Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(3)2022 Jan 30.
Article in English | MEDLINE | ID: mdl-35158994

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor in adults. Poly (ADP-ribose) polymerase inhibitors (PARPi) represent a new class of anti-neoplastic drugs. In the current study, we have characterized the mechanism by which glioblastoma cells evade the effect of PARPi as anti-tumor agents. We have found that suppression of PARP activity exerts an anti-stemness effect and has a dual impact on autophagy, inducing its activation in the first 24 h (together with down-regulation of the pro-survival mTOR pathway) and preventing autophagosomes fusion to lysosomes at later time-points, in primary glioma cells. In parallel, PARPi triggered the synthesis of lipid droplets (LDs) through ACC-dependent activation of de novo fatty acids (FA) synthesis. Notably, inhibiting ß-oxidation and blocking FA utilization, increased PARPi-induced glioma cell death while treatment with oleic acid (OA) prevented the anti-glioma effect of PARPi. Moreover, LDs fuel glioma cells by inducing pro-survival lipid consumption as confirmed by quantitation of oxygen consumption rates using Seahorse respirometry in presence or absence of OA. In summary, we uncover a novel mechanism by which glioblastoma escapes to anti-tumor agents through metabolic reprogramming, inducing the synthesis and utilization of LDs as a pro-survival strategy in response to PARP inhibition.

2.
J Exp Clin Cancer Res ; 40(1): 144, 2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33910596

ABSTRACT

Tankyrase 1 (TNKS1) and tankyrase 2 (TNKS2) are two homologous proteins that are gaining increasing importance due to their implication in multiple pathways and diseases such as cancer. TNKS1/2 interact with a large variety of substrates through the ankyrin (ANK) domain, which recognizes a sequence present in all the substrates of tankyrase, called Tankyrase Binding Motif (TBM). One of the main functions of tankyrases is the regulation of protein stability through the process of PARylation-dependent ubiquitination (PARdU). Nonetheless, there are other functions less studied that are also essential in order to understand the role of tankyrases in many pathways. In this review, we concentrate in different tankyrase substrates and we analyze in depth the biological consequences derived of their interaction with TNKS1/2. We also examine the concept of both canonical and non-canonical TBMs and finally, we focus on the information about the role of TNKS1/2 in different tumor context, along with the benefits and limitations of the current TNKS inhibitors targeting the catalytic PARP domain and the novel strategies to develop inhibitors against the ankyrin domain. Available data indicates the need for further deepening in the knowledge of tankyrases to elucidate and improve the current view of the role of these PARP family members and get inhibitors with a better therapeutic and safety profile.


Subject(s)
Neoplasms/therapy , Tankyrases/metabolism , Humans
3.
Redox Biol ; 41: 101885, 2021 05.
Article in English | MEDLINE | ID: mdl-33581682

ABSTRACT

BACKGROUND: The adaptation to hypoxia is mainly controlled by the HIF transcription factors. Increased expression/activity of HIF-1α correlates with poor prognosis in cancer patients. PARP-1 inhibitors are used in the clinic to treat BRCAness breast/ovarian cancer and have been shown to regulate the hypoxic response; therefore, their use could be expanded. METHODS: In this work by integrating molecular/cell biology approaches, genome-wide ChIP-seq, and patient samples, we elucidate the extent to which PARP-1 exerts control over HIF-1-regulated genes. RESULTS: In human melanoma, PARP-1 and HIF-1α expression are strongly associated. In response to a hypoxic challenge poly(ADP-ribose) (PAR) is synthesized, HIF-1α is post-transcriptionally modified (PTM) and stabilized by PARylation at specific K/R residues located at its C-terminus. Using an unbiased ChIP-seq approach we demonstrate that PARP-1 dictates hypoxia-dependent HIF-recruitment to chromatin in a range of HIF-regulated genes while analysis of HIF-binding motifs (RCGTG) reveals a restriction on the recognition of hypoxia responsive elements in the absence of PARP-1. Consequently, the cells are poorly adapted to hypoxia, showing a reduced fitness during hypoxic induction. CONCLUSIONS: These data characterize the fine-tuning regulation by PARP-1/PARylation of HIF activation and suggest that PARP inhibitors might have therapeutic potential against cancer types displaying HIF-1α over-activation.


Subject(s)
Breast Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Cell Hypoxia , Chromatin , Female , Humans , Hypoxia
4.
Cell Death Dis ; 11(11): 954, 2020 11 06.
Article in English | MEDLINE | ID: mdl-33159039

ABSTRACT

Parp3 is a member of the Poly(ADP-ribose) polymerase (Parp) family that has been characterized for its functions in strand break repair, chromosomal rearrangements, mitotic segregation and tumor aggressiveness. Yet its physiological implications remain unknown. Here we report a central function of Parp3 in the regulation of redox homeostasis in continuous neurogenesis in mice. We show that the absence of Parp3 provokes Nox4-induced oxidative stress and defective mTorc2 activation leading to inefficient differentiation of post-natal neural stem/progenitor cells to astrocytes. The accumulation of ROS contributes to the decreased activity of mTorc2 as a result of an oxidation-induced and Fbxw7-mediated ubiquitination and degradation of Rictor. In vivo, mTorc2 signaling is compromised in the striatum of naïve post-natal Parp3-deficient mice and 6 h after acute hypoxia-ischemia. These findings reveal a physiological function of Parp3 in the tight regulation of striatal oxidative stress and mTorc2 during astrocytic differentiation and in the acute phase of hypoxia-ischemia.


Subject(s)
Astrocytes/cytology , Cell Differentiation , Mechanistic Target of Rapamycin Complex 2/metabolism , NADPH Oxidase 4/metabolism , Neurogenesis , Poly(ADP-ribose) Polymerases/physiology , Reactive Oxygen Species/metabolism , Animals , Astrocytes/metabolism , Gene Expression Regulation , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Mice, Knockout , NADPH Oxidase 4/genetics , Signal Transduction
5.
Oxid Med Cell Longev ; 2019: 2641712, 2019.
Article in English | MEDLINE | ID: mdl-31281570

ABSTRACT

Autophagy is considered to be the primary degradative pathway that takes place in all eukaryotic cells. Morphologically, the autophagy pathway refers to a process by which cytoplasmic portions are delivered to double-membrane organelles, called autophagosomes, to fuse with lysosomes for bulk degradation. Autophagy, as a prosurvival mechanism, can be stimulated by different types of cellular stress such as nutrient deprivation, hypoxia, ROS, pH, DNA damage, or ER stress, promoting adaptation of the cell to the changing and hostile environment. The functional relevance of autophagy in many diseases such as cancer or neurodegenerative diseases remains controversial, preserving organelle function and detoxification and promoting cell growth, although in other contexts, autophagy could suppress cell expansion. Poly(ADP-ribosyl)ation (PARylation) is a covalent and reversible posttranslational modification (PTM) of proteins mediated by Poly(ADP-ribose) polymerases (PARPs) with well-described functions in DNA repair, replication, genome integrity, cell cycle, and metabolism. Herein, we review the current state of PARP1 activation and PARylation in starvation-induced autophagy.


Subject(s)
Nutrients/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Poly ADP Ribosylation/genetics , Autophagy , Humans , Signal Transduction
6.
Cell Cycle ; 18(12): 1295-1301, 2019 06.
Article in English | MEDLINE | ID: mdl-31095444

ABSTRACT

Poly(ADP-ribose) polymerase 3 (PARP3) is the third member of the PARP family that catalyze a post-translational modification of proteins to promote, control or adjust numerous cellular events including genome integrity, transcription, differentiation, cell metabolism or cell death. In the late years, PARP3 has been specified for its primary functions in programmed and stress-induced double-strand break repair, chromosomal rearrangements, transcriptional regulation in the zebrafish and mitotic segregation. Still, deciphering the therapeutic value of its inhibition awaits additional investigations. In this review, we discuss the newest advancements on the specific functions of PARP3 in cancer aggressiveness exemplifying the relevance of its selective inhibition for cancer therapy.


Subject(s)
Molecular Targeted Therapy , Neoplasms/drug therapy , Poly(ADP-ribose) Polymerases/metabolism , Adenosine Diphosphate Ribose/metabolism , Animals , DNA Repair , Epithelial-Mesenchymal Transition , Humans
7.
Cell Death Differ ; 26(9): 1615-1630, 2019 09.
Article in English | MEDLINE | ID: mdl-30442946

ABSTRACT

PARP3 has been shown to be a key driver of TGFß-induced epithelial-to-mesenchymal transition (EMT) and stemness in breast cancer cells, emerging as an attractive therapeutic target. Nevertheless, the therapeutic value of PARP3 inhibition has not yet been assessed. Here we investigated the impact of the absence of PARP3 or its inhibition on the tumorigenicity of BRCA1-proficient versus BRCA1-deficient breast cancer cell lines, focusing on the triple-negative breast cancer subtype (TNBC). We show that PARP3 knockdown exacerbates centrosome amplification and genome instability and reduces survival of BRCA1-deficient TNBC cells. Furthermore, we engineered PARP3-/- BRCA1-deficient or BRCA1-proficient TNBC cell lines using the CRISPR/nCas9D10A gene editing technology and demonstrate that the absence of PARP3 selectively suppresses the growth, survival and in vivo tumorigenicity of BRCA1-deficient TNBC cells, mechanistically via effects associated with an altered Rictor/mTORC2 signaling complex resulting from enhanced ubiquitination of Rictor. Accordingly, PARP3 interacts with and ADP-ribosylates GSK3ß, a positive regulator of Rictor ubiquitination and degradation. Importantly, these phenotypes were rescued by re-expression of a wild-type PARP3 but not by a catalytic mutant, demonstrating the importance of PARP3's catalytic activity. Accordingly, reduced survival and compromised Rictor/mTORC2 signaling were also observed using a cell-permeable PARP3-specific inhibitor. We conclude that PARP3 and BRCA1 are synthetic lethal and that targeting PARP3's catalytic activity is a promising therapeutic strategy for BRCA1-associated cancers via the Rictor/mTORC2 signaling pathway.


Subject(s)
BRCA1 Protein/genetics , Cell Cycle Proteins/genetics , Poly(ADP-ribose) Polymerases/genetics , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Signal Transduction , Transforming Growth Factor beta/genetics , Triple Negative Breast Neoplasms/pathology
8.
Oncotarget ; 7(39): 64109-64123, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27579892

ABSTRACT

Several members of the Poly(ADP-ribose) polymerase (PARP) family are essential regulators of genome integrity, actively prospected as drug targets for cancer therapy. Among them, PARP3 is well characterized for its functions in double-strand break repair and mitotis. Here we report that PARP3 also plays an integral role in TGFß and reactive oxygen species (ROS) dependent epithelial-to-mesenchymal transition (EMT) and stem-like cell properties in human mammary epithelial and breast cancer cells. PARP3 expression is higher in breast cancer cells of the mesenchymal phenotype and correlates with the expression of the mesenchymal marker Vimentin while being in inverse correlation with the epithelial marker E-cadherin. Furthermore, PARP3 expression is significantly upregulated during TGFß-induced EMT in various human epithelial cells. In line with this observation, PARP3 depletion alters TGFß-dependent EMT of mammary epithelial cells by preventing the induction of the Snail-E-cadherin axis, the dissolution of cell junctions, the acquisition of cell motility and chemoresistance. PARP3 responds to TGFß-induced ROS to promote a TG2-Snail-E-cadherin axis during EMT. Considering the link between EMT and cancer stem cells, we show that PARP3 promotes stem-like cell properties in mammary epithelial and breast cancer cells by inducing the expression of the stem cell markers SOX2 and OCT4, by increasing the proportion of tumor initiating CD44high/CD24low population and the formation of tumor spheroid bodies, and by promoting stem cell self-renewal. These findings point to a novel role of PARP3 in the control of TGFß-induced EMT and acquisition of stem-like cell features and further motivate efforts to identify PARP3 specific inhibitors.


Subject(s)
Breast Neoplasms/enzymology , Cadherins/metabolism , Cell Cycle Proteins/metabolism , Epithelial-Mesenchymal Transition , GTP-Binding Proteins/metabolism , Mammary Glands, Human/enzymology , Neoplastic Stem Cells/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Transglutaminases/metabolism , A549 Cells , Antigens, CD , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , CD24 Antigen/metabolism , Cadherins/genetics , Cell Cycle Proteins/genetics , Cell Movement , Cell Self Renewal , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition/drug effects , Etoposide/pharmacology , Female , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Hyaluronan Receptors/metabolism , Mammary Glands, Human/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Octamer Transcription Factor-3/metabolism , Phenotype , Poly(ADP-ribose) Polymerases/genetics , Protein Glutamine gamma Glutamyltransferase 2 , RNA Interference , SOXB1 Transcription Factors/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Spheroids, Cellular , Time Factors , Topoisomerase II Inhibitors/pharmacology , Transfection , Transglutaminases/genetics
9.
PLoS Genet ; 9(6): e1003531, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23785295

ABSTRACT

PARP inhibition can induce anti-neoplastic effects when used as monotherapy or in combination with chemo- or radiotherapy in various tumor settings; however, the basis for the anti-metastasic activities resulting from PARP inhibition remains unknown. PARP inhibitors may also act as modulators of tumor angiogenesis. Proteomic analysis of endothelial cells revealed that vimentin, an intermediary filament involved in angiogenesis and a specific hallmark of EndoMT (endothelial to mesenchymal transition) transformation, was down-regulated following loss of PARP-1 function in endothelial cells. VE-cadherin, an endothelial marker of vascular normalization, was up-regulated in HUVEC treated with PARP inhibitors or following PARP-1 silencing; vimentin over-expression was sufficient to drive to an EndoMT phenotype. In melanoma cells, PARP inhibition reduced pro-metastatic markers, including vasculogenic mimicry. We also demonstrated that vimentin expression was sufficient to induce increased mesenchymal/pro-metastasic phenotypic changes in melanoma cells, including ILK/GSK3-ß-dependent E-cadherin down-regulation, Snail1 activation and increased cell motility and migration. In a murine model of metastatic melanoma, PARP inhibition counteracted the ability of melanoma cells to metastasize to the lung. These results suggest that inhibition of PARP interferes with key metastasis-promoting processes, leading to suppression of invasion and colonization of distal organs by aggressive metastatic cells.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Melanoma, Experimental/genetics , Poly(ADP-ribose) Polymerases/genetics , Vimentin , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Breast Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Dogs , Endothelial Cells/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Melanoma, Experimental/pathology , Mice , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerase Inhibitors , Vimentin/genetics , Vimentin/metabolism
10.
Mol Aspects Med ; 34(6): 1153-67, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23416893

ABSTRACT

Poly(ADP-ribosyl)ation (PARylation) is a reversible protein modification carried out by the concerted actions of poly(ADP-ribose) polymerase (PARP) enzymes and poly(ADP-ribose) (PAR) decomposing enzymes such as PAR glycohydrolase (PARG) and ADP-ribosyl hydrolase 3 (ARH3). Reversible PARylation is a pleiotropic regulator of various cellular functions but uncontrolled PARP activation may also lead to cell death. The cellular demise pathway mediated by PARylation in oxidatively stressed cells has been described almost thirty years ago. However, the underlying molecular mechanisms have only begun to emerge relatively recently. PARylation has been implicated in necroptosis, autophagic cell death but its role in extrinsic and intrinsic apoptosis appears to be less predominant and depends largely on the cellular model used. Currently, three major pathways have been made responsible for PARP-mediated necroptotic cell death: (1) compromised cellular energetics mainly due to depletion of NAD, the substrate of PARPs; (2) PAR mediated translocation of apoptosis inducing factor (AIF) from mitochondria to nucleus (parthanatos) and (3) a mostly elusive crosstalk between PARylation and cell death/survival kinases and phosphatases. Here we review how these PARP-mediated necroptotic pathways are intertwined, how PARylation may contribute to extrinsic and intrinsic apoptosis and discuss recent developments on the role of PARylation in autophagy and autophagic cell death.


Subject(s)
Cell Death/physiology , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Signal Transduction , Animals , Cell Death/genetics , Gene Expression Regulation , Gene Regulatory Networks/physiology , Humans , Mitochondria/metabolism , Protein Binding , Protein Processing, Post-Translational
11.
Cell Res ; 22(7): 1181-98, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22525338

ABSTRACT

In response to nutrient stress, cells start an autophagy program that can lead to adaptation or death. The mechanisms underlying the signaling from starvation to the initiation of autophagy are not fully understood. In the current study we show that the absence or inactivation of PARP-1 strongly delays starvation-induced autophagy. We have found that DNA damage is an early event of starvation-induced autophagy as measured by γ-H2AX accumulation and comet assay, with PARP-1 knockout cells displaying a reduction in both parameters. During starvation, ROS-induced DNA damage activates PARP-1, leading to ATP depletion (an early event after nutrient deprivation). The absence of PARP-1 blunted AMPK activation and prevented the complete loss of mTOR activity, leading to a delay in autophagy. PARP-1 depletion favors apoptosis in starved cells, suggesting a pro-survival role of autophagy and PARP-1 activation after nutrient deprivation. In vivo results show that neonates of PARP-1 mutant mice subjected to acute starvation, also display deficient liver autophagy, implying a physiological role for PARP-1 in starvation-induced autophagy. Thus, the PARP signaling pathway is a key regulator of the initial steps of autophagy commitment following starvation.


Subject(s)
Autophagy/physiology , DNA Damage/physiology , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Autophagy/genetics , Blotting, Western , Cell Line , Cell Survival/genetics , Cell Survival/physiology , DNA Damage/genetics , Fluorescent Antibody Technique , Mice , Microscopy, Fluorescence , Models, Biological , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics
12.
Free Radic Biol Med ; 47(1): 13-26, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19362586

ABSTRACT

Poly(ADP-ribose) polymerases (PARPs) are defined as cell signaling enzymes that catalyze the transfer of ADP-ribose units from NAD(+) to a number of acceptor proteins. PARP-1, the best characterized member of the PARP family, which currently comprises 18 members, is an abundant nuclear enzyme implicated in cellular responses to DNA injury provoked by genotoxic stress. PARP is involved in DNA repair and transcriptional regulation and is now recognized as a key regulator of cell survival and cell death as well as a master component of a number of transcription factors involved in tumor development and inflammation. PARP-1 is essential to the repair of DNA single-strand breaks via the base excision repair pathway. Inhibitors of PARP-1 have been shown to enhance the cytotoxic effects of ionizing radiation and DNA-damaging chemotherapy agents, such as the methylating agents and topoisomerase I inhibitors. There are currently at least five PARP inhibitors in clinical trial development. Recent in vitro and in vivo evidence suggests that PARP inhibitors could be used not only as chemo/radiotherapy sensitizers, but also as single agents to selectively kill cancers defective in DNA repair, specifically cancers with mutations in the breast cancer-associated genes (BRCA1 and BRCA2). PARP becomes activated in response to oxidative DNA damage and depletes cellular energy pools, thus leading to cellular dysfunction in various tissues. The activation of PARP may also induce various cell death processes and promotes an inflammatory response associated with multiple organ failure. Inhibition of PARP activity is protective in a wide range of inflammatory and ischemia-reperfusion-associated diseases, including cardiovascular diseases, diabetes, rheumatoid arthritis, endotoxic shock, and stroke. The aim of this review is to overview the emerging data in the literature showing the role of PARP in the pathogenesis of cancer and inflammatory diseases and unravel the solid body of literature that supports the view that PARP is an important target for therapeutic intervention in critical illness.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Autoimmune Diseases/drug therapy , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , DNA Mismatch Repair/genetics , Neovascularization, Pathologic/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors , Poly(ADP-ribose) Polymerases/therapeutic use , Radiation-Sensitizing Agents/therapeutic use , Reperfusion Injury/drug therapy , BRCA1 Protein/deficiency , BRCA2 Protein/deficiency , Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/radiation effects , Clinical Trials as Topic , Combined Modality Therapy , DNA Mismatch Repair/drug effects , DNA Mismatch Repair/radiation effects , Drug-Related Side Effects and Adverse Reactions , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Oxidative Stress/drug effects , Oxidative Stress/genetics , Oxidative Stress/radiation effects , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/immunology , Radiotherapy/adverse effects
13.
Autophagy ; 5(1): 61-74, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19001878

ABSTRACT

Autophagy is a lysosome-dependent degradative pathway frequently activated in tumor cells treated with chemotherapy or radiation. PARP-1 has been implicated in different pathways leading to cell death and its inhibition potentiates chemotherapy-induced cell death. Whether PARP-1 participates in the cell's decision to commit to autophagy following DNA damage is still not known. To address this issue PARP-1 wild-type and deficient cells have been treated with a dose of doxorubicin that induces autophagy. Electron microscopy examination and GFP-LC3 transfection revealed autophagic vesicles and increased expression of genes involved in autophagy (bnip-3, cathepsin b and l and beclin-1) in wild-type cells treated with doxo but not in parp-1(-/-) cells or cells treated with a PARP inhibitor. Mechanistically the lack of autophagic features in PARP-1 deficient/PARP inhibited cells is attributed to prevention of ATP and NAD(+) depletion and to the activation of the key autophagy regulator mTOR. Pharmacological or genetical inhibition of autophagy results in increased cell death, suggesting a protective role of autophagy induced by doxorubicin. These results suggest that autophagy might be cytoprotective during the response to DNA damage and suggest that PARP-1 activation is involved in the cell's decision to undergo autophagy.


Subject(s)
Autophagy , DNA Damage , Poly(ADP-ribose) Polymerases/metabolism , 1-Naphthylamine/analogs & derivatives , 1-Naphthylamine/pharmacology , 3T3 Cells , Adenosine Triphosphate/deficiency , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Autophagy/drug effects , Autophagy/genetics , Autophagy-Related Protein 5 , Beclin-1 , Cell Survival/drug effects , Doxorubicin/pharmacology , Enzyme Activation/drug effects , Gene Deletion , Mice , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/ultrastructure , Models, Biological , NAD/deficiency , Naphthalimides/pharmacology , Necrosis/enzymology , Poly(ADP-ribose) Polymerase Inhibitors , Protein Kinases/metabolism , Proteins/metabolism , Quinolones/pharmacology , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , TOR Serine-Threonine Kinases , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...