Hostname: page-component-8448b6f56d-mp689 Total loading time: 0 Render date: 2024-04-19T05:03:50.675Z Has data issue: false hasContentIssue false

Expression Profile and Subcellular Localization of GAPDH in the Smooth Muscle Cells of Human Atherosclerotic Plaque: An Immunohistochemical and Ultrastructural Study with Biological Therapeutic Perspectives

Published online by Cambridge University Press:  23 May 2014

Ida Perrotta*
Affiliation:
Department of Biology, Ecology and Earth Science (Di.B.E.S.T.), University of Calabria – Arcavacata, Rende 87036, Cosenza, Italy
Saveria Aquila
Affiliation:
Centro Sanitario – Department of Pharmacy and Sciences of Health and Nutrition, University of Calabria – Arcavacata, Rende 87036, Cosenza, Italy
Sergio Mazzulla
Affiliation:
Department of Biology, Ecology and Earth Science (Di.B.E.S.T.), University of Calabria – Arcavacata, Rende 87036, Cosenza, Italy
*
*Corresponding author. idaperrotta@yahoo.it
Get access

Abstract

Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) has long been considered a classical glycolytic enzyme involved exclusively in cytosolic energy production. Several recent studies, however, have demonstrated that GAPDH is a multifunctional protein whose presence and activity can be regulated by disease states and/or experimental manipulation. Expression levels of GAPDH have been shown to be altered in certain tumors as well as in proliferating and differentiating cells. Since dedifferentiation and proliferation of smooth muscle cells (SMCs) are important features of human atherosclerosis, we have characterized the expression profile of GAPDH in the SMCs of atherosclerotic plaques and its putative interrelationship with the synthetic/proliferative status of these cells utilizing the proliferating cell nuclear antigen (PCNA) antibody, a valuable marker of cell proliferation. Western blot data revealed that GAPDH was significantly upregulated in atherosclerotic plaque specimens. Immunohistochemical stains demonstrated that GAPDH accumulated in the nucleus of dedifferentiated SMCs that also showed positive immunoreactivity for PCNA, but remained cytoplasmatic in the contractile SMCs (PCNA-negative), thus reflecting the proliferative, structural and synthetic differences between them. We suggest that, in human atherosclerotic plaque, GAPDH might exert additional functions that are independent of its well-documented glycolytic activity and might play key roles in development of the disease.

Type
Biological Applications
Copyright
© Microscopy Society of America 2014 

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Aledo, J.C., Segura, J.A., Barbero, L.G. & Marquez, J. (1999). Upregulation of glyceraldehyde-3-phosphate dehydrogenase mRNA in the spleen of tumor-bearing mice. Biochimie 81, 11091113.Google Scholar
Barber, R.D., Harmer, D.W., Coleman, R.A. & Clark, B.J. (2005). GAPDH as a housekeeping gene: analysis of GAPDH mRNA expression in a panel of 72 human tissues. Physiol Genomics 21, 389395.Google Scholar
Bhatia, P., Taylor, W.R., Greenberg, A.H. & Wright, J.A. (1994). Comparison of glyceraldehyde-3-phosphate dehydrogenase and 28S-ribosomal RNA gene expression as RNA loading controls for northern blot analysis of cell lines of varying malignant potential. Anal Biochem 216, 223226.CrossRefGoogle ScholarPubMed
Buhay, J.E., Moni, G., Mann, N. & Crandall, K.A. (2007). A molecular taxonomy in the dark: Evolutionary history, phylogeography, and diversity of cave crayfish in the subgenus Aviticambarus, genus Cambarus. Mol Phylogenet Evol 42, 435448.CrossRefGoogle ScholarPubMed
Butte, A.J., Dzau, V.J. & Glueck, S.B. (2001). Further defining housekeeping, or “maintenance,” genes Focus on “A compendium of gene expression in normal human tissues”. Physiol Genomics 7, 9596.Google Scholar
Canback, B., Andersson, S.G. & Kurland, C.G. (2002). The global phylogeny of glycolytic enzymes. Proc Natl Acad Sci USA 99, 60976102.Google Scholar
Chang, T.J., Juan, C.C., Yin, P.H., Chi, C.W. & Tsay, H.J. (1998). Up-regulation of beta-actin, cyclophilin and GAPDH in N1S1 rat hepatoma. Oncol Rep 5, 469471.Google Scholar
Colell, A., Green, D.R. & Ricci, J.E. (2009). Novel roles for GAPDH in cell death and carcinogenesis. Cell Death Differ 16, 15731581.Google Scholar
Dang, C.V. (2012). Links between metabolism and cancer. Genes Dev 26, 877890.Google Scholar
de Jonge, H.J.M., Fehrmann, R.S.N., de Bont, E.S.J.M., Hofstra, R.M.W., Gerbens, F., Kamps, W.A., de Vries, E.G.E., van der Zee, A.G.J., te Meerman, G.J. & ter Elst, A. (2007). Evidence based selection of housekeeping genes. PLoS One 2, e898.Google Scholar
Eisenberg, E. & Levanon, E.Y. (2013). Human housekeeping genes, revisited. Trends Genet 29, 569574.Google Scholar
Epner, D.E., Sawa, A. & Isaacs, J.T. (1999). Glyceraldehyde-3-phosphate dehydrogenase expression during apoptosis and proliferation of rat ventral prostate. Biol Reprod 61, 687691.Google Scholar
Glare, E.M., Divjak, M., Bailey, M.J. & Walters, E.H. (2002). Beta-actin and GAPDH housekeeping gene expression in asthmatic airways is variable and not suitable for normalising mRNA levels. Thorax 57, 765770.Google Scholar
Goldsworthy, S.M., Goldsworthy, T.L., Sprankle, C.S. & Butterworth, B.E. (1993). Variation in expression of genes used for normalization of Northern blots after induction of cell proliferation. Cell Prolif 26, 511518.Google Scholar
Gomez, D. & Owens, G.K. (2012). Smooth muscle cell phenotypic switching in atherosclerosis. Cardiovasc Res 95, 156164.Google Scholar
Gordon, G.B., Bush, D.E. & Weisman, H. (1988). Reduction of atherosclerosis by administration of dehydroepiandrosterone. A study in the hypercholesterolemic New Zealand white rabbit with aortic intimal injury. J Clin Invest 82, 712720.Google Scholar
Higashimura, Y., Nakajima, Y., Yamaji, R., Harada, N., Shibasaki, F., Nakano, Y. & Inui, H. (2011). Up-regulation of glyceraldehyde-3-phosphate dehydrogenase gene expression by HIF-1 activity depending on Sp1 in hypoxic breast cancer cells. Arch Biochem Biophys 509, 18.CrossRefGoogle ScholarPubMed
Jögi, A., Øra, I., Nilsson, H., Lindeheim, A., Makino, Y., Poellinger, L., Axelson, H. & Påhlman, S. (2002). Hypoxia alters gene expression in human neuroblastoma cells toward an immature and neural crest-like phenotype. Proc Natl Acad Sci USA 99, 70217026.Google Scholar
Kim, J.W., Kim, T.E., Kim, Y.K., Kim, Y.W., Kim, S.J., Lee, J.M., Kim, I.K. & Namkoong, S.E. (1999). Antisense oligodeoxynucleotide of glyceraldehyde-3-phosphate dehydrogenase gene inhibits cell proliferation and induces apoptosis in human cervical carcinoma cell lines. Antisense Nucleic Acid Drug Dev 9, 507513.Google Scholar
Kondo, S., Kubota, S., Mukudai, Y., Nishida, T., Yoshihama, Y., Shirota, T., Shintani, S. & Takigawa, M. (2011). Binding of glyceraldehyde-3-phosphate dehydrogenase to the cis-acting element of structure-anchored repression in ccn2 mRNA. Biochem Biophys Res Commun 405, 382387.Google Scholar
Lawrence, J.G., Hartl, D.L. & Ochman, H. (1991). Molecular considerations in the evolution of bacterial genes. J Mol Evol 33, 241250.Google Scholar
Li, H., Chen, T., Cottrell, J. & Wang, H. (2009). Nuclear translocation of adenoviral-enhanced yellow fluorescent protein-tagged-human constitutive androstane receptor (hCAR): A novel tool for screening hCAR activators in human primary hepatocytes. Drug Metab Dispos 37, 10981106.Google Scholar
Mansur, N.R., Meyer-Siegler, K., Wurzer, J.C. & Sirover, M.A. (1993). Cell cycle regulation of the glyceraldehyde-3-phosphate dehydrogenase/uracil DNA glycosylase gene in normal human cells. Nucleic Acids Res 21, 993998.Google Scholar
Mazzola, J.L. & Sirover, M.A. (2003). Subcellular localization of human glyceraldehyde-3-phosphate dehydrogenase is independent of its glycolytic function. Biochim Biophys Acta 1622, 5056.Google Scholar
Meyer-Siegler, K., Rahman-Mansur, N., Wurzer, J.C. & Sirover, M.A. (1992). Proliferative dependent regulation of the glyceraldehyde-3-phosphate dehydrogenase/uracil DNA glycosylase gene in human cells. Carcinogenesis 13, 21272132.Google Scholar
Nicholls, C., Pinto, A.R., Li, H., Li, L., Wang, L., Simpson, R. & Liu, J.P. (2012). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) induces cancer cell senescence by interacting with telomerase RNA component. Proc Natl Acad Sci USA 109, 1330813313.Google Scholar
Orekhov, A.N., Andreeva, E.R., Mikhailova, I.A. & Gordon, D. (1998). Cell proliferation in normal and atherosclerotic human aorta: Proliferative splash in lipid-rich lesions. Atherosclerosis 139, 4148.Google Scholar
Park, K.W., Kim, D.H., You, H.J. Sir, J.J., Jeon, S.I., Youn, S.W., Yang, H.M., Skurk, C., Park, Y.B., Walsh, K. & Kim, H.S. (2005). Activated forkhead transcription factor inhibits neointimal hyperplasia after angioplasty through induction of p27. Arterioscler Thromb Vasc Biol 25, 742747.Google Scholar
Perrotta, I. (2013). The use of electron microscopy for the detection of autophagy in human atherosclerosis. Micron 50, 713.Google Scholar
Perrotta, I., Perrotta, E., Sesti, S., Cassese, M. & Mazzulla, S. (2013). MnSOD expression in human atherosclerotic plaques: An immunohistochemical and ultrastructural study. Cardiovasc Pathol 22, 428437.Google Scholar
Perrotta, I., Sciangula, A., Perrotta, E., Donato, G. & Cassese, M. (2011). Ultrastructural analysis and electron microscopic localization of Nox4 in healthy and atherosclerotic human aorta. Ultrastruct Pathol 35, 16.Google Scholar
Persons, D.A., Schek, N., Hall, B.L. & Finn, O.J. (1989). Increased expression of glycolysis-associated genes in oncogene-transformed and growth-accelerated states. Mol Carcinog 2, 8894.Google Scholar
Phadke, M.S., Krynetskaia, N.F., Mishra, A.K. & Krynetskiy, E. (2009). Glyceraldehyde 3-phosphate dehydrogenase depletion induces cell cycle arrest and resistance to antimetabolites in human carcinoma cell lines. J Pharmacol Exp Ther 331, 7786.Google Scholar
Ranganna, K. & Yatsu, F.M. (1997). Inhibition of platelet-derived growth factor BB-induced expression of glyceraldehyde-3-phosphate dehydrogenase by sodium butyrate in rat vascular smooth muscle cells. Arterioscler Thromb Vasc Biol 17, 34203427.Google Scholar
Ripple, M.O. & Wilding, G. (1995). Alteration of glyceraldehyde-3-phosphate dehydrogenase activity and messenger RNA content by androgen in human prostate carcinoma cells. Cancer Res 55, 42344236.Google ScholarPubMed
Rivard, A. & Andrés, V. (2000). Vascular smooth muscle cell proliferation in the pathogenesis of atherosclerotic cardiovascular diseases. Histol Histopathol 15, 557571.Google Scholar
Rondinelli, R.H., Epner, D.E. & Tricoli, J.V. (1997). Increased glyceraldehyde-3-phosphate dehydrogenase gene expression in late pathological stage human prostate cancer. Prostate Cancer Prostatic Dis 1, 6672.Google Scholar
Rubie, C., Kempf, K., Hans, J., Su, T., Tilton, B., Georg, T., Brittner, B., Ludwig, B. & Schilling, M. (2005). Housekeeping gene variability in normal and cancerous colorectal, pancreatic, esophageal, gastric and hepatic tissues. Mol Cell Probes 19, 101109.Google Scholar
Ryzlak, M.T. & Pietruszko, R. (1988). Heterogeneity of glyceraldehyde-3-phosphate dehydrogenase from human brain. Biochim Biophys Acta 954, 309324.Google Scholar
Schek, N., Hall, B.L. & Finn, O.J. (1988). Increased glyceraldehyde-3- phosphate dehydrogenase gene expression in human pancreatic adenocarcinoma. Cancer Res 48, 63546359.Google Scholar
Sheng, W.Y. & Wang, T.C.V. (2009). Proteomic analysis of the differential protein expression reveals nuclear GAPDH in activated T lymphocytes. PLoS One 4, 6322.Google Scholar
Sikand, K., Singh, J., Ebron, J.S. & Shukla, G.C. (2012). Housekeeping gene selection advisory: Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) and β-actin are targets of miR-644a. PLoS One 7, e47510.Google Scholar
Sirover, M.A. (1997). Role of the glycolytic protein, glyceraldehyde-3-phosphate dehydrogenase, in normal cell function and in cell pathology. J Cell Biochem 66, 133140.Google Scholar
Sirover, M.A. (1999). New insights into an old protein: The functional diversity of mammalian glyceraldehyde-3-phosphate dehydrogenase. Biochim Biophys Acta 1432, 159184.Google Scholar
Sirover, M.A. (2005). New nuclear functions of the glycolytic protein, glyceraldehyde-3-phosphate dehydrogenase, in mammalian cells. J Cell Biochem 95, 4552.Google Scholar
Sirover, M.A. (2012). Subcellular dynamics of multifunctional protein regulation: Mechanisms of GAPDH intracellular translocation. J Cell Biochem 113, 21932200.CrossRefGoogle ScholarPubMed
Sundararaj, K.P., Wood, R.E., Ponnusamy, S., Salas, A.M., Szulc, Z., Bielawska, A., Obeid, L.M., Hannun, Y.A. & Ogretmen, B. (2004). Rapid shortening of telomere length in response to ceramide involves the inhibition of telomere binding activity of nuclear glyceraldehyde-3-phosphate dehydrogenase. J Biol Chem 279, 61526162.Google Scholar
Tang, Z., Yuan, S., Hu, Y., Zhang, H., Wu, W., Zeng, Z., Yang, J., Yun, J., Xu, R. & Huang, P. (2012). Over-expression of GAPDH in human colorectal carcinoma as a preferred target of 3-bromopyruvate propyl ester. J Bioenerg Biomembr 44, 117125.Google Scholar
Tatton, W.G., Chalmers-Redman, R.M., Elstner, M., Leesch, W., Jagodzinski, F.B., Stupak, D.P., Sugrue, M.M. & Tatton, N.A. (2000). Glyceraldehyde-3-phosphate dehydrogenase in neurodegeneration and apoptosis signaling. J Neural Transm Suppl 60, 77100.Google Scholar
Tisdale, E.J., Kelly, C. & Artalejo, C.R. (2004). Glyceraldehyde-3-phosphate dehydrogenase interacts with Rab2 and plays an essential role in endoplasmic reticulum to Golgi transport exclusive of its glycolytic activity. J Biol Chem 279, 5404654052.Google Scholar
Valenti, M.T., Bertoldo, F., Dalle Carbonare, L., Azzarello, G., Zenari, S., Zanatta, M., Balducci, E., Vinante, O. & Lo Cascio, V. (2006). The effect of bisphosphonates on gene expression: GAPDH as a housekeeping or a new target gene? BMC Cancer 6, 49.Google Scholar
Wang, D., Moothart, D.R., Lowy, D.R. & Qian, X. (2013). The expression of glyceraldehyde-3-phosphate dehydrogenase associated cell cycle (GACC) genes correlates with cancer stage and poor survival in patients with solid tumors. PLoS One 8, e61262.Google Scholar
Wong, R.S. (2011). Apoptosis in cancer: From pathogenesis to treatment. J Exp Clin Cancer Res 30, 87.Google Scholar
Zheng, L., Roeder, R.G. & Luo, Y. (2003). S phase activation of the histone H2B promoter by OCA-S, a coactivator complex that contains GAPDH as a key component. Cell 114, 255266.Google Scholar
Zhong, H. & Simons, J.W. (1999). Direct comparison of GAPDH, beta-actin, cyclophilin, and 28S rRNA as internal standards for quantifying RNA levels under hypoxia. Biochem Biophys Res Commun 259, 523526.Google Scholar