Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Phytochemistry ; 220: 113995, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38307313

RESUMO

To find structurally previously undescribed compounds with pharmacological effects from Prismatomeris tetrandra (Roxb.) K. Schum (Rubiaceae), thirteen undescribed tetrahydroanthraquinones (1⎼13) named prisconnatanones J⎼V and seven known anthraquinones (14⎼20) were isolated and characterized. The structures of these compounds were elucidated by detailed spectroscopic analyses, and their absolute configurations were established by modified Mosher's method and ECD calculations. The antitumor cell proliferative activities of prisconnatanones J⎼V were determined. Among them, prisconnatanones J possessed high antitumor cell proliferation in HGC27 cells (IC50, 0.792 µM) by blocking HGC27 cells in the S phase and significantly inducing apoptosis in HGC27 cells. Prisconnatanone J has no cytotoxicity to normal gastric cells line (GES-1) at 10 µM and showed a considerable selectivity for HGC27 cells. Prisconnatanone J can potentially inhibit tumor cell proliferation and should be further investigated.


Assuntos
Rubiaceae , Proliferação de Células , Linhagem Celular Tumoral , Rubiaceae/química , Apoptose , Estrutura Molecular
2.
Eur J Med Chem ; 264: 116015, 2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38048697

RESUMO

Simultaneous inhibition of PI3K and HDAC has shown promise for treating various cancers, leading to discovery and development of their dual inhibitors as novel anticancer agents. Herein, we disclose a new series of PI3K/HDAC dual inhibitors bearing a benzamide moiety as the pharmacophore of HDAC inhibition. Based on systematic structure-activity relationship study, compounds 36 and 51 featuring an alkyl and benzoyl linker respectively were identified with favorable potencies against both PI3K and HDAC. In cellular assays, compounds 36 and 51 showed significantly enhanced antiproliferative activities against various cancer cell lines relative to single-target inhibitors. Furthermore, western blotting analysis shows compounds 36 and 51 suppressed AKT phosphorylation and increased H3 acetylation in MV4-11 cells, while flow cytometry analysis reveals both compounds dose-dependently induced cell cycle arrest and cell apoptosis. Supported by pharmacokinetic studies, compounds 36 and 51 were subjected to the in vivo evaluation in a MV4-11 xenograft model, demonstrating significant and dose-dependent anticancer efficacies. Overall, this work provides a promising approach for the treatment of AML by simultaneously inhibiting PI3K and HDAC with a dual inhibitor.


Assuntos
Antineoplásicos , Leucemia Mieloide Aguda , Humanos , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Inibidores de Histona Desacetilases/química , Linhagem Celular Tumoral , Fosfatidilinositol 3-Quinases/metabolismo , Proliferação de Células , Antineoplásicos/química , Relação Estrutura-Atividade , Leucemia Mieloide Aguda/tratamento farmacológico , Zinco/farmacologia , Apoptose
3.
J Med Chem ; 66(20): 13946-13967, 2023 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-37698518

RESUMO

Dihydrofolate reductase (DHFR), a core enzyme of folate metabolism, plays a crucial role in the biosynthesis of purines and thymidylate for cell proliferation and growth in both prokaryotic and eukaryotic cells. However, the development of new DHFR inhibitors is challenging due to the limited number of scaffolds available for drug development. Hence, we designed and synthesized a new class of DHFR inhibitors with a 1,3-diamino-7H-pyrrol[3,2-f]quinazoline derivative (PQD) structure bearing condensed rings. Compound 6r exhibited therapeutic effects on mouse models of systemic infection and thigh infection caused by methicillin-resistant Staphylococcus aureus (MRSA) ATCC 43300. Moreover, methyl-modified PQD compound 8a showed a strong efficacy in a murine model of breast cancer, which was better than the effects of taxol. The findings showcased in this study highlight the promising capabilities of novel DHFR inhibitors in addressing bacterial infections as well as breast cancer.


Assuntos
Antagonistas do Ácido Fólico , Staphylococcus aureus Resistente à Meticilina , Neoplasias , Camundongos , Animais , Relação Estrutura-Atividade , Modelos Animais de Doenças , Tetra-Hidrofolato Desidrogenase/metabolismo
4.
Mol Med Rep ; 28(3)2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37417374

RESUMO

The Na/K­ATPase/Src complex is reportedly able to affect reactive oxygen species (ROS) amplification. However, it has remained elusive whether NADPH oxidases (NOXs) are involved in this oxidant amplification loop in renal fibrosis. To test this hypothesis, interactions between oxidative features and Na/K­ATPase/Src activation were examined in a mouse model of unilateral urethral obstruction (UUO)­induced experimental renal fibrosis. Both 1­tert­butyl­3­(4­chlorophenyl)­1H­pyrazolo[3,4­d]pyrimidin­4­amine (PP2) and apocynin significantly attenuated the development of UUO­induced renal fibrosis. Apocynin administration attenuated the expression of NOXs and oxidative markers (e.g., nuclear factor erythroid 2­related factor 2, heme oxygenase­1,4­hydroxynonenal and 3­nitrotyrosine); it also partially restored Na/K­ATPase expression and inhibited the activation of the Src/ERK cascade. Furthermore, administration of PP2 after UUO induction partially reversed the upregulation of NOX2, NOX4 and oxidative markers, while inhibiting the activation of the Src/ERK cascade. Complementary experiments in LLC­PK1 cells corroborated the in vivo observations. Inhibition of NOX2 by RNA interference attenuated ouabain­induced oxidative stress, ERK activation and E­cadherin downregulation. Thus, it is indicated that NOXs are major contributors to ROS production in the Na/K­ATPase/Src/ROS oxidative amplification loop, which is involved in renal fibrosis. The disruption of this vicious feed­forward loop between NOXs/ROS and redox­regulated Na/K­ATPase/Src may have therapeutic applicability for renal fibrosis disorders.


Assuntos
Nefropatias , NADPH Oxidases , Camundongos , Animais , NADPH Oxidases/genética , Espécies Reativas de Oxigênio/metabolismo , Oxidantes , Nefropatias/etiologia , Estresse Oxidativo , Fibrose , Adenosina Trifosfatases/metabolismo , NADPH Oxidase 4/genética , NADPH Oxidase 4/metabolismo
6.
Eur J Med Chem ; 244: 114832, 2022 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-36270090

RESUMO

In this study, a series of novel furoxan-based nitric oxide (NO) releasing derivatives of pyranocarbazole alkaloids were designed, synthesized, and biologically evaluated against human cancer cell lines. The derivatives showed considerable antiproliferative activities (IC50 = 0.05-7.55 µM) and most compounds showed higher activity in MDA-MB-231 than H460 and HeLa. Especially, the most active derivative 7a (IC50 = 0.05 µM) against MDA-MB-231 was about 60 times stronger than lead compound, as well as equivalent to positive control taxol, and produced high levels of NO in MDA-MB-231. Furthermore, 7a could significantly inhibit the growth of MDA-MB-231 tumors in vivo with low toxicity and the PI3K/Akt signaling pathway. These results indicated that compound 7a could be a promising lead for further studies.


Assuntos
Alcaloides , Antineoplásicos , Carbazóis , Desenho de Fármacos , Doadores de Óxido Nítrico , Óxido Nítrico , Humanos , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Estrutura Molecular , Óxido Nítrico/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Relação Estrutura-Atividade , Doadores de Óxido Nítrico/química , Doadores de Óxido Nítrico/farmacologia , Carbazóis/química , Carbazóis/farmacologia , Alcaloides/química , Alcaloides/farmacologia
7.
Bioorg Med Chem Lett ; 71: 128825, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35644299

RESUMO

The synergistic anti-tumor effect by simultaneous inhibitions of PI3K and HDAC has been verified to provide the rationality of PI3K/HDAC dual inhibitors for cancer treatment. Notably, the outstanding effect of PI3K/HDAC dual inhibitors against DLBCL has been paid much attention, especially for RR-DLBCL. Our previously reported 4-methylquinazoine scaffold based PI3K/HDAC dual inhibitors could suppress the growth of solid tumors and hematologic malignancies both in vitro and in vivo, validating the potential as new therapeutic agents for cancer. In this research, we further investigated the anti-tumor activity of one of our compounds against DLBCL cell lines and in vivo zebrafish xenograft model as well as the underlying mechanism, hoping to provide a novel therapeutic agent for treating DLBCL.


Assuntos
Inibidores de Histona Desacetilases , Linfoma Difuso de Grandes Células B , Animais , Apoptose , Linhagem Celular Tumoral , Inibidores de Histona Desacetilases/farmacologia , Inibidores de Histona Desacetilases/uso terapêutico , Humanos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
8.
Acta Pharm Sin B ; 12(6): 2845-2858, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-35755282

RESUMO

PD-1 and PD-L1 antibodies have brought about extraordinary clinical benefits for cancer patients, and their indications are expanding incessantly. Currently, most PD-1/PD-L1 agents are administered intravenously, which may be uncomfortable for some cancer patients. Herein, we develop a novel oral-delivered small molecular, YPD-29B, which specifically targets human PD-L1. Our data suggested that YPD-29B could potently and selectively block the interaction between PD-L1 and PD-1, but did not inhibit any other immune checkpoints. Mechanistically, YPD-29B induced human PD-L1 dimerization and internalization, which subsequently activated T lymphocytes and therefore overcomes immunity tolerance in vitro. YDP-29B was modified as the YPD-30 prodrug to improve druggability. Using humanized mice with human PD-1 xenografts of human PD-L1 knock-in mouse MC38 cancer cells, we demonstrated that YPD-30 exhibited significant antitumor activity and was well tolerated in vivo. Taken together, our results indicate that YPD-30 serves as a promising therapeutic candidate for anti-human PD-L1 cancer immunotherapy.

9.
Int J Mol Sci ; 23(10)2022 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-35628341

RESUMO

The extracellular heat shock protein 90α (eHSP90α) has been reported to promote cancer cell motility. However, whether pancreatic cancer (PC) cells expressed membrane-bound or secreted HSP90α, as well as its underlying mechanism for PC progression, were still unclear. Our study demonstrated that the amounts of secreted HSP90α proteins were discrepant in multiple PC cells. In addition, highly invasive Capan-2 cells have a higher level of secreted HSP90α compared with those of less invasive PL45 cells. The conditioned medium of Capan-2 cells or recombinant HSP90α treatment stimulated the migration and invasion of PC cells, which could be prevented with a neutralizing anti-HSP90α antibody. Furthermore, secreted HSP90α promoted elements of epithelial-mesenchymal transition in PL45 cells, including increases in vimentin and Snail expressions, decreases in E-cadherin expression, and changes in cell shape towards a mesenchymal phenotype, but these phenomena were reversed by the anti-HSP90α antibody in Capan-2 cells. In addition, high levels of low-density lipoprotein receptor-related protein 1 (LRP1) were associated with worsened patient survival in pancreatic adenocarcinoma. We demonstrated LRP1 as a receptor of eHSP90α for its stimulatory role in metastasis, by activating the AKT pathway. In addition, silencing LRP1 enhanced the chemosensitivity to gemcitabine and doxorubicin in Capan-2 cells. Therefore, our study indicated that blocking secreted HSP90α underlies an aspect of metastasis and chemoresistance in PC.


Assuntos
Adenocarcinoma , Neoplasias Pancreáticas , Receptores de Lipoproteínas , Resistencia a Medicamentos Antineoplásicos , Proteínas de Choque Térmico HSP90/metabolismo , Humanos , Proteína-1 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas
10.
Eur J Med Chem ; 234: 114252, 2022 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-35286927

RESUMO

A series of N, C-capped di- and tripeptides were designed as selective immunoproteasome inhibitors based on the known inhibitor 4-CA. Forty-eight new compounds were synthesized and evaluated, and the structure-activity relationship (SAR) of this compound class as ß5i selective inhibitors were explored. Most of these compounds showed significant inhibition against the ß5i subunit of the immunoproteasome and the most potent ß5i inhibitor (15) showed an IC50 of 0.94 nM. A selective ß5i inhibitor (54) with over 500-fold ß5i/ß5c selectivity was identified. Three of the inhibitors were found to selectively inhibit ß5i and ß5c, and showed no noticeable inhibition against the other four subunits. Six inhibitors with significant inhibitory activity against the HCT-116 cells were recognized, and the most active inhibitors, 14 and 50, showed IC50 values of 0.46 µM and 0.16 µM, respectively. Some selective ß5i inhibitors exhibited significant inhibitory effects on the release of the cytokines TNF-α and IL-6. The results not only afford effective chemical tools to elucidate the relationships between subunit selectivity and pharmacological profiles, but also offer useful clues for further optimization and development of selective immunoproteasome inhibitors.


Assuntos
Complexo de Endopeptidases do Proteassoma , Inibidores de Proteassoma , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/química , Inibidores de Proteassoma/farmacologia , Relação Estrutura-Atividade
11.
Eur J Med Chem ; 223: 113634, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34147745

RESUMO

Selective inhibition of cyclin-dependent kinase 8 (CDK8) has been recently regarded as a potential approach for cancer therapy. A series of novel CDK8 inhibitors with the pyridine core was identified via scaffold hopping from the known CDK8 inhibitor A-7. The new inhibitors were designed to improve the ligand efficiency so as to enhance drug-likeness. Most of the compounds showed significant inhibition against CDK8/cyclin C, and the most active compounds (5d, 5e and 7') displayed IC50 values of 2.4 nM, 5.0 nM and 7.7 nM, respectively. Preliminary kinase profiling of selected compounds against a panel of kinases from different families indicated that this compound class might selectively inhibit CDK8 as well as its paralog CDK19. Some compounds exhibited cellular activity in both MTT and SRB assays against a variety of tumor cells, including HCT-116, A549, MDA-MB-231, KB, KB-VIN and MCF-7. Further flow cytometry analysis revealed a dose-dependent G2/M phase arrest in MDA-MB-231 cells treated with compounds 6'a, 6'b, 6'j and 6'k. In addition, compound 6'k demonstrated moderate antitumor efficacy in HCT-116 mouse models, although unfavorable pharmacokinetic profiles were suggested by preliminary study in mice. The results provided a new structural prototype for the search of selective CDK8 inhibitors as antitumor agents.


Assuntos
Antineoplásicos/uso terapêutico , Quinase 8 Dependente de Ciclina/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/metabolismo , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Quinase 8 Dependente de Ciclina/metabolismo , Desenho de Fármacos , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Simulação de Acoplamento Molecular , Estrutura Molecular , Ligação Proteica , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacocinética , Piridinas/síntese química , Piridinas/metabolismo , Piridinas/farmacocinética , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Prostate ; 81(10): 667-682, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33956349

RESUMO

The surface expression of Na/K-ATPase α1 (NKA) is significantly reduced in primary prostate tumors and further decreased in bone metastatic lesions. Here, we show that the loss of cell surface expression of NKA induces epithelial-mesenchymal transition (EMT) and promotes metastatic potential and tumor growth of prostate cancer (PCa) by decreasing the expression of E-cadherin and increasing c-Myc expression via the activation of Src/FAK pathways. Mechanistically, reduced surface expression of NKA in PCa is due to increased endocytosis through the activation of NKA/Src receptor complex. Using a high-throughput NKA ligand-screening platform, we have discovered MB5 as an inverse agonist of the NKA/Src receptor complex, capable of blocking the endocytosis of NKA. MB5 treatment increased NKA expression and E-cadherin in PCa cells, which reversed EMT and consequently decreased the invasion and growth of spheroid models and tumor xenografts. Thus, we have identified a hitherto unrecognized mechanism that regulates EMT and invasiveness of PCa and demonstrated for the first time the feasibility of identifying inverse agonists of receptor NKA/Src complex and their potential utility as anticancer drugs. We, therefore, conclude that cell surface expression of α1 NKA can be targeted for the development of new therapeutics against aggressive PCa and that MB5 may serve as a prototype for drug development against EMT in metastatic PCa.


Assuntos
Agonismo Inverso de Drogas , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias da Próstata/metabolismo , ATPase Trocadora de Sódio-Potássio/biossíntese , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Ouabaína/farmacologia , Tiamina/análogos & derivados , Tiamina/farmacologia , Tiamina/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
13.
J Gene Med ; 22(12): e3262, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32840014

RESUMO

BACKGROUND: Long non-coding RNA (lncRNA) small nucleolar RNA host gene 6 (SNHG6) exerts a regulatory role in cancer biology, although its detailed functions and mechanisms in colorectal cancer (CRC) still remain unclear. METHODS: A quantitative reverse transcriptase-polymerase chain reaction was implemented to investigate the expression of SNHG6, miR-181 family and Janus kinase 2 (JAK2) in CRC tissues and cell lines. The proliferation of CRC cells was detected by a cell counting kit-8 assay, and the apoptosis of CRC cells was determined by flow cytometry analysis. The interaction of the miR-181 family with SNHG6 or with the 3'-untranslated region of JAK2 was validated by the luciferase reporter gene method. The effects of SNHG6 and the miR-181 family on JAK2 expression were analyzed by western blotting. RESULTS: SNHG6 was significantly up-regulated in CRC samples. The knockdown of SNHG6 reduced the proliferation of CRC cells and promoted the apoptosis, whereas the over-expression of SNHG6 had the opposite effect. SNHG6 could bind with all the four members of the miR-181 family, and expression in miR-181 family members was significantly down-regulated in CRC samples. SNHG6 expression was negatively correlated with the miR-181 family member expression in CRC samples. Moreover, over-expressed SNHG6 significantly counteracted the inhibitory effect of miR-181 mimics on CRC cell proliferation, as well as the promoting effect on apoptosis. Furthermore, SNHG6 over-expression and knockdown can promote and inhibit JAK2 expression, respectively, and miR-181 family member function is opposite to that of SNHG6 by repressing JAK2. CONCLUSIONS: SNHG6 can exert a cancer-promoting effect in CRC by targeting miR-181 family members and up-regulating JAK2.


Assuntos
Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Janus Quinase 2/metabolismo , MicroRNAs/genética , RNA Longo não Codificante/genética , Adulto , Idoso , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Feminino , Humanos , Janus Quinase 2/genética , Masculino , Pessoa de Meia-Idade , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas
14.
Mol Cell Biochem ; 472(1-2): 1-8, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32632609

RESUMO

Hypoxic-ischemic (HI) brain damage (HIBD) leads to high neonatal mortality and severe neurologic morbidity. Autophagy is involved in the pathogenesis of HIBD. This study aims to investigate the effect of long non-coding RNA colorectal neoplasia differentially expressed (CRNDE) on HIBD and to validate whether autophagy is involved in this process. A HIBD model in rat pups and a HI model in rat primary cerebrocortical neurons were established. Autophagy was evaluated by western blot. The HIBD in rats was evaluated by hematoxylin and eosin staining, TUNEL staining, triphenyl tetrazolium chloride staining, and morris water maze test. The HI injury in vitro was evaluated by determining cell viability and apoptosis. The results showed that CRNDE expression was time-dependently increased in the brain after HIBD. Administration with CRNDE shRNA-expressing lentiviruses alleviated pathological injury and apoptosis in rat hippocampus, decreased infarct volume, and improved behavior performance of rats subjected to HIBD. Furthermore, CRNDE silencing promoted cell viability and inhibited cell apoptosis in neurons exposed to HI. Moreover, CRNDE silencing promoted autophagy and the autophagy inhibitor 3-methyladenine counteracted the neuroprotective effect of CRNDE silencing on HI-induced neuronal injury both in vivo and in vitro. Collectively, CRNDE silencing alleviates HIBD, at least partially, through promoting autophagy.


Assuntos
Autofagia , Encéfalo/metabolismo , Hipóxia-Isquemia Encefálica/prevenção & controle , Neurônios/metabolismo , Fármacos Neuroprotetores , RNA Longo não Codificante/antagonistas & inibidores , Animais , Animais Recém-Nascidos , Comportamento Animal , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/etiologia , Hipóxia-Isquemia Encefálica/patologia , Neurônios/patologia , RNA Longo não Codificante/genética , Ratos , Ratos Sprague-Dawley
15.
Mol Cell Probes ; 52: 101565, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32234564

RESUMO

BACKGROUND: This study aimed to test the hypothesis that long non-coding RNA (lncRNA) colorectal neoplasia differentially expressed (CRNDE) could exacerbate brain injury caused by intrauterine infection in neonatal rats. METHODS: Intrauterine infection was induced in pregnant rats by lipopolysaccharide (LPS). After delivery, newborn rats with brain injury caused by intrauterine infection were randomly divided into control, control shRNA, and CRNDE shRNA groups. CRNDE expression in serum and amniotic fluid of pregnant rats and neonatal brain tissues were determined by quantitative real-time PCR (qRT-PCR). Morris water maze (MWM) task was used to test the spatial learning and memory ability. Histological examination and apoptosis detection were performed by hematoxylin and eosin (H&E) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, respectively. Immunohistochemistry was conducted to evaluate the activation of astrocytes and microglia. RESULTS: LncRNA CRNDE was highly expressed in serum and amniotic fluid of maternal rats and in brain tissues of offspring rats. Furthermore, shRNA-mediated CRNDE downregulation could rescue the spatial learning and memory ability, improve brain histopathological changes and cell death, and inhibit the activation of astrocytes and microglia caused by LPS. CONCLUSION: CRNDE silencing possessed a cerebral protective effect in neonatal rats with brain injury caused by interauterine infection.


Assuntos
Lesões Encefálicas/etiologia , Lesões Encefálicas/genética , RNA Longo não Codificante/metabolismo , Útero/microbiologia , Útero/patologia , Animais , Animais Recém-Nascidos , Astrócitos/patologia , Encéfalo/patologia , Lesões Encefálicas/fisiopatologia , Morte Celular , Citocinas/biossíntese , Feminino , Técnicas de Silenciamento de Genes , Humanos , Lipopolissacarídeos , Masculino , Memória , Microglia/patologia , Gravidez , RNA Longo não Codificante/genética , Ratos , Aprendizagem Espacial , Regulação para Cima/genética
16.
Molecules ; 25(6)2020 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-32210078

RESUMO

Indoleamine 2,3-dioxygenase 1 (IDO1) is a heme-containing intracellular enzyme that catalyzes the first and rate-determining step of tryptophan metabolism and is an important immunotherapeutic target for the treatment of cancer. In this study, we designed and synthesized a new series of compounds as potential IDO1 inhibitors. These compounds were then evaluated for inhibitory activity against IDO1 and tryptophan 2,3-dioxygenase (TDO). Among them, the three phenyl urea derivatives i12, i23, i24 as showed potent IDO1 inhibition, with IC50 values of 0.1-0.6 µM and no compound exhibited TDO inhibitory activity. Using molecular docking, we predicted the binding mode of compound i12 within IDO1. Compound i12 was further investigated by determining its in vivo pharmacokinetic profile and anti-tumor efficacy. The pharmacokinetic study revealed that compound i12 had satisfactory properties in mice, with moderate plasma clearance (22.45 mL/min/kg), acceptable half-life (11.2 h) and high oral bioavailability (87.4%). Compound i12 orally administered at 15 mg/kg daily showed tumor growth inhibition (TGI) of 40.5% in a B16F10 subcutaneous xenograft model and 30 mg/kg daily showed TGI of 34.3% in a PAN02 subcutaneous xenograft model. In addition, the body weight of i12-treated mice showed no obvious reduction compared with the control group. Overall, compound i12 is a potent lead compound for developing IDO1 inhibitors and anti-tumor agents.


Assuntos
Desenho de Fármacos , Inibidores Enzimáticos , Indolamina-Pirrol 2,3,-Dioxigenase , Melanoma Experimental , Proteínas de Neoplasias , Compostos de Fenilureia , Animais , Linhagem Celular Tumoral , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indolamina-Pirrol 2,3,-Dioxigenase/química , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/enzimologia , Melanoma Experimental/patologia , Camundongos , Simulação de Acoplamento Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Compostos de Fenilureia/síntese química , Compostos de Fenilureia/química , Compostos de Fenilureia/farmacocinética , Compostos de Fenilureia/farmacologia , Relação Estrutura-Atividade
17.
Eur J Med Chem ; 190: 112079, 2020 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-32028140

RESUMO

A series of novel triptolide/furoxans hybrids were designed and synthesized as analogues of triptolide, which is a naturally derived compound isolated from the thunder god vine (Tripterygium wilfordii Hook. F). Some of these synthesized compounds exhibited antiproliferative activities in the nanomolar range. Among them, compound 33 exhibited both good antiproliferative activity and NO-releasing ability and the acute toxicity of compound 33 decreased more than 160 times (LD50 = 160.9 mg/kg) than triptolide. Moreover, compound 33 significantly inhibited the growth of melanoma at a low dose (0.3 mg/kg) and showed strong anti-inflammatory activity in vitro and in vivo. These results indicate that compound 33 could be a promising candidate for further study.


Assuntos
Anti-Inflamatórios/uso terapêutico , Antineoplásicos/uso terapêutico , Diterpenos/uso terapêutico , Doadores de Óxido Nítrico/uso terapêutico , Fenantrenos/uso terapêutico , Animais , Anti-Inflamatórios/síntese química , Anti-Inflamatórios/toxicidade , Antineoplásicos/síntese química , Antineoplásicos/toxicidade , Proliferação de Células/efeitos dos fármacos , Diterpenos/síntese química , Diterpenos/toxicidade , Desenho de Fármacos , Compostos de Epóxi/síntese química , Compostos de Epóxi/uso terapêutico , Compostos de Epóxi/toxicidade , Feminino , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Estrutura Molecular , Doadores de Óxido Nítrico/síntese química , Doadores de Óxido Nítrico/toxicidade , Fenantrenos/síntese química , Fenantrenos/toxicidade , Células RAW 264.7 , Relação Estrutura-Atividade , Fator de Necrose Tumoral alfa/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Eur J Med Chem ; 188: 111985, 2020 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-31881488

RESUMO

Indoleamine 2,3-dioxygenase 1 (IDO1) and tryptophan 2,3-dioxygenase (TDO) are involved in the key steps of tryptophan metabolism and are potential new targets for tumor immunotherapy. In this work, a variety of indole-2-carboxylic acid derivatives were synthesized, and their inhibitory activities against both enzymes along with structure-activity relationships were investigated. As a result, a number of 6-acetamido-indole-2-carboxylic acid derivatives were found to be potent dual inhibitors with IC50 values at low micromolar levels. Among them, compound 9o-1 was the most potent inhibitor with an IC50 value of 1.17 µM for IDO1, and 1.55 µM for TDO, respectively. In addition, a para-benzoquinone derivative 9p-O, resulted from the oxidation of compound 9p, was also identified and it showed strong inhibition against the two enzymes with IC50 values at the double digit nanomolar level. Using molecular docking and molecular dynamic simulations, we predicted the binding modes of this class of compounds within IDO1 and TDO binding pocket. The results provide insights for further structural optimization of this series of IDO1/TDO dual inhibitors.


Assuntos
Antineoplásicos/farmacologia , Desenho de Fármacos , Inibidores Enzimáticos/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Indóis/farmacologia , Melanoma Experimental/tratamento farmacológico , Triptofano Oxigenase/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Ácidos Carboxílicos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indóis/síntese química , Indóis/química , Melanoma Experimental/metabolismo , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Estrutura Molecular , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Triptofano Oxigenase/metabolismo
19.
Molecules ; 24(11)2019 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-31195673

RESUMO

Indoleamine 2,3-dioxygenase (IDO) 1 is the key enzyme for regulating tryptophan metabolism and is an important target for interrupting tumor immune escape. In this study, we designed four series of compounds as potential IDO1 inhibitors by attaching various fragments or ligands to indole or phenylimidazole scaffolds to improve binding to IDO1. The compounds were synthesized and their inhibitory activities against IDO1 and tryptophan 2,3-dioxygenase were evaluated. The cytotoxicities of the compounds against two tumor cell lines were also determined. Two compounds with a phenylimidazole scaffold (DX-03-12 and DX-03-13) showed potent IDO1 inhibition with IC50 values of 0.3-0.5 µM. These two IDO1 inhibitors showed low cell cytotoxicity, which indicated that they may exert their anti-tumor effect via immune modulation. Compound DX-03-12 was investigated further by determining the in vivo pharmacokinetic profile and anti-tumor efficacy. The pharmacokinetic study revealed that DX-03-12 had satisfactory properties in mice, with rapid absorption, moderate plasma clearance (∼36% of hepatic blood flow), acceptable half-life (∼4.6 h), and high oral bioavailability (∼96%). Daily oral administration of 60 mg/kg of compound DX-03-12 decreased tumor growth by 72.2% after 19 days in a mouse melanoma cell B16-F10 xenograft model compared with the untreated control. Moreover, there was no obvious weight loss in DX-03-12-treated mice. In conclusion, compound DX-03-12 is a potent lead compound for developing IDO1 inhibitors and anti-tumor agents.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Administração Oral , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacocinética , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Injeções Intravenosas , Masculino , Camundongos Endogâmicos ICR , Modelos Moleculares , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Med Chem ; 62(15): 6992-7014, 2019 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-31117517

RESUMO

Polypharmacology is a promising paradigm in modern drug discovery. Herein, we have discovered a series of novel PI3K and HDAC dual inhibitors in which the hydroxamic acid moiety as the zinc binding functional group was introduced to a quinazoline-based PI3K pharmacophore through an appropriate linker. Systematic structure-activity relationship studies resulted in lead compounds 23 and 36 that simultaneously inhibited PI3K and HDAC with nanomolar potencies and demonstrated favorable antiproliferative activities. Compounds 23 and 36 efficiently modulated the expression of p-AKT and Ac-H3, arrested the cell cycle, and induced apoptosis in HCT116 cancer cells. Following pharmacokinetic studies, 23 was further evaluated in HCT116 and HGC-27 xenograft models to show significant in vivo anticancer efficacies with tumor growth inhibitions of 45.8% (po, 150 mg/kg) and 62.6% (ip, 30 mg/kg), respectively. Overall, this work shows promise in discovering new anticancer therapeutics by the approach of simultaneously targeting PI3K and HDAC pathways with a single molecule.


Assuntos
Antineoplásicos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Inibidores de Histona Desacetilases/metabolismo , Histona Desacetilases/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase/metabolismo , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Relação Dose-Resposta a Droga , Desenho de Fármacos , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Células HCT116 , Células Hep G2 , Inibidores de Histona Desacetilases/síntese química , Inibidores de Histona Desacetilases/farmacologia , Humanos , Células K562 , Células MCF-7 , Camundongos , Camundongos Endogâmicos ICR , Simulação de Acoplamento Molecular/métodos , Inibidores de Fosfoinositídeo-3 Quinase/síntese química , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA