Acessibilidade / Reportar erro

Molecular mechanisms of cell death by parthanatos: More questions than answers

Abstract

Regulated cell death by a non-apoptotic pathway known as parthanatos is increasingly recognised as a central player in pathological processes, including ischaemic tissue damage and neurodegenerative diseases. Parthanatos is activated under conditions that induce high levels of DNA damage, leading to hyperactivation of the DNA damage sensor PARP1. While this strict dependence on PARP1 activation is a defining feature of parthanatos that distinguishes it from other forms of cell death, the molecular events downstream of PARP1 activation remain poorly understood. In this mini-review, we highlight a number of important questions that remain to be answered about this enigmatic form of cell death.

Keywords:
Cell death; parthanatos; PARP1; PARG; AIF

Introduction

ADP-ribosylation (ADPr) is a covalent modification of biological macromolecules catalysed by members of the ADP-ribosyltransferase family, which transfer ADP-ribose moieties from NAD+ (nicotinamide adenine dinucleotide) to target proteins or nucleic acids (Hoch and Polo 2019Hoch NC and Polo LM (2019) ADP-ribosylation: From molecular mechanisms to human disease. Genet Mol Biol 43:e20190075.; Luscher et al., 2021Luscher B, Ahel I, Altmeyer M, Ashworth A, Bai P, Chang P, Cohen M, Corda D, Dantzer F, Daugherty MD et al. (2021) ADP-ribosyltransferases, an update on function and nomenclature. FEBS J 289:7399-7410.; Suskiewicz et al., 2023Suskiewicz MJ, Prokhorova E, Rack JGM and Ahel I (2023) ADP-ribosylation from molecular mechanisms to therapeutic implications. Cell 186:4475-4495.). ADP-ribosyltransferases can be subdivided based on the nature of the ADPr modification they catalyse, which can be in the form of single ADP-ribose units, termed mono(ADP-ribose) (MAR) or as long and sometimes branched chains of poly(ADP-ribose) (PAR). The main human PAR-catalysing enzyme is poly(ADP-ribose) polymerase 1 (PARP1), which is a highly abundant nuclear protein that consists of three DNA-binding zinc finger domains (ZnF1, ZnF2, and ZnF3), a central BRCA1 C-terminal (BRCT) domain, a DNA-binding WGR (tryptophan, glycine, arginine) domain and a bipartite C-terminal catalytic domain composed of an auto-inhibitory helical subdomain (HD) and an ADP-ribosyl transferase (ART) subdomain. PARP1 plays central roles in the cellular response to DNA damage, due to the high affinity and specificity of its DNA-binding domains for DNA strand breaks, which lead to rapid and robust activation of PARP1 catalytic activity in response to a variety of DNA lesions (Pandey and Black 2021Pandey N and Black BE (2021) Rapid detection and signaling of DNA damage by PARP-1. Trends Biochem Sci 46:744-757.; Pascal 2023Pascal JM (2023) PARP-nucleic acid interactions: Allosteric signaling, PARP inhibitor types, DNA bridges, and viral RNA surveillance. Curr Opin Struct Biol 81:102643.). Once activated, PARP1 modifies itself and a growing list of chromatin-associated proteins, including core histones, promoting the recruitment of PAR-binding DNA repair proteins to the lesion site and accelerating DNA repair (Ray Chaudhuri and Nussenzweig 2017Ray Chaudhuri A and Nussenzweig A (2017) The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol 18:610-621.; Hendriks et al., 2021Hendriks IA, Buch-Larsen SC, Prokhorova E, Elsborg JD, Rebak AKLFS, Zhu K, Ahel D, Lukas C, Ahel I and Nielsen ML (2021) The regulatory landscape of the human HPF1- and ARH3-dependent ADP-ribosylome. Nat Commun 12:5893.; Caldecott, 2022Caldecott KW (2022) DNA single-strand break repair and human genetic disease. Trends Cell Biol 32:733-745.). Interestingly, there is extensive literature on roles of PARP1 in many other cellular processes, such as chromatin remodelling, gene regulation and inflammation (Hottiger, 2015Hottiger MO (2015) Nuclear ADP-Ribosylation and its role in chromatin plasticity, cell differentiation, and epigenetics. Annu Rev Biochem 84:227-263.; Fehr et al., 2020Fehr AR, Singh SA, Kerr CM, Mukai S, Higashi H and Aikawa M (2020) The impact of PARPs and ADP-ribosylation on inflammation and host-pathogen interactions. Genes Dev 34:341-359.; Kim et al., 2020Kim DS, Challa S, Jones A and Kraus WL (2020) PARPs and ADP-ribosylation in RNA biology: From RNA expression and processing to protein translation and proteostasis. Genes Dev 34:302-320.), but whether PARP1 is also responding to some form of DNA damage under these conditions or if PARP1 can be catalytically activated in the absence of a DNA strand break is currently unclear.

In addition to its canonical role in accelerating DNA repair and, therefore, promoting cell survival in response to DNA lesions, PARP1 can become hyperactivated in response to high levels of acute DNA damage, triggering a regulated form of cell death termed parthanatos (Yu et al., 2002Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ, Poirier GG, Dawson TM and Dawson VL (2002) Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 297:259-263.; Fatokun et al., 2014Fatokun AA, Dawson VL and Dawson TM (2014) Parthanatos: Mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 171:2000-2016.). In this setting, genetic PARP1 deletion or pharmacological PARP1 inhibition are strongly cytoprotective, and this strict PARP1 dependency is the defining feature of parthanatos that distinguishes it from other forms of cell death, such as apoptosis or necrosis (Fatokun et al., 2014Fatokun AA, Dawson VL and Dawson TM (2014) Parthanatos: Mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 171:2000-2016.).

Parthanatos can be triggered by several exogenous or endogenous sources that generate a high load of PARP1-activating DNA breaks, such as the alkylating agents MNNG (N-methyl-N’-nitro-N-nitrosoguanidine) or MMS (methyl methanesulfonate), or a variety of treatments that induce high bursts of reactive oxygen or nitrogen species, such as hydrogen peroxide (H2O2) and other oxidants. In neuronal cells, this can be achieved by overstimulation of glutamate receptors via NMDA (N-methyl-D-aspartate) or glutamate, in a process also known as glutamate excitotoxicity (Mandir et al., 2000Mandir AS, Poitras MF, Berliner AR, Herring WJ, Guastella DB, Feldman A, Poirier GG, Wang ZQ, Dawson TM and Dawson VL (2000) NMDA but not non-NMDA excitotoxicity is mediated by Poly(ADP-ribose) polymerase. J Neurosci 20:8005-8011.). Several disease models that rely on DNA damage-induced tissue dysfunction, such as steptozotocin-induced diabetes and MPTP-induced Parkinsonism also rely on parthanatos for tissue demise (Yamamoto et al., 1981Yamamoto H, Uchigata Y and Okamoto H (1981) Streptozotocin and alloxan induce DNA strand breaks and poly(ADP-ribose) synthetase in pancreatic islets. Nature 294:284-286.; Wang et al., 2003Wang H, Shimoji M, Yu S-W, Dawson TM and Dawson VL (2003) Apoptosis inducing factor and PARP-Mediated injury in the MPTP mouse model of Parkinson’s disease. Ann N Y Acad Sci 991:132-139.). Ischemia-reperfusion is another well-documented process that induces a burst of oxidative DNA damage, leading to PARP1-mediated cell death (Eliasson et al., 1997Eliasson MJL, Sampei K, Mandir AS, Hurn PD, Traystman RJ, Bao J, Pieper A, Wang Z-Q, Dawson TM, Snyder SH et al. (1997) Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med 3:1089-1095.; Dawson and Dawson, 2018Dawson TM and Dawson VL (2018) Nitric Oxide signaling in neurodegeneration and cell death. Adv Pharmacol 82:57-83.) and more recently, it has become evident that PARP1 hyperactivation and parthanatos play a pathological role in neurodegenerative disorders as well, including Parkinson’s disease and Alzheimer’s disease (Hoch et al., 2017Hoch NC, Hanzlikova H, Rulten SL, Tetreault M, Komulainen E, Ju L, Hornyak P, Zeng Z, Gittens W, Rey SA et al. (2017) XRCC1 mutation is associated with PARP1 hyperactivation and cerebellar ataxia. Nature 541:87-91.; Kam et al., 2018Kam TI, Mao X, Park H, Chou SC, Karuppagounder SS, Umanah GE, Yun SP, Brahmachari S, Panicker N, Chen R et al. (2018) Poly(ADP-ribose) drives pathologic alpha-synuclein neurodegeneration in Parkinson’s disease. Science 362:eaat8407.; Park et al., 2020Park H, Kam TI, Dawson TM and Dawson VL (2020) Poly (ADP-ribose) (PAR)-dependent cell death in neurodegenerative diseases. Int Rev Cell Mol Biol 353:1-29., 2022Park H, Kam TI, Peng H, Chou SC, Mehrabani-Tabari AA, Song JJ, Yin X, Karuppagounder SS, Umanah GK, Rao AVS et al. (2022) PAAN/MIF nuclease inhibition prevents neurodegeneration in Parkinson’s disease. Cell 185:1943-1959e1921.).

The variety of pathophysiological situations that lead to PARP1 hyperactivation and the likely clinical benefit of PARP inhibitors to manage these disorders have been extensively reviewed (Fatokun et al., 2014Fatokun AA, Dawson VL and Dawson TM (2014) Parthanatos: Mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 171:2000-2016.; Berger et al., 2018Berger NA, Besson VC, Boulares AH, Burkle A, Chiarugi A, Clark RS, Curtin NJ, Cuzzocrea S, Dawson TM, Dawson VL et al. (2018) Opportunities for the repurposing of PARP inhibitors for the therapy of non-oncological diseases. Br J Pharmacol 175:192-222.; Liu et al., 2022 a Liu L, Li J, Ke Y, Zeng X, Gao J, Ba X and Wang R (2022a) The key players of parthanatos: Opportunities for targeting multiple levels in the therapy of parthanatos-based pathogenesis. Cell Mol Life Sci 79:60., bLiu S, Luo W and Wang Y (2022b) Emerging role of PARP-1 and PARthanatos in ischemic stroke. J Neurochem 160:74-87.), and will only be briefly mentioned here. Likewise, other genetic or pharmacological interventions that affect the magnitude of spontaneous or induced PARP1 hyperactivation will not be discussed (Andrabi et al., 2011Andrabi SA, Kang HC, Haince JF, Lee YI, Zhang J, Chi Z, West AB, Koehler RC, Poirier GG, Dawson TM et al. (2011) Iduna protects the brain from glutamate excitotoxicity and stroke by interfering with poly(ADP-ribose) polymer-induced cell death. Nat Med 17:692-699.; Kang et al., 2011Kang HC, Lee Y-I, Shin J-H, Andrabi SA, Chi Z, Gagné J-P, Lee Y, Ko HS, Lee BD, Poirier GG et al. (2011) Iduna is a poly(ADP-ribose) (PAR)-dependent E3 ubiquitin ligase that regulates DNA damage. Proc Natl Acad Sci U S A 108:14103-14108.; Yang et al., 2022Yang M, Wang C, Zhou M, Bao L, Wang Y, Kumar A, Xing C, Luo W and Wang Y (2022) KDM6B promotes PARthanatos via suppression of O6-methylguanine DNA methyltransferase repair and sustained checkpoint response. Nucleic Acids Res 50:6313-6331.). Instead, this short review will focus on mechanistic considerations of the downstream events that follow excessive PARP1 catalytic activity and how they contribute to cellular demise (Figure 1). At the end of each section, we will provide a list of open questions that have not been addressed so far or that are currently unclear from the literature.

Figure 1 -
Potential pathways of cell death mediated by PARP1 hyperactivation.

NAD+ depletion and inhibition of glycolysis

Since NAD+ is consumed in the process of ADP-ribosylation, donating the ADP-ribose moiety for target modification, and because PARP1 is a highly abundant and processive enzyme, PARP1 hyperactivation results in a rapid and profound depletion of cellular NAD+ pools (Berger 1985Berger NA (1985) Poly(ADP-Ribose) in the cellular response to DNA damage. Radiat Res 101:4-15.). Interestingly, this is accompanied by a depletion of cellular ATP, indicating that parthanatos could be a result of cellular energetic collapse (Berger 1985Berger NA (1985) Poly(ADP-Ribose) in the cellular response to DNA damage. Radiat Res 101:4-15.; Ha and Snyder 1999Ha HC and Snyder SH (1999) Poly(ADP-ribose) polymerase is a mediator of necrotic cell death by ATP depletion. Proc Natl Acad Sci U S A 96:13978-13982.).

One possible explanation for PARP1-dependent ATP depletion is the unavailability of NAD+ to act as an electron acceptor for core metabolic pathways such as glycolysis and the tricarboxylic acid (TCA) cycle, with the accompanying reduction of the available NADH for oxidative phosphorylation. Early studies using astrocyte cultures indicated NAD+ depletion as the primary mediator of parthanatos, as parthanatos-associated events could be induced by other NAD+-depleting treatments and prevented by supplementation with exogenous NAD+ (Alano et al., 2004Alano CC, Ying W and Swanson RA (2004) Poly(ADP-ribose) polymerase-1-mediated cell death in astrocytes requires NAD+ depletion and mitochondrial permeability transition. J Biol Chem 279:18895-18902., 2010Alano CC, Garnier P, Ying W, Higashi Y, Kauppinen TM and Swanson RA (2010) NAD+ depletion is necessary and sufficient for poly(ADP-ribose) polymerase-1-mediated neuronal death. J Neurosci 30:2967-2978.; Ying et al., 2005Ying W, Alano CC, Garnier P and Swanson RA (2005) NAD+ as a metabolic link between DNA damage and cell death. J Neurosci Res 79:216-223.). Treatment with pyruvate or α-ketoglutarate, which can support TCA cycle activity but bypass glycolysis, was also sufficient to prevent cell death, indicating a glycolytic defect. As the authors pointed out, however, this scenario could be limited to situations in which glucose is the only substrate for energy metabolism in the culture medium used (artificial cerebrospinal fluid in this case), while conventional media often contain pyruvate and other carbon sources. Similarly, Zong et al. (2004Zong W-X, Ditsworth D, Bauer DE, Wang Z-Q and Thompson CB (2004) Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev 18:1272-1282.) observed that cells that rely more heavily on glycolysis are more susceptible to cell death by parthanatos, which can be reversed by supplementation with pyruvate. Further evidence indicating a central role for NAD+ levels in parthanatos, is the observation that treatment with the NAD+ precursors nicotinamide riboside (NR) or nicotinamide mononucleotide (NMN) can prevent PARP1-dependent cell death in some settings (Nishida et al., 2022Nishida T, Naguro I and Ichijo H (2022) NAMPT-dependent NAD(+) salvage is crucial for the decision between apoptotic and necrotic cell death under oxidative stress. Cell Death Discov 8:195.; Santofimia-Castaño et al., 2022Santofimia-Castaño P, Huang C, Liu X, Xia Y, Audebert S, Camoin L, Peng L, Lomberk G, Urrutia R, Soubeyran P et al. (2022) NUPR1 protects against hyperPARylation-dependent cell death. Commun Biol 5:732.). In this context, it is worth mentioning that cellular NAD+ pools are compartmentalized, and that nuclear and cytoplasmic NAD+ are in rapid equilibrium, whereas mitochondrial NAD+ pools are maintained separately (Cambronne et al., 2016Cambronne XA, Stewart ML, Kim D, Jones-Brunette AM, Morgan RK, Farrens DL, Cohen MS and Goodman RH (2016) Biosensor reveals multiple sources for mitochondrial NAD(+). Science 352:1474-1477.; Covarrubias et al., 2021Covarrubias AJ, Perrone R, Grozio A and Verdin E (2021) NAD+ metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol 22:119-141.). This would indicate that nuclear PARP1 hyperactivation should impact the nuclear/cytoplasmic NAD+ pool more rapidly, which would be consistent with a larger impact of parthanatos-induced NAD+ depletion on cytoplasmic glycolysis than on mitochondrial TCA cycle. However, NAD+ can be transported across the mitochondrial membrane via the recently identified SLC25A51 transporter (Girardi et al., 2020Girardi E, Agrimi G, Goldmann U, Fiume G, Lindinger S, Sedlyarov V, Srndic I, Gürtl B, Agerer B, Kartnig F et al. (2020) Epistasis-driven identification of SLC25A51 as a regulator of human mitochondrial NAD import. Nat Commun 11:6145.; Kory et al., 2020Kory N, Uit de Bos J, van der Rijt S, Jankovic N, Güra M, Arp N, Pena IA, Prakash G, Chan SH, Kunchok T et al. (2020) MCART1/SLC25A51 is required for mitochondrial NAD transport. Sci Adv 6:eabe5310.; Luongo et al., 2020Luongo TS, Eller JM, Lu MJ, Niere M, Raith F, Perry C, Bornstein MR, Oliphint P, Wang L, McReynolds MR et al. (2020) SLC25A51 is a mammalian mitochondrial NAD(+) transporter. Nature 588:174-179.), and there is evidence for a mitochondrial pool of PARP1 (Szczesny et al., 2014Szczesny B, Brunyanszki A, Olah G, Mitra S and Szabo C (2014) Opposing roles of mitochondrial and nuclear PARP1 in the regulation of mitochondrial and nuclear DNA integrity: Implications for the regulation of mitochondrial function. Nucleic Acids Res 42:13161-13173.; Herrmann et al., 2021Herrmann GK, Russell WK, Garg NJ and Yin YW (2021) Poly(ADP-ribose) polymerase 1 regulates mitochondrial DNA repair in an NAD-dependent manner. J Biol Chem 296:100309.; Lee et al., 2022Lee JH, Hussain M, Kim EW, Cheng SJ, Leung AKL, Fakouri NB, Croteau DL and Bohr VA (2022) Mitochondrial PARP1 regulates NAD(+)-dependent poly ADP-ribosylation of mitochondrial nucleoids. Exp Mol Med 54:2135-2147.), suggesting that PARP1-dependent depletion of mitochondrial NAD+ may also play a role in parthanatos execution.

Other studies in cortical neurons and glioblastoma-derived cell lines have suggested that NAD+ depletion itself is not sufficient to cause ATP depletion, glycolytic defects or cell death, only being responsible for defects in mitochondrial respiration (Andrabi et al., 2014Andrabi SA, Umanah GK, Chang C, Stevens DA, Karuppagounder SS, Gagné JP, Poirier GG, Dawson VL and Dawson TM (2014 ) Poly(ADP-ribose) polymerase-dependent energy depletion occurs through inhibition of glycolysis. Proc Natl Acad Sci U S A 111:10209-10214.; Fouquerel et al., 2014Fouquerel E, Goellner Eva M, Yu Z, Gagné J-P, Barbi de Moura M, Feinstein T, Wheeler D, Redpath P, Li J, Romero G et al. (2014) ARTD1/PARP1 negatively regulates glycolysis by inhibiting hexokinase 1 independent of NAD+ Depletion. Cell Rep 8:1819-1831.). In these studies, profound NAD+ depletion in the absence of PARP1 hyperactivation was insufficient to induce parthanatos and supplementation with nicotinamide riboside (NR), an NAD+ precursor, did not prevent glycolytic dysfunction (Andrabi et al., 2014Andrabi SA, Umanah GK, Chang C, Stevens DA, Karuppagounder SS, Gagné JP, Poirier GG, Dawson VL and Dawson TM (2014 ) Poly(ADP-ribose) polymerase-dependent energy depletion occurs through inhibition of glycolysis. Proc Natl Acad Sci U S A 111:10209-10214.; Fouquerel et al., 2014Fouquerel E, Goellner Eva M, Yu Z, Gagné J-P, Barbi de Moura M, Feinstein T, Wheeler D, Redpath P, Li J, Romero G et al. (2014) ARTD1/PARP1 negatively regulates glycolysis by inhibiting hexokinase 1 independent of NAD+ Depletion. Cell Rep 8:1819-1831.). In this scenario, PARP1 activity is thought to directly inhibit the first step of glycolysis, via the release of PAR polymers from target proteins by PAR-degrading enzymes (see below), which then bind to hexokinase-1 and inhibit its catalytic activity (Andrabi et al., 2014Andrabi SA, Umanah GK, Chang C, Stevens DA, Karuppagounder SS, Gagné JP, Poirier GG, Dawson VL and Dawson TM (2014 ) Poly(ADP-ribose) polymerase-dependent energy depletion occurs through inhibition of glycolysis. Proc Natl Acad Sci U S A 111:10209-10214.; Fouquerel et al., 2014Fouquerel E, Goellner Eva M, Yu Z, Gagné J-P, Barbi de Moura M, Feinstein T, Wheeler D, Redpath P, Li J, Romero G et al. (2014) ARTD1/PARP1 negatively regulates glycolysis by inhibiting hexokinase 1 independent of NAD+ Depletion. Cell Rep 8:1819-1831.). Although at odds with the NAD+-centric model that emerged from the above studies, supplying cells with pyruvate was again sufficient to overcome PARP1-mediated metabolic dysfunction, consistent with glycolysis being a core target of parthanatos (Andrabi et al., 2014Andrabi SA, Umanah GK, Chang C, Stevens DA, Karuppagounder SS, Gagné JP, Poirier GG, Dawson VL and Dawson TM (2014 ) Poly(ADP-ribose) polymerase-dependent energy depletion occurs through inhibition of glycolysis. Proc Natl Acad Sci U S A 111:10209-10214.) (Figure 1). As hexokinase also generates substrates for the pentose phosphate pathway (PPP), this mechanism is also consistent with the recently described depletion of reduced glutathione (GSH) and NADPH during parthanatos, which are both products of the PPP (Hossain et al., 2024Hossain MI, Lee JH, Gagné JP, Khan J, Poirier GG, King PH, Dawson VL, Dawson TM and Andrabi SA (2024) Poly(ADP-ribose) mediates bioenergetic defects and redox imbalance in neurons following oxygen and glucose deprivation. FASEB J 38:e23556.).

Another model to explain the proposed uncoupling between ATP and NAD+ depletion derives from the observation that AMP, which may be generated at high levels during PAR chain degradation (see below), can inhibit the mitochondrial adenine nucleotide translocator (ANT) (Formentini et al., 2009Formentini L, Macchiarulo A, Cipriani G, Camaioni E, Rapizzi E, Pellicciari R, Moroni F and Chiarugi A (2009) Poly(ADP-ribose) catabolism triggers AMP-dependent mitochondrial energy failure. J Biol Chem 284:17668-17676.; Buonvicino et al., 2013Buonvicino D, Formentini L, Cipriani G and Chiarugi A (2013) Glucose deprivation converts poly(ADP-ribose) polymerase-1 hyperactivation into a transient energy-producing process. J Biol Chem 288:36530-36537.). This would lead to an impaired translocation of ADP into the mitochondria, inhibiting mitochondrial ATP synthesis due to the low availability of ADP for oxidative phosphorylation.

Interestingly, it has been suggested that ATP depletion is responsible for diverting cells from apoptosis to parthanatos, with PARP1 hyperactivation thus acting as a “switch” between these forms of cell death (Ha and Snyder 1999Ha HC and Snyder SH (1999) Poly(ADP-ribose) polymerase is a mediator of necrotic cell death by ATP depletion. Proc Natl Acad Sci U S A 96:13978-13982.). In agreement with this, recent findings indicate that lower (but still cytotoxic) levels of DNA damage induce an intermediate level of NAD+ consumption by PARP1 that can be matched by the NAD+ salvage pathway, leading to a transient NAD+ and ATP depletion that allows cell death to proceed by apoptosis, whereas higher DNA damage loads cause a more prolonged NAD+ and ATP depletion that precludes apoptosis (Nishida et al., 2022Nishida T, Naguro I and Ichijo H (2022) NAMPT-dependent NAD(+) salvage is crucial for the decision between apoptotic and necrotic cell death under oxidative stress. Cell Death Discov 8:195.). Conversely, PARP1 hyperactivation is also actively prevented during the apoptotic cascade via caspase-dependent cleavage of PARP1 between the DNA binding domains and the catalytic domain, which is thought to ensure that the cell can meet the energy requirements of apoptosis (D’Amours et al., 2001D’Amours D, Sallmann FR, Dixit VM and Poirier GG (2001) Gain-of-function of poly(ADP-ribose) polymerase-1 upon cleavage by apoptotic proteases: Implications for apoptosis. J Cell Sci 114:3771-3778.).

Open questions:

  • What factor(s) determine(s) whether NAD supplementation does or does not prevent parthanatos induction?

  • Is the inhibition of glycolysis necessary and/or sufficient for cell death by parthanatos?

  • How do free PAR chains inhibit hexokinase activity?

  • Are NAD+ and ATP depletion mechanistically connected?

  • Is there more extensive crosstalk between apoptosis and parthanatos, or is this limited to parthanatic NAD+/ATP depletion preventing apoptosis and apoptotic PARP1 cleavage preventing parthanatos?

PAR hydrolysis

The human genome encodes several hydrolases responsible for the reversal of ADP-ribosylation: those belonging to the macrodomain family - PARG, TARG1, MacroD1 and MacroD2; and those in the ADP-ribose-acceptor hydrolase family - ARH1 and ARH3 (O’Sullivan et al., 2019O’Sullivan J, Tedim Ferreira M, Gagné JP, Sharma AK, Hendzel MJ, Masson JY and Poirier GG (2019) Emerging roles of eraser enzymes in the dynamic control of protein ADP-ribosylation. Nat Commun 10:1182.; Rack et al., 2020Rack JGM, Palazzo L and Ahel I (2020) (ADP-ribosyl)hydrolases: Structure, function, and biology. Genes Dev 34:263-284.). Among these, PARG and ARH3 are crucial for the hydrolysis of PARP1-generated PAR chains, with PARG contributing the bulk of PAR hydrolysis activity, cleaving the O-glycosidic bond between ADP-ribose units, both in linear chains as well as at branching points (Rack et al., 2021Rack JGM, Liu Q, Zorzini V, Voorneveld J, Ariza A, Honarmand Ebrahimi K, Reber JM, Krassnig SC, Ahel D, van der Marel GA et al. (2021) Mechanistic insights into the three steps of poly(ADP-ribosylation) reversal. Nat Commun 12:4581.). While ARH3 can also contribute to PAR hydrolysis, its main role is the release of the final serine-bound mono-ADP-ribose (Abplanalp et al., 2017Abplanalp J, Leutert M, Frugier E, Nowak K, Feurer R, Kato J, Kistemaker HVA, Filippov DV, Moss J, Caflisch A et al. (2017) Proteomic analyses identify ARH3 as a serine mono-ADP-ribosylhydrolase. Nat Commun 8:2055.; Fontana et al., 2017Fontana P, Bonfiglio JJ, Palazzo L, Bartlett E, Matic I and Ahel I (2017) Serine ADP-ribosylation reversal by the hydrolase ARH3. Elife 6:e28533.), serine being the main target residue for PARP1 in response to DNA damage (Palazzo et al., 2018Palazzo L, Leidecker O, Prokhorova E, Dauben H, Matic I and Ahel I (2018) Serine is the major residue for ADP-ribosylation upon DNA damage. Elife 7:e34334.). Similar to PARP1, PARG is a highly active enzyme, making poly-ADP-ribosylation a very transient modification that is produced and degraded within minutes of an insult (Hanzlikova et al., 2018Hanzlikova H, Kalasova I, Demin AA, Pennicott LE, Cihlarova Z and Caldecott KW (2018) The Importance of Poly(ADP-Ribose) polymerase as a sensor of unligated okazaki fragments during DNA Replication. Mol Cell 71:319-331e313.). Interestingly, the reversal of DNA damage-induced mono-ADP-ribosylation, which can be generated either as a remnant of PARG activity or directly by PARP1, seems to be much slower, indicating a longer-lasting, and therefore different, cellular response (Longarini et al., 2023Longarini EJ, Dauben H, Locatelli C, Wondisford AR, Smith R, Muench C, Kolvenbach A, Lynskey ML, Pope A, Bonfiglio JJ et al. (2023) Modular antibodies reveal DNA damage-induced mono-ADP-ribosylation as a second wave of PARP1 signaling. Mol Cell 83:1743-1760.e1711.).

There is extensive but conflicting evidence as to the role of PARG in parthanatos, with several studies suggesting that PARG can either prevent or promote PARP1-dependent cell death. In favour of an inhibitory role, PARG overexpression reduced MNNG-induced cell death in mouse neuronal cultures (Andrabi et al., 2006Andrabi SA, Kim NS, Yu SW, Wang H, Koh DW, Sasaki M, Klaus JA, Otsuka T, Zhang Z, Koehler RC et al. (2006) Poly(ADP-ribose) (PAR) polymer is a death signal. Proc Natl Acad Sci U S A 103:18308-18313.) and reduced NMDA-induced AIF release from mitochondria (Yu et al., 2006Yu S-W, Andrabi SA, Wang H, Kim NS, Poirier GG, Dawson TM and Dawson VL (2006) Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci U S A 103:18314-18319.), an important step in most parthanatos models, which is covered in more detail below. Similarly, knockdown of PARG increased cell death in mouse neurons (Andrabi et al., 2006Andrabi SA, Kim NS, Yu SW, Wang H, Koh DW, Sasaki M, Klaus JA, Otsuka T, Zhang Z, Koehler RC et al. (2006) Poly(ADP-ribose) (PAR) polymer is a death signal. Proc Natl Acad Sci U S A 103:18308-18313.) and PARG deletion in trophoblast stem cells increased AIF release after UV irradiation (Zhou et al., 2011Zhou Y, Feng X and Koh DW (2011) Activation of cell death mediated by apoptosis-inducing factor due to the absence of poly(ADP-ribose) glycohydrolase. Biochemistry 50:2850-2859.). PARG +/- mice had larger infarct volumes after brain ischaemia-reperfusion injury, while mice overexpressing PARG had smaller infarct volumes (Andrabi et al., 2006Andrabi SA, Kim NS, Yu SW, Wang H, Koh DW, Sasaki M, Klaus JA, Otsuka T, Zhang Z, Koehler RC et al. (2006) Poly(ADP-ribose) (PAR) polymer is a death signal. Proc Natl Acad Sci U S A 103:18308-18313.). In contrast, other studies suggest that PARG is necessary for, or at least contributes to, the process of parthanatos. PARG inhibition protected mice against brain ischaemia (Lu et al., 2003Lu XC, Massuda E, Lin Q, Li W, Li JH and Zhang J (2003) Post-treatment with a novel PARG inhibitor reduces infarct in cerebral ischemia in the rat. Brain Res 978:99-103.), and PARG silencing rendered cells more resistant to treatment with H2O2 but not MNNG (Blenn et al., 2006Blenn C, Althaus FR and Malanga M (2006) Poly(ADP-ribose) glycohydrolase silencing protects against H2O2-induced cell death. Biochem J 396:419-429.). In the aforementioned studies proposing the hexokinase inhibition model, there is also conflicting evidence regarding the role of PARG. In (Fouquerel et al., 2014Fouquerel E, Goellner Eva M, Yu Z, Gagné J-P, Barbi de Moura M, Feinstein T, Wheeler D, Redpath P, Li J, Romero G et al. (2014) ARTD1/PARP1 negatively regulates glycolysis by inhibiting hexokinase 1 independent of NAD+ Depletion. Cell Rep 8:1819-1831.), PARG knockdown rescued the PARP1-dependent glycolytic defect and ATP depletion, while in (Andrabi et al., 2014Andrabi SA, Umanah GK, Chang C, Stevens DA, Karuppagounder SS, Gagné JP, Poirier GG, Dawson VL and Dawson TM (2014 ) Poly(ADP-ribose) polymerase-dependent energy depletion occurs through inhibition of glycolysis. Proc Natl Acad Sci U S A 111:10209-10214.) a similar rescue of glycolysis was observed after PARG overexpression. Table 1 shows a compilation of results regarding the contribution of PARG to PARP1-dependent cell death and associated effects in different models, highlighting the considerable heterogeneity currently in the literature.

Table 1 -
Contribution of PARG activity, AIF translocation and TRPM2 channel gating to cell death by parthanatos. Only studies in which the induced cell death was shown to rely on PARP1 activity have been included.

These competing roles of PARG can, at least in theory, be ascribed to two separate functions that both depend on PARG catalytic activity, but have opposing effects on parthanatos execution. One possibility is centered around the formation of free PAR chains, which are thought to inhibit hexokinase (above) and release AIF from mitochondria (below) (Figure 1). PARG activity could be required for the formation of these free PAR chains via its endoglycohydrolase activity and therefore promote parthanatos, but high PARG activity may also degrade these free PAR chains after their formation, and therefore reduce cell death by parthanatos (Mashimo et al., 2013Mashimo M, Kato J and Moss J (2013) ADP-ribosyl-acceptor hydrolase 3 regulates poly (ADP-ribose) degradation and cell death during oxidative stress. Proc Natl Acad Sci U S A 110:18964-18969.). In this context, it is worth mentioning that, while PARG acts both as an exo- and endoglycohydrolase, its exoglycohydrolase activity is thought to be predominant (Barkauskaite et al., 2015Barkauskaite E, Jankevicius G and Ahel I (2015) Structures and mechanisms of enzymes employed in the synthesis and degradation of PARP-dependent protein ADP-Ribosylation. Mol Cell 58:935-946.), indicating that PAR hydrolysis generates mostly ADP-ribose monomers, not free PAR chains. Moreover, high levels of nuclear PARG catalytic activity imply that any free PAR polymers generated in the nucleus must be protected from PARG activity in order to reach the cytosol at any significant amounts. A further complication in the interpretation of the contribution of PARG to parthanatos is that long-term PARG deletion can affect the activation of PARP1, since PARP1 auto-modification inhibits its DNA binding, such that the accumulation of spontaneously auto-modified PARP1 in PARG KO cells can reduce the population of PARP1 molecules that can engage in DNA damage-induced PARylation (Gogola et al., 2018Gogola E, Duarte AA, de Ruiter JR, Wiegant WW, Schmid JA, de Bruijn R, James DI, Guerrero Llobet S, Vis DJ, Annunziato S et al. (2018) Selective Loss of PARG Restores PARylation and Counteracts PARP Inhibitor-Mediated Synthetic Lethality. Cancer Cell 33:1078-1093.e1012.).

ARH3, on the other hand, is thought to play a protective role in parthanatos, which is ascribed to its PAR-degrading activity, which would reduce the accumulation of free PAR polymers and prevent parthanatos induction (Mashimo et al., 2013Mashimo M, Kato J and Moss J (2013) ADP-ribosyl-acceptor hydrolase 3 regulates poly (ADP-ribose) degradation and cell death during oxidative stress. Proc Natl Acad Sci U S A 110:18964-18969.) (Figure 1). In agreement with this model, ARH3-deficient mice are more sensitive to ischaemia-reperfusion injury and ARH3-deficient human patients present neurodegenerative disorders and their fibroblasts are more sensitive to H2O2-induced parthanatos (Danhauser et al., 2018Danhauser K, Alhaddad B, Makowski C, Piekutowska-Abramczuk D, Syrbe S, Gomez-Ospina N, Manning MA, Kostera-Pruszczyk A, Krahn-Peper C, Berutti R et al. (2018) Bi-allelic ADPRHL2 mutations cause neurodegeneration with developmental delay, ataxia, and axonal neuropathy. Am J Hum Genet 103:817-825.; Ghosh et al., 2018Ghosh SG, Becker K, Huang H, Dixon-Salazar T, Chai G, Salpietro V, Al-Gazali L, Waisfisz Q, Wang H, Vaux KK et al. (2018) Biallelic mutations in ADPRHL2, encoding ADP-Ribosylhydrolase 3, lead to a degenerative pediatric stress-induced epileptic ataxia syndrome. Am J Hum Genet 103:826.). However, an alternative explanation for neurodegeneration in these patients could be that failure to remove mono-ADP-ribosylation from core histones leads to epigenetic changes that culminate in transcription deregulation (Hanzlikova et al., 2020Hanzlikova H, Prokhorova E, Krejcikova K, Cihlarova Z, Kalasova I, Kubovciak J, Sachova J, Hailstone R, Brazina J, Ghosh S et al. (2020) Pathogenic ARH3 mutations result in ADP-ribose chromatin scars during DNA strand break repair. Nat Commun 11:3391.), which would be independent of parthanatos.

Open questions:

  • Do PAR hydrolases, and PARG in particular, promote or inhibit parthanatos execution?

  • How are free PAR chains generated at sufficiently high amounts, protected from hydrolytic enzymes and then transported out of the nucleus?

ADP-ribose monomers and Ca2+ release

While the nicotinamide moiety of NAD+ released during PAR synthesis is predominantly recycled back to NAD+ by the NAD+ salvage pathway (Covarrubias et al., 2021Covarrubias AJ, Perrone R, Grozio A and Verdin E (2021) NAD+ metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol 22:119-141.), the ADP-ribose moiety transferred onto target proteins and subsequently released by PAR/MAR hydrolases generates free ADP-ribose monomers (Figure 1). This free ADP-ribose can bind to the calcium channel TRPM2, which contains two ADP-ribose binding sites that regulate channel opening (Perraud et al., 2001Perraud AL, Fleig A, Dunn CA, Bagley LA, Launay P, Schmitz C, Stokes AJ, Zhu Q, Bessman MJ, Penner R et al. (2001) ADP-ribose gating of the calcium-permeable LTRPC2 channel revealed by Nudix motif homology. Nature 411:595-599.; Huang et al., 2018Huang Y, Winkler PA, Sun W, Lü W and Du J (2018) Architecture of the TRPM2 channel and its activation mechanism by ADP-ribose and calcium. Nature 562:145-149.; Szollosi 2021Szollosi A (2021) Two decades of evolution of our understanding of the Transient Receptor Potential Melastatin 2 (TRPM2) Cation Channel. Life 11:397.). In several cell types, increases in intracellular Ca2+ were observed upon oxidative stress, were accompanied by the accumulation of free ADP-ribose and relied on PARP1 activation and TRPM2 gating (Fonfria et al., 2004Fonfria E, Marshall ICB, Benham CD, Boyfield I, Brown JD, Hill K, Hughes JP, Skaper SD and McNulty S (2004) TRPM2 channel opening in response to oxidative stress is dependent on activation of poly(ADP-ribose) polymerase. Br J Pharmacol 143:186-192.; Perraud et al., 2005Perraud AL, Takanishi CL, Shen B, Kang S, Smith MK, Schmitz C, Knowles HM, Ferraris D, Li W, Zhang J et al. (2005) Accumulation of free ADP-ribose from mitochondria mediates oxidative stress-induced gating of TRPM2 cation channels. J Biol Chem 280:6138-6148.; Yang et al., 2006Yang KT, Chang WL, Yang PC, Chien CL, Lai MS, Su MJ and Wu ML (2006) Activation of the transient receptor potential M2 channel and poly(ADP-ribose) polymerase is involved in oxidative stress-induced cardiomyocyte death. Cell Death Differ 13:1815-1826.). Although in some systems there is evidence for ADP-ribose-independent, but oxidative stress-dependent TRPM2 opening (Wehage et al., 2002Wehage E, Eisfeld J, Heiner I, Jüngling E, Zitt C and Lückhoff A (2002) Activation of the cation channel long transient receptor potential channel 2 (LTRPC2) by hydrogen peroxide. A splice variant reveals a mode of activation independent of ADP-ribose. J Biol Chem 277:23150-23156.), the ADPr-dependent activation requires PARG activity (Blenn et al., 2011Blenn C, Wyrsch P, Bader J, Bollhalder M and Althaus FR (2011) Poly(ADP-ribose)glycohydrolase is an upstream regulator of Ca2+ fluxes in oxidative cell death. Cell Mol Life Sci 68:1455-1466.), arguing in favour of a PARP1/PARG-dependent route of ADP-ribose generation. Consistent with this, induction of parthanatos using MNNG can also lead to calcium influx (Chiu et al., 2011Chiu LY, Ho FM, Shiah SG, Chang Y and Lin WW (2011) Oxidative stress initiates DNA damager MNNG-induced poly(ADP-ribose)polymerase-1-dependent parthanatos cell death. Biochem Pharmacol 81:459-470.). Interestingly, in a model of renal ischaemia/reperfusion injury, cell death can be prevented both by PARP1 inhibition or Ca2+ chelation, suggesting an important role of Ca2+ influx for parthanatos execution (Zhang et al., 2014Zhang F, Xie R, Munoz FM, Lau SS and Monks TJ (2014) PARP-1 hyperactivation and reciprocal elevations in intracellular Ca2+ during ROS-induced nonapoptotic cell death. Toxicol Sci 140:118-134.). In another study, Ca2+ chelation only suppressed cell death upon H2O2 treatment, but not upon MNNG treatment, indicating that this effect could be specific to particular insults (Bentle et al., 2006Bentle MS, Reinicke KE, Bey EA, Spitz DR and Boothman DA (2006) Calcium-dependent modulation of poly(ADP-ribose) polymerase-1 alters cellular metabolism and DNA repair. J Biol Chem 281:33684-33696.). A further complication in the interpretation of this data is that Ca2+ may affect PARP1 activation by a poorly understood mechanism (Zhang et al., 2014Zhang F, Xie R, Munoz FM, Lau SS and Monks TJ (2014) PARP-1 hyperactivation and reciprocal elevations in intracellular Ca2+ during ROS-induced nonapoptotic cell death. Toxicol Sci 140:118-134.). While TRPM2 is a cell-membrane resident channel and therefore can only cause Ca2+ influxes from the extracellular space, Ca2+ release from the endoplasmic reticulum may also contribute to parthanatos (Munoz et al., 2017Munoz FM, Zhang F, Islas-Robles A, Lau SS and Monks TJ (2017) ROS-induced store-operated Ca2+ entry coupled to PARP-1 hyperactivation is independent of PARG activity in necrotic cell death. Toxicol Sci 158:444-453.; Zhong et al., 2018Zhong H, Song R, Pang Q, Liu Y, Zhuang J, Chen Y, Hu J, Hu J, Liu Y, Liu Z et al. (2018) Propofol inhibits parthanatos via ROS-ER-calcium-mitochondria signal pathway in vivo and vitro. Cell Death Dis 9:932.). Interestingly, unlike TRPM2 gating, this effect was independent of PARG, suggesting a different mechanism of channel opening (Zhang et al., 2014Zhang F, Xie R, Munoz FM, Lau SS and Monks TJ (2014) PARP-1 hyperactivation and reciprocal elevations in intracellular Ca2+ during ROS-induced nonapoptotic cell death. Toxicol Sci 140:118-134.; Munoz et al., 2017Munoz FM, Zhang F, Islas-Robles A, Lau SS and Monks TJ (2017) ROS-induced store-operated Ca2+ entry coupled to PARP-1 hyperactivation is independent of PARG activity in necrotic cell death. Toxicol Sci 158:444-453.).

Open questions:

  • Is ADP-ribose-induced TRPM2 gating necessary and/or sufficient for parthanatos execution?

  • Are there TRPM2-dependent and TRPM2-independent modes of parthanatos?

  • What are the downstream molecular effects of TRPM2-mediated increases in intracellular Ca2+?

ADP-ribose degradation into AMP

Free ADP-ribose can also be further degraded into AMP and ribose-5-phosphate by phosphodiesterases of the Nudix superfamily (Carreras-Puigvert et al., 2017Carreras-Puigvert J, Zitnik M, Jemth A-S, Carter M, Unterlass JE, Hallström B, Loseva O, Karem Z, Calderón-Montaño JM, Lindskog C et al. (2017) A comprehensive structural, biochemical and biological profiling of the human NUDIX hydrolase family. Nat Commun 8:1541.) (Figure 1). These enzymes target the phosphodiester bond in ADP-ribose and can degrade either free ADPr or leave a phosphoribose modification on previously ADP-ribosylated proteins, although the detection of protein phosphoribose modification is currently limited to in vitro reactions (Daniels et al., 2015Daniels CM, Thirawatananond P, Ong SE, Gabelli SB and Leung AK (2015) Nudix hydrolases degrade protein-conjugated ADP-ribose. Sci Rep 5:18271.; Palazzo et al., 2015Palazzo L, Thomas B, Jemth A-S, Colby TD, Leidecker O, Feijs KLH, Žaja R, Loseva OI, Puigvert JC, Matic I et al. (2015) Processing of protein ADP-ribosylation by Nudix hydrolases. Biochem J 468 2:293-301.; O’Sullivan et al., 2019O’Sullivan J, Tedim Ferreira M, Gagné JP, Sharma AK, Hendzel MJ, Masson JY and Poirier GG (2019) Emerging roles of eraser enzymes in the dynamic control of protein ADP-ribosylation. Nat Commun 10:1182.). The resultant ribose 5-phosphate could have many metabolic fates, including the formation of phosphoribosyl pyrophosphate (PRPP), which is required for the NAD+ salvage pathway (Figure 2). Interestingly, the complete cycle of NAD+ salvage, from conversion of NAD+ to an ADP-ribose unit by PARP1 back to a full NAD+ molecule using the same carbon backbones, has an energetic cost of four high-energy phosphate groups per ADP-ribose unit attached to a protein (Figure 2). Given that NAD(H) concentrations are roughly in the 0.3 mM range (Yang et al., 2007Yang H, Yang T, Baur JA, Perez E, Matsui T, Carmona JJ, Lamming DW, Souza-Pinto NC, Bohr VA, Rosenzweig A et al. (2007) Nutrient-sensitive mitochondrial NAD+ levels dictate cell survival. Cell 130:1095-1107.), while ATP concentrations are only around 10x higher, in the 3-4 mM range (Greiner and Glonek, 2021Greiner JV and Glonek T (2021) Intracellular ATP Concentration and Implication for Cellular Evolution. Biology (Basel) 10:1166.), the full consumption of cellular NAD+ by PARP1 and its subsequent salvage couId make a substantial contribution to ATP depletion during parthanatos. While the relative contributions of this Nudix-dependent salvage pathway as opposed to glycolysis inhibition (see above) to energetic collapse during parthanatos is unclear, the accumulation of ADP and particularly AMP may be an important signal in cell death after PARP1 hyperactivation (Formentini et al., 2009Formentini L, Macchiarulo A, Cipriani G, Camaioni E, Rapizzi E, Pellicciari R, Moroni F and Chiarugi A (2009) Poly(ADP-ribose) catabolism triggers AMP-dependent mitochondrial energy failure. J Biol Chem 284:17668-17676.). Illustrating this, MNNG treatment of HEK-293 cells led to activation of the AMP-activated kinase (AMPK) attributed to increased AMP/ATP ratios, which then inhibited the mTORC1 signaling pathway, involved in the regulation of cell death/survival and energy metabolism (Ethier et al., 2012Ethier C, Tardif M, Arul L and Poirier GG (2012) PARP-1 modulation of mTOR signaling in response to a DNA alkylating agent. PLoS One 7:e47978.) (Figure 1). While this would suggest the induction of an autophagic response, as observed in some parthanatos models (Zhou et al., 2013Zhou J, Ng S, Huang Q, Wu Y-T, Li Z, Yao SQ and Shen H-M (2013) AMPK mediates a pro-survival autophagy downstream of PARP-1 activation in response to DNA alkylating agents. FEBS Letters 587:170-177.; Jiang et al., 2018Jiang HY, Yang Y, Zhang YY, Xie Z, Zhao XY, Sun Y and Kong WJ (2018) The dual role of poly(ADP-ribose) polymerase-1 in modulating parthanatos and autophagy under oxidative stress in rat cochlear marginal cells of the stria vascularis. Redox Biol 14:361-370.), whether AMPK activation and autophagy contribute to cell death execution by parthanatos or are protective mechanisms is currently unclear.

Figure 2 -
A full cycle of NAD+ salvage costs four high-energy phosphate groups per NAD+ molecule consumed by PARP1.

Open questions:

  • Are Nudix hydrolases required for parthanatos execution?

  • What is the relative contribution of AMP generated from ADP-ribose hydrolysis, as opposed to ATP depletion from glycolysis inhibition (above), for the AMPK activation/autophagy observed in parthanatos?

  • How do AMPK activation and autophagy affect cell death by parthanatos?

AIF translocation and DNA fragmentation

Apoptosis-Inducing Factor (AIF) is a mitochondrial flavoprotein that plays a role in the assembly of the respiratory chain complexes, but is also involved in cell death mechanisms (Susin et al., 1999Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M et al. (1999) Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397:441-446.; Vahsen et al., 2004Vahsen N, Candé C, Brière J-J, Bénit P, Joza N, Larochette N, Mastroberardino PG, Pequignot MO, Casares N, Lazar V et al. (2004) AIF deficiency compromises oxidative phosphorylation. EMBO J 23:4679-4689.). It is normally located in the inner mitochondrial membrane, facing the inter-membrane space (Otera et al., 2005Otera H, Ohsakaya S, Nagaura Z-I, Ishihara N and Mihara K (2005) Export of mitochondrial AIF in response to proapoptotic stimuli depends on processing at the intermembrane space. EMBO J 24:1375-1386.), but can also be found loosely associated with the outer mitochondrial membrane (Yu et al., 2009Yu SW, Wang Y, Frydenlund DS, Ottersen OP, Dawson VL and Dawson TM (2009) Outer mitochondrial membrane localization of apoptosis-inducing factor: Mechanistic implications for release. ASN Neuro 1:e00021.). In response to PARP1 hyperactivation, AIF is often observed to translocate from the mitochondria to the nucleus (Yu et al., 2002Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ, Poirier GG, Dawson TM and Dawson VL (2002) Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 297:259-263.), but some models of PARP1-dependent cell death do not lead to observable AIF translocation, indicating that there may be AIF-dependent and AIF-independent forms of parthanatos (Table 1). For example, retinal cells and macrophages do not seem to exhibit AIF translocation after PARP1-dependent cell death induction (Jang et al., 2017Jang KH, Do YJ, Son D, Son E, Choi JS and Kim E (2017) AIF-independent parthanatos in the pathogenesis of dry age-related macular degeneration. Cell Death Dis 8:e2526.; Regdon et al., 2019Regdon Z, Robaszkiewicz A, Kovács K, Rygielska Ż, Hegedűs C, Bodoor K, Szabó É and Virág L (2019) LPS protects macrophages from AIF-independent parthanatos by downregulation of PARP1 expression, induction of SOD2 expression, and a metabolic shift to aerobic glycolysis. Free Radic Biol Med 131:184-196.). Interestingly, AIF translocation to the nucleus is also observed in response to some apoptotic stimuli (Daugas et al., 2000Daugas E, Susin SA, Zamzami N, Ferri KF, Irinopoulou T, Larochette N, Prévost M-C, Leber B, Andrews D, Penninger J et al. (2000) Mitochondrio-nuclear translocation of AIF in apoptosis and necrosis. FASEB J 14:729-739.), which was recently suggested to also rely on PARP1 activation (Mashimo et al., 2021Mashimo M, Onishi M, Uno A, Tanimichi A, Nobeyama A, Mori M, Yamada S, Negi S, Bu X, Kato J et al. (2021) The 89-kDa PARP1 cleavage fragment serves as a cytoplasmic PAR carrier to induce AIF-mediated apoptosis. J Biol Chem 296:100046.).

In the context of parthanatos, AIF is thought to be released from mitochondria via direct interaction with free PAR polymers (above) (Yu et al., 2006Yu S-W, Andrabi SA, Wang H, Kim NS, Poirier GG, Dawson TM and Dawson VL (2006) Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci U S A 103:18314-18319.; Wang et al., 2011Wang Y, Kim NS, Haince JF, Kang HC, David KK, Andrabi SA, Poirier GG, Dawson VL and Dawson TM (2011) Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos), Sci Signal 4:ra20.), but the molecular details of this process are currently unclear. Alternatively, AIF release may proceed via its proteolysis, which has been observed in some situations to rely on calpain I, which is a Ca2+ -dependent protease, and therefore could in principle respond to TRPM2-dependent Ca2+ influxes or endoplasmic reticulum calcium release (Polster et al., 2005Polster BM, Basañez G, Etxebarria A, Hardwick JM and Nicholls DG (2005) Calpain I induces cleavage and release of apoptosis-inducing factor from isolated mitochondria. J Biol Chem 280:6447-6454.; Norberg et al., 2008Norberg E, Gogvadze V, Ott M, Horn M, Uhlén P, Orrenius S and Zhivotovsky B (2008) An increase in intracellular Ca2+ is required for the activation of mitochondrial calpain to release AIF during cell death. Cell Death Differ 15:1857-1864.; Vosler et al., 2009Vosler PS, Sun D, Wang S, Gao Y, Kintner DB, Signore AP, Cao G and Chen J (2009) Calcium dysregulation induces apoptosis-inducing factor release: Cross-talk between PARP-1- and calpain-signaling pathways. Exp Neurol 218:213-220.; Sun et al., 2018Sun Y, Sukumaran P, Selvaraj S, Cilz NI, Schaar A, Lei S and Singh BB (2018) TRPM2 Promotes neurotoxin MPP(+)/MPTP-induced cell death. Mol Neurobiol 55:409-420.). However, there is rather strong evidence against a central role of calpain cleavage on AIF release, at least in some parthanatos models (Wang et al., 2009Wang Y, Kim NS, Li X, Greer PA, Koehler RC, Dawson VL and Dawson TM (2009) Calpain activation is not required for AIF translocation in PARP-1-dependent cell death (parthanatos). J Neurochem 110:687-696.). Another possible contributor to AIF release from mitochondria is the mitochondrial permeability transition pore, which is an ill-defined molecular entity that allows the non-selective diffusion of small molecules through the mitochondrial inner membrane, which can lead to mitochondrial swelling and rupture, and is associated to several cell death mechanisms (Yu et al., 2006Yu S-W, Andrabi SA, Wang H, Kim NS, Poirier GG, Dawson TM and Dawson VL (2006) Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci U S A 103:18314-18319.; Bernardi et al., 2023Bernardi P, Gerle C, Halestrap AP, Jonas EA, Karch J, Mnatsakanyan N, Pavlov E, Sheu S-S and Soukas AA (2023) Identity, structure, and function of the mitochondrial permeability transition pore: Controversies, consensus, recent advances, and future directions. Cell Death Differ 30:1869-1885.) (Figure 1).

AIF translocation to the nucleus is associated with large scale DNA fragmentation, culminating in cell death. Two mechanisms for AIF-induced DNA cleavage have been proposed (Figure 1). One model suggests that cytoplasmic AIF interacts with macrophage migration inhibitory factor (MIF), leading to the nuclear translocation of MIF, which was identified to have a nuclease activity (Wang et al., 2016Wang Y, An R, Umanah GK, Park H, Nambiar K, Eacker SM, Kim B, Bao L, Harraz MM, Chang C et al. (2016) A nuclease that mediates cell death induced by DNA damage and poly(ADP-ribose) polymerase-1. Science 354:aad6872.). In agreement with this model, a specific inhibitor of MIF nuclease activity was recently shown to protect cells from parthanatos in a mouse model of parkinsonism (Park et al., 2022Park H, Kam TI, Peng H, Chou SC, Mehrabani-Tabari AA, Song JJ, Yin X, Karuppagounder SS, Umanah GK, Rao AVS et al. (2022) PAAN/MIF nuclease inhibition prevents neurodegeneration in Parkinson’s disease. Cell 185:1943-1959e1921.). The second model is based on the recent identification of a nuclease activity in AIF itself, which is proposed to degrade DNA in a complex formed between AIF, cyclophilin A and histone H2AX (Artus et al., 2010Artus C, Boujrad H, Bouharrour A, Brunelle M-N, Hoos S, Yuste VJ, Lenormand P, Rousselle J-C, Namane A, England P et al. (2010) AIF promotes chromatinolysis and caspase-independent programmed necrosis by interacting with histone H2AX. EMBO J 29:1585-1599.; Novo et al., 2022Novo N, Romero-Tamayo S, Marcuello C, Boneta S, Blasco-Machin I, Velázquez-Campoy A, Villanueva R, Moreno-Loshuertos R, Lostao A, Medina M et al. (2022) Beyond a platform protein for the degradosome assembly: The Apoptosis-Inducing Factor as an efficient nuclease involved in chromatinolysis. PNAS Nexus 2:pgac312.).

Open questions:

  • What is the precise sequence of molecular events that promotes AIF release from mitochondria?

  • How does translocation of AIF promote DNA fragmentation and what protein(s) catalyse(s) DNA cleavage?

  • What factor(s) define(s) AIF-dependent and AIF-independent parthanatos and what process leads to DNA fragmentation in AIF-independent parthanatos?

  • What are the differences and similarities between apoptotic and parthanatic AIF translocation?

Conclusions

In the late 1970s, Goodwin and colleagues first showed that DNA damage can induce the depletion of NAD and ATP levels, and that PARP1 activity is central to this effect (Goodwin et al., 1978Goodwin PM, Lewis PJ, Davies MI, Skidmore CJ and Shall S (1978) The effect of gamma radiation and neocarzinostatin of NAD and ATP levels in mouse leukaemia cells. Biochim Biophys Acta Gen Subj 543:576-582.). Almost 50 years of research since then have led to the identification of a range of different stimuli that induce PARP1 hyperactivation and a variety of pathological situations in which PARP1 activation seems to contribute to cell death and tissue damage. However, several questions and inconsistencies still remain regarding the sequence of molecular events that drive cell death by parthanatos. While a number of key mechanisms have already been described, it remains unclear which events are necessary and sufficient for parthanatos execution and how each of these steps connects to the next one in the cascade. Complicating matters even further, there seem to be clear differences in how parthanatos proceeds in different cell types and at different metabolic states. With this review, we aim to highlight the urgent need for studies that determine the contribution of several steps along the cascade in single, well-defined model systems. Only by comparing all of these steps between different models in which NAD+ supplementation, PARG activity, AIF translocation or TRPM2 gating play differential roles, can we hope to shed light on whether there are multiple pathways of parthanatos or if a single pathway integrates all of these events. Although technically difficult and inherently multidisciplinary, this will be critical to better understand not only how this pathway operates, but also how other cell death mechanisms, such as apoptosis, are interconnected to parthanatos. A better definition of these mechanisms will be central to clarify the contribution of PARP1-dependent cell death to human pathology, particularly in a variety of neurodegenerative disorders, such as Parkinson’s and Alzheimer’s disease, which are of rising concern in an aging human population.

Acknowledgements

The authors wish to apologize to the colleagues whose work could not be mentioned here. Work in the NCH lab is funded by FAPESP grant 2018/18007-5.

References

  • Abplanalp J, Leutert M, Frugier E, Nowak K, Feurer R, Kato J, Kistemaker HVA, Filippov DV, Moss J, Caflisch A et al (2017) Proteomic analyses identify ARH3 as a serine mono-ADP-ribosylhydrolase. Nat Commun 8:2055.
  • Alano CC, Garnier P, Ying W, Higashi Y, Kauppinen TM and Swanson RA (2010) NAD+ depletion is necessary and sufficient for poly(ADP-ribose) polymerase-1-mediated neuronal death. J Neurosci 30:2967-2978.
  • Alano CC, Ying W and Swanson RA (2004) Poly(ADP-ribose) polymerase-1-mediated cell death in astrocytes requires NAD+ depletion and mitochondrial permeability transition. J Biol Chem 279:18895-18902.
  • An X, Fu Z, Mai C, Wang W, Wei L, Li D, Li C and Jiang LH (2019) Increasing the TRPM2 channel expression in human neuroblastoma SH-SY5Y cells augments the susceptibility to ROS-Induced Cell Death. Cells 8:28.
  • Andrabi SA, Kang HC, Haince JF, Lee YI, Zhang J, Chi Z, West AB, Koehler RC, Poirier GG, Dawson TM et al (2011) Iduna protects the brain from glutamate excitotoxicity and stroke by interfering with poly(ADP-ribose) polymer-induced cell death. Nat Med 17:692-699.
  • Andrabi SA, Kim NS, Yu SW, Wang H, Koh DW, Sasaki M, Klaus JA, Otsuka T, Zhang Z, Koehler RC et al (2006) Poly(ADP-ribose) (PAR) polymer is a death signal. Proc Natl Acad Sci U S A 103:18308-18313.
  • Andrabi SA, Umanah GK, Chang C, Stevens DA, Karuppagounder SS, Gagné JP, Poirier GG, Dawson VL and Dawson TM (2014 ) Poly(ADP-ribose) polymerase-dependent energy depletion occurs through inhibition of glycolysis. Proc Natl Acad Sci U S A 111:10209-10214.
  • Artus C, Boujrad H, Bouharrour A, Brunelle M-N, Hoos S, Yuste VJ, Lenormand P, Rousselle J-C, Namane A, England P et al (2010) AIF promotes chromatinolysis and caspase-independent programmed necrosis by interacting with histone H2AX. EMBO J 29:1585-1599.
  • Barkauskaite E, Jankevicius G and Ahel I (2015) Structures and mechanisms of enzymes employed in the synthesis and degradation of PARP-dependent protein ADP-Ribosylation. Mol Cell 58:935-946.
  • Bentle MS, Reinicke KE, Bey EA, Spitz DR and Boothman DA (2006) Calcium-dependent modulation of poly(ADP-ribose) polymerase-1 alters cellular metabolism and DNA repair. J Biol Chem 281:33684-33696.
  • Berger NA (1985) Poly(ADP-Ribose) in the cellular response to DNA damage. Radiat Res 101:4-15.
  • Berger NA, Besson VC, Boulares AH, Burkle A, Chiarugi A, Clark RS, Curtin NJ, Cuzzocrea S, Dawson TM, Dawson VL et al (2018) Opportunities for the repurposing of PARP inhibitors for the therapy of non-oncological diseases. Br J Pharmacol 175:192-222.
  • Bernardi P, Gerle C, Halestrap AP, Jonas EA, Karch J, Mnatsakanyan N, Pavlov E, Sheu S-S and Soukas AA (2023) Identity, structure, and function of the mitochondrial permeability transition pore: Controversies, consensus, recent advances, and future directions. Cell Death Differ 30:1869-1885.
  • Blenn C, Althaus FR and Malanga M (2006) Poly(ADP-ribose) glycohydrolase silencing protects against H2O2-induced cell death. Biochem J 396:419-429.
  • Blenn C, Wyrsch P, Bader J, Bollhalder M and Althaus FR (2011) Poly(ADP-ribose)glycohydrolase is an upstream regulator of Ca2+ fluxes in oxidative cell death. Cell Mol Life Sci 68:1455-1466.
  • Buonvicino D, Formentini L, Cipriani G and Chiarugi A (2013) Glucose deprivation converts poly(ADP-ribose) polymerase-1 hyperactivation into a transient energy-producing process. J Biol Chem 288:36530-36537.
  • Caldecott KW (2022) DNA single-strand break repair and human genetic disease. Trends Cell Biol 32:733-745.
  • Cambronne XA, Stewart ML, Kim D, Jones-Brunette AM, Morgan RK, Farrens DL, Cohen MS and Goodman RH (2016) Biosensor reveals multiple sources for mitochondrial NAD(+). Science 352:1474-1477.
  • Carreras-Puigvert J, Zitnik M, Jemth A-S, Carter M, Unterlass JE, Hallström B, Loseva O, Karem Z, Calderón-Montaño JM, Lindskog C et al (2017) A comprehensive structural, biochemical and biological profiling of the human NUDIX hydrolase family. Nat Commun 8:1541.
  • Chiu LY, Ho FM, Shiah SG, Chang Y and Lin WW (2011) Oxidative stress initiates DNA damager MNNG-induced poly(ADP-ribose)polymerase-1-dependent parthanatos cell death. Biochem Pharmacol 81:459-470.
  • Covarrubias AJ, Perrone R, Grozio A and Verdin E (2021) NAD+ metabolism and its roles in cellular processes during ageing. Nat Rev Mol Cell Biol 22:119-141.
  • Cuzzocrea S, Di Paola R, Mazzon E, Cortes U, Genovese T, Muià C, Li W, Xu W, Li JH, Zhang J et al (2005) PARG activity mediates intestinal injury induced by splanchnic artery occlusion and reperfusion. FASEB J 19:558-566.
  • D’Amours D, Sallmann FR, Dixit VM and Poirier GG (2001) Gain-of-function of poly(ADP-ribose) polymerase-1 upon cleavage by apoptotic proteases: Implications for apoptosis. J Cell Sci 114:3771-3778.
  • Danhauser K, Alhaddad B, Makowski C, Piekutowska-Abramczuk D, Syrbe S, Gomez-Ospina N, Manning MA, Kostera-Pruszczyk A, Krahn-Peper C, Berutti R et al (2018) Bi-allelic ADPRHL2 mutations cause neurodegeneration with developmental delay, ataxia, and axonal neuropathy. Am J Hum Genet 103:817-825.
  • Daniels CM, Thirawatananond P, Ong SE, Gabelli SB and Leung AK (2015) Nudix hydrolases degrade protein-conjugated ADP-ribose. Sci Rep 5:18271.
  • Daugas E, Susin SA, Zamzami N, Ferri KF, Irinopoulou T, Larochette N, Prévost M-C, Leber B, Andrews D, Penninger J et al (2000) Mitochondrio-nuclear translocation of AIF in apoptosis and necrosis. FASEB J 14:729-739.
  • Dawson TM and Dawson VL (2018) Nitric Oxide signaling in neurodegeneration and cell death. Adv Pharmacol 82:57-83.
  • Eliasson MJL, Sampei K, Mandir AS, Hurn PD, Traystman RJ, Bao J, Pieper A, Wang Z-Q, Dawson TM, Snyder SH et al (1997) Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nat Med 3:1089-1095.
  • Ethier C, Tardif M, Arul L and Poirier GG (2012) PARP-1 modulation of mTOR signaling in response to a DNA alkylating agent. PLoS One 7:e47978.
  • Fatokun AA, Dawson VL and Dawson TM (2014) Parthanatos: Mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 171:2000-2016.
  • Fehr AR, Singh SA, Kerr CM, Mukai S, Higashi H and Aikawa M (2020) The impact of PARPs and ADP-ribosylation on inflammation and host-pathogen interactions. Genes Dev 34:341-359.
  • Fonfria E, Marshall ICB, Benham CD, Boyfield I, Brown JD, Hill K, Hughes JP, Skaper SD and McNulty S (2004) TRPM2 channel opening in response to oxidative stress is dependent on activation of poly(ADP-ribose) polymerase. Br J Pharmacol 143:186-192.
  • Fonfria E, Marshall IC, Boyfield I, Skaper SD, Hughes JP, Owen DE, Zhang W, Miller BA, Benham CD and McNulty S (2005) Amyloid beta-peptide(1-42) and hydrogen peroxide-induced toxicity are mediated by TRPM2 in rat primary striatal cultures. J Neurochem 95:715-723.
  • Fontana P, Bonfiglio JJ, Palazzo L, Bartlett E, Matic I and Ahel I (2017) Serine ADP-ribosylation reversal by the hydrolase ARH3. Elife 6:e28533.
  • Formentini L, Macchiarulo A, Cipriani G, Camaioni E, Rapizzi E, Pellicciari R, Moroni F and Chiarugi A (2009) Poly(ADP-ribose) catabolism triggers AMP-dependent mitochondrial energy failure. J Biol Chem 284:17668-17676.
  • Fouquerel E, Goellner Eva M, Yu Z, Gagné J-P, Barbi de Moura M, Feinstein T, Wheeler D, Redpath P, Li J, Romero G et al (2014) ARTD1/PARP1 negatively regulates glycolysis by inhibiting hexokinase 1 independent of NAD+ Depletion. Cell Rep 8:1819-1831.
  • Ghosh SG, Becker K, Huang H, Dixon-Salazar T, Chai G, Salpietro V, Al-Gazali L, Waisfisz Q, Wang H, Vaux KK et al (2018) Biallelic mutations in ADPRHL2, encoding ADP-Ribosylhydrolase 3, lead to a degenerative pediatric stress-induced epileptic ataxia syndrome. Am J Hum Genet 103:826.
  • Girardi E, Agrimi G, Goldmann U, Fiume G, Lindinger S, Sedlyarov V, Srndic I, Gürtl B, Agerer B, Kartnig F et al (2020) Epistasis-driven identification of SLC25A51 as a regulator of human mitochondrial NAD import. Nat Commun 11:6145.
  • Gogola E, Duarte AA, de Ruiter JR, Wiegant WW, Schmid JA, de Bruijn R, James DI, Guerrero Llobet S, Vis DJ, Annunziato S et al (2018) Selective Loss of PARG Restores PARylation and Counteracts PARP Inhibitor-Mediated Synthetic Lethality. Cancer Cell 33:1078-1093.e1012.
  • Goodwin PM, Lewis PJ, Davies MI, Skidmore CJ and Shall S (1978) The effect of gamma radiation and neocarzinostatin of NAD and ATP levels in mouse leukaemia cells. Biochim Biophys Acta Gen Subj 543:576-582.
  • Greiner JV and Glonek T (2021) Intracellular ATP Concentration and Implication for Cellular Evolution. Biology (Basel) 10:1166.
  • Ha HC and Snyder SH (1999) Poly(ADP-ribose) polymerase is a mediator of necrotic cell death by ATP depletion. Proc Natl Acad Sci U S A 96:13978-13982.
  • Hanzlikova H, Kalasova I, Demin AA, Pennicott LE, Cihlarova Z and Caldecott KW (2018) The Importance of Poly(ADP-Ribose) polymerase as a sensor of unligated okazaki fragments during DNA Replication. Mol Cell 71:319-331e313.
  • Hanzlikova H, Prokhorova E, Krejcikova K, Cihlarova Z, Kalasova I, Kubovciak J, Sachova J, Hailstone R, Brazina J, Ghosh S et al (2020) Pathogenic ARH3 mutations result in ADP-ribose chromatin scars during DNA strand break repair. Nat Commun 11:3391.
  • Hendriks IA, Buch-Larsen SC, Prokhorova E, Elsborg JD, Rebak AKLFS, Zhu K, Ahel D, Lukas C, Ahel I and Nielsen ML (2021) The regulatory landscape of the human HPF1- and ARH3-dependent ADP-ribosylome. Nat Commun 12:5893.
  • Herrmann GK, Russell WK, Garg NJ and Yin YW (2021) Poly(ADP-ribose) polymerase 1 regulates mitochondrial DNA repair in an NAD-dependent manner. J Biol Chem 296:100309.
  • Hoch NC, Hanzlikova H, Rulten SL, Tetreault M, Komulainen E, Ju L, Hornyak P, Zeng Z, Gittens W, Rey SA et al (2017) XRCC1 mutation is associated with PARP1 hyperactivation and cerebellar ataxia. Nature 541:87-91.
  • Hoch NC and Polo LM (2019) ADP-ribosylation: From molecular mechanisms to human disease. Genet Mol Biol 43:e20190075.
  • Hossain MI, Lee JH, Gagné JP, Khan J, Poirier GG, King PH, Dawson VL, Dawson TM and Andrabi SA (2024) Poly(ADP-ribose) mediates bioenergetic defects and redox imbalance in neurons following oxygen and glucose deprivation. FASEB J 38:e23556.
  • Hottiger MO (2015) Nuclear ADP-Ribosylation and its role in chromatin plasticity, cell differentiation, and epigenetics. Annu Rev Biochem 84:227-263.
  • Huang Y, Winkler PA, Sun W, Lü W and Du J (2018) Architecture of the TRPM2 channel and its activation mechanism by ADP-ribose and calcium. Nature 562:145-149.
  • Ishii M, Hagiwara T, Mori Y and Shimizu S (2014) Involvement of TRPM2 and L-type Ca²⁺ channels in Ca²⁺ entry and cell death induced by hydrogen peroxide in rat β-cell line RIN-5F. J Toxicol Sci 39:199-209.
  • Jang KH, Do YJ, Son D, Son E, Choi JS and Kim E (2017) AIF-independent parthanatos in the pathogenesis of dry age-related macular degeneration. Cell Death Dis 8:e2526.
  • Jiang HY, Yang Y, Zhang YY, Xie Z, Zhao XY, Sun Y and Kong WJ (2018) The dual role of poly(ADP-ribose) polymerase-1 in modulating parthanatos and autophagy under oxidative stress in rat cochlear marginal cells of the stria vascularis. Redox Biol 14:361-370.
  • Kam TI, Mao X, Park H, Chou SC, Karuppagounder SS, Umanah GE, Yun SP, Brahmachari S, Panicker N, Chen R et al (2018) Poly(ADP-ribose) drives pathologic alpha-synuclein neurodegeneration in Parkinson’s disease. Science 362:eaat8407.
  • Kang HC, Lee Y-I, Shin J-H, Andrabi SA, Chi Z, Gagné J-P, Lee Y, Ko HS, Lee BD, Poirier GG et al (2011) Iduna is a poly(ADP-ribose) (PAR)-dependent E3 ubiquitin ligase that regulates DNA damage. Proc Natl Acad Sci U S A 108:14103-14108.
  • Kim DS, Challa S, Jones A and Kraus WL (2020) PARPs and ADP-ribosylation in RNA biology: From RNA expression and processing to protein translation and proteostasis. Genes Dev 34:302-320.
  • Koh DW, Lawler AM, Poitras MF, Sasaki M, Wattler S, Nehls MC, Stöger T, Poirier GG, Dawson VL and Dawson TM (2004) Failure to degrade poly(ADP-ribose) causes increased sensitivity to cytotoxicity and early embryonic lethality. Proc Natl Acad Sci U S A 101:17699-17704.
  • Kolthur-Seetharam U, Dantzer F, McBurney MW, de Murcia G and Sassone-Corsi P (2006) Control of AIF-mediated cell death by the functional interplay of SIRT1 and PARP-1 in response to DNA damage. Cell Cycle 5:873-877.
  • Kory N, Uit de Bos J, van der Rijt S, Jankovic N, Güra M, Arp N, Pena IA, Prakash G, Chan SH, Kunchok T et al (2020) MCART1/SLC25A51 is required for mitochondrial NAD transport. Sci Adv 6:eabe5310.
  • Lee JH, Hussain M, Kim EW, Cheng SJ, Leung AKL, Fakouri NB, Croteau DL and Bohr VA (2022) Mitochondrial PARP1 regulates NAD(+)-dependent poly ADP-ribosylation of mitochondrial nucleoids. Exp Mol Med 54:2135-2147.
  • Li X, Yang W and Jiang LH (2017) Alteration in intracellular Zn(2+) homeostasis as a result of TRPM2 channel activation contributes to ROS-induced hippocampal cell death. Front Mol Neurosci 10:414.
  • Liu L, Li J, Ke Y, Zeng X, Gao J, Ba X and Wang R (2022a) The key players of parthanatos: Opportunities for targeting multiple levels in the therapy of parthanatos-based pathogenesis. Cell Mol Life Sci 79:60.
  • Liu S, Luo W and Wang Y (2022b) Emerging role of PARP-1 and PARthanatos in ischemic stroke. J Neurochem 160:74-87.
  • Longarini EJ, Dauben H, Locatelli C, Wondisford AR, Smith R, Muench C, Kolvenbach A, Lynskey ML, Pope A, Bonfiglio JJ et al (2023) Modular antibodies reveal DNA damage-induced mono-ADP-ribosylation as a second wave of PARP1 signaling. Mol Cell 83:1743-1760.e1711.
  • Lu XC, Massuda E, Lin Q, Li W, Li JH and Zhang J (2003) Post-treatment with a novel PARG inhibitor reduces infarct in cerebral ischemia in the rat. Brain Res 978:99-103.
  • Luongo TS, Eller JM, Lu MJ, Niere M, Raith F, Perry C, Bornstein MR, Oliphint P, Wang L, McReynolds MR et al (2020) SLC25A51 is a mammalian mitochondrial NAD(+) transporter. Nature 588:174-179.
  • Luscher B, Ahel I, Altmeyer M, Ashworth A, Bai P, Chang P, Cohen M, Corda D, Dantzer F, Daugherty MD et al (2021) ADP-ribosyltransferases, an update on function and nomenclature. FEBS J 289:7399-7410.
  • Ma D, Lu B, Feng C, Wang C, Wang Y, Luo T, Feng J, Jia H, Chi G, Luo Y et al (2016) Deoxypodophyllotoxin triggers parthanatos in glioma cells via induction of excessive ROS. Cancer Lett 371:194-204.
  • Mandir AS, Poitras MF, Berliner AR, Herring WJ, Guastella DB, Feldman A, Poirier GG, Wang ZQ, Dawson TM and Dawson VL (2000) NMDA but not non-NMDA excitotoxicity is mediated by Poly(ADP-ribose) polymerase. J Neurosci 20:8005-8011.
  • Mashimo M, Kato J and Moss J (2013) ADP-ribosyl-acceptor hydrolase 3 regulates poly (ADP-ribose) degradation and cell death during oxidative stress. Proc Natl Acad Sci U S A 110:18964-18969.
  • Mashimo M, Onishi M, Uno A, Tanimichi A, Nobeyama A, Mori M, Yamada S, Negi S, Bu X, Kato J et al (2021) The 89-kDa PARP1 cleavage fragment serves as a cytoplasmic PAR carrier to induce AIF-mediated apoptosis. J Biol Chem 296:100046.
  • Munoz FM, Zhang F, Islas-Robles A, Lau SS and Monks TJ (2017) ROS-induced store-operated Ca2+ entry coupled to PARP-1 hyperactivation is independent of PARG activity in necrotic cell death. Toxicol Sci 158:444-453.
  • Nishida T, Naguro I and Ichijo H (2022) NAMPT-dependent NAD(+) salvage is crucial for the decision between apoptotic and necrotic cell death under oxidative stress. Cell Death Discov 8:195.
  • Norberg E, Gogvadze V, Ott M, Horn M, Uhlén P, Orrenius S and Zhivotovsky B (2008) An increase in intracellular Ca2+ is required for the activation of mitochondrial calpain to release AIF during cell death. Cell Death Differ 15:1857-1864.
  • Novo N, Romero-Tamayo S, Marcuello C, Boneta S, Blasco-Machin I, Velázquez-Campoy A, Villanueva R, Moreno-Loshuertos R, Lostao A, Medina M et al (2022) Beyond a platform protein for the degradosome assembly: The Apoptosis-Inducing Factor as an efficient nuclease involved in chromatinolysis. PNAS Nexus 2:pgac312.
  • O’Sullivan J, Tedim Ferreira M, Gagné JP, Sharma AK, Hendzel MJ, Masson JY and Poirier GG (2019) Emerging roles of eraser enzymes in the dynamic control of protein ADP-ribosylation. Nat Commun 10:1182.
  • Otera H, Ohsakaya S, Nagaura Z-I, Ishihara N and Mihara K (2005) Export of mitochondrial AIF in response to proapoptotic stimuli depends on processing at the intermembrane space. EMBO J 24:1375-1386.
  • Palazzo L, Leidecker O, Prokhorova E, Dauben H, Matic I and Ahel I (2018) Serine is the major residue for ADP-ribosylation upon DNA damage. Elife 7:e34334.
  • Palazzo L, Thomas B, Jemth A-S, Colby TD, Leidecker O, Feijs KLH, Žaja R, Loseva OI, Puigvert JC, Matic I et al (2015) Processing of protein ADP-ribosylation by Nudix hydrolases. Biochem J 468 2:293-301.
  • Pandey N and Black BE (2021) Rapid detection and signaling of DNA damage by PARP-1. Trends Biochem Sci 46:744-757.
  • Park H, Kam TI, Dawson TM and Dawson VL (2020) Poly (ADP-ribose) (PAR)-dependent cell death in neurodegenerative diseases. Int Rev Cell Mol Biol 353:1-29.
  • Park H, Kam TI, Peng H, Chou SC, Mehrabani-Tabari AA, Song JJ, Yin X, Karuppagounder SS, Umanah GK, Rao AVS et al (2022) PAAN/MIF nuclease inhibition prevents neurodegeneration in Parkinson’s disease. Cell 185:1943-1959e1921.
  • Pascal JM (2023) PARP-nucleic acid interactions: Allosteric signaling, PARP inhibitor types, DNA bridges, and viral RNA surveillance. Curr Opin Struct Biol 81:102643.
  • Perraud AL, Fleig A, Dunn CA, Bagley LA, Launay P, Schmitz C, Stokes AJ, Zhu Q, Bessman MJ, Penner R et al (2001) ADP-ribose gating of the calcium-permeable LTRPC2 channel revealed by Nudix motif homology. Nature 411:595-599.
  • Perraud AL, Takanishi CL, Shen B, Kang S, Smith MK, Schmitz C, Knowles HM, Ferraris D, Li W, Zhang J et al (2005) Accumulation of free ADP-ribose from mitochondria mediates oxidative stress-induced gating of TRPM2 cation channels. J Biol Chem 280:6138-6148.
  • Polster BM, Basañez G, Etxebarria A, Hardwick JM and Nicholls DG (2005) Calpain I induces cleavage and release of apoptosis-inducing factor from isolated mitochondria. J Biol Chem 280:6447-6454.
  • Rack JGM, Liu Q, Zorzini V, Voorneveld J, Ariza A, Honarmand Ebrahimi K, Reber JM, Krassnig SC, Ahel D, van der Marel GA et al (2021) Mechanistic insights into the three steps of poly(ADP-ribosylation) reversal. Nat Commun 12:4581.
  • Rack JGM, Palazzo L and Ahel I (2020) (ADP-ribosyl)hydrolases: Structure, function, and biology. Genes Dev 34:263-284.
  • Ray Chaudhuri A and Nussenzweig A (2017) The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol 18:610-621.
  • Regdon Z, Robaszkiewicz A, Kovács K, Rygielska Ż, Hegedűs C, Bodoor K, Szabó É and Virág L (2019) LPS protects macrophages from AIF-independent parthanatos by downregulation of PARP1 expression, induction of SOD2 expression, and a metabolic shift to aerobic glycolysis. Free Radic Biol Med 131:184-196.
  • Santofimia-Castaño P, Huang C, Liu X, Xia Y, Audebert S, Camoin L, Peng L, Lomberk G, Urrutia R, Soubeyran P et al (2022) NUPR1 protects against hyperPARylation-dependent cell death. Commun Biol 5:732.
  • Shimizu T, Macey TA, Quillinan N, Klawitter J, Perraud AL, Traystman RJ and Herson PS (2013) Androgen and PARP-1 regulation of TRPM2 channels after ischemic injury. J Cereb Blood Flow Metab 33:1549-1555.
  • Sun Y, Sukumaran P, Selvaraj S, Cilz NI, Schaar A, Lei S and Singh BB (2018) TRPM2 Promotes neurotoxin MPP(+)/MPTP-induced cell death. Mol Neurobiol 55:409-420.
  • Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M et al (1999) Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397:441-446.
  • Suskiewicz MJ, Prokhorova E, Rack JGM and Ahel I (2023) ADP-ribosylation from molecular mechanisms to therapeutic implications. Cell 186:4475-4495.
  • Szczesny B, Brunyanszki A, Olah G, Mitra S and Szabo C (2014) Opposing roles of mitochondrial and nuclear PARP1 in the regulation of mitochondrial and nuclear DNA integrity: Implications for the regulation of mitochondrial function. Nucleic Acids Res 42:13161-13173.
  • Szollosi A (2021) Two decades of evolution of our understanding of the Transient Receptor Potential Melastatin 2 (TRPM2) Cation Channel. Life 11:397.
  • Tang JB, Goellner EM, Wang XH, Trivedi RN, St Croix CM, Jelezcova E, Svilar D, Brown AR and Sobol RW (2010) Bioenergetic metabolites regulate base excision repair-dependent cell death in response to DNA damage. Mol Cancer Res 8:67-79.
  • Vahsen N, Candé C, Brière J-J, Bénit P, Joza N, Larochette N, Mastroberardino PG, Pequignot MO, Casares N, Lazar V et al (2004) AIF deficiency compromises oxidative phosphorylation. EMBO J 23:4679-4689.
  • Vosler PS, Sun D, Wang S, Gao Y, Kintner DB, Signore AP, Cao G and Chen J (2009) Calcium dysregulation induces apoptosis-inducing factor release: Cross-talk between PARP-1- and calpain-signaling pathways. Exp Neurol 218:213-220.
  • Wang H, Shimoji M, Yu S-W, Dawson TM and Dawson VL (2003) Apoptosis inducing factor and PARP-Mediated injury in the MPTP mouse model of Parkinson’s disease. Ann N Y Acad Sci 991:132-139.
  • Wang Y, An R, Umanah GK, Park H, Nambiar K, Eacker SM, Kim B, Bao L, Harraz MM, Chang C et al (2016) A nuclease that mediates cell death induced by DNA damage and poly(ADP-ribose) polymerase-1. Science 354:aad6872.
  • Wang Y, Kim NS, Haince JF, Kang HC, David KK, Andrabi SA, Poirier GG, Dawson VL and Dawson TM (2011) Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos), Sci Signal 4:ra20.
  • Wang Y, Kim NS, Li X, Greer PA, Koehler RC, Dawson VL and Dawson TM (2009) Calpain activation is not required for AIF translocation in PARP-1-dependent cell death (parthanatos). J Neurochem 110:687-696.
  • Wehage E, Eisfeld J, Heiner I, Jüngling E, Zitt C and Lückhoff A (2002) Activation of the cation channel long transient receptor potential channel 2 (LTRPC2) by hydrogen peroxide. A splice variant reveals a mode of activation independent of ADP-ribose. J Biol Chem 277:23150-23156.
  • Yamamoto H, Uchigata Y and Okamoto H (1981) Streptozotocin and alloxan induce DNA strand breaks and poly(ADP-ribose) synthetase in pancreatic islets. Nature 294:284-286.
  • Yang H, Yang T, Baur JA, Perez E, Matsui T, Carmona JJ, Lamming DW, Souza-Pinto NC, Bohr VA, Rosenzweig A et al (2007) Nutrient-sensitive mitochondrial NAD+ levels dictate cell survival. Cell 130:1095-1107.
  • Yang KT, Chang WL, Yang PC, Chien CL, Lai MS, Su MJ and Wu ML (2006) Activation of the transient receptor potential M2 channel and poly(ADP-ribose) polymerase is involved in oxidative stress-induced cardiomyocyte death. Cell Death Differ 13:1815-1826.
  • Yang M, Wang C, Zhou M, Bao L, Wang Y, Kumar A, Xing C, Luo W and Wang Y (2022) KDM6B promotes PARthanatos via suppression of O6-methylguanine DNA methyltransferase repair and sustained checkpoint response. Nucleic Acids Res 50:6313-6331.
  • Ying W, Alano CC, Garnier P and Swanson RA (2005) NAD+ as a metabolic link between DNA damage and cell death. J Neurosci Res 79:216-223.
  • Yu S-W, Andrabi SA, Wang H, Kim NS, Poirier GG, Dawson TM and Dawson VL (2006) Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci U S A 103:18314-18319.
  • Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ, Poirier GG, Dawson TM and Dawson VL (2002) Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 297:259-263.
  • Yu SW, Wang Y, Frydenlund DS, Ottersen OP, Dawson VL and Dawson TM (2009) Outer mitochondrial membrane localization of apoptosis-inducing factor: Mechanistic implications for release. ASN Neuro 1:e00021.
  • Zhang F, Xie R, Munoz FM, Lau SS and Monks TJ (2014) PARP-1 hyperactivation and reciprocal elevations in intracellular Ca2+ during ROS-induced nonapoptotic cell death. Toxicol Sci 140:118-134.
  • Zhong H, Song R, Pang Q, Liu Y, Zhuang J, Chen Y, Hu J, Hu J, Liu Y, Liu Z et al (2018) Propofol inhibits parthanatos via ROS-ER-calcium-mitochondria signal pathway in vivo and vitro. Cell Death Dis 9:932.
  • Zhou J, Ng S, Huang Q, Wu Y-T, Li Z, Yao SQ and Shen H-M (2013) AMPK mediates a pro-survival autophagy downstream of PARP-1 activation in response to DNA alkylating agents. FEBS Letters 587:170-177.
  • Zhou Y, Feng X and Koh DW (2011) Activation of cell death mediated by apoptosis-inducing factor due to the absence of poly(ADP-ribose) glycohydrolase. Biochemistry 50:2850-2859.
  • Zong W-X, Ditsworth D, Bauer DE, Wang Z-Q and Thompson CB (2004) Alkylating DNA damage stimulates a regulated form of necrotic cell death. Genes Dev 18:1272-1282.

Edited by

Associate Editor:

Mercedes Rodriguez Teja

Publication Dates

  • Publication in this collection
    30 Aug 2024
  • Date of issue
    2024

History

  • Received
    17 Dec 2023
  • Accepted
    16 June 2024
Sociedade Brasileira de Genética Rua Cap. Adelmio Norberto da Silva, 736, 14025-670 Ribeirão Preto SP Brazil, Tel.: (55 16) 3911-4130 / Fax.: (55 16) 3621-3552 - Ribeirão Preto - SP - Brazil
E-mail: editor@gmb.org.br