Figure 1 - uploaded by Janos Hunyadi
Content may be subject to copyright.
Basal and hydrogen peroxide-induced poly-ADP ribosylation in J774 macrophages. Untreated ( A,B ) and hydrogen peroxide- treated J774 cells were stained with the bio-NAD method as described in Materials and Methods. A low-intensity nuclear staining could be visualized with the assay in untreated cells, reflecting baseline PARP activity ( A,B ). Mitotic cells displayed increased poly-ADP ribosylating activity (arrows). In response to hydrogen peroxide (500 ␮ M), a markedly increased nuclear staining could be detected in J774 cells ( C ) but not in cells pretreated with the PARP inhibitor PJ34 (5 ␮ M) ( D ). Bars: A,C,D ϭ 8 ␮ m; B ϭ 3 ␮ m. 

Basal and hydrogen peroxide-induced poly-ADP ribosylation in J774 macrophages. Untreated ( A,B ) and hydrogen peroxide- treated J774 cells were stained with the bio-NAD method as described in Materials and Methods. A low-intensity nuclear staining could be visualized with the assay in untreated cells, reflecting baseline PARP activity ( A,B ). Mitotic cells displayed increased poly-ADP ribosylating activity (arrows). In response to hydrogen peroxide (500 ␮ M), a markedly increased nuclear staining could be detected in J774 cells ( C ) but not in cells pretreated with the PARP inhibitor PJ34 (5 ␮ M) ( D ). Bars: A,C,D ϭ 8 ␮ m; B ϭ 3 ␮ m. 

Source publication
Article
Full-text available
Poly(ADP-ribose) polymerase (PARP) is a nuclear enzyme activated by DNA damage. Activated PARP cleaves NAD(+) into nicotinamide and (ADP-ribose) and polymerizes the latter on nuclear acceptor proteins. Over-activation of PARP by reactive oxygen and nitrogen intermediates represents a pathogenetic factor in various forms of inflammation, shock, and...

Context in source publication

Context 1
... polymerase (PARP) is a nuclear enzyme that becomes activated in response to DNA damage (de Murcia and Menissier de Murcia 1994). Activated PARP cleaves NAD ϩ to nicotinamide and ADP-ribose and polymerizes the latter on nuclear acceptor proteins such as histones, transcription factors, and PARP itself. Poly-ADP ribosylation contributes to DNA repair and to the maintenance of genomic stability (Wang et al. 1997; Simbulan–Rosenthal et al. 1999; Muiras and Bürkle 2000). During inflammation, ischemia–reperfusion, or shock, free radical/oxi- dant-induced DNA single-strand breakage triggers the over-activation of PARP, leading to depletion of NAD ϩ (Szabó and Dawson 1998; Szabó 2000). In an effort to re-synthesize NAD ϩ , ATP is also consumed, resulting in necrotic type cell death (Schraufstatter et al. 1986; Virág et al. 1998a,b). This PARP-mediated pathway of cell suicide has been implicated in the death of immune-stimulated macrophages as well as in peroxynitrite- or hydrogen peroxide-induced dysfunction or cell death of thymocytes, macrophages, endothelial cells, neuronal cells, and fibroblasts (Zingarelli et al. 1996; Szabó et al. 1998; Virág et al. 1998b; Soriano et al. 2001). Inhibition of PARP activity by pharmacological inhibitors or the absence of functional PARP enzyme in PARP knockout animals provided significant protection in animal models of a wide variety of diseases, including various forms of inflammation, shock, stroke, myocardial ischemia, dia- betes, and diabetic endothelial dysfunction (Szabó and Dawson 1998; Szabó 2000). For measurement of PARP activity, the incorporation of radioactivity from isotope-labeled NAD ϩ into TCA-precipitable proteins is considered the gold standard (Schraufstatter et al. 1986). Alternatively, poly(ADP-ribose), the product of the PARP-catalyzed reaction can be purified from cells and tissues by a te- dious procedure and the polymer can be quantitated by HPLC (Kiehlbauch et al. 1993; Shah et al. 1995). A more convenient approach to assessment of PARP activation is the detection of poly(ADP-ribose) by a monoclonal anti-poly(ADP-ribose) antibody in West- ern blots, dot-blots, immunocytochemistry, and flow cytometry (Affar et al. 1998,1999). However, the use of the murine monoclonal anti-poly(ADP-ribose) antibody in mouse tissues, especially in inflamed tissues, often results in high background staining (unpub- lished observations). Moreover, the amount of the polymer synthesized does not necessarily reflect the degree of PARP activation, because in the cells and tissues poly(ADP-ribose) is rapidly metabolized by poly(ADP- ribose) glycohydrolase (PARG) (Ueda et al. 1972). Recently, a novel non-radioactive assay has been marketed by Trevigen (Gaithersburg, MD) for screen- ing of potential PARP inhibitors. The assay utilizes a novel PARP substrate, 6-biotin-17-nicotinamide-ade- nine-dinucleotide (bio-NAD ϩ ), which was originally developed to detect and isolate mono-ADP-ribosylated proteins (Zhang and Snyder 1993). In this assay, 96- well plates are coated with histones as acceptor proteins and biotinyl-ADP ribose is incorporated from bio-NAD ϩ into the histones by purified PARP. Biotin incorporation is then detected by streptavidin–peroxidase and a suitable peroxidase substrate. The potency of PARP inhibitors is assessed on the basis of their inhibition of biotinyl–ADP-ribose incorporation. We set out to investigate whether this commercially available novel PARP substrate, bio-NAD ϩ , can be used to detect cellular PARP activation. We have developed a cellular ELISA (CELISA) assay to quantify PARP activation in cultured cells and an enzyme cytochemical/ histochemical reaction to detect PARP activation in oxidatively stressed cells and tissues. J774 macrophages were stained for PARP activity with the bio-NAD ϩ substrate (Figure 1). All cells showed a predominantly nuclear staining (Figure 1A), indicating that the bio-NAD ϩ -metabolizing enzyme localizes in the nucleus. The intensity of nuclear staining in the vast majority of cells was moderate, reflecting basal PARP activity. In sharp contrast to interphase cells, mitotic cells displayed an intense nuclear staining (Figures 1A and 1B). These cells appeared to be in the metaphase or ana–telophase of the mitotic cycle. Treatment of J774 cells with 500 ␮ M H 2 O 2 induced a marked enhancement of ADP ribosylating activity, as indicated by the strong nuclear staining of H 2 O 2 -treated cells (Figure 1C). Pretreatment of cells with the novel potent PARP inhibitor PJ34 (5 ␮ M) (Abdelkarim et al. 2001; Soriano et al. 2001) 30 min before H 2 O 2 exposure prevented enhancement of bio- NAD ϩ staining (Figure 1D). Similar results were also obtained with other types of cells, including fibroblasts and human keratinocytes (data not shown). To prove the identity of the product synthesized by the cells from bio-NAD ϩ , we used wild-type (PARP ϩ / ϩ ) and PARP-deficient (PARP Ϫ / Ϫ ) macrophages (Figure 2). Exposure of cells to 200 ␮ M hydrogen peroxide induced bio-NAD ϩ incorporation into the nuclei of wild-type but not of PARP-deficient macrophages. Bio-NAD ϩ incorporation could be inhibited with PJ34 (Figure 2) or 3-aminobenzamide (not shown). These results indicate that PARP-1 is responsible for bio-NAD ϩ incorporation into hydrogen peroxide-treated macrophages. To demonstrate the ability of the bio-NAD method to detect PARP activation in tissues, we applied hydrogen peroxide (250 nmol/50 ␮ l) to the skin of mice for 30 min. Skin was excised and immediately frozen in cryoembedding medium to preserve enzyme activity. Frozen sections (10 ␮ m) permeabilized with Triton X-100 were incubated with the bio-NAD ϩ substrate followed by biotin detection with streptavidin–per- oxidase. In control (vehicle-treated) skin, no detect- able ADP ribosylation was found (Figure 3A). Peroxynitrite treatment activated PARP in the skin, as indicated by the appearance of darkly stained cells (Figure 3B). Staining was nuclear and was most intense in keratinocytes. However, some scattered cells in the dermis also showed nuclear PARP activity (Figure 3B). The presence of the PARP inhibitor PJ34 (5 ␮ M) (Figure 3C) or 3-aminobenzamide (5 mM) (not shown) abolished peroxynitrite-induced bio-ADP–ribose incorporation, demonstrating that PARP activation was responsible for the staining. A cellular ELISA method allows the quantification of PARP activity. Furthermore, the potency of pharmacological PARP inhibitors in cells can also be deter- mined in a CELISA. J774 macrophages seeded in 96-well plates were exposed to hydrogen peroxide (50–400 ␮ M) in the presence or absence of PJ34 (5 ␮ M). Hydrogen peroxide induced a dose-dependent PARP activation in J774 cells, and pretreatment with the PARP inhibitor PJ-34 suppressed hydrogen peroxide- induced PARP activation (Figure 4). As a reference method, [ 3 H]-NAD incorporation was also used to measure PARP activity and the two assays showed good correlation ( r 2 ϭ 0.92) with the bio-NAD ϩ method, giving higher induction results. We have demonstrated that bio-NAD can be used as a substrate for PARP in cells and tissues. In untreated J774 macrophage, bio-NAD ϩ metabolizing activity could be detected in the nuclei. The nuclear staining pattern indicates that bio-NAD is metabolized most likely by PARP, a nuclear enzyme polymerizing ADP- ribose units on nuclear acceptor proteins. It has been shown previously that bio-NAD ϩ can also serve as a substrate for mono-ADP ribosylation in vitro (Zhang and Snyder 1993). However, this G-protein-coupled process takes place in the plasma membrane. Because bio-NAD staining is localized to the nuclei, mono- ADP ribosylation is not likely to contribute to bio- NAD ϩ metabolism in our system. Furthermore, the polymeric nature of the PARP product, as opposed to mono-ADP-ribose, may also be responsible for its good detectability with streptavidin–peroxidase. We observed an interesting phenomenon in untreated J774 cells. Mitotic cells displayed strong nuclear posi- tivity compared to interphase cells. Many cells repre- senting various stages of mitosis could be found with strong reactivity localized to the condensed chromatin but not to the interchromatin areas. This observation, however, is not surprising in light of previous observations linking PARP to the process of replication. PARP has been shown to associate with and to regulate the activity of topoisomerase I, an enzyme-uncoiling DNA, by temporarily cutting into one of the DNA strands (Ferro et al. 1984; Bauer et al. 2000; Bauer and Kun 2000). Association of PARP with other proteins in- volved in replication, such as the DNA polymerase I–pri- mase complex, has also been demonstrated (Dantzer et al. 1998). Furthermore, it has also been reported that in PARP knockout cells, but not in ...

Citations

... The detection of PARP-1 activity is crucial as PARP-1 may serve as a potential biomarker. Frequently used techniques for detecting PARP-1 include enzyme-linked immunosorbent assay (ELISA), biotin labeling, immunoblotting, fluorescence, and colorimetry [102][103][104][105][106]. Recently, novel methods have been developed for more efficient detection of PARP-1 activity. ...
Article
Full-text available
Advancements in technology have resulted in increasing concerns over the safety of eye exposure to light illumination, since prolonged exposure to intensive visible light, especially to short-wavelength light in the visible spectrum, can cause photochemical damage to the retina through a photooxidation-triggered cascade reaction. Poly(ADP-ribose) polymerase-1 (PARP-1) is the ribozyme responsible for repairing DNA damage. When damage to DNA occurs, including nicks and breaks, PARP-1 is rapidly activated, synthesizing a large amount of PAR and recruiting other nuclear factors to repair the damaged DNA. However, retinal photochemical damage may lead to the overactivation of PARP-1, triggering PARP-dependent cell death, including parthanatos, necroptosis, and autophagy. In this review, we retrieved targeted articles with the keywords such as “PARP-1,” “photoreceptor,” “retinal light damage,” and “photooxidation” from databases and summarized the molecular mechanisms involved in retinal photooxidation, PARP activation, and DNA repair to clarify the key regulatory role of PARP-1 in retinal light injury and to determine whether PARP-1 may be a potential marker in response to retinal photooxidation. The highly sensitive detection of PARP-1 activity may facilitate early evaluation of the effects of light on the retina, which will provide an evidentiary basis for the future assessment of the safety of light illumination from optoelectronic products and medical devices.
... For example, PARP inhibitors have been reported to exert anti-inflammatory effects in a wide range of systemic models of inflammation (endotoxemia, peritonitis, colitis, streptozotocin-induced diabetes) (40) and to protect endothelial-dependent responses from oxidant stress associated with aging (41), streptozotocin-induced diabetes (42), hypochlorite (43), and H 2 O 2 (44). Exposing macrophages to oxidants stimulates their PARP activity (45). Exposing cultured BV2 microglial cells to lipopolysaccharide induces NF-κB, inducible nitric oxide synthase (iNOS), nitrite formation, reactive oxygen species formation, and tumor necrosis factorα (TNFα), all of which are attenuated by the PARP inhibitor PJ34 (46). ...
Article
Full-text available
Parthanatos is a cell death signaling pathway in which excessive oxidative damage to DNA leads to over-activation of poly(ADP-ribose) polymerase (PARP). PARP then generates the formation of large poly(ADP-ribose) polymers that induce the release of apoptosis-inducing factor from the outer mitochondrial membrane. In the cytosol, apoptosis-inducing factor forms a complex with macrophage migration inhibitory factor that translocates into the nucleus where it degrades DNA and produces cell death. In a review of the literature, we identified 24 publications from 13 laboratories that support a role for parthanatos in young male mice and rats subjected to transient and permanent middle cerebral artery occlusion (MCAO). Investigators base their conclusions on the use of nine different PARP inhibitors (19 studies) or PARP1-null mice (7 studies). Several studies indicate a therapeutic window of 4–6 h after MCAO. In young female rats, two studies using two different PARP inhibitors from two labs support a role for parthanatos, whereas two studies from one lab do not support a role in young female PARP1-null mice. In addition to parthanatos, a body of literature indicates that PARP inhibitors can reduce neuroinflammation by interfering with NF-κB transcription, suppressing matrix metaloproteinase-9 release, and limiting blood-brain barrier damage and hemorrhagic transformation. Overall, most of the literature strongly supports the scientific premise that a PARP inhibitor is neuroprotective, even when most did not report behavior outcomes or address the issue of randomization and treatment concealment. Several third-generation PARP inhibitors entered clinical oncology trials without major adverse effects and could be repurposed for stroke. Evaluation in aged animals or animals with comorbidities will be important before moving into clinical stroke trials.
... This process is accompanied by downregulation of PARP1 expression via transcription factors, specifically factors 1 and 3 (Sp1 and Sp3), as demonstrated in various primary cells [53] and in keratinocytes [54]. This may be related to intrinsic cell cycle-related gene regulation [55,56] and/or integrin signaling [57]. How PARP1 expression changes when neoplastic cells are in contact with each other (e.g., in 2D or 3D cell culture models or in vivo tumors) and how adhesion factors in the tumor stroma affect PARP1 expression are largely unexplored. ...
Article
Full-text available
Simple Summary Poly (ADP-ribose) polymerase (PARP) proteins regulate DNA damage correction, replication, and gene transcription. By controlling pivotal aspects of these processes, PARPs are heavily implicated in cancer development. Inhibitors of PARPs, approved for cancer chemotherapy a few years ago, have achieved great success against tumors of the breast and ovary carrying mutations in the BRCA1/2 genes. The spectrum of the inhibitors is avidly sought to be extended to tumors with different genetic backgrounds and cancers of other origins. This pursuit requires thorough apprehension of PARP-dependent processes affecting cancer development. The hallmarks of cancer are acquired by defining capabilities that differentiate cancer cells from their normal counterparts. Here, in two joint papers, we walk through the connections between these cancer traits and PARP functions. The present review focuses on how PARPs affect the features of cancer that can be attributed to cell-intrinsic changes increasing proliferative potential and survival capabilities. In a kindred paper, we explore the PARP association of cancer hallmarks that derive from tissue-level reorganization in tumors and intercellular interactions of cancer cells. Abstract The 17-member poly (ADP-ribose) polymerase enzyme family, also known as the ADP-ribosyl transferase diphtheria toxin-like (ARTD) enzyme family, contains DNA damage-responsive and nonresponsive members. Only PARP1, 2, 5a, and 5b are capable of modifying their targets with poly ADP-ribose (PAR) polymers; the other PARP family members function as mono-ADP-ribosyl transferases. In the last decade, PARP1 has taken center stage in oncology treatments. New PARP inhibitors (PARPi) have been introduced for the targeted treatment of breast cancer 1 or 2 (BRCA1/2)-deficient ovarian and breast cancers, and this novel therapy represents the prototype of the synthetic lethality paradigm. Much less attention has been paid to other PARPs and their potential roles in cancer biology. In this review, we summarize the roles played by all PARP enzyme family members in six intrinsic hallmarks of cancer: uncontrolled proliferation, evasion of growth suppressors, cell death resistance, genome instability, reprogrammed energy metabolism, and escape from replicative senescence. In a companion paper, we will discuss the roles of PARP enzymes in cancer hallmarks related to cancer-host interactions, including angiogenesis, invasion and metastasis, evasion of the anticancer immune response, and tumor-promoting inflammation. While PARP1 is clearly involved in all ten cancer hallmarks, an increasing body of evidence supports the role of other PARPs in modifying these cancer hallmarks (e.g., PARP5a and 5b in replicative immortality and PARP2 in cancer metabolism). We also highlight controversies, open questions, and discuss prospects of recent developments related to the wide range of roles played by PARPs in cancer biology. Some of the summarized findings may explain resistance to PARPi therapy or highlight novel biological roles of PARPs that can be therapeutically exploited in novel anticancer treatment paradigms.
... [217] An NAD analog called Bio-NAD (6-biotin-17-nicotinamide-adenine-dinucleotide) has been used for the isolation of ribosylated proteins and for the quantification of PARP activity in cells. [218,219] An orthogonal NAD ana-log has been used in combination with genetically engineered members of the ARTD family to label and identify specific target proteins. [220] Recently many modified NAD analogs have been employed to visualize PARylation in living cells using FLIM microscopy. ...
Thesis
Nucleotides are biomolecules that not only act as a cellular constituent but are also crucial for cellular functioning and homeostasis. A nucleotide consists of a nitrogenous base, sugar, and phosphate groups. Nucleotides play a critical role in regulatory processes including cellular signaling, metabolism, and energy transfer. Adenosine triphosphate (ATP) is a mononucleotide that acts as an intermediate energy source. Nicotinamide adenine dinucleotide (NAD+) is a dinucleotide central to metabolism and is involved in electron transfer in various redox reactions. ATP and NAD+ are, however, also involved in posttranslational protein modifications (PTM). The present work involves the study of two prominent nucleotides and their dynamics in living cells using fluorescence microscopy. We have used the Förster resonance energy transfer (FRET) microscopy based on fluorescence lifetime imaging (FLIM) to monitor the activity and kinetics of two nucleotide analogs of ATP and NAD+. The activity of ATP hydrolysis was visualized by using fluorescence microscopy in combination with fluorescent ATP analogs. The ATP analog adenosine tetraphosphate (Ap4) was modified with the rhodamine derivative Atto-488 at the terminal phosphate (donar) and a non-fluorescent quencher group at the adenosine base (acceptor). In the intact molecule, the fluorescence of the dye is quenched due to the Förster resonance energy transfer (FRET) because of the spatial proximity between the donor and acceptor. Enzymatic cleavage results in an increase in the fluorescence intensity as well as an increase in the fluorescence lifetime of the dye. This principle was used to quantify the hydrolysis of analogs in vitro as well as in living cells by using fluorescence lifetime imaging (FLIM) microscopy and confocal scanning microscopy. Our experiments revealed that the hydrolysis of Ap4 analogs largely takes place in lysosomes. It was observed that the analog is hydrolyzed and is used as a potential energy source in lysosomes and autolysosomes during the process of autophagy. We thus have been able to successfully monitor the hydrolysis of a nucleotide in living cells. Protein Poly(ADP-ribosyl)ation (PARylation) is a primary step in DNA damage response. NAD+ is used as a donor for the process of protein PARylation. A novel fluorescent analog of NAD+, TMR-NAD, was synthesized with tetramethylrhodamine dye attached to it. The analog was used in combination with EGFP fusion proteins to monitor the PARylation of ARTD1 which is primarily involved in PARylation. With the application of FLIM-FRET imaging, the real-time visualization of PARylation in living cells using this analog was achieved. The decrease in the fluorescence lifetime of the donor fluorophore is measured to visualize and quantify protein PARylation and the associated interactions. The kinetics of the recruitment and PARylation of two proteins i.e. ARTD1 and mH2A were determined in DNA damage response. To induce the DNA damage, ultra-short laser pulses from the near-infrared (NIR) laser were used for the micro-irradiation of the nucleus in a well-defined region. Using this analog approach, it is conceivable to monitor the dynamics of molecular events involved in the DNA damage response with incredible spatial and temporal resolution.
... H 2 O 2 causes DNA damage (31), which stimulates PARP in RPE cells (32). Excessive PARP activity can deplete NAD + by using it as a substrate to synthesize poly(ADP ribose) (PAR) (Fig. 3G) (33,34). To determine if PARP is responsible for the effect of H 2 O 2 on NAD + in RPE cells, we treated them with 10 μM PJ34, a potent cell-permeable PARP inhibitor (EC 50 = 20 nM). ...
Article
Full-text available
Significance In the vertebrate eye, a monolayer of cells, called the retinal pigment epithelium (RPE), is between the choroidal blood supply and the retina. The RPE provides metabolic support for the retina, including delivery of glucose and other nutrients. Here, we show that reductive carboxylation of α-ketoglutarate, a type of metabolism that supports growth and survival of cancer cells, is a prominent feature of RPE cells. We show that extreme oxidative stress can overwhelm the reductive carboxylation pathway. However, we also found that the RPE can be protected from extreme oxidative stress by supplementation with an NAD ⁺ precursor or α-ketoglutarate.
... The purpose of the current study was to measure the physiological levels of PAR, which are quite low, in cultured cells. Various methods have been reported for this purpose, including radioimmunoassay [24], radioisotope labeling methods [25,26], the use of nonisotopic compounds [27,28], enzyme-linked immunosorbent assay (ELISA) [29,30], and stable isotope dilution mass spectrometry [31]. However, some current assays have low sensitivity for measuring PAR in intact cells, are not amenable to high-throughput measurements, or need expensive instrumentation. ...
... First, we developed a sensitive and specific ELISA and successfully measured small amounts of PAR. The sensitivity is quite high as compared with previous methods [28][29][30], and the limit of detection is 5 pg of PAR, which corresponds to 9 fmol of ADP-ribose residues in PAR. Further improvement of the sensitivity awaits further study. ...
Article
Full-text available
PolyADP-ribosylation is mediated by poly(ADP-ribose) (PAR) polymerases (PARPs) and may be involved in various cellular events, including chromosomal stability, DNA repair, transcription, cell death, and differentiation. The physiological level of PAR is difficult to determine in intact cells because of the rapid synthesis of PAR by PARPs and breakdown of PAR by PAR-degrading enzymes, including poly(ADP-ribose) glycohydrolase (PARG) and ADP-ribosylhydrolase 3. Artifactual synthesis and/or degradation of PAR likely occurs during lysis of cells in culture. We developed a sensitive enzyme-linked immunosorbent assay (ELISA) to measure the physiological levels of PAR in cultured cells. We immediately inactivated enzymes that catalyze the synthesis and degradation of PAR. We validated that trichloroacetic acid is suitable for inactivating PARPs, PARG, and other enzymes involved in metabolizing PAR in cultured cells during cell lysis. The PAR level in cells harvested with the standard radio-immunoprecipitation assay buffer was increased by 450-fold compared to trichloroacetic acid for lysis, presumably because of activation of PARPs by DNA damage that occurred during cell lysis. This ELISA can be used to analyze the biological functions of polyADP-ribosylation under various physiological conditions in cultured cells.
... Proteolytic activity in plant cells undergoing PCD has also been studied using poly(ADP-ribose) polymerase (PARP), a well-characterized substrate for human caspase-3. Endogenous PARP cleavage occurs during menadione-induced PCD (Sun et al., 1999b;Sirisha et al., 2014); H 2 O 2 -induced PCD (Amor et al., 1998;Bakondi et al., 2002;Zákány et al., 2007) and in heat-shock-treated tobacco suspension cells (Tian et al., 2000). Having used the anti-PARP-1 from humans for our assays, we have also shown that it cross-reacts to a PARP-1-like enzyme from C. reinhardtii. ...
Article
Full-text available
Eukaryotic microalgae serve as indicators of environmental change when exposed to severe seasonal fluctuations. Several environmental stress conditions are known to produce reactive oxygen species in cellular compartments, resulting in oxidative damage and apoptosis. The study of cell death in higher plants and animals has revealed the existence of an active ‘programmed cell death’ (PCD) process and similarities between such processes suggest an evolutionary origin. A study was undertaken to examine the morphological, biochemical and molecular responses of the unicellular green alga Chlamydomonas reinhardtii after exposure to oxidative (10 mM H2O2) and osmotic (200 mM NaCl and 360 mM sorbitol) stress. Concentrations of H2O2 (2–50 mM), NaCl and sorbitol (100–800 mM) were negatively correlated with growth. Biochemical analyses showed an increase in intracellular H2O2 production (2.2-fold with H2O2 and ~1.2–1.4-fold with NaCl and sorbitol) and activities of some antioxidant enzymes [super oxide dismutase (SOD), catalase (CAT) and ascorbate peroxidase (APX)]. Alteration of mitochondrial membrane potential (MMP) was observed upon treatment with H2O2 and NaCl, but not with sorbitol, indicating that the ionic stress component of NaCl altered the MMP. In addition, H2O2 led to the activation of a caspase-3-like protein, increase in the cleavage of a poly(ADP) ribose polymerase-1 (PARP-1)-like enzyme and formation of DNA nicks and laddering. With NaCl and sorbitol, no caspase activation, nor oligonucleosomal DNA laddering was observed, indicating non-apoptotic death. However, genomic DNA of NaCl (800 mM)-stressed cells, but not those of sorbitol-treated cells showed complete shearing. We conclude that the ionic rather than the osmotic component of NaCl leads to necrosis. These results unequivocally suggest that the vegetative cells of C. reinhardtii respond differentially to various stress agents, leading to different death types in the same organism. Moreover, unlike most other organisms, when exposed to NaCl this alga does not undergo PCD.
... PARP-1 activity in chondrocytes was determined using a colorimetric assay, as described previously [18]. Briefly, cells were stimulated with 500 mM H 2 O 2 , and then the medium was replaced with PARP-1 reaction buffer. ...
Article
Osteoarthritis (OA) is an age-related joint disease that is characterized by the degeneration of articular chondrocytes. Nuclear enzyme poly(ADP-ribose) polymerase 1 (PARP-1) is associated with inflammation response. We investigated the role of PARP-1 in interleukin-1β (IL-1β)-stimulated human articular chondrocytes and its underlying mechanism. Cell viability and apoptosis were evaluated by using 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide assay and flow cytometry, respectively. Tumor necrosis factor-α (TNF-α) level was measured by enzyme-linked immunosorbent assay. The mRNA and protein expression levels of PARP-1, IL-receptor (IL-1R), inducible nitric oxide synthase (iNOS), matrix metalloproteinases (MMPs), and tissue inhibitor of metalloproteinases-1 (TIMP-1) were determined by real-time reverse transcriptase-polymerase chain reaction and western blot analysis, respectively. The expression and phosphorylation of NF-кB p65 were measured by western blot analysis. Results showed that stimulation of chondrocytes with IL-1β caused a significant up-regulation of PARP-1 and IL-1R, resulting in NF-кB p65 nuclear translocation and phosphorylation associated with an increase of TNF-α secretion and iNOS expression. PARP-1 was inhibited by siRNA transfection. Results showed that PARP-1 inhibition suppressed IL-1β-induced reduction of cell viability and up-regulation of cell apoptosis, with a reduced IL-1R expression. PARP-1 inhibition also effectively reversed IL-1β-induced inflammatory response through inhibiting the IL-1R/NF-кB pathway. These data suggested that PARP-1 inhibition prevents IL-1β-induced inflammation response at least partly by inhibiting the IL-1R/NF-кB signaling pathway in human articular chondrocytes. Moreover, PARP-1 inhibition reduced MMPs expression and increased TIMP-1 expression, suggesting that PARP-1 inhibition could suppress cartilage destruction by modulating the balance between MMPs and TIMP-1. Inhibition of PARP-1 might be useful in the treatment of OA. © The Author 2015. Published by ABBS Editorial Office in association with Oxford University Press on behalf of the Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences.
... PARP1 is the most abundant isoform of the PARP enzyme family and, upon activation by genotoxic stimuli, cleaves nicotinamide adenine dinucleotide (NAD + ) into nicotinamide (NAM), resulting in the formation of ADP-ribose moieties; these moieties covalently attach to various acceptor proteins, including PARP itself. The continued activation of PARP leads to depletion of its substrate, NAD + and, consequently, adenosine-5′-triphosphate (ATP), energy failure and cell death (11)(12)(13)(14)(15)(16)(17)(18)(19)(20). The benefits conferred by pharmacological inhibitors of poly(ADP-ribosyl)ation in several experimental disease models, including sepsis, further emphasize the potential importance of PARP1 as a pharmacological target . ...
Article
Full-text available
Pathophysiological conditions that lead to the release of the prototypic damage-associated molecular pattern molecule high mobility group box-1 (HMGB1) also result in activation of poly (ADP-ribose) polymerase (now known as ADP-ribosyl transferases (ARTD))-1 or PARP1. Persistent activation of PARP1 promotes energy failure and cell death. The role of poly(ADP-ribosyl)ation in HMGB1 release has been previously explored; however, PARP1 is a versatile enzyme and performs several other functions including cross-talk with another nicotinamide adenine dinucleotide (NAD(+)) dependent member of the Class III histone deacetylases (HDACs)-sirtuin-1 (SIRT1). Previously, it has been shown that the hyperacetylation of HMGB1 is a seminal event prior to its secretion, a process that is also dependent on HDACs. Therefore, in this study, we seek to determine if PARP1 inhibition alters LPS-mediated HMGB1 hyperacetylation and subsequent secretion due to its effect on SIRT1.We demonstrate in an in vitro model that LPS treatment leads to hyperacetylated HMGB1with concomitant reduction in nuclear HDAC activity. Treatment with PARP1 inhibitors mitigates the LPS-mediated reduction in nuclear HDAC activity and decreases HMGB1 acetylation. By utilizing an NAD(+) based mechanism, PARP1 inhibition increases the activity of SIRT1. Consequently, there is an increased nuclear retention and decreased extracellular secretion of HMGB1. We also demonstrate that PARP1 physically interacts with SIRT1. Further confirmation of this data was obtained in a murine model of sepsis i.e., administration of PJ-34, a specific PARP1 inhibitor, led to decreased serum HMGB1 concentrations in mice subjected to CLP as compared to untreated mice. In conclusion, our study provides new insights in understanding the molecular mechanisms of HMGB1 secretion in sepsis.
... Intracellular PARP activity was measured by cell ELISA as Bakondi et al. (18) described. A total of 5 3 10 4 BMDMs were planted in 96-well plates. ...
Article
The high-mobility group box protein 1 (HMGB1) is increasingly recognized as an important inflammatory mediator. In some cases, the release of HMGB1 is regulated by poly(ADP-ribose) polymerase-1 (PARP-1), but the mechanism is still unclear. In this study, we report that PARP-1 activation contributes to LPS-induced PARylation of HMGB1, but the PARylation of HMGB1 is insufficient to direct its migration from the nucleus to the cytoplasm; PARP-1 regulates the translocation of HMGB1 to the cytoplasm through upregulating the acetylation of HMGB1. In mouse bone marrow-derived macrophages, genetic and pharmacological inhibition of PARP-1 suppressed LPS-induced translocation and release of HMGB1. Increased PARylation was accompanied with the nucleus-to-cytoplasm translocation and release of HMGB1 upon LPS exposure, but PARylated HMGB1 was located at the nucleus, unlike acetylated HMGB1 localized at the cytoplasm in an import assay. PARP inhibitor and PARP-1 depletion decreased the activity ratio of histone acetyltransferases to histone deacetylases that elevated after LPS stimulation and impaired LPS-induced acetylation of HMGB1. In addition, PARylation of HMGB1 facilitates its acetylation in an in vitro enzymatic reaction. Furthermore, reactive oxygen species scavenger (N-acetyl-l-cysteine) and the ERK inhibitor (FR180204) impaired LPS-induced PARP activation and HMGB1 release. Our findings suggest that PARP-1 regulates LPS-induced acetylation of HMGB1 in two ways: PARylating HMGB1 to facilitate the latter acetylation and increasing the activity ratio of histone acetyltransferases to histone deacetylases. These studies revealed a new mechanism of PARP-1 in regulating the inflammatory response to endotoxin.