ADP-ribosylation

Last updated

ADP-ribose ADP ribose.svg
ADP-ribose

ADP-ribosylation is the addition of one or more ADP-ribose moieties to a protein. [1] [2] It is a reversible post-translational modification that is involved in many cellular processes, including cell signaling, DNA repair, gene regulation and apoptosis. [3] [4] Improper ADP-ribosylation has been implicated in some forms of cancer. [5] It is also the basis for the toxicity of bacterial compounds such as cholera toxin, diphtheria toxin, and others. [6]

Contents

History

The first suggestion of ADP-ribosylation surfaced during the early 1960s. At this time, Pierre Chambon and coworkers observed the incorporation of ATP into hen liver nuclei extract. [7] After extensive studies on the acid insoluble fraction, several different research laboratories were able to identify ADP-ribose, derived from NAD+, as the incorporated group. Several years later, the enzymes responsible for this incorporation were identified and given the name poly(ADP-ribose)polymerase. Originally, this group was thought to be a linear sequence of ADP-ribose units covalently bonded through a ribose glycosidic bond. It was later reported that branching can occur every 20 to 30 ADP residues. [8]

The first appearance of mono(ADP-ribosyl)ation occurred a year later during a study of toxins: the diphtheria toxin of Corynebacterium diphtheriae was shown to be dependent on NAD+ in order for it to be completely effective, [9] leading to the discovery of enzymatic conjugation of a single ADP-ribose group by mono(ADP-ribosyl)transferase.

It was initially thought that ADP-ribosylation was a post translational modification involved solely in gene regulation. However, as more enzymes with the ability to ADP-ribosylate proteins were discovered, the multifunctional nature of ADP-ribosylation became apparent. The first mammalian enzyme with poly(ADP-ribose)transferase activity was discovered during the late 1980s. For the next 15 years, it was thought to be the only enzyme capable of adding a chain of ADP-ribose in mammalian cells. [10] During the late 1980s, ADP-ribosyl cyclases, which catalyze the addition of cyclic-ADP-ribose groups to proteins, were discovered. Finally, sirtuins, a family of enzymes that also possess NAD+-dependent deacylation activity, were discovered to also possess mono(ADP-ribosyl)transferase activity. [11] [12]

Catalytic mechanism

Mechanism for ADP-ribosylation, with residues of the catalyzing enzyme shown in blue. ADP-Ribosylation-Mechanism.png
Mechanism for ADP-ribosylation, with residues of the catalyzing enzyme shown in blue.

The source of ADP-ribose for most enzymes that perform this modification is the redox cofactor NAD+. In this transfer reaction, the N-glycosidic bond of NAD+ that bridges the ADP-ribose molecule and the nicotinamide group is cleaved, followed by nucleophilic attack by the target amino acid side chain. (ADP-ribosyl)transferases can perform two types of modifications: mono(ADP-ribosyl)ation and poly(ADP-ribosyl)ation.

Mono(ADP-ribosyl)ation

Mono(ADP-ribosyl)transferases commonly catalyze the addition of ADP-ribose to arginine side chains using a highly conserved R-S-EXE motif of the enzyme. [13] The reaction proceeds by breaking the bond between nicotinamide and ribose to form an oxonium ion. Next, the arginine side chain of the target protein then acts a nucleophile, attacking the electrophilic carbon adjacent to the oxonium ion. In order for this step to occur, the arginine nucleophile is deprotonated by a glutamate residue on the catalyzing enzyme[ disputed ]. Another conserved glutamate residue forms a hydrogen bond with one of the hydroxyl groups on the ribose chain to further facilitate this nucleophilic attack. As a result of the cleavage reaction, nicotinamide is released. The modification can be reversed by (ADP-ribosyl)hydrolases, which cleave the N-glycosidic bond between arginine and ribose to release ADP-ribose and unmodified protein; NAD+ is not restored by the reverse reaction.

Poly(ADP-ribosyl)ation

Poly(ADP-ribose)polymerases (PARPs) are found mostly in eukaryotes and catalyze the transfer of multiple ADP-ribose molecules to target proteins. As with mono(ADP-ribosyl)ation, the source of ADP-ribose is NAD+. PARPs use a catalytic triad of His-Tyr-Glu to facilitate binding of NAD+ and positioning of the end of the existing poly(ADP-ribose) chain on the target protein; the Glu facilitates catalysis and formation of a (1''→2') O-glycosidic linkage between two ribose molecules. There are several other enzymes that recognize poly(ADP-ribose) chains, hydrolyse them or form branches; over 800 proteins have been annotated to contain the loosely defined poly(ADP-ribose) binding motif; therefore, in addition to this modification altering target protein conformation and structure, it may also be used as a tag to recruit other proteins or for regulation of the target protein. [14]

Amino acid specificity

Many different amino acid side chains have been described as ADP-ribose acceptors. From a chemical perspective, this modification represents protein glycosylation: the transfer of ADP-ribose occurs onto amino acid side chains with a nucleophilic oxygen, nitrogen, or sulfur, resulting in N-, O-, or S-glycosidic linkage to the ribose of the ADP-ribose. [15] Originally, acidic amino acids (glutamate and aspartate) were described as the main sites of ADP-ribosylation. However, many other ADP-ribose acceptor sites such as serine, [16] [17] arginine, [18] cysteine, [19] lysine, [20] diphthamide, [21] phosphoserine, [22] and asparagine [23] have been identified in subsequent works.

Function

Apoptosis

During DNA damage or cellular stress PARPs are activated, leading to an increase in the amount of poly(ADP-ribose) and a decrease in the amount of NAD+. [24] For over a decade it was thought that PARP1 was the only poly(ADP-ribose)polymerase in mammalian cells, therefore this enzyme has been the most studied. Caspases are a family of cysteine proteases that are known to play an essential role in programmed cell death. This protease cleaves PARP-1 into two fragments, leaving it completely inactive, to limit poly(ADP-ribose) production. One of its fragments migrates from the nucleus to the cytoplasm and is thought to become a target of autoimmunity.

During caspase-independent apoptosis, also called parthanatos, poly(ADP-ribose) accumulation can occur due to activation of PARPs or inactivation of poly(ADP-ribose)glycohydrolase, an enzyme that hydrolyses poly(ADP-ribose) to produce free ADP-ribose. Studies have shown poly(ADP-ribose) drives the translocation of the apoptosis inducing factor protein to the nucleus where it will mediate DNA fragmentation. It has been suggested that if a failure of caspase activation under stress conditions were to occur, necroptosis would take place. Overactivation of PARPs has led to a necrotic cell death regulated by the tumor necrosis factor protein. Though the mechanism is not yet understood, PARP inhibitors have been shown to affect necroptosis. [25]

Gene regulation

ADP-ribosylation can affect gene expression at nearly every level of regulation, including chromatin organization, transcription factor recruitment and binding, and mRNA processing.

The organization of nucleosomes is key to regulation of gene expression: the spacing and organization of nucleosomes changes what regions of DNA are available for transcription machinery to bind and transcribe DNA. PARP1, a poly-ADP ribose polymerase, has been shown to affect chromatin structure and promote changes in the organization of nucleosomes through modification of histones.

Crystal structure of PARP1 zinc finger domain bound to DNA (purple). PDB: 4AV1 PARP1 with DNA.png
Crystal structure of PARP1 zinc finger domain bound to DNA (purple). PDB: 4AV1

PARPs have been shown to affect transcription factor structure and cause recruitment of many transcription factors to form complexes at DNA and elicit transcription. Mono(ADP-ribosyl)transferases are also shown to affect transcription factor binding at promoters. For example, PARP14, a mono (ADP-ribosyl)transferase, has been shown to affect STAT transcription factor binding.

Other (ADP-ribosyl)transferases have been shown to modify proteins that bind mRNA, which can cause silencing of that gene transcript. [26]

DNA repair

Poly(ADP-ribose)polymerases (PARPs) can function in DNA repair of single strand breaks as well as double strand breaks. In single-strand break repair (base excision repair) the PARP can either facilitate removal of an oxidized sugar or strand cleavage. PARP1 binds the single-strand breaks and pulls any nearby base excision repair intermediates close. These intermediates include XRCC1 and APLF and they can be recruited directly or through the PBZ domain of the APLF. [27] This leads to the synthesis of poly(ADP-ribose). The PBZ domain is present in many proteins involved in DNA repair and allows for the binding of the PARP and thus ADP-ribosylation which recruits repair factors to interact at the break site. PARP2 is a secondary responder to DNA damage but serves to provide functional redundancy in DNA repair. [28]

DNA repair facilitated by PARP1 recruitment of repair enzymes. Repair of a single strand break in DNA is initiated by the binding of PARP1. PARP1 binds single-strand breaks and pulls base excision repair intermediates close, leading to the synthesis of poly(ADP-ribose). XRCC1 is the X-ray repair cross complementing protein 1. XRCC1 complexes with polynucleotide kinase (PNK) which processes DNA termini. PCNA is the proliferating cell nuclear antigen that serves as a DNA clamp that aids in the DNA polymerase activity (DNA pol) FEN1 (Flap endonuclease 1) is then recruited to remove the overhanging 5' flap. The last step of DNA repair involves DNA ligase which brings the final DNA strands together in a phosphodiester bond. WikiDiagram1.pdf
DNA repair facilitated by PARP1 recruitment of repair enzymes. Repair of a single strand break in DNA is initiated by the binding of PARP1. PARP1 binds single-strand breaks and pulls base excision repair intermediates close, leading to the synthesis of poly(ADP-ribose). XRCC1 is the X-ray repair cross complementing protein 1. XRCC1 complexes with polynucleotide kinase (PNK) which processes DNA termini. PCNA is the proliferating cell nuclear antigen that serves as a DNA clamp that aids in the DNA polymerase activity (DNA pol) FEN1 (Flap endonuclease 1) is then recruited to remove the overhanging 5' flap. The last step of DNA repair involves DNA ligase which brings the final DNA strands together in a phosphodiester bond.

There are many mechanisms for the repair of damaged double stranded DNA. PARP1 may function as a synapsis factor in alternative non-homologous end joining. Additionally, it has been proposed that PARP1 is required to slow replication forks following DNA damage and promotes homologous recombination at replication forks that may be dysfunctional. It is possible that PARP1 and PARP3 work together in repair of double-stranded DNA and it has been shown that PARP3 is critical for double-stranded break resolution. There are two hypotheses by which PARP1 and PARP3 coincide. The first hypothesis states that the two (ADP-ribosyl)transferases serve to function for each other's inactivity. If PARP3 is lost, this results in single-strand breaks, and thus the recruitment of PARP1. A second hypothesis suggests that the two enzyme work together; PARP3 catalyzes mono(ADP-ribosyl)ation and short poly(ADP-ribosyl)ation and serves to activate PARP1. [28]

The PARPs have many protein targets at the site of DNA damage. KU protein and DNA-PKcs are both double-stranded break repair components with unknown sites of ADP-ribosylation. Histones are another protein target of the PARPs. All core histones and linker histone H1 are ADP-ribosylated following DNA damage. The function of these modifications is still unknown, but it has been proposed that ADP-ribosylation modulates higher-order chromatin structure in efforts to facilitate more accessible sites for repair factors to migrate to the DNA damage.

Protein degradation

The ubiquitin-proteasome system (UPS) figures prominently in protein degradation. The 26S proteasome consists of a catalytic subunit (the 20S core particle), and a regulatory subunit (the 19S cap). [29] Poly-ubiquitin chains tag proteins for degradation by the proteasome, which causes hydrolysis of tagged proteins into smaller peptides.

Physiologically, PI31 attacks 20S catalytic domain of 26S Proteasome that results in decreased proteasome activity. (ADP-ribosyl)transferase Tankyrase (TNKS) causes ADP-ribosylation of PI31 which in turn increases the proteasome activity. Inhibition of TNKs further shows the reduced 26S Proteasome assembly. Therefore, ADP-ribosylation promotes 26S Proteasome activity in both Drosophila and human cells. [30]

Enzyme regulation

The activity of some enzymes is regulated by ADP-ribosylation. For instance, the activity of Rodospirillum rubrum di-nitrogenase-reductase is turned off by ADP-ribosylation of an arginine residue, and reactivated by the removal of the ADP-ribosyl group. [31]

Clinical significance

Cancer

PARP1 is involved in base excision repair (BER), single- and double-strand break repair, and chromosomal stability. It is also involved in transcriptional regulation through its facilitation of protein–protein interactions. PARP1 uses NAD+ in order to perform its function in apoptosis. If a PARP becomes overactive the cell will have decreased levels of NAD+ cofactor as well as decreased levels of ATP and thus will undergo necrosis. This is important in carcinogenesis because it could lead to the selection of PARP1 deficient cells (but not depleted) due to their survival advantage during cancer growth. [32]

Susceptibility to carcinogenesis under PARP1 deficiency depends significantly on the type of DNA damage incurred. There are many implications that various PARPs are involved in preventing carcinogenesis. As stated previously, PARP1 and PARP2 are involved in BER and chromosomal stability. PARP3 is involved in centrosome regulation. Tankyrase is another (ADP-ribosyl)polymerase that is involved in telomere length regulation. [5]

PARP1 inhibition has also been widely studied in anticancer therapeutics. The mechanism of action of a PARP1 inhibitor is to enhance the damage done by chemotherapy on the cancerous DNA by disallowing the reparative function of PARP1 in BRCA1/2 deficient individuals .

PARP14 is another ADP-ribosylating enzyme that has been well-studied in regards to cancer therapy targets; it is a signal transducer and activator of STAT6 transcription-interacting protein, and was shown to be associated with the aggressiveness of B-cell lymphomas. [32]

Bacterial toxins

Bacterial ADP-ribosylating exotoxins (bAREs) covalently transfer an ADP-ribose moiety of NAD+ to target proteins of infected eukaryotes, to yield nicotinamide and a free hydrogen ion. bAREs are produced as enzyme precursors, consisting of a "A" and "B" domains: the "A" domain is responsible for ADP-ribosylation activity; and, the "B" domain for translocation of the enzyme across the membrane of the cell. These domains can exist in concert in three forms: first, as single polypeptide chains with A and B domains covalently linked; second, in multi-protein complexes with A and B domains bound by non-covalent interactions; and, third, in multi-protein complexes with A and B domains not directly interacting, prior to processing. [6]

Crystal structure of the diphtheria toxin. PDB: 1MDT Diphtheria tox.png
Crystal structure of the diphtheria toxin. PDB: 1MDT

Upon activation, bAREs ADP-ribosylate any number of eukaryotic proteins; such mechanism is crucial to the instigation of the diseased states associated with ADP-ribosylation. GTP-binding proteins, in particular, are well-established in bAREs pathophysiology. For examples, cholera and heat-labile enterotoxin target the α-subunit of Gs of heterotrimeric GTP-binding proteins. As the α-subunit is ADP-ribosylated, it is permanently in an "active", GTP-bound state; subsequent activation of intracellular cyclic AMP stimulates the release of fluid and ions from intestinal epithelial cells. Furthermore, C. Botulinum C3 ADP-ribosylates GTP-binding proteins Rho and Ras, and Pertussis toxin ADP-ribosylates Gi, Go, and Gt. Diphtheria toxin ADP-ribosylates ribosomal elongation factor EF-2, which attenuates protein synthesis. [6]

There are a variety of bacteria which employ bAREs in infection: CARDS toxin of Mycoplasma pneumoniae , cholera toxin of Vibrio cholerae ; heat-labile enterotoxin of E. coli ; exotoxin A of Pseudomonas aeruginosa ; pertussis toxin of B. pertussis ; C3 toxin of C. botulinum ; and diphtheria toxin of Corynebacterium diphtheriae . [33]

See also

Related Research Articles

<span class="mw-page-title-main">Nicotinamide adenine dinucleotide</span> Chemical compound which is reduced and oxidized

Nicotinamide adenine dinucleotide (NAD) is a coenzyme central to metabolism. Found in all living cells, NAD is called a dinucleotide because it consists of two nucleotides joined through their phosphate groups. One nucleotide contains an adenine nucleobase and the other, nicotinamide. NAD exists in two forms: an oxidized and reduced form, abbreviated as NAD+ and NADH (H for hydrogen), respectively.

<span class="mw-page-title-main">Poly (ADP-ribose) polymerase</span> Family of proteins

Poly (ADP-ribose) polymerase (PARP) is a family of proteins involved in a number of cellular processes such as DNA repair, genomic stability, and programmed cell death.

<span class="mw-page-title-main">Histone-modifying enzymes</span> Type of enzymes

Histone-modifying enzymes are enzymes involved in the modification of histone substrates after protein translation and affect cellular processes including gene expression. To safely store the eukaryotic genome, DNA is wrapped around four core histone proteins, which then join to form nucleosomes. These nucleosomes further fold together into highly condensed chromatin, which renders the organism's genetic material far less accessible to the factors required for gene transcription, DNA replication, recombination and repair. Subsequently, eukaryotic organisms have developed intricate mechanisms to overcome this repressive barrier imposed by the chromatin through histone modification, a type of post-translational modification which typically involves covalently attaching certain groups to histone residues. Once added to the histone, these groups elicit either a loose and open histone conformation, euchromatin, or a tight and closed histone conformation, heterochromatin. Euchromatin marks active transcription and gene expression, as the light packing of histones in this way allows entry for proteins involved in the transcription process. As such, the tightly packed heterochromatin marks the absence of current gene expression.

<span class="mw-page-title-main">ETS1</span> Protein-coding gene in the species Homo sapiens

Protein C-ets-1 is a protein that in humans is encoded by the ETS1 gene. The protein encoded by this gene belongs to the ETS family of transcription factors.

<span class="mw-page-title-main">PARP1</span> Mammalian protein found in Homo sapiens

Poly [ADP-ribose] polymerase 1 (PARP-1) also known as NAD+ ADP-ribosyltransferase 1 or poly[ADP-ribose] synthase 1 is an enzyme that in humans is encoded by the PARP1 gene. It is the most abundant of the PARP family of enzymes, accounting for 90% of the NAD+ used by the family. PARP1 is mostly present in cell nucleus, but cytosolic fraction of this protein was also reported.

<span class="mw-page-title-main">NAD(P)(+)—protein-arginine ADP-ribosyltransferase</span>

In enzymology, a NAD(P)+-protein-arginine ADP-ribosyltransferase (EC 2.4.2.31) is an enzyme that catalyzes the chemical reaction using nicotinamide adenine dinucleotide

In enzymology, a NAD+-diphthamide ADP-ribosyltransferase (EC 2.4.2.36) is an enzyme that catalyzes the chemical reaction

<span class="mw-page-title-main">Tankyrase</span> Enzyme

Tankyrase, also known as tankyrase 1, is an enzyme that in humans is encoded by the TNKS gene. It inhibits the binding of TERF1 to telomeric DNA. Tankyrase attracts substantial interest in cancer research through its interaction with AXIN1 and AXIN2, which are negative regulators of pro-oncogenic β-catenin signaling. Importantly, activity in the β-catenin destruction complex can be increased by tankyrase inhibitors and thus such inhibitors are a potential therapeutic option to reduce the growth of β-catenin-dependent cancers.

<span class="mw-page-title-main">NMNAT1</span> Protein-coding gene in the species Homo sapiens

Nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) is an enzyme that in humans is encoded by the nmnat1 gene. It is a member of the nicotinamide-nucleotide adenylyltransferases (NMNATs) which catalyze nicotinamide adenine dinucleotide (NAD) synthesis.

<span class="mw-page-title-main">PARP4</span> Enzyme

Poly [ADP-ribose] polymerase 4 is an enzyme that in humans is encoded by the PARP4 gene.

<span class="mw-page-title-main">PARP3</span> Protein-coding gene in the species Homo sapiens

Poly [ADP-ribose] polymerase 3 is an enzyme that in humans is encoded by the PARP3 gene.

<span class="mw-page-title-main">PARP2</span> Protein-coding gene in the species Homo sapiens

Poly [ADP-ribose] polymerase 2 is an enzyme that in humans is encoded by the PARP2 gene. It is one of the PARP family of enzymes.

<span class="mw-page-title-main">ADP-ribosylhydrolase 3</span> Protein-coding gene in the species Homo sapiens

(ADP-ribosyl)hydrolase 3 (ARH3) is an enzyme that in humans is encoded by the ADPRHL2 gene (also called ADPRS). This enzyme reverses the proteins’ post-translational addition of ADP-ribose to serine residues as part of the DNA damage response The enzyme is also known to cleave poly(ADP-ribose) polymers, 1''-O-acetyl-ADP-ribose and alpha-NAD+

<span class="mw-page-title-main">Sirtuin 6</span> Protein-coding gene in the species Homo sapiens

Sirtuin 6 is a stress responsive protein deacetylase and mono-ADP ribosyltransferase enzyme encoded by the SIRT6 gene. In laboratory research, SIRT6 appears to function in multiple molecular pathways related to aging, including DNA repair, telomere maintenance, glycolysis and inflammation. SIRT6 is member of the mammalian sirtuin family of proteins, which are homologs to the yeast Sir2 protein.

<span class="mw-page-title-main">PARP inhibitor</span> Pharmacological enzyme inhibitors of poly (ADP-ribose) polymerases

PARP inhibitors are a group of pharmacological inhibitors of the enzyme poly ADP ribose polymerase (PARP).

<span class="mw-page-title-main">ADP-ribosylhydrolase</span>

In molecular biology, the (ADP-ribosyl)hydrolase (ARH) family contains enzymes which catalyses the hydrolysis of ADP-ribosyl modifications from proteins, nucleic acids and small molecules.

<span class="mw-page-title-main">Macro domain</span>

In molecular biology, the Macro domain or A1pp domain is a module of about 180 amino acids which can bind ADP-ribose, an NAD metabolite, or related ligands. Binding to ADP-ribose can be either covalent or non-covalent: in certain cases it is believed to bind non-covalently, while in other cases it appears to bind both non-covalently through a zinc finger motif, and covalently through a separate region of the protein.

<span class="mw-page-title-main">Clostridium botulinum C3 toxin</span>

Clostridium botulinum C3 exoenzyme is a toxin that causes the addition of one or more ADP-ribose moieties to Rho-like proteins. Many bacterial toxins nucleotide-binding modify by ADP-ribosylation proteins involved in essential cell functions, leading to their toxic effects.

<span class="mw-page-title-main">ADP-ribosylhydrolase 1</span>

(ADP-ribosyl)hydrolase 1, also termed [Protein ADP-ribosylarginine] hydrolase and protein-Nω-(ADP-D-ribosyl)-L-arginine ADP-ribosylhydrolase (EC 3.2.2.19), is an enzyme that in humans is encoded by the ADPRH gene. This enzyme is a specific mono(ADP-ribosyl)hydrolase that catalyses the removal of an ADP-ribosyl modification from target arginine residues of protein substrates. The chemical reactions can formally be described as follows:

Parthanatos is a form of programmed cell death that is distinct from other cell death processes such as necrosis and apoptosis. While necrosis is caused by acute cell injury resulting in traumatic cell death and apoptosis is a highly controlled process signalled by apoptotic intracellular signals, parthanatos is caused by the accumulation of Poly(ADP ribose) (PAR) and the nuclear translocation of apoptosis-inducing factor (AIF) from mitochondria. Parthanatos is also known as PARP-1 dependent cell death. PARP-1 mediates parthanatos when it is over-activated in response to extreme genomic stress and synthesizes PAR which causes nuclear translocation of AIF. Parthanatos is involved in diseases that afflict hundreds of millions of people worldwide. Well known diseases involving parthanatos include Parkinson's disease, stroke, heart attack, and diabetes. It also has potential use as a treatment for ameliorating disease and various medical conditions such as diabetes and obesity.

References

  1. Belenky P, Bogan KL, Brenner C (2007). "NAD+ metabolism in health and disease" (PDF). Trends Biochem. Sci. 32 (1): 12–9. doi:10.1016/j.tibs.2006.11.006. PMID   17161604.
  2. Ziegler M (2000). "New functions of a long-known molecule. Emerging roles of NAD in cellular signaling". Eur. J. Biochem. 267 (6): 1550–64. doi: 10.1046/j.1432-1327.2000.01187.x . PMID   10712584.
  3. Berger F, Ramírez-Hernández MH, Ziegler M (2004). "The new life of a centenarian: signalling functions of NAD(P)". Trends Biochem. Sci. 29 (3): 111–8. doi:10.1016/j.tibs.2004.01.007. PMID   15003268. S2CID   8820773.
  4. Corda D, Di Girolamo M (2003). "NEW EMBO MEMBER'S REVIEW: Functional aspects of protein mono-ADP-ribosylation". EMBO J. 22 (9): 1953–8. doi:10.1093/emboj/cdg209. PMC   156081 . PMID   12727863.
  5. 1 2 Scarpa ES, Fabrizio G, Di Girolamo M (2013). ". A role in intracellular mono-ADP-ribosylation in cancer biology". FEBS Journal. 280 (15): 3551–3562. doi: 10.1111/febs.12290 . PMID   23590234.
  6. 1 2 3 Krueger, KM; Barbieri, JT (January 1995). "The family of bacterial ADP-ribosylating exotoxins". Clinical Microbiology Reviews. 8 (1): 34–47. doi:10.1128/CMR.8.1.34. PMC   172848 . PMID   7704894.
  7. Chambon, P; Weill, J. D.; Mandel, P. (1963). "Nicotinamide mononucleotide activation of new DNA-dependent polyadenylic acid synthesizing nuclear enzyme". Biochem. Biophys. Res. Commun. 11: 39–43. doi:10.1016/0006-291x(63)90024-x. PMID   14019961.
  8. Hayaishi, O.; Ueda, K. (2012). Poly- and Mono(ADP-ribosyl)ation Reactions: Their Significance in Molecular Biology. In ADP-Ribosylation Reactions: Biology and Medicine. New York: Academic Press.
  9. Collier RJ, Pappenheimer, Jr AM (1964). "STUDIES ON THE MODE OF ACTION OF DIPHTHERIA TOXIN* II. EFFECT OF TOXIN ON AMINO ACID INCORPORATION IN CELL-FREE SYSTEM". Journal of Experimental Medicine. 120 (6): 1019–1039. doi: 10.1084/jem.120.6.1019 . PMC   2137798 . PMID   14238922.
  10. Hassa, P. O.; Haenni, S. S.; Elser, M.; Hottiger, M. O. (2006). "Hassa, P. O.; Haenni, S. S.; Elser, M.; Hottiger, M. O. (2006) "Nuclear ADP-Ribozylation Reactions in Mammalian Cells: Where Are We Today and Where Are We Going". Microbiol. Mol. Biol. Rev. 70 (3): 789–829. doi:10.1128/mmbr.00040-05. PMC   1594587 . PMID   16959969.
  11. Frye, RA (June 24, 1999). "Characterization of five human cDNAs with homology to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD and may have protein ADP-ribosyltransferase activity". Biochemical and Biophysical Research Communications. 260 (1): 273–9. doi:10.1006/bbrc.1999.0897. PMID   10381378.
  12. Rack, Johannes Gregor Matthias; Morra, Rosa; Barkauskaite, Eva; Kraehenbuehl, Rolf; Ariza, Antonio; Qu, Yue; Ortmayer, Mary; Leidecker, Orsolya; Cameron, David R. (July 16, 2015). "Identification of a Class of Protein ADP-Ribosylating Sirtuins in Microbial Pathogens". Molecular Cell. 59 (2): 309–320. doi:10.1016/j.molcel.2015.06.013. ISSN   1097-4164. PMC   4518038 . PMID   26166706.
  13. Laing, Sabrina; Unger, Mandy; Koch-Nolte, Friedrich; Haag, Friedrich (July 21, 2010). "ADP-ribosylation of arginine". Amino Acids. 41 (2): 257–269. doi:10.1007/s00726-010-0676-2. PMC   3102197 . PMID   20652610.
  14. Žaja, Roko; Mikoč, Andreja; Barkauskaite, Eva; Ahel, Ivan (December 21, 2012). "Molecular Insights into Poly(ADP-ribose) Recognition and Processing". Biomolecules. 3 (1): 1–17. doi: 10.3390/biom3010001 . PMC   4030884 . PMID   24970154.
  15. Liu, Qiang; Florea, Bogdan I.; Filippov, Dmitri V. (2017). "ADP-Ribosylation Goes Normal: Serine as the Major Site of the Modification". Cell Chemical Biology. 24 (4): 431–432. doi: 10.1016/j.chembiol.2017.04.003 . PMID   28431224.
  16. Leidecker, Orsolya; Bonfiglio, Juan José; Colby, Thomas; Zhang, Qi; Atanassov, Ilian; Zaja, Roko; Palazzo, Luca; Stockum, Anna; Ahel, Ivan; Matic, Ivan (2016). "Serine is a new target residue for endogenous ADP-ribosylation on histones". Nature Chemical Biology. 12 (12): 998–1000. doi:10.1038/nchembio.2180. PMC   5113755 . PMID   27723750.
  17. Bonfiglio, Juan José; Fontana, Pietro; Zhang, Qi; Colby, Thomas; Gibbs-Seymour, Ian; Atanassov, Ilian; Bartlett, Edward; Zaja, Roko; Ahel, Ivan; Matic, Ivan (2017). "Serine ADP-Ribosylation Depends on HPF1". Molecular Cell. 65 (5): 932–940.e6. doi: 10.1016/j.molcel.2017.01.003 . PMC   5344681 . PMID   28190768.
  18. Laing S, Koch-Nolte F, Haag F, Buck F. "Strategies for the identification of arginine ADP-ribosylation sites". Journal of proteomics. 2011;75:169–176.
  19. McDonald LJ, Moss J. "Enzymatic and nonenzymatic ADP-ribosylation of cysteine". Mol Cell Biochem. 1994;138:221–226.
  20. Messner, Simon; Altmeyer, Matthias; Zhao, Hongtao; Pozivil, Andrea; Roschitzki, Bernd; Gehrig, Peter; Rutishauser, Dorothea; Huang, Danzhi; Caflisch, Amedeo; Hottiger, Michael O. (2010). "PARP1 ADP-ribosylates lysine residues of the core histone tails". Nucleic Acids Research. 38 (19): 6350–6362. doi:10.1093/nar/gkq463. PMC   2965223 . PMID   20525793.
  21. Oppenheimer NJ, Bodley JW. Diphtheria toxin. "Site and configuration of ADP-ribosylation of diphthamide in elongation factor 2". J Biol Chem. 1981;256:8579–8581.
  22. Smith JA, Stocken LA. "Chemical and metabolic properties of adenosine diphosphate ribose derivatives of nuclear proteins". Biochem J. 1975;147:523–529.
  23. Manning DR, Fraser BA, Kahn RA, Gilman AG. "ADP-ribosylation of transducin by islet-activation protein. Identification of asparagine as the site of ADP-ribosylation". J Biol Chem. 1984;259:749–756.
  24. Scovassi, AI; Denegri, M; Donzelli, M; Rossi, L; Bernardi, R; Mandarino, A; Frouin, I; Negri, C (1998). "Poly(ADP-ribose) synthesis in cells undergoing apoptosis: an attempt to face death before PARP degradation". European Journal of Histochemistry. 42 (4): 251–8. PMID   10068897.
  25. Aredia, F; Scovassi, AI (June 1, 2014). "Involvement of PARPs in cell death". Frontiers in Bioscience. 6 (2): 308–17. doi: 10.2741/707 . PMID   24896207.
  26. Ryu, Keun Woo; Kim, Dae-Seok; Kraus, W. Lee (January 9, 2015). "New Facets in the Regulation of Gene Expression by ADP-Ribosylation and Poly(ADP-ribose) Polymerases". Chemical Reviews. 115 (6): 2453–2481. doi:10.1021/cr5004248. PMC   4378458 . PMID   25575290.
  27. Schreiber, V; Amé, JC; Dollé, P; Schultz, I; Rinaldi, B; Fraulob, V; Ménissier-de Murcia, J; de Murcia, G (June 21, 2002). "Poly(ADP-ribose) polymerase-2 (PARP-2) is required for efficient base excision DNA repair in association with PARP-1 and XRCC1". The Journal of Biological Chemistry. 277 (25): 23028–36. doi: 10.1074/jbc.m202390200 . PMID   11948190.
  28. 1 2 Pears, Catherine J.; Couto, C. Anne-Marie; Wang, Hong-Yu; Borer, Christine; Kiely, Rhian; Lakin, Nicholas D. (October 28, 2014). "The role of ADP-ribosylation in regulating DNA double-strand break repair". Cell Cycle. 11 (1): 48–56. doi:10.4161/cc.11.1.18793. PMC   3272231 . PMID   22186780.
  29. Cheng, Yifan (April 2009). "Toward an atomic model of the 26S proteasome". Current Opinion in Structural Biology. 19 (2): 203–208. doi:10.1016/j.sbi.2009.02.004. PMC   2743420 . PMID   19286367.
  30. Cho-Park, Park F.; Steller, Hermann (April 25, 2013). "Proteasome regulation by ADP-ribosylation". Cell. 153 (3): 614–627. doi:10.1016/j.cell.2013.03.040. ISSN   0092-8674. PMC   3676968 . PMID   23622245.
  31. Pope, M. R.; Saari, L. L.; Ludden, P. W. (August 5, 1986). "N-glycohydrolysis of adenosine diphosphoribosyl arginine linkages by dinitrogenase reductase activating glycohydrolase (activating enzyme) from Rhodospirillum rubrum". The Journal of Biological Chemistry. 261 (22): 10104–10111. doi: 10.1016/S0021-9258(18)67497-6 . ISSN   0021-9258. PMID   3090031.
  32. 1 2 Boulares HA, Yakovlev AG, Smulson ME (2000). "Genome Degradation by DNAS1L3 Endonuclease: A Key PARP-1-Regulated Event in Apoptosis". Madame Curie Bioscience Database.
  33. Deng, Qing; Barbieri, Joseph T. (October 2008). "Molecular Mechanisms of the Cytotoxicity of ADP-Ribosylating Toxins". Annual Review of Microbiology. 62 (1): 271–288. doi:10.1146/annurev.micro.62.081307.162848. PMID   18785839.

Further reading