Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38895204

RESUMO

Mutations in coiled-coil-helix-coiled-coil-helix domain containing 10( CHCHD10 ) have been identified as a genetic cause of amyotrophic lateral sclerosis and/or frontotemporal dementia(ALS-FTD). In our previous studies using in vivo Drosophila model expressing C2C10H S81L , and human cell models expressing CHCHD10 S59L , we have identified that the PINK1/Parkin pathway is activated and causes cellular toxicity. Furthermore, we demonstrated that pseudo-substrate inhibitors for PINK1 and mitofusin2 agonists mitigated the cellular toxicity of CHCHD10 S59L . Evidences using in vitro/ in vivo genetic and chemical tools indicate that inhibiting PINK1 would be the most promising treatment for CHCHD10 S59L -induced diseases. Therefore, we have investigated cellular pathways that can modulate the PINK1/Parkin pathway and reduce CHCHD10 S59L -induced cytotoxicity. Here, we report that FDA-approved PDE4 inhibitors reduced CHCHD10 S59L -induced morphological and functional mitochondrial defects in human cells and an in vivo Drosophila model expressing C2C10H S81L . Multiple PDE4 inhibitors decreased PINK1 accumulation and downstream mitophagy induced by CHCHD10 S59L . These findings suggest that PDE4 inhibitors currently available in the market may be repositioned to treat CHCHD10 S59L -induced ALS-FTD and possibly other related diseases.

2.
iScience ; 25(9): 105017, 2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36105584

RESUMO

The heterotypic CIC structures formed of cancer and immune cells have been observed in tumor tissues. We aimed to assess the feasibility of using heterotypic CICs as a functional biomarker to predict NK susceptibility and drug resistance. The heterotypic CIC-forming cancer cells showed a lower response to NK cytotoxicity and higher proliferative ability than non-CIC cancer cells. After treatment with anticancer drugs, cancer cells that formed heterotypic CICs showed a higher resistance to anticancer drugs than non-CIC cancer cells. We also observed the formation of more CIC structures in cancer cells treated with anticancer drugs than in the non-treated group. Our results confirm the association between heterotypic CIC structures and anticancer drug resistance in CICs formed from NK and cancer cells. These results suggest a mechanism underlying immune evasion in heterotypic CIC cancer cells and provide insights into the anticancer drug resistance of cancer cells.

3.
JACS Au ; 1(8): 1158-1177, 2021 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-34467355

RESUMO

NO3 • can compete with omnipotent •OH/SO4 •- in decomposing aqueous pollutants because of its lengthy lifespan and significant tolerance to background scavengers present in H2O matrices, albeit with moderate oxidizing power. The generation of NO3 •, however, is of grand demand due to the need of NO2 •/O3, radioactive element, or NaNO3/HNO3 in the presence of highly energized electron/light. This study has pioneered a singular pathway used to radicalize surface NO3 - functionalities anchored on polymorphic α-/γ-MnO2 surfaces (α-/γ-MnO2-N), in which Lewis acidic Mn2+/3+ and NO3 - served to form •OH via H2O2 dissection and NO3 • via radical transfer from •OH to NO3 - (•OH → NO3 •), respectively. The elementary steps proposed for the •OH → NO3 • route could be energetically favorable and marginal except for two stages such as endothermic •OH desorption and exothermic •OH-mediated NO3 - radicalization, as verified by EPR spectroscopy experiments and DFT calculations. The Lewis acidic strength of the Mn2+/3+ species innate to α-MnO2-N was the smallest among those inherent to α-/ß-/γ-MnO2 and α-/γ-MnO2-N. Hence, α-MnO2-N prompted the rate-determining stage of the •OH → NO3 • route (•OH desorption) in the most efficient manner, as also evidenced by the analysis on the energy barrier required to proceed with the •OH → NO3 • route. Meanwhile, XANES and in situ DRIFT spectroscopy experiments corroborated that α-MnO2-N provided a larger concentration of surface NO3 - species with bi-dentate binding arrays than γ-MnO2-N. Hence, α-MnO2-N could outperform γ-MnO2-N in improving the collision frequency between •OH and NO3 - species and in facilitating the exothermic transition of NO3 - functionalities to surface NO3 • analogues per unit time. These were corroborated by a greater efficiency of α-MnO2-N in decomposing phenol, in addition to scavenging/filtration control runs and DFT calculations. Importantly, supported NO3 • species provided 5-7-fold greater efficiency in degrading textile wastewater than conventional •OH and supported SO4 •- analogues we discovered previously.

5.
Nat Commun ; 12(1): 1924, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33772006

RESUMO

Mutations in coiled-coil-helix-coiled-coil-helix domain containing 10 (CHCHD10) can cause amyotrophic lateral sclerosis and frontotemporal dementia (ALS-FTD). However, the underlying mechanisms are unclear. Here, we generate CHCH10S59L-mutant Drosophila melanogaster and HeLa cell lines to model CHCHD10-associated ALS-FTD. The CHCHD10S59L mutation results in cell toxicity in several tissues and mitochondrial defects. CHCHD10S59L independently affects the TDP-43 and PINK1 pathways. CHCHD10S59L expression increases TDP-43 insolubility and mitochondrial translocation. Blocking TDP-43 mitochondrial translocation with a peptide inhibitor reduced CHCHD10S59L-mediated toxicity. While genetic and pharmacological modulation of PINK1 expression and activity of its substrates rescues and mitigates the CHCHD10S59L-induced phenotypes and mitochondrial defects, respectively, in both Drosophila and HeLa cells. Our findings suggest that CHCHD10S59L-induced TDP-43 mitochondrial translocation and chronic activation of PINK1-mediated pathways result in dominant toxicity, providing a mechanistic insight into the CHCHD10 mutations associated with ALS-FTD.


Assuntos
Esclerose Lateral Amiotrófica/genética , Proteínas de Ligação a DNA/genética , Proteínas de Drosophila/genética , Demência Frontotemporal/genética , Proteínas Mitocondriais/genética , Mutação , Proteínas Serina-Treonina Quinases/genética , Sequência de Aminoácidos , Esclerose Lateral Amiotrófica/metabolismo , Animais , Animais Geneticamente Modificados , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila/metabolismo , Demência Frontotemporal/metabolismo , Células HEK293 , Células HeLa , Humanos , Microscopia Confocal , Mitocôndrias/genética , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Transporte Proteico/genética , Homologia de Sequência de Aminoácidos
7.
Oncotarget ; 8(41): 69691-69708, 2017 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-29050234

RESUMO

The Aurora kinases, Aurora A (AURKA), Aurora B (AURKB), and Aurora C (AURKC), are serine/threonine kinases required for the control of mitosis (AURKA and AURKB) or meiosis (AURKC). Several Aurora kinase inhibitors are being investigated as novel anticancer therapeutics. Recent studies demonstrated that AURKC activation contributes to breast cancer cell transformation. Therefore, AURKC is both a promising marker and therapeutic target for breast cancer; however, its signaling network has not been fully characterized. Using translocation-based cellular assays, we identified IκBα as a binding partner of AURKC, and found that AURKC phosphorylates IκBα at Ser32, thereby activating it. In silico modeling and computational analyses revealed a small-molecule inhibitor (AKCI) that blocked the AURKC-IκBα interaction and exerted antitumor activity in MDA-MB-231 breast cancer cells. Specifically, AKCI induced G2/M cell-cycle arrest through modulation of the p53/p21/CDC2/cyclin B1 pathways. In addition, the drug significantly inhibited MDA-MB-231 cell migration and invasion, as well as decreasing colony formation and tumor growth. Via its interaction with IκBα, AURKC indirectly induced NF-κB activation; accordingly, AKCI decreased PMA-induced activation of NF-κB. Thus, the small-molecule inhibitor AKCI represents a first step towards developing targeted inhibitors of AURKC protein binding, which may lead to further advances in the treatment of breast cancer.

8.
Int J Oncol ; 45(2): 675-82, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24867259

RESUMO

Nutlin-3 which occupies the p53 binding pocket in HDM2, has been reported to activate apoptosis through both the transcriptional activity-dependent and -independent programs of p53. Transcription-independent apoptosis by nutlin-3 is triggered by p53 which is translocated to mitochondria. However, we previously demonstrated that the nutlin-3-induced mitochondrial translocation of p53 stimulates ERK1/2 activation, an anti-apoptosis signal, via mitochondrial ROS generation. We report on how nutlin-3-stimulated ERK1/2 activity inhibits p53-induced apoptosis. Among the anti-apoptotic BCL2 family proteins, BCL2A1 expression was increased by nutlin-3 at both the mRNA and protein levels, and this increase was prevented by the inhibition of ERK1/2. TEMPO, a ROS scavenger, and PFT-µ , a blocker of the mitochondrial translocation of p53, also inhibited BCL2A1 expression as well as ERK1/2 phosphorylation. In addition, nutlin-3 stimulated phosphorylation of ELK1, which was prevented by all compounds that inhibited nutlin-3-induced ERK1/2 such as U0126, PFT-µ and TEMPO. Moreover, an increase in BCL2A1 expression was weakened by the knockdown of ELK1. Finally, nutlin-3-induced apoptosis was found to be potentiated by the knockdown of BCL2A1, as demonstrated by an increase of in hypo-diploidic cells and Annexin V-positive cells. Parallel to the increase in apoptotic cells, the knockdown of BCL2A1 augmented the cleavage of poly(ADP-ribose) polymerase-1. It is noteworthy that the augmented levels of apoptosis induced by the knockdown of BCL2A1 were comparable to those of apoptosis induced by U0126. Collectively, these results suggest that nutlin-3-activated ERK1/2 may stimulate the transcription of BCL2A1 via the activation of ELK1, and BCL2A1 expression may contribute to the inhibitory effect of ERK1/2 on nutlin-3-induced apoptosis, thereby constituting a negative feedback loop of p53-induced apoptosis.


Assuntos
Apoptose/fisiologia , Imidazóis/farmacologia , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Transdução de Sinais/fisiologia , Proteínas Elk-1 do Domínio ets/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Humanos , Immunoblotting , Sistema de Sinalização das MAP Quinases/fisiologia , Antígenos de Histocompatibilidade Menor , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Osteossarcoma/metabolismo , RNA Interferente Pequeno , Espécies Reativas de Oxigênio/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transfecção , Proteína Supressora de Tumor p53/metabolismo
9.
Oncol Rep ; 31(1): 131-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24190574

RESUMO

Nutlin-3, a human double minute 2 (HDM2) antagonist, induces cell cycle arrest or apoptosis by upregulating p53 in cancer cells. WT1, the product of Wilms' tumor gene 1, has been shown to interact with p53, but the effect of WT1 on nutlin-3-induced apoptosis has yet to be examined. To address this issue, we analyzed the inhibitory effect of nutlin-3 on cell growth as a function of Wt1 expression status using a Wt1-inducible U2OS cell line. In the absence of Wt1 expression, nutlin-3 induced cell cycle arrest with marginal cytotoxicity. Furthermore, upon Wt1 expression, nutlin-3 exerted a marked degree of cell death, as evidenced by the accumulation of hypo-diploid cells and LDH release. During cell death induction, cytochrome c was released into the cytosol, and caspase-9 and -3 were activated, suggesting that an intrinsic apoptotic pathway may be involved in this cell death. Consistent with this, z-VAD-Fmk, a pan-caspase inhibitor and the overexpression of BCL-XL attenuated the cell death. Nutlin-3 caused an increase in the mRNA levels of both BCL-XL and BAK, as well as their corresponding protein levels in mitochondria. In the presence of Wt1, nutlin-3-induced BCL-XL expression was attenuated while the expression of nutlin-3-induced BAK was potentiated. Collectively, these results suggest that WT1 potentiates nutlin-3-induced apoptosis by downregulating the expression of BCL-XL while upregulating that of BAK, which leads to the activation of an intrinsic apoptotic pathway.


Assuntos
Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Imidazóis/farmacologia , Piperazinas/farmacologia , Proteínas WT1/biossíntese , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/genética , Caspase 3/metabolismo , Caspase 9/metabolismo , Inibidores de Caspase/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocromos c/metabolismo , Regulação para Baixo , Ativação Enzimática , Humanos , Mitocôndrias , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , RNA Mensageiro/biossíntese , Proteína Supressora de Tumor p53/biossíntese , Regulação para Cima , Proteínas WT1/genética , Proteína Killer-Antagonista Homóloga a bcl-2/biossíntese , Proteína Killer-Antagonista Homóloga a bcl-2/genética , Proteína bcl-X/biossíntese , Proteína bcl-X/genética
10.
J Biol Chem ; 289(1): 112-21, 2014 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-24275661

RESUMO

Tight regulation of autophagy is critical for the fate of pancreatic ß cells. The autophagy protein ATG5 is essential for the formation of autophagosomes by promoting the lipidation of microtubule-associated protein LC3 (light chain 3). However, little is known about the mechanisms that regulate ATG5 expression levels. In this study, we investigated the regulation of ATG5 expression by HuD. The association of HuD with ATG5 mRNA was analyzed by ribonucleoprotein complex immunoprecipitation and biotin pulldown assays. HuD expression levels in pancreatic ß cells were knocked down via siRNA, elevated by overexpression of a HuD-expressing plasmid. The expression levels of HuD, ATG5, LC3, and ß-actin were determined by Western blot and quantitative RT-PCR analysis. Autophagosome formation was assessed by fluorescence microscopy in GFP-LC3-expressing cells and in pancreatic tissues from WT and HuD-null mice. We identified ATG5 mRNA as a post-transcriptional target of the mammalian RNA-binding protein HuD in pancreatic ß cells. HuD associated with the 3'-UTR of the ATG5 mRNA. Modulating HuD abundance did not alter ATG5 mRNA levels, but HuD silencing decreased ATG5 mRNA translation, and, conversely, HuD overexpression enhanced ATG5 mRNA translation. Through its effect on ATG5, HuD contributed to the lipidation of LC3 and the formation of LC3-positive autophagosomes. In keeping with this regulatory paradigm, HuD-null mice displayed lower ATG5 and LC3 levels in pancreatic ß cells. Our results reveal HuD to be an inducer of ATG5 expression and hence a critical regulator of autophagosome formation in pancreatic ß cells.


Assuntos
Proteínas ELAV/metabolismo , Regulação da Expressão Gênica/fisiologia , Células Secretoras de Insulina/metabolismo , Proteínas Associadas aos Microtúbulos/biossíntese , Fagossomos/metabolismo , Biossíntese de Proteínas/fisiologia , Regiões 3' não Traduzidas/fisiologia , Actinas/genética , Actinas/metabolismo , Animais , Proteína 5 Relacionada à Autofagia , Linhagem Celular , Proteínas ELAV/genética , Proteína Semelhante a ELAV 4 , Lipoilação/fisiologia , Camundongos , Camundongos Mutantes , Proteínas Associadas aos Microtúbulos/genética , Fagossomos/genética
11.
Int J Oncol ; 44(3): 761-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24366007

RESUMO

A recent study reported that p53 can induce HO-1 by directly binding to the putative p53 responsive element in the HO-1 promoter. In this study, we report that nutlin-3, a small molecule antagonist of HDM2, induces the transcription of HO-1 in a transcription-independent manner of p53. Nutlin-3 induced HO-1 expression at the level of transcription in human cancer cells such as U2OS and RKO cells. This induction of HO-1 did not occur in SAOS cells in which p53 was mutated and was prevented by knocking down the p53 protein using p53 siRNA transfection, but not by PFT-α, an inhibitor of the transcriptional activity of p53. Accompanying HO-1 expression, nutlin-3 stimulated the accumulation of ROS and the phosphorylation of MAPKs such as JNK, p38 MAPK and ERK1/2. Nutlin-3-induced HO-1 expression was suppressed by TEMPO, a ROS scavenger, and chemical inhibitors of JNK and p38 MAPK but not ERK1/2. In addition, nutlin­3-induced phosphorylation of JNK but not p38 MAPK was inhibited by TEMPO. Notably, the levels of nutlin-3-induced ROS were correlated with the mitochondrial translocation of p53 and this induction was prevented by PFT-µ, an inhibitor of the mitochondrial translocation of p53. Consistent with the effect of the ROS scavenger and MAPK inhibitors, PFT-µ reduced HO-1 expression and the phosphorylation of JNK induced by nutlin-3. In the experiments of analyzing cell death, the knockdown of HO-1 augmented nutlin-3-induced apoptosis. Collectively, these results suggest that nutlin-3 induces HO-1 expression via the activation of both JNK which is dependent on ROS generated by p53 translocated to the mitochondria and p38 MAPK which appears to be stimulated by a ROS-independent mechanism, and this HO-1 induction may inhibit nutlin-3-induced apoptosis, constituting a negative feedback loop of p53-induced apoptosis.


Assuntos
Heme Oxigenase-1/biossíntese , Imidazóis/administração & dosagem , MAP Quinase Quinase 4/biossíntese , Piperazinas/administração & dosagem , Proteína Supressora de Tumor p53/genética , Apoptose/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Heme Oxigenase-1/genética , Humanos , MAP Quinase Quinase 4/genética , Fosforilação , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , RNA Interferente Pequeno , Espécies Reativas de Oxigênio/metabolismo , Transcrição Gênica , Proteínas Quinases p38 Ativadas por Mitógeno/genética
12.
Oncol Rep ; 28(6): 2049-56, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22992992

RESUMO

Anthocyanins (ATCs) have been reported to induce apoptosis in various types of cancer cells, stimulating the development of ATCs as a cancer chemotherapeutic or chemopreventive agent. It was recently reported that ATCs can induce autophagy, however, the mechanism for this remains unclear. In the present report, we carried out mechanistic studies of the mechanism involved in ATC-induced autophagy using ATCs extracted from black soybeans (cv. Cheongja 3, Glycine max L.). ATCs clearly induced hallmarks of autophagy, including LC3 puncta formation and the conversion of LC3-I to LC3-II in U2OS human osteosarcoma cells. The induction of autophagy was accompanied by the phosphorylation of multiple protein kinases including extracellular signal-regulated kinase (ERK)1/2, p38 mitogen-activated protein kinase (MAPK), c-Jun N-terminal kinase (JNK), protein kinase B (AKT) and adenosyl mono-phosphate-dependent protein kinase (AMPK). While chemical inhibitors against ERK1/2, p38 MAPK, JNK and AKT failed to inhibit ATC-induced autophagy, the suppression of AMPK by compound C (CC) as well as siRNA against AMPK reduced ATC-induced autophagy. The treatment of ATCs resulted in a decrease in intracellular ATP contents and the activation of AMPK by AICAR treatment also induced autophagy. It is noteworthy that the reduction of autophagy via the inhibition of AMPK resulted in enhanced apoptosis in ATC-treated cells. In addition, siRNA against forkhead box O3A (FOXO3a), a downstream target of AMPK, suppressed ATC-induced autophagy and p27KIP1 siRNA increased apoptosis in ATC-treated cells. Collectively, it can be concluded that ATCs induce autophagy in U2OS cells via activation of the AMPK-FOXO3a pathway and protect cells from ATC-induced apoptosis via the AMPK-p27KIP1 pathway. These results also suggest that autophagy-modulating agents could contribute to the efficient development of ATCs as anticancer therapy.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antocianinas/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Glycine max/química , Osteossarcoma/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Apoptose/genética , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/genética , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Humanos , Osteossarcoma/patologia , Fosforilação , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais
13.
Korean J Physiol Pharmacol ; 14(6): 407-12, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21311682

RESUMO

3-Deazaadenosine (DZA), a potent inhibitor of S-adenosylhomocysteine hydrolase, was previously proposed to induce intrinsic apoptosis in human leukemic cells. In the present study, we analyzed the mechanism underlying the DZA-induced intrinsic apoptotic pathway. DZA activated typical caspase-dependent apoptosis in HL-60 cells, as demonstrated by an accumulation of hypo-diploidic cells, the processing of multiple procaspases and an inhibitory effect of z-VAD-Fmk on this cell death. During DZA-induced apoptosis, cytochrome c (cyt c) was released into the cytosol. This was neither prevented by z-VAD-Fmk and nor was it associated with the dissipation of mitochondrial membrane potential (ΔΨ(m)). Prior to the release of cyt c, BAX was translocated from the cytosol to mitochondria and underwent oligomerization. Finally, the overexpression of BCL-XL protected HL-60 cells from apoptosis by blocking both the cyt c release and BAX oligomerization. Collectively, these findings suggest that DZA may activate intrinsic apoptosis by stimulating BAX activation and thereby the release of cyt c.

14.
Arch Pharm (Weinheim) ; 341(10): 661-4, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18816587

RESUMO

3,4-Dihydroquinazoline derivatives have been known to be the novel and potent T-type calcium channel blockers. From a systematic variation of 3,4-dihydroquinazoline derivative 5c (KYS05043), plausible SAR results were established. It was revealed that a 5-(dimethylamino)pentylamino group at R(1), a biphenyl group at R(2), and a benzyl amido group at R(3)in the 3,4-dihydroquinazoline backbone are closely related with the channel selectivity (T/N-type) as well as the potency based on the discovery of 6k (KYS05090).


Assuntos
Bloqueadores dos Canais de Cálcio/síntese química , Canais de Cálcio Tipo T/metabolismo , Desenho de Fármacos , Quinazolinas/síntese química , Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , Humanos , Estrutura Molecular , Quinazolinas/química , Quinazolinas/farmacologia , Relação Estrutura-Atividade
15.
Bioorg Med Chem Lett ; 18(14): 3899-901, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18585035

RESUMO

In order to further clarify the role of T-type Ca(2+) channels in cell proliferation, we have measured the growth inhibition of human cancer cells by using our potent T-type Ca(2+) channel blockers. As a result, KYS05090, a most potent T-type Ca(2+) channel blocker, was found to be as potent as doxorubicin against some human cancer cells without acute toxicity. Therefore, this letter provides the biological results that T-type calcium channel is important in regulating the important cellular phenotype transition leading to cell proliferation, and thus novel T-type Ca(2+) channel blocker presents new prospects for cancer treatment.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Bloqueadores dos Canais de Cálcio/síntese química , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/química , Ensaios de Seleção de Medicamentos Antitumorais , Quinazolinas/farmacologia , Animais , Canais de Cálcio Tipo T/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Química Farmacêutica/métodos , Doxorrubicina/farmacologia , Desenho de Fármacos , Humanos , Concentração Inibidora 50 , Camundongos , Modelos Químicos , Quinazolinas/síntese química
16.
Bioorg Med Chem Lett ; 17(21): 5740-3, 2007 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-17869104

RESUMO

The intensive SAR study of 3,4-dihydroquinazoline series led to the most potent compound 10 (KYS05090: IC(50)=41+/-1 nM) against T-type calcium channel and its potency is nearly comparable to that of Kurtoxin. As a small organic molecule, this compound showed the highest blocking activity reported to date.


Assuntos
Bloqueadores dos Canais de Cálcio/química , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo T/efeitos dos fármacos , Quinazolinas/química , Quinazolinas/farmacologia , Linhagem Celular , Humanos , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA