Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 131
Filtrar
1.
Bioorg Chem ; 130: 106234, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36375353

RESUMO

Ras protein has been considered a fascinating target for anticancer therapy because its malfunction is closely related to cancer. However, Ras has been considered undruggable because of the failure to regulate its malfunction by controlling the Ras activation mechanism. Recently, Lumakras targeting the G12C mutation was approved, and therapeutic interest in Ras for anticancer therapy has been rejuvenated. Here, we present a series of compounds that inhibit Ras via a unique mechanism of action that exploits the relationship between the Wnt/ß-catenin pathway and Ras. KYA1797K (1) binds to axin to stabilize the ß-catenin destruction complex that causes the phosphorylation and subsequent degradation of Ras, similar to canonical ß-catenin regulation. Based on the chemical structure of 1, we performed a structural optimization and identified 3-(2-hydroxyethyl)-5-((6-(4-nitrophenyl)pyridin-2-yl)methylene)thiazolidine-2,4-dione (13d) as the most potent compound. 13d displayed antitumor effects in a colorectal cancer model with enhanced inhibition activity on Ras. The results of this study suggest that the further development of 13d could contribute to the development of Ras inhibitors with novel mechanisms of action.


Assuntos
Neoplasias Colorretais , beta Catenina , Proteínas ras , Humanos , Proteína Axina/química , Proteína Axina/genética , Proteína Axina/metabolismo , beta Catenina/química , beta Catenina/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Proteínas ras/efeitos dos fármacos , Proteínas ras/metabolismo , Via de Sinalização Wnt
2.
Int J Mol Sci ; 24(5)2023 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-36901935

RESUMO

Oral squamous cell carcinoma (OSCC) is the sixth most common type of cancer worldwide. Despite advancement in treatment, advanced-stage OSCC is associated with poor prognosis and high mortality. The present study aimed to investigate the anticancer activities of semilicoisoflavone B (SFB), which is a natural phenolic compound isolated from Glycyrrhiza species. The results revealed that SFB reduces OSCC cell viability by targeting cell cycle and apoptosis. The compound caused cell cycle arrest at the G2/M phase and downregulated the expressions of cell cycle regulators including cyclin A and cyclin-dependent kinase (CDK) 2, 6, and 4. Moreover, SFB induced apoptosis by activating poly-ADP-ribose polymerase (PARP) and caspases 3, 8, and 9. It increased the expressions of pro-apoptotic proteins Bax and Bak, reduced the expressions of anti-apoptotic proteins Bcl-2 and Bcl-xL, and increased the expressions of the death receptor pathway protein Fas cell surface death receptor (FAS), Fas-associated death domain protein (FADD), and TNFR1-associated death domain protein (TRADD). SFB was found to mediate oral cancer cell apoptosis by increasing reactive oxygen species (ROS) production. The treatment of the cells with N-acetyl cysteine (NAC) caused a reduction in pro-apoptotic potential of SFB. Regarding upstream signaling, SFB reduced the phosphorylation of AKT, ERK1/2, p38, and JNK1/2 and suppressed the activation of Ras, Raf, and MEK. The human apoptosis array conducted in the study identified that SFB downregulated survivin expression to induce oral cancer cell apoptosis. Taken together, the study identifies SFB as a potent anticancer agent that might be used clinically to manage human OSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Bucais , Humanos , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Quinases de Proteína Quinase Ativadas por Mitógeno , Neoplasias Bucais/tratamento farmacológico , Neoplasias Bucais/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas ras/efeitos dos fármacos , Proteínas ras/metabolismo , Proteínas Proto-Oncogênicas c-raf/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-raf/metabolismo
3.
Int J Neurosci ; 131(10): 975-983, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32378973

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common aggressive primary cancer occurring in the brain tissue. GBM accounts 16% of primary brain tumors and half of gliomas. Additionally, the incidence of GBM is increases with aging, and reaches the peak at the age of 75 to 84 years. The survival of patients with GBM remains at a low level, only less than 5% patients diagnosed with GBM survive for 5 years. Temozolomide (TMZ) is a DNA alkylating agent and is currently a first line chemotherapeutic treatment for GBM. TMZ combined with radiation therapy has been shown to prolong the overall survival (OS) to 14.6 months compared with 12.1 months for radiation therapy alone. NF-E2-related factor 2 (Nrf2) is a transcription factor that contains seven functional domains. The binding of Keap1 to Nrf2 is a central regulator of the cellular defense mechanism against environmental stresses. METHODS: First, Nrf2 overexpression and inhibition models were constructed in U251 cells using transfection. The percentage of viable cells was detected using the MTT assay. Then, the expression of the HO-1 regulator was detected using qPCR, and the concentrations of oxidative stress related factors were detected using ELISAs. The levels of proteins related to oxidative stress and the Ras/Raf/MEK signaling pathway was detected using western blotting analysis. RESULTS: We initially established Nrf2 inhibition and activation cell models in U251 cells and found that the inhibition of Nrf2 expression decreased the mRNA and protein levels of the anti-oxidative enzymes, as well as the secretion of these enzymes into the cellular microenvironment. These effects might be mediated by the inhibition of Ras/Raf/MEK signaling pathway, leading to the inhibition of cellular proliferation. CONCLUSIONS: Inhibition of Nrf2 expression might enhance the effect of TMZ on the treatment of GBM and might be a new therapeutic strategy.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Glioma/tratamento farmacológico , MAP Quinase Quinase Quinases/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/efeitos dos fármacos , Temozolomida/farmacologia , Quinases raf/efeitos dos fármacos , Proteínas ras/efeitos dos fármacos , Humanos , MAP Quinase Quinase Quinases/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Quinases raf/antagonistas & inibidores , Proteínas ras/antagonistas & inibidores
4.
Org Biomol Chem ; 18(16): 3069-3081, 2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32101243

RESUMO

The use of selective covalent inhibitors with low binding affinity and high reactivity with the target enzyme is a promising way to solve a long-standing problem of the "undruggable" RAS-like proteins. Specifically, compounds of the ARS family that prevent the activation of the GDP-bound G12C mutant of Kirsten RAS (KRAS) are in the focus of recent experimental research. We report the first computational characterization of the entire reaction mechanism of the covalent binding of ARS-853 to the KRASG12C·GDP complex. The application of molecular dynamics, molecular docking and quantum mechanics/molecular mechanics approaches allowed us to model the inhibitor binding to the protein and the chemical reaction of ARS-853 with Cys12 in the enzyme binding site. We estimated a full set of kinetic constants and carried out numerical kinetic analysis of the process. Thus, we were able to compare directly the physicochemical parameters of the reaction obtained in silico and the macroscopic parameters observed in experimental studies. From our computational results, we explain the observed unusual dependence of the rate constant of covalent complex formation, kobs, on the ARS concentration. The latter depends both on the non-covalent binding step with the equilibrium constant, Ki, and on the rate constant of covalent adduct formation, kinact. The calculated ratio kinact/Ki = 213 M-1 s-1 reproduces the corresponding experimental value of 250 ± 30 M-1 s-1 for the interaction of ARS-853 with KRASG12C. Electron density analysis in the reactive region demonstrates that covalent bond formation occurs efficiently according to the Michael addition mechanism, which assumes the activation of the C[double bond, length as m-dash]C bond of ARS-853 by a water molecule and Lys16 in the binding site of KRASG12C. We also refine the kinact and Ki constants of the ARS-107 compound, which shares common features with ARS-853, and show that the decrease in the kinact/Ki ratio in the case of ARS-107 is explained by changes in both Ki and kinact constants.


Assuntos
Azetidinas/metabolismo , Piperazinas/metabolismo , Proteínas ras/antagonistas & inibidores , Azetidinas/farmacologia , Sítios de Ligação , Guanosina Difosfato/metabolismo , Humanos , Cinética , Simulação de Acoplamento Molecular , Mutação , Piperazinas/farmacologia , Estudo de Prova de Conceito , Ligação Proteica , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética , Proteínas ras/metabolismo
5.
BMC Anesthesiol ; 20(1): 43, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32085741

RESUMO

BACKGROUND: Substantial clinical and preclinical evidence have indicated the association between amide-linked local anesthesia and the long-term outcomes of cancer patients. However, the potential effects of local anesthesia on cancer recurrence are inconclusive and the underlying mechanisms remain poorly understood. METHODS: We systematically examined the effects of three commonly used local anesthetics in melanoma cells and analyzed the underlying mechanisms focusing on small GTPases. RESULTS: Ropivacaine and lidocaine but not bupivacaine inhibited migration and proliferation, and induced apoptosis in melanoma cells. In addition, ropivacaine and lidocaine but not bupivacaine significantly augmented the in vitro efficacy of vemurafenib (a B-Raf inhibitor for melanoma with BRAF V600E mutation) and dacarbazine (a chemotherapeutic drug). Mechanistically, ropivacaine but not bupivacaine decreased the activities of Ras superfamily members with the dominant inhibitory effects on RhoA and Ras, independent of sodium channel blockade. Rescue studies using constitutively active Ras and Rho activator calpeptin demonstrated that ropivacaine inhibited migration mainly through RhoA whereas growth and survival were mainly inhibited through Ras in melanoma cells. We further detected a global reduction of downstream signaling of Ras and RhoA in ropivacaine-treated melanoma cells. CONCLUSION: Our study is the first to demonstrate the anti-melanoma activity of ropivacaine and lidocaine but not bupivacaine, via targeting small GTPases. Our findings provide preclinical evidence on how amide-linked local anesthetics could affect melanoma patients.


Assuntos
Anestésicos Locais/farmacologia , Melanoma/metabolismo , Bloqueadores dos Canais de Sódio/metabolismo , Proteínas ras/efeitos dos fármacos , Proteína rhoA de Ligação ao GTP/efeitos dos fármacos , Bupivacaína/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Técnicas In Vitro , Lidocaína/farmacologia , Melanoma/tratamento farmacológico , Ropivacaina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
6.
Respir Res ; 20(1): 194, 2019 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-31443651

RESUMO

BACKGROUND: The aim of this study was to investigate the effects and mechanisms of ectonucleoside triphosphate phosphohydrolase-7 (ENTPD7) on lung cancer cells. METHODS: The expression characteristics of ENTPD7 and its effect on the survival of lung cancer patients were analyzed by referring to The Cancer Genome Atlas (TCGA). Streptavidin-peroxidase (SP) staining was performed to detect the ENTPD7 protein in tumor tissues and adjacent tissues. Plasmid transfection technology was also applied to silence ENTPD7 gene. Crystal violet staining and flow cytometry were performed to determine cell proliferation and apoptosis. Tumor-bearing nude mice model was established to investigate the effect of sh-ENTPD7 on tumors. RESULTS: The results showed that patients with low levels of ENTPD7 had higher survival rates. ENTPD7 was up-regulated in lung cancer tissues and cells. Down-regulation of the expression of ENTPD7 inhibited proliferation but promoted apoptosis of lung cancer cell. Silencing ENTPD7 also inhibited the expression levels of Ras and Raf proteins and the phosphorylation of mitogen-activated protein kinase (MEK) and extracellular signal-regulated kinase (ERK). Tumor-bearing nude mice experiments showed that silencing ENTPD7 had an inhibitory effect on lung cancer cells. CONCLUSIONS: ENTPD7 was overexpressed in lung cancer cells. Down-regulating ENTPD7 could inhibit lung cancer cell proliferation and promote apoptosis via inhibiting the Ras/Raf/MEK/ERK pathway.


Assuntos
Apirase/antagonistas & inibidores , Apirase/genética , Neoplasias Pulmonares/terapia , Transdução de Sinais/efeitos dos fármacos , Adulto , Idoso , Animais , Apoptose , Biomarcadores , Linhagem Celular Tumoral , Proliferação de Células , Células Cultivadas , Feminino , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Neoplasias Pulmonares/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Plasmídeos , Análise de Sobrevida , Quinases raf/antagonistas & inibidores , Quinases raf/genética , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética
7.
Tumour Biol ; 39(10): 1010428317734947, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28990465

RESUMO

Recently, statins have been demonstrated to improve cancer-related mortality or prognosis in patients of various cancers. However, the details of the apoptosis-inducing mechanisms remain unknown. This study showed that the induction of apoptosis by statins in hematopoietic tumor cells is mediated by mitochondrial apoptotic signaling pathways, which are activated by the suppression of mevalonate or geranylgeranyl pyrophosphate biosynthesis. In addition, statins decreased the levels of phosphorylated extracellular signal-regulated kinase 1/2 and mammalian target of rapamycin through suppressing Ras prenylation. Furthermore, inhibition of extracellular signal-regulated kinase 1/2 and mammalian target of rapamycin by statins induced Bim expression via inhibition of Bim phosphorylation and ubiquitination and cell-cycle arrest at G1 phase via enhancement of p27 expression. Moreover, combined treatment of U0126, a mitogen-activated protein kinase kinase 1/2 inhibitor, and rapamycin, a mammalian target of rapamycin inhibitor, induced Bim and p27 expressions. The present results suggested that statins induce apoptosis by decreasing the mitochondrial transmembrane potential, increasing the activation of caspase-9 and caspase-3, enhancing Bim expression, and inducing cell-cycle arrest at G1 phase through inhibition of Ras/extracellular signal-regulated kinase and Ras/mammalian target of rapamycin pathways. Therefore, our findings support the use of statins as potential anticancer agents or concomitant drugs of adjuvant therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Hematológicas/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2/efeitos dos fármacos , Proteína 11 Semelhante a Bcl-2/metabolismo , Western Blotting , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Imunoprecipitação , Antígeno Nuclear de Célula em Proliferação/efeitos dos fármacos , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas ras/efeitos dos fármacos , Proteínas ras/metabolismo
8.
Mutagenesis ; 32(1): 77-90, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28011749

RESUMO

We used the marine bivalve (Mytilus galloprovincialis) to assess a range of biological or biomarker responses following exposure to a model-engineered nanoparticle, C60 fullerene, either alone or in combination with a model polycyclic aromatic hydrocarbon, benzo(α)pyrene [B(α)P]. An integrated biomarker approach was used that included: (i) determination of 'clearance rates' (a physiological indicator at individual level), (ii) histopathological alterations (at tissue level), (iii) DNA strand breaks using the comet assay (at cellular level) and (iv) transcriptional alterations of p53 (anti-oncogene) and ras (oncogene) determined by real-time quantitative polymerase chain reaction (at the molecular/genetic level). In addition, total glutathione in the digestive gland was measured as a proxy for oxidative stress. Here, we report that mussels showed no significant changes in 'clearance rates' after 1 day exposure, however significant increases in 'clearance rates' were found following exposure for 3 days. Histopathology on selected organs (i.e. gills, digestive glands, adductor muscles and mantles) showed increased occurrence of abnormalities in all tissues types, although not all the exposed organisms showed these abnormalities. Significantly, increased levels of DNA strand breaks were found after exposure for 3-days in most individuals tested. In addition, a significant induction for p53 and ras expression was observed in a tissue and chemical-specific pattern, although large amounts of inter-individual variability, compared with other biomarkers, were clearly apparent. Overall, biological responses at different levels showed variable sensitivity, with DNA strand breaks and gene expression alterations exhibiting higher sensitivities. Furthermore, the observed genotoxic responses were reversible after a recovery period, suggesting the ability of mussels to cope with the toxicants C60 and/or B(α)P under our experimental conditions. Overall, in this comprehensive study, we have demonstrated mussels as a suitable model marine invertebrate species to study the potential detrimental effects induced by possible genotoxicants and toxicants, either alone or in combinations at different levels of biological organisation (i.e. molecular to individual levels).


Assuntos
Bivalves/efeitos dos fármacos , Dano ao DNA , Fulerenos/toxicidade , Proteína Supressora de Tumor p53/efeitos dos fármacos , Proteínas ras/efeitos dos fármacos , Animais , Benzo(a)pireno/farmacologia , Benzo(a)pireno/toxicidade , Bivalves/genética , Bivalves/metabolismo , Ensaio Cometa , DNA/efeitos dos fármacos , Fulerenos/farmacologia , Regulação da Expressão Gênica , Glutationa/análise , Glutationa/efeitos dos fármacos , Modelos Animais , Especificidade de Órgãos , Estresse Oxidativo/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteínas ras/genética
9.
Curr Atheroscler Rep ; 17(2): 474, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25433554

RESUMO

Prostate cancer is the second most common cause of cancer and, when it metastasizes, has a high mortality rate. Statins may affect prostate cancer progression through cholesterol- and pleiotropic-mediated effects. The data on statin effects on prostate cancer has been mixed with benefit most likely occurring in reducing prostate cancer recurrence after radiation therapy and reduced mortality due to prostate cancer. More research is needed in this area to better characterize potential statin-mediated mechanisms that affect cancer. Also, future studies should report patients' anatomic/prognostic stage based on the updated staging system of the American Joint Committee on Cancer, which is a more effective predictor of recurrence and mortality than anatomic stage alone.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Neoplasias da Próstata/patologia , Humanos , Masculino , Microdomínios da Membrana/efeitos dos fármacos , Recidiva Local de Neoplasia , Prostatectomia , Proteínas ras/efeitos dos fármacos
10.
Am J Respir Crit Care Med ; 188(7): 770-5, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23841470

RESUMO

The remarkable success of epidermal growth factor receptor (EGFR) and anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitors in patients with EGFR mutations and ALK rearrangements, respectively, introduced the era of targeted therapy in advanced non-small cell lung cancer (NSCLC), shifting treatment from platinum-based combination chemotherapy to molecularly tailored therapy. Recent genomic studies in lung adenocarcinoma identified other potential therapeutic targets, including ROS1 rearrangements, RET fusions, MET amplification, and activating mutations in BRAF, HER2, and KRAS in frequencies exceeding 1%. Lung cancers that harbor these genomic changes can potentially be targeted with agents approved for other indications or under clinical development. The need to generate increasing amounts of genomic information should prompt health-care providers to be mindful of the amounts of tissue needed for these assays when planning diagnostic procedures. In this review, we summarize oncogenic drivers in NSCLC that can be currently detected, highlight their potential therapeutic implications, and discuss practical considerations for successful application of tumor genotyping in clinical decision making.


Assuntos
Adenocarcinoma/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Proteínas Oncogênicas/genética , Proteínas Proto-Oncogênicas B-raf/genética , Receptores Proteína Tirosina Quinases/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma de Pulmão , Quinase do Linfoma Anaplásico , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Receptores ErbB/uso terapêutico , Genômica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Mutação/efeitos dos fármacos , Mutação/genética , Proteínas Oncogênicas/efeitos dos fármacos , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas B-raf/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-met/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-ret/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-ret/genética , Proteínas Proto-Oncogênicas p21(ras) , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Receptores Proteína Tirosina Quinases/uso terapêutico , Receptor ErbB-2/efeitos dos fármacos , Receptor ErbB-2/genética , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética
11.
Ann Dermatol Venereol ; 141(11): 689-93, 2014 Nov.
Artigo em Francês | MEDLINE | ID: mdl-25442474

RESUMO

BACKGROUND: Acantholytic dyskeratosis under BRAF inhibitors are dermatological diseases rarely reported to date. PATIENTS AND METHODS: We report 2 cases of acantholytic dyskeratosis, reaching the trunk and the seborrheic zones, not itchy, appeared one month after the introduction of vemurafenib. The histological analysis was typical of a "Grover-like rash" for the 2 patients. DISCUSSION: The appearance of acantholytic dyskeratosis under vemurafenib, a BRAF inhibitor, seems related with a paradoxical activation of the MAP-kinases pathway and with a growth acceleration of lesions in which RAS mutations of keratinocytes. Theses dermatoses seem also to occur with dabrafenib. CONCLUSION: The patients treated by BRAF inhibitors (vemurafenib and dabrafenib) can present acantholytic dyskeratosis. The arisen of this mild dermatosis does not question, of course, the continuation of the treatment. These cutaneous manifestations can be managed with emollients.


Assuntos
Acantólise/induzido quimicamente , Antineoplásicos/efeitos adversos , Toxidermias/etiologia , Ictiose/induzido quimicamente , Indóis/efeitos adversos , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Sulfonamidas/efeitos adversos , Idoso , Exantema/induzido quimicamente , Feminino , Humanos , Imidazóis/efeitos adversos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/secundário , Pessoa de Meia-Idade , Mutação/efeitos dos fármacos , Mutação/genética , Oximas/efeitos adversos , Proteínas Proto-Oncogênicas B-raf/genética , Vemurafenib , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética
12.
Oncology ; 83(5): 241-7, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22948721

RESUMO

PURPOSE: This study investigated the impact of specific mutations in codon 12 of the Kirsten-ras (KRAS) gene on treatment efficacy in patients with metastatic colorectal cancer (mCRC). PATIENTS: Overall, 119 patients bearing a KRAS mutation in codon 12 were evaluated. All patients received cetuximab-based first-line chemotherapy within the Central European Cooperative Oncology Group (CECOG), AIO KRK-0104 or AIO KRK-0306 trials. RESULTS: Patients with KRAS codon 12 mutant mCRC showed a broad range of outcome when treated with cetuximab-based first-line regimens. Patients with tumors bearing a KRAS p.G12D mutation showed a strong trend to a more favorable outcome compared to other mutations (overall survival 23.3 vs. 14-18 months; hazard ratio 0.66, range 0.43-1.03). An interaction model illustrated that KRAS p.G12C was associated with unfavorable outcome when treated with oxaliplatin plus cetuximab. CONCLUSION: The present analysis suggests that KRAS codon 12 mutation may not represent a homogeneous entity in mCRC when treated with cetuximab-based first-line therapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Mutação , Proteínas Proto-Oncogênicas/genética , Proteínas ras/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Capecitabina , Cetuximab , Códon/efeitos dos fármacos , Neoplasias Colorretais/genética , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Esquema de Medicação , Feminino , Fluoruracila/administração & dosagem , Fluoruracila/análogos & derivados , Alemanha , Humanos , Irinotecano , Leucovorina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Estudos Multicêntricos como Assunto , Razão de Chances , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Prognóstico , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas p21(ras) , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento , Proteínas ras/efeitos dos fármacos
13.
Nat Chem Biol ; 6(6): 449-56, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20418879

RESUMO

Cycles of depalmitoylation and repalmitoylation critically control the steady-state localization and function of various peripheral membrane proteins, such as Ras proto-oncogene products. Interference with acylation using small molecules is a strategy to modulate cellular localization--and thereby unregulated signaling--caused by palmitoylated Ras proteins. We present the knowledge-based development and characterization of a potent inhibitor of acyl protein thioesterase 1 (APT1), a bona fide depalmitoylating enzyme that is, so far, poorly characterized in cells. The inhibitor, palmostatin B, perturbs the cellular acylation cycle at the level of depalmitoylation and thereby causes a loss of the precise steady-state localization of palmitoylated Ras. As a consequence, palmostatin B induces partial phenotypic reversion in oncogenic HRasG12V-transformed fibroblasts. We identify APT1 as one of the thioesterases in the acylation cycle and show that this protein is a cellular target of the inhibitor.


Assuntos
Inibidores Enzimáticos/farmacologia , Propiolactona/análogos & derivados , Tioléster Hidrolases/antagonistas & inibidores , Tioléster Hidrolases/química , Proteínas ras/fisiologia , Animais , Linhagem Celular , Cães , Regulação para Baixo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Humanos , Rim/efeitos dos fármacos , Rim/fisiologia , Ligantes , Lipase/química , Lipase/metabolismo , Lipoilação/efeitos dos fármacos , Modelos Moleculares , Propiolactona/síntese química , Propiolactona/química , Propiolactona/farmacologia , Conformação Proteica , Proto-Oncogene Mas , Transdução de Sinais , Estômago/enzimologia , Tioléster Hidrolases/genética , Proteínas ras/efeitos dos fármacos , Proteínas ras/metabolismo
14.
Dis Model Mech ; 15(2)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35244677

RESUMO

Several cancers and rare genetic diseases are caused by dysregulation in the RAS signaling pathway. RAS proteins serve as molecular switches that regulate pathways involved in cellular growth, differentiation and survival. These pathways have been an intense area of investigation for four decades, since the initial identification of somatic RAS mutations linked to human cancers. In the past few years, inhibitors against several RAS effectors, as well as direct inhibitors of the K-RAS mutant G12C, have been developed. This Special Issue in DMM includes original Research articles on RAS-driven cancers and RASopathies. The articles provide insights into mechanisms and biomarkers, and evaluate therapeutic targets. Several articles also present new disease models, whereas others describe technologies or approaches to evaluate the function of RAS in vivo. The collection also includes a series of Review articles on RAS biology and translational aspects of defining and treating RAS-driven diseases. In this Editorial, we summarize this collection and discuss the potential impact of the articles within this evolving area of research. We also identify areas of growth and possible future developments.


Assuntos
Neoplasias , Proteínas ras , Antineoplásicos/farmacologia , Humanos , Mutação/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/metabolismo , Processamento de Proteína Pós-Traducional , Transdução de Sinais/genética , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética , Proteínas ras/metabolismo
15.
Aging (Albany NY) ; 13(9): 13333-13348, 2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33982674

RESUMO

This study aimed to explore effects of Sevoflurane on ischemia-reperfusion (I/R) injury after total knee arthroplasty (TKA). To explore potential molecular mechanism, Ras related dexamethasone induced 1 (RASD1), a Protein kinase A (PKA) activator, frequently associated with various models of I/R injury, was also investigated. In vivo mouse models with I/R injury after TKA and in vitro cell models with I/R injury were induced. Contents of creatinine kinase (CK), lactic dehydrogenase (LDH), superoxide dismutase (SOD), and malondialdehyde (MDA), serum levels of inflammatory factors, expression of PKA pathway-related genes and cell proliferation and apoptosis were measured. RASD1 was altered and PKA pathway was inhibited in mice and cells to elucidate the involvement of RASD1 and PKA pathway in Sevoflurane treatment on I/R injury. RASD1 was upregulated in I/R injury after TKA. Sevoflurane treatment or silencing RASD1 reduced RASD1 expression, CK, LDH and MDA contents, inflammation, apoptosis, but increased proliferation, SOD content, cAMP expression, and extents of PKA and cAMP responsive element binding protein (CREB) phosphorylation in skeletal muscle cells of I/R injury. Additionally, PKA pathway activation potentiated the therapeutic effect of Sevoflurane on I/R injury after TKA. Altogether, Sevoflurane treatment confines I/R injury after TKA via RASD1-mediated PKA pathway activation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Sevoflurano/farmacologia , Proteínas ras/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Artroplastia do Joelho/métodos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Camundongos , Substâncias Protetoras/farmacologia , Traumatismo por Reperfusão/metabolismo
16.
Biomolecules ; 11(6)2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34208655

RESUMO

The RTK-RAS-MAPK axis is one of the most extensively studied signaling cascades and is related to the development of both cancers and RASopathies. In the last 30 years, many ideas and approaches have emerged for directly targeting constituent members of this cascade, predominantly in the context of cancer treatment. These approaches are still insufficient due to undesirable drug toxicity, resistance, and low efficacy. Significant advances have been made in understanding the spatiotemporal features of the constituent members of the RTK-RAS-MAPK axis, which are linked and modulated by many accessory proteins. Given that the majority of such modulators are now emerging as attractive therapeutic targets, a very small number of accessory inhibitors have yet to be discovered.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/efeitos dos fármacos , Receptores Proteína Tirosina Quinases/efeitos dos fármacos , Proteínas ras/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/metabolismo
17.
Br J Cancer ; 102(7): 1137-44, 2010 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-20234366

RESUMO

BACKGROUND: The validation of KRAS mutations as a negative marker of response to anti-epidermal growth factor receptor (EGFR) antibodies has meant a seminal advance towards treatment individualisation of colorectal cancer (CRC) patients. However, as a KRAS wild-type status does not guarantee a response to anti-EGFR antibodies, a current challenge is the identification of other biomarkers of response. On the basis of pre-clinical evidence, we hypothesised that mitogen-activated protein kinase phosphatase-1 (MKP-1), a phosphatase that inactivates MAPKs, could be a mediator of resistance to anti-EGFR antibodies. METHODS: Tumour specimens from 48 metastatic CRC patients treated with cetuximab-based chemotherapy were evaluated for KRAS and BRAF mutational status and MKP-1 expression as assessed by immunohistochemistry. RESULTS: As expected, clinical benefit was confined to wild-type KRAS and BRAF patients. Mitogen-activated protein kinase phosphatase-1 was overexpressed in 16 patients (33%) and was not associated with patient baseline clinicopathological characteristics and KRAS mutational status. All patients with BRAF mutations (n=3) had MKP-1 overexpression. Among KRAS wild-type patients, MKP-1 overexpressors had a 7% response rate (RR), whereas patients not overexpressing MKP-1 had a 44% RR (P=0.03). Moreover, median time to progression was significantly longer in MKP-1 non-overexpressing patients (32 vs 13 weeks, P=0.009). CONCLUSION: These results support the concept of MKP-1 as a promising negative marker of response to cetuximab-based treatment in CRC patients with wild-type KRAS.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Fosfatase 1 de Especificidade Dupla/metabolismo , Idoso , Anticorpos Monoclonais Humanizados , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Cetuximab , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/antagonistas & inibidores , Feminino , Humanos , Masculino , Mutação , Metástase Neoplásica , Proteínas Proto-Oncogênicas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras) , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética , Proteínas ras/metabolismo
18.
ACS Chem Biol ; 15(6): 1604-1612, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32378881

RESUMO

Protein-protein interactions (PPIs) play a critical role in fundamental biological processes. Competitive inhibition of these interfaces requires compounds that can access discontinuous binding epitopes along a large, shallow binding surface area. Conformationally defined protein surface mimics present a viable route to target these interactions. However, the development of minimal protein mimics that engage intracellular targets with high affinity remains a major challenge because mimicry of a portion of the binding interface is often associated with the loss of critical binding interactions. Covalent targeting provides an attractive approach to overcome the loss of noncovalent contacts but have the inherent risk of dominating noncovalent contacts and increasing the likelihood of nonselective binding. Here, we report the iterative design of a proteolytically stable α3ß chimeric helix mimic that covalently targets oncogenic Ras G12C as a model system. We explored several electrophiles to optimize preferential alkylation with the desired C12 on Ras. The designed lead peptide modulates nucleotide exchange, inhibits activation of the Ras-mediated signaling cascade, and is selectively toxic toward mutant Ras G12C cancer cells. The relatively high frequency of acquired cysteines as missense mutations in cancer and other diseases suggests that covalent peptides may offer an untapped therapeutic approach for targeting aberrant protein interactions.


Assuntos
Sistemas de Liberação de Medicamentos , Desenho de Fármacos , Peptidomiméticos/farmacologia , Proteínas ras/efeitos dos fármacos , Fenômenos Biofísicos , Linhagem Celular Tumoral , Humanos , Ligantes , Peptidomiméticos/química , Conformação Proteica , Mapas de Interação de Proteínas , Proteólise , Transdução de Sinais
19.
Folia Neuropathol ; 58(1): 45-56, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32337957

RESUMO

Trauma is the main cause of death for people aged 1-45, and among them, traumatic brain injury (TBI) is the major condition, which causes over 50,000 deaths each year and costs over 80 billion per year. Tetrahydroxystilbene glucoside (TSG) is the active ingredient of polygonum multiflorum, a traditional Chinese herbal medicine, which presented multiple pharmacological effects, including antioxidative, anti-inflammatory, reducing blood fat and neuroprotection effects. However, the effect of TSG in promoting the recovery of the nerve system after TBI is not fully understood. PARP1 is a key enzyme in repair of the damage in DNA, which is activated by binding to DNA breaks, initiating both single-strand and double-strand DNA break repair. And we thought that overexpression of TSG might enhance the effect of TSG in TBI treatment. In this study, we firstly detected the oxidative stress response related molecules in serum samples of TBI patients and a TBI mice model, and found that oxidative stress response was activated after TBI, and TSG would reduce this effect. We further noticed that inflammation related molecules presented a similar trend with oxidative stress response related molecules. These results indicated that inflammatory response and oxidative stress processes were both activated after TBI, and reduced after TSG treatment. We further detected that the apoptosis related proteins and anti-oxidative proteins were increased after TSG treatment, and these effects were enlarged after overexpression of PARP1. We further noticed that these effects might be mediated by inhibition of the Ras/JNK signalling pathway. Thus, we thought overexpression of PARP1 might enhance the therapeutic effect of TSG in TBI treatment.


Assuntos
Lesões Encefálicas Traumáticas/patologia , Glucosídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/metabolismo , Estilbenos/farmacologia , Proteínas ras/efeitos dos fármacos , Adulto , Animais , Lesões Encefálicas Traumáticas/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/fisiologia , Espécies Reativas de Oxigênio/sangue , Proteínas ras/metabolismo
20.
Clin Epigenetics ; 12(1): 76, 2020 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-32493461

RESUMO

BACKGROUND: A long-term opioid use has been associated with hypermethylation of the opioid receptor mu 1 (OPRM1) promoter. Very little is currently known about the early epigenetic response to therapeutic opioids. Here, we examine whether we can detect DNA methylation changes associated with a few days' use of prescribed opioids. Genome-wide DNA methylation was assayed in a cohort of 33 opioid-naïve participants who underwent standard dental surgery followed by opioid self-administration. Saliva samples were collected before surgery (visit 1), and at two postsurgery visits at 2.7 ± 1.5 days (visit 2), and 39 ± 10 days (visit 3) after the discontinuation of opioid analgesics. RESULTS: The perioperative methylome underwent significant changes over the three visits that were primarily due to postoperative inflammatory response and cell heterogeneity. To specifically examine the effect of opioids, we started with a candidate gene approach and evaluated 10 CpGs located in the OPRM1 promoter. There was a significant cross-sectional variability in opioid use, and for participants who self-administered the prescribed drugs, the total dosage ranged from 5-210 morphine milligram equivalent (MME). Participants were categorized by cumulative dosage into three groups: < 25 MME, 25-90 MME, and ≥ 90 MME. Using mixed-effects modeling, 4 CpGs had significant positive associations with opioid dose at two-tailed p value < 0.05, and overall, 9 of the 10 OPRM1 promoter CpGs showed the predicted higher methylation in the higher dose groups relative to the lowest dose group. After adjustment for age, cellular heterogeneity, and past tobacco use, the promoter mean methylation also had positive associations with cumulative MME (regression coefficient = 0.0002, one-tailed p value = 0.02) and duration of opioid use (regression coefficient = 0.003, one-tailed p value = 0.001), but this effect was significant only for visit 3. A preliminary epigenome-wide association study identified a significant CpG in the promoter of the RAS-related signaling gene, RASL10A, that may be predictive of opioid dosage. CONCLUSION: The present study provides evidence that the hypermethylation of the OPRM1 promoter is in response to opioid use and that epigenetic differences in OPRM1 and other sites are associated with a short-term use of therapeutic opioids.


Assuntos
Analgésicos Opioides/farmacologia , Metilação de DNA/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/genética , Regiões Promotoras Genéticas/efeitos dos fármacos , Receptores Opioides mu/efeitos dos fármacos , Adulto , Analgésicos Opioides/administração & dosagem , Estudos de Casos e Controles , Ilhas de CpG/genética , Epigênese Genética , Epigenoma/efeitos dos fármacos , Epigenoma/genética , Feminino , Estudo de Associação Genômica Ampla/métodos , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Relacionados ao Uso de Opioides/metabolismo , Período Perioperatório , Variantes Farmacogenômicos/genética , Regiões Promotoras Genéticas/genética , Receptores Opioides mu/metabolismo , Saliva/metabolismo , Proteínas ras/efeitos dos fármacos , Proteínas ras/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa