Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Med Chem ; 66(5): 3411-3430, 2023 03 09.
Article de Anglais | MEDLINE | ID: mdl-36823782

RÉSUMÉ

The unique bisubunit structure of Leishmania donovani topoisomerase 1B (LdTop1) is a potential drug target in the parasites unlike the monomeric Top1 from its human host counterpart. Here, we report the design, synthesis, and validation of a chimeric pyrido[2',1':2,3]imidazo[4,5-c]quinoline derivative (C17) as a novel antileishmanial agent that poisons topoisomerase 1-DNA covalent complexes (LdTop1cc) inside the parasites and inhibits Top1 religation activity both in the drug sensitive and antimony-resistant L. donovani clinical isolates. Importantly, the human Top1 is not sensitive to C17. Further, C17 overcomes the chemical instability of camptothecin (CPT) by generating persistent LdTop1cc-induced DNA breaks inside the parasites even after 12 h of drug removal. Intraperitoneal administration of C17 results in marked reduction of the Leishmania amastigotes from the infected spleen and liver of BALB/c mice. C17 confers a host protective immune-response up-regulating the Th1 cytokines facilitating parasite clearance which can be exploited for treating drug-resistant leishmaniasis.


Sujet(s)
Antiprotozoaires , Leishmania donovani , Leishmaniose viscérale , Leishmaniose , Toxiques , Quinoléines , Animaux , Souris , Humains , Leishmaniose viscérale/traitement médicamenteux , Leishmaniose viscérale/parasitologie , Antimoine/pharmacologie , Antimoine/usage thérapeutique , Toxiques/usage thérapeutique , Antiprotozoaires/pharmacologie , Antiprotozoaires/usage thérapeutique , Leishmaniose/traitement médicamenteux , ADN/composition chimique , Quinoléines/pharmacologie , Quinoléines/usage thérapeutique , Souris de lignée BALB C
2.
Trends Cell Biol ; 33(3): 235-246, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-35963793

RÉSUMÉ

The MYC protooncogene functions as a universal amplifier of transcription through interaction with numerous factors and complexes that regulate almost every cellular process. However, a comprehensive model that explains MYC's actions and the interplay governing the complicated dynamics of components of the transcription and replication machinery is still lacking. Here, we review the potency of MYC as an oncogenic driver and how it regulates the broad spectrum of complexes (effectors and regulators). We propose a 'hand-over model' for differential partitioning and trafficking of unstructured MYC via a loose interaction network between various gene-regulatory complexes and factors. Additionally, the article discusses how unstructured-MYC energetically favors efficient modulation of the energy landscape of the transcription cycle.


Sujet(s)
Régulation de l'expression des gènes , Protéines proto-oncogènes c-myc , Humains , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme
3.
FASEB J ; 36(4): e22265, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35319800

RÉSUMÉ

Leishmania donovani, a unicellular protozoan parasite, causes a wide range of human diseases including fatal visceral leishmaniasis. Tyrosyl DNA-phosphodiesterase 1 (TDP1) hydrolyzes the phosphodiester bond between DNA 3'-end and a tyrosyl moiety of trapped topoisomerase I-DNA covalent complexes (Top1cc). We have previously shown Leishmania harbors a TDP1 gene (LdTDP1), however, the biological role of TDP1 remains largely unknown. In the present study, we have generated TDP1 knockout L. donovani (LdTDP1-/- ) promastigotes and have shown that LdTDP1-/- parasites are deficient in 3'-phosphodiesterase activities and were hypersensitive to Top1-poison like camptothecin (CPT), DNA alkylation agent like methyl methanesulfonate, and oxidative DNA lesions generated by hydrogen peroxide but were not sensitive to etoposide. We also detected elevated levels of CPT-induced reactive oxygen species triggering cell cycle arrest and cell death in LdTDP1-/- promastigotes. LdTDP1-/- promastigotes accumulate a significant change in the membrane morphology with the accumulation of membrane pores, which is associated with oxidative stress and lipid peroxidation. To our surprise, we detected that LdTDP1-/- parasites were hypersensitive to antileishmanial drugs like amphotericin B and miltefosine, which could be rescued by complementation of wild-type TDP1 gene in the LdTDP1-/- parasites. Notably, multidrug-resistant L. donovani clinical isolates showed a marked reduction in TDP1 expression and were sensitive to Top1 poisons. Taken together, our study provides a new role of LdTDP1 in protecting L. donovani parasites from oxidative stress-induced DNA damage and resistance to amphotericin B and miltefosine.


Sujet(s)
Esterases , Leishmania donovani , Protéines de protozoaire , Amphotéricine B , Camptothécine/pharmacologie , ADN , Altération de l'ADN , Réparation de l'ADN , ADN topoisomérases de type I/génétique , ADN topoisomérases de type I/métabolisme , Esterases/génétique , Leishmania donovani/enzymologie , Leishmania donovani/génétique , Phosphodiesterases/métabolisme , Protéines de protozoaire/génétique
4.
Mol Cell ; 82(1): 140-158.e12, 2022 01 06.
Article de Anglais | MEDLINE | ID: mdl-34890565

RÉSUMÉ

High-intensity transcription and replication supercoil DNA to levels that can impede or halt these processes. As a potent transcription amplifier and replication accelerator, the proto-oncogene MYC must manage this interfering torsional stress. By comparing gene expression with the recruitment of topoisomerases and MYC to promoters, we surmised a direct association of MYC with topoisomerase 1 (TOP1) and TOP2 that was confirmed in vitro and in cells. Beyond recruiting topoisomerases, MYC directly stimulates their activities. We identify a MYC-nucleated "topoisome" complex that unites TOP1 and TOP2 and increases their levels and activities at promoters, gene bodies, and enhancers. Whether TOP2A or TOP2B is included in the topoisome is dictated by the presence of MYC versus MYCN, respectively. Thus, in vitro and in cells, MYC assembles tools that simplify DNA topology and promote genome function under high output conditions.


Sujet(s)
ADN topoisomérases de type II/métabolisme , Tumeurs/enzymologie , Protéines liant le poly-adp-ribose/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Transcription génétique , Animaux , Réplication de l'ADN , ADN topoisomérases de type I/génétique , ADN topoisomérases de type I/métabolisme , ADN topoisomérases de type II/génétique , ADN tumoral/biosynthèse , ADN tumoral/génétique , ADN superhélicoïdal/biosynthèse , ADN superhélicoïdal/génétique , Activation enzymatique , Régulation de l'expression des gènes tumoraux , Cellules HCT116 , Humains , Cellules K562 , Complexes multienzymatiques , Tumeurs/génétique , Tumeurs/anatomopathologie , Protéines liant le poly-adp-ribose/génétique , Régions promotrices (génétique) , Liaison aux protéines , Protéines proto-oncogènes c-myc/génétique , Rats
5.
Elife ; 92020 07 27.
Article de Anglais | MEDLINE | ID: mdl-32715994

RÉSUMÉ

Supraphysiological MYC levels are oncogenic. Originally considered a typical transcription factor recruited to E-boxes (CACGTG), another theory posits MYC a global amplifier increasing output at all active promoters. Both models rest on large-scale genome-wide "-omics'. Because the assumptions, statistical parameter and model choice dictates the '-omic' results, whether MYC is a general or specific transcription factor remains controversial. Therefore, an orthogonal series of experiments interrogated MYC's effect on the expression of synthetic reporters. Dose-dependently, MYC increased output at minimal promoters with or without an E-box. Driving minimal promoters with exogenous (glucocorticoid receptor) or synthetic transcription factors made expression more MYC-responsive, effectively increasing MYC-amplifier gain. Mutations of conserved MYC-Box regions I and II impaired amplification, whereas MYC-box III mutations delivered higher reporter output indicating that MBIII limits over-amplification. Kinetic theory and experiments indicate that MYC activates at least two steps in the transcription-cycle to explain the non-linear amplification of transcription that is essential for global, supraphysiological transcription in cancer.


Sujet(s)
Amplification de gène , Protéines proto-oncogènes c-myc/génétique , Facteurs de transcription/génétique , Activation de la transcription , Animaux , Lignée cellulaire , Humains , Protéines proto-oncogènes c-myc/métabolisme , Rats , Facteurs de transcription/métabolisme
6.
Eur J Med Chem ; 202: 112551, 2020 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-32682183

RÉSUMÉ

We have recently reported a new chemotype of a potent topoisomerase I poison with compound 1 as a potential anticancer chemotherapeutic agent. During further optimization, it has been observed that compound 1 suffers from high intrinsic clearance in human liver microsomes. To overcome the metabolic instability of compound 1, we report design and synthesis of metabolically stable Top1 poison 3. Newly identified Top1 poison 3 exhibits t1/2 of 69.1 min in human liver microsomes in comparison to compound 1 with t1/2 of 9.9 min. Molecular dynamic study of the newly optimized Top1 poison 3 was performed to get the insight into the stability of the binding pose in the active site. Compound 3 was able to trap DNA-Top1 cleavage complex and found to be less cytotoxic in non-cancerous cell line as compared to compound 1.


Sujet(s)
Antinéoplasiques/pharmacologie , ADN topoisomérases de type I/métabolisme , Développement de médicament , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Humains , Microsomes du foie/composition chimique , Microsomes du foie/métabolisme , Modèles moléculaires , Structure moléculaire , Protéines recombinantes/métabolisme , Relation structure-activité
7.
J Med Chem ; 62(7): 3428-3446, 2019 04 11.
Article de Anglais | MEDLINE | ID: mdl-30897325

RÉSUMÉ

To overcome chemical limitations of camptothecin (CPT), we report design, synthesis, and validation of a quinoline-based novel class of topoisomerase 1 (Top1) inhibitors and establish that compound 28 ( N-(3-(1 H-imidazol-1-yl)propyl)-6-(4-methoxyphenyl)-3-(1,3,4-oxadiazol-2-yl)quinolin-4-amine) exhibits the highest potency in inhibiting human Top1 activity with an IC50 value of 29 ± 0.04 nM. Compound 28 traps Top1-DNA cleavage complexes (Top1ccs) both in the in vitro cleavage assays and in live cells. Point mutation of Top1-N722S fails to trap compound 28-induced Top1cc because of its inability to form a hydrogen bond with compound 28. Unlike CPT, compound 28 shows excellent plasma serum stability and is not a substrate of P-glycoprotein 1 (permeability glycoprotein) advancing its potential anticancer activity. Finally, we provide evidence that compound 28 overcomes the chemical instability of CPT in human breast adenocarcinoma cells through generation of persistent and less reversible Top1cc-induced DNA double-strand breaks as detected by γH2AX foci immunostaining after 5 h of drug removal.


Sujet(s)
Antinéoplasiques/pharmacologie , Découverte de médicament/méthodes , Inhibiteurs de la topoisomérase-I/pharmacologie , Antinéoplasiques/composition chimique , Lignée cellulaire tumorale , Cassures double-brin de l'ADN/effets des médicaments et des substances chimiques , ADN topoisomérases de type I/génétique , ADN topoisomérases de type I/métabolisme , Tests de criblage d'agents antitumoraux , Humains , Mutation ponctuelle , Inhibiteurs de la topoisomérase-I/composition chimique
8.
Nucleic Acids Res ; 46(11): 5601-5617, 2018 06 20.
Article de Anglais | MEDLINE | ID: mdl-29718323

RÉSUMÉ

Human tyrosyl-DNA phosphodiesterases (TDP) hydrolyze the phosphodiester bond between DNA and the catalytic tyrosine of Top1 to excise topoisomerase I cleavage complexes (Top1cc) that are trapped by camptothecin (CPT) and by genotoxic DNA alterations. Here we show that the protein arginine methyltransferase PRMT5 enhances the repair of Top1cc by direct binding to TDP1 and arginine dimethylation of TDP1 at residues R361 and R586. Top1-induced replication-mediated DNA damage induces TDP1 arginine methylation, enhancing its 3'- phosphodiesterase activity. TDP1 arginine methylation also increases XRCC1 association with TDP1 in response to CPT, and the recruitment of XRCC1 to Top1cc DNA damage foci. PRMT5 knockdown cells exhibit defective TDP1 activity with marked elevation in replication-coupled CPT-induced DNA damage and lethality. Finally, methylation of R361 and R586 stimulate TDP1 repair function and promote cell survival in response to CPT. Together, our findings provide evidence for the importance of PRMT5 for the post-translational regulation of TDP1 and repair of Top1cc.


Sujet(s)
Réparation de l'ADN , ADN topoisomérases de type I/métabolisme , Phosphodiesterases/métabolisme , Protein-arginine N-methyltransferases/métabolisme , Animaux , Arginine/métabolisme , Lignée cellulaire tumorale , Cellules cultivées , Altération de l'ADN , Réplication de l'ADN , Cellules HEK293 , Humains , Méthylation , Souris , Phosphodiesterases/composition chimique , Protéine-1 de complémentation croisée de la réparation des lésions induites par les rayons X/métabolisme
9.
J Med Chem ; 61(3): 804-817, 2018 02 08.
Article de Anglais | MEDLINE | ID: mdl-29290109

RÉSUMÉ

Camptothecin (CPT) selectively traps topoisomerase 1-DNA cleavable complexes (Top1cc) to promote anticancer activity. Here, we report the design and synthesis of a new class of neutral porphyrin derivative 5,10-bis(4-carboxyphenyl)-15, 20-bis(4-dimethylaminophenyl)porphyrin (compound 8) as a potent catalytic inhibitor of human Top1. In contrast to CPT, compound 8 reversibly binds with the free enzyme and inhibits the formation of Top1cc and promotes reversal of the preformed Top1cc with CPT. Compound 8 induced inhibition of Top1cc formation in live cells was substantiated by fluorescence recovery after photobleaching (FRAP) assays. We established that MCF7 cells treated with compound 8 trigger proteasome-mediated Top1 degradation, accumulate higher levels of reactive oxygen species (ROS), PARP1 cleavage, oxidative DNA fragmentation, and stimulate apoptotic cell death without stabilizing apoptotic Top1-DNA cleavage complexes. Finally, compound 8 shows anticancer activity by targeting cellular Top1 and preventing the enzyme from directly participating in the apoptotic process.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Clivage de l'ADN/effets des médicaments et des substances chimiques , ADN topoisomérases de type I/métabolisme , Porphyrines/composition chimique , Porphyrines/pharmacologie , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Biocatalyse/effets des médicaments et des substances chimiques , Cassures de l'ADN/effets des médicaments et des substances chimiques , Humains , Cellules MCF-7 , Espèces réactives de l'oxygène/métabolisme , Inhibiteurs de la topoisomérase-I/composition chimique , Inhibiteurs de la topoisomérase-I/pharmacologie
10.
Nucleic Acids Res ; 44(17): 8363-75, 2016 09 30.
Article de Anglais | MEDLINE | ID: mdl-27466387

RÉSUMÉ

Topoisomerase 1 (Top1) is essential for removing the DNA supercoiling generated during replication and transcription. Anticancer drugs like camptothecin (CPT) and its clinical derivatives exert their cytotoxicity by reversibly trapping Top1 in covalent complexes on the DNA (Top1cc). Poly(ADP-ribose) polymerase (PARP) catalyses the addition of ADP-ribose polymers (PAR) onto itself and Top1. PARP inhibitors enhance the cytotoxicity of CPT in the clinical trials. However, the molecular mechanism by which PARylation regulates Top1 nuclear dynamics is not fully understood. Using live-cell imaging of enhanced green fluorescence tagged-human Top1, we show that PARP inhibitors (Veliparib, ABT-888) delocalize Top1 from the nucleolus to the nucleoplasm, which is independent of Top1-PARP1 interaction. Using fluorescence recovery after photobleaching and subsequent fitting of the data employing kinetic modelling we demonstrate that ABT-888 markedly increase CPT-induced bound/immobile fraction of Top1 (Top1cc) across the nuclear genome, suggesting Top1-PARylation counteracts CPT-induced stabilization of Top1cc. We further show Trp205 and Asn722 of Top1 are critical for subnuclear dynamics. Top1 mutant (N722S) was restricted to the nucleolus in the presence of CPT due to its deficiency in the accumulation of CPT-induced Top1-PARylation and Top1cc formation. This work identifies ADP-ribose polymers as key determinant for regulating Top1 subnuclear dynamics.


Sujet(s)
Camptothécine/pharmacologie , Noyau de la cellule/métabolisme , ADN topoisomérases de type I/métabolisme , Poly adénosine diphosphate ribose/métabolisme , Benzimidazoles/pharmacologie , Nucléole/effets des médicaments et des substances chimiques , Nucléole/métabolisme , Noyau de la cellule/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , ADN/métabolisme , Diffusion , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Redistribution de fluorescence après photoblanchiment , Protéines à fluorescence verte/métabolisme , Cellules HCT116 , Humains , Cinétique , Protéines mutantes/métabolisme , Plasmides/métabolisme , Inhibiteurs de poly(ADP-ribose) polymérases/pharmacologie
11.
Eur J Med Chem ; 102: 540-51, 2015 Sep 18.
Article de Anglais | MEDLINE | ID: mdl-26312433

RÉSUMÉ

DNA topoisomerase I is a potential chemotherapeutic target. Here, we designed and synthesized a library comprising of hydantoin and thiohydantoin derivatives and tested them against human and Leishmania Top1. One of the thiohydantoin compounds with substituted thiophenyl as the central moiety (compound 15) exhibited potent inhibition of human Top1 (HTop1) through stabilization of Top1-DNA cleavage complexes and showed selective anticancer activity against human cervical carcinoma (HeLa) and breast carcinoma (MCF-7) cell lines. Molecular modeling studies with HTop1 rationalized the inhibitory mechanism of compound 15.


Sujet(s)
Antinéoplasiques/pharmacologie , ADN topoisomérases de type I/métabolisme , Conception de médicament , Thiohydantoïnes/pharmacologie , Inhibiteurs de la topoisomérase-I/pharmacologie , Antinéoplasiques/synthèse chimique , Antinéoplasiques/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Tests de criblage d'agents antitumoraux , Cellules HEK293 , Cellules HeLa , Humains , Cellules MCF-7 , Modèles moléculaires , Structure moléculaire , Relation structure-activité , Thiohydantoïnes/synthèse chimique , Thiohydantoïnes/composition chimique , Inhibiteurs de la topoisomérase-I/synthèse chimique , Inhibiteurs de la topoisomérase-I/composition chimique
12.
Nucleic Acids Res ; 42(7): 4435-49, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24493735

RÉSUMÉ

Poly(ADP-ribose) polymerases (PARP) attach poly(ADP-ribose) (PAR) chains to various proteins including themselves and chromatin. Topoisomerase I (Top1) regulates DNA supercoiling and is the target of camptothecin and indenoisoquinoline anticancer drugs, as it forms Top1 cleavage complexes (Top1cc) that are trapped by the drugs. Endogenous and carcinogenic DNA lesions can also trap Top1cc. Tyrosyl-DNA phosphodiesterase 1 (TDP1), a key repair enzyme for trapped Top1cc, hydrolyzes the phosphodiester bond between the DNA 3'-end and the Top1 tyrosyl moiety. Alternative repair pathways for Top1cc involve endonuclease cleavage. However, it is unknown what determines the choice between TDP1 and the endonuclease repair pathways. Here we show that PARP1 plays a critical role in this process. By generating TDP1 and PARP1 double-knockout lymphoma chicken DT40 cells, we demonstrate that TDP1 and PARP1 are epistatic for the repair of Top1cc. The N-terminal domain of TDP1 directly binds the C-terminal domain of PARP1, and TDP1 is PARylated by PARP1. PARylation stabilizes TDP1 together with SUMOylation of TDP1. TDP1 PARylation enhances its recruitment to DNA damage sites without interfering with TDP1 catalytic activity. TDP1-PARP1 complexes, in turn recruit X-ray repair cross-complementing protein 1 (XRCC1). This work identifies PARP1 as a key component driving the repair of trapped Top1cc by TDP1.


Sujet(s)
Altération de l'ADN , Réparation de l'ADN , Protéines de liaison à l'ADN/métabolisme , Phosphodiesterases/métabolisme , Poly(ADP-ribose) polymerases/métabolisme , Animaux , Lignée cellulaire tumorale , ADN topoisomérases de type I/métabolisme , Épistasie , Humains , Phosphodiesterases/composition chimique , Phosphodiesterases/génétique , Poly(ADP-ribose) polymerases/composition chimique , Poly(ADP-ribose) polymerases/génétique , Motifs et domaines d'intéraction protéique , Sumoylation , Protéine-1 de complémentation croisée de la réparation des lésions induites par les rayons X
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE