Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 75(2): 252-266.e8, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31202577

RESUMEN

Topoisomerase II (TOP2) relieves torsional stress by forming transient cleavage complex intermediates (TOP2ccs) that contain TOP2-linked DNA breaks (DSBs). While TOP2ccs are normally reversible, they can be "trapped" by chemotherapeutic drugs such as etoposide and subsequently converted into irreversible TOP2-linked DSBs. Here, we have quantified etoposide-induced trapping of TOP2ccs, their conversion into irreversible TOP2-linked DSBs, and their processing during DNA repair genome-wide, as a function of time. We find that while TOP2 chromatin localization and trapping is independent of transcription, it requires pre-existing binding of cohesin to DNA. In contrast, the conversion of trapped TOP2ccs to irreversible DSBs during DNA repair is accelerated 2-fold at transcribed loci relative to non-transcribed loci. This conversion is dependent on proteasomal degradation and TDP2 phosphodiesterase activity. Quantitative modeling shows that only two features of pre-existing chromatin structure-namely, cohesin binding and transcriptional activity-can be used to predict the kinetics of TOP2-induced DSBs.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo II/química , ADN/genética , Complejos Multiproteicos/química , Proteínas de Unión a Poli-ADP-Ribosa/química , Rotura Cromosómica , Cromosomas/genética , ADN/química , Reparación del ADN/genética , ADN-Topoisomerasas de Tipo II/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Etopósido/química , Conversión Génica/genética , Células HCT116 , Humanos , Cinética , Complejos Multiproteicos/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Inhibidores de Topoisomerasa II/química , Inhibidores de Topoisomerasa II/farmacología , Torsión Mecánica , Transcripción Genética , Translocación Genética/genética
2.
Mol Cell ; 74(3): 466-480.e4, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30930055

RESUMEN

The mTOR pathway integrates both extracellular and intracellular signals and serves as a central regulator of cell metabolism, growth, survival, and stress responses. Neurotropic viruses, such as herpes simplex virus-1 (HSV-1), also rely on cellular AKT-mTORC1 signaling to achieve viral latency. Here, we define a novel genotoxic response whereby spatially separated signals initiated by extracellular neurotrophic factors and nuclear DNA damage are integrated by the AKT-mTORC1 pathway. We demonstrate that endogenous DNA double-strand breaks (DSBs) mediated by Topoisomerase 2ß-DNA cleavage complex (TOP2ßcc) intermediates are required to achieve AKT-mTORC1 signaling and maintain HSV-1 latency in neurons. Suppression of host DNA-repair pathways that remove TOP2ßcc trigger HSV-1 reactivation. Moreover, perturbation of AKT phosphorylation dynamics by downregulating the PHLPP1 phosphatase led to AKT mis-localization and disruption of DSB-induced HSV-1 reactivation. Thus, the cellular genome integrity and environmental inputs are consolidated and co-opted by a latent virus to balance lifelong infection with transmission.


Asunto(s)
ADN-Topoisomerasas de Tipo II/genética , Herpesvirus Humano 1/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-akt/genética , Latencia del Virus/genética , Animales , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN/genética , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Herpesvirus Humano 1/patogenicidad , Humanos , Proteína Homóloga de MRE11/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Neuronas/metabolismo , Neuronas/virología , Fosforilación , Ratas , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética
3.
J Biol Chem ; 300(7): 107446, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38844130

RESUMEN

Meiosis reduces ploidy through two rounds of chromosome segregation preceded by one round of DNA replication. In meiosis I, homologous chromosomes segregate, while in meiosis II, sister chromatids separate from each other. Topoisomerase II (Topo II) is a conserved enzyme that alters DNA structure by introducing transient double-strand breaks. During mitosis, Topo II relieves topological stress associated with unwinding DNA during replication, recombination, and sister chromatid segregation. Topo II also plays a role in maintaining mitotic chromosome structure. However, the role and regulation of Topo II during meiosis is not well-defined. Previously, we found an allele of Topo II, top-2(it7), disrupts homologous chromosome segregation during meiosis I of Caenorhabditis elegans spermatogenesis. In a genetic screen, we identified different point mutations in 5'-tyrosyl-DNA phosphodiesterase two (Tdp2, C. elegans tdpt-1) that suppress top-2(it7) embryonic lethality. Tdp2 removes trapped Top-2-DNA complexes. The tdpt-1 suppressing mutations rescue embryonic lethality, ameliorate chromosome segregation defects, and restore TOP-2 protein levels of top-2(it7). Here, we show that both TOP-2 and TDPT-1 are expressed in germ line nuclei but occupy different compartments until late meiotic prophase. We also demonstrate that tdpt-1 suppression is due to loss of function of the protein and that the tdpt-1 mutations do not have a phenotype independent of top-2(it7) in meiosis. Lastly, we found that the tdpt-1 suppressing mutations either impair the phosphodiesterase activity, affect the stability of TDPT-1, or disrupt protein interactions. This suggests that the WT TDPT-1 protein is inhibiting chromosome biological functions of an impaired TOP-2 during meiosis.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Segregación Cromosómica , ADN-Topoisomerasas de Tipo II , Espermatogénesis , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Espermatogénesis/genética , Masculino , ADN-Topoisomerasas de Tipo II/metabolismo , ADN-Topoisomerasas de Tipo II/genética , Meiosis , Hidrolasas Diéster Fosfóricas/metabolismo , Hidrolasas Diéster Fosfóricas/genética , Mutación
4.
J Biol Chem ; 299(8): 104988, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37392847

RESUMEN

Topoisomerases are enzymes that relax DNA supercoiling during replication and transcription. Camptothecin, a topoisomerase 1 (TOP1) inhibitor, and its analogs trap TOP1 at the 3'-end of DNA as a DNA-bound intermediate, resulting in DNA damage that can kill cells. Drugs with this mechanism of action are widely used to treat cancers. It has previously been shown that tyrosyl-DNA phosphodiesterase 1 (TDP1) repairs TOP1-induced DNA damage generated by camptothecin. In addition, tyrosyl-DNA phosphodiesterase 2 (TDP2) plays critical roles in repairing topoisomerase 2 (TOP2)-induced DNA damage at the 5'-end of DNA and in promoting the repair of TOP1-induced DNA damage in the absence of TDP1. However, the catalytic mechanism by which TDP2 processes TOP1-induced DNA damage has not been elucidated. In this study, we found that a similar catalytic mechanism underlies the repair of TOP1- and TOP2-induced DNA damage by TDP2, with Mg2+-TDP2 binding playing a role in both repair mechanisms. We show chain-terminating nucleoside analogs are incorporated into DNA at the 3'-end and abort DNA replication to kill cells. Furthermore, we found that Mg2+-TDP2 binding also contributes to the repair of incorporated chain-terminating nucleoside analogs. Overall, these findings reveal the role played by Mg2+-TDP2 binding in the repair of both 3'- and 5'-blocking DNA damage.


Asunto(s)
Proteínas de Unión al ADN , Magnesio , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Magnesio/metabolismo , Nucleósidos , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Daño del ADN , ADN-Topoisomerasas de Tipo I/genética , ADN-Topoisomerasas de Tipo I/metabolismo , Inhibidores de Topoisomerasa , Camptotecina/farmacología , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , ADN , Reparación del ADN
5.
Genes Cells ; 28(1): 53-67, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36415926

RESUMEN

Steroid hormones induce the transcription of target genes by activating nuclear receptors. Early transcriptional response to various stimuli, including hormones, involves the active catalysis of topoisomerase II (TOP2) at transcription regulatory sequences. TOP2 untangles DNAs by transiently generating double-strand breaks (DSBs), where TOP2 covalently binds to DSB ends. When TOP2 fails to rejoin, called "abortive" catalysis, the resulting DSBs are repaired by tyrosyl-DNA phosphodiesterase 2 (TDP2) and non-homologous end-joining (NHEJ). A steroid, cortisol, is the most important glucocorticoid, and dexamethasone (Dex), a synthetic glucocorticoid, is widely used for suppressing inflammation in clinics. We here revealed that clinically relevant concentrations of Dex and physiological concentrations of cortisol efficiently induce DSBs in G1 phase cells deficient in TDP2 and NHEJ. The DSB induction depends on glucocorticoid receptor (GR) and TOP2. Considering the specific role of TDP2 in removing TOP2 adducts from DSB ends, induced DSBs most likely represent stalled TOP2-DSB complexes. Inhibition of RNA polymerase II suppressed the DSBs formation only modestly in the G1 phase. We propose that cortisol and Dex frequently generate DSBs through the abortive catalysis of TOP2 at transcriptional regulatory sequences, including promoters or enhancers, where active TOP2 catalysis occurs during early transcriptional response.


Asunto(s)
Roturas del ADN de Doble Cadena , Factores de Transcripción , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/metabolismo , Glucocorticoides/farmacología , Reparación del ADN , Proteínas Nucleares/metabolismo , Hidrocortisona/farmacología , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , ADN/genética
6.
Neurol Sci ; 45(6): 2881-2885, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38433132

RESUMEN

TDP2 gene encodes tyrosyl DNA phosphodiesterase 2, an enzyme required for effective repair of the DNA double-strand breaks (DSBs). Spinocerebellar ataxia autosomal recessive 23 (SCAR23) is a rare disease caused by the pathogenic mutation of TDP2 gene and characterized by intellectual disability, progressive ataxia and refractory epilepsy. Thus far, merely nine patients harboring five different variants (c.425 + 1G > A; c.413_414delinsAA, p. Ser138*; c.400C > T, p. Arg134*; c.636 + 3_ 636 + 6 del; c.4G > T, p. Glu2*) in TDP2 gene have been reported. Here, we describe the tenth patient with a novel variant (c.650del, p. Gly217GlufsTer7) and new phenotype (pituitary tumor and hyperhidrosis).


Asunto(s)
Hiperhidrosis , Hidrolasas Diéster Fosfóricas , Neoplasias Hipofisarias , Femenino , Humanos , Proteínas de Unión al ADN/genética , Hiperhidrosis/genética , Mutación , Hidrolasas Diéster Fosfóricas/genética , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/complicaciones , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/complicaciones , Adolescente , Lactante
7.
Molecules ; 29(3)2024 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-38338326

RESUMEN

Deoxycholic acid derivatives containing various heterocyclic functional groups at C-3 on the steroid scaffold were designed and synthesized as promising dual tyrosyl-DNA phosphodiesterase 1 and 2 (TDP1 and TDP2) inhibitors, which are potential targets to potentiate topoisomerase poison antitumor therapy. The methyl esters of DCA derivatives with benzothiazole or benzimidazole moieties at C-3 demonstrated promising inhibitory activity in vitro against TDP1 with IC50 values in the submicromolar range. Furthermore, methyl esters 4d-e, as well as their acid counterparts 3d-e, inhibited the phosphodiesterase activity of both TDP1 and TDP2. The combinations of compounds 3d-e and 4d-e with low-toxic concentrations of antitumor drugs topotecan and etoposide showed significantly greater cytotoxicity than the compounds alone. The docking of the derivatives into the binding sites of TDP1 and TDP2 predicted plausible binding modes of the DCA derivatives.


Asunto(s)
Inhibidores de Fosfodiesterasa , Hidrolasas Diéster Fosfóricas , Inhibidores de Fosfodiesterasa/química , Hidrolasas Diéster Fosfóricas/metabolismo , Modelos Moleculares , Ácido Desoxicólico/farmacología , Relación Estructura-Actividad
8.
Genes Cells ; 25(7): 450-465, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32277721

RESUMEN

Androgens stimulate the proliferation of epithelial cells in the prostate by activating topoisomerase 2 (TOP2) and regulating the transcription of target genes. TOP2 resolves the entanglement of genomic DNA by transiently generating double-strand breaks (DSBs), where TOP2 homodimers covalently bind to 5' DSB ends, called TOP2-DNA cleavage complexes (TOP2ccs). When TOP2 fails to rejoin TOP2ccs generating stalled TOP2ccs, tyrosyl DNA phosphodiesterase-2 (TDP2) removes 5' TOP2 adducts from stalled TOP2ccs prior to the ligation of the DSBs by nonhomologous end joining (NHEJ), the dominant DSB repair pathway in G0 /G1 phases. We previously showed that estrogens frequently generate stalled TOP2ccs in G0 /G1 phases. Here, we show that physiological concentrations of androgens induce several DSBs in individual human prostate cancer cells during G1 phase, and loss of TDP2 causes a five times higher number of androgen-induced chromosome breaks in mitotic chromosome spreads. Intraperitoneally injected androgens induce several DSBs in individual epithelial cells of the prostate in TDP2-deficient mice, even at 20 hr postinjection. In conclusion, physiological concentrations of androgens have very strong genotoxicity, most likely by generating stalled TOP2ccs.


Asunto(s)
Andrógenos/toxicidad , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Inestabilidad Genómica/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Próstata/metabolismo , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Rotura Cromosómica , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/genética , Proteínas de Unión al ADN/genética , Células Epiteliales/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Inestabilidad Genómica/efectos de los fármacos , Histonas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hidrolasas Diéster Fosfóricas/genética , Próstata/efectos de los fármacos , Neoplasias de la Próstata/genética , ARN Interferente Pequeño , Receptores Androgénicos/metabolismo
9.
EMBO Rep ; 20(10): e47517, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31393064

RESUMEN

In Caenorhabditis elegans, the JNK MAP kinase (MAPK) pathway is important for axon regeneration. The JNK pathway is activated by a signaling cascade consisting of the growth factor SVH-1 and its receptor tyrosine kinase SVH-2. Expression of the svh-2 gene is induced by axonal injury in a process involving the transcription factors ETS-4 and CEBP-1. Here, we find that svh-14/mxl-1, a gene encoding a Max-like transcription factor, is required for activation of svh-2 expression in response to axonal injury. We show that MXL-1 binds to and inhibits the function of TDPT-1, a C. elegans homolog of mammalian tyrosyl-DNA phosphodiesterase 2 [TDP2; also called Ets1-associated protein II (EAPII)]. Deletion of tdpt-1 suppresses the mxl-1 defect, but not the ets-4 defect, in axon regeneration. TDPT-1 induces SUMOylation of ETS-4, which inhibits ETS-4 transcriptional activity, and MXL-1 counteracts this effect. Thus, TDPT-1 interacts with two different transcription factors in axon regeneration.


Asunto(s)
Axones/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Regeneración Nerviosa , Hidrolasas Diéster Fosfóricas/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Sumoilación , Factores de Transcripción/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Modelos Biológicos , Neuronas Motoras/metabolismo , Fosforilación , Unión Proteica , Transcripción Genética
10.
Cell Mol Life Sci ; 77(1): 81-91, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31728578

RESUMEN

The compaction of DNA and the continuous action of DNA transactions, including transcription and DNA replication, create complex DNA topologies that require Type IIA Topoisomerases, which resolve DNA topological strain and control genome dynamics. The human TOP2 enzymes catalyze their reactions via formation of a reversible covalent enzyme DNA-protein crosslink, the TOP2 cleavage complex (TOP2cc). Spurious interactions of TOP2 with DNA damage, environmental toxicants and chemotherapeutic "poisons" perturbs the TOP2 reaction cycle, leading to an accumulation of DNA-protein crosslinks, and ultimately, genomic instability and cell death. Emerging evidence shows that TOP2-DNA protein crosslink (DPC) repair entails multiple strand break repair activities, such as removal of the poisoned TOP2 protein and rejoining of the DNA ends through homologous recombination (HR) or non-homologous end joining (NHEJ). Herein, we discuss the molecular mechanisms of TOP2-DPC resolution, with specific emphasis on the recently uncovered ZATTZnf451-licensed TDP2-catalyzed TOP2-DPC reversal mechanism.


Asunto(s)
Roturas del ADN , Reparación del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , ADN/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Aminoaciltransferasas/química , Aminoaciltransferasas/metabolismo , Animales , ADN/química , ADN/genética , ADN-Topoisomerasas de Tipo II/química , Humanos , Proteínas de Unión a Poli-ADP-Ribosa/química , Conformación Proteica , Sumoilación , Factores de Transcripción/química , Factores de Transcripción/metabolismo
11.
Cell Mol Life Sci ; 77(1): 1-2, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31712993

RESUMEN

DNA strand breaks present a complex challenge for our cells, and the integrity of the DNA damage response machinery is critical for preventing cancer, premature aging, and neurodegenerative syndromes amongst other ailments. This multi-author review issue presents emerging topics relevant to understanding the fundamental structural mechanisms of DNA strand break sensing, signaling, and repair.


Asunto(s)
Roturas del ADN , Reparación del ADN , Animales , ADN/genética , Inestabilidad Genómica , Humanos , Neoplasias/genética
12.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-34768766

RESUMEN

Tyrosyl-DNA phosphodiesterase 1 (TDP1) catalyzes the cleavage of the phosphodiester bond between the tyrosine residue of topoisomerase 1 (TOP1) and the 3' phosphate of DNA in the single-strand break generated by TOP1. TDP1 promotes the cleavage of the stable DNA-TOP1 complexes with the TOP1 inhibitor topotecan, which is a clinically used anticancer drug. This article reports the synthesis and study of usnic acid thioether and sulfoxide derivatives that efficiently suppress TDP1 activity, with IC50 values in the 1.4-25.2 µM range. The structure of the heterocyclic substituent introduced into the dibenzofuran core affects the TDP1 inhibitory efficiency of the compounds. A five-membered heterocyclic fragment was shown to be most pharmacophoric among the others. Sulfoxide derivatives were less cytotoxic than their thioester analogs. We observed an uncompetitive type of inhibition for the four most effective inhibitors of TDP1. The anticancer effect of TOP1 inhibitors can be enhanced by the simultaneous inhibition of PARP1, TDP1, and TDP2. Some of the compounds inhibited not only TDP1 but also TDP2 and/or PARP1, but at significantly higher concentration ranges than TDP1. Leader compound 10a showed promising synergy on HeLa cells in conjunction with the TOP1 inhibitor topotecan.


Asunto(s)
Benzofuranos/química , Proteínas de Unión al ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Sulfuros/química , Benzofuranos/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN/metabolismo , Inhibidores Enzimáticos/síntesis química , Humanos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Relación Estructura-Actividad , Sulfuros/farmacología , Sulfóxidos/química , Sulfóxidos/farmacología , Inhibidores de Topoisomerasa I/farmacología , Topotecan/farmacología
13.
Molecules ; 27(1)2021 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-35011303

RESUMEN

A series of deoxycholic acid (DCA) amides containing benzyl ether groups on the steroid core were tested against the tyrosyl-DNA phosphodiesterase 1 (TDP1) and 2 (TDP2) enzymes. In addition, 1,2,4- and 1,3,4-oxadiazole derivatives were synthesized to study the linker influence between a para-bromophenyl moiety and the steroid scaffold. The DCA derivatives demonstrated promising inhibitory activity against TDP1 with IC50 in the submicromolar range. Furthermore, the amides and the 1,3,4-oxadiazole derivatives inhibited the TDP2 enzyme but at substantially higher concentration. Tryptamide 5 and para-bromoanilide 8 derivatives containing benzyloxy substituent at the C-3 position and non-substituted hydroxy group at C-12 on the DCA scaffold inhibited both TDP1 and TDP2 as well as enhanced the cytotoxicity of topotecan in non-toxic concentration in vitro. According to molecular modeling, ligand 5 is anchored into the catalytic pocket of TDP1 by one hydrogen bond to the backbone of Gly458 as well as by π-π stacking between the indolyl rings of the ligand and Tyr590, resulting in excellent activity. It can therefore be concluded that these derivatives contribute to the development of specific TDP1 and TDP2 inhibitors for adjuvant therapy against cancer in combination with topoisomerase poisons.


Asunto(s)
Ácido Desoxicólico/análogos & derivados , Ácido Desoxicólico/química , Inhibidores de Fosfodiesterasa/química , Hidrolasas Diéster Fosfóricas/química , Sitios de Unión , Línea Celular , Fenómenos Químicos , Técnicas de Química Sintética , Ácido Desoxicólico/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Modelos Moleculares , Conformación Molecular , Estructura Molecular , Inhibidores de Fosfodiesterasa/síntesis química , Inhibidores de Fosfodiesterasa/farmacología , Hidrolasas Diéster Fosfóricas/metabolismo , Unión Proteica , Proteínas Recombinantes/química , Relación Estructura-Actividad
14.
Med Chem Res ; 30(2): 371-386, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33776385

RESUMEN

Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs topoisomerase II (Top2) mediated DNA damages, including double-strand breaks (DSBs) that underpin the anticancer mechanism of clinical TOP2 poisons such as etoposide (ETP). Inhibition of TDP2 could sensitize cancer cells toward TOP2 poisons by increasing Top2 cleavage complex. We have previously identified isoquinoline-1,3-dione as a selective inhibitor type of TDP2. However, the reported structure-activity relationship (SAR) was limited to simple substitutions on the isoquinoline-1,3-dione core. Herein, we report the extended SAR consisting of the synthesis and testing of a total of 50 analogs featuring N-2 and C-4 modifications. Major SAR observations include the loss of potency upon N-2 substitution, the lack of inhibition with C-4 enamine analogs (subtype 11), or any other C-4 modifications (subtypes 13-15) except for the benzylidene substitution (subtype 12), where eight analogs showed low micromolar potency. The best analog, 12q, inhibited TDP2 with an IC50 of 4.8 µM. Molecular modeling was performed to help understand the observed SAR trends. Overall, these SAR observations which could significantly benefit future work on the design of improved TDP2 inhibitors.

15.
J Biol Chem ; 294(8): 2665-2677, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30593505

RESUMEN

Tyrosyl-DNA phosphodiesterase 2 (TDP2) is a multifunctional protein that has been implicated in a myriad of cellular pathways. Although most well-known for its phosphodiesterase activity removing stalled topoisomerase 2 from DNA, TDP2 has also been shown to interact with both survival and apoptotic mitogen-activated protein kinase (MAPK) signaling cascades. Moreover, it facilitates enterovirus replication and has been genetically linked to neurological disorders ranging from Parkinson's disease to dyslexia. To accurately evaluate TDP2 as a therapeutic target, we need to understand how TDP2 performs such a wide diversity of functions. Here, we use cancer cell lines modified with CRISPR/Cas9 or stably-expressed TDP2-targeted shRNA and transfection of various TDP2 mutants to show that its expression is regulated at the translational level via an internal ribosome entry site (IRES) that initiates translation at codon 54, the second in-frame methionine of the TDP2 coding sequence. We observed that this IRES drives expression of a shorter, N-terminally truncated isoform of TDP2, ΔN-TDP2, which omits a nuclear localization sequence. Additionally, we noted that ΔN-TDP2 retains phosphodiesterase activity and is protective against etoposide-induced cell death, but co-immunoprecipitates with fewer high-molecular-weight ubiquitinated peptide species, suggesting partial loss-of-function of TDP2's ubiquitin-association domain. In summary, our findings suggest the existence of an IRES in the 5' coding sequence of TDP2 that translationally regulates expression of an N-terminally truncated, cytoplasmic isoform of TDP2. These results shed light on the regulation of this multifunctional protein and may inform the design of therapies targeting TDP2 and associated pathways.


Asunto(s)
Empalme Alternativo , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Sitios Internos de Entrada al Ribosoma/genética , Neoplasias/genética , Proteínas Nucleares/genética , Ribosomas/metabolismo , Factores de Transcripción/genética , Secuencia de Aminoácidos , Proteínas de Unión al ADN , Humanos , Neoplasias/enzimología , Neoplasias/patología , Iniciación de la Cadena Peptídica Traduccional , Hidrolasas Diéster Fosfóricas , Isoformas de Proteínas , Ribosomas/genética , Homología de Secuencia , Células Tumorales Cultivadas , Ubiquitina/metabolismo
16.
Cerebellum ; 18(5): 972-975, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31410782

RESUMEN

Spinocerebellar Ataxia 23 (SCAR23) is a newly described condition caused by mutations in TDP2 gene. To date, only four patients from two families have been reported, all carrying the same homozygous mutation. We describe a fifth patient, carrying a novel mutation in the same gene, thus confirming the role of TDP2 mutations in determining the disease and defining the main features SCAR23: pediatric onset ataxia and drug-resistant epilepsy and intellectual disability. We further show the clinical presentation which is associated with the neuroradiological evidence of progressive cerebellar atrophy, giving the evidence that SCAR23 can be classified as a degenerative condition.


Asunto(s)
Proteínas de Unión al ADN/genética , Epilepsia Refractaria/genética , Discapacidad Intelectual/genética , Mutación/genética , Hidrolasas Diéster Fosfóricas/genética , Ataxias Espinocerebelosas/genética , Adolescente , Epilepsia Refractaria/complicaciones , Epilepsia Refractaria/diagnóstico por imagen , Femenino , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/diagnóstico por imagen , Ataxias Espinocerebelosas/complicaciones , Ataxias Espinocerebelosas/diagnóstico por imagen , Secuenciación del Exoma/métodos
17.
Bioorg Med Chem Lett ; 29(2): 257-261, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30522956

RESUMEN

Tyrosyl-DNA phosphodiesterase 2 (TDP2) repairs topoisomerase II (TOP2) mediated DNA damages and causes cellular resistance to clinically used TOP2 poisons. Inhibiting TDP2 can potentially sensitize cancer cells toward TOP2 poisons. Commercial compound P10A10, to which the structure was assigned as 7-phenyl triazolopyrimidine analogue 6a, was previously identified as a TDP2 inhibitor hit in our virtual and fluorescence-based biochemical screening campaign. We report herein that the hit validation through resynthesis and structure elucidation revealed the correct structure of P10A10 (Chembridge ID 7236827) to be the 5-phenyl triazolopyrimidine regioisomer 7a. Subsequent structure-activity relationship (SAR) via the synthesis of a total of 47 analogues of both the 5-phenyl triazolopyrimidine scaffold (7) and its bioisosteric triazolopyridine scaffold (17) identified four derivatives (7a, 17a, 17e, and 17z) with significant TDP2 inhibition (IC50 < 50 µM), with 17z showing excellent cell permeability and no cytotoxicity.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Proteínas Nucleares/antagonistas & inhibidores , Piridinas/farmacología , Pirimidinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Triazoles/farmacología , Proteínas de Unión al ADN , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Estructura Molecular , Proteínas Nucleares/metabolismo , Hidrolasas Diéster Fosfóricas , Piridinas/síntesis química , Piridinas/química , Pirimidinas/síntesis química , Pirimidinas/química , Relación Estructura-Actividad , Factores de Transcripción/metabolismo , Triazoles/síntesis química , Triazoles/química
18.
Int J Mol Sci ; 20(12)2019 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-31226795

RESUMEN

Mammalian mitochondria contain four topoisomerases encoded in the nuclear genome: TOP1MT, TOP2α, TOP2ß, and TOP3α. They also contain the two known tyrosyl-DNA phosphodiesterases (TDPs): TDP1 and TDP2, including a specific TDP2S isoform. Both TDP1 and TDP2 excise abortive topoisomerase cleavage complexes (TOPccs), yet their molecular structures and mechanisms are different. TDP1 is present across eukaryotes, from yeasts to humans and belongs to the phospholipase D family. It functions without a metal cofactor and has a broad activity range, as it also serves to cleanse blocking 3'-DNA ends bearing phosphoglycolate, deoxyribose phosphate, nucleoside, nucleoside analogs (zidovudine), abasic moieties, and with a lower efficiency, TOP2ccs. Found in higher vertebrates, TDP2 is absent in yeast where TDP1 appears to perform its functions. TDP2 belongs to the exonuclease/endonuclease/phosphodiesterase family and requires magnesium as a cofactor to excise TOP2ccs, and it also excises TOP1ccs, albeit with a lower efficiency. Here, we review TDP1 and TDP2 in the context of mitochondrial DNA repair and discuss potential new research areas centered on the mitochondrial TDPs.


Asunto(s)
Reparación del ADN , ADN Mitocondrial/genética , Proteínas Nucleares/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Factores de Transcripción/metabolismo , Animales , Daño del ADN , ADN Mitocondrial/metabolismo , Proteínas de Unión al ADN , Humanos , Mitocondrias/genética , Mitocondrias/metabolismo
19.
Int J Mol Sci ; 19(7)2018 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-30011940

RESUMEN

DNA topoisomerase II (TOP2) activity involves a normally transient double-strand break intermediate in which the enzyme is coupled to DNA via a 5'-phosphotyrosyl bond. However, etoposide and other topoisomerase drugs poison the enzyme by stabilising this enzyme-bridged break, resulting in the accumulation of TOP2-DNA covalent complexes with cytotoxic consequences. The phosphotyrosyl diesterase TDP2 appears to be required for efficient repair of this unusual type of DNA damage and can remove 5'-tyrosine adducts from a double-stranded oligonucleotide substrate. Here, we adapt the trapped in agarose DNA immunostaining (TARDIS) assay to investigate the role of TDP2 in the removal of TOP2-DNA complexes in vitro and in cells. We report that TDP2 alone does not remove TOP2-DNA complexes from genomic DNA in vitro and that depletion of TDP2 in cells does not slow the removal of TOP2-DNA complexes. Thus, if TDP2 is involved in repairing TOP2 adducts, there must be one or more prior steps in which the protein-DNA complex is processed before TDP2 removes the remaining 5' tyrosine DNA adducts. While this is partly achieved through the degradation of TOP2 adducts by the proteasome, a proteasome-independent mechanism has also been described involving the SUMOylation of TOP2 by the ZATT E3 SUMO ligase. The TARDIS assay was also adapted to measure the effect of TDP2 knockdown on levels of SUMOylated TOP2-DNA complexes, which together with levels of double strand breaks were unaffected in K562 cells following etoposide exposure and proteasomal inhibition.


Asunto(s)
Reparación del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , ADN/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Factores de Transcripción/metabolismo , ADN/genética , Aductos de ADN/genética , Aductos de ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/genética , Proteínas de Unión al ADN , Etopósido/farmacología , Humanos , Células K562 , Proteínas Nucleares/genética , Hidrolasas Diéster Fosfóricas , Proteínas de Unión a Poli-ADP-Ribosa/antagonistas & inhibidores , Proteínas de Unión a Poli-ADP-Ribosa/genética , Interferencia de ARN , Sumoilación , Inhibidores de Topoisomerasa II/farmacología , Factores de Transcripción/genética
20.
Adv Exp Med Biol ; 1007: 157-178, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28840557

RESUMEN

Yeast research has been tremendously contributing to the understanding of a variety of molecular pathways due to the ease of its genetic manipulation, fast doubling time as well as being cost-effective. The understanding of these pathways did not only help scientists learn more about the cellular functions but also assisted in deciphering the genetic and cellular defects behind multiple diseases. Hence, yeast research not only opened the doors for transforming basic research into applied research, but also paved the roads for improving diagnosis and innovating personalized therapy of different diseases. In this chapter, we discuss how yeast research has contributed to understanding major genome maintenance pathways such as the S-phase checkpoint activation pathways, repair via homologous recombination and non-homologous end joining as well as topoisomerases-induced protein linked DNA breaks repair. Defects in these pathways lead to neurodegenerative diseases and cancer. Thus, the understanding of the exact genetic defects underlying these diseases allowed the development of personalized medicine, improving the diagnosis and treatment and overcoming the detriments of current conventional therapies such as the side effects, toxicity as well as drug resistance.


Asunto(s)
Daño del ADN , Reparación del ADN , Genoma , Medicina de Precisión , Saccharomyces cerevisiae/genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA