Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
FASEB J ; 36(8): e22398, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35792869

RESUMO

Conjugated bile acids (CBAs) play major roles in hepatic gene regulation via nuclear S1P-inhibited histone deacetylase (HDACs). Gut microbiota modifies bile acid pool to generate CBAs and then CBAs returned to liver to regulate hepatic genes, fatty liver, and non-alcoholic fatty liver disease (NAFLD). However, it is not yet known how the gut microbiota was modified under the environment of inflammatory bowel disease (IBD). Here, we revealed that aberrant intestinal sphingosine kinases (SphKs), a major risk factor of IBD, modified gut microbiota by increasing the proportions of Firmicutes and Verrucomicrobia, which were associated with the increase in CBAs. When exposed to a high-fat diet (HFD), sphingosine kinases 2 knockout (SphK2KO) mice developed more severity of intestinal inflammation and hepatic steatosis than their wild-type (WT) littermates. Due to knockdown of nuclear SphK2, Sphk2KO mice exhibited an increase in sphingosine kinases 1 (SphK1) and sphingosine-1-phosphate (S1P) in intestinal epithelial cells. Therefore, the microbiota was modified in the environment of the SphK1/S1P-induced IBD. 16S rDNA amplicon sequencing of cecal contents indicated an increase of Firmicutes and Verrucomicrobia. Ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS) measured an increase in CBAs, including taurocholic acid (TCA), taurodeoxycholic acid (TDCA), and glycocholic acid (GCA), in cecal contents and liver tissues of Sphk2KO mice. These CBAs accumulated in the liver promoted hepatic steatosis through downregulating the acetylation of H3K9, H3K14, H3K18 and H3K27 due to the CBAs-S1PR2-nuclear SphK2-S1P signaling pathway was blocked in HFD-SphK2KO mice. In summary, intestinal aberrant sphingolipid metabolism developed hepatic steatosis through the increase in CBAs associated with an increase in Firmicutes and Verrucomicrobia.


Assuntos
Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , Hepatopatia Gordurosa não Alcoólica , Animais , Ácidos e Sais Biliares , Cromatografia Líquida , Firmicutes , Metaboloma , Camundongos , Esfingolipídeos , Esfingosina , Espectrometria de Massas em Tandem , Verrucomicrobia
2.
Bioorg Chem ; 131: 106318, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36527992

RESUMO

Targeting sphingosine-1-phosphate receptor 2 (S1PR2) has been proved as a promising strategy to reverse 5-fluorouracil (5-FU) resistance. Here, we report the discovery of the novel JTE-013 derivative compound 37 h as a more effective S1PR2 antagonist to reverse 5-FU resistance in SW620/5-FU and HCT116DPD cells than JTE-013 and previously reported compound 5. Compound 37 h could effectively bind S1PR2 and reduce its expression, thus leading to decreased expression of JMJD3 and dihydropyrimidine dehydrogenase (DPD), while also increasing the level of H3K27me3 to decrease the degradation of 5-FU and thereby increase its intracellular concentration in SW620/5-FU, HCT116DPD, and L02 cells. Furthermore, compound 37 h showed good selectivity to other S1PRs and normal colon cell line NCM460. Western blot analysis demonstrated that compound 37 h could abrogate the FBAL-stimulated upregulation of DPD expression by S1PR2. Importantly, compound 37 h also showed favorable metabolic stability with a long half-life (t1/2) of 7.9 h. Moreover, compound 37 h significantly enhanced the antitumor efficacy of 5-FU in the SW620/5-FU animal model. Thus, the JTE-013-based derivative compound 37 h represents a promising lead compound for the development of novel 5-FU sensitizers for colorectal cancer (CRC) therapy.


Assuntos
Neoplasias Colorretais , Fluoruracila , Animais , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Receptores de Esfingosina-1-Fosfato , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos , Di-Hidrouracila Desidrogenase (NADP)/metabolismo
3.
Acta Pharmacol Sin ; 43(11): 2967-2976, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35365782

RESUMO

Atypical chemokine receptor 3 (ACKR3) has emerged as a key player in various biological processes. Its atypical "intercepting receptor" properties have established ACKR3 as the major regulator in the pathophysiological processes in many diseases. In this study, we investigated the role of ACKR3 activation in promoting colorectal tumorigenesis. We showed that ACKR3 expression levels were significantly increased in human colon cancer tissues, and high levels of ACKR3 predicted the increased severity of cancer. In Villin-ACKR3 transgenic mice with a high expression level of CKR3 in their intestinal epithelial cells, administration of AOM/DSS induced more severe colorectal tumorigenesis than their WT littermates. Cancer cells of Villin-ACKR3 transgenic mice were characterised by the nuclear ß-arrestin-1 (ß-arr1)-activated perturbation of rRNA biogenesis. In HCT116 cells, cotreatment with CXCL12 and AMD3100 selectively activated ACKR3 and induced nuclear translocation of ß-arr1, leading to an interaction of ß-arr1 with nucleolar and coiled-body phosphoprotein 1 (NOLC1). NOLC1, as the phosphorylated protein, further interacted with fibrillarin, a conserved nucleolar methyltransferase responsible for ribosomal RNA methylation in the nucleolus, thereby increasing the methylation in histone H2A and promoting rRNA transcription in ribosome biogenesis. In conclusion, ACKR3 promotes colorectal tumorigenesis through the perturbation of rRNA biogenesis by the ß-arr1-induced interaction of NOLC1 with fibrillarin.


Assuntos
Transformação Celular Neoplásica , Neoplasias Colorretais , Receptores CXCR , Animais , Humanos , Camundongos , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo , Carcinogênese/genética , Transformação Celular Neoplásica/genética , Quimiocina CXCL12 , Neoplasias Colorretais/genética , Camundongos Transgênicos , Proteínas Nucleares/genética , Fosfoproteínas/metabolismo , Receptores CXCR/metabolismo
4.
Acta Pharmacol Sin ; 42(3): 460-469, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32647340

RESUMO

Sphingosine-1-phosphate (S1P), the backbone of most sphingolipids, activating S1P receptors (S1PRs) and the downstream G protein signaling has been implicated in chemoresistance. In this study we investigated the role of S1PR2 internalization in 5-fluorouracil (5-FU) resistance in human colorectal cancer (CRC). Clinical data of randomly selected 60 CRC specimens showed the correlation between S1PR2 internalization and increased intracellular uracil (P < 0.001). Then we explored the regulatory mechanisms in CRC model of villin-S1PR2-/- mice and CRC cell lines. We showed that co-administration of S1P promoted S1PR2 internalization from plasma membrane (PM) to endoplasmic reticulum (ER), thus blunted 5-FU efficacy against colorectal tumors in WT mice, compared to that in S1PR2-/- mice. In HCT116 and HT-29 cells, application of S1P (10 µM) empowered S1PR2 to internalize from PM to ER, thus inducing 5-FU resistance, whereas the specific S1PR2 inhibitor JTE-013 (10 µM) effectively inhibited S1P-induced S1PR2 internalization. Using Mag-Fluo-AM-labeling [Ca2+]ER and LC-ESI-MS/MS, we revealed that internalized S1PR2 triggered elevating [Ca2+]ER levels to activate PERK-eLF2α-ATF4 signaling in HCT116 cells. The activated ATF4 upregulated RNASET2-mediated uracil generation, which impaired exogenous 5-FU uptake to blunt 5-FU therapy. Overall, this study reveals a previously unrecognized mechanism of 5-FU resistance resulted from S1PR2 internalization-upregulated uracil generation in colorectal cancer, and provides the novel insight into the significance of S1PR2 localization in predicting the benefit of CRC patients from 5-FU-based chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Fluoruracila/uso terapêutico , Lisofosfolipídeos/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Uracila/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Animais , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/fisiologia , Retículo Endoplasmático/metabolismo , Feminino , Células HCT116 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Ribonucleases/metabolismo , Transdução de Sinais/fisiologia , Esfingosina/metabolismo , Proteínas Supressoras de Tumor/metabolismo
5.
Carcinogenesis ; 39(7): 889-899, 2018 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-29757351

RESUMO

Chronic gut inflammation disposes to an increased risk of colitis-associated cancer. Chemoprevention is an attractive complementary strategy. We aimed to evaluate the chemopreventive effects of M10, a novel derivative of Myricetin, in the murine azoxymethane/dextran sodium sulfate model. Oral administration of M10 at 50-100 mg/kg once a day for consecutive 12 weeks significantly prevented ulcerative colitis (UC) and colorectal tumor. Pathological analysis of intestines showed that M10 reduced the degree of chronic inflammation and prevented the progression of colorectal tumorigenesis. Flow cytometry analysis of the immunocytes isolated from intraepithelial and lamina propria showed that M10 prevented the infiltration of myeloid-derived suppressor cells and increased CD8+T and CD4+T cells in colorectal tissues. Enzyme-linked immunosorbent analysis revealed the reduction of pro-inflammatory mediators granulocyte-macrophage colony-stimulating factor/macrophage colony-stimulating factor, IL-6 and TNF-α in colonic mucosa. Western blot assay also showed M10 prevention of the NF-κB/IL-6/STAT3 pathways and the biomarkers of inflammation and colorectal tumorigenesis. Electron microscopy analysis revealed that M10 prevent robust endoplasmic reticulum (ER) stress-induced autophagy in inflamed colonic mucosal cells. In conclusion, oral administration of Myricetin derivative M10 exerts chemoprevention of UC and colorectal tumor in mice. The mechanism of chemoprevention is associated with the reduction of biomarkers of chronic inflammation and proliferation through attenuating robust ER stress in inflamed colonic mucosal cells. M10 exerts chemoprevention activity without evidence of toxicity in mice. These results justify further evaluation of M10 in clinical trials. M10 could develop a promising regimen in the chemoprevention of colitis and colorectal cancer.


Assuntos
Alanina/análogos & derivados , Colite Ulcerativa/prevenção & controle , Neoplasias Colorretais/prevenção & controle , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Flavonoides/farmacologia , Hidroxiquinolinas/farmacologia , Alanina/farmacologia , Animais , Anticarcinógenos/farmacologia , Colo/efeitos dos fármacos , Colo/metabolismo , Neoplasias Colorretais/metabolismo , Modelos Animais de Doenças , Inflamação/metabolismo , Inflamação/prevenção & controle , Mediadores da Inflamação/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
6.
Cancer Invest ; 36(1): 10-18, 2018 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-29381400

RESUMO

CXCL12 is an extracellular chemokine binding to cell surface receptor CXCR4. We found that activation of CXCL12/CXCR4 axis stimulated angiogenesis in endothelial cells. Knockdown of CXCR4 in endothelial cells prevented the branch points of angiogenesis. Endothelial cells exposed to CXCL12 presented high level of epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF), and matrix metalloproteinase MMP-2, but not in CXCR4 knockdown cells. Further studies revealed that activation of CXCL12/CXCR4 axis in vascular endothelial cells stimulates the angiogenesis through upregulation of the MAPK/ERK and PI3K/AKT and Wnt/ß-catenin pathways. Conclusion, downregulation of CXCR4 could inhibit angiogenesis in cancer tissues.


Assuntos
Quimiocina CXCL12/genética , Regulação para Baixo/genética , Regulação Neoplásica da Expressão Gênica/genética , Neovascularização Patológica/genética , Receptores CXCR4/genética , Transdução de Sinais/genética , Linhagem Celular , Células Endoteliais/metabolismo , Receptores ErbB/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Sistema de Sinalização das MAP Quinases/genética , Metaloproteinase 2 da Matriz/genética , Proteínas Quinases Ativadas por Mitógeno/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Regulação para Cima/genética , Fator A de Crescimento do Endotélio Vascular/genética , Via de Sinalização Wnt/genética , beta Catenina/genética
7.
Cancer Invest ; 34(9): 459-464, 2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27673353

RESUMO

Des-γ-carboxyprothrombin (DCP), an abnormal prothrombin produced in human hepatocellular carcinoma (HCC), plays crucial roles in the progression of HCC. DCP binding to cellular mesenchymal-epithelial transition factor (c-Met) is an initial event and consequently stimulates HCC through the increase of c-Met-Janus kinase 1- signal transducers and activators of transcription pathways. DCP stimulates HCC invasion through activation of matrix metalloproteinase via upregulation of extracellular signal-regulated kinase-mitogen-activated protein kinase (MAPK) pathway. DCP stimulates HCC angiogenesis through activation of the DCP-kinase insert domain receptor-phospholipaseC-γ-MAPK pathway. Identification of these pathways is important for designing the therapeutic strategy for HCC.


Assuntos
Biomarcadores/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Precursores de Proteínas/metabolismo , Protrombina/metabolismo , Transdução de Sinais , Progressão da Doença , Humanos , Janus Quinase 1/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Metástase Neoplásica , Neovascularização Patológica , Fosfolipase C gama/metabolismo , Ligação Proteica , Precursores de Proteínas/biossíntese , Protrombina/biossíntese , Proteínas Proto-Oncogênicas c-met/metabolismo , Fator de Transcrição STAT3/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
8.
Cell Physiol Biochem ; 35(1): 201-12, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25591763

RESUMO

BACKGROUND/AIMS: Des-gamma-carboxy prothrombin (DCP), an aberrant prothrombin produced by hepatocellular carcinoma (HCC) cells, is known as a marker for HCC. Recent studies indicated that high levels of DCP are associated with the malignant potential of HCC. In this study, we aimed to investigate the association of DCP with gefitinib treatment failure in HCC and whether DCP counteracts gefitinib-induced growth inhibition and apoptosis of HCC. METHODS: The experiments were performed in HCC cell lines HepG2 and PLC/PRF/5. The effects of gefitinib on HCC in the presence or absence of DCP were evaluated by the 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyl-tetrazolium bromide (MTT) assay. Apoptotic cells were identified by Annexin V-FITC/PI staining. Western blotting was performed to analyze the expressions of molecules related to the apoptotic caspase-dependent pathway and epidermal growth factor receptor (EGFR) pathway. RESULTS: Gefitinib inhibited HCC cell proliferation and induced apoptosis in HCC cells. The effects of gefitinib on HCC cells were antagonized by DCP. In the presence of DCP, HCC cells were resistant to the gefitinib-induced inhibition of proliferation and stimulation of apoptosis. DCP prevented the activation of the apoptotic caspase-dependent pathway induced by gefitinib. These antagonistic effects of DCP also arose from its ability to up-regulate EGFR, c-Met and hepatocyte growth factor (HGF) in HCC cells. CONCLUSION: DCP antagonized gefitinib-induced HCC cell growth inhibition by counteracting apoptosis and up-regulating the EGFR pathway. High levels of DCP might thus lead to low response rates or possibly no response to gefitinib in patients with HCC.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Precursores de Proteínas/farmacologia , Protrombina/farmacologia , Quinazolinas/farmacologia , Biomarcadores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Receptores ErbB/metabolismo , Gefitinibe , Células Hep G2 , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas Proto-Oncogênicas c-met/metabolismo , Regulação para Cima/efeitos dos fármacos
9.
Biochim Biophys Acta ; 1830(10): 4907-16, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23850473

RESUMO

BACKGROUND: Acetyl-11-keto-beta-boswellic acid (AKBA) is a derivative of boswellic acid. We have previously reported that AKBA can reduce the number and size of colonic adenomatous polyps in the APC(Min/+) mouse model. In this study, we evaluated the effect of AKBA on human colonic adenocarcinoma growth. Its efficacy and toxicity were compared with those of the non-steroidal anti-inflammatory drug aspirin. METHODS: The inhibition of cancer cell growth was estimated by colorimetric and clonogenic assay. Cell cycle distribution was analyzed by the flow cytometry assay. Annexin V-FITC/PI staining and JC-1 fluorescence probe assays were performed to determine the apoptotic cells. Further experiment was carried out in mice with HT-29 xenografts. AKBA was orally administered for 24days. The HT-29 xenografts were removed for TUNEL staining and western blotting analysis. Blood was obtained for clinical chemical analysis, and samples of organs were sectioned for microscopic assessment. RESULTS: AKBA significantly inhibited human colon adenocarcinoma growth, showing arrest of the cell cycle in G1-phase and induction of apoptosis. AKBA administration in mice effectively delayed the growth of HT-29 xenografts without signs of toxicity. The activity of AKBA was more potent than that of aspirin. Western blotting suggested that this activity may arise from its multiple effects on the activation of apoptotic proteins, suppression of inflammatory cytokines and modulation of EGFR and ATM/P53 signaling pathways in the HT-29 xenografts. CONCLUSIONS: AKBA prevents the growth of colonic adenocarcinoma through modulation of multiple signaling pathways. GENERAL SIGNIFICANCE: AKBA could be a promising agent in the prevention of colonic adenocarcinomas.


Assuntos
Adenocarcinoma/patologia , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/patologia , Transdução de Sinais/efeitos dos fármacos , Triterpenos/farmacologia , Adenocarcinoma/metabolismo , Animais , Apoptose/efeitos dos fármacos , Neoplasias do Colo/metabolismo , Receptores ErbB/metabolismo , Células HT29 , Humanos , Marcação In Situ das Extremidades Cortadas , Camundongos , Triterpenos/toxicidade
10.
Cell Physiol Biochem ; 34(3): 903-15, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25200250

RESUMO

Des-γ-carboxy prothrombin (DCP) is a prothrombin precursor produced in hepatocellular carcinoma (HCC). Because of deficiency of vitamin K or γ-glutamyl carboxylase in HCC cells, the 10 glutamic acid (Glu) residues in prothrombin precursor did not completely carboxylate to γ-carboxylated glutamic acid (Gla) residues, leaving some Glu residues remained in N-terminal domain. These prothrombin precursors with Glu residues are called DCPs. DCP displays insufficient coagulation activity. Since Liebman reported an elevated plasma DCP in patients with HCC, DCP has been used in the diagnosis of HCC. Recently, its biological malignant potential has been specified to describe DCP as an autologous growth factor to stimulate HCC growth and a paracrine factor to integrate HCC with vascular endothelial cells. DCP was found to stimulate HCC growth through activation of the DCP-Met-JAK1-STAT3 signaling pathway. DCP might increase HCC invasion and metastasis through activation of matrix metalloproteinase (MMPs) and the ERK1/2 MAPK signaling pathway. DCP has also been found to play a crucial role in the formation of angiogenesis. DCP could increase the angiogenic factors released from HCC and vascular endothelial cells. These effects of DCP in angiogenesis might be related to activation of the DCP-KDR-PLC-γ-MAPK signaling pathway. In this article, we summarized recent studies on DCP in biological roles related to cancer progression and angiogenesis in HCC.


Assuntos
Carcinoma Hepatocelular/fisiopatologia , Substâncias de Crescimento/fisiologia , Neoplasias Hepáticas/fisiopatologia , Precursores de Proteínas/fisiologia , Protrombina/fisiologia , Biomarcadores/química , Biomarcadores Tumorais , Carcinoma Hepatocelular/diagnóstico , Humanos , Neoplasias Hepáticas/diagnóstico , Estrutura Molecular , Precursores de Proteínas/química , Protrombina/química
11.
Int J Cancer ; 132(11): 2667-81, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23132636

RESUMO

Acetyl-11-keto-beta-boswellic acid (AKBA) is a derivative of boswellic acid, which is an active component of the gum resin of Boswellia serrata. AKBA has been used as an adjuvant medication for treatment of inflammatory diseases. In this study, we aimed to evaluate the efficacy of AKBA as a chemopreventive agent against intestinal adenomatous polyposis in the adenomatous polyposis coli multiple intestinal neoplasia (APC(Min/+) ) mouse model. APC(Min/+) mice were administered AKBA by p.o. gavage for 8 consecutive weeks. The mice were sacrificed and the number, size and histopathology of intestinal polyps were examined by light microscopy. AKBA decreased polyp numbers by 48.9% in the small intestine and 60.4% in the colon. An even greater AKBA effect was observed in preventing the malignant progression of these polyps. The number of large (>3 cm) colonic polyposis was reduced by 77.8%. Histopathologic analysis demonstrated a significant reduction in the number of dysplastic cells and in the degree of dysplasia in each polyp after AKBA treatment. There was no evidence of high grade dysplasia or intramucosal carcinoma in any of the polyps examined within the treated group. More interestingly, interdigitated normal appearing intestinal villi were observed in the polyps of the treated group. During the course of the study, AKBA was well tolerated by the mice with no obvious signs of toxicity. Results from immunohistochemical staining, Western blotting and enzyme-linked immunosorbent assay indicated that the chemopreventive effect of AKBA was attributed to a collection of activities including antiproliferation, apoptosis induction, antiangiogenesis and anti-inflammation. AKBA was found to exert its chemopreventive action through the inhibition of the Wnt/ß-catenin and NF-κB/cyclooxygenase-2 signaling pathways. Our findings suggest that AKBA could be a promising regimen in chemoprevention against intestinal tumorigenesis.


Assuntos
Adenoma/prevenção & controle , Proteína da Polipose Adenomatosa do Colo/fisiologia , Polipose Adenomatosa do Colo/prevenção & controle , Apoptose/efeitos dos fármacos , Polipose Intestinal/prevenção & controle , Neovascularização Patológica/prevenção & controle , Triterpenos/uso terapêutico , Adenoma/genética , Adenoma/patologia , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Animais , Western Blotting , Boswellia/química , Proliferação de Células , Ciclo-Oxigenase 2/química , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Técnicas Imunoenzimáticas , Mediadores da Inflamação/metabolismo , Polipose Intestinal/genética , Polipose Intestinal/patologia , Leucotrieno B4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Transdução de Sinais , beta Catenina/metabolismo
12.
Biomed Chromatogr ; 27(6): 802-6, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23303567

RESUMO

Brazilin is a major homoisoflavonoid component isolated from the dried heartwood of traditional Chinese medicine Caesalpinia sappan L., which is a natural red pigment used for histological staining. Herein a sensitive, specific and rapid analytical LC-MS/MS method was established and validated for brazilin in rat plasma. After a simple step of protein precipitation using acetonitrile, plasma samples were analyzed using an LC-MS/MS system. Brazilin and the IS (protosappanin B) were separated on a Diamonsil C18 analytical column (150 × 4.6 mm, 5 µm) using a mixture of water and 10 mm ammonium acetate in methanol (20:80, v/v) as mobile phase at a flow rate of 0.6 mL/min. The method was sensitive with a lower limit of quantitation of 10.0 ng/mL, with good linearity (r(2) ≥ 0.99) over the linear range 10.0-5000 ng/mL. All the validation data, such as accuracy and precision, matrix effect, extraction recovery and stability tests were within the required limits. The assay method was successfully applied to evaluate the pharmacokinetics parameters of brazilin after an oral dose of 100 mg/kg brazilin in rats.


Assuntos
Benzopiranos/sangue , Benzopiranos/farmacocinética , Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas em Tandem/métodos , Animais , Benzopiranos/química , Estabilidade de Medicamentos , Masculino , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
13.
Eur J Med Chem ; 260: 115763, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37659196

RESUMO

ProTide prodrug technology has emerged as a promising way for the development of anti-viral and anti-tumor drugs, whereas, there are fewer applications for the treatment of liver cancer. Herein, a series of distinct 3'-ester ProTide prodrugs of 5-fluoro-2'-deoxyuridine (FdUR) were synthesized and evaluated for their anti-liver cancer activity. The most efficient prodrug 11b reached a sub-micromolar activity (IC50 = 0.42 ± 0.13 µM) against HepG2 and over 100-fold and 200-fold improvements compared to 5-FU, respectively. 11b also demonstrated favorable selectivity towards normal liver cells L-02 (IC50 > 100 µM). In vitro metabolic stability studies revealed that 11b is stable in the plasma and could be activated rapidly in the liver, which supported that 11b is liver-targeted. Importantly, to more accurately evaluate the anti-HCC activity of 11b, the liver orthotopic model was built and 11b significantly suppressed tumor growth (TGI = 75.5%) at a dose of 60 mg/kg/2d in vivo without obvious toxicity. Overall, these promising results indicated that 11b could serve as a safe and effective prodrug of 5-FU nucleoside for liver cancer therapy.


Assuntos
Neoplasias Hepáticas , Pró-Fármacos , Humanos , Pró-Fármacos/farmacologia , Desoxiuridina/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico
14.
Acta Pharm Sin B ; 13(7): 2963-2975, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37521868

RESUMO

Insulin-like growth factor-1 receptor (IGF-1R) has been made an attractive anticancer target due to its overexpression in cancers. However, targeting it has often produced the disappointing results as the role played by cross talk with numerous downstream signalings. Here, we report a disobliging IGF-1R signaling which promotes growth of cancer through triggering the E3 ubiquitin ligase MEX3A-mediated degradation of RIG-I. The active ß-arrestin-2 scaffolds this disobliging signaling to talk with MEX3A. In response to ligands, IGF-1Rß activated the basal ßarr2 into its active state by phosphorylating the interdomain domain on Tyr64 and Tyr250, opening the middle loop (Leu130‒Cys141) to the RING domain of MEX3A through the conformational changes of ßarr2. The models of ßarr2/IGF-1Rß and ßarr2/MEX3A could interpret the mechanism of the activated-IGF-1R in triggering degradation of RIG-I. The assay of the mutants ßarr2Y64A and ßarr2Y250A further confirmed the role of these two Tyr residues of the interlobe in mediating the talk between IGF-1Rß and the RING domain of MEX3A. The truncated-ßarr2 and the peptide ATQAIRIF, which mimicked the RING domain of MEX3A could prevent the formation of ßarr2/IGF-1Rß and ßarr2/MEX3A complexes, thus blocking the IGF-1R-triggered RIG-I degradation. Degradation of RIG-I resulted in the suppression of the IFN-I-associated immune cells in the TME due to the blockade of the RIG-I-MAVS-IFN-I pathway. Poly(I:C) could reverse anti-PD-L1 insensitivity by recovery of RIG-I. In summary, we revealed a disobliging IGF-1R signaling by which IGF-1Rß promoted cancer growth through triggering the MEX3A-mediated degradation of RIG-I.

15.
Acta Pharm Sin B ; 13(9): 3744-3755, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37719369

RESUMO

The well-known insulin-like growth factor 1 (IGF1)/IGF-1 receptor (IGF-1R) signaling pathway is overexpressed in many tumors, and is thus an attractive target for cancer treatment. However, results have often been disappointing due to crosstalk with other signals. Here, we report that IGF-1R signaling stimulates the growth of hepatocellular carcinoma (HCC) cells through the translocation of IGF-1R into the ER to enhance sarco-endoplasmic reticulum calcium ATPase 2 (SERCA2) activity. In response to ligand binding, IGF-1Rß is translocated into the ER by ß-arrestin2 (ß-arr2). Mass spectrometry analysis identified SERCA2 as a target of ER IGF-1Rß. SERCA2 activity is heavily dependent on the increase in ER IGF-1Rß levels. ER IGF-1Rß phosphorylates SERCA2 on Tyr990 to enhance its activity. Mutation of SERCA2-Tyr990 disrupted the interaction of ER IGF-1Rß with SERCA2, and therefore ER IGF-1Rß failed to promote SERCA2 activity. The enhancement of SERCA2 activity triggered Ca2+ER perturbation, leading to an increase in autophagy. Thapsigargin blocked the interaction between SERCA2 and ER IGF-1Rß and therefore SERCA2 activity, resulting in inhibition of HCC growth. In conclusion, the translocation of IGF-1R into the ER triggers Ca2+ER perturbation by enhancing SERCA2 activity through phosphorylating Tyr990 in HCC.

16.
Biomed Chromatogr ; 26(7): 857-62, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22860257

RESUMO

A sensitive, rapid and specific LC-MS-MS method was established and validated for determination of methyl kulonate, a major bioactive constituent isolated from Meliae Cortex, in rat plasma. Plasma samples were treated by precipitating protein with methanol and were chromatographed using a Capcell Pak C18 column (100 x 4.6 mm, 5 µm) with the mobile phase comprising a mixture of methanol, 10 mM ammonium formate and formic acid (95:5:0.1, v/v/v). Detection and quantification were performed by mass spectrometry in the multiple reaction monitoring mode with positive atmospheric ionization at m/z 467 --> 311 for methyl kulonate, and m/z 469 --> 451 for dubione B (internal standard), respectively. A good linear response was observed over the concentration range 1.00-500 ng/mL with the lower limit of quantification 1.00 ng/mL in rat plasma. The method also afforded satisfactory results base on sensitivity, specificity, precision, accuracy, recovery, freeze-thaw and long-time stability. The validated method was successfully applied to determine the pharmacokinetic properties of methyl kulonate in rats after oral administration at dose of 100 mg/kg. This pharmacokinetic study of methyl kulonate is reported here for the first time.


Assuntos
Cromatografia Líquida/métodos , Limoninas/sangue , Espectrometria de Massas em Tandem/métodos , Administração Oral , Animais , Cromatografia Líquida/normas , Estabilidade de Medicamentos , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/farmacocinética , Limoninas/administração & dosagem , Limoninas/farmacocinética , Modelos Lineares , Masculino , Melia azedarach/química , Ratos , Ratos Sprague-Dawley , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrometria de Massas em Tandem/normas
17.
J Asian Nat Prod Res ; 14(1): 76-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22263597

RESUMO

A new chromone polymorchromone B was isolated and characterized from the roots of Angelica polymorpha Maxim, a commonly used traditional Chinese medicine. On the basis of spectroscopic analysis, including IR, HR-ESI-MS, 1D and 2D NMR spectral data, and hydrolysis followed by chromatographic analysis, the structure of the new chromone was elucidated as 5-hydroxy-2-[(angeloyloxy)methyl]-2'-(dimethyl)-3'-(2S,3S-epoxy-2-methylbutanoate)-dihydropyran [3',2':6,7]chromone. Moreover, it was found that polymorchromone B showed remarkable cytotoxic activity against A-549 cell lines.


Assuntos
Angelica/química , Antineoplásicos Fitogênicos/isolamento & purificação , Antineoplásicos Fitogênicos/farmacologia , Cromonas/isolamento & purificação , Cromonas/farmacologia , Antineoplásicos Fitogênicos/química , Cromonas/química , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Raízes de Plantas/química
18.
Environ Int ; 158: 106923, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34634619

RESUMO

As the use of zinc oxide nanoparticles (ZnO NPs) in everyday products grows, so does concern about health risks. However, no findings on the gastrointestinal toxicity of ZnO NPs have been published. We investigated the possible malignant transformation of ZnO NPs in the mice's colonic tissues using the APCmin/+ mouse model with a premalignant lesion in intestinal epithelial cells. Higher doses and long-term oral exposure to ZnO NPs were found to mildly promote colonic inflammation in WT mice, while they moderately or strongly exacerbated the severity of chronic inflammation and tumorigenesis in APCmin/+ mice with intestinal adenomatous polyposis. The ZnO NPs-induced inflammation and tumorigenesis in colonic epithelial cells was linked to the activation of CXCR2/NF-κB/STAT3/ERK and AKT pathways. Analysis of the ZnO NPs-exacerbated intestinal adenomatous polyposis in APCmin/+ mice revealed that ZnO NPs could activate the APC-driven Wnt/ß-catenin signaling pathway, exacerbating intestinal tumorigenesis. In fact, ZnO NPs have been shown to increase intestinal inflammation and tumorigenesis in APCmin/+ mice by releasing free Zn2+. In WT mice, a low dose of ZnO NPs (26 mg/kg/day) did not cause intestinal inflammation. In conclusion, higher doses and prolonged exposure to ZnO NPs promote the malignant transformation of precancerous epithelial cells.


Assuntos
Polipose Adenomatosa do Colo , Nanopartículas , Óxido de Zinco , Animais , Transformação Celular Neoplásica , Células Epiteliais , Camundongos , Nanopartículas/toxicidade , Óxido de Zinco/toxicidade
19.
Cell Death Dis ; 13(4): 380, 2022 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-35443745

RESUMO

C-X-C motif chemokine receptor 7 (CXCR7) is a newly discovered atypical chemokine receptor that binds to C-X-C motif chemokine ligand 12 (CXCL12) with higher affinity than CXCR4 and is associated with the metastasis of colorectal cancer (CRC). Cancer-associated fibroblasts (CAFs) have been known to promote tumor progression. However, whether CAFs are involved in CXCR7-mediated metastasis of CRC remains elusive. We found a significant positive correlation between CXCR7 expression and CAF activation markers in colonic tissues from clinical specimens and in villin-CXCR7 transgenic mice. RNA sequencing revealed a coordinated increase in the levels of miR-146a-5p and miR-155-5p in CXCR7-overexpressing CRC cells and their exosomes. Importantly, these CRC cell-derived miR-146a-5p and miR-155-5p could be uptaken by CAFs via exosomes and promote the activation of CAFs through JAK2-STAT3/NF-κB signaling by targeting suppressor of cytokine signaling 1 (SOCS1) and zinc finger and BTB domain containing 2 (ZBTB2). Reciprocally, activated CAFs further potently enhanced the invasive capacity of CRC cells. Mechanistically, CAFs transfected with miR-146a-5p and miR-155-5p exhibited a robust increase in the levels of inflammatory cytokines interleukin-6, tumor necrosis factor-α, transforming growth factor-ß, and CXCL12, which trigger the epithelial-mesenchymal transition and pro-metastatic switch of CRC cells. More importantly, the activation of CAFs by miR-146a-5p and miR-155-5p facilitated tumor formation and lung metastasis of CRC in vivo using tumor xenograft models. Our work provides novel insights into CXCR7-mediated CRC metastasis from tumor-stroma interaction and serum exosomal miR-146a-5p and miR-155-5p could serve as potential biomarkers and therapeutic targets for inhibiting CRC metastasis.


Assuntos
Fibroblastos Associados a Câncer , Neoplasias Colorretais , Exossomos , MicroRNAs , Animais , Fibroblastos Associados a Câncer/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Quimiocina CXCL12/genética , Quimiocina CXCL12/metabolismo , Neoplasias Colorretais/patologia , Exossomos/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas Repressoras/metabolismo
20.
Eur J Med Chem ; 227: 113923, 2022 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-34688013

RESUMO

Sphingosine-1-phosphate receptor 2 (S1PR2) has been identified as a brand-new GPCR target for designing antagonists to reverse 5-FU resistance. We herein report the structural optimization and structure-activity relationship of JTE-013 derivatives as S1PR2 antagonists. Compound 9d was the most potent S1PR2 antagonist (KD = 34.8 nM) among developed compounds. Here, compound 9d could significantly inhibit the expression of dihydropyrimidine dehydrogenase (DPD) to reverse 5-FU-resistance in HCT116DPD and SW620/5-FU cells. Further mechanism studies demonstrated that compound 9d not only inhibited S1PR2 but also affected the transcription of S1PR2. In addition, compound 9d also showed acceptable selectivity to normal cells (NCM460). Importantly, compound 9d with suitable pharmacokinetic properties could significantly reverse 5-FU-resistance in the HCT116DPD and SW620/5-FU xenograft models without obvious toxicity, in which the inhibition rates of 5-FU were increased from 23.97% to 65.29% and 27.23% to 60.81%, respectively. Further immunohistochemistry and western blotting analysis also demonstrated that compound 9d significantly decreases the expression of DPD in tumor and liver tissues. These results indicated that compound 9d is a promising lead compound to reverse 5-FU-resistance for colorectal cancer therapy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Desenho de Fármacos , Fluoruracila/farmacologia , Receptores de Esfingosina-1-Fosfato/antagonistas & inibidores , Animais , Antimetabólitos Antineoplásicos/síntese química , Antimetabólitos Antineoplásicos/química , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Relação Dose-Resposta a Droga , Ensaios de Seleção de Medicamentos Antitumorais , Fluoruracila/síntese química , Fluoruracila/química , Humanos , Masculino , Camundongos , Camundongos Nus , Simulação de Acoplamento Molecular , Estrutura Molecular , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Ratos , Ratos Sprague-Dawley , Receptores de Esfingosina-1-Fosfato/metabolismo , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA