864
Views
6
CrossRef citations to date
0
Altmetric
Article Addendum

Helicobacter pylori activation of PARP-1

Usurping a versatile regulator of host cellular health

&
Pages 373-378 | Received 21 Jun 2010, Accepted 08 Sep 2010, Published online: 01 Nov 2010

Abstract

Chronic infection of the human stomach by Helicobacter pylori is an important risk factor for gastric cancer. H. pylori produces a cache of virulence factors that promote colonization and persistence, which, in turn, contributes to a robust inflammatory response at the host-pathogen interface. Recently, we reported that H. pylori activates the abundant nuclear regulator poly(ADP-ribose) polymerase (PARP)-1, resulting in the production of the catabolite poly(ADP-ribose) (PAR). PARP-1 is emerging as a key player in establishing homeostasis at the host-pathogen interface. In this article, we summarize the discovery of H. pylori-dependent PARP-1 activation, and discuss potential roles for PARP-1 in H. pylori-mediated gastric disease. In light of the remarkable successes that have reported for treating inflammatory disorders and cancers with PARP-1 inhibitors, we discuss the prospects of targeting PARP-1 for treatment of H. pylori-associated gastric disease.

This article refers to:

Prologue

Successful pathogens often remodel host cells and tissues to create a more suitable niche for colonization.Citation1 Active remodeling at the site of colonization brings about changes that can promote bacterial adherence, invasion and nutrient acquisition, while at the same time subverting host mechanisms for pathogen clearance. A common and effective strategy for pathogen-initiated remodeling is to specifically target key regulators of host cell function for modulation.

Helicobacter pylori, a human gastric pathogen, is an example of a bacterium that is very successful at remodeling the host-pathogen interface to promote long-term persistence.Citation2 However, persistent infection with Helicobacter pylori is a well-established risk factor for the development of serious gastric disease in humans, including stomach cancer, which is the second leading cause of cancer-related death worldwide.Citation3 Thus, there is considerable interest in identifying the molecular mechanisms that underlie gastric remodeling during chronic H. pylori infection, as well as how these changes contribute to persistence and disease.

Recently, we reported that the abundant nuclear protein, poly(ADP-ribose) polymerase (PARP)-1, becomes activated in H. pylori infected gastric epithelial cells and, moreover, that purified recombinant PARP-1 is directly activated by a protein factor present within the H. pylori culture broth after removal of intact bacterial cells (e.g., culture filtrate).Citation4 PARP-1, which is the most abundant of the PARP super-family,Citation5 is a 113-kDa protein with three well-defined functional domains: a DNA-binding domain that recognizes DNA strand breaks, a central automodification domain that is an acceptor for PAR and a NAD-binding catalytic domain.Citation6,Citation7 The catalytic domain of PARP-1 is typically activated by sensing and binding damaged DNA, which can result in a 500-fold increase over low basal levels of the synthesis of long, branched, negatively charged polymers of the metabolite poly-ADP ribose (PAR).Citation8,Citation9

In the presence of damaged DNA, PAR synthesis facilitates PARP-1 mediated base excision repair (BER), which is normally important for the long-term maintenance of genomic stability. However, under certain conditions, inappropriate PARP-1 activation, by alternative mechanisms, such as interaction with other proteins,Citation10Citation12 may promote necrotic or apoptotic cell death, as well enhanced inflammatory signaling and secondary damage to host tissues.Citation13 In this addendum, we discuss H. pylori-induced PARP-1 activation as a potential mechanism by which this pathogen might contribute to gastric remodeling by inducing inflammatory and apoptotic signaling pathways.

Discovery of the H. pylori PARP-1-activating Factor

The PARP-1 activating factor produced by H. pylori was discovered during a screen of H. pylori culture filtrates for enzymatic activities normally associated with bacterial toxins or effectors. The screen yielded a high molecular protein within the lysates of several mammalian cell lines, identified as PARP-1,Citation4 that had been covalently modified in a manner consistent with ADP-ribosylation.Citation14 Because ADP-ribosylation is an important modifying activity associated with several potent exotoxins (e.g., diphtheria and cholera toxins) and type III effectors (e.g., Pseudomonas aeruginosa ExoS and ExoT), these findings suggested that H. pylori might generate an ADP-ribosylating factor that specifically targets PARP-1 for modification.

However, further studies revealed that our initial hypothesis was incorrect. Efforts to map the ADP-ribose acceptor domain of PARP-1 indicated that all three domains of PARP-1Citation6,Citation7 were required for H. pylori-dependent modification.Citation4 Moreover, an active PARP-1 catalytic domain was required for H. pylori-dependent PARP-1 modification.Citation4 As described above, the catalytic domain of PARP-1 synthesizes large quantities of PAR in response to activation, which can result in auto-modification or trans-modification of other nuclear proteins.Citation6,Citation9 Indeed, we found that purified recombinant PARP-1 was modified with PAR in an H. pylori dependent fashion. Thus, rather than generating a toxin that ADP-ribosylates PARP-1, H. pylori produces a novel protein factor that directly activates PARP-1, resulting in the synthesis of PAR. These studies are the first to describe a bacterial factor that directly activates PARP-1.Citation4

Importantly, PAR was detected not only under highly defined conditions using recombinant proteins, but also within intact gastric epithelial cells that had been infected with H. pylori.Citation4 Intracellular PAR production was nearly eliminated in PARP-1 knockdown cells and, moreover, there was little evidence of H. pylori-dependent ADP-ribosylation within lysates prepared from parp-1-/- mouse embryonic fibroblasts.Citation4 These data strongly suggested that PARP-1 is the target for H. pylori-mediated PAR synthesis and, moreover, suggested that other members of the PARP super-family (e.g., PARP-2) are not likely to be significant contributors to H. pylori-dependent PAR synthesis.

The Relationship between Persistent H. pylori Infection, Chronic Inflammation and Apoptosis at the Epithelial Barrier

A variety of diseases, including a number of cancers, are linked to chronic inflammation.Citation15 Although both strain-specific virulence traits and host responses contribute to the risk that H. pylori infections will progress to disease, chronic H. pylori-induced gastric inflammation is the single most important risk factor for the development of stomach cancer. Subsequent to initial H. pylori infection, a well-characterized inflammatory response develops,Citation16,Citation17 which includes the secretion of pro-inflammatory cytokines by epithelialCitation17Citation20 or mucosal antigen presenting cells,Citation21 increased recruitment of neutrophils,Citation22 and an IgA response.Citation23 However, this robust inflammatory response is not protective, as H. pylori infection both promotes and limits the extent of inflammation, in part because regulatory Th cells (Tregs) are induced.Citation24Citation28 The Tregs appear to protect the host against excessive tissue damage, while at the same time promoting persistent infection,Citation28,Citation29 thereby allowing H. pylori to exist in a long-term, dynamic equilibrium with its human host. Persistent inflammation contributes directly to tissue damage and increased apoptotic cell death within the gastric mucosa, both of which are hallmarks of persistent H. pylori infection.Citation30Citation34 For a subset of individuals, the biological cost of the H. pylori-human long-term relationship is the development of one of several gastric diseases, including cancer.

The Very Talented PARP-1

PARP-1 was originally discovered as an important contributor to the maintenance of genome integrity, with functional roles in the modulation of chromatin compaction and decondensation, repair of DNA single strand breaks (base excision repair), repair of DNA double strand breaks, DNA replication and telomere homeostasis or integrity.Citation6 More recently, PARP-1 has been functionally linked to several processes central to cell division, proliferation, differentiation and death.Citation6 The normal physiological functions of PARP-1 continue to expand and the full range of PARP-1 activities has not likely been identified.Citation35

In addition to the potential roles for PARP in normal physiology and function, PARP-1 has also been associated with a surprising number of disease states. These pathologies include, but are not limited to, carcinogenesis, tumor progression and hypoxia.Citation6 As discussed below, a unifying feature is that many of these PARP-1 associated pathologies are inflammatory in nature.

PARP-1 as a Regulator of Inflammation

The importance of PARP-1 in inflammatory-mediated pathologies is accentuated by the success of using PARP-1 inhibitors in treating a number of inflammatory disorders, including ischemia/reperfusion,Citation36,Citation37 heart failure,Citation38 hypertension,Citation39 arthritis,Citation40,Citation41 atherosclerosis,Citation42 and LPS-induced inflammation.Citation43 PARP-1 inhibitors decrease the in vivo production of inflammatory cytokines and chemokines, decrease antigen-specific Th1-cell expansion and induce the production of the anti-inflammatory cytokine IL-10.Citation40 PARP-1 inhibition also reduced neutrophil recruitment and multiple organ failure in a rodent model of inflammation.Citation44 Finally, monocytes/macrophage-mediated release of TNFalpha, IL-1 and IL-6 was reduced by PARP-1 inhibition.Citation45

Causal associations between H. pylori-mediated PARP-1 activation and host inflammatory responses have not yet been explored, although potential mechanistic links do exist. For example, overactivation of PARP-1 is associated with increased activity of pro-inflammatory factors like activator protein-1 (AP-1), mitogen-activated protein (MAP) kinases (MAPKs) and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB).Citation46 At the same time, activation of AP-1, MAPK and NFκB are hallmarks of H. pylori infection, and are thought to contribute to increased inflammatory cytokine expression, an increased rate of apoptosis, an increased proliferation rate and altered cell cycle in gastric epithelial cells.Citation47

PARP-1-mediated inflammation may occur via indirect mechanisms, such as necrotic cell death that occurs as a result of NAD depletion.Citation48 However, direct mechanistic roles for PARP-1 in regulating pro-inflammatory processes at the transcriptional level have also emerged. PARP-1 can directly interact with NFκB and promote NFκB interactions with other proteins as well as DNA to promote the expression of proinflammatory cytokines as well as the activity of nitric oxide synthases.Citation49Citation51 Moreover, PARP-1/NFκB interactions also regulate cellular differentiation, proliferation and death.Citation52 PARP-1/NFκB interactions are regulated at several levels, and requires PARP-1 acetylation by a p300/cAMP response element-binding-binding protein.Citation53,Citation54 Acetylation-dependent PARP-1 trans-activation functions are balanced by the sumoylation of PARP-1. It remains to be seen which, if any, of these pathways are involved in mediating the consequences of H. pylori-dependent PARP-1 activation.

PARP-1 as a Regulator of Cell Death

While PARP-1 activation under mild genotoxic stress normally promotes cell survival, inappropriate PARP-1 activation and excessive PAR production can instead promote cell death. The importance of PARP-1-mediated cell death is accentuated by genetic evidence in some animal models that over-activation of PARP-1 contributes significantly to the cell death that accompanies stroke, diabetes, inflammation and neurodegeneration.Citation55

PARP-1 activation can induce cell death by several overall mechanisms. Necrotic cell death can result from the depletion of cellular NAD that accompanies excessive PAR production.Citation48 PARP-1 activation has also been linked to a more direct pathway of cell death called parthanatos, which is derived from “PAR” and “thanatos,” which is the Greek reference to death and mortality.Citation55Citation57 “Parthanatic” cell death shares characteristics with both apoptosis and necrosis.Citation56 However, one of the distinguishing characteristics of parthanatos is that this pathway is triggered by excessive PARP-1 activation and PAR production. A key event in parthanatic cell death is the release of the mitochondrial localized apoptosis-inducing factor (AIF) by a PAR-dependent mechanism.Citation58,Citation59 Several lines of evidence suggest that PAR and AIF directly interact,Citation60 and that both of these factors have important roles in PARP-1-dependent cell death,Citation61,Citation62 AIF is a mitochondrial-localized flavoproteinCitation63 that normally has an important role in cellular bioenergetics.Citation64,Citation65 During parthanatos, PARP-1 synthesized PAR translocates from the nucleus to the mitochondria and, by a poorly understood mechanism, promotes release of AIF to the cytosol.Citation61,Citation62 AIF in turn, localizes back to the nucleus and participates in killing of the cell.

Although it is currently unclear whether the characteristic increase in cell death within the gastric epithelium accompanying H. pylori infection involves PAR-dependent mechanisms of cell death, it is noteworthy that AIF release from mitochondria during H. pylori infection has been reported in both cultured gastric epithelial cellsCitation66 and T and B cells.Citation67 In preliminary studies,Citation4 we discovered that within H. pylori-infected gastric epithelial cells, AIF release was significantly inhibited in the presence of a PARP-1 inhibitor, suggesting a potential link between H. pylori-mediated PARP-1 activation and parthanatos.

PARP-1 as a Modulator of Host Responses to Pathogens

Contributions of PARP-1 towards the outcome of human infections with pathogenic microorganisms are only beginning to be explored. However, the importance of PARP-1 in mediating important physiological responses to cellular stress suggests that this nuclear factor could potentially have a functional role in the outcome of some host infections with pathogenic microbes. Several studies suggest that PARP-1 may have multiple and sometimes paradoxical roles in both regulating and promoting infections.

PARP-1 was recently reported to function as a transcriptional repressor of the anti-inflammatory cytokine interleukin (IL)-10Citation68 in humans with a single nucleotide polymorphism in the promoter region associated with increased susceptibility to septic shock during pneumococcal infection.Citation69 In another recent study, PARP-1 was implicated in a mouse model for enterocolitis induced by S. typhimurium.Citation70 Specifically, in response to Salmonella infection, PARP-1 deficient animals demonstrated reduced infiltration of immune cells into the gut, along with severely delayed inflammation.

PARP-1 has been demonstrated to be important for the life cycle of several viruses. PARP-1 is important for retroviral integration into the host genome,Citation71 including efficient human immunodeficiency virus type 1 (HIV-1) integration.Citation72 HIV-1 infection was nearly abolished using PARP-1 knockout fibroblasts.Citation72 PARP-1 is rapidly activated following Sindbis virus infection and contributes to apoptotic cell death.Citation73 For Epstein-Barr virus (EBV), PARP-1 has apparently two roles, both to protect EBV from excess DNA damage, while exerting antiviral activity to protect the cell from excessive EBV proliferation.Citation74

PARP-1 as a Pathogen Target?

H. pylori is the first pathogenic bacteria reported to directly modulate PARP-1 activity. However, given the increasing evidence for the significance of PARP-1 during host-pathogen interactions, it is perhaps not surprising that there is an expanding body of evidence indicating that PARP-1 may be an important target for modulation by some pathogens. In fact, several pathogenic viruses have been recently reported to produce proteins that directly interact with and activate PARP-1. For example, two capsid proteins of the Simian virus 40 (SV40) directly interact with PARP-1.Citation75 PARP-1 interaction with the minor capsid protein VP3 directly activates PARP-1 enzymatic activity. VP3-mediated PARP activation was linked to necrotic death of infected cells and the release of SV40 to the culture supernatant, suggesting a possible role for PARP activation in SV40 dissemination.Citation75 The polyomavirus capsid protein viral protein 1 (VP1) has been demonstrated to activate PAR synthesis by interacting directly with PARP-1 and knocking down or knocking PARP-1 in target cells reduced early viral transcription.Citation76 The Sindbis virus non-structural protein 3 interacts with PARP-1 within infected neuronal cells.Citation77

Other pathogens may act to prevent PARP-1 function. For example, the host immune response to viral infections was blocked by hepatitis B virus-mediated inhibition of PARP-1-dependent type I interferon receptor 1 transcription.Citation78 A second example is the prevention of PARP-1 nuclear localization by the Vpr protein of HIV-1, in complex with the glucocorticoid receptor, resulting in suppression of NFκB.Citation79 These few examples seem to suggest that some pathogens specifically target PARP-1 for activation, while others prevent PARP-1 activation as virulence strategies.

Exploiting H. pylori-activation of PARP-1 Therapeutically?

PARP-1 inhibitors have emerged as promising and effective therapeutics for breast, ovarian, melanoma and lymphoid cancers,Citation80 but the use of PARP-1 inhibitors for the treatment of H. pylori-associated stomach cancers remains to be evaluated clinically. However, using pre-clinical murine models of H. felis induced atrophic gastritis, epithelial hyperplasia and metaplasia, the small PARP-1 inhibitor PJ34 was recently reported to prevent the formation of Helicobacter-induced precancerous lesions.Citation81 Perhaps more exciting, however, was that PJ34, when combined with antibiotics, effectively reversed existing Helicobacter-induced pre-cancerous lesions.Citation81 These results are potentially highly significant because while antibiotics often effectively eliminate H. pylori infections in patients presenting with clinical symptoms, eradication therapy alone does not reverse H. pylori-associated metaplasia or dysplasia, which are strong predictors for the development of gastric cancer.Citation82

As mentioned above, PARP-1 is an important component of the BER pathway to repair damaged DNA. Thus, impairment of BER through PARP-1 inhibition may enhance the cytotoxicity of radiation or chemotherapy drugs that generate single-strand breaks in DNA.Citation83Citation85 In addition, PARP-1 inhibitors are emerging as “stand-alone” cancer therapeutics in their own right by inducing “synthetic lethality” in cancer cells in which DNA repair mechanisms that normally rescue BER-deficient cells are themselves impaired.Citation86 Some PARP-associated cancers have been correlated to mutations in the genes encoding the tumor suppressors BRCA1 and BRCA2 (breast cancer 1 and 2, early onset), which are important components of the homologous recombination repair pathway. Damage to DNA in BRCA1- or BRCA2-deficient cells is often not sufficiently repaired and can thus evolve into cytotoxic double stranded breaks. Thus, inhibition of a second, alternative DNA repair pathway, such as the BER pathway with PARP-1 inhibitors, can be highly effective at inducing selective lethal cytotoxicity in the tumor cells of BRCA1- or BRCA2-deficient individuals. This principle is thought to underlie the highly encouraging anti-tumor activity resulting from PARP-1 inhibition in breast cancer, as well as other BRCA deficient cancers such as ovarian cancer.Citation86,Citation87 Interestingly, BRCA mutations have now also been linked to increased risk in gastric cancers.Citation88,Citation89 It remains to be tested whether PARP-1 inhibition might also be an effective strategy for killing tumor cells and reversing pre-neoplastic lesions in patients with BRCA-deficient stomach cancers.

Future Prospects

There are a number of key issues surrounding the discovery of H. pylori-dependent PARP-1 activation that have yet to be explored. Detailed mechanistic studies will require that the PARP-1 activating factor be identified, likely via a combination of biochemical and genetic approaches. Consistent with the idea that the factor is released, direct contact of H. pylori with epithelial cells is not required for PARP-1 activation, as the application of sterilized culture filtrates alone, in the absence of bacteria cells, was sufficient for inducing activation.Citation4 PARP-1 was not activated when mammalian cells were incubated with bacteria that had been seperated from culture filtrate and then killed by mild heat treatment,Citation4 further suggesting that the activating factor is not normally associated with the bacterial outer membrane surface. Currently, it is not known whether the PARP-1 activating factor is specifically secreted or released non-specifically, although very little in vitro PARP-1 activating activity was detected within whole cell bacterial lysates,Citation14 suggesting that the activity does not accumulate intracellularly. The PARP-1 activating factor is not likely to be a small peptide, as activity is retained in dialysis membranes with molecular mass cutoffs of either 8 or 20 kDa (unpublished observation).

The mechanism of H. pylori-mediated PARP-1 activation is unknown, although preliminary studies suggest that a protein factor released by the bacterium may interact with the DNA-binding domain of PARP-1.Citation4 Consistent with the notion that the activating factor must access intracellular PARP-1 for activation to occur, the presence of a protein transfection reagent that promotes protein entry into cells resulted in significantly more PAR production than cells incubated with culture filtrate alone. Nothing is known about the pathways involved in the uptake and intracellular transport of the H. pylori PARP-1 activating factor, but it is likely that localization to the nucleus will be required for the bacterial factor to access PARP-1.

Ultimately, the importance of the PARP-1 activating factor for H. pylori colonization, persistence or disease pathogenesis will be determined using human epidemiologic studies coupled with animal studies employing an isogenic bacterial mutant lacking only the gene encoding the PARP-1 activating factor. The in vivo expression patterns of the PARP-1 activating factor in animal models of H. pylori colonization and disease, as well as during human infections may also provide clues as to the importance of PARP-1 modulation in establishing and maintaining homeostasis at the host-pathogen interface. Finally, identifying inflammatory and apoptotic signaling affected by PARP-1 activation will provide insights into the functional roles for PARP-1 during chronic infection.

Conclusions

PARP-1 is emerging as a key player in establishing homeostasis at the host-pathogen interface. Because of increasing evidence of the importance of PARP-1 in regulating host responses to infection, it is perhaps not surprising that a number of pathogens, primarily viruses to date, have been discovered to specifically target PARP-1 for modulation. The discovery of H. pylori-mediated PARP-1 activation suggests a novel pathway by which a bacterium may modulate host responses to establish and maintain the long-term dynamic relationship within its gastric environment. The use of PARP-1 inhibitors in successfully treating a broad spectrum of inflammatory diseases and cancers raises the interesting possibility of PARP-1 targeted therapeutic intervention for treating H. pylori-mediated gastric diseases.

Acknowledgements

This work was supported from a grant from the National Institutes of Health R01 AI045928 (S.R.B.).

Addendum to:

References

  • Blanke SR. Portals and pathways: principles of bacterial toxin entry into cells. Microbe 2006; 1:26 - 32
  • Atherton JC, Blaser MJ. Coadaptation of Helicobacter pylori and humans: ancient history, modern implications. J Clin Invest 2009; 119:2475 - 2487
  • Polk DB, Peek RM Jr. Helicobacter pylori: gastric cancer and beyond. Nat Rev Cancer 2010; 10:403 - 414
  • Nossa CW, Jain P, Tamilselvam B, Gupta VR, Chen LF, Schreiber V, et al. Activation of the abundant nuclear factor poly(ADP-ribose) polymerase-1 by Helicobacter pylori. Proc Natl Acad Sci USA 2009; 106:19998 - 20003
  • Ame JC, Spenlehauer C, de Murcia G. The PARP superfamily. Bioessays 2004; 26:882 - 893
  • Hakme A, Wong HK, Dantzer F, Schreiber V. The expanding field of poly(ADP-ribosyl)ation reactions. ‘Protein modifications: beyond the usual suspects’ Review Series. EMBO Rep 2008; 9:1094 - 1100
  • de Murcia G, Schreiber V, Molinete M, Saulier B, Poch O, Masson M, et al. Structure and function of poly(ADP-ribose) polymerase. Mol Cell Biochem 1994; 138:15 - 24
  • Jagtap P, Szabo C. Poly(ADP-ribose) polymerase and the therapeutic effects of its inhibitors. Nat Rev Drug Discov 2005; 4:421 - 440
  • Schreiber V, Dantzer F, Ame JC, de Murcia G. Poly(ADP-ribose): novel functions for an old molecule. Nat Rev Mol Cell Biol 2006; 7:517 - 528
  • Cohen-Armon M, Visochek L, Rozensal D, Kalal A, Geistrikh I, Klein R, et al. DNA-independent PARP-1 activation by phosphorylated ERK2 increases Elk1 activity: a link to histone acetylation. Mol Cell 2007; 25:297 - 308
  • Griesenbeck J, Ziegler M, Tomilin N, Schweiger M, Oei SL. Stimulation of the catalytic activity of poly(ADP-ribosyl) transferase by transcription factor Yin Yang 1. FEBS Lett 1999; 443:20 - 24
  • Kun E, Kirsten E, Mendeleyev J, Ordahl CP. Regulation of the enzymatic catalysis of poly(ADP-ribose) polymerase by dsDNA, polyamines, Mg2+, Ca2+, histones H1 and H3 and ATP. Biochemistry 2004; 43:210 - 216
  • Kirkland JB. Poly ADP-ribose polymerase-1 and health. Exp Biol Med (Maywood) 2010; 235:561 - 568
  • Nossa CW, Blanke SR. Modification of a mammalian cell protein in the presence of [32P-adenylate] NAD: evidence for ADP ribosylation activity associated with Helicobacter pylori. Infect Immun 2006; 74:3071 - 3076
  • Maeda S, Omata M. Inflammation and cancer: role of nuclear factorkappaB activation. Cancer Sci 2008; 99:836 - 842
  • Crabtree JE, Lindley IJ. Mucosal interleukin-8 and Helicobacter pylori-associated gastroduodenal disease. Eur J Gastroenterol Hepatol 1994; 6:33 - 38
  • Noach LA, Bosma NB, Jansen J, Hoek FJ, van Deventer SJ, Tytgat GN. Mucosal tumor necrosis factor-alpha, interleukin-1 beta and interleukin-8 production in patients with Helicobacter pylori infection. Scand J Gastroenterol 1994; 29:425 - 429
  • Crabtree JE, Xiang Z, Lindley IJ, Tompkins DS, Rappuoli R, Covacci A. Induction of interleukin-8 secretion from gastric epithelial cells by a cagA negative isogenic mutant of Helicobacter pylori. J Clin Pathol 1995; 48:967 - 969
  • Crowe SE, Alvarez L, Dytoc M, Hunt RH, Muller M, Sherman P, et al. Expression of interleukin 8 and CD54 by human gastric epithelium after Helicobacter pylori infection in vitro. Gastroenterology 1995; 108:65 - 74
  • Ding SZ, O'Hara AM, Denning TL, Dirden-Kramer B, Mifflin RC, Reyes VE, et al. Helicobacter pylori and H2O2 increase AP endonuclease-1/redox factor-1 expression in human gastric epithelial cells. Gastroenterology 2004; 127:845 - 858
  • Wilcox CM, Harris PR, Redman TK, Kawabata S, Hiroi T, Kiyono H, et al. High mucosal levels of tumor necrosis factor alpha messenger RNA in AIDS-associated cytomegalovirus-induced esophagitis. Gastroenterology 1998; 114:77 - 82
  • Wyatt JI, Rathbone BJ. Immune response of the gastric mucosa to Campylobacter pylori. Scand J Gastroenterol Suppl 1988; 142:44 - 49
  • Perez-Perez GI, Dworkin BM, Chodos JE, Blaser MJ. Campylobacter pylori antibodies in humans. Ann Intern Med 1988; 109:11 - 17
  • Beswick EJ, Pinchuk IV, Das S, Powell DW, Reyes VE. Expression of the programmed death ligand 1, B7-H1, on gastric epithelial cells after Helicobacter pylori exposure promotes development of CD4+ CD25+ FoxP3+ regulatory T cells. Infect Immun 2007; 75:4334 - 4341
  • Enarsson K, Lundgren A, Kindlund B, Hermansson M, Roncador G, Banham AH, et al. Function and recruitment of mucosal regulatory T cells in human chronic Helicobacter pylori infection and gastric adenocarcinoma. Clin Immunol 2006; 121:358 - 368
  • Harris PR, Wright SW, Serrano C, Riera F, Duarte I, Torres J, et al. Helicobacter pylori gastritis in children is associated with a regulatory T-cell response. Gastroenterology 2008; 134:491 - 499
  • Lundgren A, Stromberg E, Sjoling A, Lindholm C, Enarsson K, Edebo A, et al. Mucosal FOXP3-expressing CD4+ CD25 high regulatory T cells in Helicobacter pylori-infected patients. Infect Immun 2005; 73:523 - 531
  • Rad R, Brenner L, Bauer S, Schwendy S, Layland L, da Costa CP, et al. CD25+/Foxp3+ T cells regulate gastric inflammation and Helicobacter pylori colonization in vivo. Gastroenterology 2006; 131:525 - 537
  • Alam MS, Kurtz CC, Rowlett RM, Reuter BK, Wiznerowicz E, Das S, et al. CD73 is expressed by human regulatory T helper cells and suppresses proinflammatory cytokine production and Helicobacter felis-induced gastritis in mice. J Infect Dis 2009; 199:494 - 504
  • Fan X, Crowe SE, Behar S, Gunasena H, Ye G, Haeberle H, et al. The effect of class II major histocompatibility complex expression on adherence of Helicobacter pylori and induction of apoptosis in gastric epithelial cells: a mechanism for T helper cell type 1-mediated damage. J Exp Med 1998; 187:1659 - 1669
  • Jones NL, Shannon PT, Cutz E, Yeger H, Sherman PM. Increase in proliferation and apoptosis of gastric epithelial cells early in the natural history of Helicobacter pylori infection. Am J Pathol 1997; 151:1695 - 1703
  • Mannick EE, Bravo LE, Zarama G, Realpe JL, Zhang XJ, Ruiz B, et al. Inducible nitric oxide synthase, nitrotyrosine and apoptosis in Helicobacter pylori gastritis: effect of antibiotics and antioxidants. Cancer Res 1996; 56:3238 - 3243
  • Moss SF, Calam J, Agarwal B, Wang S, Holt PR. Induction of gastric epithelial apoptosis by Helicobacter pylori. Gut 1996; 38:498 - 501
  • Wagner S, Beil W, Westermann J, Logan RP, Bock CT, Trautwein C, et al. Regulation of gastric epithelial cell growth by Helicobacter pylori: evidence for a major role of apoptosis. Gastroenterology 1997; 113:1836 - 1847
  • Hassa PO, Haenni SS, Elser M, Hottiger MO. Nuclear ADP-ribosylation reactions in mammalian cells: where are we today and where are we going?. Microbiol Mol Biol Rev 2006; 70:789 - 829
  • Hauser B, Groger M, Ehrmann U, Albicini M, Bruckner UB, Schelzig H, et al. The Parp-1 inhibitor INO-1001 facilitates hemodynamic stabilization without affecting DNA repair in porcine thoracic aortic cross-clamping-induced ischemia/reperfusion. Shock 2006; 25:633 - 640
  • Black JH, Casey PJ, Albadawi H, Cambria RP, Watkins MT. Poly adenosine diphosphate-ribose polymerase inhibitor PJ34 abolishes systemic proinflammatory responses to thoracic aortic ischemia and reperfusion. J Am Coll Surg 2006; 203:44 - 53
  • Pacher P, Liaudet L, Mabley JG, Cziraki A, Hasko G, Szabo C. Beneficial effects of a novel ultrapotent poly(ADP-ribose) polymerase inhibitor in murine models of heart failure. Int J Mol Med 2006; 17:369 - 375
  • Bartha E, Solti I, Kereskai L, Lantos J, Plozer E, Magyar K, et al. PARP inhibition delays transition of hypertensive cardiopathy to heart failure in spontaneously hypertensive rats. Cardiovasc Res 2009; 83:501 - 510
  • Gonzalez-Rey E, Martinez-Romero R, O'Valle F, Aguilar-Quesada R, Conde C, Delgado M, et al. Therapeutic effect of a poly(ADP-ribose) polymerase-1 inhibitor on experimental arthritis by downregulating inflammation and Th1 response. PLoS One 2007; 2:1071
  • Szabo C, Virag L, Cuzzocrea S, Scott GS, Hake P, O'Connor MP, et al. Protection against peroxynitrite-induced fibroblast injury and arthritis development by inhibition of poly(ADP-ribose) synthase. Proc Natl Acad Sci USA 1998; 95:3867 - 3872
  • von Lukowicz T, Hassa PO, Lohmann C, Boren J, Braunersreuther V, Mach F, et al. PARP1 is required for adhesion molecule expression in atherogenesis. Cardiovasc Res 2008; 78:158 - 166
  • Czapski GA, Cakala M, Gajkowska B, Strosznajder JB. Poly(ADP-ribose) polymerase-1 inhibition protects the brain against systemic inflammation. Neurochem Int 2006; 49:751 - 755
  • Szabo C, Lim LH, Cuzzocrea S, Getting SJ, Zingarelli B, Flower RJ, et al. Inhibition of poly (ADP-ribose) synthetase attenuates neutrophil recruitment and exerts antiinflammatory effects. J Exp Med 1997; 186:1041 - 1049
  • Hauschildt S, Scheipers P, Bessler W, Schwarz K, Ullmer A, Flad HD, et al. Role of ADP-ribosylation in activated monocytes/macrophages. Adv Exp Med Biol 1997; 419:249 - 252
  • Giansanti V, Dona F, Tillhon M, Scovassi AI. PARP inhibitors: New tools to protect from inflammation. Biochem Pharmacol 2010; 80:1869 - 1877
  • Ernst PB, Peura DA, Crowe SE. The translation of Helicobacter pylori basic research to patient care. Gastroenterology 2006; 130:188 - 206
  • Scovassi AI, Poirier GG. Poly(ADP-ribosylation) and apoptosis. Mol Cell Biochem 1999; 199:125 - 137
  • Haddad M, Rhinn H, Bloquel C, Coqueran B, Szabo C, Plotkine M, et al. Anti-inflammatory effects of PJ34, a poly(ADP-ribose) polymerase inhibitor, in transient focal cerebral ischemia in mice. Br J Pharmacol 2006; 149:23 - 30
  • Huang D, Yang CZ, Yao L, Wang Y, Liao YH, Huang K. Activation and overexpression of PARP-1 in circulating mononuclear cells promote TNFalpha and IL-6 expression in patients with unstable angina. Arch Med Res 2008; 39:775 - 784
  • Naura AS, Datta R, Hans CP, Zerfaoui M, Rezk BM, Errami Y, et al. Reciprocal regulation of iNOS and PARP-1 during allergen-induced eosinophilia. Eur Respir J 2009; 33:252 - 262
  • Beneke S. Poly(ADP-ribose) polymerase activity in different pathologies—the link to inflammation and infarction. Exp Gerontol 2008; 43:605 - 614
  • Hassa PO, Buerki C, Lombardi C, Imhof R, Hottiger MO. Transcriptional coactivation of nuclear factorkappaB-dependent gene expression by p300 is regulated by poly(ADP)-ribose polymerase-1. J Biol Chem 2003; 278:45145 - 45153
  • Hassa PO, Haenni SS, Buerki C, Meier NI, Lane WS, Owen H, et al. Acetylation of poly(ADP-ribose) polymerase-1 by p300/CREB-binding protein regulates coactivation of NFkappaB-dependent transcription. J Biol Chem 2005; 280:40450 - 40464
  • David KK, Andrabi SA, Dawson TM, Dawson VL. Parthanatos, a messenger of death. Front Biosci 2009; 14:1116 - 1128
  • Wang Y, Dawson VL, Dawson TM. Poly(ADP-ribose) signals to mitochondrial AIF: A key event in parthanatos. Exp Neurol 2009;
  • Andrabi SA, Dawson TM, Dawson VL. Mitochondrial and nuclear cross talk in cell death: parthanatos. Ann NY Acad Sci 2008; 1147:233 - 241
  • Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ, et al. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 2002; 297:259 - 263
  • Ruchalski K, Mao H, Singh SK, Wang Y, Mosser DD, Li F, et al. HSP72 inhibits apoptosis-inducing factor release in ATP-depleted renal epithelial cells. Am J Physiol Cell Physiol 2003; 285:1483 - 1493
  • Gagne JP, Isabelle M, Lo KS, Bourassa S, Hendzel MJ, Dawson VL, et al. Proteome-wide identification of poly(ADP-ribose) binding proteins and poly(ADP-ribose)-associated protein complexes. Nucleic Acids Res 2008; 36:6959 - 6976
  • Andrabi SA, Kim NS, Yu SW, Wang H, Koh DW, Sasaki M, et al. Poly(ADP-ribose) (PAR) polymer is a death signal. Proc Natl Acad Sci USA 2006; 103:18308 - 18313
  • Yu SW, Andrabi SA, Wang H, Kim NS, Poirier GG, Dawson TM, et al. Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci USA 2006; 103:18314 - 18319
  • Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, et al. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 1999; 397:441 - 446
  • Cheung EC, Joza N, Steenaart NA, McClellan KA, Neuspiel M, McNamara S, et al. Dissociating the dual roles of apoptosis-inducing factor in maintaining mitochondrial structure and apoptosis. EMBO J 2006; 25:4061 - 4073
  • Modjtahedi N, Giordanetto F, Madeo F, Kroemer G. Apoptosis-inducing factor: vital and lethal. Trends Cell Biol 2006; 16:264 - 272
  • Ashktorab H, Dashwood RH, Dashwood MM, Zaidi SI, Hewitt SM, Green WR, et al. H. pylori-induced apoptosis in human gastric cancer cells mediated via the release of apoptosis-inducing factor from mitochondria. Helicobacter 2008; 13:506 - 517
  • Singh M, Prasad KN, Saxena A, Yachha SK. Helicobacter pylori induces apoptosis of T- and B-cell lines and translocates mitochondrial apoptosis-inducing factor to nucleus. Curr Microbiol 2006; 52:254 - 260
  • Oberholzer A, Oberholzer C, Moldawer LL. Interleukin-10: A complex role in the pathogenesis of sepsis syndromes and its potential as an anti-inflammatory drug. Crit Care Med 2002; 30:58 - 63
  • Kang X, Kim HJ, Ramirez M, Salameh S, Ma X. The septic shock-associated IL-10-1082 A > G polymorphism mediates allele-specific transcription via poly(ADP-Ribose) polymerase 1 in macrophages engulfing apoptotic cells. J Immunol 2010; 184:3718 - 3724
  • Altmeyer M, Barthel M, Eberhard M, Rehrauer H, Hardt WD, Hottiger MO. Absence of PARP1 delays the onset of Salmonella-induced gut inflammation. Infect Immun 2010; 78:3420 - 3431
  • Gaken JA, Tavassoli M, Gan SU, Vallian S, Giddings I, Darling DC, et al. Efficient retroviral infection of mammalian cells is blocked by inhibition of poly(ADP-ribose) polymerase activity. J Virol 1996; 70:3992 - 4000
  • Ha HC, Juluri K, Zhou Y, Leung S, Hermankova M, Snyder SH. Poly(ADP-ribose) polymerase-1 is required for efficient HIV-1 integration. Proc Natl Acad Sci USA 2001; 98:3364 - 3368
  • Nargi-Aizenman JL, Simbulan-Rosenthal CM, Kelly TA, Smulson ME, Griffin DE. Rapid activation of poly(ADP-ribose) polymerase contributes to Sindbis virus and staurosporine-induced apoptotic cell death. Virology 2002; 293:164 - 171
  • Tempera I, Deng Z, Atanasiu C, Chen CJ, D'Erme M, Lieberman PM. Regulation of Epstein-Barr virus OriP replication by poly(ADP-ribose) polymerase 1. J Virol 84:4988 - 4997
  • Gordon-Shaag A, Yosef Y, Abd El-Latif M, Oppenheim A. The abundant nuclear enzyme PARP participates in the life cycle of simian virus 40 and is stimulated by minor capsid protein VP3. J Virol 2003; 77:4273 - 4282
  • Carbone M, Reale A, Di Sauro A, Sthandier O, Garcia MI, Maione R, et al. PARP-1 interaction with VP1 capsid protein regulates polyomavirus early gene expression(dagger). J Mol Biol 2006; 363:773 - 785
  • Park E, Griffin DE. Interaction of Sindbis virus non-structural protein 3 with poly(ADP-ribose) polymerase 1 in neuronal cells. J Gen Virol 2009; 90:2073 - 2080
  • Zhou J, Huang JD, Poon VK, Chen DQ, Chan CC, Ng F, et al. Functional dissection of an IFNalpha/beta receptor 1 promoter variant that confers higher risk to chronic hepatitis B virus infection. J Hepatol 2009; 51:322 - 332
  • Muthumani K, Choo AY, Zong WX, Madesh M, Hwang DS, Premkumar A, et al. The HIV-1 Vpr and glucocorticoid receptor complex is a gain-of-function interaction that prevents the nuclear localization of PARP-1. Nat Cell Biol 2006; 8:170 - 179
  • Drew Y, Plummer R. PARP inhibitors in cancer therapy: two modes of attack on the cancer cell widening the clinical applications. Drug Resist Updat 2009; 12:153 - 156
  • Toller IM, Altmeyer M, Kohler E, Hottiger MO, Muller A. Inhibition of ADP ribosylation prevents and cures Helicobacter-induced gastric preneoplasia. Cancer Res 2010; 70:5912 - 5922
  • Rokkas T, Pistiolas D, Sechopoulos P, Robotis I, Margantinis G. The long-term impact of Helicobacter pylori eradication on gastric histology: a systematic review and meta-analysis. Helicobacter 2007; 12:32 - 38
  • Rodon J, Iniesta MD, Papadopoulos K. Development of PARP inhibitors in oncology. Expert Opin Investig Drugs 2009; 18:31 - 43
  • Lord CJ, Ashworth A. Targeted therapy for cancer using PARP inhibitors. Curr Opin Pharmacol 2008; 8:363 - 369
  • Kinsella TJ. Coordination of DNA mismatch repair and base excision repair processing of chemotherapy and radiation damage for targeting resistant cancers. Clin Cancer Res 2009; 15:1853 - 1859
  • Boss DS, Beijnen JH, Schellens JH. Inducing Synthetic Lethality using PARP Inhibitors. Curr Clin Pharmacol 2010; 5:192 - 195
  • Comen EA, Robson M. Poly(ADP-ribose) polymerase inhibitors in triple-negative breast cancer. Cancer J 16:48 - 52
  • Friedenson B. BRCA1 and BRCA2 pathways and the risk of cancers other than breast or ovarian. MedGenMed 2005; 7:60
  • Mohamad HB, Apffelstaedt JP. Counseling for male BRCA mutation carriers: a review. Breast 2008; 17:441 - 450