Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 419
Filtrer
1.
Proc Natl Acad Sci U S A ; 121(25): e2322689121, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38865276

RÉSUMÉ

Poly(ADP-ribose) polymerase 1 (PARP1) has emerged as a central target for cancer therapies due to the ability of PARP inhibitors to specifically kill tumors deficient for DNA repair by homologous recombination. Upon DNA damage, PARP1 quickly binds to DNA breaks and triggers ADP-ribosylation signaling. ADP-ribosylation is important for the recruitment of various factors to sites of damage, as well as for the timely dissociation of PARP1 from DNA breaks. Indeed, PARP1 becomes trapped at DNA breaks in the presence of PARP inhibitors, a mechanism underlying the cytotoxitiy of these inhibitors. Therefore, any cellular process influencing trapping is thought to impact PARP inhibitor efficiency, potentially leading to acquired resistance in patients treated with these drugs. There are numerous ADP-ribosylation targets after DNA damage, including PARP1 itself as well as histones. While recent findings reported that the automodification of PARP1 promotes its release from the DNA lesions, the potential impact of other ADP-ribosylated proteins on this process remains unknown. Here, we demonstrate that histone ADP-ribosylation is also crucial for the timely dissipation of PARP1 from the lesions, thus contributing to cellular resistance to PARP inhibitors. Considering the crosstalk between ADP-ribosylation and other histone marks, our findings open interesting perspectives for the development of more efficient PARP inhibitor-driven cancer therapies.


Sujet(s)
ADP-Ribosylation , Histone , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Histone/métabolisme , Altération de l'ADN , Résistance aux médicaments antinéoplasiques/génétique , Lignée cellulaire tumorale , Poly(ADP-ribose) polymerases/métabolisme , Poly(ADP-ribose) polymerases/génétique
2.
Molecules ; 29(12)2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38930900

RÉSUMÉ

The malignancy of breast cancer poses a global challenge, with existing treatments often falling short of desired efficacy. Extensive research has underscored the effectiveness of targeting the metabolism of nicotinamide adenine dinucleotide (NAD), a pivotal molecule crucial for cancer cell survival and growth, as a promising anticancer strategy. Within mammalian cells, sustaining optimal NAD concentrations relies on two key enzymes, namely nicotinamide phosphoribosyltransferase (NAMPT) and poly(ADP-ribose) polymer 1 (PARP1). Recent studies have accentuated the potential benefits of combining NAMPT inhibitors and PARP1 inhibitors to enhance therapeutic outcomes, particularly in breast cancer. In this study, we designed and synthesized eleven novel NAMPT/PARP1 dual-target inhibitors. Among them, compound DDY02 exhibited acceptable inhibitory activities against both NAMPT and PARP1, with IC50 values of 0.01 and 0.05 µM, respectively. Moreover, in vitro evaluations revealed that treatment with DDY02 resulted in proliferation inhibition, NAD depletion, DNA damage, apoptosis, and migration inhibition in MDA-MB-468 cells. These results posit DDY02, by targeting NAD metabolism through inhibiting both NAMPT and PARP1, as a promising lead compound for the development of breast cancer therapy.


Sujet(s)
Antinéoplasiques , Tumeurs du sein , Prolifération cellulaire , NAD , Nicotinamide phosphoribosyltransferase , Poly (ADP-Ribose) polymerase-1 , Nicotinamide phosphoribosyltransferase/antagonistes et inhibiteurs , Nicotinamide phosphoribosyltransferase/métabolisme , Humains , NAD/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Femelle , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Conception de médicament , Cytokines/métabolisme , Antienzymes/pharmacologie , Antienzymes/synthèse chimique , Antienzymes/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Simulation de docking moléculaire
3.
Nat Aging ; 4(6): 771-782, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38724734

RÉSUMÉ

Excessive amounts of reactive oxygen species (ROS) lead to macromolecular damage and high levels of cell death with consequent pathological sequelae. We hypothesized that switching cell death to a tissue regenerative state could potentially improve the short-term and long-term detrimental effects of ROS-associated acute tissue injury, although the mechanisms regulating oxidative stress-induced cell fate decisions and their manipulation for improving repair are poorly understood. Here, we show that cells exposed to high oxidative stress enter a poly (ADP-ribose) polymerase 1 (PARP1)-mediated regulated cell death, and that blocking PARP1 activation promotes conversion of cell death into senescence (CODIS). We demonstrate that this conversion depends on reducing mitochondrial Ca2+ overload as a consequence of retaining the hexokinase II on mitochondria. In a mouse model of kidney ischemia-reperfusion damage, PARP inhibition reduces necrosis and increases transient senescence at the injury site, alongside improved recovery from damage. Together, these data provide evidence that converting cell death into transient senescence can therapeutically benefit tissue regeneration.


Sujet(s)
Mort cellulaire , Vieillissement de la cellule , Stress oxydatif , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Animaux , Stress oxydatif/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Souris , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Mort cellulaire/effets des médicaments et des substances chimiques , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Espèces réactives de l'oxygène/métabolisme , Humains , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Mitochondries/anatomopathologie , Calcium/métabolisme , Modèles animaux de maladie humaine
4.
J Med Chem ; 67(11): 8877-8901, 2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38776379

RÉSUMÉ

Designing selective PARP-1 inhibitors has become a new strategy for anticancer drug development. By sequence comparison of PARP-1 and PARP-2, we identified a possible selective site (S site) consisting of several different amino acid residues of α-5 helix and D-loop. Targeting this S site, 140 compounds were designed, synthesized, and characterized for their anticancer activities and mechanisms. Compound I16 showed the highest PARP-1 enzyme inhibitory activity (IC50 = 12.38 ± 1.33 nM) and optimal selectivity index over PARP-2 (SI = 155.74). Oral administration of I16 (25 mg/kg) showed high inhibition rates of Hela and SK-OV-3 tumor cell xenograft models, both of which were higher than those of the oral positive drug Olaparib (50 mg/kg). In addition, I16 has an excellent safety profile, without significant toxicity at high oral doses. These findings provide a novel design strategy and chemotype for the development of safe, efficient, and highly selective PARP-1 inhibitors.


Sujet(s)
Antinéoplasiques , Conception de médicament , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Animaux , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Souris , Relation structure-activité , Lignée cellulaire tumorale , Souris nude , Femelle , Tests d'activité antitumorale sur modèle de xénogreffe , Cellules HeLa , Simulation de docking moléculaire , Souris de lignée BALB C , Prolifération cellulaire/effets des médicaments et des substances chimiques , Phtalazines/pharmacologie , Phtalazines/composition chimique , Phtalazines/synthèse chimique
5.
Bioorg Chem ; 148: 107455, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38772289

RÉSUMÉ

Poly(ADP-ribose) polymerase-1 (PARP-1) is a crucial member of DNA repair enzymes responsible for repairing DNA single-strand breaks. Developing PARP inhibitors based on synthetic lethality strategies is an effective approach for treating breast cancer and other diseases. In this study, a series of novel piperidine-based benzamide derivatives were designed and synthesized using structure-based drug design principles. The anticancer activities of these compounds were evaluated against five human cancer cell lines (MDA-MB-436, CAPAN-1, SW-620, HepG2, SKOV3, and PC3) and the preliminary structure-activity relationships were delineated. Among the compounds, 6a and 15d demonstrated potent antiproliferative effects against MDA-MB-436 cells with IC50 values of 8.56 ± 1.07 µM and 6.99 ± 2.62 µM, respectively. Furthermore, both compounds exhibited excellent inhibitory activity against PARP-1, with IC50 values of 8.33 nM and 12.02 nM, respectively. Mechanistic investigations revealed that 6a and 15d effectively inhibited colony formation and cell migration of HCT116 cells. Moreover, they induced apoptosis by upregulating the expression of Bax and cleaved Caspase-3, while downregulating the expression of Caspase-3 and Bcl-2 in HCT116 cells. Based on its impressive pharmacodynamic data in vitro, we conducted a study to evaluate the efficacy of 15d in a xenograft tumor model in mice when used in combination with cytotoxic agents. Collectively, these findings suggest that 15d could be promising drug candidates worthy of further investigation.


Sujet(s)
Antinéoplasiques , Apoptose , Prolifération cellulaire , Relation dose-effet des médicaments , Conception de médicament , Tests de criblage d'agents antitumoraux , Pipéridines , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Pipéridines/pharmacologie , Pipéridines/composition chimique , Pipéridines/synthèse chimique , Relation structure-activité , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Animaux , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Structure moléculaire , Souris , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris nude , Souris de lignée BALB C
6.
Bioorg Chem ; 148: 107480, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38772291

RÉSUMÉ

A novel series of erythrina derivatives as PARP-1/FTase inhibitors were synthesized, and evaluated for their biological activities. Compound T9 had excellent inhibitory effects on cell viability (A549: IC50 = 1.74 µM; A549/5-Fu: IC50 = 1.03 µM) and in vitro enzyme activities (PARP-1: IC50 = 0.40 µM; FTase: IC50 = 0.067 µM). Molecular docking and point mutation assays demonstrated the interaction of compound T9 with key amino acid residues. The compound T9 exhibited potent anti-proliferation and anti-migration capabilities against A549 and A549/5-Fu cells. PCR array and western blot results showed that compound T9 could effectively inhibit EMT-related proteins in A549 and A549/5-Fu cells, thereby inhibiting the development of lung cancer. Importantly, compound T9 could significantly inhibit tumor growth in the A549 xenograft tumor model (TGI = 65.3 %). In conclusion, this study was the first presentation of the concept of dual-target inhibitors of the PARP-1/FTase enzymes. It also provides the basis for further research and development of novel PARP-1/FTase inhibitors.


Sujet(s)
Antinéoplasiques , Prolifération cellulaire , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Transition épithélio-mésenchymateuse , Erythrina , Tumeurs du poumon , Poly (ADP-Ribose) polymerase-1 , Humains , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Relation structure-activité , Erythrina/composition chimique , Animaux , Structure moléculaire , Souris , Simulation de docking moléculaire , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Souris nude , Tumeurs expérimentales/traitement médicamenteux , Tumeurs expérimentales/anatomopathologie , Tumeurs expérimentales/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Mouvement cellulaire/effets des médicaments et des substances chimiques
8.
J Hematol Oncol ; 17(1): 36, 2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38783389

RÉSUMÉ

Oncolytic viruses (OVs) offer a novel approach to treat solid tumors; however, their efficacy is frequently suboptimal due to various limiting factors. To address this challenge, we engineered an OV containing targets for neuron-specific microRNA-124 and Granulocyte-macrophage colony-stimulating factor (GM-CSF), significantly enhancing its neuronal safety while minimally compromising its replication capacity. Moreover, we identified PARP1 as an HSV-1 replication restriction factor using genome-wide CRISPR screening. In models of glioblastoma (GBM) and triple-negative breast cancer (TNBC), we showed that the combination of OV and a PARP inhibitor (PARPi) exhibited superior efficacy compared to either monotherapy. Additionally, single-cell RNA sequencing (scRNA-seq) revealed that this combination therapy sensitized TNBC to immune checkpoint blockade, and the incorporation of an immune checkpoint inhibitor (ICI) further increased the survival rate of tumor-bearing mice. The combination of PARPi and ICI synergistically enhanced the ability of OV to establish durable tumor-specific immune responses. Our study effectively overcomes the inherent limitations of OV therapy, providing valuable insights for the clinical treatment of TNBC, GBM, and other malignancies.


Sujet(s)
Thérapie virale de cancers , Thérapie virale de cancers/méthodes , Animaux , Humains , Souris , Facteur de stimulation des colonies de granulocytes et de macrophages/génétique , Glioblastome/thérapie , Glioblastome/génétique , Virus oncolytiques/génétique , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Tumeurs du sein triple-négatives/thérapie , Tumeurs du sein triple-négatives/génétique , Femelle , Poly (ADP-Ribose) polymerase-1/génétique , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Herpèsvirus humain de type 1/génétique , Lignée cellulaire tumorale , Clustered regularly interspaced short palindromic repeats/génétique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , microARN/génétique , Tests d'activité antitumorale sur modèle de xénogreffe , Systèmes CRISPR-Cas
9.
Chin J Nat Med ; 22(5): 455-465, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38796218

RÉSUMÉ

In this study, we reported the discovery and structure-activity relationship analysis of chrysin derivatives as a new class of inhibitors targeting poly (ADP-ribose) polymerase 1 (PARP1). Among these derivatives, compound 5d emerged as the most effective chrysin-based inhibitor of PARP1, with an IC50 value of 108 nmol·L-1. This compound significantly inhibited the proliferation and migration of breast cancer cell lines HCC-1937 and MDA-MB-436 by inducing DNA damage. Furthermore, 5d induced apoptosis and caused an extended G1/S-phase in these cell lines. Molecular docking studies revealed that 5d possesses a strong binding affinity toward PARP1. In vivo, in a xenograft model, 5d effectively reduced tumor growth by downregulating PARP1 expression. Overall, compound 5d shows promise as a potential therapeutic agent for the treatment of BRCA wild-type breast cancer.


Sujet(s)
Apoptose , Tumeurs du sein , Prolifération cellulaire , Flavonoïdes , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Flavonoïdes/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Femelle , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Lignée cellulaire tumorale , Animaux , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation structure-activité , Apoptose/effets des médicaments et des substances chimiques , Simulation de docking moléculaire , Souris , Conception de médicament , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Souris nude , Souris de lignée BALB C , Structure moléculaire
10.
Chem Biol Interact ; 395: 111010, 2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38679114

RÉSUMÉ

The incidence and mortality rate of myocardial infarction are increasing per year in China. The polarization of macrophages towards the classically activated macrophages (M1) phenotype is of utmost importance in the progression of inflammatory stress subsequent to myocardial infarction. Poly (ADP-ribose) polymerase 1(PARP1) is the ubiquitous and best characterized member of the PARP family, which has been reported to support macrophage polarization towards the pro-inflammatory phenotype. Yet, the role of PARP1 in myocardial ischemic injury remains to be elucidated. Here, we demonstrated that a myocardial infarction mouse model induced cardiac damage characterized by cardiac dysfunction and increased PARP1 expression in cardiac macrophages. Inhibition of PARP1 by the PJ34 inhibitors could effectively alleviate M1 macrophage polarization, reduce infarction size, decrease inflammation and rescue the cardiac function post-MI in mice. Mechanistically, the suppression of PARP1 increase NLRC5 gene expression, and thus inhibits the NF-κB pathway, thereby decreasing the production of inflammatory cytokines such as IL-1ß and TNF-α. Inhibition of NLRC5 promote infection by effectively abolishing the influence of this mechanism discussed above. Interestingly, inhibition of NLRC5 promotes cardiac macrophage polarization toward an M1 phenotype but without having major effects on M2 macrophages. Our results demonstrate that inhibition of PARP1 increased NLRC5 gene expression, thereby suppressing M1 polarization, improving cardiac function, decreasing infarct area and attenuating inflammatory injury. The aforementioned findings provide new insights into the proinflammatory mechanisms that drive macrophage polarization following myocardial infarction, thereby introducing novel potential targets for future therapeutic interventions in individuals affected by myocardial infarction.


Sujet(s)
Protéines et peptides de signalisation intracellulaire , Macrophages , Infarctus du myocarde , Facteur de transcription NF-kappa B , Poly (ADP-Ribose) polymerase-1 , Animaux , Mâle , Souris , Modèles animaux de maladie humaine , Protéines et peptides de signalisation intracellulaire/métabolisme , Protéines et peptides de signalisation intracellulaire/génétique , Macrophages/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Infarctus du myocarde/métabolisme , Infarctus du myocarde/traitement médicamenteux , Infarctus du myocarde/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Phénanthrènes/pharmacologie , Phénanthrènes/usage thérapeutique , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Régulation positive/effets des médicaments et des substances chimiques
11.
EBioMedicine ; 103: 105129, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38640836

RÉSUMÉ

BACKGROUND: Poly(ADP-ribose) polymerase (PARP) inhibitors have emerged as promising chemotherapeutic drugs primarily against BRCA1/2-associated tumours, known as synthetic lethality. However, recent clinical trials reported patients' survival benefits from PARP inhibitor treatments, irrelevant to homologous recombination deficiency. Therefore, revealing the therapeutic mechanism of PARP inhibitors beyond DNA damage repair is urgently needed, which can facilitate precision medicine. METHODS: A CRISPR-based knock-in technology was used to establish stable BRCA1 mutant cancer cells. The effects of PARP inhibitors on BRCA1 mutant cancer cells were evaluated by biochemical and cell biological experiments. Finally, we validated its in vivo effects in xenograft and patient-derived xenograft (PDX) tumour mice. FINDINGS: In this study, we uncovered that the majority of clinical BRCA1 mutations in breast cancers were in and near the middle of the gene, rather than in essential regions for DNA damage repair. Representative mutations such as R1085I and E1222Q caused transient extra spindle poles during mitosis in cancer cells. PAR, which is synthesized by PARP2 but not PARP1 at mitotic centrosomes, clustered these transient extra poles, independent of DNA damage response. Common PARP inhibitors could effectively suppress PARP2-synthesized PAR and induce cell senescence by abrogating the correction of mitotic extra-pole error. INTERPRETATION: Our findings uncover an alternative mechanism by which PARP inhibitors efficiently suppress tumours, thereby pointing to a potential new therapeutic strategy for centrosome error-related tumours. FUNDING: Funded by National Natural Science Foundation of China (NSFC) (T2225006, 82272948, 82103106), Beijing Municipal Natural Science Foundation (Key program Z220011), and the National Clinical Key Specialty Construction Program, P. R. China (2023).


Sujet(s)
Protéine BRCA1 , Vieillissement de la cellule , Centrosome , Altération de l'ADN , Inhibiteurs de poly(ADP-ribose) polymérases , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Humains , Animaux , Centrosome/métabolisme , Centrosome/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Souris , Protéine BRCA1/génétique , Lignée cellulaire tumorale , Femelle , Mutation , Réparation de l'ADN/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Poly(ADP-ribose) polymerases/métabolisme , Poly(ADP-ribose) polymerases/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/génétique
12.
Eur J Pharmacol ; 972: 176557, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38574839

RÉSUMÉ

Cerebral ischemia-reperfusion injury (CIRI) can induce massive death of ischemic penumbra neurons via oxygen burst, exacerbating brain damage. Parthanatos is a form of caspase-independent cell death involving excessive activation of PARP-1, closely associated with intense oxidative stress following CIRI. 4'-O-methylbavachalcone (MeBavaC), an isoprenylated chalcone component in Fructus Psoraleae, has potential neuroprotective effects. This study primarily investigates whether MeBavaC can act on SIRT3 to alleviate parthanatos of ischemic penumbra neurons induced by CIRI. MeBavaC was oral gavaged to the middle cerebral artery occlusion-reperfusion (MCAO/R) rats after occlusion. The effects of MeBavaC on cerebral injury were detected by the neurological deficit score and cerebral infarct volume. In vitro, PC-12 cells were subjected to oxygen and glucose deprivation/reoxygenation (OGD/R), and assessed cell viability and cell injury. Also, the levels of ROS, mitochondrial membrane potential (MMP), and intracellular Ca2+ levels were detected to reflect mitochondrial function. We conducted western blotting analyses of proteins involved in parthanatos and related signaling pathways. Finally, the exact mechanism between the neuroprotection of MeBavaC and parthanatos was explored. Our results indicate that MeBavaC reduces the cerebral infarct volume and neurological deficit scores in MCAO/R rats, and inhibits the decreased viability of PC-12 cells induced by OGD/R. MeBavaC also downregulates the expression of parthanatos-related death proteins PARP-1, PAR, and AIF. However, this inhibitory effect is weakened after the use of a SIRT3 inhibitor. In conclusion, the protective effect of MeBavaC against CIRI may be achieved by inhibiting parthanatos of ischemic penumbra neurons through the SIRT3-PARP-1 axis.


Sujet(s)
Chalcones , Neuroprotecteurs , Parthanatos , Rat Sprague-Dawley , Lésion d'ischémie-reperfusion , Sirtuines , Animaux , Rats , Mâle , Chalcones/pharmacologie , Chalcones/usage thérapeutique , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Lésion d'ischémie-reperfusion/métabolisme , Parthanatos/effets des médicaments et des substances chimiques , Accident vasculaire cérébral ischémique/traitement médicamenteux , Accident vasculaire cérébral ischémique/anatomopathologie , Accident vasculaire cérébral ischémique/métabolisme , Espèces réactives de l'oxygène/métabolisme , Cellules PC12 , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neurones/anatomopathologie , Neurones/métabolisme , Calcium/métabolisme , Infarctus du territoire de l'artère cérébrale moyenne/traitement médicamenteux , Infarctus du territoire de l'artère cérébrale moyenne/anatomopathologie , Infarctus du territoire de l'artère cérébrale moyenne/complications , Survie cellulaire/effets des médicaments et des substances chimiques , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme
13.
Eur J Med Chem ; 271: 116405, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38678823

RÉSUMÉ

PARPi have been explored and applied in the treatment of various cancers with remarkable efficacy, especially BRCA1/2 mutated ovarian, breast, prostate, and pancreatic cancers. However, PARPi renders inevitable drug resistance and showed high toxicity because of PARP-Trapping with long-term clinic tracking. To overcome the drug resistance and the high toxicity of PARPi, many novel methods have been developed including PROTACs. Being an event-driven technology, PROTACs needs a high affinity, low toxicity warhead with no steric hindrance in binding process. Veliparib shows the lowest PARP-Trapping effect but could hardly to be the warhead of PROTACs because of the strong steric hindrance. Other PARP1 inhibitors showed less steric hindrance but owns high PARP-Trapping effect. Thus, the development of novel warhead with high PARP1 affinity, low PARP1-Trapping, and no steric hindrance would be valuable. In this work, we reserved benzimidazole as the motif to reserve the low PARP1-Trapping effect and substituted the pyrrole by aromatic ring to avoiding the steric hindrance in PARP1 binding cave. Thus, a series of benzimidazole derivates were designed and synthesized, and some biological activities in vitro were evaluated including the inhibition for PARP1 enzyme and the PARP-Trapping effect using MDA-MB-436 cell line. Results showed that the compound 19A10 has higher PARP1 affinity(IC50 = 4.62 nM)) and similar low PARP-Trapping effect compared with Veliparib(IC50 (MDA-MB-436) >100 µM). Docking study showed that the compound 19A10 could avoiding the steric hindrance which was much better than Veliparib. So, the compound 19A10 could potentially be a perfect warhead for PARP1 degraders. Besides, because of the depletion of the PARP1 and the decreasing of the binding capability, we suppose that the PROTACs using 19A10 as the warhead would be no-PARP-Trapping effect. Furthermore, QSAR study showed that to develop novel compounds with high PARP1 binding affinity and low PARP-Trapping, we can choose the skeleton with substituent R1H, R2 = piperiazine, and R3 with large tPSA. And, if we want to develop the compounds with high PARP1 binding affinity and high PARP-Trapping which can possibly improve the lethality against tumor cells, we can choose the skeleton with substituent R1F, R2 = 3-methy-piperiazine, and R3 with large tPSA.


Sujet(s)
Antinéoplasiques , Benzimidazoles , Tests de criblage d'agents antitumoraux , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Benzimidazoles/composition chimique , Benzimidazoles/pharmacologie , Benzimidazoles/synthèse chimique , Humains , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Relation structure-activité , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/synthèse chimique , Structure moléculaire , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Simulation de docking moléculaire
14.
Arch Biochem Biophys ; 756: 110010, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38642632

RÉSUMÉ

PARP1 plays a pivotal role in DNA repair within the base excision pathway, making it a promising therapeutic target for cancers involving BRCA mutations. Current study is focused on the discovery of PARP inhibitors with enhanced selectivity for PARP1. Concurrent inhibition of PARP1 with PARP2 and PARP3 affects cellular functions, potentially causing DNA damage accumulation and disrupting immune responses. In step 1, a virtual library of 593 million compounds has been screened using a shape-based screening approach to narrow down the promising scaffolds. In step 2, hierarchical docking approach embedded in Schrödinger suite was employed to select compounds with good dock score, drug-likeness and MMGBSA score. Analysis supplemented with decomposition energy, molecular dynamics (MD) simulations and hydrogen bond frequency analysis, pinpointed that active site residues; H862, G863, R878, M890, Y896 and F897 are crucial for specific binding of ZINC001258189808 and ZINC000092332196 with PARP1 as compared to PARP2 and PARP3. The binding of ZINC000656130962, ZINC000762230673, ZINC001332491123, and ZINC000579446675 also revealed interaction involving two additional active site residues of PARP1, namely N767 and E988. Weaker or no interaction was observed for these residues with PARP2 and PARP3. This approach advances our understanding of PARP-1 specific inhibitors and their mechanisms of action, facilitating the development of targeted therapeutics.


Sujet(s)
Antinéoplasiques , Conception de médicament , Simulation de dynamique moléculaire , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Simulation de docking moléculaire , Domaine catalytique , Poly(ADP-ribose) polymerases/métabolisme , Poly(ADP-ribose) polymerases/composition chimique , Liaison hydrogène
15.
J Virol ; 98(5): e0048324, 2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38639486

RÉSUMÉ

Alphaherpesvirus pseudorabies virus (PRV) causes severe economic losses to the global pig industry and has garnered increasing attention due to its broad host range including humans. PRV has developed a variety of strategies to antagonize host antiviral innate immunity. However, the underlying mechanisms have not been fully elucidated. In our previous work, we demonstrated that non-muscle myosin heavy chain IIA (NMHC-IIA), a multifunctional cytoskeleton protein, attenuates innate immune responses triggered by RNA viruses. In the current study, we reported a previously unrecognized role of NMHC-IIA in counteracting PRV-induced cyclic GMP-AMP synthase (cGAS)-dependent type I interferon (IFN-I) production. Mechanistically, PRV infection led to an elevation of NMHC-IIA, strengthening the interaction between poly (ADP-ribose) polymerase 1 (PARP1) and cGAS. This interaction impeded cGAS recognition of PRV DNA and hindered downstream signaling activation. Conversely, inhibition of NMHC-IIA by Blebbistatin triggered innate immune responses and enhanced resistance to PRV proliferation both in vitro and in vivo. Taken together, our findings unveil that PRV utilizes NMHC-IIA to antagonize host antiviral immune responses via impairing DNA sensing by cGAS. This in-depth understanding of PRV immunosuppression not only provides insights for potential PRV treatment strategies but also highlights NMHC-IIA as a versatile immunosuppressive regulator usurped by both DNA and RNA viruses. Consequently, NMHC-IIA holds promise as a target for the development of broad-spectrum antiviral drugs.IMPORTANCECyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) axis plays a vital role in counteracting alphaherpesvirus infections. Alphaherpesviruses exploit various strategies for antagonizing cGAS-STING-mediated antiviral immune responses. However, limited examples of pseudorabies virus (PRV)-caused immunosuppression have been documented. Our findings reveal a novel role of non-muscle myosin heavy chain IIA (NMHC-IIA) in suppressing PRV-triggered innate immune responses to facilitate viral propagation both in vitro and in vivo. In detail, NMHC-IIA recruits poly (ADP-ribose) polymerase 1 (PARP1) to augment its interaction with cGAS, which impairs cGAS recognition of PRV DNA. Building on our previous demonstration of NMHC-IIA's immunosuppressive role during RNA virus infections, these findings indicate that NMHC-IIA acts as a broad-spectrum suppressor of host antiviral innate immunity in response to both DNA and RNA viruses. Therefore, NMHC-IIA will be a promising target for the development of comprehensive antiviral strategies.


Sujet(s)
Herpèsvirus porcin de type 1 , Immunité innée , Myosine non-musculaire de type IIA , Maladie d'Aujeszky , Animaux , Humains , Souris , Lignée cellulaire , ADN viral/immunologie , Cellules HEK293 , Herpèsvirus porcin de type 1/immunologie , Interféron de type I/métabolisme , Interféron de type I/immunologie , Chaînes lourdes de myosine/métabolisme , Chaînes lourdes de myosine/immunologie , Myosine non-musculaire de type IIA/métabolisme , Nucleotidyltransferases/métabolisme , Poly (ADP-Ribose) polymerase-1/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Maladie d'Aujeszky/immunologie , Maladie d'Aujeszky/virologie , Transduction du signal , Suidae
16.
ChemMedChem ; 19(11): e202400093, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38482564

RÉSUMÉ

Inhibition of poly (ADP-ribose) polymerase-1 (PARP1), a DNA repair enzyme, has proven to be a successful strategy for the treatment of various cancers. With the appropriate selection conditions and protein design, DNA-encoded library (DEL) technology provides a powerful avenue to identify small molecules with the desired mechanism of action towards a target of interest. However, DNA-binding proteins, such as PARP1, can be challenging targets for DEL screening due to non-specific protein-DNA interactions. To overcome this, we designed and screened a PARP1 catalytic domain construct without the autoinhibitory helical domain. This allowed us to interrogate an active, functionally-relevant form of the protein resulting in the discovery of novel isoindolinone PARP1 inhibitors with single-digit nanomolar potency. These inhibitors also demonstrated little to no PARP1-DNA trapping, a property that could be advantageous in the clinic.


Sujet(s)
ADN , Poly (ADP-Ribose) polymerase-1 , Inhibiteurs de poly(ADP-ribose) polymérases , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , ADN/composition chimique , ADN/métabolisme , Relation structure-activité , Découverte de médicament , Structure moléculaire , Bibliothèques de petites molécules/composition chimique , Bibliothèques de petites molécules/pharmacologie , Bibliothèques de petites molécules/synthèse chimique , Relation dose-effet des médicaments , Isoindoles/composition chimique , Isoindoles/pharmacologie , Isoindoles/synthèse chimique , Domaine catalytique
17.
JAMA Oncol ; 10(5): 658-670, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38512229

RÉSUMÉ

Importance: Poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitors have revolutionized the treatment of patients with germline BRCA1/2-associated breast cancer, representing the first targeted therapy capable of improving outcomes in patients with hereditary tumors. However, resistance to PARP inhibitors occurs in almost all patients. Observations: This narrative review summarizes the biological rationale behind the use of PARP inhibitors in breast cancer, as well as the available evidence, recent progress, and potential future applications of these agents. Recent studies have shown that the benefit of PARP inhibitors extends beyond patients with germline BRCA1/2-associated metastatic breast cancer to patients with somatic BRCA1/2 variants and to those with germline PALB2 alterations. Moreover, these agents proved to be effective both in the metastatic and adjuvant settings. However, patients with metastatic breast cancer usually do not achieve the long-term benefit from PARP inhibitors observed in other tumor types. Mechanisms of resistance have been identified, but how to effectively target them is largely unknown. Ongoing research is investigating both novel therapeutics and new combination strategies to overcome resistance. PARP1-selective inhibitors, by sparing the hematological toxic effects induced by the PARP2 blockade, are promising agents to be combined with chemotherapy, antibody-drug conjugates, and other targeted therapies. Conclusions and Relevance: Although the efficacy of PARP inhibitors is well established, many questions persist. Future research should focus on identifying predictive biomarkers and therapeutic strategies to overcome resistance. Integrating well-designed translational efforts into all clinical studies is thereby crucial to laying the groundwork for future insights from ongoing research.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Inhibiteurs de poly(ADP-ribose) polymérases , Humains , Inhibiteurs de poly(ADP-ribose) polymérases/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Femelle , Protéine BRCA1/génétique , Protéine BRCA2/génétique , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Résultat thérapeutique
18.
Biochem Pharmacol ; 221: 116045, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38336156

RÉSUMÉ

Cancer is a disease with a high mortality rate characterized by uncontrolled proliferation of abnormal cells. The hallmarks of cancer evidence the acquired cells characteristics that promote the growth of malignant tumours, including genomic instability and mutations, the ability to evade cellular death and the capacity of sustaining proliferative signalization. Poly(ADP-ribose) polymerase-1 (PARP1) is a protein that plays key roles in cellular regulation, namely in DNA damage repair and cell survival. The inhibition of PARP1 promotes cellular death in cells with homologous recombination deficiency, and therefore, the interest in PARP protein has been rising as a target for anticancer therapies. There are already some PARP1 inhibitors approved by Food and Drug Administration (FDA), such as Olaparib and Niraparib. The last compound presents in its structure an indazole core. In fact, pyrazoles and indazoles have been raising interest due to their various medicinal properties, namely, anticancer activity. Derivatives of these compounds have been studied as inhibitors of PARP1 and presented promising results. Therefore, this review aims to address the importance of PARP1 in cell regulation and its role in cancer. Moreover, it intends to report a comprehensive literature review of PARP1 inhibitors, containing the pyrazole and indazole scaffolds, published in the last fifteen years, focusing on structure-activity relationship aspects, thus providing important insights for the design of novel and more effective PARP1 inhibitors.


Sujet(s)
Tumeurs , Poly (ADP-Ribose) polymerase-1 , Pyrazoles , Adénosine diphosphate ribose , Cycle cellulaire , Indazoles/pharmacologie , Indazoles/usage thérapeutique , Tumeurs/traitement médicamenteux , Tumeurs/enzymologie , Pyrazoles/pharmacologie , Pyrazoles/usage thérapeutique , États-Unis , Humains , Animaux , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme
19.
Sci Rep ; 14(1): 3875, 2024 02 16.
Article de Anglais | MEDLINE | ID: mdl-38365924

RÉSUMÉ

ADP-ribosyltransferases PARP1 and PARP2 play a major role in DNA repair mechanism by detecting the DNA damage and inducing poly-ADP-ribosylation dependent chromatin relaxation and recruitment of repair proteins. Catalytic PARP inhibitors are used as anticancer drugs especially in the case of tumors arising from sensitizing mutations. Recently, a study showed that Histone PARylation Factor (HPF1) forms a joint active site with PARP1/2. The interaction of HPF1 with PARP1/2 alters the modification site from Aspartate/Glutamate to Serine, which has been shown to be a key ADP-ribosylation event in the context of DNA damage. Therefore, disruption of PARP1/2-HPF1 interaction could be an alternative strategy for drug development to block the PARP1/2 activity. In this study, we describe a FRET based high-throughput screening assay to screen inhibitor libraries against PARP-HPF1 interaction. We optimized the conditions for FRET signal and verified the interaction by competing the FRET pair in multiple ways. The assay is robust and easy to automate. Validatory screening showed the robust performance of the assay, and we discovered two compounds Dimethylacrylshikonin and Alkannin, with µM inhibition potency against PARP1/2-HPF1 interaction. The assay will facilitate the discovery of inhibitors against HPF1-PARP1/2 complex and to develop potentially new effective anticancer agents.


Sujet(s)
Antinéoplasiques , Histone , Inhibiteurs de poly(ADP-ribose) polymérases , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Altération de l'ADN , Réparation de l'ADN , Tests de criblage à haut débit , Histone/effets des médicaments et des substances chimiques , Histone/métabolisme , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly(ADP-ribosylation) , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie
20.
Mol Inform ; 43(4): e202300183, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38258328

RÉSUMÉ

De novo design has been a hotly pursued topic for many years. Most recent developments have involved the use of deep learning methods for generative molecular design. Despite increasing levels of algorithmic sophistication, the design of molecules that are synthetically accessible remains a major challenge. Reaction-based de novo design takes a conceptually simpler approach and aims to address synthesisability directly by mimicking synthetic chemistry and driving structural transformations by known reactions that are applied in a stepwise manner. However, the use of a small number of hand-coded transformations restricts the chemical space that can be accessed and there are few examples in the literature where molecules and their synthetic routes have been designed and executed successfully. Here we describe the application of reaction-based de novo design to the design of synthetically accessible and biologically active compounds as proof-of-concept of our reaction vector-based software. Reaction vectors are derived automatically from known reactions and allow access to a wide region of synthetically accessible chemical space. The design was aimed at producing molecules that are active against PARP1 and which have improved brain penetration properties compared to existing PARP1 inhibitors. We synthesised a selection of the designed molecules according to the provided synthetic routes and tested them experimentally. The results demonstrate that reaction vectors can be applied to the design of novel molecules of biological relevance that are also synthetically accessible.


Sujet(s)
Conception de médicament , Inhibiteurs de poly(ADP-ribose) polymérases , Inhibiteurs de poly(ADP-ribose) polymérases/composition chimique , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie , Inhibiteurs de poly(ADP-ribose) polymérases/synthèse chimique , Humains , Poly (ADP-Ribose) polymerase-1/antagonistes et inhibiteurs , Poly (ADP-Ribose) polymerase-1/métabolisme , Logiciel
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...