Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27.854
Filtrar
1.
Med Oncol ; 40(7): 196, 2023 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-37284891

RESUMO

Anaplastic thyroid cancer (ATC) represents the type with the worst prognosis among thyroid cancers. In ATC with a highly invasive phenotype, selective targeting of TERT with BIBR1532 may be a goal-driven approach to preserving healthy tissues. In present study, it was aimed to investigate the effects of treatment of SW1736 cells with BIBR1532 on apoptosis, cell cycle progression, and migration. The apoptotic effect of BIBR1532 on SW1736 cells was examined using the Annexin V method, the cytostatic effect using cell cycle test, migration properties using wound healing assay. Gene expression differences were determined by real-time qRT-PCR and differences in protein level by ELISA test. BIBR1532-treated SW1736 cells had 3.1-fold increase in apoptosis compared to their untreated counterpart. There was 58.1% arrest in the G0/G1 phase and 27.6% arrest in the S phase of the cell cycle in untreated group, treatment with BIBR1532 increased cell population in G0/G1 phase to 80.9% and decreased in S phase to 7.1%. Treatment with the TERT inhibitor resulted in a 50.8% decrease in cell migration compared to the untreated group. After BIBR1532 treatment of SW1736 cells, upregulation of BAD, BAX, CASP8, CYCS, TNFSF10, CDKN2A genes, and downregulation of BCL2L11, XIAP, CCND2 genes were detected. BIBR1532 treatment resulted in an increase in BAX and p16 proteins, and a decrease in concentration of BCL-2 protein compared to untreated group. Targeting TERT with BIBR1532 as a mono drug or using of BIBR1532 at "priming stage" prior to chemotherapy treatment in ATC may present a novel and promising treatment strategy.


Assuntos
Antineoplásicos , Apoptose , Ciclo Celular , Movimento Celular , Inibidores Enzimáticos , Telomerase , Carcinoma Anaplásico da Tireoide , Neoplasias da Glândula Tireoide , Humanos , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Telomerase/antagonistas & inibidores , Carcinoma Anaplásico da Tireoide/tratamento farmacológico , Carcinoma Anaplásico da Tireoide/genética , Carcinoma Anaplásico da Tireoide/patologia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/genética , Neoplasias da Glândula Tireoide/patologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/genética , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos
2.
J Virol ; 97(6): e0037023, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37219458

RESUMO

DNA replication of E1-deleted first-generation adenoviruses (AdV) in cultured cancer cells has been reported repeatedly and it was suggested that certain cellular proteins could functionally compensate for E1A, leading to the expression of the early region 2 (E2)-encoded proteins and subsequently virus replication. Referring to this, the observation was named E1A-like activity. In this study, we investigated different cell cycle inhibitors with respect to their ability to increase viral DNA replication of dl70-3, an E1-deleted adenovirus. Our analyses of this issue revealed that in particular inhibition of cyclin-dependent kinases 4/6 (CDK4/6i) increased E1-independent adenovirus E2-expression and viral DNA replication. Detailed analysis of the E2-expression in dl70-3 infected cells by RT-qPCR showed that the increase in E2-expression originated from the E2-early promoter. Mutations of the two E2F-binding sites in the E2-early promoter (pE2early-LucM) caused a significant reduction in E2-early promoter activity in trans-activation assays. Accordingly, mutations of the E2F-binding sites in the E2-early promoter in a virus named dl70-3/E2Fm completely abolished CDK4/6i induced viral DNA replication. Thus, our data show that E2F-binding sites in the E2-early promoter are crucial for E1A independent adenoviral DNA replication of E1-deleted vectors in cancer cells. IMPORTANCE E1-deleted AdV vectors are considered replication deficient and are important tools for the study of virus biology, gene therapy, and large-scale vaccine development. However, deletion of the E1 genes does not completely abolish viral DNA replication in cancer cells. Here, we report, that the two E2F-binding sites in the adenoviral E2-early promoter contribute substantially to the so-called E1A-like activity in tumor cells. With this finding, on the one hand, the safety profile of viral vaccine vectors can be increased and, on the other hand, the oncolytic property for cancer therapy might be improved through targeted manipulation of the host cell.


Assuntos
Adenoviridae , Ciclo Celular , Replicação do DNA , Replicação Viral , Adenoviridae/genética , Adenoviridae/metabolismo , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Sítios de Ligação , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células/efeitos dos fármacos , Células/virologia , Replicação do DNA/efeitos dos fármacos , DNA Viral/metabolismo , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Mutação , Regiões Promotoras Genéticas/genética , Inibidores de Proteínas Quinases/farmacologia , Replicação Viral/fisiologia , Humanos
3.
J Enzyme Inhib Med Chem ; 38(1): 2198163, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37036011

RESUMO

Novel series of aminopyrimidines bearing a biologically active cyclohexenone 3a-f and oxo-selaneylidene moiety 4, besides selenadiazolopyrimidines (5a-e and 7), were synthesised using 5,6-diaminouracils as starting materials. Compound 3a exhibited strong anti-proliferative activity against three cell lines: HepG-2 (IC50 14.31 ± 0.83 µM), A-549 (IC50 30.74 ± 0.76 µM), and MCF-7 (IC50 27.14 ± 1.91 µM). Also, it was four times more selectively cytotoxic against WI-38 cell lines than doxorubicin. Furthermore, Topoisomerase II (IC50 4.48 ± 0.65 µM) and HSP90 (IC50 1.78 ± 0.11 µM) were both strongly inhibited in vitro by 3a. The cell cycle was halted at the G1-S phase, and total apoptotic cells were 65 times more than control Hep-G2 cells. Besides, it increased caspase-3 gene expression, triggering mitochondrial cell death. Molecular docking study indicated that it could bind to Topoisomerase II and HSP90 binding sites in an inhibitory mode. Its geometric properties were investigated using the density functional theory (DFT). Furthermore, compound 3a demonstrated in silico good oral bioavailability.


Assuntos
Antineoplásicos , Proteínas de Choque Térmico HSP90 , Pirimidinas , Inibidores da Topoisomerase II , Pirimidinas/síntese química , Pirimidinas/farmacologia , Inibidores da Topoisomerase II/síntese química , Inibidores da Topoisomerase II/farmacologia , Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Células Hep G2 , Células A549 , Células MCF-7 , Humanos , Proliferação de Células/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Simulação por Computador , Antineoplásicos/síntese química , Antineoplásicos/farmacologia
4.
Mol Psychiatry ; 28(4): 1571-1584, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36385168

RESUMO

Prenatal alcohol exposure is the foremost preventable etiology of intellectual disability and leads to a collection of diagnoses known as Fetal Alcohol Spectrum Disorders (FASD). Alcohol (EtOH) impacts diverse neural cell types and activity, but the precise functional pathophysiological effects on the human fetal cerebral cortex are unclear. Here, we used human cortical organoids to study the effects of EtOH on neurogenesis and validated our findings in primary human fetal neurons. EtOH exposure produced temporally dependent cellular effects on proliferation, cell cycle, and apoptosis. In addition, we identified EtOH-induced alterations in post-translational histone modifications and chromatin accessibility, leading to impairment of cAMP and calcium signaling, glutamatergic synaptic development, and astrocytic function. Proteomic spatial profiling of cortical organoids showed region-specific, EtOH-induced alterations linked to changes in cytoskeleton, gliogenesis, and impaired synaptogenesis. Finally, multi-electrode array electrophysiology recordings confirmed the deleterious impact of EtOH on neural network formation and activity in cortical organoids, which was validated in primary human fetal tissues. Our findings demonstrate progress in defining the human molecular and cellular phenotypic signatures of prenatal alcohol exposure on functional neurodevelopment, increasing our knowledge for potential therapeutic interventions targeting FASD symptoms.


Assuntos
Córtex Cerebral , Etanol , Vias Neurais , Neurogênese , Neurônios , Organoides , Feminino , Humanos , Masculino , Gravidez , Astrócitos/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/citologia , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Montagem e Desmontagem da Cromatina/genética , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/genética , Etanol/farmacologia , Transtornos do Espectro Alcoólico Fetal/etiologia , Transtornos do Espectro Alcoólico Fetal/genética , Feto/citologia , Perfilação da Expressão Gênica , Rede Nervosa/efeitos dos fármacos , Transtornos do Neurodesenvolvimento/induzido quimicamente , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Neurogênese/efeitos dos fármacos , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Organoides/citologia , Organoides/efeitos dos fármacos , Organoides/patologia , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Efeitos Tardios da Exposição Pré-Natal/genética , Proteômica , Sinapses/efeitos dos fármacos , Vias Neurais/efeitos dos fármacos
5.
ACS Chem Biol ; 17(12): 3290-3297, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36469692

RESUMO

Autophagy is a conserved metabolic pathway that is central to many diseases. Recently, there has been a lot of interest in targeting autophagy with small molecule inhibitors as a possible therapeutic strategy. However, many of the compounds used for autophagy are nonselective. Here, we explored the inhibition of autophagy in pancreatic cancer cells using established selective small molecule inhibitors and discovered an unexpected link between the autophagy pathway and progression through the cell cycle. Our findings revealed that treatments with inhibitors that have different autophagy pathway targets block cell replication and activate other metabolic pathways to compensate for the blockade in autophagy. An unbiased screen looking for known drugs that might synergize with autophagy inhibition revealed new combination treatments that might provide a blueprint for therapeutic approaches to pancreatic cancer. The drugs quizartinib and THZ1 showed a strong synergistic effect in pancreatic cells with autophagy inhibition.


Assuntos
Autofagia , Ciclo Celular , Neoplasias Pancreáticas , Humanos , Autofagia/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Combinação de Medicamentos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas
6.
Nature ; 608(7924): 733-740, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35978187

RESUMO

Single-cell transcriptomics (scRNA-seq) has greatly advanced our ability to characterize cellular heterogeneity1. However, scRNA-seq requires lysing cells, which impedes further molecular or functional analyses on the same cells. Here, we established Live-seq, a single-cell transcriptome profiling approach that preserves cell viability during RNA extraction using fluidic force microscopy2,3, thus allowing to couple a cell's ground-state transcriptome to its downstream molecular or phenotypic behaviour. To benchmark Live-seq, we used cell growth, functional responses and whole-cell transcriptome read-outs to demonstrate that Live-seq can accurately stratify diverse cell types and states without inducing major cellular perturbations. As a proof of concept, we show that Live-seq can be used to directly map a cell's trajectory by sequentially profiling the transcriptomes of individual macrophages before and after lipopolysaccharide (LPS) stimulation, and of adipose stromal cells pre- and post-differentiation. In addition, we demonstrate that Live-seq can function as a transcriptomic recorder by preregistering the transcriptomes of individual macrophages that were subsequently monitored by time-lapse imaging after LPS exposure. This enabled the unsupervised, genome-wide ranking of genes on the basis of their ability to affect macrophage LPS response heterogeneity, revealing basal Nfkbia expression level and cell cycle state as important phenotypic determinants, which we experimentally validated. Thus, Live-seq can address a broad range of biological questions by transforming scRNA-seq from an end-point to a temporal analysis approach.


Assuntos
Sobrevivência Celular , Perfilação da Expressão Gênica , Macrófagos , RNA-Seq , Análise de Célula Única , Transcriptoma , Tecido Adiposo/citologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Diferenciação Celular , Perfilação da Expressão Gênica/métodos , Perfilação da Expressão Gênica/normas , Genoma/efeitos dos fármacos , Genoma/genética , Lipopolissacarídeos/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/metabolismo , Inibidor de NF-kappaB alfa/genética , Especificidade de Órgãos , Fenótipo , RNA/genética , RNA/isolamento & purificação , RNA-Seq/métodos , RNA-Seq/normas , Reprodutibilidade dos Testes , Análise de Sequência de RNA/métodos , Análise de Sequência de RNA/normas , Análise de Célula Única/métodos , Células Estromais/citologia , Células Estromais/metabolismo , Fatores de Tempo , Transcriptoma/genética
7.
Med Oncol ; 39(9): 124, 2022 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-35716217

RESUMO

Both pro-oncogenic and anti-oncogenic effects of E2F2 have been revealed in different malignancies. However, the precise role of E2F2 in pancreatic cancer, in particular in relation to therapeutic intervention with gemcitabine, remains unclear. In this study, the effect of E2F2 on the proliferation and cell cycle modulation of pancreatic cancer cells, and whether E2F2 plays a role in the treatment of pancreatic cancer cells by gemcitabine, were investigated. The expression of E2F2 in pancreatic cancer was assessed by various methods including bioinformatics prediction, Western blotting, and real-time PCR. The effect of E2F2 on the proliferation and cell cycling of pancreatic cancer cells was analyzed by tissue culture and flow cytometry. In addition, the effect of E2F2 on the intervention of pancreatic cancer by gemcitabine was investigated using both in vitro and in vivo approaches. The expression of E2F2 was found to be significantly increased in pancreatic cancer tissues and cell lines. The pathogenic capacity of E2F2 lied in the fact that this transcription factor promoted the transformation of pancreatic cancer cell cycle from G1-phase to S-phase, thus enhancing the proliferation of pancreatic cancer cells. Furthermore, the expression of E2F2 was increased in pancreatic cancer cells in the presence of gemcitabine, and the augmented expression of E2F2 upregulated the gemcitabine resistance-related gene RRM2 and its downstream signaling molecule deoxycytidine kinase (DCK). The resistance of pancreatic cancer cells to gemcitabine was confirmed using both in vitro and in vivo models. In this study, E2F2 has been demonstrated for the first time to play a pro-oncogenic role in pancreatic cancer by promoting the transition of the cell cycle from G1-phase to S-phase and, therefore, enhancing the proliferation of pancreatic cancer cells. E2F2 has also been demonstrated to enhance the chemotherapy resistance of pancreatic cancer cells to gemcitabine by upregulating the expression of RRM2 and DCK that is downstream of RRM2.


Assuntos
Desoxicitidina , Resistencia a Medicamentos Antineoplásicos , Fator de Transcrição E2F2 , Neoplasias Pancreáticas , Antimetabólitos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Fator de Transcrição E2F2/genética , Fator de Transcrição E2F2/metabolismo , Humanos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Ribonucleosídeo Difosfato Redutase/biossíntese , Ribonucleosídeo Difosfato Redutase/genética , Ribonucleosídeo Difosfato Redutase/metabolismo , Regulação para Cima/efeitos dos fármacos , Gencitabina
8.
Ars pharm ; 63(2)abr.-jun. 2022. ilus, tab, graf
Artigo em Inglês | IBECS | ID: ibc-202812

RESUMO

Introduction: Urothelial carcinoma is a significant public health problem. Transitional cell carcinoma (TCC) is the most common subtype, accounting for approximately 90 % of all bladder cancers. Chemotherapeutic protocols have been studied, but some present high toxicity and low tolerability. Naringin is a polyphenolic compound found mainly in citrus fruits, which antitumor activity has been studied in several types of cancer. However, there is little information about naringin effects on bladder cancer. This study aimed to evaluate the antitumor potential of naringin in silico and in vitro using two bladder cancer cell lines. Method: In silico analysis was carried out by PASS Online software. In vitro , the effects of naringin treatment (12.5 - 400 µM) were evaluated regarding its cytotoxicity, clonogenic survival, morphological alterations, cell cycle progression, migration, and mutagenicity Results: In silico analyses predicted antitumor activity through several mechanisms of action. In vitro results showed naringin presented cytotoxic effects, reduced the number of colonies, inhibited cell migration, and changed the morphology and cell cycle progression of the two cell lines evaluated. However, naringin did not present mutagenic effects. Conclusions: Naringin has antiproliferative activity and is a promising candidate for bladder cancer treatment.(AU)


Introducción: El carcinoma urotelial es un problema de salud pública importante. El carcinoma de células de transición es el subtipo más común y representa aproximadamente el 90 % de todos los cánceres de vejiga. Se han estudiado protocolos quimioterapéuticos, pero algunos presentan alta toxicidad y baja tolerabilidad. La naringina es un compuesto polifenólico que se encuentra principalmente en los cítricos, cuya actividad antitumoral se ha estudiado en varios tipos de cáncer. Sin embargo, hay poca información sobre los efectos de la naringina en el cáncer de vejiga. Este estudio tuvo como objetivo evaluar el potencial antitumoral de la naringina in silico e in vitro utilizando dos líneas celulares de cáncer de vejiga. Método: El análisis in silico se llevó a cabo mediante el software PASS Online. In vitro, se evaluaron los efectos del tratamiento con naringina (12,5 - 400 µM) en cuanto a su citotoxicidad, supervivencia clonogénica, alteraciones morfológicas, progresión del ciclo celular, migración y mutagenicidad. Resultados: los análisis in silico predijeron la actividad antitumoral a través de varios mecanismos de acción. Los resultados in vitro mostraron que la naringina presentó efectos citotóxicos, redujo el número de colonias, inhibió la migración celular y cambió la morfología y la progresión del ciclo celular de las dos líneas celulares evaluadas. Sin embargo, la naringina no presentó efectos mutagénicos. Conclusiones: la naringina tiene actividad antiproliferativa y es un candidato prometedor para el tratamiento del cáncer de vejiga.(AU)


Assuntos
Humanos , Compostos de Silício/farmacologia , Antineoplásicos/farmacologia , Neoplasias da Bexiga Urinária , Flavanonas/farmacologia , Células Tumorais Cultivadas/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos
9.
Cancer Res ; 82(7): 1170-1171, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35373292

RESUMO

Cyclin-dependent kinases (CDK), such as CDK4 and CDK6, phosphorylate RB1 to release the transcription factor E2F and drive the transition from G1 to S-phase of the cell cycle. Inhibitors of these kinases thereby block cell-cycle progression and presumably exert their therapeutic effect. While this mechanism is straight forward, several aspects have seemed problematic, not the least of which is that these drugs seem to have therapeutic effects on a relatively small number of human cancers. Tong and colleagues took an open-ended approach to this mechanistic question, and their results raise the possibility that inhibition of phosphorylation of the transcription factor p73 is a key mechanism of action of these drugs. They show that p73 inhibition and the resultant upregulation of the cell surface receptor DR5 are necessary for the anticancer effects of CDK4/6 inhibitors, including enhancement of immune-mediated cell killing, and that therapeutic benefit relies largely on their use in conjunction with other agents. While many questions remain to be answered, these findings demonstrate the importance of keeping an open mind to mechanistic aspects of therapeutic agents already in clinical use and highlight how rigorous mechanistic studies can answer both basic and translational questions. See related article by Tong et al., p. 1340.


Assuntos
Quinase 4 Dependente de Ciclina , Ciclo Celular/efeitos dos fármacos , Divisão Celular , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Fatores de Transcrição E2F , Humanos , Fosforilação
10.
In Vitro Cell Dev Biol Anim ; 58(3): 220-231, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35386089

RESUMO

4-Vinylcyclohexene diepoxide (VCD) is a potentially hazardous industrial chemical that may enter a goat's body in various ways during industrial breeding. Ovarian granulosa cells (GCs) play a critical role in supporting follicle development and hormone synthesis. However, there are few studies on the effect of VCD on goat ovarian GCs. In this study, goat ovarian GCs were isolated and treated with VCD. The results showed that treatment with VCD increased the proportion of S phase and G2/M cells, but decreased the proportion of G1 phase. VCD treatment significantly inhibited the expression of cyclin A and cyclin-dependent kinase 2 (CDK2). But the expression levels of p21 and p27 were increased. VCD could induce an apparent increase in the proportion of apoptosis and the level of cleaved caspase 3. Treatment with VCD significantly reduced the progesterone and estrogen concentration in the medium in which goat ovarian GCs were cultured. Correspondingly, the expression level of steroidogenic acute regulatory protein (STAR) was significantly downregulated. Treatment with 0.25 and 0.5 mM VCD, the protein expression level of insulin-like growth factor 1 receptor (IGF1R) and Akt were significantly decreased. Moreover, treatment with 0.25 mM VCD significantly inhibited the phosphorylation of Akt. In conclusion, VCD exposure had cytotoxic effects such as decreased cell viability, disordered cell cycle, increased apoptosis, and interference with steroid hormone synthesis on goat GCs. These cytotoxic effects of VCD on goat GCs may be due to the downregulation of IGF1R and the inhibition of IGF1R/Akt signaling pathway.


Assuntos
Apoptose , Ciclo Celular , Cicloexenos , Cabras , Proteínas Proto-Oncogênicas c-akt , Esteroides , Compostos de Vinila , Animais , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Cicloexenos/farmacologia , Feminino , Cabras/metabolismo , Células da Granulosa/metabolismo , Hormônios , Proteínas Proto-Oncogênicas c-akt/metabolismo , Esteroides/metabolismo , Compostos de Vinila/farmacologia
11.
Mar Drugs ; 20(3)2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35323484

RESUMO

Marine alkaloid fascaplysin and its derivatives are known to exhibit promising anticancer properties in vitro and in vivo. However, toxicity of these molecules to non-cancer cells was identified as a main limitation for their clinical use. Here, for the very first time, we synthesized a library of fascaplysin derivatives covering all possible substituent introduction sites, i.e., cycles A, C and E of the 12H-pyrido[1-2-a:3,4-b']diindole system. Their selectivity towards human prostate cancer versus non-cancer cells, as well as the effects on cellular metabolism, membrane integrity, cell cycle progression, apoptosis induction and their ability to intercalate into DNA were investigated. A pronounced selectivity for cancer cells was observed for the family of di- and trisubstituted halogen derivatives (modification of cycles A and E), while a modification of cycle C resulted in a stronger activity in therapy-resistant PC-3 cells. Among others, 3,10-dibromofascaplysin exhibited the highest selectivity, presumably due to the cytostatic effects executed via the targeting of cellular metabolism. Moreover, an introduction of radical substituents at C-9, C-10 or C-10 plus C-3 resulted in a notable reduction in DNA intercalating activity and improved selectivity. Taken together, our research contributes to understanding the structure-activity relationships of fascaplysin alkaloids and defines further directions of the structural optimization.


Assuntos
Antineoplásicos , Indóis , Antineoplásicos/química , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , DNA/metabolismo , Humanos , Indóis/química , Indóis/farmacologia , Substâncias Intercalantes/química , Substâncias Intercalantes/farmacologia , Relação Estrutura-Atividade
12.
J Enzyme Inhib Med Chem ; 37(1): 832-843, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35260020

RESUMO

This paper described our efforts to develop dianilinopyrimidines as novel EGFR inhibitors. All the target compounds were tested for inhibitory effects against wild type EGFR (EGFRwt) and three tumour cells, including A549, PC-3, and HepG2. Some of the compounds performed well in antitumor activities. Especially, compound 4c 2-((2-((4-(3-fluorobenzamido)phenyl)amino)-5-(trifluoromethyl) pyrimidin-4-yl)amino)-N-methylthiophene-3-carboxamide showed higher anti-tumour activities than Gefitinib. The IC50 values of compound 4c against A549, PC-3, and HepG2. reached 0.56 µM, 2.46 µM, and 2.21 µM, respectively. In addition, further studies indicated that compound 4c could induce apoptosis against A549 cells and arrest A549 cells in the G2/M phase. Molecular docking studies showed that compound 4c could closely interact with EGFR. Generally, compound 4c was the potential for developing into an anti-tumour drug.


Assuntos
Compostos de Anilina/farmacologia , Antineoplásicos/farmacologia , Desenho de Fármacos , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Compostos de Anilina/síntese química , Compostos de Anilina/química , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Humanos , Simulação de Acoplamento Molecular , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Pirimidinas/síntese química , Pirimidinas/química , Relação Estrutura-Atividade
13.
Chem Biol Interact ; 358: 109881, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35307378

RESUMO

Stomach cancer causes the third-highest cancer-related deaths worldwide. Limited availability of anticancer measures with higher efficiency and low unwanted toxicities necessitates the development of better cancer chemotherapeutics. Naphthalene diimide (NDI) derivatives have gained significant attention owing to their excellent anticancer potential. We evaluated the anticancer properties of NDI derivatives, 1a and 2a in cancer cell lines and found that 1a showed higher efficacy as compared to 2a exhibiting a remarkable difference in activity upon single atom substitution of C with N. Particularly, NDI 1a showed potent inhibitory activity against gastric cancer cell line AGS with IC50 of 2.0 µM. NDI 1a induced remarkable morphological changes and reduced clonogenicity as well as the migratory ability of AGS cells. The reduction in AGS cell migration was mediated through inhibition of Tyr397 p-FAK dephosphorylation at focal adhesion points leading to enhanced attachment of cells at contact points. NDI 1a caused extensive DNA double-strand-breaks (DSBs) leading to activation of p53 and its transcriptional target p21. Reduced nuclear BRCA1 but enhanced nuclear p53BP1 foci formation upon 1a treatment suggests that DNA DSB repair is mediated through error-prone NHEJ which led to the accumulation of extensive DNA damage. Combinatorial effects mediated by interactions of 1a with double-stranded DNA through minor groove binding as well as induction of intracellular ROS exacerbated the loss of genomic integrity induced by 1a. NDI 1a mediated DNA damage-induced S phase arrest; however, cells experiencing extensive and irreparable DNA damage underwent mitochondrial apoptosis through downregulation of anti-apoptotic protein p21. Furthermore, proliferation inhibitory activity of 1a is also attributed to inhibition of ß-catenin/c-Myc axis in AGS cells with constitutively active ß-catenin pathway. In vivo toxicity analysis of 1a revealed minimal systemic toxicity suggesting that compound 1a is a safe and potential candidate for the development of gastric cancer chemotherapeutics.


Assuntos
Apoptose , Ciclo Celular , Dano ao DNA , Imidas , Naftalenos , Neoplasias Gástricas , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Imidas/farmacologia , Naftalenos/farmacologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , beta Catenina
14.
BMC Cancer ; 22(1): 238, 2022 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-35241028

RESUMO

BACKGROUND: Circular RNAs (circRNAs) are well-known regulators of cancer progression and chemoresistance in various types of cancers. This study was performed to investigate the function of hsa_circ_0000277 in esophageal squamous cell carcinoma (ESCC). METHODS: RNA levels were analyzed via the reverse transcription-quantitative polymerase chain reaction (RT-qPCR). Cell Counting Kit-8 (CCK-8) assay was applied to determine cell proliferation and half maximal inhibitory concentration (IC50) of cisplatin (DDP). Colony formation ability was evaluated by colony formation assay. Cell cycle and apoptosis were measured using flow cytometry. RNA immunoprecipitation (RIP), pull-down assay and dual-luciferase reporter assays were performed for target interaction analysis. The protein levels were determined through western blot. Xenograft models were established for researching hsa_circ_0000277 function in vivo. RESULTS: Hsa_circ_0000277 expression was increased in ESCC cells and tissues, and it had important clinical significance. Downregulation of hsa_circ_0000277 repressed ESCC cell proliferation, colony formation, cell cycle, and DDP resistance. Hsa_circ_0000277 acted as a microRNA-873-5p (miR-873-5p) sponge and Sry-related high-mobility group box 4 (SOX4) was validated as a target of miR-873-5p. Moreover, hsa_circ_0000277/miR-873-5p axis and miR-873-5p/SOX4 axis regulated ESCC cell progression and DDP resistance. Hsa_circ_0000277/miR-873-5p axis activated SOX4/Wnt/ß-catenin signaling pathway. Hsa_circ_0000277 facilitated tumorigenesis and DDP resistance by miR-873-5p/SOX4 axis in vivo. CONCLUSION: These findings unraveled that hsa_circ_0000277 promoted ESCC progression and DDP resistance via miR-873-5p/SOX4/Wnt/ß-catenin axis, showing a specific molecular mechanism of carcinogenesis and chemoresistance in ESCC.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas do Esôfago/genética , RNA Circular/genética , Apoptose/efeitos dos fármacos , Apoptose/genética , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Regulação para Baixo/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Esofágicas/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Humanos , MicroRNAs/efeitos dos fármacos , Fatores de Transcrição SOXC/efeitos dos fármacos , Proteínas Wnt/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/efeitos dos fármacos
15.
Life Sci ; 296: 120442, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-35245520

RESUMO

AIMS: This study measured the effects of 5-Fluorouracil (5-FU), calcitriol (VD3), and/or thymoquinone (TQ) single/dual/triple therapies on cell cycle progression, apoptosis, inhibition of the PI3K/AKT/mTOR pathway, and oxidative stress against colorectal cancer (CRC). MAIN METHODS: The HT29, SW480 and SW620 cell lines were treated with 5-FU (50 µM), VD3 (25 µM), and TQ (75 µM), alone or combined for 12 h, prior to cell cycle/apoptosis analyses. KEY FINDINGS: TQ monotherapy had greater anticancer effects to active VD3 or 5-FU, revealing higher expression of p21/p27/PTEN/BAX/Cyto-C/Casp-3 and increased levels of total glutathione, with inhibitions in CCND1/CCND3/BCL-2 and PI3K/AKT/mTOR molecules, alongside higher rates of apoptosis in HT29, SW480 and SW620 cells (P < 0.005 for all markers). Additionally, all combination protocols revealed enhanced modulations of the PI3K/PTEN/Akt/mTOR pathway, higher expression of p21/p27/PTEN/BAX/Cyto-C/Casp-3, and better anti-oxidant effects, than the monotherapies. Although TQ/5-FU and TQ/VD3 co-therapies were better relative to the VD3/5-FU regimen, the best tumoricidal effects were observed with triple therapy in the HT29 and SW480 cell lines, possibly by boosted attenuations of the PI3K/AKT/mTOR oncogenic pathway. In contrast, TQ single treatment was more effective than the triple therapy regimen in metastatic SW620 cells, suggesting that this protocol would be more useful therapeutically in late-stage CRC. SIGNIFICANCE: In conclusion, this study is the first to demonstrated enhanced anti-tumorigenic effects for VD3, TQ, and 5-FU triple therapy against CRC cells and could represent the best strategy for treating early stages of malignancy, whereas TQ monotherapy could be a better approach for treating metastatic forms of the disease.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzoquinonas/farmacologia , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Benzoquinonas/administração & dosagem , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Colecalciferol/administração & dosagem , Neoplasias do Colo/patologia , Fluoruracila/administração & dosagem , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutationa/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
16.
Mol Med Rep ; 25(4)2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35137920

RESUMO

Pentraxin 3 (PTX3), a member of the c­reactive protein family, is a long pentraxin protein and a pro­inflammatory marker. However, the role of PTX3 in preeclampsia (PE) remains to be elucidated. Thus, the present study aimed to investigate the biological role and mechanisms underlying PTX3 in PE. In the present study, PTX3 was overexpressed in trophoblasts and the subsequent changes in cell proliferation, cycle distribution and invasion were observed using Cell Counting Kit­8, flow cytometry and Transwell assays, respectively. Moreover, the expression levels of MMP2 and MMP9, proteins associated with the development of PE, were detected using reverse transcription­quantitative PCR and western blot analysis. Following treatment with interleukin (IL)­1ß, the expression levels of PTX3 were measured. Furthermore, subsequent changes in cell proliferation, cycle distribution and invasion were investigated following overexpression of PTX3 and treatment with IL­1 receptor antagonist (IL­1Ra). Overexpression of PTX3 inhibited the proliferation, cycle and invasion of HTR­8/SV neo and JEG3 cells. Moreover, treatment with IL­1ß increased the expression of PTX3 in HTR­8/SV neo and JEG3 cells, which was suppressed following treatment with the IL­1ß antagonist. Following PTX3 overexpression and treatment with IL­1Ra, the inhibitory effects of PTX3 overexpression alone on the invasion of HTR­8/SV neo and JEG3 cells were attenuated. In conclusion, these results indicated that IL­1ß could induce PTX3 upregulation, which led to the inhibition of the proliferation, invasion and cell cycle of trophoblasts, thereby promoting the progression of PE.


Assuntos
Proteína C-Reativa/metabolismo , Ciclo Celular/genética , Movimento Celular/genética , Proliferação de Células/genética , Interleucina-1beta/farmacologia , Pré-Eclâmpsia/metabolismo , Componente Amiloide P Sérico/metabolismo , Trofoblastos/metabolismo , Proteína C-Reativa/genética , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/antagonistas & inibidores , Pré-Eclâmpsia/genética , Gravidez , Componente Amiloide P Sérico/genética , Trofoblastos/efeitos dos fármacos
17.
Sci Rep ; 12(1): 1848, 2022 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-35115608

RESUMO

WDR5 nucleates the assembly of histone-modifying complexes and acts outside this context in a range of chromatin-centric processes. WDR5 is also a prominent target for pharmacological inhibition in cancer. Small-molecule degraders of WDR5 have been described, but most drug discovery efforts center on blocking the WIN site of WDR5, an arginine binding cavity that engages MLL/SET enzymes that deposit histone H3 lysine 4 methylation (H3K4me). Therapeutic application of WIN site inhibitors is complicated by the disparate functions of WDR5, but is generally guided by two assumptions-that WIN site inhibitors disable all functions of WDR5, and that changes in H3K4me drive the transcriptional response of cancer cells to WIN site blockade. Here, we test these assumptions by comparing the impact of WIN site inhibition versus WDR5 degradation on H3K4me and transcriptional processes. We show that WIN site inhibition disables only a specific subset of WDR5 activity, and that H3K4me changes induced by WDR5 depletion do not explain accompanying transcriptional responses. These data recast WIN site inhibitors as selective loss-of-function agents, contradict H3K4me as a relevant mechanism of action for WDR5 inhibitors, and indicate distinct clinical applications of WIN site inhibitors and WDR5 degraders.


Assuntos
Antineoplásicos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Linfoma de Células B/tratamento farmacológico , Sítios de Ligação , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Montagem e Desmontagem da Cromatina , Regulação Neoplásica da Expressão Gênica , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Metilação , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Proteólise , Transdução de Sinais , Transcrição Gênica
18.
J Enzyme Inhib Med Chem ; 37(1): 686-700, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-35139719

RESUMO

Inhibiting Cyclin-dependent kinase 2 (CDK2) has been established as a therapeutic strategy for the treatment of many cancers. Accordingly, this study aimed at developing a new set of quinazolinone-based derivatives as CDK2 inhibitors. The new compounds were evaluated for their anticancer activity against sixty tumour cell lines. Compounds 5c and 8a showed excellent growth inhibition against the melanoma cell line MDA-MB-435 with GI% of 94.53 and 94.15, respectively. Cell cycle analysis showed that compound 5c led to cell cycle cessation at S phase and G2/M phase revealing that CDK2 could be the plausible biological target. Thus, the most cytotoxic candidates 5c and 8a were evaluated in vitro for their CDK2 inhibitory activity and were able to display significant inhibitory action. The molecular docking study confirmed the obtained results. ADME study predicted that 5c had appropriate drug-likeness properties. These findings highlight a rationale for further development and optimisation of novel CDK2 inhibitors.


Assuntos
Antineoplásicos/farmacologia , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Desenvolvimento de Medicamentos , Melanoma/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Quinazolinonas/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Quinase 2 Dependente de Ciclina/metabolismo , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Melanoma/metabolismo , Melanoma/patologia , Simulação de Acoplamento Molecular , Estrutura Molecular , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/química , Quinazolinonas/síntese química , Quinazolinonas/química , Relação Estrutura-Atividade
19.
Toxicol Appl Pharmacol ; 438: 115909, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-35134436

RESUMO

Emodin-8-O-ß-D-glucopyranoside (EG), a natural hydroxyanthraquinone glycoside from some traditional medicinal plants, has been demonstrated to have potential antitumor effects in our previous studies. Herein, we confirm that EG remains stable in the cell culture medium. It suppresses cell viability and proliferation and induces G1 cell cycle arrest in human colorectal cancer and neuroblastoma cells in vitro. EG inhibits tumor growth in human colorectal cancer cell HCT 116-bearing xenograft mice with low toxicity in the liver and kidney. The transcriptome analysis shows that the p53 signaling pathway is the most enriched cellular pathway and EG affects the proliferation of HCT 116 cells through modulating cell cycle related genes, such as CDKN1A and Cyclin-dependent kinases (CDKs). We demonstrate that the protein expression level of p21 was up-regulated, and CDK1/CDK2 were reduced significantly in both HCT 116 and SH-SY5Y cells after EG treatment. The switch from hypo- to hyper-phosphorylated Retinoblastoma (Rb), which is believed as a result of activated CDKs, was inhibited when cells were treated with EG. These findings indicate that EG suppresses cancer cell proliferation via p21-CDKs-Rb axis.


Assuntos
Antraquinonas/farmacologia , Proliferação de Células/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Quinases Ciclina-Dependentes/metabolismo , Emodina/farmacologia , Glicosídeos/farmacologia , Proteína do Retinoblastoma/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Células HCT116 , Células HT29 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação/efeitos dos fármacos , Retinoblastoma/tratamento farmacológico , Retinoblastoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
20.
BMC Cancer ; 22(1): 171, 2022 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-35168555

RESUMO

BACKGROUND: Serotonin (or 5-Hydroxytryptamine, 5-HT) signals in mammary gland becomes dysregulated in cancer, also contributing to proliferation, metastasis, and angiogenesis. Thus, the discovery of novel compounds targeting serotonin signaling may contribute to tailor new therapeutic strategies usable in combination with endocrine therapies. We have previously synthesized serotoninergic receptor ligands (SER) with high affinity and selectivity towards 5-HT2A and 5-HT2C receptors, the main mediators of mitogenic effect of serotonin in breast cancer (BC). Here, we investigated the effect of 10 SER on viability of MCF7, SKBR3 and MDA-MB231 BC cells and focused on their potential ability to affect Tamoxifen responsiveness in ER+ cells. METHODS: Cell viability has been assessed by sulforhodamine B assay. Cell cycle has been analyzed by flow cytometry. Gene expression of 5-HT receptors and Connective Tissue Growth Factor (CTGF) has been checked by RT-PCR; mRNA levels of CTGF and ABC transporters have been further measured by qPCR. Protein levels of 5-HT2C receptors have been analyzed by Western blot. All data were statistically analyzed using GraphPad Prism 7. RESULTS: We found that treatment with SER for 72 h reduced viability of BC cells. SER were more effective on MCF7 ER+ cells (IC50 range 10.2 µM - 99.2 µM) compared to SKBR3 (IC50 range 43.3 µM - 260 µM) and MDA-MB231 BC cells (IC50 range 91.3 µM - 306 µM). This was paralleled by accumulation of cells in G0/G1 phase of cell cycle. Next, we provided evidence that two ligands, SER79 and SER68, improved the effectiveness of Tamoxifen treatment in MCF7 cells and modulated the expression of CTGF, without affecting viability of MCF10A non-cancer breast epithelial cells. In a cell model of Tamoxifen resistance, SER68 also restored drug effect independently of CTGF. CONCLUSIONS: These results identified serotoninergic receptor ligands potentially usable in combination with Tamoxifen to improve its effectiveness on ER+ BC patients.


Assuntos
Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/tratamento farmacológico , Serotonina/metabolismo , Tamoxifeno/farmacologia , Neoplasias da Mama/metabolismo , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator de Crescimento do Tecido Conjuntivo/metabolismo , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Ligantes , Células MCF-7 , Receptores de Estrogênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...