Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Acta Pharmacol Sin ; 41(3): 394-403, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31645658

RESUMO

RNF6, a RING-type ubiquitin ligase, has been identified as an oncogene in various cancers but its role in multiple myeloma (MM) remains elusive. In the present study we first showed that the expression levels of RNF6 in MM were significantly elevated compared with the bone marrow cells of healthy donors. Overexpression of RNF6 in LP1 and PRMI-8266 MM cell lines promoted cell proliferation, whereas knockdown of RNF6 led to apoptosis of MM cells. Furthermore, we revealed that RNF6, as a ubiquitin ligase, interacted with glucocorticoid receptor (GR) and induced its K63-linked polyubiquitination. Different from current knowledge, RNF6 increased GR stability at both endogenous and exogenous contexts. Such an action greatly promoted GR transcriptional activity, which was confirmed by luciferase assays and by the increased expression levels of prosurvival genes including Bcl-xL and Mcl-1, two typical downstream genes of the GR pathway. Consistent with these findings, ectopic expression of RNF6 in MM cells conferred resistance to dexamethasone, a typical anti-myeloma agent. In conclusion, we demonstrate that RNF6 promotes MM cell proliferation and survival by inducing atypical polyubiquitination to GR, and RNF6 could be a promising therapeutic target for the treatment of MM.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Mieloma Múltiplo/metabolismo , Receptores de Glucocorticoides/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Proteínas de Ligação a DNA/genética , Relação Dose-Resposta a Droga , Humanos , Estrutura Molecular , Mieloma Múltiplo/patologia , Receptores de Glucocorticoides/genética , Relação Estrutura-Atividade , Ubiquitinação
2.
Cancer Sci ; 109(4): 1220-1229, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29453896

RESUMO

Nuclear factor kappa B (NF-κB) signaling pathway is activated in many colorectal cancer (CRC) cells and in the tumor microenvironment, which plays a critical role in cancer initiation, development, and response to therapies. In the present study, we found that the widely used antimalarial drug mefloquine was a NF-κB inhibitor that blocked the activation of IκBα kinase, leading to reduction of IκBα degradation, decrease of p65 phosphorylation, and suppressed expression of NF-κB target genes in CRC cells. We also found that mefloquine induced growth arrest and apoptosis of CRC cells harboring phosphorylated p65 in culture and in mice. Furthermore, expression of constitutive active IKKß kinase significantly attenuated the cytotoxic effect of the compound. These results showed that mefloquine could exert antitumor action through inhibiting the NF-κB signaling pathway, and indicated that the antimalarial drug might be repurposed for anti-CRC therapy in the clinic as a single agent or in combination with other anticancer drugs.


Assuntos
Antimaláricos/farmacologia , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Mefloquina/farmacologia , NF-kappa B/metabolismo , Animais , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Quinase I-kappa B/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
3.
J Biol Chem ; 291(18): 9617-28, 2016 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-26971355

RESUMO

RNF6 is a little-studied ring finger protein. In the present study, we found that RNF6 was overexpressed in various leukemia cells and that it accelerated leukemia cell proliferation, whereas knockdown of RNF6 delayed tumor growth in xenografts. To find out the mechanism of RNF6 overexpression in leukemia, we designed a series of truncated constructs of RNF6 regulatory regions in the luciferase reporter system. The results revealed that the region between -144 and -99 upstream of the RNF6 transcription start site was critical and that this region contained a PBX1 recognition element (PRE). PBX1 modulated RNF6 expression by binding to the specific PRE. When PRE was mutated, RNF6 transcription was completely abolished. Further studies showed that PBX1 collaborated with PREP1 but not MEIS1 to modulate RNF6 expression. Moreover, RNF6 expression could be suppressed by doxorubicin, a major anti-leukemia agent, via down-regulating PBX1. This study thus suggests that RNF6 overexpression in leukemia is under the direction of PBX1 and that the PBX1/RNF6 axis can be developed as a novel therapeutic target of leukemia.


Assuntos
Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/metabolismo , Regulação Leucêmica da Expressão Gênica , Leucemia/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Elementos de Resposta , Animais , Proteínas de Ligação a DNA/genética , Doxorrubicina/farmacologia , Células HL-60 , Xenoenxertos , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Células Jurkat , Células K562 , Leucemia/tratamento farmacológico , Leucemia/genética , Leucemia/patologia , Camundongos , Proteína Meis1 , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Fator de Transcrição 1 de Leucemia de Células Pré-B , Proteínas Proto-Oncogênicas/genética
4.
J Biol Chem ; 288(47): 34181-34189, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24114842

RESUMO

The antiparasitic clioquinol (CQ) represents a class of novel anticancer drugs by interfering with proteasome activity. In the present study, we found that CQ induced blood cancer cell apoptosis by inhibiting histone deacetylases (HDACs). CQ accumulated the acetylation levels of several key proteins including histone H3 (H3), p53, HSP90, and α-tubulin. In the mechanistic study, CQ was found to down-regulate HDAC1, -3, -4, and -5 in both myeloma and leukemia cells. Computer modeling analysis revealed that CQ was well docked into the active pocket of the enzyme, where the oxygen and nitrogen atoms in CQ formed stable coordinate bonds with the zinc ion, and the hydroxyl group from CQ formed an effective hydrogen bond with Asp-267. Moreover, co-treatment with CQ and zinc/copper chloride led to decreased Ac-H3. Furthermore, CQ inhibited the activity of Class I and IIa HDACs in the cell-free assays, demonstrating that CQ interfered with HDAC activity. By inhibiting HDAC activity, CQ induced expression of p21, p27, and p53, cell cycle arrest at G1 phase, and cell apoptosis. This study suggested that the HDAC enzymes are targets of CQ, which provided a novel insight into the molecular mechanism of CQ in the treatment of hematological malignancies.


Assuntos
Antipruriginosos/farmacologia , Apoptose/efeitos dos fármacos , Clioquinol/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Leucemia/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Cloretos/farmacologia , Regulação para Baixo/efeitos dos fármacos , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , Células K562 , Leucemia/enzimologia , Leucemia/patologia , Masculino , Antissépticos Bucais/farmacologia , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/metabolismo , Células U937 , Compostos de Zinco/farmacologia
5.
J Biochem Mol Toxicol ; 28(5): 232-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24615755

RESUMO

Dihydrorotenone (DHR) is a natural pesticide used for farming including organic produces. We recently found that DHR induces human plasma cell apoptosis by provoking endoplasmic reticulum stress. In the present study, we found that DHR arrested human plasma cancer cells at the G0/G1 phase of the cell cycle. Mechanistical studies demonstrated that cell cycle arrest was associated with downregulated cell cycle promotors including cyclin D2, cyclin D3, cyclin-dependent kinases (CDK4, CKD6), and phosphorylated-Rb. DHR inhibited cyclin D2 transactivation, thus inhibiting its mRNA expression. In addition, DHR upregulated the cell cycle repressors p21 and p53. DHR also increased the phosphorylation level of p53, suggesting the upregulated transactivation function of p53, which was confirmed by the induction of p21, a substrate of activated p53. Moreover, DHR downregulated AKT and ERK phosphorylation, an incentive of cell cycle progression. Therefore, these results collectively demonstrated that DHR disrupts the cell cycle progress, which suggests that DHR is toxic to human plasma cells. Caution is thus suggested when handling with this agent.


Assuntos
Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Fase G1/efeitos dos fármacos , Inseticidas/toxicidade , Plasmócitos/efeitos dos fármacos , Fase de Repouso do Ciclo Celular/efeitos dos fármacos , Rotenona/análogos & derivados , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Ciclina D2/genética , Quinases Ciclina-Dependentes/metabolismo , Humanos , Inseticidas/química , Estrutura Molecular , Plasmócitos/enzimologia , Plasmócitos/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rotenona/química , Rotenona/toxicidade , Transdução de Sinais
6.
Bioorg Med Chem Lett ; 23(11): 3314-9, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23601711

RESUMO

The small chemical compound 8-ethoxy-2-(4-fluorophenyl)-3-nitro-2H-chromene (S14161) was recently identified as an inhibitor of the phosphoinositide 3-kinase (PI3K). In the present study, we designed a novel synthesis of S14161 and prepared a series of its analogues via the oxa-Michael-Henry reaction in the presence of catalytic amounts of l-proline and triethylamine. Further structural simplification led to the identification of 6-bromo-8-ethoxy-3-nitro-2H-chromene (BENC-511) that exhibited potent antiproliferative activities against a panel of 12 tumor cell lines. Compared with S14161, BENC-511 was more potent in blocking the AKT phosphorylation and inducing cancer cell apoptosis. BENC-511 also displayed more potent effects on human umbilical vein epithelial cells (HUVEC) migration, suggesting its anti-angiogenesis activity.


Assuntos
Antineoplásicos/síntese química , Benzopiranos/química , Benzopiranos/síntese química , Inibidores de Fosfoinositídeo-3 Quinase , Antineoplásicos/química , Antineoplásicos/toxicidade , Benzopiranos/farmacologia , Sítios de Ligação , Catálise , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Células Endoteliais da Veia Umbilical Humana , Humanos , Simulação de Acoplamento Molecular , Neovascularização Fisiológica/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Piperazinas/química , Prolina/química , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/metabolismo
7.
Am J Cancer Res ; 13(11): 5382-5393, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38058843

RESUMO

In the effort to identify deubiquitinating enzymes required for the growth of colorectal cancer (CRC) cells, we found that OTUB2 knockdown markedly inhibited the viability of these cancer cells in culture and in xenografted mice. It was also found that the level of OTUB2 was elevated in primary CRCs, and its high expression was a poor prognostic indicator for the patients. Interestingly, immunoprecipitation and LC-MS/MS analyses suggested that ß-Catenin was an OTUB2-interacting protein, and there was a positive correlation between OTUB2 and ß-Catenin expression in both CRC tissues and cell lines. We then performed reciprocal co-immunoprecipitations and demonstrated that OTUB2 and ß-Catenin bound to each other. Enforced expression of OTUB2 decreased ubiquitination of ß-Catenin and increased the half-life and intracellular level of ß-Catenin, whereas the catalytic inactive OTUB2 did not. OTUB2 also enhanced ß-Catenin-mediated transactivation as measured by TCF-luciferase and expression of endogenous CCND1 and MYC in CRC cells. These results indicated that OTUB2 was a potential target for therapeutic intervention for CRC.

8.
Int J Cancer ; 131(10): 2411-9, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22362357

RESUMO

The pan-Src family kinase inhibitor dasatinib has been approved for chronic myeloid leukemia treatment but displays limited activity in lung cancer patients. In this study, we used a deuterium substitution strategy to develop a class of novel chemicals based on dasatinib and found that these compounds maintain inhibition on c-Src activity and display anti-non-small cell lung cancer activity in vitro and in vivo. BRP800, one of these compounds, was chosen for further studies. BRP800 mainly displayed antiproliferative but not proapoptotic activity. Molecularly, BRP800 did not show significant effects on the expression of antiapoptotic genes, such as Bcl-2 and Mcl1, or on the activation of apoptotic enzymes, such as caspase-3, -8 or 9. However, BRP800 decreased expression of cell cycle promoting genes such as cyclins D1, D3, E, A and CDK4 and 6, and increased the expression of cell cycle negative regulators including p21, p27 and p53. Consistent with these findings, BRP800 arrested cells at the G0/G1 phase in a concentration-dependent manner, and the G0/G1 fraction was increased from 64% in control to 85% in BRP800-treated cells. We also evaluated the effects of BRP800 on NSCLC xenografts using H460 as a model in nude mice. Compared with the known NSCLC drug docetaxel, BRP800 displayed potent and similar antitumor activity but with less toxicity. These findings suggest that the deuterated analog of dasatinib is antiproliferative by inhibiting c-Src and disrupting cell cycle progression, and could be further developed as a novel drug for non-small lung cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Ciclo Celular/efeitos dos fármacos , Neoplasias Pulmonares/metabolismo , Pirimidinas/farmacologia , Tiazóis/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dasatinibe , Deutério/química , Ativação Enzimática/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Camundongos Nus , Pirimidinas/administração & dosagem , Pirimidinas/química , Transdução de Sinais/efeitos dos fármacos , Tiazóis/administração & dosagem , Tiazóis/química , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Quinases da Família src/antagonistas & inibidores
9.
J Nanosci Nanotechnol ; 12(12): 8955-62, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23447944

RESUMO

Using poly (ethylene oxide)-poly(propylene oxide)-poly(ethylene oxide) triblock copolymer (P123) as template and tetraethyl orthosilicate as silica source, well-ordered cross-linked bimodal mesoporous SBA-15 was easily prepared via an one-pot synthetic strategy, where a pre-self-assembly of P123 with TEOS in strong acidic media was introduced followed by pH-adjusting. The pH-adjusting can effectively change the copolymer micelle size, which directly lead to the formation of the bimodal mesoporous structure at pH range of 6-7.5, with most typical value at pH = 7.5. Meanwhile, these bimodal mesopores are interconnected, which generates a 3D intra-particle porosity with many pore-openings on the surfaces of particles. After modified with tetraethylenepentamine, the as-synthesized bimodal mesoporous SBA-15 exhibited the CO2 adsorption capacities much higher than as-synthesized conventional SBA-15. These bimodal mesoporous SBA-15 materials have great potential application in CO2 capture.

10.
J Nanosci Nanotechnol ; 11(5): 4079-88, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21780409

RESUMO

Mesoporous silica/cotton fiber composite materials have been prepared in situ by using pluronics P123 (EO20PO70EO20) as template, tetraethyl orthosilicate as silica source and degreasing cotton as supporter. In order to avoid the hydrolysis of cotton fiber in a strong acidic media during the hydrothermal treatment, two kinds of methods were used to control the acidity of the reaction media. One was to adjust the pH to 5 after self-assembly in a strong acidic media; the other was a two-step route containing the pre-hydrolysis of TEOS and self-assembly in a weak acidic media. The resulting shaped composite materials presented the morphology of cotton fiber, and the silica particles mainly covered the surfaces of cotton fibers. These silica particles possessed a slightly ordered pore structure or a well ordered SBA-15 structure according to the difference in the synthetic methods. After modification with tetraethylenepentamine, these shaped composite materials exhibited considerable CO2 adsorption capacity. The use of cotton fiber has the advantages of shaping the powdery materials, dispersing the silica particles and avoiding the formation of moisture and sticky solid surfaces by overloaded tetraethylenepentamine.

11.
Theranostics ; 9(14): 4208-4220, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31281542

RESUMO

Ubiquitin specific peptidase 5 (USP5) is a ubiquitous expressed deubiquitinating enzyme (DUB). It has been shown involved in DNA repair, apoptosis, inflammation, and tumor cell growth. However, the function and molecular mechanism of USP5 in colorectal cancer (CRC) are still unclear. In the present study, we asked how it affected the growth of colorectal cancer cells. Methods: A shRNA-based high-content screening was performed to identify DUBs affecting the growth of CRC cells. CCK-8 assay and xenografts were used to assess CRC cell growth, survival and tumorigenesis. RT-qPCR, immunoblotting and immunohistochemistry were carried out to quantitate USP5 expression in CRC tissues and cell lines. Immunoprecipitation and mass spectrometry analysis were performed to identify USP5-interacting proteins. Cycloheximide chase was performed to assess Tu translation elongation factor (TUFM) stability. Dual luciferase reporter assay was utilized for USP5 promoter analysis. Results: We found that USP5 was highly expressed in a group of primary CRC tissues, and the increased USP5 was correlated with clinical stages and shorter overall survival. While USP5 knockdown effectively inhibited CRC cell growth, overexpressed USP5 promoted the growth of CRC cells and made them more resistant to doxorubicin (DOX). TUFM was discovered as a substrate of USP5. USP5 deubiquitinated TUFM and increased its level in CRC cells. Enforced expression of TUFM was able to alleviate the growth inhibition induced by USP5 knockdown. Further analyses showed that EBF transcription factor 1 (EBF1) was a major regulator for USP5 transcription, and DOX inhibited EBF1-USP5-TUFM axis in CRC cells. Conclusions: USP5 was required for CRC cells and promoted their growth and resistance to chemotherapeutics. TUFM was a USP5 deubiquitinating substrate that mediated the cellular effects of USP5. The transcription of USP5 was regulated by EBF1. Thus, targeting EBF1-USP5-TUFM axis is a potential novel strategy for CRC treatment.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proteínas Mitocondriais/metabolismo , Fator Tu de Elongação de Peptídeos/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Neoplasias Colorretais/genética , Feminino , Células HCT116 , Células HT29 , Humanos , Immunoblotting , Lentivirus/genética , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Proteínas Mitocondriais/genética , Fator Tu de Elongação de Peptídeos/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteases Específicas de Ubiquitina/genética
12.
Front Oncol ; 9: 1406, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31921663

RESUMO

Mutations and altered expression of deubiquitinating enzymes (DUBs) have been found associated with many human diseases including cancers. In this study, Ubiquitin specific protease 1 (USP1) expression was found significantly increased in some colorectal cancers (CRC). The elevated USP1 level was associated with short overall survival of patients and with advanced stages of cancers. In cultured CRC cells, knockdown of USP1 induced growth arrest at G2/M of cell cycle and reduced the expression of anti-apoptotic proteins Bcl-2 and Mcl-1. Its knockdown also led to reduction of DNA-repair related substrates FANCD2 and ID1. Further investigations found that small molecular inhibitor of USP1 ML323 sensitized CRC cells to DNA-targeting chemotherapeutics, including doxorubicin, TOPI/II inhibitors, and PARP inhibitor, but not to 5-Fu. These results indicate that USP1 plays a critical in colorectal cancer cell survival and is a promising target for anti-colorectal cancer chemotherapy. Targeting USP1 may represent an effective strategy to regulate the DNA-repairing system.

14.
Oncotarget ; 8(12): 20103-20112, 2017 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-28223545

RESUMO

Ring finger protein 6 (RNF6) is a key oncogene in both prostate cancer and leukemia, but its role is elusive in breast cancer. In the present study, we found that RNF6 was overexpressed in more than 70% of breast cancer tissues and it was associated with overall survival. RNF6 increased breast cancer cell proliferation, migration and reduced cell sensitivity to doxorubicin. Further studies showed that RNF6 was closely associated with increased expression of estrogen receptor, a critical factor in the development of breast cancers. RNF6 was found to induce ERα expression and increased its stability. In doxorubicin-resistant breast cancer cells, RNF6 was found to be elevated in association with increased ERα and anti-apoptotic Bcl-xL, but not pro-apoptotic Bim-1. In consistence with this finding, overexpression of ERα led to increased Bcl-xL but had no effects on Bim-1. Therefore, this study demonstrated that there exists an RNF6/ERα/Bcl-xL axle in breast cancer which promotes cancer cell proliferation and survival. Targeting the RNF6/ERα/Bcl-xL axle could be a promising strategy in the treatment of breast cancer.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/secundário , Proliferação de Células , Proteínas de Ligação a DNA/metabolismo , Receptor alfa de Estrogênio/metabolismo , Proteína bcl-X/metabolismo , Apoptose , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Estudos de Casos e Controles , Proteínas de Ligação a DNA/genética , Receptor alfa de Estrogênio/genética , Feminino , Seguimentos , Humanos , Metástase Linfática , Pessoa de Meia-Idade , Gradação de Tumores , Invasividade Neoplásica , Estadiamento de Neoplasias , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas , Proteína bcl-X/genética
15.
Oncotarget ; 7(46): 75539-75550, 2016 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-27705908

RESUMO

The activated JAK2-STAT3 signaling pathway is a high risk factor for multiple myeloma (MM), a fatal malignancy of plasma cells. In the present study, SC09, a potential inhibitor of cholesterol absorption, was identified in a STAT3-targeted drug screen. SC09 suppressed the activation of STAT3 in a time-course and concentration-dependent manner but did not affect its family members STAT1 and STAT5. SC09 inhibited STAT3 transcriptional activity and downregulated the expression of STAT3-regulated genes. Further studies showed that SC09 selectively inhibited JAK2 activation but not other kinases including c-Src, ERK, p38 and mTOR that are all associated with STAT3 activation. Moreover, SC09 obviously induced MM cell death in vitro and delayed MM tumor growth in vivo. SC09-induced MM cell death was dependent on the endogenous STAT3 status, and this effect could be attenuated by enforced expression of STAT3. All the results collectively indicated that SC09 blocks the JAK2-STAT3 signaling thus displaying anti-MM activity. Given its well tolerance and anti-MM potency, SC09 is credited for further investigation as a promising drug for MM treatment.


Assuntos
Antineoplásicos/farmacologia , Colesterol/metabolismo , Janus Quinase 2/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Mieloma Múltiplo/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Humanos , Janus Quinase 2/química , Camundongos , Camundongos Nus , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Fosforilação , Fator de Transcrição STAT3/genética , Ativação Transcricional/efeitos dos fármacos
16.
Oncotarget ; 7(8): 9296-308, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26814430

RESUMO

The oncogenic STAT3 signaling pathway is emerging as a promising target for the treatment of multiple myeloma (MM). In the present study, we identified a novel STAT3 inhibitor SC99 in a target-based high throughput screen. SC99 inhibited JAK2-STAT3 activation but had no effects on other transcription factors such as NF-κB, and kinases such as AKT, ERK, and c-Src that are in association with STAT3 signaling pathway. Furthermore, SC99 downregulated the expression of STAT3-modulated genes, including Bcl-2, Bcl-xL, VEGF, cyclin D2, and E2F-1. By inhibiting the STAT3 signaling, SC99 induced MM cell apoptosis which could be partly abolished by the ectopic expression of STAT3. Furthermore, SC99 displayed potent anti-MM activity in two independent MM xenograft models in nude mice. Oral administration of SC99 led to marked decrease of tumor growth within 10 days at a daily dosage of 30 mg/kg, but did not raise toxic effects. Taken together, this study identified a novel oral JAK2/STAT3 inhibitor that could be developed as an anti-myeloma agent.


Assuntos
Antineoplásicos/uso terapêutico , Hidrazonas/uso terapêutico , Janus Quinase 2/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Células 3T3 , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Ciclina D2/biossíntese , Fator de Transcrição E2F1/biossíntese , Ativação Enzimática/efeitos dos fármacos , Feminino , Células HeLa , Humanos , Camundongos , Camundongos Nus , Mieloma Múltiplo/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína bcl-X/biossíntese
17.
Sci Rep ; 5: 12809, 2015 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-26329846

RESUMO

The mammalian target of rapamycin (mTOR) is extensively involved in multiple myeloma (MM) pathophysiology. In the present study, we reported a novel small molecule SC06 that induced MM cell apoptosis and delayed MM xenograft growth in vivo. Oral administration of SC06 to mice bearing human MM xenografts resulted in significant inhibition of tumor growth at doses that were well tolerated. Mechanistic studies revealed that SC06 selectively inhibited the mTOR signaling pathway but had no effects on other associated kinases, such as AKT, ERK, p38, c-Src and JNK. Further studies showed that SC06-decreased mTOR activation was associated with the downregulation of Raptor, a key component of the mTORC1 complex. SC06 also suppressed the phosphorylation of 4E-BP1 and P70S6K, two typical substrates in the mTORC1 signaling pathway. Notably, expression of Raptor, phosphorylation of mTOR and phosphorylated 4E-BP1 was also decreased in the tumor tissues from SC06-treated mice, which was consistent with the cellular studies. Therefore, given the potency and low toxicity, SC06 could be developed as a potential anti-MM drug candidate by disrupting the mTOR signaling.


Assuntos
Aminopiridinas/uso terapêutico , Hidrazonas/uso terapêutico , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/uso terapêutico , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aminopiridinas/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Feminino , Células HEK293 , Humanos , Hidrazonas/farmacologia , Lisossomos/efeitos dos fármacos , Lisossomos/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos Nus , Mieloma Múltiplo/patologia , Complexos Multiproteicos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Regulatória Associada a mTOR , Bibliotecas de Moléculas Pequenas/farmacologia
18.
Oncotarget ; 6(1): 185-95, 2015 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-25474140

RESUMO

Recent clinical trials have demonstrated targeting PI3K pathway is a promising strategy for the treatment of blood cancers. To identify novel PI3K inhibitors, we performed a high throughput virtual screen and identified several novel small molecule compounds, including PIK-C98 (C98). The cell-free enzymatic studies showed that C98 inhibited all class I PI3Ks at nano- or low micromolar concentrations but had no effects on AKT or mTOR activity. Molecular docking analysis revealed that C98 interfered with the ATP-binding pockets of PI3Ks by forming H-bonds and arene-H interactions with specific amino acid residues. The cellular assays demonstrated that C98 specifically inhibited PI3K/AKT/mTOR signaling pathway, but had no effects on other kinases and proteins including IGF-1R, ERK, p38, c-Src, PTEN, and STAT3. Inhibition of PI3K by C98 led to myeloma cell apoptosis. Furthermore, oral administration of C98 delayed tumor growth in two independent human myeloma xenograft models in nude mice but did not show overt toxicity. Pharmacokinetic analyses showed that C98 was well penetrated into myeloma tumors. Therefore, through a high throughput virtual screen we identified a novel PI3K inhibitor that is orally active against multiple myeloma with great potential for further development.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Furanos/farmacologia , Regulação Neoplásica da Expressão Gênica , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Tiocarbamatos/farmacologia , Administração Oral , Animais , Apoptose , Linhagem Celular Tumoral , Sobrevivência Celular , Feminino , Humanos , Ligação de Hidrogênio , Camundongos , Camundongos Nus , Camundongos SCID , Simulação de Dinâmica Molecular , Mieloma Múltiplo/tratamento farmacológico , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais
19.
Toxicol Lett ; 228(3): 133-9, 2014 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-24831963

RESUMO

Recent studies demonstrated that targeting the phosphatidylinositide 3-kinase (PI3K)/AKT signaling pathway is a major strategy for the treatment of androgen-independent prostate cancer. In the present study, we developed an analog BENC-511 from a recently reported PI3K inhibitor S14161 by structural optimization. Using PC3 and DU145 as the model cell lines, we found PTEN-deficient PC3 cells were more sensitive than PTEN-expressing DU145 ones in terms of cell proliferation, apoptosis, and caspase-3 activation. These findings were consistent with the inhibition on PI3K/AKT signals. BENC-511 preferably suppressed AKT activation in PC3 over DU145 cells. Notably, PTEN restoration attenuated BENC-511 induced apoptosis. Moreover, BENC-511 displayed great therapeutic efficacy in a PC3-derived prostate cancer model in nude mice. With an oral dosage of 50mg/kg, BENC-511 decreased tumor growth more than 50% in 27 days, which was accompanied with PARP cleavage, but did not show overt toxicity. This study lays a solid rationale for the development of BENC-511 as a drug for the treatment of PTEN-deficient and androgen-independent prostate cancers.


Assuntos
Antineoplásicos/farmacologia , Benzopiranos/farmacologia , PTEN Fosfo-Hidrolase/deficiência , Inibidores de Fosfoinositídeo-3 Quinase , Neoplasias da Próstata/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Benzopiranos/administração & dosagem , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ativação Enzimática , Humanos , Masculino , Camundongos , Camundongos Nus , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Neoplasias da Próstata/enzimologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Inibidores de Proteínas Quinases/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Hematol Oncol ; 7: 9, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24428908

RESUMO

BACKGROUND: We previously reported a PI3K inhibitor S14161 which displays a promising preclinical activity against multiple myeloma (MM) and leukemia, but the chiral structure and poor solubility prevent its further application. METHODS: Six S14161 analogs were designed based on the structure-activity relationship; activity of the compounds in terms of cell death and inhibition of PI3K were analyzed by flow cytometry and Western blotting, respectively; anti-myeloma activity in vivo was performed on two independent xenograft models. RESULTS: Among the six analogs, BENC-511 was one of the most potent compounds which significantly inhibited PI3K activity and induced MM cell apoptosis. BENC-511 was able to inactivate PI3K and its downstream signals AKT, mTOR, p70S6K, and 4E-BP1 at 1 µM but had no effects on their total protein expression. Consistent with its effects on PI3K activity, BENC-511 induced MM cell apoptosis which was evidenced by the cleavage of Caspase-3 and PARP. Notably, addition of insulin-like growth factor 1 and interleukin-6, two important triggers for PI3K activation in MM cells, partly blocked BENC-511-induced MM cell death, which further demonstrated that PI3K signaling pathway was critical for the anti-myeloma activity of BENC-511. Moreover, BENC-511 also showed potent oral activity against myeloma in vivo. Oral administration of BENC-511 decreased tumor growth up to 80% within 3 weeks in two independent MM xenograft models at a dose of 50 mg/kg body weight, but presented minimal toxicity. Suppression of BENC-511 on MM tumor growth was associated with decreased PI3K/AKT activity and increased cell apoptosis. CONCLUSIONS: Because of its potent anti-MM activity, low toxicity (LD50 oral >1.5 g/kg), and easy synthesis, BENC-511 could be developed as a promising agent for the treatment of MM via suppressing the PI3K/AKT signaling pathway.


Assuntos
Antineoplásicos/farmacologia , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Benzopiranos/química , Benzopiranos/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Humanos , Immunoblotting , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-6/farmacologia , Camundongos , Camundongos Nus , Estrutura Molecular , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Poli(ADP-Ribose) Polimerases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Carga Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA