يعرض 1 - 10 نتائج من 383 نتيجة بحث عن '"Katargin A"', وقت الاستعلام: 2.17s تنقيح النتائج
  1. 1
    دورية أكاديمية
  2. 2
    دورية أكاديمية
  3. 3
    دورية أكاديمية
  4. 4
    دورية أكاديمية

    المساهمون: The research was funded by the Russian Science Foundation (grant No. 19-15-00245, https://rscf.ru/project/22-15-35008Test/)., Исследование финансировалось Российским научным фондом (грант № 19-15-00245, https://rscf.ru/project/22-15-35008Test).

    المصدر: Advances in Molecular Oncology; Том 10, № 4 (2023); 149-156 ; Успехи молекулярной онкологии; Том 10, № 4 (2023); 149-156 ; 2413-3787 ; 2313-805X ; 10.17650/2313-805X-2023-10-4

    وصف الملف: application/pdf

    العلاقة: https://umo.abvpress.ru/jour/article/view/605/322Test; Mansoori B., Mohammadi A., Davudian S. et al. The Different mechanisms of cancer drug resistance: a brief review. Adv Pharm Bull 2017;7(3):339–48. DOI:10.15171/apb.2017.041; Aleksakhina S.N., Kashyap A., Imyanitov E.N. Mechanisms of acquired tumor drug resistance. Biochim Biophys Acta Rev Cancer 2019;1872(2):188310. DOI:10.1016/j.bbcan.2019.188310; Cree I.A., Charlton P. Molecular chess? Hallmarks of anti-cancer drug resistance. BMC Cancer 2017;17(1):10. DOI:10.1186/s12885-016-2999-1; Konjevic G., Mirjacic Martinovic K., Vuletic A. et al. Low expression of CD161 and NKG2D activating NK receptor is associated with impaired NK cell cytotoxicity in metastatic melanoma patients. Clin Exp Metastasis 2007;24(1):1–11. DOI: https://doi.org/10.1007/s10585-006-9043-9Test; Scherbakov A.M., Krasil’nikov M.A., Kushlinskii N.E. Molecular mechanisms of hormone resistance of breast cancer. Bull Exp Biol Med 2013;155(3):384–95. DOI: https://doi.org/10.1007/s10517-013-2160-yTest; Clarke R., Tyson J.J., Dixon J.M. Endocrine resistance in breast cancer – an overview and update. Mol Cell Endocrinol 2015;418 Pt. 3(03):220–34. DOI: https://doi.org/10.1016/j.mce.2015.09.035Test; Araki K., Miyoshi Y. Mechanism of resistance to endocrine therapy in breast cancer: the important role of PI3K/Akt/mTOR in estrogen receptor-positive, HER2-negative breast cancer. Breast Cancer 2018;25(4)392–401. DOI: https://doi.org/10.1007/s12282-017-0812-xTest; Citi V., Del Re M., Martelli A. et al. Phosphorylation of AKT and ERK1/2 and mutations of PIK3CA and PTEN are predictive of breast cancer cell sensitivity to everolimus in vitro. Cancer Chem Pharmacol 2018;81(4):745–54. DOI: https://doi.org/10.1007/s00280-018-3543-6Test; Xie W., Sun H., Li X. et al. Ovarian cancer: epigenetics, drug resistance, and progression. Cancer Cell Int 2021;21(1):1–16. DOI: https://doi.org/10.1186/s12935-021-02136-yTest; Hazra A., Bose P., Sunita P. et al. Molecular epigenetic dynamics in breast carcinogenesis. Arch Pharm Res 2021;44(8):741–63. DOI: https://doi.org/10.1007/s12272-021-01348-0Test; Andreeva O.E., Shchegolev Y.Y., Scherbakov A.M. et al. The phenomenon of the cross-resistance of breast cancer to target and hormonal drugs: the role of epigenetic reconstruction. Med Sci Forum 2023;20(1):5. DOI: https://doi.org/10.3390/IECC2023-14220Test; Vinokurova S. Epigenetics of virus-induced tumors: perspectives for therapeutic targeting. Curr Pharm Des 2017;23(32):4842–61. DOI: https://doi.org/10.2174/1381612823666170822100627Test; Ariazi E.A., Taylor J.C., Black M.A. et al. A new role for ERalpha: silencing via DNA methylation of basal, stem cell, and emt genes. Mol Cancer Res 2017;15(2):152–64. DOI: https://doi.org/10.1158/1541-7786.MCR-16-0283Test; Lin X., Li J., Yin G. et al. Integrative analyses of gene expression and DNA methylation profiles in breast cancer cell line models of tamoxifen-resistance indicate a potential role of cells with stem-like properties. Breast Cancer Res 2013;15(6):R119. DOI: https://doi.org/10.1186/bcr3588Test; Jin B., Robertson K.D. DNA methyltransferases, DNA damage repair, and cancer. Adv Exp Med Biol 2013;754:3–29. DOI: https://doi.org/10.1007/978-1-4419-9967-2_1Test; Moore L.D., Le T., Fan G. DNA methylation and its basic function. Neuropsychopharmacology 2013;38(1):23–38. DOI: https://doi.org/10.1038/npp.2012.112Test; Jahangiri R., Mosaffa F., Emami Razavi A. et al. Altered DNA methyltransferases promoter methylation and mRNA expression are associated with tamoxifen response in breast tumors. J Cell Physiol 2018;233(9):7305–19. DOI: https://doi.org/10.1002/jcp.26562Test; He D., Wang X., Zhang Y. et al. DNMT3A/3B overexpression might be correlated with poor patient survival, hypermethylation and low expression of ESR1/PGR in endometrioid carcinoma: an analysis of The Cancer Genome Atlas. Chin Med J (Engl) 2019;132(2): 161–70. DOI: https://doi.org/10.1097/CM9.0000000000000054Test; Chen B.F., Chan W.Y. The de novo DNA methyltransferase DNMT3A in development and cancer. Epigenetics 2014;9(5):669–77. DOI: https://doi.org/10.4161/epi.28324Test; Shchegolev Y.Y., Sorokin D.V., Scherbakov A.M. et al. Exosomes are involved in the intercellular transfer of rapamycin resistance in the breast cancer cells. Bioimpacts 2023;13(4):313–21. DOI: https://doi.org/10.34172/bi.2023.27490Test; Scherbakov A.M., Sorokin D.V., Tatarskiy V.V.Jr. et al. The phenomenon of acquired resistance to metformin in breast cancer cells: the interaction of growth pathways and estrogen receptor signaling. IUBMB life 2016;68(4):281–92. DOI: https://doi.org/10.1002/iub.1481Test; Semina S.E., Scherbakov A.M., Kovalev S.V. et al. Horizontal transfer of tamoxifen resistance in MCF-7 cell derivates: proteome study. Cancer Invest 2017;35(8):506–18. DOI: https://doi.org/10.1080/07357907.2017.1368081Test; Iselt M., Holtei W., Hilgard P. The tetrazolium dye assay for rapid in vitro assessment of cytotoxicity. Arzneimittel-Forschung 1989;39(7):747–9.; Scherbakov A.M., Vorontsova S.K., Khamidullina A.I. et al. Novel pentacyclic derivatives and benzylidenes of the progesterone series cause anti-estrogenic and antiproliferative effects and induce apoptosis in breast cancer cells. Invest New Drugs 2023;41(1): 142–52. DOI: https://doi.org/10.1007/s10637-023-01332-zTest; Livak K.J., Schmittgen T.D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods (San Diego, Calif) 2001;25(4):402–8. DOI: https://doi.org/10.1006/meth.2001.1262Test; Scherbakov A.M., Komkov A.V., Komendantova A.S. et al. Steroidal pyrimidines and dihydrotriazines as novel classes of anticancer agents against hormone-dependent breast cancer cells. Front Pharmacol 2017;8:979. DOI: https://doi.org/10.3389/fphar.2017.00979Test; Scherbakov A.M., Lobanova Y.S., Shatskaya V.A. et al. Activation of mitogenic pathways and sensitization to estrogen-induced apoptosis: two independent characteristics of tamoxifen-resistant breast cancer cells? Breast Cancer Res Treat 2006;100(1):1–11. DOI: https://doi.org/10.1007/s10549-005-9075-xTest; Mruk D.D., Cheng C.Y. Enhanced chemiluminescence (ECL) for routine immunoblotting: an inexpensive alternative to commercially available kits. Spermatogenesis 2011;1(2):121–2. DOI: https://doi.org/10.4161/spmg.1.2.16606Test; Taylor S.C., Berkelman T., Yadav G. et al. A defined methodology for reliable quantification of Western blot data. Mol Biotechnol 2013;55(3):217–26. DOI: https://doi.org/10.1007/s12033-013-9672-6Test PMID: 23709336; Lander E.S., Linton L.M., Birren B. et al. Initial sequencing and analysis of the human genome. Nature 2001;409(6822):860–921. DOI:10.1038/35057062; Yang A.S., Estecio M.R., Doshi K. et al. A simple method for estimating global DNA methylation using bisulfite PCR of repetitive DNA elements. Nucleic Acids Res 2004;32(3):e38. DOI: https://doi.org/10.1093/nar/gnh032Test; Dong C., Wu J., Chen Y. et al. Activation of PI3K/AKT/mTOR pathway causes drug resistance in breast cancer. Front Pharmacol 2021;12:628690. DOI: https://doi.org/10.3389/fphar.2021Test; Liu R., Chen Y., Liu G. et al. PI3K/AKT pathway as a key link modulates the multidrug resistance of cancers. Cell Death Dis 2020;11(9):797. DOI: https://doi.org/10.1038/s41419-020-02998-6Test; Scherbakov A.M., Basharina A.A., Sorokin D.V. et al. Targeting hormone-resistant breast cancer cells with docetaxel: a look inside the resistance. Cancer Drug Resist 2023;6(1):103–15. DOI: https://doi.org/10.20517/cdr.2022.96Test; Meng D., Li Z., Ma X. et al. MicroRNA-1280 modulates cell growth and invasion of thyroid carcinoma through targeting estrogen receptor alpha. Cell Mol Biol (Noisy-le-grand) 2016;62 (3):1–6.; Xu C.G., Yang M.F., Ren Y.Q. et al. Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells. Eur Rev Med Pharmacol Sci 2016;20(20):4362–8.; Romero-Garcia S., Prado-Garcia H., Carlos-Reyes A. Role of DNA methylation in the resistance to therapy in solid tumors. Front Oncol 2020;10:1152. DOI: https://doi.org/10.3389/fonc.2020.01152Test; Nayak S.R., Harrington E., Boone D. et al. A role for histone H2B variants in endocrine-resistant breast cancer. Horm Cancer 2015;6(5–6):214–24. DOI: https://doi.org/10.1007/s12672-015-0230-5Test; Hamadneh L., Al-Lakkis L., Alhusban A.A. et al. Changes in Lactate production, lactate dehydrogenase genes expression and DNA methylation in response to tamoxifen resistance development in MCF-7 cell line. Genes (Basel) 2021;12(5). DOI: https://doi.org/10.3390/genes12050777Test; Андреева О.Е., Сигин В.О., Стрельников В.В. и др. Изменение профиля метилирования ДНК в тамоксифен-резистентных сублиниях клеток MCF-7. Сибирский онкологический журнал 2019;18(5):45–53. DOI: https://doi.org/10.21294/1814-4861-2019-18-5-45-53Test; https://umo.abvpress.ru/jour/article/view/605Test

  5. 5
    دورية أكاديمية
  6. 6
    دورية أكاديمية
  7. 7
  8. 8
    دورية أكاديمية

    المساهمون: The study was performed as a part of an experimental state task from the Ministry of Health of the Russian Federation under supervision of the Center for Strategic Planning and Management of Medical and Biological Health Risks, Ministry of Health of the Russian Federation, Данное исследование выполняется в рамках экспериментального государственного задания Министерства здравоохранения Российской Федерации при координации ФГБУ «Центр стратегического планирования и управления медико-биологическими рисками здоровью» Минздрава России

    المصدر: Advances in Molecular Oncology; Том 6, № 4 (2019); 26-37 ; Успехи молекулярной онкологии; Том 6, № 4 (2019); 26-37 ; 2413-3787 ; 2313-805X ; 10.17650/2313-805X-2019-6-4

    وصف الملف: application/pdf

    العلاقة: https://umo.abvpress.ru/jour/article/view/239/190Test; Bray F., Ferlay J., Soerjomataram I. et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortal ity worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018. DOI:10.3322/caac.21492.; Llovet J.M., Zucman-Rossi J., Pikarsky E. et al. Hepatocellular carcinoma. Nat Rev Dis Primers 2016;2:16018. DOI:10.1038/nrdp.2016.18.; Kulik L., El-Serag H.B. Epidemiology and management of hepatocellular carcinoma. Gastroenterology 2019;156(2):477–91.e1. DOI:10.1053/j.gastro.2018.08.065.; Abelev G.I., Eraiser T.L. Cellular aspects of alpha-fetoprotein reexpression in tumors. Semin Cancer Biol 1999;9(2):95–107.; Rich N., Singal A.G. Hepatocellular carcinoma tumour markers: current role and expectations. Best Pract Res Clin Gastroenterol 2014;28(5):843–53. DOI:10.1016/j.bpg.2014.07.018.; Gai W., Sun K. Epigenetic Biomarkers in Cell-Free DNA and Applications in Liq uid Biopsy. Genes (Basel) 2019;10(1). DOI:10.3390/genes10010032.; Кустова И.Ф., Макарова А.С., Лазаревич Н.Л. Потенциал использования биомаркеров метилирования для диагностики и прогноза гепатоцеллюлярной карциномы методом жидкостной биопсии. Успехи молекулярной онкологии 2018;5(4):8–19. DOI:10.17650/2313-805X-2018.; De Vos T., Tetzner R., Model F. et al. Circulating methylated SEPT9 DNA in plasma is a biomarker for colorectal cancer. Clin Chem 2009;55(7):1337–46. DOI:10.1373/clinchem.2008.115808.; Wasserkort R., Kalmar A., Valcz G. et al. Aberrant septin 9 DNA methylation in colorectal cancer is restricted to a single CpG island. BMC Cancer 2013;13:398. DOI:10.1186/1471-2407-13-398.; Epigenomics, 2016. Epi proColon®. Available at: https://www.accessdata.fda.gov/cdrh_docs/pdf13/P130001C.pdfTest. Accessed date: 1 October 2019.; Shen N., Wang T., Li D. et al. Hyper methylation of the Sept9 gene suggests significantly poor prognosis in cancer patients: a systematic review and metaanalysis. Front Genet 2019;10:887. DOI:10.3389/fgene.2019.00887.; Villanueva A., Portela A., Sayols S. et al. DNA methylation-based prognosis and epidrivers in hepatocellular carcinoma. Hepatology 2015;61(6):1945–56. DOI:10.1002/hep.27732.; Oussalah A., Rischer S., Bensenane M. et al. Plasma mSEPT9: a novel circulating cell-free DNA-based epigenetic biomarker to diagnose hepatocellular carcinoma. EBioMedicine 2018;30:138–47. DOI:10.1016/j.ebiom.2018.03.029.; Li J., Zhou X., Liu X. Detection of colorectal cancer in circulating cell-free DNA by methylated CpG tandem amplification and sequencing. Clin Chem 2019;65(7):916–26. DOI:10.1373/clinchem.2019.301804.; Cancer Genome Atlas Research Network. Comprehensive and Integrative Genomic Characterization of Hepatocellular Carcinoma. Cell 2017;169(7):1327–41.e23. DOI:10.1016/j.cell.2017.05.046.; Li L.C., Dahiya R. MethPrimer: designing primers for methylation PCRs. Bio informatics 2002;18(11):1427–31. DOI:10.1093/bioinformatics/18.11.1427.; Illumina, 2011. Available at: https://support.illumina.com/array/array_kits/infinium_humanmethylation450_beadchip_kit/downloads.htmlTest. Accessed date: 1 October 2019.; Xiong Y., Wei Y., Gu Y. et al. DiseaseMeth version 2.0: a major expansion and update of the human disease methylation database. Nucleic Acids Res 2017;45(D1):D888–95. DOI:10.1093/nar/gkw1123.; Li R., Liang F., Li M. et al. MethBank 3.0: a database of DNA methylomes across a variety of species. Nucleic Acids Res 2018;46(D1):D288–95. DOI:10.1093/nar/gkx1139.; Состояние онкологической помощи населению России в 2017 году. Под ред. А.Д. Каприна, В.В. Старинского, Г.В. Петровой. М.: МНИОИ им. П.А. Герцена – филиал ФГБУ «НМИЦ радиологии» Минздрава России, 2018. 236 с.; Wittwer C.T., Herrmann M.G., Moss A.A., Rasmussen R.P. Continuous fluorescence monitoring of rapid cycle DNA amplification. Biotechniques 1997;22(1):130–1. DOI:10.2144/97221bi01.; Malentacchi F., Forni G., Vinci S., Orlando C. Quantitative evaluation of DNA methylation by optimization of a differentialhigh resolution melt analysis protocol. Nucleic Acids Res 2009;37(12):e86. DOI:10.1093/nar/gkp383.; Bergheim J., Semaan A., Gevensleben H. et al. Potential of quantitative SEPT9 and SHOX2 methylation in plasmatic circulating cell-free DNA as auxiliary staging parameter in colorectal cancer: a prospective observational cohort study. Br J Cancer 2018;118(9):1217–28. DOI:10.1038/s41416-018-0035-8.; Song L., Yu H., Jia J., Li Y. A systematic review of the performance of the SEPT9 gene methylation assay in colorectal cancer screening, monitoring, diagnosis and prognosis. Cancer Biomark 2017;18(4):425–32. DOI:10.3233/CBM-160321.; https://umo.abvpress.ru/jour/article/view/239Test

  9. 9
  10. 10