Pokaż uproszczony rekord

dc.contributor.authorCzarny, Piotr
dc.contributor.authorPawlowska, Elzbieta
dc.contributor.authorBialkowska-Warzecha, Jolanta
dc.contributor.authorKaarniranta, Kai
dc.date.accessioned2015-04-20T08:42:22Z
dc.date.available2015-04-20T08:42:22Z
dc.date.issued2015-01-23
dc.identifier.issn1422-0067
dc.identifier.urihttp://hdl.handle.net/11089/8050
dc.description.abstractDNA damage response (DDR) involves DNA repair, cell cycle regulation and apoptosis, but autophagy is also suggested to play a role in DDR. Autophagy can be activated in response to DNA-damaging agents, but the exact mechanism underlying this activation is not fully understood, although it is suggested that it involves the inhibition of mammalian target of rapamycin complex 1 (mTORC1). mTORC1 represses autophagy via phosphorylation of the ULK1/2–Atg13–FIP200 complex thus preventing maturation of pre-autophagosomal structures. When DNA damage occurs, it is recognized by some proteins or their complexes, such as poly(ADP)ribose polymerase 1 (PARP-1), Mre11–Rad50–Nbs1 (MRN) complex or FOXO3, which activate repressors of mTORC1. SQSTM1/p62 is one of the proteins whose levels are regulated via autophagic degradation. Inhibition of autophagy by knockout of FIP200 results in upregulation of SQSTM1/p62, enhanced DNA damage and less efficient damage repair. Mitophagy, one form of autophagy involved in the selective degradation of mitochondria, may also play role in DDR. It degrades abnormal mitochondria and can either repress or activate apoptosis, but the exact mechanism remains unknown. There is a need to clarify the role of autophagy in DDR, as this process may possess several important biomedical applications, involving also cancer therapy.pl_PL
dc.language.isoenpl_PL
dc.publisherMDPI AGpl_PL
dc.relation.ispartofseriesInternational Journal of Molecular Sciences;2015, 16(2)
dc.rightsUznanie autorstwa 3.0 Polska*
dc.rights.urihttp://creativecommons.org/licenses/by/3.0/pl/*
dc.subjectautophagypl_PL
dc.subjectDNA damage responsepl_PL
dc.subjectDNA repairpl_PL
dc.subjectapoptosispl_PL
dc.subjectsignal transductionpl_PL
dc.subjectsenescencepl_PL
dc.subjectcancer therapypl_PL
dc.titleAutophagy in DNA Damage Responsepl_PL
dc.typeArticlepl_PL
dc.page.number2641-2662pl_PL
dc.contributor.authorAffiliationCzarny, Piotr, University of Lodz, Department of Molecular Geneticspl_PL
dc.contributor.authorAffiliationPawlowska, Elzbieta, Medical University of Lodz, Department of Orthodonticspl_PL
dc.contributor.authorAffiliationBialkowska-Warzecha Jolanta, Medical University of Lodz, Department of Infectious and Liver Diseasespl_PL
dc.contributor.authorAffiliationKaarniranta, Kai, University of Eastern Finland, Department of Ophthalmology, Institute of Clinical Medicinepl_PL
dc.referencesCiccia, A.; Elledge, S.J. The DNA damage response: Making it safe to play with knives. Mol. Cell 2010, 40, 179–204.pl_PL
dc.referencesFerguson, L.R. Chronic inflammation and mutagenesis. Mutat. Res. 2010, 690, 3–11.pl_PL
dc.referencesJackson, S.P.; Bartek, J. The DNA-damage response in human biology and disease. Nature 2009, 461, 1071–1078.pl_PL
dc.referencesKaarniranta, K.; Sinha, D.; Blasiak, J.; Kauppinen, A.; Veréb, Z.; Salminen, A.; Boulton, M.E.; Petrovski, G. Autophagy and heterophagy dysregulation leads to retinal pigment epithelium dysfunction and development of age-related macular degeneration. Autophagy 2013, 9, 973–984.pl_PL
dc.referencesSimpson, P.T.; Vargas, A.C.; Al-Ejeh, F.; Khanna, K.K.; Chenevix-Trench, G.; Lakhani, S.R. Application of molecular findings to the diagnosis and management of breast disease: Recent advances and challenges. Hum. Pathol. 2011, 42, 153–165.pl_PL
dc.referencesEker, A.P.; Quayle, C.; Chaves, I.; van der Horst, G.T. DNA repair in mammalian cells. Cell. Mol. Life Sci. 2009, 66, 968–980.pl_PL
dc.referencesLindahl, T.; Barnes, D.E. Repair of endogenous DNA damage. Cold Spring Harb. Symp. Quant. Biol. 2000, 65, 127–133.pl_PL
dc.referencesHoeijmakers, J.H. DNA damage, aging, and cancer. N. Engl. J. Med. 2009, 361, 1475–1485.pl_PL
dc.referencesJiricny, J. The multifaceted mismatch-repair system. Nat. Rev. Mol. Cell Biol. 2006, 7, 335–346.pl_PL
dc.referencesChapman, J.R.; Taylor., M.R.; Boulton, S.J. Playing the end game: DNA double-strand break repair pathway choice. Mol. Cell 2012, 47, 497–510.pl_PL
dc.referencesYakes, F.M.; van Houten, B. Mitochondrial DNA damage is more extensive and persists longer than nuclear DNA damage in human cells following oxidative stress. Proc. Natl. Acad. Sci. USA 1997, 94, 514–519.pl_PL
dc.referencesReddy, V.N.; Kasahara, E.; Hiraoka, M.; Lin, L.R.; Ho, Y.S. Effects of variation in superoxide dismutases (SOD) on oxidative stress and apoptosis in lens epithelium. Exp. Eye Res. 2004, 79, 859–868.pl_PL
dc.referencesBanmeyer, I.C.; Clippe, A.; Knoops, B. Human mitochondrial peroxiredoxin 5 protects from mitochondrial DNA damages induced by hydrogen peroxide. FEBS Lett. 2005, 579, 2327–2333.pl_PL
dc.referencesPascucci, B.; Versteegh, A.; van Hoffen, A.; van Zeeland, A.A.; Mullenders, L.H.; Dogliotti, E. DNA repair of UV photoproducts and mutagenesis in human mitochondrial DNA. J. Mol. Biol. 1997, 273, 417–427.pl_PL
dc.referencesBoesch, P.; Weber-Lotfi, F.; Ibrahim, N.; Tarasenko, V.; Cosset, A.; Paulus, F.; Lightowlers, R.N.; Dietrich, A. DNA repair in organelles: Pathways, organization, regulation, relevance in disease and aging. Biochim. Biophys. Acta 2011, 1813, 186–200.pl_PL
dc.referencesLe Doux, S.P.; Wilson, G.L. Base excision repair of mitochondrial DNA damage in mammalian cells. Prog. Nucleic Acid Res. Mol. Biol. 2001, 68, 273–284.pl_PL
dc.referencesDe Souza-Pinto, N.C.; Mason, P.A.; Hashiguchi, K.; Weissman, L.; Tian, J.; Guay, D.; Lebel, M.; Stevnsner, T.V.; Rasmussen, L.J.; Bohr, V.A.; et al. Novel DNA mismatch-repair activity involving YB-1 in human mitochondria. DNA Repair 2009, 8, 704–719.pl_PL
dc.referencesKraytsberg, Y.; Schwartz, M.; Brown, T.A.; Ebralidse, K.; Kunz, W.S.; Clayton, D.A.; Vissing, J.; Khrapko, K. Recombination of human mitochondrial DNA. Science 2004, 304, 981.pl_PL
dc.referencesSage, J.M.; Gildemeister, O.S.; Knight, K.L. Discovery of a novel function for human Rad51: Maintenance of the mitochondrial genome. J. Biol. Chem. 2010, 285, 18984–18990.pl_PL
dc.referencesLakshmipathy, U.; Campbell, C. Double strand break rejoining by mammalian mitochondrial extracts. Nucleic Acids Res. 1999, 27, 11198–11204.pl_PL
dc.referencesCui, R.; Widlund, H.R.; Feige, E.; Lin, J.Y.; Wilensky, D.L.; Igras, V.E.; D’Orazio, J.; Fung, C.Y.; Schanbacher, C.F.; Granter, S.R.; et al. Central role of p53 in the suntan response and pathologic hyperpigmentation. Cell 2007, 128, 853–864.pl_PL
dc.referencesSchreiber, V.; Dantzer, F.; Ame, J.C.; de Murcia, G. Poly(ADP-ribose): Novel functions for an old molecule. Nat. Rev. Mol. Cell Biol. 2006, 7, 517–528.pl_PL
dc.referencesSancar, A.; Lindsey-Boltz, L.A.; Unsal-Kacmaz, K.; Linn, S. Molecular mechanisms of mammalian DNA repair and the DNA damage checkpoints. Annu. Rev. Biochem. 2004, 73, 39–85.pl_PL
dc.referencesMeek, K.; Dang, V.; Lees-Miller, S.P. DNA-PK: The means to justify the ends? Adv. Immunol. 2008, 99, 33–58.pl_PL
dc.referencesHarpe, J.W.; Elledge, S.J. The DNA damage response: Ten years after. Mol. Cell 2007, 28, 739–745.pl_PL
dc.referencesMatsuoka, S.; Ballif, B.A.; Smogorzewska, A.; McDonald, E.R., 3rd; Hurov, K.E.; Luo, J.; Bakalarski, C.E.; Zhao, Z.; Solimini, N.; Lerenthal, Y.; et al. ATM and ATR substrate analysis reveals extensive protein networks responsive to DNA damage. Science 2007, 316, 1160–1166.pl_PL
dc.referencesPaulsen, R.D.; Soni, D.V.; Wollman, R.; Hahn, A.T.; Yee, M.C.; Guan, A.; Hesley, J.A.; Miller, S.C.; Cromwell, E.F.; Solow-Cordero, D.E.; et al. A genome-wide siRNA screen reveals diverse cellular processes and pathways that mediate genome stability. Mol. Cell 2009, 35, 228–239.pl_PL
dc.referencesCimprich, K.A.; Cortez, D. ATR: An essential regulator of genome integrity. Nat. Rev. Mol. Cell Biol. 2008, 9, 616–627.pl_PL
dc.referencesPaulovich, A.G.; Margulies, R.U.; Garvik, B.M.; Hartwell, L.H. RAD9, RAD17, and RAD24 are required for S phase regulation in Saccharomyces cerevisiae in response to DNA damage. Genetics 1997, 145, 45–62.pl_PL
dc.referencesVolkmer, E.; Karnitz, L.M. Human homologs of Schizosaccharomyces pombe Rad1, Hus1, and Rad9 form a DNA damage-responsive protein complex. J. Biol. Chem. 1999, 274, 567–570.pl_PL
dc.referencesThelen, M.P.; Venclovas, C.; Fidelis, K. A sliding clamp model for the Rad1 family of cell cycle checkpoint proteins. Cell 1999, 96, 769–770.pl_PL
dc.referencesGriffiths, D.J.; Barbet, N.C.; McCready, S.; Lehmann, A.R.; Carr, A.M. Fission yeast rad17: A homologue of budding yeast RAD24 that shares regions of sequence similarity with DNA polymerase accessory proteins. EMBO J. 1995, 14, 5812–5823.pl_PL
dc.referencesGreen, C.M.; Erdjument-Bromage, H.; Tempst, P.; Lowndes, N.F. A novel Rad24 checkpoint protein complex closely related to replication factor C. Curr. Biol. 2000, 10, 39–42.pl_PL
dc.referencesNaiki, T.; Shimomura, T.; Kondo, T.; Matsumoto, K.; Sugimoto, K. Rfc5, in cooperation with Rad24, controls DNA damage checkpoints throughout the cell cycle in Saccharomyces cerevisiae. Mol. Cell. Biol. 2000, 20, 5888–5896.pl_PL
dc.referencesLindsey-Boltz, L.A.; Bermudez, V.P.; Hurwitz, J; Sancar, A. Purification and characterization of human DNA damage checkpoint Rad complexes. Proc. Natl. Acad. Sci. USA 2001, 98, 11236–11241.pl_PL
dc.referencesGolia, B.; Singh, H.R.; Timinszki, G. Poly-ADP-ribosylation signaling during DNA damage repair. Front. Biosci. 2015, 20, 440–457.pl_PL
dc.referencesBakkenist, C.J.; Kastan, M.B. DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature 2003, 421, 499–506.pl_PL
dc.referencesWang, Y.; Cortez, D.; Yazdi, P.; Neff, N.; Elledge, S.J.; Qin, J. BASC, a super complex “of BRCA1-associated proteins involved in the recognition and repair of aberrant DNA structures. Genes Dev. 2000, 14, 927–939.pl_PL
dc.referencesKim, G.D.; Choi, Y.H.; Dimtchev, A.; Jeong, S.J.; Dritschilo, A.; Jung, M. Sensing of ionizing radiation-induced DNA damage by ATM through interaction with histone deacetylase. J. Biol. Chem. 1999, 274, 31127–33130.pl_PL
dc.referencesSchmidt, D.R.; Schreiber, S.L. Molecular association between ATR and two components of the nucleosome remodeling and deacetylating complex, HDAC2 and CHD4. Biochemistry 1999, 38, 14711–14717.pl_PL
dc.referencesTran, H.; Brunet, A.; Grenier, J.M.; Datta, S.R.; Fornace, A.J., Jr.; DiStefano, P.S.; Chiang, L.W.; Greenberg, M.E. DNA repair pathway stimulated by the forkhead transcription factor FOXO3a through the Gadd45 protein. Science 2002, 296, 530–534.pl_PL
dc.referencesThurn, K.T.; Thomas, S.; Raha, P.; Qureshi, I.; Munster, P.N. Histone deacetylase regulation of ATM-mediated DNA damage signaling. Mol. Cancer Ther. 2013, 12, 2078–2087.pl_PL
dc.referencesRodriguez-Rochaa, H.; Garcia-Garciaa, A.; Panayiotidisb, M.I.; Francoa, R. DNA damage and autophagy. Mutat. Res. 2011, 711, 158–166.pl_PL
dc.referencesFestjens, N.; VandenBerghe, T.; Vandenabeele, P. Necrosis, a well-orchestrated form of cell demise: Signaling cascades, important mediators and concomitant immune response. Biochim. Biophys. Acta 2006, 1757, 1371–1387.pl_PL
dc.referencesHuang, C.; Freter, C. Lipid metabolism, apoptosis and cancer therapy. Int. J. Mol. Sci. 2015, 16, 924–949.pl_PL
dc.referencesHsu, H.; Xiong, J.; Goeddel, D.V. The TNF receptor 1-associated protein TRADD signals cell death and NFκB activation. Cell 1995, 81, 495–504.pl_PL
dc.referencesWajant, H. The Fas signaling pathway: More than a paradigm. Science 2002, 29, 1635–1636.pl_PL
dc.referencesKischkel, F.C.; Hellbardt, S.; Behrmann, I.; Germer, M.; Pawlita, M.; Krammer, P.H.; Peter, M.E. Cytotoxicity dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 1995, 14, 5579–5588.pl_PL
dc.referencesSaelens, X.; Festjens, N.; vande Walle, L.; van Gurp, M.; van Loo, G.; Vandenabeele, P. Toxic proteins released from mitochondria in cell death. Oncogene 2004, 23, 2861–2874.pl_PL
dc.referencesParsons, M.J.; Green, D.R. Mitochondria in cell death. Essays Biochem. 2010, 47, 99–114.pl_PL
dc.referencesElmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516.pl_PL
dc.referencesSchuler, M.; Green, D.R. Mechanisms of p53-dependent apoptosis. Biochem. Soc. Trans. 2001, 29, 684–688.pl_PL
dc.referencesRoos, W.P.; Kaina, B. DNA damage-induced cell death by apoptosis. Trends Mol. Med. 2006, 12, 440–450.pl_PL
dc.referencesChipuk, J.E.; Green, D.R. Dissecting p53-dependent apoptosis. Cell Death Differ. 2006, 13, 994–1002.pl_PL
dc.referencesKung, C.P.; Budina, A.; Balaburski, G.; Bergenstock, M.K.; Murphy, M. Autophagy in tumor suppression and cancer therapy. Crit. Rev. Eukaryot. Gene Expr. 2011, 21, 71–100.pl_PL
dc.referencesLevine, B.; Klionsky, D.J. Development by self-digestion: Molecular mechanisms and biological functions of autophagy. Cell 2004, 6, 463–477.pl_PL
dc.referencesCuervo, A.M. Autophagy: Many paths to the same end. Mol. Cell. Biochem. 2004, 263, 55–72.pl_PL
dc.referencesMarzella, L.; Ahlberg, J.; Glaumann, H. Autophagy, heterophagy, microautophagy and crinophagy as the means for intracellular degradation. Virchows Arch. B 1981, 36, 219–234.pl_PL
dc.referencesShao, N.; Chai, Y.L.; Shyam, E.; Reddy, P.; Rao, V.N. Induction of apoptosis by the tumor suppressor protein BRCA1. Oncogene 1996, 13, 1–7.pl_PL
dc.referencesMartin, S.A.; Ouchi, T. BRCA1 phosphorylation regulates caspase-3 activation in UV-induced apoptosis. Cancer Res. 2005, 65, 10657–10662.pl_PL
dc.referencesBurma, S.; Chen, D.J. Role of DNA-PK in the cellular response to DNA double-strand breaks. DNA Repair 2004, 3, 909–918.pl_PL
dc.referencesEspejel, S.; Franco, S.; Sgura, A.; Gae, D.; Bailey, S.M.; Taccioli, G.E.; Blasco, M.A. Functional interaction between DNA-PKcs and telomerase in telomere length maintenance. EMBO J. 2002, 21, 6275–6287.pl_PL
dc.referencesEspejel, S.; Martín, M.; Klatt, P.; Martín-Caballero, J.; Flores, J.M.; Blasco, M.A. Shorter telomeres, accelerated ageing and increased lymphoma in DNA-PKcs deficient mice. EMBO Rep. 2004, 5, 503–509.pl_PL
dc.referencesLuo, X.; Kraus, W.L. On PAR with PARP: Cellular stress signaling through poly (ADP-ribose) and PARP-1. Genes Dev. 2012, 26, 417–432.pl_PL
dc.referencesNowsheen, S.; Bonner, J.A.; Lo Buglio, A.F.; Trummell, H.; Whitley, A.C.; Dobelbower, M.C.; Yang, E.S. Cetuximab augments cytotoxicity with poly (ADP-ribose) polymerase inhibition in head and neck cancer. PLoS One 2011, 6, e24148.pl_PL
dc.referencesAl-Ejeh, F.; Shi, W.; Miranda, M.; Simpson, P.T.; Vargas, A.C.; Song, S.; Wiegmans, A.P.; Swarbrick, A.; Welm, A.L.; Brown, M.P.; et al. Treatment of triple-negative breast cancer using anti-EGFR-directed radioimmunotherapy combined with radiosensitizing chemotherapy and PARP inhibitor. J. Nucl. Med. 2013, 54, 913–921.pl_PL
dc.referencesYu, S.W.; Wang, H.; Poitras, M.F.; Coombs, C.; Bowers, W.J.; Federoff, H.J.; Poirier, G.G.; Dawson, T.M.; Dawson, V.L. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 2002, 297, 259–263.pl_PL
dc.referencesCregan, S.P.; Dawson, V.L.; Slack, R.S. Role of AIF in caspase-dependent and caspase-independent cell death. Oncogene 2004, 23, 2785–2796.pl_PL
dc.referencesAl-Ejeh, F.; Kumar, R.; Wiegmans, A.; Lakhani, S.R.; Brown, M.P.; Khanna, K.K. Harnessing the complexity of DNA-damage response pathways to improve cancer treatment outcomes. Oncogene 2010, 29, 6085–6098.pl_PL
dc.referencesMortimore, G.E.; Lardeux, B.R.; Adams, C.E. Regulation of microautophagy and basal protein turnover in rat liver. Effects of short-term starvation. J. Biol. Chem. 1988, 263, 2506–2512.pl_PL
dc.referencesAgarraberes, F.; Dice, J.F. A molecular chaperone complex at the lysosomal membrane is required for protein translocation. J. Cell Sci. 2001, 114, 2491–2499.pl_PL
dc.referencesMajeski, A.E.; Dice, J.F. Mechanisms of chaperone-mediated autophagy. Int. J. Biochem. Cell Biol. 2004, 36, 2435–2444.pl_PL
dc.referencesMassey, A.C.; Zhang, C.; Cuervo, A.M. Chaperone-mediated autophagy in aging and disease. Curr. Top. Dev. Biol. 2006, 73, 205–235.pl_PL
dc.referencesKlionsky, D.J.; Emr, S.D. Autophagy as a regulated pathway of cellular degradation. Science 2000, 290, 1717–1721.pl_PL
dc.referencesBaehrecke, E.H. Autophagy: Dual roles in life and death? Nat. Rev. Mol. Cell Biol 2005, 6, 505–510.pl_PL
dc.referencesKlionsky, D.J.; Abdalla, F.C.; Abeliovich, H.; Abraham, R.T.; Acevedo-Arozena, A.; Adeli, K.; Agholme, L.; Agnello, M.; Agostinis, P.; Aguirre-Ghiso, J.A.; et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 2012, 8, 445–544.pl_PL
dc.referencesFilomeni, G.; de Zio, D.; Cecconi, F. Oxidative stress and autophagy: the clash between damage and metabolic needs. Cell Death Differ. 2014.pl_PL
dc.referencesKroemer, G.; Mariño, G.; Levine, B. Autophagy and the integrated stress response. Mol. Cell 2010, 40, 280–293.pl_PL
dc.referencesGozuacik, D.; Kimchi, A. Autophagy and cell death. Curr. Top. Dev. Biol. 2007, 78, 217–245.pl_PL
dc.referencesWei, H.; Wang, C.; Croce, C.M.; Guan, J.L. p62/SQSTM1 synergizes with autophagy for tumor growth in vivo. Genes Dev. 2014, 28, 1204–1216.pl_PL
dc.referencesKamada, Y.; Funakoshi, T.; Shintani, T.; Nagano, K.; Ohsumi, M.; Ohsumi, Y. Tor-mediated induction of autophagy via an Apg1 protein kinase complex. J. Cell Biol. 2000, 150, 1507–1513.pl_PL
dc.referencesMizushima, N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr. Opin. Cell Biol. 2010, 22, 132–139.pl_PL
dc.referencesWei, H.; Guan, J.L. Pro-tumorigenic function of autophagy in mammalian oncogenesis. Autophagy 2012, 8, 129–131.pl_PL
dc.referencesDunlop, E.A.; Tee, A.R. mTOR and autophagy: A dynamic relationship governed by nutrients and energy. Semin. Cell Dev. Biol. 2014, 36, 121–129.pl_PL
dc.referencesRoberts, D.J.; Miyamoto, S. Hexokinase II integrates energy metabolism and cellular protection: Akting on mitochondria and TORcing to autophagy. Cell Death Differ. 2015, 22, 248–257.pl_PL
dc.referencesMizushima, N.; Noda, T.; Ohsumi, Y. Apg16p is required for the function of the Apg12p–Apg5p conjugate in the yeast autophagy pathway. EMBO J. 1999, 18, 3888–3896.pl_PL
dc.referencesKuma, A.; Mizuchima, N.; Ishihara, N.; Ohsumi, Y. Formation of the approximately 350 kDa Apg12–Apg5–Apg16 multimeric complex, mediated by Apg16 oligomerization, is essential for autophagy in yeast. J. Biol. Chem. 2002, 277, 18619–18625.pl_PL
dc.referencesScott, S.V.; Nice, D.C., 3rd; Nau, J.J.; Weisman, L.S.; Kamada, Y.; Keizer-Gunnink, I.; Funakoshi, T.; Veenhuis, M.; Ohsumi, Y.; Klionsky, D.J.; et al. Apg13p and Vac8p are part of a complex of phosphoproteins that are required for cytoplasm to vacuole targeting. J. Biol. Chem. 2000, 275, 25840–25849.pl_PL
dc.referencesItakura, E.; Kishi, C.; Inoue, K.; Mizushima, N. Beclin-1 forms two distinct phosphatidylinositol 3-kinase complexes with mammalian Atg14 and UVRAG. Mol. Biol. Cell 2008, 19, 5360–5372.pl_PL
dc.referencesLiang, C.; Feng, P.; Ku, B.; Dotan, I.; Canaani, D.; Oh, B.H.; Jung, J.U. Autophagic and tumour suppressor activity of a novel Beclin1-binding protein UVRAG. Nat. Cell Biol. 2006, 8, 688–699.pl_PL
dc.referencesSun, Q.; Fan, W.; Chen, K.; Ding, X.; Chen, S.; Zhong, Q. Identification of Barkor as a mammalian autophagy-specific factor for Beclin 1 and class III phosphatidylinositol 3-kinase. Proc. Natl. Acad. Sci. USA 2008, 105, 19211–19216.pl_PL
dc.referencesTakahashi, Y.; Coppola, D.; Matsushita, N.; Cualing, H.D.; Sun, M.; Sato, Y.; Liang, C.; Jung, J.U.; Cheng, J.Q.; Mulé, J.J.; et al. Bif-1 interacts with Beclin 1 through UVRAG and regulates autophagy and tumorigenesis. Nat. Cell Biol. 2007, 9, 1142–1151.pl_PL
dc.referencesKihara, A.; Noda, T.; Ishihara, N.; Ohsumi, Y. Two distinct Vps34 phosphatidylinositol 3-kinase complexes function in autophagy and carboxypeptidase Y sorting in Saccharomyces cerevisiae. J. Cell Biol. 2001, 152, 519–530.pl_PL
dc.referencesIchimura, Y.; Kirisako, T.; Takao, T.; Satomi, Y.; Shimonishi, Y.; Ishihara, N.; Mizushima, N.; Tanida, I.; Kominami, E.; Ohsumi, M.; et al. An ubiquitin-like system mediates protein lipidation. Nature 2000, 408, 488–492.pl_PL
dc.referencesKawai, A.; Uchiyama, H.; Takano, S.; Nakamura, N.; Ohkuma, S. Autophagosome-lysosome fusion depends on the pH in acidic compartments in CHO cells. Autophagy 2007, 3, 154–157.pl_PL
dc.referencesRieber, M.; Rieber, M.S. Sensitization to radiation-induced DNA damage accelerates loss of Bcl-2 and increases apoptosis and autophagy. Cancer Biol. Ther. 2008, 7, 1561–1566.pl_PL
dc.referencesKatayama, M.; Kawaguchi, T.; Berger, M.S.; Pieper, R.O. DNA damaging agent induced autophagy produces a cytoprotective adenosine triphosphate surge in malignant glioma cells. Cell Death Differ. 2007, 14, 548–558.pl_PL
dc.referencesAbedin, M.J.; Wang, D.; McDonnell, M.A.; Lehmann, U.; Kelekar, A. Autophagy delays apoptotic death in breast cancer cells following DNA damage. Cell Death Differ. 2007, 14, 500–510.pl_PL
dc.referencesElliott, A.; Reiners, J.J., Jr. Suppression of autophagy enhances the cytotoxicity of the DNA-damaging aromatic amine p-anilinoaniline. Toxicol. Appl. Pharmacol. 2008, 232, 169–179.pl_PL
dc.referencesApel, A.; Herr, I.; Schwarz, H.; Rodemann, H.P.; Mayer, A. Blocked autophagy sensitizes resistant carcinoma cells to radiation therapy. Cancer Res. 2008, 68, 1485–1494.pl_PL
dc.referencesQadir, M.A.; Kwok, B.; Dragowska, W.H.; To, K.H.; Le, D.; Bally, M.B.; Gorski, S.M. Macroautophagy inhibition sensitizes tamoxifen-resistant breast cancer cells and enhances mitochondrial depolarization. Breast Cancer Res. Treat. 2008, 112, 389–403.pl_PL
dc.referencesAmaravadi, R.K.; Yu, D.; Lum, J.J.; Bui, T.; Christophorou, M.A.; Evan, G.I.; Thomas-Tikhonenko, A.; Thompson, C.B. Autophagy inhibition enhances therapy-induced apoptosis in a Myc-induced model of lymphoma. J. Clin. Investig. 2007, 117, 326–336.pl_PL
dc.referencesZhao, Z.; Ni, D.; Ghozalli, I.; Piroz, S.D.; Ma, B.; Liang, C. UVARG at the crossroad of autophagy and genomic stability. Autophagy 2012, 8, 1392–1393.pl_PL
dc.referencesKarantza-Wadsworth, V.; Patel, S.; Kravchuk, O.; Chen, G.; Mathew, R.; Jin, S.; White, E. Autophagy mitigates metabolic stress and genome damage in mammary tumorigenesis. Genes Dev. 2007, 21, 1621–1635.pl_PL
dc.referencesAlexander, A.; Kim, J.; Walker, C.L. ATM engages the TSC2/mTORC1 signaling node to regulate autophagy. Autophagy 2010, 6, 672–673.pl_PL
dc.referencesAlexander, A.; Cai, S.L.; Kim, J.; Nanez, A.; Sahin, M.; MacLean, K.H.; Inoki, K.; Guan, K.L.; Shen, J.; Person, M.D.; et al. ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS. Proc. Natl. Acad. Sci. USA 2010, 107, 4153–4158.pl_PL
dc.referencesInoki, K.; Zhu, T.; Guan, K.L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 2003, 115, 577–590.pl_PL
dc.referencesZhao, M.; Klionsky, D.J. AMPK-dependent phosphorylation of ULK1 induces autophagy. Cell Metab. 2011, 13, 119–120.pl_PL
dc.referencesTsai, W.B.; Chung, Y.M.; Takahashi, Y.; Xu, Z.; Hu, M.C. Functional interaction between FOXO3a and ATM regulates DNA damage response. Nat. Cell Biol. 2008, 10, 460–467.pl_PL
dc.referencesMammucari, C.; Milan, G.; Romanello, V.; Masiero, E.; Rudolf, R.; del Piccolo, P.; Burden, S.J.; di Lisi, R.; Sandri, C.; Zhao, J.; et al. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab. 2007, 6, 458–471.pl_PL
dc.referencesSalminen, A.; Kaarniranta, K. Regulation of the aging process by autophagy. Trends Mol. Med. 2009, 15, 217–224.pl_PL
dc.referencesChiacchiera, F.; Simone, C. The AMPK-FoxO3A axis as a target for cancer treatment. Cell Cycle 2010, 9, 1091–1096.pl_PL
dc.referencesTasdemir, E.; Chiara Maiuri, M.; Morselli, E.; Criollo, A.; D’Amelio, M.; Djavaheri-Mergny, M.; Cecconi, F.; Tavernarakis, N.; Kroemer, G. A dual role of p53 in the control of autophagy. Autophagy 2008, 4, 810–814.pl_PL
dc.referencesZong, W.X.; Moll, U. p53 in autophagy control. Cell Cycle 2008, 7, 2947–2948.pl_PL
dc.referencesKang, K.B.; Zhu, C.; Yong, S.K.; Gao, Q.; Wong, M.C. Enhanced sensitivity of celecoxib in human glioblastoma cells: Induction of DNA damage leading to p53-dependent G1 cell cycle arrest and autophagy. Mol. Cancer 2009, 8, 66.pl_PL
dc.referencesFeng, Z.; Zhang, H.; Levine, A.J.; Jin, S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc. Natl. Acad. Sci. USA 2005, 102, 8204–8209.pl_PL
dc.referencesFortini, P.; Dogliotti, E. Mechanisms of dealing with DNA damage in terminally differentiated cells. Mutat. Res. 2010, 685, 38–44.pl_PL
dc.referencesJin, S. p53, Autophagy and tumor suppression. Autophagy 2005, 1, 171–173.pl_PL
dc.referencesCrighton, D.; Wilkinson, S.; O’Prey, J.; Syed, N.; Smith, P.; Harrison, P.R.M.; Garrone, O.; Crook, T.; Ryan, K.M. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 2006, 126, 121–134.pl_PL
dc.referencesCrighton, D.; Wilkinson, S.; Ryan, K.M. DRAM links autophagy to p53 and programmed cell death. Autophagy 2007, 3, 72–74.pl_PL
dc.referencesValbuena, A.; Castro-Obregón, S.; Lazo, P.A. Down-regulation of VRK1 by p53 in response to DNA damage is mediated by the autophagic pathway. PLoS One 2011, 6, e17320.pl_PL
dc.referencesKlerkx, E.P.; Lazo, P.A.; Askjaer, P. Emerging biological functions of the vaccinia-related kinase (VRK) family. Histol. Histopathol. 2009, 24, 749–759.pl_PL
dc.referencesSanz-Garcia, M.; Valbuena González, M.; López-Sánchez, A.; Blanco, I.; Fernández, S.; Vázquez Cedeira, I.F.; Lazo, M.; Pedro, A. Vaccinia-related kinase (VRK) signaling in cell and tumor biology. In Emerging Signaling Pathways in Tumor Biology; Lazo, P.A., Ed.; Transworld Research Networks: Kerala, India, 2010; pp. 135–156.pl_PL
dc.referencesDyavaiah, M.; Rooney, J.P.; Chittur, S.V.; Lin, Q.; Begley, T.J. Autophagy-dependent regulation of the DNA damage response protein ribonucleotide reductase 1. Mol. Cancer Res. 2011, 9, 462–475.pl_PL
dc.referencesHa, H.C.; Snyder, S.H. Poly(ADP-ribose) polymerase is a mediator of necrotic cell death by ATP depletion. Proc. Natl. Acad. Sci. USA 1999, 96, 13978–13982.pl_PL
dc.referencesRodríguez-Vargas, J.M.; Ruiz-Magaña, M.J.; Ruiz-Ruiz, C.; Majuelos-Melguizo, J.; Peralta-Leal, A.; Rodríguez, M.I.; Muñoz-Gámez, J.A.; de Almodóvar, M.R.; Siles, E.; Rivas, A.L.; et al. ROS-induced DNA damage and PARP-1 are required for optimal induction of starvation-induced autophagy. Cell Res. 2012, 22, 1181–1198.pl_PL
dc.referencesHoyer-Hansen, M.; Jaattela, M. AMP-activated protein kinase: A universal regulator of autophagy? Autophagy 2007, 3, 381–383.pl_PL
dc.referencesMunoz-Gamez, J.A.; Rodríguez-Vargas, J.M.; Quiles-Pérez, R.; Aguilar-Quesada, R.; Martín-Oliva, D.; de Murcia, G.; Menissier de Murcia, J.; Almendros, A.; Ruiz de Almodóvar, M.; Oliver, F.J.; et al. PARP-1 is involved in autophagy induced by DNA damage. Autophagy 2009, 5, 61–74.pl_PL
dc.referencesHuang, Q.; Shen, H.M. To die or to live: The dual role of poly(ADP-ribose) polymerase-1 in autophagy and necrosis under oxidative stress and DNA damage. Autophagy 2009, 5, 273–276.pl_PL
dc.referencesYoon, J.H.; Ahn, S.G.; Lee, B.H.; Jung, S.H.; Oh, S.H. Role of autophagy in chemoresistance: Regulation of the ATM-mediated DNA-damage signaling pathway through activation of DNA-PKcs and PARP-1. Biochem. Pharmacol. 2012, 83, 747–757.pl_PL
dc.referencesAbbi, S.; Ueda, H.; Zheng, C.; Cooper, L.A.; Zhao, J.; Christopher, R.; Guan, J.L. Regulation of focal adhesion kinase by a novel protein inhibitor FIP200. Mol. Biol. Cell 2002, 13, 3178–3191.pl_PL
dc.referencesRajendran, R.; Garva, R.; Krstic-Demonacos, M.; Demonacos, C. Sirtuins: Molecular traffic lights in the crossroad of oxidative stress, chromatin remodeling, and transcription. J. Biomed. Biotechnol. 2011, 2011, 368276.pl_PL
dc.referencesKitada, M.; Kume, S.; Takeda-Watanabe, A.; Kanasaki, K.; Koya, D. Sirtuins and renal diseases: Relationship with aging and diabetic nephropathy. Clin. Sci. 2013, 124, 153–164.pl_PL
dc.referencesHariharan, N.; Maejima, Y.; Nakae, J.; Paik, J.; Depinho, R.A.; Sadoshima, J. Deacetylation of FoxO by Sirt1 plays an essential role in mediating starvation-induced autophagy in cardiac myocytes. Circ. Res. 2010, 107, 1470–1482.pl_PL
dc.referencesHands, S.L.; Proud, C.G.; Wyttenbach, A. mTOR’s role in ageing: Protein synthesis or autophagy? Aging 2009, 1, 586–597.pl_PL
dc.referencesYi, J.; Luo, J. SIRT1 and p53, effect on cancer, senescence and beyond. Biochim. Biophys. Acta 2010, 1804, 1684–1689.pl_PL
dc.referencesUeda, H.; Abbi, S.; Zheng, C.; Guan, J.L. Suppression of Pyk2 kinase and cellular activities by FIP200. J. Cell Biol. 2000, 149, 423–430.pl_PL
dc.referencesGan, B.; Melkoumian, Z.K.; Wu, X.; Guan, K.L.; Guan, J.L. Identification of FIP200 interaction with the TSC1–TSC2 complex and its role in regulation of cell size control. J. Cell Biol. 2005, 170, 379–389.pl_PL
dc.referencesMelkoumian, Z.K.; Peng, X.; Gan, B.; Wu, X.; Guan, J.L. Mechanism of cell cycle regulation by FIP200 in human breast cancer cells. Cancer Res. 2005, 65, 6676–6684.pl_PL
dc.referencesGan, B.; Peng, X.; Nagy, T.; Alcaraz, A.; Gu, H.; Guan, J.L. Role of FIP200 in cardiac and liver development and its regulation of TNFα and TSC–mTOR signaling pathways. J. Cell Biol. 2006, 175, 121–133.pl_PL
dc.referencesGanley, I.G.; du Lam, H.; Wang, J.; Ding, X.; Chen, S.; Jiang, X. ULK1–ATG13–FIP200 complex mediates mTOR signaling and is essential for autophagy. J. Biol. Chem. 2009, 284, 12297–12305.pl_PL
dc.referencesHosokawa, N.; Hara, T.; Kaizuka, T.; Kishi, C.; Takamura, A.; Miura, Y.; Iemura, S.; Natsume, T.; Takehana, K.; Yamada, N.; et al. Nutrient-dependent mTORC1 association with the ULK1–Atg13–FIP200 complex required for autophagy. Mol. Biol. Cell 2009, 20, 1981–1991.pl_PL
dc.referencesJung, C.H.; Jun, C.B.; Ro, S.H.; Kim, Y.M.; Otto, N.M.; Cao, J.; Kundu, M.; Kim, D.H. ULK–Atg13–FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol. Biol. Cell 2009, 20, 1992–2003.pl_PL
dc.referencesAlers, S.; Löffler, A.S.; Wesselborg, S.; Stork, B. The incredible ULKs. Cell Commun. Signal. 2012, 10, 7.pl_PL
dc.referencesBae, H.; Guan, J.L. Suppression of autophagy by FIP200 deletion impairs DNA damage repair and increases cell death upon treatments with anticancer agents. Mol. Cancer Res. 2011, 9, 1232–1241.pl_PL
dc.referencesMoscat, J.; Diaz-Meco, M.T.; Wooten, M.W. Signal integration and diversification through the p62 scaffold protein. Trends Biochem. Sci. 2007, 32, 95–100.pl_PL
dc.referencesVadlamudi, R.K.; Joung, I.; Strominger, J.L.; Shin, J. p62, a phosphotyrosine-independent ligand of the SH2 domain of p56lck, belongs to a new class of ubiquitin-binding proteins. J. Biol. Chem. 1996, 271, 20235–20237.pl_PL
dc.referencesPohl, C.; Jentsch, S. Midbody ring disposal by autophagy is a post-abscission event of cytokinesis. Nat. Cell Biol. 2009, 11, 65–70.pl_PL
dc.referencesPankiv, S.; Clausen, T.H.; Lamark, T.; Brech, A.; Bruun, J.A.; Outzen, H.; Øvervatn, A.; Bjørkøy, G.; Johansen, T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J. Biol. Chem. 2007, 282, 24131–24145.pl_PL
dc.referencesWooten, M.W.; Geetha, T.; Babu, J.R.; Seibenhener, M.L.; Peng, J.; Cox, N.; Diaz-Meco, M.T.; Moscat, J. Essential role of sequestosome 1/p62 in regulating accumulation of Lys63-ubiquitinated proteins. J. Biol. Chem. 2008, 283, 6783–6789.pl_PL
dc.referencesPankiv, S.; Lamark, T.; Bruun, J.A.; Øvervatn, A.; Bjørkøy, G.; Johansen, T. Nucleocytoplasmic shuttling of p62/SQSTM1 and its role in recruitment of nuclear polyubiquitinated proteins to promyelocytic leukemia bodies. J. Biol. Chem. 2010, 285, 5941–5953.pl_PL
dc.referencesLallemand-Breitenbach, V.; de The, H. PML nuclear bodies. Cold Spring Harb. Perspect. Biol. 2010, 2, a000661.pl_PL
dc.referencesMathew, R.; Kongara, S.; Beaudoin, B.; Karp, C.M.; Bray, K.; Degenhardt, K.; Chen, G.; Jin, S.; White, E. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev. 2007, 21, 1367–1381.pl_PL
dc.referencesScheffler, I.E. Mitochondria,, 2nd ed.; Wiley: Hoboken, NJ, USA, 2008.pl_PL
dc.referencesGreaves, L.C.; Reeve, A.K.; Taylor, R.W.; Turnbull, D.M. Mitochondrial DNA and disease. J. Pathol. 2012, 226, 274–286.pl_PL
dc.referencesKim, I.; Rodriguez-Enriquez, S.; Lemasters, J.J. Selective degradation of mitochondria by mitophagy. Arch. Biochem. Biophys. 2007, 462, 245–253.pl_PL
dc.referencesAshford, T.P.; Porter, K.R. Cytoplasmic components in hepatic cell lysosomes. J. Cell Biol. 1962, 12, 198–202.pl_PL
dc.referencesNovak, I.; Kirkin, V.; McEwan, D.G.; Zhang, J.; Wild, P.; Rozenknop, A.; Rogov, V.; Löhr, F.; Popovic, D.; Occhipinti, A.; et al. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep. 2010, 11, 45–51.pl_PL
dc.referencesDing, W.X.; Ni, H.M.; Li, M.; Liao, Y.; Chen, X.; Stolz, D.B.; Dorn, G.W., 2nd.; Yin, X.M. Nix is critical to two distinct phases of mitophagy, reactive oxygen species-mediated autophagy induction and Parkin-ubiquitin-p62-mediated mitochondrial priming. J. Biol. Chem. 2010, 285, 27879–27890.pl_PL
dc.referencesMatsuda, S.; nakanishi, A.; Minami, A.; Wada, Y.; Kitagishi, Y. Functions and characteristics of PINK1 and Parkin in cancer. Front. Biosci. 2015, 20, 491–501.pl_PL
dc.referencesGeisler, S.; Holmström, K.M.; Treis, A.; Skujat, D.; Weber, S.S.; Fiesel., F.C.; Kahle, P.J.; Springer, W. The PINK1/Parkin-mediated mitophagy is compromised by PD-associated mutations. Autophagy 2010, 6, 871–878.pl_PL
dc.referencesNarendra, D.; Tanaka, A.; Suen, D.F.; Youle, R.J. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J. Cell Biol. 2008, 183, 795–803.pl_PL
dc.referencesVives-Bauza, C.; Zhou, C.; Huang, Y.; Cui, M.; de Vries, R.L.; Kim, J.; May, J.; Tocilescu, M.A.; Liu, W.; Ko, H.S.; et al. PINK1-dependent recruitment of Parkin to mitochondria in mitophagy. Proc. Natl. Acad. Sci. USA 2010, 107, 378–383.pl_PL
dc.referencesVila, M.; Ramonet, D.; Perier, C. Mitochondrial alterations in Parkinson’s disease: New clues. J. Neurochem. 2008, 107, 317–328.pl_PL
dc.referencesGil, J.M.; Rego, A.C. Mechanisms of neurodegeneration in Huntington’s disease. Eur. J. Neurosci. 2008, 27, 2803–2820.pl_PL
dc.referencesBess, A.S.; Ryde, I.T.; Hinton, D.E.; Meyer, J.N. UVC-induced mitochondrial degradation via autophagy correlates with mtDNA damage removal in primary human fibroblasts. J. Biochem. Mol. Toxicol. 2013, 27, 28–41.pl_PL
dc.referencesScheibye-Knudsen, M.; Ramamoorthy, M.; Sykora, P.; Maynard, S.; Lin, P.C.; Minor, R.K.; Wilson, D.M., 3rd.; Cooper, M.; Spencer, R.; de Cabo, R.; et al. Cockayne syndrome group B protein prevents the accumulation of damaged mitochondria by promoting mitochondrial autophagy. J. Exp. Med. 2012, 209, 855–869.pl_PL
dc.referencesMonick, M.M.; Powers, L.S.; Walters, K.; Lovan, N.; Zhang, M.; Gerke, A.; Hansdottir, S.; Hunninghake, G.W. Identification of an autophagy defect in smokers’ alveolar macrophages. J. Immunol. 2010, 185, 5425–5435.pl_PL
dc.referencesChen, L.H.; Chu, P.M.; Lee, Y.J.; Tu, P.H.; Chi, C.W.; Lee, H.C.; Chiou, S.H. Targeting protective autophagy exacerbates UV-triggered apoptotic cell death. Int. J. Mol. Sci. 2012, 13, 1209–1224.pl_PL
dc.referencesRodriguez-Hernandez, A.; Cordero, M.D.; Salviati, L.; Artuch, R.; Pineda, M.; Briones, P.; Gómez Izquierdo, L.; Cotán, D.; Navas, P.; Sánchez-Alcázar, J.A.; et al. Coenzyme Q deficiency triggers mitochondria degradation by mitophagy. Autophagy 2009, 5, 19–32.pl_PL
dc.referencesCotan, D.; Cordero, M.D.; Garrido-Maraver, J.; Oropesa-Ávila, M.; Rodríguez-Hernández, A.; Gómez Izquierdo, L.; de la Mata, M.; de Miguel, M.; Lorite, J.B.; Infante, E.R.; et al. Secondary coenzyme Q10 dficiency triggers mitochondria degradation by mitophagy in MELAS fibroblasts. FASEB J. 2011, 25, 2669–2687.pl_PL
dc.referencesPriault, M.; Salin, B.; Schaeffer, J.; Vallette, F.M.; di Rago, J.P.; Martinou, J.C. Impairing the bioenergetic status and the biogenesis of mitochondria triggers mitophagy in yeast. Cell Death Differ. 2005, 12, 1613–1621.pl_PL
dc.referencesElmore, S.P.; Qian, T.; Grissom, S.F.; Lemasters, J.J. The mitochondrial permeability transition initiates autophagy in rat hepatocytes. FASEB J. 2001, 15, 2286–2287.pl_PL
dc.referencesGu, Y.; Wang, C.; Cohen, A. Effect of IGF-1 on the balance between autophagy of dysfunctional mitochondria and apoptosis. FEBS Lett. 2004, 577, 357–360.pl_PL
dc.referencesAmaravadi, R.K.; Lippincott-Schwartz, J.; Yin, X.M.; Weiss, W.A.; Takebe, N.; Timmer, W.; di Paola, R.S.; Lotze, M.T.; White, E. Principles and current strategies for targeting autophagy for cancer treatment. Clin. Cancer Res. 2011, 17, 654–666.pl_PL
dc.referencesEdinger, A.L.; Thompson, C.B. Death by design: Apoptosis, necrosis and autophagy. Curr. Opin. Cell Biol. 2004, 16, 663–669.pl_PL
dc.referencesMaycotte, P.; Thorburn, A. Autophagy and cancer therapy. Cancer Biol. Ther. 2011, 11, 127–137.pl_PL
dc.referencesKenzelmann Broz, D.; Spano Mello, S.; Bieging, K.T.; Jiang, D.; Dusek, R.L.; Brady, C.A.; Sidow, A.; Attardi, L.D. Global genomic profiling reveals an extensive p53-regulated autophagy program contributing to key p53 responses. Genes Dev. 2013, 27, 1016–1031.pl_PL
dc.referencesLiu, H.; He, Z.; Simon, H.U. Targeting autophagy as a potential therapeutic approach for melanoma therapy. Semin. Cancer Biol. 2013, 23, 352–360.pl_PL
dc.referencesCui, J.; Gong, Z.; Shen, H.M. The role of autophagy in liver cancer: Molecular mechanisms and potential therapeutic targets. Biochim. Biophys. Acta 2013, 1836, 15–26.pl_PL
dc.contributor.authorEmailpczarny@biol.uni.lodz.plpl_PL


Pliki tej pozycji

Thumbnail
Thumbnail

Pozycja umieszczona jest w następujących kolekcjach

Pokaż uproszczony rekord

Uznanie autorstwa 3.0 Polska
Poza zaznaczonymi wyjątkami, licencja tej pozycji opisana jest jako Uznanie autorstwa 3.0 Polska