Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 67(10): 8161-8171, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38690856

RESUMO

The mediator kinases CDK8 and CDK19 control the dynamic transcription of selected genes in response to various signals and have been shown to be hijacked to sustain hyperproliferation by various solid and liquid tumors. CDK8/19 is emerging as a promising anticancer therapeutic target. Here, we report the discovery of compound 12, a novel small molecule CDK8/19 inhibitor. This molecule demonstrated not only decent enzymatic and cellular activities but also remarkable selectivity in CDK and kinome panels. Besides, compound 12 also displayed favorable ADME profiles including low CYP1A2 inhibition, acceptable clearance, and high oral bioavailability in multiple preclinical species. Robust in vivo PD and efficacy studies in mice models further demonstrated its potential use as mono- and combination therapy for the treatment of cancers.


Assuntos
Antineoplásicos , Quinase 8 Dependente de Ciclina , Quinases Ciclina-Dependentes , Inibidores de Proteínas Quinases , Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Quinase 8 Dependente de Ciclina/metabolismo , Humanos , Animais , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/uso terapêutico , Inibidores de Proteínas Quinases/síntese química , Quinases Ciclina-Dependentes/antagonistas & inibidores , Quinases Ciclina-Dependentes/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Antineoplásicos/síntese química , Camundongos , Descoberta de Drogas , Linhagem Celular Tumoral , Relação Estrutura-Atividade , Proliferação de Células/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Ratos
2.
Oncogene ; 43(11): 804-820, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38279062

RESUMO

HJURP is overexpressed in several cancer types and strongly correlates with patient survival. However, the mechanistic basis underlying the association of HJURP with cancer aggressiveness is not well understood. HJURP promotes the loading of the histone H3 variant, CENP-A, at the centromeric chromatin, epigenetically defining the centromeres and supporting proper chromosome segregation. In addition, HJURP is associated with DNA repair but its function in this process is still scarcely explored. Here, we demonstrate that HJURP is recruited to DSBs through a mechanism requiring chromatin PARylation and promotes epigenetic alterations that favor the execution of DNA repair. Incorporation of HJURP at DSBs promotes turnover of H3K9me3 and HP1, facilitating DNA damage signaling and DSB repair. Moreover, HJURP overexpression in glioma cell lines also affected global structure of heterochromatin independently of DNA damage induction, promoting genome-wide reorganization and assisting DNA damage response. HJURP overexpression therefore extensively alters DNA damage signaling and DSB repair, and also increases radioresistance of glioma cells. Importantly, HJURP expression levels in tumors are also associated with poor response of patients to radiation. Thus, our results enlarge the understanding of HJURP involvement in DNA repair and highlight it as a promising target for the development of adjuvant therapies that sensitize tumor cells to irradiation.


Assuntos
Cromatina , Glioma , Humanos , Centrômero/metabolismo , Proteína Centromérica A/genética , Proteína Centromérica A/metabolismo , Cromatina/genética , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Glioma/genética
3.
J Med Chem ; 67(1): 420-432, 2024 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-38146659

RESUMO

Breast and gynecological cancers are among the leading causes of death in women worldwide, illustrating the urgent need for innovative treatment options. We identified MYT1 as a promising new therapeutic target for breast and gynecological cancer using PandaOmics, an AI-driven target discovery platform. The synthetic lethal relationship of MYT1 in tumor cell lines with CCNE1 amplification enhanced this rationale. Through structure-based drug design, we developed a series of novel, potent, and highly selective inhibitors specifically targeting MYT1. Importantly, our lead compound, featuring a tetrahydropyrazolopyrazine ring, exhibits remarkable selectivity over WEE1, a related kinase associated with bone marrow suppression upon inhibition. Optimization of potency and physical properties resulted in the discovery of compound 21, a novel MYT1 inhibitor, exhibiting optimal pharmacokinetic properties and promising in vivo antitumor efficacy.


Assuntos
Antineoplásicos , Neoplasias , Feminino , Humanos , Linhagem Celular Tumoral , Mama , Desenho de Fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Proliferação de Células , Neoplasias/tratamento farmacológico , Proteínas de Ligação a DNA/metabolismo , Fatores de Transcrição/metabolismo
4.
J Med Chem ; 66(8): 5439-5452, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37029334

RESUMO

Cyclin-dependent kinase 8 (CDK8), as a kinase subunit of the Mediator complex, is involved in the regulation of RNA polymerase II-mediated transcription, thereby modulating multiple signaling pathways and multiple transcription factors involved in oncogenic control. CDK8 deregulation has been implicated in human diseases, particularly in acute myeloid leukemia (AML) and advanced solid tumors, where it has been reported as a putative oncogene. Here, we report the successful optimization of an azaindole series of CDK8 inhibitors that were identified and further progressed through a structure-based generative chemistry approach. In several optimization cycles, we improved in vitro microsomal stability, kinase selectivity, and in vivo pharmacokinetic profile cross-species, leading to the discovery of compound 23, which demonstrated robust tumor growth inhibition in multiple in vivo efficacy models after oral administration.


Assuntos
Quinase 8 Dependente de Ciclina , Neoplasias , Humanos , Neoplasias/genética , Complexo Mediador/metabolismo , Oncogenes , Inibidores de Proteínas Quinases/farmacologia
5.
Cell Rep ; 41(12): 111866, 2022 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-36543120

RESUMO

DNA damage leads to rapid synthesis of poly(ADP-ribose) (pADPr), which is important for damage signaling and repair. pADPr chains are removed by poly(ADP-ribose) glycohydrolase (PARG), releasing free mono(ADP-ribose) (mADPr). Here, we show that the NUDIX hydrolase NUDT5, which can hydrolyze mADPr to ribose-5-phosphate and either AMP or ATP, is recruited to damage sites through interaction with PARG. NUDT5 does not regulate PARP or PARG activity. Instead, loss of NUDT5 reduces basal cellular ATP levels and exacerbates the decrease in cellular ATP that occurs during DNA repair. Further, loss of NUDT5 activity impairs RAD51 recruitment, attenuates the phosphorylation of key DNA-repair proteins, and reduces both H2A.Z exchange at damage sites and repair by homologous recombination. The ability of NUDT5 to hydrolyze mADPr, and/or regulate cellular ATP, may therefore be important for efficient DNA repair. Targeting NUDT5 to disrupt PAR/mADPr and energy metabolism may be an effective anti-cancer strategy.


Assuntos
Adenosina Difosfato Ribose , Reparo do DNA , Adenosina Difosfato Ribose/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Glicosídeo Hidrolases/metabolismo , Dano ao DNA , Trifosfato de Adenosina
6.
Appl Microbiol Biotechnol ; 106(19-20): 6701-6711, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36097173

RESUMO

Highland birds evolve multiple adaptive abilities to cope with the harsh environments; however, how they adapt to the high-altitude habitats via the gut microbiota remains understudied. Here we integrated evidences from comparative analysis of gut microbiota to explore the adaptive mechanism of black-necked crane, a typical highland bird in the Qinghai-Tibet Plateau. Firstly, the gut microbiota diversity and function was compared among seven crane species (one high-altitude species and six low-altitude species), and then among three populations of contrasting altitudes for the black-necked crane. Microbiota community diversity in black-necked crane was significantly lower than its low-altitude relatives, but higher microbiota functional diversity was observed in black-necked crane, suggesting that unique bacteria are developed and acquired due to the selection pressure of high-altitude environments. The functional microbial genes differed significantly between the low- and high-altitude black-necked cranes, indicating that altitude significantly impacted microbial communities' composition and structure. Adaptive changes in microbiota diversity and function are observed in response to high-altitude environments. These findings provide us a new insight into the adaptation mechanism to the high-altitude environment for birds via the gut microbiota. KEY POINTS: • The diversity and function of gut microbiota differed significantly between the low- and high-altitude crane species. • Black-necked crane adapts to the high-altitude environment via specific gut microbiota. • Altitude significantly impacted microbial communities' composition and structure.


Assuntos
Microbioma Gastrointestinal , Aclimatação , Altitude , Animais , Aves , Microbioma Gastrointestinal/fisiologia , Tibet
7.
J Colloid Interface Sci ; 599: 370-380, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33962198

RESUMO

It is imperative but challenging to develop non-noble metal-based bifunctional electrocatalysts for oxygen evolution reaction (OER) and hydrogen evolution reaction (HER). Our work reports a core-shell nanostructure that is constructed by the electrodeposition of ultrathin NiFe-LDH nanosheets (NiFe-LDHNS) on Cu2Se nanowires, which are obtained by selenizing Cu(OH)2 nanowires in situ grown on Cu foam. The obtained Cu2Se@NiFe-LDHNS electrocatalyst provides more exposed edges and catalytic active sites, thus exhibiting excellent OER and HER electrocatalytic performance in alkaline electrolytes. This catalyst needs only an overpotential of 197 mV for OER at 50 mA cm-2 and 195 mV for HER at 10 mA cm-2. Besides, when employed as a bifunctional catalyst for overall water-splitting, it requires a cell voltage of 1.67 V to reach 10 mA cm-2 in alkaline media. Furthermore, the corresponding water electrolyzer demonstrates robust durability for at least 40 h. The excellent performance of Cu2Se@NiFe-LDHNS might be ascribed to the synergistic effect from the ultrathin NiFe-LDHNS, the Cu2Se nanowires anchored on the Cu foam, and the formed core-shell nanostructure, which offers large surface area, ample active sites, and sufficient channels for gas and electrolyte diffusion. This work provides an efficient strategy for the fabrication of self-supported electrocatalysts for efficient overall water-splitting.

8.
Trends Genet ; 35(2): 159-172, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30595401

RESUMO

Although stress-induced synthesis of mono(ADP-ribose) (mADPr) and poly(ADP-ribose) (pADPr) conjugates by pADPr polymerase (PARP) enzymes has been studied extensively, the removal and degradation of pADPr, as well as the fate of ADPr metabolites, have received less attention. The observations that stress-induced pADPr undergoes rapid turnover, and that deficiencies in ADPr degradation phenocopy loss of pADPr synthesis, suggest that ADPr degradation is fundamentally important to the cellular stress response. Recent work has identified several distinct families of pADPr hydrolases that can degrade pADPr to release pADPr or mADPr into the cytoplasm. Further, many stress-response proteins contain ADPr-binding domains that can interact with these metabolites. We discuss how pADPr metabolites generated during pADPr degradation can function as signaling intermediates in processes such as inflammation, apoptosis, and DNA damage responses. These studies highlight that the full cycle of ADPr metabolism, including both synthesis and degradation, is necessary for responses to genotoxic stress.


Assuntos
ADP-Ribosilação/genética , Poli Adenosina Difosfato Ribose/genética , Proteínas/genética , Estresse Fisiológico/genética , Dano ao DNA/genética , Domínios Proteicos/genética , Processamento de Proteína Pós-Traducional/genética , Transdução de Sinais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...