Structural basis for allosteric PARP-1 retention on DNA breaks

DNA death grip Poly(ADP-ribose) polymerase–1 (PARP-1) binds to DNA breaks and recruits DNA repair components. Cancer-killing PARP-1 inhibitor (PARPi) compounds all block the same catalytic site but exhibit vastly different efficacy. Zandarashvili et al. investigated the molecular impact of PARPi binding to PARP-1 (see the Perspective by Slade and Eustermann). Different PARPi molecules perturb PARP-1 allostery in diverse manners: Some drive allostery to promote release of PARP-1 from DNA, and others drive allostery to promote retention. These insights help explain the different efficacies in the clinic and enable conversion of a pro-release, ineffective cancer-killing compound to a pro-retention, more effective PARPi. Science, this issue p. eaax6367; see also p. 30 Different poly(ADP-ribose) polymerase–1 (PARP-1) inhibitors used to treat cancer either trap or release PARP-1 at DNA break sites. INTRODUCTION Poly(ADP-ribose) polymerase–1 (PARP-1) is an abundant enzyme in the cell nucleus that regulates genome repair by binding to DNA damage sites and creating the poly(ADP-ribose) posttranslational modification. PARP-1 hyperactivity leads to cell stress or death associated with many prominent diseases (e.g., cardiovascular disease and several common neurodegenerative disorders). PARP-1 has notably emerged as an effective clinical target for a growing list of cancers. Clinical PARP-1 inhibitor (PARPi) compounds all bind at the same location at the catalytic center of the enzyme to block the binding of substrate nicotinamide adenine dinucleotide (NAD+) and prevent poly(ADP-ribose) production, yet they exhibit vastly different outcomes in tumor cell killing and efficacy in the clinic—a paradox that has confounded the development of PARPi. The resolution of this paradox likely lies in the realization that the most effective PARPi compounds trap PARP-1 at the site of a DNA break, generating a lesion that becomes cytotoxic, especially in tumor cells with deficiencies in the repair of DNA strand breaks. RATIONALE The molecular roots of PARP-1 trapping on DNA remain poorly understood. We focused on the retention of PARP-1 on damaged DNA, examining a panel of PARPi that included those currently approved for clinical use. Solution biophysical approaches, especially hydrogen/deuterium exchange mass spectrometry (HXMS), combined with x-ray structures and a battery of biochemical assays, were used to interrogate the molecular impact of PARPi binding to PARP-1 engaged on sites of DNA damage. Structure-guided modification of PARPi through medicinal chemistry was combined with chromatin fractionation to monitor trapped PARP-1 and with cell survival assays to assess PARPi efficacy, so as to probe the molecular underpinnings of the variable outcomes between clinical PARPi. RESULTS HXMS experiments revealed that a critical allosteric regulatory domain of PARP-1, the helical domain (HD), is affected in distinct ways depending on the particular PARPi engaged in the NAD+-binding site adjacent to the HD. Certain PARPi destabilized specific HD regions, some had no effect on the HD, and others actually stabilized regions of the HD. PARPi that destabilized the HD increased PARP-1 affinity for DNA and retained PARP-1 on DNA breaks. Conversely, PARPi that stabilized the HD decreased PARP-1 affinity for DNA breaks. PARPi molecules were thus classified into three types: type I, allosteric pro-retention on DNA; type II, non-allosteric; and type III, allosteric pro-release from DNA. X-ray structure analysis identified PARPi contacts with the HD structural element helix αF, which was established to be the discriminating factor between the types of PARPi. We found that type I PARPi contact helix αF to initiate an allosteric chain reaction that travels ~40 Å through the multidomain PARP-1 molecule and culminates in increased DNA binding affinity. Structure-guided mutagenesis of helix αF disrupted PARPi contacts and abrogated the allosteric effects of a type I inhibitor, transforming it into a non-allosteric type II inhibitor. Other mutations that disrupted PARP-1 allostery, including one identified in a de novo PARPi-resistant patient with ovarian cancer, also prevented type I PARPi from retaining PARP-1 on a DNA break. Type III PARPi influenced PARP-1 allostery in a manner that reduced DNA binding and favored DNA release. Structure-inspired modification of a pro-release (type III) inhibitor converted it to a pro-retention (type I) inhibitor that conferred potent PARP-1 trapping within the cellular context and increased its ability to kill cancer cells. CONCLUSION Our findings establish the impact of clinical PARPi on PARP-1 allostery and demonstrate that allostery plays a critical role in cellular PARP-1 trapping and can increase potency toward cancer cell killing. The results illuminate the molecular basis for the fine-tuning of PARPi to achieve allosteric effects and to influence PARP-1 retention on DNA damage and trapping on chromatin in cells. In contrast to cancer, other diseases would seem to benefit from PARP-1 inhibition but not cell death (e.g., cardiovascular disease, neurodegenerative diseases, and inflammation). Our studies provide the molecular understanding and the appropriate toolset to create and evaluate tunable PARPi for clinical applications where PARP-1 trapping and associated cytotoxicity are either desirable or undesirable in specific patients. PARPi impact on PARP-1 allostery. PARP-1 (tan) uses multiple domains to detect DNA breaks, and DNA damage detection is allosterically coupled to poly(ADP-ribose) production. PARPi bind to the catalytic domain to inhibit PARP-1 activity. Type I PARPi influence PARP-1 allostery and retain PARP-1 on DNA (left, UKTT15 in green), whereas type III PARPi perturb PARP-1 allostery and release PARP-1 from DNA (right, veliparib in red). Type II PARPi do not influence PARP-1 allostery. The success of poly(ADP-ribose) polymerase–1 (PARP-1) inhibitors (PARPi) to treat cancer relates to their ability to trap PARP-1 at the site of a DNA break. Although different forms of PARPi all target the catalytic center of the enzyme, they have variable abilities to trap PARP-1. We found that several structurally distinct PARPi drive PARP-1 allostery to promote release from a DNA break. Other inhibitors drive allostery to retain PARP-1 on a DNA break. Further, we generated a new PARPi compound, converting an allosteric pro-release compound to a pro-retention compound and increasing its ability to kill cancer cells. These developments are pertinent to clinical applications where PARP-1 trapping is either desirable or undesirable.

[1]  A. Ashworth,et al.  PARP inhibition enhances tumor cell–intrinsic immunity in ERCC1-deficient non–small cell lung cancer , 2019, The Journal of clinical investigation.

[2]  S. C. Panchal,et al.  PARP1 Trapping by PARP Inhibitors Drives Cytotoxicity in Both Cancer Cells and Healthy Bone Marrow , 2018, Molecular Cancer Research.

[3]  A. Ashworth,et al.  Synthetic lethal therapies for cancer: what’s next after PARP inhibitors? , 2018, Nature Reviews Clinical Oncology.

[4]  A. Ashworth,et al.  Coupling bimolecular PARylation biosensors with genetic screens to identify PARylation targets , 2018, Nature Communications.

[5]  W. Symmans,et al.  Addition of the PARP inhibitor veliparib plus carboplatin or carboplatin alone to standard neoadjuvant chemotherapy in triple-negative breast cancer (BrighTNess): a randomised, phase 3 trial. , 2018, The Lancet. Oncology.

[6]  B. E. Black,et al.  NAD+ analog reveals PARP-1 substrate-blocking mechanism and allosteric communication from catalytic center to DNA-binding domains , 2018, Nature Communications.

[7]  Simon C Watkins,et al.  PARP1 changes from three-dimensional DNA damage searching to one-dimensional diffusion after auto-PARylation or in the presence of APE1 , 2017, Nucleic acids research.

[8]  A. Ashworth,et al.  Genome-wide and high-density CRISPR-Cas9 screens identify point mutations in PARP1 causing PARP inhibitor resistance , 2017, bioRxiv.

[9]  A. Heijink,et al.  Progression through mitosis promotes PARP inhibitor-induced cytotoxicity in homologous recombination-deficient cancer cells , 2017, Nature Communications.

[10]  A. Ashworth,et al.  Modeling Therapy Resistance in BRCA1/2-Mutant Cancers , 2017, Molecular Cancer Therapeutics.

[11]  Alan Ashworth,et al.  PARP inhibitors: Synthetic lethality in the clinic , 2017, Science.

[12]  Ziying Liu,et al.  PARPs and ADP-ribosylation: recent advances linking molecular functions to biological outcomes , 2017, Genes & development.

[13]  Y. Pommier,et al.  Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action , 2016, Science Translational Medicine.

[14]  Yonghao Yu,et al.  Chemical genetic discovery of PARP targets reveals a role for PARP-1 in transcription elongation , 2016, Science.

[15]  J. Pascal,et al.  Structural Basis of Detection and Signaling of DNA Single-Strand Breaks by Human PARP-1 , 2015, Molecular cell.

[16]  Jamin D Steffen,et al.  PARP-1 Activation Requires Local Unfolding of an Autoinhibitory Domain. , 2015, Molecular cell.

[17]  T. Ellenberger,et al.  The rise and fall of poly(ADP-ribose): An enzymatic perspective. , 2015, DNA Repair.

[18]  Yan Shi,et al.  Mechanistic Dissection of PARP1 Trapping and the Impact on In Vivo Tolerability and Efficacy of PARP Inhibitors , 2015, Molecular Cancer Research.

[19]  B. Wang,et al.  Trapping Poly(ADP-Ribose) Polymerase , 2015, The Journal of Pharmacology and Experimental Therapeutics.

[20]  E. Pai,et al.  Insights into the binding of PARP inhibitors to the catalytic domain of human tankyrase-2 , 2014, Acta crystallographica. Section D, Biological crystallography.

[21]  G. Sica,et al.  Poly (ADP) ribose polymerase enzyme inhibitor, veliparib, potentiates chemotherapy and radiation in vitro and in vivo in small cell lung cancer , 2014, Cancer medicine.

[22]  J. Pascal,et al.  PARP-2 and PARP-3 are selectively activated by 5′ phosphorylated DNA breaks through an allosteric regulatory mechanism shared with PARP-1 , 2014, Nucleic acids research.

[23]  James H. Doroshow,et al.  Rationale for Poly(ADP-ribose) Polymerase (PARP) Inhibitors in Combination Therapy with Camptothecins or Temozolomide Based on PARP Trapping versus Catalytic Inhibition , 2014, The Journal of Pharmacology and Experimental Therapeutics.

[24]  Y. Pommier,et al.  Stereospecific PARP Trapping by BMN 673 and Comparison with Olaparib and Rucaparib , 2013, Molecular Cancer Therapeutics.

[25]  A. Ashworth,et al.  BMN 673, a Novel and Highly Potent PARP1/2 Inhibitor for the Treatment of Human Cancers with DNA Repair Deficiency , 2013, Clinical Cancer Research.

[26]  J. Pascal,et al.  PARP-1 mechanism for coupling DNA damage detection to poly(ADP-ribose) synthesis. , 2013, Current opinion in structural biology.

[27]  Y. Pommier,et al.  Trapping of PARP1 and PARP2 by Clinical PARP Inhibitors. , 2012, Cancer research.

[28]  J. Pascal,et al.  Structural Basis for DNA Damage–Dependent Poly(ADP-ribosyl)ation by Human PARP-1 , 2012, Science.

[29]  R. Martínez-Zamudio,et al.  Histone ADP-Ribosylation Facilitates Gene Transcription by Directly Remodeling Nucleosomes , 2012, Molecular and Cellular Biology.

[30]  R. Hromas,et al.  Synthetic lethality: exploiting the addiction of cancer to DNA repair. , 2011, Blood.

[31]  A. Ashworth,et al.  Synthetic lethality of PARP inhibition in cancers lacking BRCA1 and BRCA2 mutations , 2011, Cell cycle.

[32]  N. Pannu,et al.  REFMAC5 for the refinement of macromolecular crystal structures , 2011, Acta crystallographica. Section D, Biological crystallography.

[33]  Randy J. Read,et al.  Overview of the CCP4 suite and current developments , 2011, Acta crystallographica. Section D, Biological crystallography.

[34]  J. Pascal,et al.  Crystal Structures of Poly(ADP-ribose) Polymerase-1 (PARP-1) Zinc Fingers Bound to DNA , 2011, The Journal of Biological Chemistry.

[35]  H. Kotani,et al.  Synthetic lethal interactions for the development of cancer therapeutics: biological and methodological advancements , 2010, Human Genetics.

[36]  J. Pascal,et al.  The Zn3 Domain of Human Poly(ADP-ribose) Polymerase-1 (PARP-1) Functions in Both DNA-dependent Poly(ADP-ribose) Synthesis Activity and Chromatin Compaction* , 2010, The Journal of Biological Chemistry.

[37]  S. Kaufmann,et al.  PARP inhibition: PARP1 and beyond , 2010, Nature Reviews Cancer.

[38]  P. Emsley,et al.  Features and development of Coot , 2010, Acta crystallographica. Section D, Biological crystallography.

[39]  Randy J. Read,et al.  Acta Crystallographica Section D Biological , 2003 .

[40]  Eric F. Johnson,et al.  Discovery of the Poly(ADP-ribose) polymerase (PARP) inhibitor 2-[(R)-2-methylpyrrolidin-2-yl]-1H-benzimidazole-4-carboxamide (ABT-888) for the treatment of cancer. , 2009, Journal of medicinal chemistry.

[41]  A. Ashworth,et al.  A high-throughput RNA interference screen for DNA repair determinants of PARP inhibitor sensitivity. , 2008, DNA repair.

[42]  Randy J. Read,et al.  Phaser crystallographic software , 2007, Journal of applied crystallography.

[43]  P. Katsamba,et al.  Analyzing a kinetic titration series using affinity biosensors. , 2006, Analytical biochemistry.

[44]  H. Leonhardt,et al.  Recruitment of DNA methyltransferase I to DNA repair sites. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[45]  Csaba Szabó,et al.  Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors , 2005, Nature Reviews Drug Discovery.

[46]  M. Y. Kim,et al.  NAD+-Dependent Modulation of Chromatin Structure and Transcription by Nucleosome Binding Properties of PARP-1 , 2004, Cell.

[47]  Csaba Szabó,et al.  The discovery and synthesis of novel adenosine substituted 2,3-dihydro-1H-isoindol-1-ones: potent inhibitors of poly(ADP-ribose) polymerase-1 (PARP-1). , 2004, Bioorganic & medicinal chemistry letters.

[48]  P. Chambon,et al.  Functional interaction between PARP‐1 and PARP‐2 in chromosome stability and embryonic development in mouse , 2003, The EMBO journal.

[49]  K. Schlemmer,et al.  Substituted Uracil Derivatives as Potent Inhibitors of Poly(ADP‐ribose)polymerase‐1 (PARP‐1) , 2003 .

[50]  A. Spradling,et al.  Chromatin Loosening by Poly(ADP)-Ribose Polymerase (PARP) at Drosophila Puff Loci , 2003, Science.

[51]  G. Poirier,et al.  Poly(ADP-ribosyl)ation reactions in the regulation of nuclear functions. , 1999, The Biochemical journal.

[52]  D. Myszka,et al.  Improving biosensor analysis , 1999, Journal of molecular recognition : JMR.

[53]  Masahiko S. Satoh,et al.  Role of poly(ADP-ribose) formation in DNA repair , 1992, Nature.

[54]  S. Kaufmann,et al.  PARP inhibition: PARP1 and beyond , 2010, Nature Reviews Cancer.