Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.827
Filtrer
1.
Biochemistry (Mosc) ; 89(7): 1202-1210, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39218019

RÉSUMÉ

Poly(ADP-ribose) polymerase 1 (PARP1) plays a major role in the DNA damage repair and transcriptional regulation, and is targeted by a number of clinical inhibitors. Despite this, catalytic mechanism of PARP1 remains largely underexplored because of the complex substrate/product structure. Using molecular modeling and metadynamics simulations we have described in detail elongation of poly(ADP-ribose) chain in the PARP1 active site. It was shown that elongation reaction proceeds via the SN1-like mechanism involving formation of the intermediate furanosyl oxocarbenium ion. Intriguingly, nucleophilic 2'A-OH group of the acceptor substrate can be activated by the general base Glu988 not directly but through the proton relay system including the adjacent 3'A-OH group.


Sujet(s)
Poly (ADP-Ribose) polymerase-1 , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/composition chimique , Humains , Modèles moléculaires , Simulation de dynamique moléculaire , Domaine catalytique , Poly adénosine diphosphate ribose/métabolisme , Poly adénosine diphosphate ribose/composition chimique
2.
J Nutr Sci Vitaminol (Tokyo) ; 70(4): 295-304, 2024.
Article de Anglais | MEDLINE | ID: mdl-39218690

RÉSUMÉ

Cellular NAD+ is continuously degraded and synthesized under resting conditions. In mammals, NAD+ synthesis is primarily initiated from nicotinamide (Nam) by Nam phosphoribosyltransferase, whereas poly(ADP-ribose) polymerase 1 (PARP1) and 2 (PARP2), sirtuin1 (SIRT1), CD38, and sterile alpha and TIR motif containing 1 (SARM1) are involved in NAD+ breakdown. Using flux analysis with 2H-labeled Nam, we found that when mammalian cells were cultured in the absence of Nam, cellular NAD+ levels were maintained and NAD+ breakdown was completely suppressed. In the presence of Nam, the rate of NAD+ breakdown (RB) did not significantly change upon PARP1, PARP2, SIRT1, or SARM1 deletion, whereas stable expression of CD38 did not increase RB. However, RB in PARP1-deleted cells was much higher compared with that in wild-type cells, in which PARP1 activity was blocked with a selective inhibitor. In contrast, RB in CD38-overexpressing cells in the presence of a specific CD38 inhibitor was much lower compared with that in control cells. The results indicate that PARP1 deletion upregulates the activity of other NADases, whereas CD38 expression downregulates the activity of endogenous NADases, including PARP1 and PARP2. The rate of cellular NAD+ breakdown and the resulting NAD+ concentration may be maintained at a constant level, despite changes in the NAD+-degrading enzyme expression, through the compensatory regulation of NADase activity.


Sujet(s)
Antigènes CD38 , NAD , Poly (ADP-Ribose) polymerase-1 , Sirtuine-1 , NAD/métabolisme , Antigènes CD38/métabolisme , Antigènes CD38/génétique , Animaux , Poly (ADP-Ribose) polymerase-1/métabolisme , Sirtuine-1/métabolisme , Sirtuine-1/génétique , Nicotinamide/pharmacologie , Nicotinamide/métabolisme , Souris , Poly(ADP-ribose) polymerases/métabolisme , Humains , Nicotinamide phosphoribosyltransferase/métabolisme , Nicotinamide phosphoribosyltransferase/génétique , Délétion de gène
3.
BMC Biol ; 22(1): 188, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39218869

RÉSUMÉ

BACKGROUND: The histone variant macroH2A (mH2A), the most deviant variant, is about threefold larger than the conventional histone H2A and consists of a histone H2A-like domain fused to a large Non-Histone Region responsible for recruiting PARP-1 to chromatin. The available data suggest that the histone variant mH2A participates in the regulation of transcription, maintenance of heterochromatin, NAD+ metabolism, and double-strand DNA repair. RESULTS: Here, we describe a novel function of mH2A, namely its implication in DNA oxidative damage repair through PARP-1. The depletion of mH2A affected both repair and cell survival after the induction of oxidative lesions in DNA. PARP-1 formed a specific complex with mH2A nucleosomes in vivo. The mH2A nucleosome-associated PARP-1 is inactive. Upon oxidative damage, mH2A is ubiquitinated, PARP-1 is released from the mH2A nucleosomal complex, and is activated. The in vivo-induced ubiquitination of mH2A, in the absence of any oxidative damage, was sufficient for the release of PARP-1. However, no release of PARP-1 was observed upon treatment of the cells with either the DNA alkylating agent MMS or doxorubicin. CONCLUSIONS: Our data identify a novel pathway for the repair of DNA oxidative lesions, requiring the ubiquitination of mH2A for the release of PARP-1 from chromatin and its activation.


Sujet(s)
Altération de l'ADN , Réparation de l'ADN , Histone , Poly (ADP-Ribose) polymerase-1 , Ubiquitination , Histone/métabolisme , Histone/génétique , Humains , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Stress oxydatif , Nucléosomes/métabolisme
4.
Nat Commun ; 15(1): 6641, 2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-39103378

RÉSUMÉ

DNA-protein crosslinks (DPCs) are toxic lesions that inhibit DNA related processes. Post-translational modifications (PTMs), including SUMOylation and ubiquitylation, play a central role in DPC resolution, but whether other PTMs are also involved remains elusive. Here, we identify a DPC repair pathway orchestrated by poly-ADP-ribosylation (PARylation). Using Xenopus egg extracts, we show that DPCs on single-stranded DNA gaps can be targeted for degradation via a replication-independent mechanism. During this process, DPCs are initially PARylated by PARP1 and subsequently ubiquitylated and degraded by the proteasome. Notably, PARP1-mediated DPC resolution is required for resolving topoisomerase 1-DNA cleavage complexes (TOP1ccs) induced by camptothecin. Using the Flp-nick system, we further reveal that in the absence of PARP1 activity, the TOP1cc-like lesion persists and induces replisome disassembly when encountered by a DNA replication fork. In summary, our work uncovers a PARP1-mediated DPC repair pathway that may underlie the synergistic toxicity between TOP1 poisons and PARP inhibitors.


Sujet(s)
Réparation de l'ADN , Réplication de l'ADN , ADN topoisomérases de type I , Poly (ADP-Ribose) polymerase-1 , Poly(ADP-ribosylation) , Animaux , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , ADN topoisomérases de type I/métabolisme , Xenopus laevis , Ubiquitination , Humains , ADN/métabolisme , Altération de l'ADN , Camptothécine/pharmacologie , Maturation post-traductionnelle des protéines , ADN simple brin/métabolisme , Protéines de Xénope/métabolisme
5.
Biochem J ; 481(17): 1097-1123, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39178157

RÉSUMÉ

ADP-ribosylation is a prominent and versatile post-translational modification, which regulates a diverse set of cellular processes. Poly-ADP-ribose (PAR) is synthesised by the poly-ADP-ribosyltransferases PARP1, PARP2, tankyrase (TNKS), and tankyrase 2 (TNKS2), all of which are linked to human disease. PARP1/2 inhibitors have entered the clinic to target cancers with deficiencies in DNA damage repair. Conversely, tankyrase inhibitors have continued to face obstacles on their way to clinical use, largely owing to our limited knowledge of their molecular impacts on tankyrase and effector pathways, and linked concerns around their tolerability. Whilst detailed structure-function studies have revealed a comprehensive picture of PARP1/2 regulation, our mechanistic understanding of the tankyrases lags behind, and thereby our appreciation of the molecular consequences of tankyrase inhibition. Despite large differences in their architecture and cellular contexts, recent structure-function work has revealed striking parallels in the regulatory principles that govern these enzymes. This includes low basal activity, activation by intra- or inter-molecular assembly, negative feedback regulation by auto-PARylation, and allosteric communication. Here we compare these poly-ADP-ribosyltransferases and point towards emerging parallels and open questions, whose pursuit will inform future drug development efforts.


Sujet(s)
Poly (ADP-Ribose) polymerase-1 , Tankyrases , Tankyrases/métabolisme , Tankyrases/antagonistes et inhibiteurs , Tankyrases/génétique , Tankyrases/composition chimique , Humains , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/génétique , Poly(ADP-ribose) polymerases/métabolisme , Poly(ADP-ribose) polymerases/composition chimique , Poly(ADP-ribose) polymerases/génétique , Animaux , Maturation post-traductionnelle des protéines , Réparation de l'ADN , ADP-Ribosylation , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly(ADP-ribosylation)/génétique
6.
Epigenetics Chromatin ; 17(1): 26, 2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39118189

RÉSUMÉ

Poly (ADP-ribose) polymerase 1 (PARP1) is a multifunctional nuclear enzyme that catalyzes poly-ADP ribosylation in eukaryotic cells. In addition to maintaining genomic integrity, this nuclear enzyme is also involved in transcriptional regulation. PARP1 can trigger and maintain changes in the chromatin structure and directly recruit transcription factors. PARP1 also prevents DNA methylation. However, most previous reviews on PARP1 have focused on its involvement in maintaining genome integrity, with less focus on its transcriptional regulatory function. This article comprehensively reviews the transcriptional regulatory function of PARP1 and its application in disease treatment, providing new ideas for targeting PARP1 for the treatment of diseases other than cancer.


Sujet(s)
Poly (ADP-Ribose) polymerase-1 , Transcription génétique , Humains , Poly (ADP-Ribose) polymerase-1/métabolisme , Animaux , Tumeurs/génétique , Tumeurs/métabolisme , Régulation de l'expression des gènes , Méthylation de l'ADN , Chromatine/métabolisme
7.
Cell Rep ; 43(8): 114626, 2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39167487

RÉSUMÉ

The majority of severe early-onset and juvenile cases of amyotrophic lateral sclerosis (ALS) are caused by mutations in the FUS gene, resulting in rapid disease progression. Mutant FUS accumulates within stress granules (SGs), thereby affecting the dynamics of these ribonucleoprotein complexes. Here, we define the interactome of the severe mutant FUSP525L variant in human induced pluripotent stem cell (iPSC)-derived motor neurons. We find increased interaction of FUSP525L with the PARP1 enzyme, promoting poly-ADP-ribosylation (PARylation) and binding of FUS to histone H1.2. Inhibiting PARylation or reducing H1.2 levels alleviates mutant FUS aggregation, SG alterations, and apoptosis in human motor neurons. Conversely, elevated H1.2 levels exacerbate FUS-ALS phenotypes, driven by the internally disordered terminal domains of H1.2. In C. elegans models, knockdown of H1.2 and PARP1 orthologs also decreases FUSP525L aggregation and neurodegeneration, whereas H1.2 overexpression worsens ALS-related changes. Our findings indicate a link between PARylation, H1.2, and FUS with potential therapeutic implications.


Sujet(s)
Sclérose latérale amyotrophique , Caenorhabditis elegans , Histone , Mutation , Poly (ADP-Ribose) polymerase-1 , Protéine FUS de liaison à l'ARN , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/anatomopathologie , Humains , Histone/métabolisme , Protéine FUS de liaison à l'ARN/métabolisme , Protéine FUS de liaison à l'ARN/génétique , Caenorhabditis elegans/métabolisme , Caenorhabditis elegans/génétique , Animaux , Mutation/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Motoneurones/métabolisme , Motoneurones/anatomopathologie , Poly(ADP-ribosylation) , Cellules souches pluripotentes induites/métabolisme , Liaison aux protéines
8.
Int J Mol Sci ; 25(16)2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39201718

RÉSUMÉ

Poly (ADP-Ribose) Polymerase (PARP) inhibitors have changed the outcomes and therapeutic strategy for several cancer types. As a targeted therapeutic mainly for patients with BRCA1/2 mutations, PARP inhibitors have commonly been exploited for their capacity to prevent DNA repair. In this review, we discuss the multifaceted roles of PARP-1 and PARP-2 beyond DNA repair, including the impact of PARP-1 on chemokine signalling, immune modulation, and transcriptional regulation of gene expression, particularly in the contexts of angiogenesis and epithelial-to-mesenchymal transition (EMT). We evaluate the pre-clinical role of PARP inhibitors, either as single-agent or combination therapies, to block the metastatic process. Efficacy of PARP inhibitors was demonstrated via DNA repair-dependent and independent mechanisms, including DNA damage, cell migration, invasion, initial colonization at the metastatic site, osteoclastogenesis, and micrometastasis formation. Finally, we summarize the recent clinical advancements of PARP inhibitors in the prevention and progression of distant metastases, with a particular focus on specific metastatic sites and PARP-1 selective inhibitors. Overall, PARP inhibitors have demonstrated great potential in inhibiting the metastatic process, pointing the way for greater use in early cancer settings.


Sujet(s)
Métastase tumorale , Tumeurs , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Poly(ADP-ribose) polymerases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Tumeurs/traitement médicamenteux , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Tumeurs/génétique , Poly(ADP-ribose) polymerases/métabolisme , Animaux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Réparation de l'ADN/effets des médicaments et des substances chimiques
9.
Genome Med ; 16(1): 107, 2024 Aug 26.
Article de Anglais | MEDLINE | ID: mdl-39187844

RÉSUMÉ

BACKGROUND: Poly (ADP-ribose) polymerase 1 and 2 (PARP1/2) inhibitors (PARPi) are targeted therapies approved for homologous recombination repair (HRR)-deficient breast, ovarian, pancreatic, and prostate cancers. Since inhibition of PARP1 is sufficient to cause synthetic lethality in tumors with homologous recombination deficiency (HRD), PARP1 selective inhibitors such as saruparib (AZD5305) are being developed. It is expected that selective PARP1 inhibition leads to a safer profile that facilitates its combination with other DNA damage repair inhibitors. Here, we aimed to characterize the antitumor activity of AZD5305 in patient-derived preclinical models compared to the first-generation PARP1/2 inhibitor olaparib and to identify mechanisms of resistance. METHODS: Thirteen previously characterized patient-derived tumor xenograft (PDX) models from breast, ovarian, and pancreatic cancer patients harboring germline pathogenic alterations in BRCA1, BRCA2, or PALB2 were used to evaluate the efficacy of AZD5305 alone or in combination with carboplatin or an ataxia telangiectasia and Rad3 related (ATR) inhibitor (ceralasertib) and compared it to the first-generation PARPi olaparib. We performed DNA and RNA sequencing as well as protein-based assays to identify mechanisms of acquired resistance to either PARPi. RESULTS: AZD5305 showed superior antitumor activity than the first-generation PARPi in terms of preclinical complete response rate (75% vs. 37%). The median preclinical progression-free survival was significantly longer in the AZD5305-treated group compared to the olaparib-treated group (> 386 days vs. 90 days). Mechanistically, AZD5305 induced more replication stress and genomic instability than the PARP1/2 inhibitor olaparib in PARPi-sensitive tumors. All tumors at progression with either PARPi (39/39) showed increase of HRR functionality by RAD51 foci formation. The most prevalent resistance mechanisms identified were the acquisition of reversion mutations in BRCA1/BRCA2 and the accumulation of hypomorphic BRCA1. AZD5305 did not sensitize PDXs with acquired resistance to olaparib but elicited profound and durable responses when combined with carboplatin or ceralasertib in 3/6 and 5/5 models, respectively. CONCLUSIONS: Collectively, these results show that the novel PARP1 selective inhibitor AZD5305 yields a potent antitumor response in PDX models with HRD and delays PARPi resistance alone or in combination with carboplatin or ceralasertib, which supports its use in the clinic as a new therapeutic option.


Sujet(s)
Protéine BRCA1 , Protéine BRCA2 , Inhibiteurs de poly(ADP-ribose) polymérases , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Animaux , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Souris , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Femelle , Phtalazines/pharmacologie , Phtalazines/usage thérapeutique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Indoles/usage thérapeutique , Indoles/pharmacologie , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Carboplatine/pharmacologie , Carboplatine/usage thérapeutique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique
10.
Cell Death Dis ; 15(8): 610, 2024 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-39174499

RÉSUMÉ

PARP1 is crucial in DNA damage repair, chromatin remodeling, and transcriptional regulation. The principle of synthetic lethality has effectively guided the application of PARP inhibitors in treating tumors carrying BRCA1/2 mutations. Meanwhile, PARP inhibitors have exhibited efficacy in BRCA-proficient patients, further highlighting the necessity for a deeper understanding of PARP1 function and its inhibition in cancer therapy. Here, we unveil PIN2/TRF1-interacting telomerase inhibitor 1 (PINX1) as an uncharacterized PARP1-interacting protein that synergizes with PARP inhibitors upon its depletion across various cancer cell lines. Loss of PINX1 compromises DNA damage repair capacity upon etoposide treatment. The vulnerability of PINX1-deficient cells to etoposide and PARP inhibitors could be effectively restored by introducing either a full-length or a mutant form of PINX1 lacking telomerase inhibitory activity. Mechanistically, PINX1 is recruited to DNA lesions through binding to the ZnF3-BRCT domain of PARP1, facilitating the downstream recruitment of the DNA repair factor XRCC1. In the absence of DNA damage, PINX1 constitutively binds to PARP1, promoting PARP1-chromatin association and transcription of specific DNA damage repair proteins, including XRCC1, and transcriptional regulators, including GLIS3. Collectively, our findings identify PINX1 as a multifaceted partner of PARP1, crucial for safeguarding cells against genotoxic stress and emerging as a potential candidate for targeted tumor therapy.


Sujet(s)
Protéines du cycle cellulaire , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Lignée cellulaire tumorale , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Protéines suppresseurs de tumeurs/métabolisme , Protéines suppresseurs de tumeurs/génétique , Altération de l'ADN , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Réparation de l'ADN/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/anatomopathologie , Tumeurs/métabolisme , Étoposide/pharmacologie , Protéine-1 de complémentation croisée de la réparation des lésions induites par les rayons X
11.
Cells ; 13(16)2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39195238

RÉSUMÉ

Uveal melanoma (UM) is the most common primary intraocular tumor in adults, with no standardized treatment for advanced disease. Based on preliminary bioinformatical analyses DTYMK and PARP1 were selected as potential therapeutic targets. High levels of both proteins were detected in uveal melanoma cells and correlated with increased tumor growth and poor prognosis. In vitro tests on MP41 (BAP1 positive) and MP46 (BAP1 negative) cancer cell lines using inhibitors pamiparib (PARP1) and Ymu1 (DTYMK) demonstrated significant cytotoxic effects. Combined treatment had synergistic effects in MP41 and additive in MP46 cell lines, reducing cell proliferation and inhibiting the mTOR signaling pathway. Furthermore, the applied inhibitors in combination decreased cell motility and migration speed, especially for BAP1-negative cell lines. Our hypothesis of the double hit into tumoral DNA metabolism as a possible therapeutic option in uveal melanoma was confirmed since combined targeting of DTYMK and PARP1 affected all tested cytophysiological parameters with the highest efficiency. Our in vitro findings provide insights into novel therapeutic avenues for managing uveal melanoma, warranting further exploration in preclinical and clinical settings.


Sujet(s)
Prolifération cellulaire , Mélanome , Poly (ADP-Ribose) polymerase-1 , Tumeurs de l'uvée , Humains , Tumeurs de l'uvée/traitement médicamenteux , Tumeurs de l'uvée/anatomopathologie , Tumeurs de l'uvée/métabolisme , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Mélanome/métabolisme , Lignée cellulaire tumorale , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique
12.
CNS Neurosci Ther ; 30(9): e70012, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39215404

RÉSUMÉ

AIMS: Poly (ADP-ribose) polymerase (PARP) has been extensively investigated in human cancers. Recent studies verified that current available PARP inhibitors (Olaparib or Veliparib) provided clinical palliation of clinical patients suffering from paclitaxel-induced neuropathic pain (PINP). However, the underlying mechanism of PARP overactivation in the development of PINP remains to be investigated. METHODS AND RESULTS: We reported induction of DNA oxidative damage, PARP-1 overactivation, and subsequent nicotinamide adenine dinucleotide (NAD+) depletion as crucial events in the pathogenesis of PINP. Therefore, we developed an Olaparib PROTAC to achieve the efficient degradation of PARP. Continuous intrathecal injection of Olaparib PROTAC protected against PINP by inhibiting the activity of PARP-1 in rats. PARP-1, but not PARP-2, was shown to be a crucial enzyme in the development of PINP. Specific inhibition of PARP-1 enhanced mitochondrial redox metabolism partly by upregulating the expression and deacetylase activity of sirtuin-3 (SIRT3) in the dorsal root ganglions and spinal cord in the PINP rats. Moreover, an increase in the NAD+ level was found to be a crucial mechanism by which PARP-1 inhibition enhanced SIRT3 activity. CONCLUSION: The findings provide a novel insight into the mechanism of DNA oxidative damage in the development of PINP and implicate PARP-1 as a possible therapeutic target for clinical PINP treatment.


Sujet(s)
Altération de l'ADN , Mitochondries , Névralgie , Paclitaxel , Poly (ADP-Ribose) polymerase-1 , Animaux , Mâle , Rats , Modèles animaux de maladie humaine , Altération de l'ADN/effets des médicaments et des substances chimiques , Ganglions sensitifs des nerfs spinaux/effets des médicaments et des substances chimiques , Ganglions sensitifs des nerfs spinaux/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , NAD/métabolisme , Névralgie/induit chimiquement , Névralgie/métabolisme , Névralgie/traitement médicamenteux , Stress oxydatif/effets des médicaments et des substances chimiques , Paclitaxel/toxicité , Phtalazines/pharmacologie , Pipérazines/pharmacologie , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Rat Sprague-Dawley , Moelle spinale/effets des médicaments et des substances chimiques , Moelle spinale/métabolisme
13.
Immunohorizons ; 8(8): 586-597, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-39186692

RÉSUMÉ

Neutrophil extracellular traps (NETs) function to control infectious agents as well as to propagate inflammatory response in a variety of disease conditions. DNA damage associated with chromatin decondensation and NACHT domain-leucine-rich repeat-and pyrin domain-containing protein 3 (NLRP3) inflammasome activation have emerged as crucial events in NET formation, but the link between the two processes is unknown. In this study, we demonstrate that poly(ADP-ribose) polymerase-1 (PARP-1), a key DNA repair enzyme, regulates NET formation triggered by NLRP3 inflammasome activation in neutrophils. Activation of mouse neutrophils with canonical NLRP3 stimulants LPS and nigericin induced NET formation, which was significantly abrogated by pharmacological inhibition of PARP-1. We found that PARP-1 is required for NLRP3 inflammasome assembly by regulating post-transcriptional levels of NLRP3 and ASC dimerization. Importantly, this PARP-1-regulated NLRP3 activation for NET formation was independent of inflammasome-mediated pyroptosis, because caspase-1 and gasdermin D processing as well as IL-1ß transcription and secretion remained intact upon PARP-1 inhibition in neutrophils. Accordingly, pharmacological inhibition or genetic ablation of caspase-1 and gasdermin D had no effect on NLRP3-mediated NET formation. Mechanistically, PARP-1 inhibition increased p38 MAPK activity, which was required for downmodulation of NLRP3 and NETs, because concomitant inhibition of p38 MAPK with PARP-1 restored NLRP3 activation and NET formation. Finally, mice undergoing bacterial peritonitis exhibited increased survival upon treatment with PARP-1 inhibitor, which correlated with increased leukocyte influx and improved intracellular bacterial clearance. Our findings reveal a noncanonical pyroptosis-independent role of NLRP3 in NET formation regulated by PARP-1 via p38 MAPK, which can be targeted to control NETosis in inflammatory diseases.


Sujet(s)
Pièges extracellulaires , Inflammasomes , Protéine-3 de la famille des NLR contenant un domaine pyrine , Granulocytes neutrophiles , Poly (ADP-Ribose) polymerase-1 , Pyroptose , Animaux , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Pièges extracellulaires/métabolisme , Souris , Poly (ADP-Ribose) polymerase-1/métabolisme , Inflammasomes/métabolisme , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/immunologie , Souris de lignée C57BL , Nigéricine/pharmacologie , Souris knockout , Péritonite/métabolisme , Péritonite/immunologie , Lipopolysaccharides/pharmacologie , Caspase-1/métabolisme
14.
Physiol Rep ; 12(15): e16181, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39138135

RÉSUMÉ

This study aimed to evaluate the influence of combined intermittent fasting (IF) and high-intensity interval training (HIIT) on morphology, caspase-independent apoptosis signaling pathway, and myostatin expression in soleus and gastrocnemius (white portion) muscles from healthy rats. Sixty-day-old male Wistar rats (n = 60) were divided into four groups: control (C), IF, high-intensity-interval training (T), and high-intensity-interval training and intermittent fasting (T-IF). The C and T groups received ad libitum chow daily; IF and T-IF received the same standard chow every other day. Animals from T and T-IF underwent a HIIT protocol five times a week for 12 weeks. IF reduced gastrocnemius mass and increased pro-apoptotic proteins apoptosis-inducing factor (AIF) and endonuclease G (EndoG) in soleus and cleaved-to-non-cleaved PARP-1 ratio and myostatin expression in gastrocnemius white portion. HIIT increased AIF and apoptosis repressor with caspase recruitment domain expression in soleus and cleaved-to-total PARP-1 ratio in gastrocnemius muscle white portion. The combination of IF and HIIT reduced fiber cross-sectional area in both muscles, increased EndoG and AIF expression, and decreased cleaved-to-non-cleaved PARP-1 ratio in gastrocnemius muscle white portion. Muscle responses to IF and HIIT are directly impacted by the muscle fiber type composition and are modulated, at least in part, by myostatin and caspase-independent apoptosis signaling.


Sujet(s)
Facteur inducteur d'apoptose , Apoptose , Jeûne , Entrainement fractionné de haute intensité , Fibres musculaires à contraction lente , Amyotrophie , Myostatine , Rat Wistar , Transduction du signal , Animaux , Mâle , Apoptose/physiologie , Jeûne/métabolisme , Jeûne/physiologie , Myostatine/métabolisme , Entrainement fractionné de haute intensité/méthodes , Rats , Transduction du signal/physiologie , Amyotrophie/métabolisme , Amyotrophie/anatomopathologie , Facteur inducteur d'apoptose/métabolisme , Fibres musculaires à contraction lente/métabolisme , Fibres musculaires à contraction rapide/métabolisme , Fibres musculaires à contraction rapide/anatomopathologie , Endodeoxyribonucleases/métabolisme , Conditionnement physique d'animal/méthodes , Conditionnement physique d'animal/physiologie , Muscles squelettiques/métabolisme , Jeûne intermittent , Poly (ADP-Ribose) polymerase-1
15.
Eur J Med Chem ; 277: 116726, 2024 Nov 05.
Article de Anglais | MEDLINE | ID: mdl-39116535

RÉSUMÉ

Structural modification based on natural privileged scaffolds has proven to be an attractive approach to generate potential antitumor candidates with high potency and specific targeting. As a continuation of our efforts to identify potent PARP-1 inhibitors, natural 3-arylcoumarin scaffold was served as the starting point for the construction of novel structural unit for PARP-1 inhibition. Herein, a series of novel 8-carbamyl-3-arylcoumarin derivatives were designed and synthesized. The antiproliferative activities of target compounds against four BRCA-mutated cancer cells (SUM149PT, HCC1937, MDA-MB-436 and Capan-1) were evaluated. Among them, compound 9b exhibited excellent antiproliferative effects against SUM149PT, HCC1937 and Capan-1 cells with IC50 values of 0.62, 1.91 and 4.26 µM, respectively. Moreover, 9b could significantly inhibit the intracellular PARP-1/2 activity in SUM149PT cells with IC50 values of 2.53 nM and 6.45 nM, respectively. Further mechanism studies revealed that 9b could aggravate DNA double-strand breaks, increase ROS production, decrease mitochondrial membrane potential, arrest cell cycle at G2/M phase and ultimately induce apoptosis in SUM149PT cells. In addition, molecular docking study demonstrated that the binding mode of 9b with PARP-1 was similar to that of niraparib, forming multiple hydrogen bond interactions with the active site of PARP-1. Taken together, these findings suggest that 8-carbamyl-3-arylcoumarin scaffold could serve as an effective structural unit for PARP-1 inhibition and offer a valuable paradigm for the structural modification of natural products.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Coumarines , Tests de criblage d'agents antitumoraux , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Coumarines/pharmacologie , Coumarines/composition chimique , Coumarines/synthèse chimique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques , Structure moléculaire , Relation dose-effet des médicaments , Découverte de médicament , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Simulation de docking moléculaire , Produits biologiques/pharmacologie , Produits biologiques/composition chimique , Produits biologiques/synthèse chimique
16.
Proc Natl Acad Sci U S A ; 121(30): e2303642121, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39012819

RÉSUMÉ

Glutamyl-prolyl-tRNA synthetase (EPRS1) is a bifunctional aminoacyl-tRNA-synthetase (aaRS) essential for decoding the genetic code. EPRS1 resides, with seven other aaRSs and three noncatalytic proteins, in the cytoplasmic multi-tRNA synthetase complex (MSC). Multiple MSC-resident aaRSs, including EPRS1, exhibit stimulus-dependent release from the MSC to perform noncanonical activities distinct from their primary function in protein synthesis. Here, we show EPRS1 is present in both cytoplasm and nucleus of breast cancer cells with constitutively low phosphatase and tensin homolog (PTEN) expression. EPRS1 is primarily cytosolic in PTEN-expressing cells, but chemical or genetic inhibition of PTEN, or chemical or stress-mediated activation of its target, AKT, induces EPRS1 nuclear localization. Likewise, preferential nuclear localization of EPRS1 was observed in invasive ductal carcinoma that were also P-Ser473-AKT+. EPRS1 nuclear transport requires a nuclear localization signal (NLS) within the linker region that joins the catalytic glutamyl-tRNA synthetase and prolyl-tRNA synthetase domains. Nuclear EPRS1 interacts with poly(ADP-ribose) polymerase 1 (PARP1), a DNA-damage sensor that directs poly(ADP-ribosyl)ation (PARylation) of proteins. EPRS1 is a critical regulator of PARP1 activity as shown by markedly reduced ADP-ribosylation in EPRS1 knockdown cells. Moreover, EPRS1 and PARP1 knockdown comparably alter the expression of multiple tumor-related genes, inhibit DNA-damage repair, reduce tumor cell survival, and diminish tumor sphere formation by breast cancer cells. EPRS1-mediated regulation of PARP1 activity provides a mechanistic link between PTEN loss in breast cancer cells, PARP1 activation, and cell survival and tumor growth. Targeting the noncanonical activity of EPRS1, without inhibiting canonical tRNA ligase activity, provides a therapeutic approach potentially supplementing existing PARP1 inhibitors.


Sujet(s)
Tumeurs du sein , Noyau de la cellule , Poly (ADP-Ribose) polymerase-1 , Protéines proto-oncogènes c-akt , Humains , Tumeurs du sein/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Noyau de la cellule/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/génétique , Lignée cellulaire tumorale , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique , Amino acyl-tRNA synthetases/métabolisme , Amino acyl-tRNA synthetases/génétique , Transport nucléaire actif , Signaux de localisation nucléaire/métabolisme
17.
J Enzyme Inhib Med Chem ; 39(1): 2383886, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-39072709

RÉSUMÉ

Specifically inducing the degradation of acidic nucleoplasmic DNA-binding protein 1 (And1) is a promising antitumor strategy. Our previous study identified Bazedoxifene (BZA) and CH3 as specific And1 degraders and validated their activity in reversing radiotherapy resistance in vitro and in vivo. However, unelucidated structure-activity relationships and moderate activity have limited their application. In this study, 27 novel CH3 derivatives were designed and synthesised based on the cavity topology of the WD40 domain of And1. Among them, A15 with a "V" conformation significantly induced And1 degradation in NSCLC cells. In addition, this study demonstrated a potential synthetic lethal effect of And1 degraders and PARP1 inhibitors. 1 µM of Olaparib in combination with 5 µM of A15 significantly inhibited the proliferation of A549 and H460 cells. Overall, these compounds are valuable tools for elucidating And1 biology, and their special spatial conformation make them promising candidates for future optimisation studies.


Sujet(s)
Antinéoplasiques , Carcinome pulmonaire non à petites cellules , Prolifération cellulaire , Relation dose-effet des médicaments , Conception de médicament , Tests de criblage d'agents antitumoraux , Tumeurs du poumon , Poly (ADP-Ribose) polymerase-1 , Stilbènes , Humains , Relation structure-activité , Prolifération cellulaire/effets des médicaments et des substances chimiques , Structure moléculaire , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/métabolisme , Stilbènes/pharmacologie , Stilbènes/composition chimique , Stilbènes/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Lignée cellulaire tumorale
18.
Cell Rep ; 43(7): 114433, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38985679

RÉSUMÉ

ADP-ribosylation (ADPr) signaling plays a crucial role in DNA damage response. Inhibitors against the main enzyme catalyzing ADPr after DNA damage, poly(ADP-ribose) polymerase 1 (PARP1), are used to treat patients with breast cancer harboring BRCA1/2 mutations. However, resistance to PARP inhibitors (PARPi) is a major obstacle in treating patients. To understand the role of ADPr in PARPi sensitivity, we use liquid chromatography-tandem mass spectrometry (LC-MS/MS) to analyze ADPr in six breast cancer cell lines exhibiting different PARPi sensitivities. We identify 1,632 sites on 777 proteins across all cell lines, primarily on serine residues, with site-specific overlap of targeted residues across DNA-damage-related proteins across all cell lines, demonstrating high conservation of serine ADPr-signaling networks upon DNA damage. Furthermore, we observe site-specific differences in ADPr intensities in PARPi-sensitive BRCA mutants and unique ADPr sites in PARPi-resistant BRCA-mutant HCC1937 cells, which have low poly(ADP-ribose) glycohydrolase (PARG) levels and longer ADPr chains on PARP1.


Sujet(s)
ADP-Ribosylation , Protéine BRCA1 , Tumeurs du sein , Altération de l'ADN , Sérine , Humains , Femelle , Lignée cellulaire tumorale , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Sérine/métabolisme , Protéine BRCA1/métabolisme , Protéine BRCA1/génétique , Protéine BRCA2/métabolisme , Protéine BRCA2/génétique , Mutation/génétique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Glycosidases/métabolisme , Glycosidases/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique
19.
Sci Rep ; 14(1): 17555, 2024 07 30.
Article de Anglais | MEDLINE | ID: mdl-39080338

RÉSUMÉ

Performing accurate Fluorescence Correlation Spectroscopy (FCS) measurements in cells can be challenging due to cellular motion or other intracellular processes. In this respect, it has recently been shown that analysis of FCS data in short temporal segments (segmented FCS) can be very useful to increase the accuracy of FCS measurements inside cells. Here, we demonstrate that segmented FCS can be performed on a commercial laser scanning microscope (LSM), even in the absence of the dedicated FCS module. We show how data can be acquired on a Leica SP8 confocal microscope and then exported and processed with a custom software in MATLAB. The software performs segmentation of the data to extract an average ACF and measure the diffusion coefficient in specific subcellular regions. First of all, we measure the diffusion of fluorophores of different size in solution, to show that good-quality ACFs can be obtained in a commercial LSM. Next, we validate the method by measuring the diffusion coefficient of GFP in the nucleus of HeLa cells, exploiting variations of the intensity to distinguish between nucleoplasm and nucleolus. As expected, the measured diffusion coefficient of GFP is slower in the nucleolus relative to nucleoplasm. Finally, we apply the method to HeLa cells expressing a PARP1 chromobody to measure the diffusion coefficient of PARP1 in different subcellular regions. We find that PARP1 diffusion is slower in the nucleolus compared to the nucleoplasm.


Sujet(s)
Microscopie confocale , Spectrométrie de fluorescence , Humains , Cellules HeLa , Microscopie confocale/méthodes , Spectrométrie de fluorescence/méthodes , Protéines à fluorescence verte/métabolisme , Diffusion , Noyau de la cellule/métabolisme , Logiciel , Poly (ADP-Ribose) polymerase-1/métabolisme
20.
Reprod Biol Endocrinol ; 22(1): 92, 2024 Jul 31.
Article de Anglais | MEDLINE | ID: mdl-39085882

RÉSUMÉ

BACKGROUND: Endometriosis is a gynecological disease characterized by the presence of endometrial tissue in abnormal locations, leading to severe symptoms, inflammation, pain, organ dysfunction, and infertility. Surgical removal of endometriosis lesions is crucial for improving pain and fertility outcomes, with the goal of complete lesion removal. This study aimed to analyze the location and expression patterns of poly (ADP-ribose) polymerase 1 (PARP-1), epithelial cell adhesion molecule (EpCAM), and folate receptor alpha (FRα) in endometriosis lesions and evaluate their potential for targeted imaging. METHODS: Gene expression analysis was performed using the Turku endometriosis database (EndometDB). By immunohistochemistry, we investigated the presence and distribution of PARP-1, EpCAM, and FRα in endometriosis foci and adjacent tissue. We also applied an ad hoc platform for the analysis of images to perform a quantitative immunolocalization analysis. Double immunofluorescence analysis was carried out for PARP-1 and EpCAM, as well as for PARP-1 and FRα, to explore the expression of these combined markers within endometriosis foci and their potential simultaneous utilization in surgical treatment. RESULTS: Gene expression analysis revealed that PARP-1, EpCAM, and FOLR1 (FRα gene) are more highly expressed in endometriotic lesions than in the peritoneum, which served as the control tissue. The results of the immunohistochemical study revealed a significant increase in the expression levels of all three biomarkers inside the endometriosis foci compared to the adjacent tissues. Additionally, the double immunofluorescence analysis consistently demonstrated the presence of PARP-1 in the nucleus and the expression of EpCAM and FRα in the cell membrane and cytoplasm. CONCLUSION: Overall, these three markers demonstrate significant potential for effective imaging of endometriosis. In particular, the results emphasize the importance of PARP-1 expression as a possible indicator for distinguishing endometriotic lesions from adjacent tissue. PARP-1, as a potential biomarker for endometriosis, offers promising avenues for further investigation in terms of both pathophysiology and diagnostic-therapeutic approaches.


Sujet(s)
Endométriose , Molécule d'adhérence des cellules épithéliales , Récepteur-1 des folates , Poly (ADP-Ribose) polymerase-1 , Endométriose/métabolisme , Endométriose/chirurgie , Endométriose/génétique , Endométriose/diagnostic , Endométriose/anatomopathologie , Femelle , Humains , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Récepteur-1 des folates/génétique , Récepteur-1 des folates/métabolisme , Molécule d'adhérence des cellules épithéliales/génétique , Molécule d'adhérence des cellules épithéliales/métabolisme , Adulte , Marqueurs biologiques/métabolisme , Immunohistochimie , Endomètre/métabolisme , Endomètre/anatomopathologie , Endomètre/chirurgie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE