Artículo

Estamos trabajando para incorporar este artículo al repositorio
Consulte el artículo en la página del editor
Consulte la política de Acceso Abierto del editor

Abstract:

DMD gene mutations have been associated with the development of Dystrophinopathies. Interestingly, it has been recently reported that DMD is involved in the development and progression of myogenic tumors, assigning DMD a tumor suppressor activity in these types of cancer. However, there are only few reports that analyze DMD in non-myogenic tumors. Our study was designed to examine DMD expression and genetic alterations in non-myogenic tumors using public repositories. We also evaluated the overall survival of patients with and without DMD mutations. We studied 59 gene expression microarrays (GEO database) and RNAseq (cBioPortal) datasets that included 9817 human samples. We found reduced DMD expression in 15/27 (56%) pairwise comparisons performed (Fold-Change (FC) ≤ 0.70; p-value range = 0.04-1.5x10-20). The analysis of RNAseq studies revealed a median frequency of DMD genetic alterations of 3.4%, higher or similar to other well-known tumor suppressor genes. In addition, we observed significant poorer overall survival for patients with DMD mutations. The analyses of paired tumor/normal tissues showed that the majority of tumor specimens had lower DMD expression compared to their normal adjacent counterpart. Interestingly, statistical significant over-expression of DMD was found in 6/27 studies (FC ≥ 1.4; p-value range = 0.03-3.4x10-15). These results support that DMD expression and genetic alterations are frequent and relevant in non-myogenic tumors. The study and validation of DMD as a new player in tumor development and as a new prognostic factor for tumor progression and survival are warranted.

Registro:

Documento: Artículo
Título:Non-myogenic tumors display altered expression of dystrophin (DMD) and a high frequency of genetic alterations
Autor:Luce, L.N.; Abbate, M.; Cotignola, J.; Giliberto, F.
Filiación:INIGEM, CONICET/Cátedra de Genética y Biología Molecular, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Argentina
IQUIBICEN, CONICET/Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina
Palabras clave:DMD; Dystrophin; Gene expression; Genetic alteration; Survival; dystrophin; Article; benign neoplasm; breast cancer; cancer growth; cancer prognosis; cancer survival; controlled study; disease association; DMD gene; gene expression regulation; gene identification; gene mutation; gene overexpression; genetic association; genetic disorder; microarray analysis; mutational analysis; non myogenic tumor; overall survival; RNA sequence; survival rate; survival time; tumor suppressor gene; uterus cancer
Año:2017
Volumen:8
Número:1
Página de inicio:145
Página de fin:155
DOI: http://dx.doi.org/10.18632/oncotarget.10426
Título revista:Oncotarget
Título revista abreviado:Oncotarget
ISSN:19492553
CAS:dystrophin, 116978-02-0
Registro:https://bibliotecadigital.exactas.uba.ar/collection/paper/document/paper_19492553_v8_n1_p145_Luce

Referencias:

  • Blake, D.J., Weir, A., Newey, S.E., Davies, K.E., Function and genetics of dystrophin and dystrophin-related proteins in muscle (2002) Physiol Rev, 82, pp. 291-329
  • Sgambato, A., Migaldi, M., Montanari, M., Camerini, A., Brancaccio, A., Rossi, G., Cangiano, R., Cittadini, A., Dystroglycan expression is frequently reduced in human breast and colon cancers and is associated with tumor progression (2003) Am J Pathol, 162, pp. 849-860
  • Korner, H., Epanchintsev, A., Berking, C., Schuler-Thurner, B., Speicher, M.R., Menssen, A., Hermeking, H., Digital karyotyping reveals frequent inactivation of the dystrophin/DMD gene in malignant melanoma (2007) Cell Cycle, 6, pp. 189-198
  • Schmidt, W.M., Uddin, M.H., Dysek, S., Moser-Thier, K., Pirker, C., Hoger, H., Ambros, I.M., Bittner, R.E., DNA damage, somatic aneuploidy, and malignant sarcoma susceptibility in muscular dystrophies (2011) PLoS genetics, 7
  • Hosur, V., Kavirayani, A., Riefler, J., Carney, L.M., Lyons, B., Gott, B., Cox, G.A., Shultz, L.D., Dystrophin and dysferlin double mutant mice: a novel model for rhabdomyosarcoma (2012) Cancer genetics, 205, pp. 232-241
  • Wang, Y., Marino-Enriquez, A., Bennett, R.R., Zhu, M., Shen, Y., Eilers, G., Lee, J.C., Fletcher, C.D., Dystrophin is a tumor suppressor in human cancers with myogenic programs (2014) Nat Genet, 46, pp. 601-606
  • Barrett, T., Wilhite, S.E., Ledoux, P., Evangelista, C., Kim, I.F., Tomashevsky, M., Marshall, K.A., Davis, S., NCBI GEO: archive for functional genomics data sets update (2013) Nucleic acids research, 41, pp. D991-D995
  • Edgar, R., Domrachev, M., Lash, A.E., Gene Expression Omnibus: NCBI gene expression and hybridization array data repository (2002) Nucleic acids research, 30, pp. 207-210
  • Gentleman, R.C., Carey, V.J., Bates, D.M., Bolstad, B., Dettling, M., Dudoit, S., Ellis, B., Huber, W., Bioconductor: open software development for computational biology and bioinformatics (2004) Genome biology, 5, p. R80
  • Huber, W., Carey, V.J., Gentleman, R., Anders, S., Carlson, M., Carvalho, B.S., Bravo, H.C., Hansen, K.D., Orchestrating highthroughput genomic analysis with Bioconductor (2015) Nature methods, 12, pp. 115-121
  • Irizarry, R.A., Hobbs, B., Collin, F., Beazer-Barclay, Y.D., Antonellis, K.J., Scherf, U., Speed, T.P., Exploration, normalization, and summaries of high density oligonucleotide array probe level data (2003) Biostatistics, 4, pp. 249-264
  • Ritchie, M.E., Phipson, B., Wu, D., Hu, Y., Law, C.W., Shi, W., Smyth, G.K., limma powers differential expression analyses for RNA-sequencing and microarray studies (2015) Nucleic acids research, 43
  • Cerami, E., Gao, J., Dogrusoz, U., Gross, B.E., Sumer, S.O., Aksoy, B.A., Jacobsen, A., Schultz, N., The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data (2012) Cancer discovery, 2, pp. 401-404
  • Gao, J., Aksoy, B.A., Dogrusoz, U., Dresdner, G., Gross, B., Sumer, S.O., Sun, Y., Schultz, N., Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal (2013) Science signaling, 6
  • Pantaleo, M.A., Astolfi, A., Urbini, M., Fuligni, F., Saponara, M., Nannini, M., Lolli, C., Biasco, G., Dystrophin deregulation is associated with tumor progression in KIT/PDGFRA mutant gastrointestinal stromal tumors (2014) Clinical sarcoma research, 4, p. 9
  • Stephens, N.A., Skipworth, R.J., Gallagher, I.J., Greig, C.A., Guttridge, D.C., Ross, J.A., Fearon, K.C., Evaluating potential biomarkers of cachexia and survival in skeletal muscle of upper gastrointestinal cancer patients (2015) Journal of cachexia, sarcopenia and muscle, 6, pp. 53-61
  • Nikitin, E.A., Malakho, S.G., Biderman, B.V., Baranova, A.V., Lorie, Y.Y., Shevelev, A.Y., Peklo, M.M., Sudarikov, A.B., Expression level of lipoprotein lipase and dystrophin genes predict survival in B-cell chronic lymphocytic leukemia (2007) Leukemia & lymphoma, 48, pp. 912-922
  • Tan, S., Tan, S., Chen, Z., Cheng, K., Chen, Z., Wang, W., Wen, Q., Zhang, W., Knocking down Dp71 expression in A549 cells reduces its malignancy in vivo and in vitro (2016) Cancer investigation, 34, pp. 16-25
  • Talantov, D., Mazumder, A., Yu, J.X., Briggs, T., Jiang, Y., Backus, J., Atkins, D., Wang, Y., Novel genes associated with malignant melanoma but not benign melanocytic lesions (2005) Clin Cancer Res, 11, pp. 7234-7242
  • Chandran, U.R., Ma, C., Dhir, R., Bisceglia, M., Lyons-Weiler, M., Liang, W., Michalopoulos, G., Monzon, F.A., Gene expression profiles of prostate cancer reveal involvement of multiple molecular pathways in the metastatic process (2007) BMC Cancer, 7, p. 64
  • Yu, Y.P., Landsittel, D., Jing, L., Nelson, J., Ren, B., Liu, L., McDonald, C., Luo, J.H., Gene expression alterations in prostate cancer predicting tumor aggression and preceding development of malignancy (2004) J Clin Oncol, 22, pp. 2790-2799
  • Landi, M.T., Dracheva, T., Rotunno, M., Figueroa, J.D., Liu, H., Dasgupta, A., Mann, F.E., Pesatori, A.C., Gene expression signature of cigarette smoking and its role in lung adenocarcinoma development and survival (2008) PLoS One, 3
  • Lu, T.P., Tsai, M.H., Lee, J.M., Hsu, C.P., Chen, P.C., Lin, C.W., Shih, J.Y., Chuang, E.Y., Identification of a novel biomarker, SEMA5A, for non-small cell lung carcinoma in nonsmoking women (2010) Cancer Epidemiol Biomarkers Prev, 19, pp. 2590-2597
  • Kabbout, M., Garcia, M.M., Fujimoto, J., Liu, D.D., Woods, D., Chow, C.W., Mendoza, G., Kadara, H., ETS2 mediated tumor suppressive function and MET oncogene inhibition in human non-small cell lung cancer (2013) Clin Cancer Res, 19, pp. 3383-3395
  • Casey, T., Bond, J., Tighe, S., Hunter, T., Lintault, L., Patel, O., Eneman, J., Plaut, K., Molecular signatures suggest a major role for stromal cells in development of invasive breast cancer (2009) Breast Cancer Res Treat, 114, pp. 47-62
  • Merdad, A., Karim, S., Schulten, H.J., Dallol, A., Buhmeida, A., Al-Thubaity, F., Gari, M.A., Al-Qahtani, M.H., Expression of matrix metalloproteinases (MMPs) in primary human breast cancer: MMP-9 as a potential biomarker for cancer invasion and metastasis (2014) Anticancer Res, 34, pp. 1355-1366
  • Badea, L., Herlea, V., Dima, S.O., Dumitrascu, T., Popescu, I., Combined gene expression analysis of whole-tissue and microdissected pancreatic ductal adenocarcinoma identifies genes specifically overexpressed in tumor epithelia (2008) Hepatogastroenterology, 55, pp. 2016-2027
  • Sole, X., Crous-Bou, M., Cordero, D., Olivares, D., Guino, E., Sanz-Pamplona, R., Rodriguez-Moranta, F., Moreno, V., Discovery and validation of new potential biomarkers for early detection of colon cancer (2014) PLoS One, 9
  • Closa, A., Cordero, D., Sanz-Pamplona, R., Sole, X., Crous-Bou, M., Pare-Brunet, L., Berenguer, A., Moreno, V., Identification of candidate susceptibility genes for colorectal cancer through eQTL analysis (2014) Carcinogenesis, 35, pp. 2039-2046
  • Griesinger, A.M., Birks, D.K., Donson, A.M., Amani, V., Hoffman, L.M., Waziri, A., Wang, M., Foreman, N.K., Characterization of distinct immunophenotypes across pediatric brain tumor types (2013) J Immunol, 191, pp. 4880-4888
  • Brune, V., Tiacci, E., Pfeil, I., Doring, C., Eckerle, S., van Noesel, C.J., Klapper, W., Kuppers, R., Origin and pathogenesis of nodular lymphocyte-predominant Hodgkin lymphoma as revealed by global gene expression analysis (2008) The Journal of experimental medicine, 205, pp. 2251-2268
  • Giefing, M., Winoto-Morbach, S., Sosna, J., Doring, C., Klapper, W., Kuppers, R., Bottcher, S., Schutze, S., Hodgkin-Reed-Sternberg cells in classical Hodgkin lymphoma show alterations of genes encoding the NADPH oxidase complex and impaired reactive oxygen species synthesis capacity (2013) PLoS One, 8
  • Cramer-Morales, K., Nieborowska-Skorska, M., Scheibner, K., Padget, M., Irvine, D.A., Sliwinski, T., Haas, K., Wasik, M.A., Personalized synthetic lethality induced by targeting RAD52 in leukemias identified by gene mutation and expression profile (2013) Blood, 122, pp. 1293-1304
  • Gutierrez, A., Jr., Tschumper, R.C., Wu, X., Shanafelt, T.D., Eckel-Passow, J., Huddleston, P.M., III, Slager, S.L., Jelinek, D.F., LEF-1 is a prosurvival factor in chronic lymphocytic leukemia and is expressed in the preleukemic state of monoclonal B-cell lymphocytosis (2010) Blood, 116, pp. 2975-2983
  • Wang, L., Shalek, A.K., Lawrence, M., Ding, R., Gaublomme, J.T., Pochet, N., Stojanov, P., Sievers, Q.L., Somatic mutation as a mechanism of Wnt/beta-catenin pathway activation in CLL (2014) Blood, 124, pp. 1089-1098
  • Von Roemeling, C.A., Radisky, D.C., Marlow, L.A., Cooper, S.J., Grebe, S.K., Anastasiadis, P.Z., Tun, H.W., Copland, J.A., Neuronal pentraxin 2 supports clear cell renal cell carcinoma by activating the AMPA-selective glutamate receptor-4 (2014) Cancer Res, 74, pp. 4796-4810
  • Stirewalt, D.L., Meshinchi, S., Kopecky, K.J., Fan, W., Pogosova-Agadjanyan, E.L., Engel, J.H., Cronk, M.R., Radich, J.P., Identification of genes with abnormal expression changes in acute myeloid leukemia (2008) Genes, chromosomes & cancer, 47, pp. 8-20

Citas:

---------- APA ----------
Luce, L.N., Abbate, M., Cotignola, J. & Giliberto, F. (2017) . Non-myogenic tumors display altered expression of dystrophin (DMD) and a high frequency of genetic alterations. Oncotarget, 8(1), 145-155.
http://dx.doi.org/10.18632/oncotarget.10426
---------- CHICAGO ----------
Luce, L.N., Abbate, M., Cotignola, J., Giliberto, F. "Non-myogenic tumors display altered expression of dystrophin (DMD) and a high frequency of genetic alterations" . Oncotarget 8, no. 1 (2017) : 145-155.
http://dx.doi.org/10.18632/oncotarget.10426
---------- MLA ----------
Luce, L.N., Abbate, M., Cotignola, J., Giliberto, F. "Non-myogenic tumors display altered expression of dystrophin (DMD) and a high frequency of genetic alterations" . Oncotarget, vol. 8, no. 1, 2017, pp. 145-155.
http://dx.doi.org/10.18632/oncotarget.10426
---------- VANCOUVER ----------
Luce, L.N., Abbate, M., Cotignola, J., Giliberto, F. Non-myogenic tumors display altered expression of dystrophin (DMD) and a high frequency of genetic alterations. Oncotarget. 2017;8(1):145-155.
http://dx.doi.org/10.18632/oncotarget.10426