles of T-cadherin in mammalian biology

  • Received: 21 November 2016 Accepted: 26 January 2017 Published: 13 February 2017
  • T-cadherin is a cadherin and cell adhesion molecule that is anchored to the cell surface membrane through a glycosylphosphatidylinositol moiety. T-cadherin lacks a transmembrane and cytoskeletal domain, suggesting that it must interact with other membrane-bound molecules to elicit cellular signaling to modulate normal cellular functions, and alternatively its absence can be a factor in promoting neoplasia. Moreover, apart from binding to itself it can sequester adiponectin to the cell surface. Consistent with these observations, recent research has expanded the scope of T-cadherin’s role in cancer, neuronal function, metabolism and cardiovascular disease. In this context, we highlight the experimental and genomic evidence that links T-cadherin with these diseases. In particular, we discuss how T-cadherin homophilic and heterophilic interactions impact on signaling pathways and cellular behavior.

    Citation: Jade Sternberg, Miriam Wankell, V. Nathan Subramaniam, Lionel W. Hebbard. The functional roles of T-cadherin in mammalian biology[J]. AIMS Molecular Science, 2017, 4(1): 62-81. doi: 10.3934/molsci.2017.1.62

    Related Papers:

  • T-cadherin is a cadherin and cell adhesion molecule that is anchored to the cell surface membrane through a glycosylphosphatidylinositol moiety. T-cadherin lacks a transmembrane and cytoskeletal domain, suggesting that it must interact with other membrane-bound molecules to elicit cellular signaling to modulate normal cellular functions, and alternatively its absence can be a factor in promoting neoplasia. Moreover, apart from binding to itself it can sequester adiponectin to the cell surface. Consistent with these observations, recent research has expanded the scope of T-cadherin’s role in cancer, neuronal function, metabolism and cardiovascular disease. In this context, we highlight the experimental and genomic evidence that links T-cadherin with these diseases. In particular, we discuss how T-cadherin homophilic and heterophilic interactions impact on signaling pathways and cellular behavior.


    加载中
    [1] Berx G, van Roy F (2009) Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol 1: a003129.
    [2] Ranscht B, Dours-Zimmermann MT (1991) T-cadherin, a novel cadherin cell adhesion molecule in the nervous system lacks the conserved cytoplasmic region. Neuron 7: 391-402. doi: 10.1016/0896-6273(91)90291-7
    [3] Pfaff D, Philippova M, Kyriakakis E, et al. (2011) Paradoxical effects of T-cadherin on squamous cell carcinoma: up- and down-regulation increase xenograft growth by distinct mechanisms. J Pathol 225: 512-524. doi: 10.1002/path.2900
    [4] Fredette BJ, Ranscht B (1994) T-cadherin expression delineates specific regions of the developing motor axon-hindlimb projection pathway. J Neurosci 14: 7331-7346.
    [5] Philippova M, Joshi MB, Kyriakakis E, et al. (2009) A guide and guard: the many faces of T-cadherin. Cell Signal 21: 1035-1044. doi: 10.1016/j.cellsig.2009.01.035
    [6] Rubina KA, Surkova EI, Semina EV, et al. (2015) T-Cadherin Expression in Melanoma Cells Stimulates Stromal Cell Recruitment and Invasion by Regulating the Expression of Chemokines, Integrins and Adhesion Molecules. Cancers (Basel) 7: 1349-1370. doi: 10.3390/cancers7030840
    [7] Wyder L, Vitaliti A, Schneider H, et al. (2000) Increased expression of H/T-cadherin in tumor-penetrating blood vessels. Cancer Res 60: 4682-4688.
    [8] Dames SA, Bang E, Haussinger D, et al. (2008) Insights into the low adhesive capacity of human T-cadherin from the NMR structure of Its N-terminal extracellular domain. J Biol Chem 283: 23485-23495. doi: 10.1074/jbc.M708335200
    [9] Teng MS, Hsu LA, Wu S, et al. (2015) Association of CDH13 genotypes/haplotypes with circulating adiponectin levels, metabolic syndrome, and related metabolic phenotypes: the role of the suppression effect. PLoS One 10: e0122664. doi: 10.1371/journal.pone.0122664
    [10] Org E, Eyheramendy S, Juhanson P, et al. (2009) Genome-wide scan identifies CDH13 as a novel susceptibility locus contributing to blood pressure determination in two European populations. Hum Mol Genet 18: 2288-2296. doi: 10.1093/hmg/ddp135
    [11] Angst BD, Marcozzi C, Magee AI (2001) The cadherin superfamily: diversity in form and function. J Cell Sci 114: 629-641.
    [12] Sato M, Mori Y, Sakurada A, et al. (1998) The H-cadherin (CDH13) gene is inactivated in human lung cancer. Hum Genet 103: 96-101. doi: 10.1007/s004390050790
    [13] Behrens J, Lowrick O, Klein-Hitpass L, et al. (1991) The E-cadherin promoter: functional analysis of a G.C-rich region and an epithelial cell-specific palindromic regulatory element. Proc Natl Acad Sci U S A 88: 11495-11499.
    [14] Jarrard DF, Paul R, van Bokhoven A, et al. (1997) P-Cadherin is a basal cell-specific epithelial marker that is not expressed in prostate cancer. Clin Cancer Res 3: 2121-2128.
    [15] Bromhead C, Miller JH, McDonald FJ (2006) Regulation of T-cadherin by hormones, glucocorticoid and EGF. Gene 374: 58-67. doi: 10.1016/j.gene.2006.01.013
    [16] Niermann T, Schmutz S, Erne P, et al. (2003) Aryl hydrocarbon receptor ligands repress T-cadherin expression in vascular smooth muscle cells. Biochem Biophys Res Commun 300: 943-949. doi: 10.1016/S0006-291X(02)02970-4
    [17] Ellmann L, Joshi MB, Resink TJ, et al. (2012) BRN2 is a transcriptional repressor of CDH13 (T-cadherin) in melanoma cells. Lab Invest 92: 1788-1800. doi: 10.1038/labinvest.2012.140
    [18] Kuzmenko YS, Stambolsky D, Kern F, et al. (1998) Characteristics of smooth muscle cell lipoprotein binding proteins (p105/p130) as T-cadherin and regulation by positive and negative growth regulators. Biochem Biophys Res Commun 246: 489-494. doi: 10.1006/bbrc.1998.8645
    [19] Ciatto C, Bahna F, Zampieri N, et al. (2010) T-cadherin structures reveal a novel adhesive binding mechanism. Nat Struct Mol Biol 17: 339-347. doi: 10.1038/nsmb.1781
    [20] Vestal DJ, Ranscht B (1992) Glycosyl phosphatidylinositol--anchored T-cadherin mediates calcium-dependent, homophilic cell adhesion. J Cell Biol 119: 451-461. doi: 10.1083/jcb.119.2.451
    [21] Sacristan MP, Vestal DJ, Dours-Zimmermann MT, et al. (1993) T-cadherin 2: molecular characterization, function in cell adhesion, and coexpression with T-cadherin and N-cadherin. J Neurosci Res 34: 664-680. doi: 10.1002/jnr.490340610
    [22] Harrison OJ, Bahna F, Katsamba PS, et al. (2010) Two-step adhesive binding by classical cadherins. Nat Struct Mol Biol 17: 348-357. doi: 10.1038/nsmb.1784
    [23] Winterhalter PR, Lommel M, Ruppert T, et al. (2013) O-glycosylation of the non-canonical T-cadherin from rabbit skeletal muscle by single mannose residues. FEBS Lett 587: 3715-3721. doi: 10.1016/j.febslet.2013.09.041
    [24] Stambolsky DV, Kuzmenko YS, Philippova MP, et al. (1999) Identification of 130 kDa cell surface LDL-binding protein from smooth muscle cells as a partially processed T-cadherin precursor. Biochim Biophys Acta 1416: 155-160. doi: 10.1016/S0005-2736(98)00218-1
    [25] Mavroconstanti T, Johansson S, Winge I, et al. (2013) Functional properties of rare missense variants of human CDH13 found in adult attention deficit/hyperactivity disorder (ADHD) patients. PLoS One 8: e71445. doi: 10.1371/journal.pone.0071445
    [26] Wang Y, Lam KS, Yau MH, et al. (2008) Post-translational modifications of adiponectin: mechanisms and functional implications. Biochem J 409: 623-633. doi: 10.1042/BJ20071492
    [27] Fruebis J, Tsao TS, Javorschi S, et al. (2001) Proteolytic cleavage product of 30-kDa adipocyte complement-related protein increases fatty acid oxidation in muscle and causes weight loss in mice. Proc Natl Acad Sci U S A 98: 2005-2010. doi: 10.1073/pnas.98.4.2005
    [28] Pajvani UB, Hawkins M, Combs TP, et al. (2004) Complex distribution, not absolute amount of adiponectin, correlates with thiazolidinedione-mediated improvement in insulin sensitivity. J Biol Chem 279: 12152-12162. doi: 10.1074/jbc.M311113200
    [29] Scherer PE, Williams S, Fogliano M, et al. (1995) A novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem 270: 26746-26749. doi: 10.1074/jbc.270.45.26746
    [30] Yamauchi T, Kamon J, Ito Y, et al. (2003) Cloning of adiponectin receptors that mediate antidiabetic metabolic effects. Nature 423: 762-769. doi: 10.1038/nature01705
    [31] Yamauchi T, Kadowaki T (2013) Adiponectin receptor as a key player in healthy longevity and obesity-related diseases. Cell Metab 17: 185-196. doi: 10.1016/j.cmet.2013.01.001
    [32] Hebbard L, Ranscht B (2014) Multifaceted roles of adiponectin in cancer. Best Pract Res Clin Endocrinol Metab 28: 59-69. doi: 10.1016/j.beem.2013.11.005
    [33] Hug C, Wang J, Ahmad NS, et al. (2004) T-cadherin is a receptor for hexameric and high-molecular-weight forms of Acrp30/adiponectin. Proc Natl Acad Sci U S A 101: 10308-10313. doi: 10.1073/pnas.0403382101
    [34] Hebbard LW, Garlatti M, Young LJ, et al. (2008) T-cadherin supports angiogenesis and adiponectin association with the vasculature in a mouse mammary tumor model. Cancer Res 68: 1407-1416. doi: 10.1158/0008-5472.CAN-07-2953
    [35] Denzel MS, Scimia MC, Zumstein PM, et al. (2010) T-cadherin is critical for adiponectin-mediated cardioprotection in mice. J Clin Invest 120: 4342-4352. doi: 10.1172/JCI43464
    [36] Parker-Duffen JL, Nakamura K, Silver M, et al. (2013) T-cadherin is essential for adiponectin-mediated revascularization. J Biol Chem 288: 24886-24897. doi: 10.1074/jbc.M113.454835
    [37] Rivero O, Sich S, Popp S, et al. (2013) Impact of the ADHD-susceptibility gene CDH13 on development and function of brain networks. Eur Neuropsychopharmacol 23: 492-507. doi: 10.1016/j.euroneuro.2012.06.009
    [38] Fredette BJ, Miller J, Ranscht B (1996) Inhibition of motor axon growth by T-cadherin substrata. Development 122: 3163-3171.
    [39] Hayano Y, Zhao H, Kobayashi H, et al. (2014) The role of T-cadherin in axonal pathway formation in neocortical circuits. Development 141: 4784-4793. doi: 10.1242/dev.108290
    [40] Treutlein J, Cichon S, Ridinger M, et al. (2009) Genome-wide association study of alcohol dependence. Arch Gen Psychiatry 66: 773-784. doi: 10.1001/archgenpsychiatry.2009.83
    [41] Johnson C, Drgon T, Liu QR, et al. (2006) Pooled association genome scanning for alcohol dependence using 104,268 SNPs: validation and use to identify alcoholism vulnerability loci in unrelated individuals from the collaborative study on the genetics of alcoholism. Am J Med Genet B Neuropsychiatr Genet 141B: 844-853. doi: 10.1002/ajmg.b.30346
    [42] Arias-Vasquez A, Altink ME, Rommelse NN, et al. (2011) CDH13 is associated with working memory performance in attention deficit/hyperactivity disorder. Genes Brain Behav 10: 844-851. doi: 10.1111/j.1601-183X.2011.00724.x
    [43] Rivero O, Selten MM, Sich S, et al. (2015) Cadherin-13, a risk gene for ADHD and comorbid disorders, impacts GABAergic function in hippocampus and cognition. Transl Psychiatry 5: e655. doi: 10.1038/tp.2015.152
    [44] Drgonova J, Walther D, Hartstein GL, et al. (2016) Cadherin 13: human cis-regulation and selectively-altered addiction phenotypes and cerebral cortical dopamine in knockout mice. Mol Med 22.
    [45] Shen LH, Liao MH, Tseng YC (2012) Recent advances in imaging of dopaminergic neurons for evaluation of neuropsychiatric disorders. J Biomed Biotechnol 2012: 259349.
    [46] Poliak S, Norovich AL, Yamagata M, et al. (2016) Muscle-type Identity of Proprioceptors Specified by Spatially Restricted Signals from Limb Mesenchyme. Cell 164: 512-525. doi: 10.1016/j.cell.2015.12.049
    [47] Huang ZY, Wu Y, Hedrick N, et al. (2003) T-cadherin-mediated cell growth regulation involves G2 phase arrest and requires p21(CIP1/WAF1) expression. Mol Cell Biol 23: 566-578. doi: 10.1128/MCB.23.2.566-578.2003
    [48] Matsuda K, Fujishima Y, Maeda N, et al. (2015) Positive feedback regulation between adiponectin and T-cadherin impacts adiponectin levels in tissue and plasma of male mice. Endocrinology 156: 934-946. doi: 10.1210/en.2014-1618
    [49] Niermann T, Kern F, Erne P, et al. (2000) The glycosyl phosphatidylinositol anchor of human T-cadherin binds lipoproteins. Biochem Biophys Res Commun 276: 1240-1247. doi: 10.1006/bbrc.2000.3465
    [50] Joshi MB, Philippova M, Ivanov D, et al. (2005) T-cadherin protects endothelial cells from oxidative stress-induced apoptosis. FASEB J 19: 1737-1739.
    [51] Joshi MB, Ivanov D, Philippova M, et al. (2007) Integrin-linked kinase is an essential mediator for T-cadherin-dependent signaling via Akt and GSK3beta in endothelial cells. FASEB J 21: 3083-3095. doi: 10.1096/fj.06-7723com
    [52] Philippova M, Ivanov D, Joshi MB, et al. (2008) Identification of proteins associating with glycosylphosphatidylinositol- anchored T-cadherin on the surface of vascular endothelial cells: role for Grp78/BiP in T-cadherin-dependent cell survival. Mol Cell Biol 28: 4004-4017. doi: 10.1128/MCB.00157-08
    [53] Kyriakakis E, Philippova M, Joshi MB, et al. (2010) T-cadherin attenuates the PERK branch of the unfolded protein response and protects vascular endothelial cells from endoplasmic reticulum stress-induced apoptosis. Cell Signal 22: 1308-1316. doi: 10.1016/j.cellsig.2010.04.008
    [54] Resink TJ, Kuzmenko YS, Kern F, et al. (1999) LDL binds to surface-expressed human T-cadherin in transfected HEK293 cells and influences homophilic adhesive interactions. FEBS Lett 463: 29-34. doi: 10.1016/S0014-5793(99)01594-X
    [55] Rubina K, Talovskaya E, Cherenkov V, et al. (2005) LDL induces intracellular signalling and cell migration via atypical LDL-binding protein T-cadherin. Mol Cell Biochem 273: 33-41. doi: 10.1007/s11010-005-0250-5
    [56] Kipmen-Korgun D, Osibow K, Zoratti C, et al. (2005) T-cadherin mediates low-density lipoprotein-initiated cell proliferation via the Ca(2+)-tyrosine kinase-Erk1/2 pathway. J Cardiovasc Pharmacol 45: 418-430. doi: 10.1097/01.fjc.0000157458.91433.86
    [57] Ivanov D, Philippova M, Tkachuk V, et al. (2004) Cell adhesion molecule T-cadherin regulates vascular cell adhesion, phenotype and motility. Exp Cell Res 293: 207-218. doi: 10.1016/j.yexcr.2003.09.030
    [58] Ivanov D, Philippova M, Allenspach R, et al. (2004) T-cadherin upregulation correlates with cell-cycle progression and promotes proliferation of vascular cells. Cardiovasc Res 64: 132-143. doi: 10.1016/j.cardiores.2004.06.010
    [59] Philippova M, Banfi A, Ivanov D, et al. (2006) Atypical GPI-anchored T-cadherin stimulates angiogenesis in vitro and in vivo. Arterioscler Thromb Vasc Biol 26: 2222-2230. doi: 10.1161/01.ATV.0000238356.20565.92
    [60] Frismantiene A, Pfaff D, Frachet A, et al. (2014) Regulation of contractile signaling and matrix remodeling by T-cadherin in vascular smooth muscle cells: constitutive and insulin-dependent effects. Cell Signal 26: 1897-1908. doi: 10.1016/j.cellsig.2014.05.001
    [61] Kostopoulos CG, Spiroglou SG, Varakis JN, et al. (2014) Adiponectin/T-cadherin and apelin/APJ expression in human arteries and periadventitial fat: implication of local adipokine signaling in atherosclerosis? Cardiovasc Pathol 23: 131-138. doi: 10.1016/j.carpath.2014.02.003
    [62] Fujishima Y, Maeda N, Matsuda K, et al. (2017) Adiponectin association with T-cadherin protects against neointima proliferation and atherosclerosis. FASEB J.
    [63] Philippova M, Suter Y, Toggweiler S, et al. (2011) T-cadherin is present on endothelial microparticles and is elevated in plasma in early atherosclerosis. Eur Heart J 32: 760-771. doi: 10.1093/eurheartj/ehq206
    [64] Tyrberg B, Miles P, Azizian KT, et al. (2011) T-cadherin (Cdh13) in association with pancreatic beta-cell granules contributes to second phase insulin secretion. Islets 3: 327-337. doi: 10.4161/isl.3.6.17705
    [65] Andreeva AV, Kutuzov MA (2010) Cadherin 13 in cancer. Genes Chromosomes Cancer 49: 775-790.
    [66] Kong DD, Yang J, Li L, et al. (2015) T-cadherin association with clinicopathological features and prognosis in axillary lymph node-positive breast cancer. Breast Cancer Res Treat 150: 119-126. doi: 10.1007/s10549-015-3302-x
    [67] Lee SW (1996) H-cadherin, a novel cadherin with growth inhibitory functions and diminished expression in human breast cancer. Nat Med 2: 776-782. doi: 10.1038/nm0796-776
    [68] Toyooka KO, Toyooka S, Virmani AK, et al. (2001) Loss of expression and aberrant methylation of the CDH13 (H-cadherin) gene in breast and lung carcinomas. Cancer Res 61: 4556-4560.
    [69] Miki Y, Katagiri T, Nakamura Y (1997) Infrequent mutation of the H-cadherin gene on chromosome 16q24 in human breast cancers. Jpn J Cancer Res 88: 701-704. doi: 10.1111/j.1349-7006.1997.tb00439.x
    [70] Celebiler Cavusoglu A, Kilic Y, Saydam S, et al. (2009) Predicting invasive phenotype with CDH1, CDH13, CD44, and TIMP3 gene expression in primary breast cancer. Cancer Sci 100: 2341-2345. doi: 10.1111/j.1349-7006.2009.01333.x
    [71] Toyooka S, Toyooka KO, Harada K, et al. (2002) Aberrant methylation of the CDH13 (H-cadherin) promoter region in colorectal cancers and adenomas. Cancer Res 62: 3382-3386.
    [72] Wei B, Shi H, Lu X, et al. (2015) Association between the expression of T-cadherin and vascular endothelial growth factor and the prognosis of patients with gastric cancer. Mol Med Rep 12: 2075-2081.
    [73] Hibi K, Kodera Y, Ito K, et al. (2004) Methylation pattern of CDH13 gene in digestive tract cancers. Br J Cancer 91: 1139-1142.
    [74] Hibi K, Nakayama H, Kodera Y, et al. (2004) CDH13 promoter region is specifically methylated in poorly differentiated colorectal cancer. Br J Cancer 90: 1030-1033. doi: 10.1038/sj.bjc.6601647
    [75] Scarpa M, Scarpa M, Castagliuolo I, et al. (2016) Aberrant gene methylation in non-neoplastic mucosa as a predictive marker of ulcerative colitis-associated CRC. Oncotarget 7: 10322-10331.
    [76] Ren JZ, Huo JR (2012) Correlation between T-cadherin gene expression and aberrant methylation of T-cadherin promoter in human colon carcinoma cells. Med Oncol 29: 915-918. doi: 10.1007/s12032-011-9836-9
    [77] Zhong Y, Delgado Y, Gomez J, et al. (2001) Loss of H-cadherin protein expression in human non-small cell lung cancer is associated with tumorigenicity. Clin Cancer Res 7: 1683-1687.
    [78] Brock MV, Hooker CM, Ota-Machida E, et al. (2008) DNA methylation markers and early recurrence in stage I lung cancer. N Engl J Med 358: 1118-1128. doi: 10.1056/NEJMoa0706550
    [79] Zhou S, Matsuyoshi N, Liang SB, et al. (2002) Expression of T-cadherin in Basal keratinocytes of skin. J Invest Dermatol 118: 1080-1084. doi: 10.1046/j.1523-1747.2002.01795.x
    [80] Takeuchi T, Liang SB, Matsuyoshi N, et al. (2002) Loss of T-cadherin (CDH13, H-cadherin) expression in cutaneous squamous cell carcinoma. Lab Invest 82: 1023-1029. doi: 10.1097/01.LAB.0000025391.35798.F1
    [81] Mukoyama Y, Zhou S, Miyachi Y, et al. (2005) T-cadherin negatively regulates the proliferation of cutaneous squamous carcinoma cells. J Invest Dermatol 124: 833-838. doi: 10.1111/j.0022-202X.2005.23660.x
    [82] Mukoyama Y, Utani A, Matsui S, et al. (2007) T-cadherin enhances cell-matrix adhesiveness by regulating beta1 integrin trafficking in cutaneous squamous carcinoma cells. Genes Cells 12: 787-796.
    [83] Pfaff D, Philippova M, Buechner SA, et al. (2010) T-cadherin loss induces an invasive phenotype in human keratinocytes and squamous cell carcinoma (SCC) cells in vitro and is associated with malignant transformation of cutaneous SCC in vivo. Br J Dermatol 163: 353-363. doi: 10.1111/j.1365-2133.2010.09801.x
    [84] Kyriakakis E, Maslova K, Philippova M, et al. (2012) T-Cadherin is an auxiliary negative regulator of EGFR pathway activity in cutaneous squamous cell carcinoma: impact on cell motility. J Invest Dermatol 132: 2275-2285. doi: 10.1038/jid.2012.131
    [85] Philippova M, Pfaff D, Kyriakakis E, et al. (2013) T-cadherin loss promotes experimental metastasis of squamous cell carcinoma. Eur J Cancer 49: 2048-2058. doi: 10.1016/j.ejca.2012.12.026
    [86] Wang XD, Wang BE, Soriano R, et al. (2007) Expression profiling of the mouse prostate after castration and hormone replacement: implication of H-cadherin in prostate tumorigenesis. Differentiation 75: 219-234. doi: 10.1111/j.1432-0436.2006.00135.x
    [87] Dasen B, Vlajnic T, Mengus C, et al. (2016) T-cadherin in prostate cancer: relationship with cancer progression, differentiation and drug resistance. J Pathol Clin Res 3: 44-57.
    [88] Thomas G, Jacobs KB, Yeager M, et al. (2008) Multiple loci identified in a genome-wide association study of prostate cancer. Nat Genet 40: 310-315. doi: 10.1038/ng.91
    [89] Maslova K, Kyriakakis E, Pfaff D, et al. (2015) EGFR and IGF-1R in regulation of prostate cancer cell phenotype and polarity: opposing functions and modulation by T-cadherin. FASEB J 29: 494-507. doi: 10.1096/fj.14-249367
    [90] Lin Y, Sun G, Liu X, et al. (2011) Clinical significance of T-cadherin tissue expression in patients with bladder transitional cell carcinoma. Urol Int 86: 340-345. doi: 10.1159/000322962
    [91] Lin YL, Liu XQ, Li WP, et al. (2012) Promoter methylation of H-cadherin is a potential biomarker in patients with bladder transitional cell carcinoma. Int Urol Nephrol 44: 111-117. doi: 10.1007/s11255-011-9961-6
    [92] Lin YL, Sun G, Liu XQ, et al. (2011) Clinical significance of CDH13 promoter methylation in serum samples from patients with bladder transitional cell carcinoma. J Int Med Res 39: 179-186. doi: 10.1177/147323001103900119
    [93] Lin YL, Xie PG, Ma JG (2014) Aberrant methylation of CDH13 is a potential biomarker for predicting the recurrence and progression of non muscle invasive bladder cancer. Med Sci Monit 20: 1572-1577. doi: 10.12659/MSM.892130
    [94] Lin YL, He ZK, Li ZG, et al. (2013) Downregulation of CDH13 expression promotes invasiveness of bladder transitional cell carcinoma. Urol Int 90: 225-232. doi: 10.1159/000345054
    [95] Roman-Gomez J, Castillejo JA, Jimenez A, et al. (2003) Cadherin-13, a mediator of calcium-dependent cell-cell adhesion, is silenced by methylation in chronic myeloid leukemia and correlates with pretreatment risk profile and cytogenetic response to interferon alfa. J Clin Oncol 21: 1472-1479. doi: 10.1200/JCO.2003.08.166
    [96] Sakai M, Hibi K, Koshikawa K, et al. (2004) Frequent promoter methylation and gene silencing of CDH13 in pancreatic cancer. Cancer Sci 95: 588-591. doi: 10.1111/j.1349-7006.2004.tb02491.x
    [97] Jee SH, Sull JW, Lee JE, et al. (2010) Adiponectin concentrations: a genome-wide association study. Am J Hum Genet 87: 545-552. doi: 10.1016/j.ajhg.2010.09.004
    [98] Choi JR, Jang Y, Kim Yoon S, et al. (2015) The Impact of CDH13 Polymorphism and Statin Administration on TG/HDL Ratio in Cardiovascular Patients. Yonsei Med J 56: 1604-1612. doi: 10.3349/ymj.2015.56.6.1604
    [99] Wu Y, Li Y, Lange EM, et al. (2010) Genome-wide association study for adiponectin levels in Filipino women identifies CDH13 and a novel uncommon haplotype at KNG1-ADIPOQ. Hum Mol Genet 19: 4955-4964. doi: 10.1093/hmg/ddq423
    [100] Nicolas A, Aubert R, Bellili-Munoz N, et al. (2016) T-cadherin gene variants are associated with type 2 diabetes and the Fatty Liver Index in the French population. Diabetes Metab.
    [101] Chung CM, Lin TH, Chen JW, et al. (2011) A genome-wide association study reveals a quantitative trait locus of adiponectin on CDH13 that predicts cardiometabolic outcomes. Diabetes 60: 2417-2423. doi: 10.2337/db10-1321
    [102] Fava C, Danese E, Montagnana M, et al. (2011) A variant upstream of the CDH13 adiponectin receptor gene and metabolic syndrome in Swedes. Am J Cardiol 108: 1432-1437. doi: 10.1016/j.amjcard.2011.06.068
    [103] Park J, Kim I, Jung KJ, et al. (2015) Gene-gene interaction analysis identifies a new genetic risk factor for colorectal cancer. J Biomed Sci 22: 73. doi: 10.1186/s12929-015-0180-9
    [104] Kudrjashova E, Bashtrikov P, Bochkov V, et al. (2002) Expression of adhesion molecule T-cadherin is increased during neointima formation in experimental restenosis. Histochem Cell Biol 118: 281-290.
  • Reader Comments
  • © 2017 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(5318) PDF downloads(1421) Cited by(8)

Article outline

Figures and Tables

Figures(1)  /  Tables(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog