Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 36(8): e22398, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35792869

RESUMEN

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.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Enfermedad del Hígado Graso no Alcohólico , Animales , Ácidos y Sales Biliares , Cromatografía Liquida , Firmicutes , Metaboloma , Ratones , Esfingolípidos , Esfingosina , Espectrometría de Masas en Tándem , Verrucomicrobia
2.
Acta Pharmacol Sin ; 43(11): 2967-2976, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35365782

RESUMEN

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.


Asunto(s)
Transformación Celular Neoplásica , Neoplasias Colorrectales , Receptores CXCR , Animales , Humanos , Ratones , beta-Arrestina 1/genética , beta-Arrestina 1/metabolismo , beta-Arrestinas/metabolismo , Carcinogénesis/genética , Transformación Celular Neoplásica/genética , Quimiocina CXCL12 , Neoplasias Colorrectales/genética , Ratones Transgénicos , Proteínas Nucleares/genética , Fosfoproteínas/metabolismo , Receptores CXCR/metabolismo
3.
Acta Pharmacol Sin ; 42(3): 460-469, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32647340

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Fluorouracilo/uso terapéutico , Lisofosfolípidos/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Uracilo/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Retículo Endoplásmico/metabolismo , Femenino , Células HCT116 , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Ribonucleasas/metabolismo , Transducción de Señal/fisiología , Esfingosina/metabolismo , Proteínas Supresoras de Tumor/metabolismo
4.
Pharmacol Res ; 155: 104717, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32088343

RESUMEN

In this study, S1PR2 was reckoned as a brand-new GPCR target for designing inhibitors to reverse 5-FU resistance. Herein a series of pyrrolidine pyrazoles as the S1PR2 inhibitors were designed, synthesized and evaluated for their activities of anti-FU-resistance. Among them, the most promising compound JTE-013, exhibited excellent inhibition on DPD expression and potent anti-FU-resistance activity in various human cancer cell lines, along with the in vivo HCT116DPD cells xenograft model, in which the inhibition rate of 5-FU was greatly increased from 13.01%-75.87%. The underlying mechanism was uncovered that JTE-013 demonstrated an anti-FU-resistance activity by blocking S1PR2 internalization to the endoplasmic reticulum (ER), which inhibited the degradation of 5-FU into α-fluoro-ß-alanine (FBAL) by downregulating tumoral DPD expression. Overall, JTE-013 could serve as the lead compound for the discovery of new anti-FU-resistance drugs. SIGNIFICANCE: This study provides novel insights that S1PR2 inhibitors could sensitize 5-FU therapy in colorectal cancer.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Fluorouracilo/uso terapéutico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Receptores de Esfingosina-1-Fosfato/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Dihidrouracilo Deshidrogenasa (NADP)/genética , Regulación hacia Abajo/efectos de los fármacos , Humanos , Ratones Desnudos , Simulación del Acoplamiento Molecular , Pirazoles/farmacología , Piridinas/farmacología , Receptores de Esfingosina-1-Fosfato/metabolismo
5.
Carcinogenesis ; 39(7): 889-899, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29757351

RESUMEN

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.


Asunto(s)
Alanina/análogos & derivados , Colitis Ulcerosa/prevención & control , Neoplasias Colorrectales/prevención & control , Estrés del Retículo Endoplásmico/efectos de los fármacos , Flavonoides/farmacología , Hidroxiquinolinas/farmacología , Alanina/farmacología , Animales , Anticarcinógenos/farmacología , Colon/efectos de los fármacos , Colon/metabolismo , Neoplasias Colorrectales/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Inflamación/prevención & control , Mediadores de Inflamación/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
6.
Cancer Invest ; 36(1): 10-18, 2018 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-29381400

RESUMEN

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.


Asunto(s)
Quimiocina CXCL12/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Neovascularización Patológica/genética , Receptores CXCR4/genética , Transducción de Señal/genética , Línea Celular , Células Endoteliales/metabolismo , Receptores ErbB/genética , Células Endoteliales de la Vena Umbilical Humana , Humanos , Sistema de Señalización de MAP Quinasas/genética , Metaloproteinasa 2 de la Matriz/genética , Proteínas Quinasas Activadas por Mitógenos/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/genética , Vía de Señalización Wnt/genética , beta Catenina/genética
7.
Cancer Invest ; 34(9): 459-464, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27673353

RESUMEN

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.


Asunto(s)
Biomarcadores/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Precursores de Proteínas/metabolismo , Protrombina/metabolismo , Transducción de Señal , Progresión de la Enfermedad , Humanos , Janus Quinasa 1/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Neovascularización Patológica , Fosfolipasa C gamma/metabolismo , Unión Proteica , Precursores de Proteínas/biosíntesis , Protrombina/biosíntesis , Proteínas Proto-Oncogénicas c-met/metabolismo , Factor de Transcripción STAT3/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Am J Ther ; 23(1): e198-207, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-24621642

RESUMEN

Arginine-glycine-aspartate (RGD)-binding integrins, including αvß1, αvß3, αvß5, αvß6, αvß8, α5ß1, αIIbß3, and α8ß1, recognize the tripeptide motif RGD in their ligands. RGD-binding integrins are involved in various cell functions, including cell proliferation, survival, differentiation, and motility that are critically important to both health and disease. The diagnostic and therapeutic value of some RGD-binding integrin inhibitors are either clinically proven or at different stages of development. In this review, we first summarized the structure and signaling characteristics of RGD-binding integrins. We then discussed the functions of RGD-binding integrins and their association with human disease. Finally, we recapitulated the research efforts and clinical trials of targeting RGD-binding integrins for the diagnosis and treatment of human disease. This comprehensive review of the current advances in RGD-binding integrins could assist scientists and clinicians in gaining a complete understanding of this group of molecules. It can also contribute to the design of new projects to further advance this field of research and to better apply the research results to benefit patients in clinical practice.


Asunto(s)
Integrinas/antagonistas & inhibidores , Oligopéptidos/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Diagnóstico por Imagen , Humanos , Integrinas/química , Integrinas/fisiología , Neoplasias/tratamiento farmacológico , Transducción de Señal
9.
Cell Physiol Biochem ; 35(1): 201-12, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25591763

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Precursores de Proteínas/farmacología , Protrombina/farmacología , Quinazolinas/farmacología , Biomarcadores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Caspasas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/metabolismo , Gefitinib , Células Hep G2 , Factor de Crecimiento de Hepatocito/metabolismo , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Regulación hacia Arriba/efectos de los fármacos
11.
Biochim Biophys Acta ; 1830(10): 4907-16, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23850473

RESUMEN

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.


Asunto(s)
Adenocarcinoma/patología , División Celular/efectos de los fármacos , Neoplasias del Colon/patología , Transducción de Señal/efectos de los fármacos , Triterpenos/farmacología , Adenocarcinoma/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias del Colon/metabolismo , Receptores ErbB/metabolismo , Células HT29 , Humanos , Etiquetado Corte-Fin in Situ , Ratones , Triterpenos/toxicidad
12.
Biochim Biophys Acta ; 1830(6): 3604-15, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23500016

RESUMEN

BACKGROUND: Acetyl-11-keto-beta-boswellic acid (AKBA) is a derivative of boswellic acid, an active component of Boswellia serrata gum resin. We examined the effect of AKBA on human gastric carcinoma growth and explored the underlying molecular mechanisms. METHODS: Inhibition of cancer cell growth was estimated by colorimetric and clonogenic assays. Cell cycle distribution was analyzed by flow cytometry and apoptosis determined using Annexin V-FITC/PI staining and DNA ladder quantification. After three weeks of oral AKBA administration in nude mice bearing cancer xenografts, animals were sacrificed and xenografts removed for TUNEL staining and western blot analysis. RESULTS: AKBA exhibited anti-cancer activity in vitro and in vivo. With oral application in mice, AKBA significantly inhibited SGC-7901 and MKN-45 xenografts without toxicity. This effect might be associated with its roles in cell cycle arrest and apoptosis induction. The results also showed activation of p21(Waf1/Cip1) and p53 in mitochondria and increased cleaved caspase-9, caspase-3, and PARP and Bax/Bcl-2 ratio after AKBA treatment. Further analysis suggested that these effects might arise from AKBA's modulation of the aberrant Wnt/ß-catenin signaling pathway. Upon AKBA treatment, ß-catenin expression in nuclei was inhibited, and membrane ß-catenin was activated. In the same sample, active GSK3ß was increased and its non-active form decreased. Levels of cyclin D1, PCNA, survivin, c-Myc, MMP-2, and MMP-7, downstream targets of Wnt/ß-catenin, were inhibited. CONCLUSIONS: AKBA effects on human gastric carcinoma growth were associated with its activity in modulating the Wnt/ß-catenin signaling pathway. GENERAL SIGNIFICANCE: AKBA could be useful in the treatment of gastric cancers.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/metabolismo , Triterpenos/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Animales , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Línea Celular Tumoral , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/patología , Ciclina D1/genética , Ciclina D1/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Neoplasias Gástricas/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína X Asociada a bcl-2/genética , Proteína X Asociada a bcl-2/metabolismo , beta Catenina/biosíntesis , beta Catenina/genética
13.
Biochim Biophys Acta ; 1830(1): 2194-203, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23085067

RESUMEN

BACKGROUND: Riccardin D-26, a synthesized macrocyclic bisbibenzyl compound, might possess anti-cancer properties. We aimed to evaluate the efficacy of Riccardin D-26 as a candidate compound for treatment of cancers with sensitive or drug resistant cells. METHODS: Experiments were performed on human oral squamous carcinoma KB cells and vincristin-selected MDR KB/VCR cells. The inhibition of cell growth was evaluated by colorimetric and clonogenic assays. The apoptotic cells were determined by the Annexin V-FITC/PI staining assay. JC-1 fluorescence probe was used to examine the mitochondria membrane potential (MMP). Further experiments were performed in nude mice bearing KB or KB/VCR xenografts. Riccardin D-26 was administered by injection for 2weeks. The specimens of KB and KB/VCR xenografts were removed for TUNEL staining and Western blotting analysis. RESULTS: Riccardin D-26 significantly inhibited cancer growth in both KB and KB/VCR cells. Riccardin D-26's activity in cancer cells was greater than that in human normal liver cells. In mice, Riccardin D-26 effectively prevented the growth of KB and KB/VCR xenografts without significant toxicity. Further studies suggested that Riccardin D-26 inhibited cancer growth by inducing apoptosis in the activation of mitochondria-mediated intrinsic apoptosis pathway. Riccardin D-26 also possessed this activity in regulation of mitogen-related protein kinases such as MAPK and PI3K/Akt, which is associated with its inhibitory effect on KB/VCR cells. CONCLUSIONS: Riccardin D-26 possessed an anti-proliferation activity against both sensitive KB and MDR KB/VCR cancer cells. GENERAL SIGNIFICANCE: Riccardin D-26 could be a promising agent for treatment of cancers with sensitive or drug resistant cells.


Asunto(s)
Antineoplásicos , Carcinoma de Células Escamosas/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Compuestos Macrocíclicos , Neoplasias de la Boca/tratamiento farmacológico , Éteres Fenílicos , Estilbenos , Animales , Anexina A5/metabolismo , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Compuestos Macrocíclicos/síntesis química , Compuestos Macrocíclicos/química , Compuestos Macrocíclicos/farmacología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Desnudos , Mitocondrias/metabolismo , Mitocondrias/patología , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Éteres Fenílicos/síntesis química , Éteres Fenílicos/química , Éteres Fenílicos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Estilbenos/síntesis química , Estilbenos/química , Estilbenos/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Physiol Biochem ; 34(3): 903-15, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25200250

RESUMEN

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.


Asunto(s)
Carcinoma Hepatocelular/fisiopatología , Sustancias de Crecimiento/fisiología , Neoplasias Hepáticas/fisiopatología , Precursores de Proteínas/fisiología , Protrombina/fisiología , Biomarcadores/química , Biomarcadores de Tumor , Carcinoma Hepatocelular/diagnóstico , Humanos , Neoplasias Hepáticas/diagnóstico , Estructura Molecular , Precursores de Proteínas/química , Protrombina/química
15.
Anticancer Drugs ; 25(10): 1107-21, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25010394

RESUMEN

Integrins are a large family of cell surface receptors that bind extracellular matrix proteins. The interaction of integrins with extracellular matrix activates a number of intracellular signaling pathways involved in cell proliferation, differentiation, motility, and other essential cell functions. Integrins are critically important to both health and disease. In this review, we first describe the structure, functions, and signaling characteristics of integrins. We then discuss the roles of integrins in cancer progression. Finally, we recapitulate the laboratory and clinical efforts of targeting integrins as effective means of cancer therapy and diagnosis. This comprehensive review could help scientists and clinicians gain a complete understanding of integrins. It could also contribute toward the development of new drugs, new methods of diagnostics, and new treatment of cancers to benefit the patients in clinical practice.


Asunto(s)
Antineoplásicos/uso terapéutico , Integrinas/metabolismo , Neoplasias/diagnóstico , Neoplasias/patología , Animales , Movimiento Celular , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Integrinas/química , Invasividad Neoplásica , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica/metabolismo , Pronóstico , Transducción de Señal
16.
Int J Cancer ; 132(11): 2667-81, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23132636

RESUMEN

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.


Asunto(s)
Adenoma/prevención & control , Proteína de la Poliposis Adenomatosa del Colon/fisiología , Poliposis Adenomatosa del Colon/prevención & control , Apoptosis/efectos de los fármacos , Poliposis Intestinal/prevención & control , Neovascularización Patológica/prevención & control , Triterpenos/uso terapéutico , Adenoma/genética , Adenoma/patología , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Animales , Western Blotting , Boswellia/química , Proliferación Celular , Ciclooxigenasa 2/química , Ciclooxigenasa 2/metabolismo , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Técnicas para Inmunoenzimas , Mediadores de Inflamación/metabolismo , Poliposis Intestinal/genética , Poliposis Intestinal/patología , Leucotrieno B4/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Transducción de Señal , beta Catenina/metabolismo
17.
Biochem Biophys Res Commun ; 438(2): 402-9, 2013 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-23899521

RESUMEN

UNLABELLED: SL-01 is an oral derivative of gemcitabine that was synthesized by introducing the moiety of 3-(dodecyloxycarbonyl) pyrazine-2-carbonyl at N4-position on cytidine ring of gemcitabine. We aimed to evaluate the efficacy of SL-01 on human breast cancer growth. SL-01 significantly inhibited MCF-7 proliferation as estimated by colorimetric assay. Flow cytometry assay indicated the apoptotic induction and cell cycle arrest in G1 phase. SL-01 modulated the expressions of p-ATM, p53 and p21 and decrease of cyclin D1 in MCF-7 cells. Further experiments were performed in a MCF-7 xenografts mouse model. SL-01 by oral administration strongly inhibited MCF-7 xenografts growth. This effect of SL-01 might arise from its roles in the induction of apoptosis. Immunohistochemistry assay showed the increase of TUNEL staining cells. Western blotting indicated the modulation of apoptotic proteins in SL-01-treated xenografts. During the course of study, there was no evidence of toxicity to mice. In contrast, the decrease of neutrophil cells in peripheral and increase of AST and ALT levels in serum were observed in the gemcitabine-treated mice. CONCLUSION: SL-01 possessed similar activity against human breast cancer growth with gemcitabine, whereas, with lower toxicity to gemcitabine. SL-01 is a potent oral agent that may supplant the use of gemcitabine.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis , Neoplasias de la Mama/patología , Desoxicitidina/análogos & derivados , Administración Oral , Animales , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Desoxicitidina/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto , Gemcitabina
18.
Bioorg Med Chem Lett ; 23(7): 1989-92, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23454017

RESUMEN

New series of indazole based diarylureas were synthesized and their anticancer activity against cancer cells H460, A549, OS-RC-2, HT-29, Lovo, HepG2, Bel-7402, SGC-7901 and MDA-MB-231 were examined. These derivatives of diarylureas, except azaindazole based diarylureas 5f, 5l and 5m, showed superior or similar activity against most of these selected cancer cell lines to the reference compound sorafenib. The effect of substituents on the indazole ring was also investigated. Derivatives with trifluoromenthy or halogen substituent on the indazole ring showed higher activity against the selected cancer cell lines than sorafenib. The acute toxicity assay showed that compounds 5a, 5b and 5i possessed lower toxicity than sorafenib. Compound 5i with 4-(trifluoromenthy)-1H-indazole and 4-(trifluoromenthy) benzene moieties exhibited the most potent anticancer activity.


Asunto(s)
Antineoplásicos/farmacología , Indazoles/química , Urea/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HT29 , Células Hep G2 , Humanos , Estructura Molecular , Relación Estructura-Actividad , Urea/análogos & derivados , Urea/síntesis química
19.
Invest New Drugs ; 30(1): 212-22, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20924640

RESUMEN

We studied the effect of riccardin D, a macrocyclic bisbibenzyl, which was isolated from the Chinese liverwort plant, on human leukemia cells and the underlying molecular mechanism. Riccardin D had a significant antiproliferative effect on human leukemia cell lines HL-60, K562 and its multidrug resistant (MDR) counterpart K562/A02 cells, but showed no effect on the topoisomerase-II-deficient HL-60/MX2 cells, as measured by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay. The pBR322 DNA relaxation assay revealed that riccardin D selectively inhibited the activity of topoisomerase II (topo II). The suppression of topo II activity by riccardin D was stronger than that of etoposide, a known topo II inhibitor. After treatment with riccardin D, nuclear extracts of leukemia K562 and K562/A02 cells left the majority of pBR322 DNA in a supercoiled form. Further examination showed that riccardin D effectively induced HL-60, K562 and K562/A02 apoptosis as evidenced by externalization of phosphatidylserine and formation of DNA ladder fragments. The activation of cytochrome c, caspase-9, caspase-3 and cleaved poly ADP-ribose polymerase (PARP) was also enhanced, as estimated by Western blot analysis. By contrast, riccardin D was unable to induce apoptosis in the topoisomerase-II-deficient HL-60/MX2 cells, indicating that the induction of apoptosis by riccardin D was due to the inhibition of topo II activity. In addition, riccardin D was able to significantly decrease P-glycoprotein (P-gp) expression in K562/A02 cells. Taken together, our data demonstrate that riccardin D is a novel DNA topo II inhibitor which can induce apoptosis of human leukemia cells and that it has therapeutic potential for both regular and MDR strains of leukemia cells.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/metabolismo , Leucemia/enzimología , Éteres Fenílicos/farmacología , Estilbenos/farmacología , Inhibidores de Topoisomerasa II/farmacología , Subfamilia B de Transportador de Casetes de Unión a ATP , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Proliferación Celular/efectos de los fármacos , Citocromos c/metabolismo , Fragmentación del ADN , ADN Superhelicoidal/metabolismo , Relación Dosis-Respuesta a Droga , Resistencia a Múltiples Medicamentos , Resistencia a Antineoplásicos , Células HL-60 , Humanos , Células K562 , Leucemia/genética , Leucemia/patología , Fosfatidilserinas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factores de Tiempo
20.
Invest New Drugs ; 29(4): 574-82, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20111888

RESUMEN

LYP is a bestatin dimethylaminoethyl ester which inhibits aminopeptidase N (APN/CD13). Our goal in this study was to evaluate LYP as a candidate compound for cancer treatment, beginning by studying its inhibitory effects on tumors and then comparing it to bestatin. Experiments were performed on human ovarian carcinoma (OVCA) ES-2 and SKOV-3 cell lines, which have high and low levels of APN/CD13 respectively. LYP effectively inhibited ES-2 cell growth as estimated by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and the trypan blue dye-exclusion test. LYP significantly suppressed APN/CD13 activity on the surface of ES-2 cells as measured by quantifying the enzymatic cleavage of the substrate L-leucine-p-nitroanilide. The inhibitory effects of LYP were greater than those of bestatin at the same concentrations. In contrast, LYP was a weak inhibitor of SKOV-3 cell growth, suggesting that LYP may inhibit ES-2 cell growth via suppression of APN/CD13. Inhibition of APN/CD13 expression was also demonstrated with immunofluorescent flow cytometry and Western blot analysis. Inhibitory effects of LYP were confirmed by using a mouse model in which LYP delayed the growth of ES-2 xenografts in mice after 2 weeks of LYP injections. Inhibition of APN/CD13 expression was demonstrated in the ES-2 xenografts using Western blot analysis. The inhibitory effects of LYP on the ES-2 xenografts were stronger than those of bestatin. These results suggest that LYP has a powerful inhibitory effect on the growth of OVCA cells and that the mechanism may be via a decrease in the expression of APN/CD13.


Asunto(s)
Antígenos CD13/antagonistas & inhibidores , Leucina/análogos & derivados , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Animales , Antígenos CD13/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Proliferación Celular/efectos de los fármacos , Ésteres , Femenino , Humanos , Leucina/química , Leucina/farmacología , Ratones , Neoplasias Ováricas/sangre , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA