parp activity
Recently Published Documents


TOTAL DOCUMENTS

95
(FIVE YEARS 27)

H-INDEX

19
(FIVE YEARS 3)

Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 322-322
Author(s):  
Dang Hai Nguyen ◽  
Zhiyan Silvia Liu ◽  
Sayantani Sinha ◽  
Maxwell Bannister ◽  
Erica Arriaga-Gomez ◽  
...  

Abstract Somatic heterozygous mutations in spliceosome genes SRSF2, U2AF1, and SF3B1 commonly occur in patients with myeloid malignancies such as myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Moreover, SRSF2 and U2AF1 mutations associate with poor survival and high risk of progression to AML, representing a unique genetic vulnerability for targeted therapy. We and others previously found that R-loops, a group of transcription intermediates containing RNA:DNA hybrids and displaced single-stranded DNA, are a source of genomic instability induced by different spliceosome mutants. We further showed that inhibition of ATR kinase activity preferentially kills spliceosome mutant cells in an R-loop-dependent manner. Inspired by ATR inhibition results, we performed a focused drug screen with inhibitors targeting additional DNA damage response pathways to identify novel therapeutic vulnerabilities generated by spliceosome mutations. We generated a murine leukemia model by overexpressing the MLL-AF9 fusion oncogene on an Srsf2 P95H/+background, a mutational combination that is found in ~10% of MLL-rearranged leukemias. Surprisingly, we found that Srsf2 P95H/+cells are more sensitive to five inhibitors targeting ADP-ribosyltransferases or PARP (olaparib, talazoparib, rucaparib, niraparib, veliparib) (Figs 1A-B). Olaparib (PARPi)-treated Srsf2 P95H/+cells exhibited increased apoptosis compared to Srsf2 +/+ cells as determined by AnnexinV (Fig 1C). PARPi sensitivity was also observed in isogenic murine MLL-AF9 U2af1 S34F/+cells compared to MLL-AF9 U2af1 +/+ cells (Fig 1D). These data highlight that both SRSF2 P95H and U2AF1 S34F mutations create a common vulnerability that is dependent on PARP activity for survival. To evaluate PARP activity in cells, we used isogenic K562 leukemia cells expressing SRSF2 P95H and U2AF1 S34F mutations from their endogenous loci and monitored PAR (poly(ADP-ribose)) chain levels, a marker of PARP activity. Both SRSF2 P95H and U2AF1 S34F cells exhibited elevated PAR levels compared to wildtype cells (Figs 1E-F). PARPi treatment significantly suppressed PAR signals in SRSF2 P95H and U2AF1 S34F cells. PARP inhibitors target both PARP1 and PARP2 enzymes, of which PARP1 plays a key role in DNA damage response. We used CRISPR-Cas9 to knockout PARP1 gene to determine the major PARP responsible for elevated PAR level in these leukemia cells. PARP1 deletion abrogated elevated PAR levels in U2AF1 S34F (Fig 1G) and SRSF2 P95H cells (data not shown). Altogether, we demonstrated that SRSF2 P95H and U2AF1 S34F cells trigger a PARP1 response critical for cell survival. To test whether increased PAR level arises from U2AF1 S34F-induced R-loops, we generated U2AF1 S34F cells that inducibly express RNaseH1, an enzyme that specifically cleaves the RNA moiety within RNA:DNA hybrids. Induction of RNaseH1 in U2AF1 S34F cells significantly reduced PAR levels, showing that U2AF1 S34F-induced PAR chains is R-loop-dependent (Fig 1H). Moreover, RNaseH1 overexpression suppressed the growth inhibition of PARPi-treated U2AF1 S34F cells (Fig 1I). Collectively, these results suggest that U2AF1 S34F mutants induce R-loop accumulation and elicit an R-loop-associated PARP1 signaling to promote cell survival. We next tested whether combining ATR inhibitor (ATRi) can further exacerbate PARPi sensitivity in spliceosome mutant cells. To examine ATR activity, we monitored phosphorylated RPA (Replication Protein A, or pRPA), a known ATR substrate. pRPA level was enhanced in PARPi-treated SRSF2 P95H cells compared to PARPi-treated SRSF2 WT cells but was suppressed when treated with ATRi (Fig 1J), suggesting that splicing factor mutant cells are more reliant on ATR function in the context of PARPi. Importantly, the combination of PARPi with ATRi, but not with ATMi, significantly promoted cell growth inhibition in SRSF2 P95H cells compared to SRSF2 WT cells or to SRSF2 P95H cells treated with individual compounds alone (Fig 1K). Collectively, these data provide a pre-clinical rationale that splicing factor mutant leukemias are preferentially sensitive to PARP1 modulation compared to their wildtype counterpart. Moreover, combining PARPi and ATRi may further sensitize spliceosome mutant cells and could represent a new therapeutic strategy in myeloid leukemia patients harboring these mutations (Fig 1L). Figure 1 Figure 1. Disclosures Graubert: Calico: Research Funding; Janssen: Research Funding; astrazeneca: Research Funding.


Author(s):  
JUN TANG ◽  
Xuejie Wang ◽  
Liyun Qin ◽  
Coral Ampurdanés ◽  
Xavier Duran ◽  
...  
Keyword(s):  

Author(s):  
Nicolas C. Hoch

The COVID-19 pandemic has prompted intense research efforts into elucidating mechanisms of coronavirus pathogenesis and to propose antiviral interventions. The interferon (IFN) response is the main antiviral component of human innate immunity and is actively suppressed by several non-structural SARS-CoV-2 proteins, allowing viral replication within human cells. Differences in IFN signalling efficiency and timing have emerged as central determinants of the variability of COVID-19 disease severity between patients, highlighting the need for an improved understanding of host–pathogen interactions that affect the IFN response. ADP-ribosylation is an underexplored post-translational modification catalyzed by ADP-ribosyl transferases collectively termed poly(ADP-ribose) polymerases (PARPs). Several human PARPs are induced by the IFN response and participate in antiviral defences by regulating IFN signalling itself, modulating host processes such as translation and protein trafficking, as well as directly modifying and inhibiting viral target proteins. SARS-CoV-2 and other viruses encode a macrodomain that hydrolyzes ADP-ribose modifications, thus counteracting antiviral PARP activity. This mini-review provides a brief overview of the known targets of IFN-induced ADP-ribosylation and the functions of viral macrodomains, highlighting several open questions in the field.


2021 ◽  
Author(s):  
Alina Vaitsiankova ◽  
Kamila Burdova ◽  
Hana Hanzlikova ◽  
Keith Caldecott

PARP1 is implicated in the detection and repair of unligated Okazaki fragment intermediates, highlighting these structures as a potential source of genome breakage induced by PARP inhibition. In agreement with this, we show here that PARP1 activity is greatly elevated in chicken and human S phase cells in which FEN1 nuclease is genetically deleted, and that PARP activity is highest tens of kilobases behind DNA replication forks. Importantly, PARP inhibitor reduces the integrity of nascent DNA strands in both wild type chicken and human cells during DNA replication, and does so in FEN1-/- cells to an even greater extent that can be detected as post-replicative single-strand gaps within individual DNA fibres. Collectively, these data show that PARP inhibitors impede the maturation of Okazaki fragments in nascent DNA, implicating these canonical DNA replication intermediates in the cytotoxicity of these compounds.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. TPS5089-TPS5089
Author(s):  
Neeraj Agarwal ◽  
Arun Azad ◽  
Neal D. Shore ◽  
Joan Carles ◽  
Andre P. Fay ◽  
...  

TPS5089 Background: TALA blocks poly(ADP-ribose) polymerase (PARP) activity and traps PARP on single-strand DNA breaks, preventing DNA damage repair (DDR) and causing death of cells with DDR alterations (eg, BRCA1/2).a TALA is approved in multiple countries as monotherapy for germline BRCA1/2-mutated human epidermal growth factor receptor 2 (HER2)-negative advanced breast cancer. Olaparib and rucaparib are PARP inhibitors approved for use in mCRPC. ENZA is an androgen receptor (AR) inhibitor and an established therapy for mCRPC. As PARP activity has been shown to support AR function, inhibition of PARP is expected to increase sensitivity to AR-directed therapies. In addition, AR blockade downregulates homologous recombination repair gene transcription, which induces a “ BRCAness” phenotype. A proof-of-concept study combining olaparib and abiraterone (abi) in pts with mCRPC demonstrated improved median radiographic progression-free survival (rPFS) vs placebo plus abi (13.8 vs 8.2 months) and a tolerable safety profile. Therefore, ENZA may be efficacious regardless of DDR alterations. TALAPRO-2 (NCT03395197) is a Phase 3, 2-part study evaluating the efficacy, safety, pharmacokinetics, and patient-reported outcomes (PROs) of TALA plus ENZA in pts with mCRPC with or without DDR alterations. Methods: Enrollment goal is 1037 patients (pts; 19 pts, part 1 dose-finding [completed]; 1018 pts, part 2 placebo-controlled [ongoing; accrual completed in unselected cohort]). Key eligibility criteria: age ≥18 years; asymptomatic/mildly symptomatic mCRPC; ECOG performance status ≤1; metastatic disease (no brain metastases); and no prior life-prolonging systemic therapy for nonmetastatic CRPC or mCRPC. Prior therapies (excluding novel AR inhibitors) in the castration-sensitive (CSPC) setting are allowed. ADT must continue throughout the study. The randomized double-blind portion (part 2) will evaluate safety, efficacy, and PROs of TALA (0.5 mg once daily [QD]) + ENZA (160 mg QD) vs placebo + ENZA (160 mg QD). Pts are stratified by prior novel hormonal therapy or docetaxel for CSPC or mCSPC (yes or no) and DDR alteration status (deficient vs nondeficient/unknown). The primary endpoint is rPFS, defined as time to progression in soft tissue per RECIST v.1.1 or in bone per PCWG3 criteria by independent central review or death. The key secondary endpoint is overall survival. Efficacy is assessed radiographically every 8 weeks up to Week 25 and every 8–12 weeks thereafter. rPFS will be compared between the two arms by a one-sided stratified log-rank test. Pt recruitment is ongoing at 223 sites in 26 countries, including 32 states across the US, and Europe, Israel, South America, South Africa, and Asia-Pacific region. aDDR alterations are defined as known/likely pathogenic variants or homozygous deletions. Clinical trial information: NCT03395197.


2021 ◽  
Author(s):  
Scott H Greenwald ◽  
Emily E Brown ◽  
Michael J Scandura ◽  
Erin Hennessey ◽  
Raymond Farmer ◽  
...  

Abstract Nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) is required for nuclear nicotinamide adenine mononucleotide (NAD+) biosynthesis in all nucleated cells, and despite its functional ubiquity, mutations in this gene lead to an isolated retinal degeneration. The mechanisms underlying how mutant NMNAT1 causes disease are not well understood, nor is the reason why the pathology is confined to the retina. Using a mouse model of NMNAT1-associated retinal degeneration that harbors the p.Val9Met mutation, we tested the hypothesis that decreased function of mutant NMNAT1 has a greater effect on the levels of NAD+ in the retina than elsewhere in the body. Measurements by liquid chromatography with tandem mass spectrometry showed an early and sustained decrease of NAD+ in mutant retinas that was not observed in other tissues. To understand how consumers of nuclear NAD+ are affected by the reduced availability of NAD+ in mutant retinas, poly(ADP-ribose) polymerase (PARP) and nuclear sirtuin activity were evaluated. PARP activity was elevated during disease progression, as evidenced by overproduction of poly(ADP-ribose) (PAR) in photoreceptors, whereas histone deacetylation activity of nuclear sirtuins was not altered. We hypothesized that PARP could be activated because of elevated levels of oxidative stress; however, we did not observe oxidative DNA damage, lipid peroxidation, or a low glutathione to oxidized glutathione ratio. Terminal deoxynucleotidyl transferase dUTP nick end labeling staining revealed that photoreceptors appear to ultimately die by apoptosis, although the low NAD+ levels and overproduction of PAR suggest that cell death may include aspects of the parthanatos cell death pathway.


2021 ◽  
Vol 204 ◽  
pp. 108448
Author(s):  
Furong Gao ◽  
Zongyi Li ◽  
Ziwei Kang ◽  
Dandan Liu ◽  
Peng Li ◽  
...  

2021 ◽  
pp. 166949
Author(s):  
Nicolás Luis Calzetta ◽  
Marina Alejandra González Besteiro ◽  
Vanesa Gottifredi

2021 ◽  
Vol 2 (1) ◽  
pp. 100344
Author(s):  
Ilsa T. Kirby ◽  
Daniel J. Sanderson ◽  
Michael S. Cohen

2021 ◽  
Author(s):  
Magali VanLinden ◽  
Lena Høyland ◽  
Jörn Dietze ◽  
Ingvill Tolås ◽  
Lars Sverkeli ◽  
...  

Abstract The coenzyme NAD is consumed by signaling enzymes including poly-ADP-ribose-polymerases (PARPs) and sirtuins. Understanding the mechanisms of aging-associated NAD decline and how cells cope with decreased NAD concentrations requires model systems reflecting chronic NAD deficiency. To evoke compartment-specific over-consumption of NAD, we have engineered cell lines expressing PARP activity in mitochondria, the cytosol, endoplasmic reticulum, or peroxisomes. Irrespective of the compartment targeted, total cellular NAD concentrations declined by ~40%. Isotope-tracer flux measurements and mathematical modeling showed that the lowered NAD concentration limits total NAD consumption kinetically. Moreover, NAD biosynthesis rate and capacity remained unchanged, thereby also precluding an increase of total NAD turnover. The chronic NAD deficiency was surprisingly well tolerated unless the mitochondria were targeted. Oxidative phosphorylation and glycolysis were little affected by NAD over-consumption in the other compartments. Likewise, peroxisomal NAD over-consumption was balanced by mitochondrial NAD decrease to maintain beta-oxidation of very long chain fatty acids in peroxisomes. We propose that subcellular NAD pools are interconnected, with mitochondria acting as a rheostat to facilitate NAD-dependent processes in organelles with excessive consumption.


Sign in / Sign up

Export Citation Format

Share Document