Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Intervalo de año de publicación
1.
Biomed Pharmacother ; 169: 115849, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37976890

RESUMEN

Tumor cells and macrophages communicate through the secretion of various cytokines to jointly promote the malignant development of cancers. We synthesized and characterized an oxoaporphine Pr(III) complex (PrL3(NO3)3) and found that it inhibits hepatocellular carcinoma (HCC) progression and metastasis by disrupting HCC cell-macrophage crosstalk. PrL3(NO3)3 treatment upregulated CD86, TNF-α, and IL-1ß and downregulated CD163, CD206, CCL2, and VEGFA in macrophages. Our mRNA-Seq results demonstrated that PrL3(NO3)3 inhibited macrophage M2-like polarization by inhibiting the AMPK pathway and activating the NF-κB pathway by upregulating RelA/p65 Ser536 phosphorylation. This kind of macrophage polarization significantly inhibited HCC cell proliferation, migration, and invasion. In addition, PrL3(NO3)3 inhibited the migration, invasion, and chemotaxis of HCC cells by downregulating the expression of EMT-related markers and CCL2. hTFtarget database analysis revealed that PrL3(NO3)3 inhibited NF-κB nuclear translocation by upregulating RelA/p65 Ser536 phosphorylation in HCC cells, thereby downregulating the expression of Snail and CCL2. HCC tissue microarray analysis revealed that downregulation of RelA/p65 Ser536 phosphorylation is a driving event in HCC malignant progression. In conclusion, PrL3(NO3)3 effectively inhibits HCC cell-macrophage crosstalk by upregulating RelA/p65 Ser536 phosphorylation. This is the first report of a lanthanide complex exerting regulatory effects on both tumors and tumor-associated macrophages, providing a new strategy for the development of effective antitumor drugs.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patología , FN-kappa B/metabolismo , Neoplasias Hepáticas/patología , Línea Celular Tumoral , Macrófagos/metabolismo
2.
Dalton Trans ; 52(11): 3287-3294, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36691961

RESUMEN

Three copper(II) complexes C1-C3 were synthesized and fully characterized as chemodynamic therapy (CDT) anticancer agents. C1-C3 showed greater cytotoxicity than their ligands toward SK-OV-3 and T24 cells. Particularly, C2 showed high cytotoxicity toward T24 cells and low cytotoxicity toward normal human HL-7702 and WI-38 cells. Mechanistic studies demonstrated that C2 oxidized GSH to GSSG and produced ˙OH, which induced mitochondrial dysfunction and ER stress, finally leading to apoptosis of T24 cells. In addition, C2 inhibited autophagy by blocking autophagy flow, thereby closing the self-protection pathway of oxidative stress to enhance CDT. Importantly, C2 significantly inhibited T24 tumor growth with 57.1% inhibition in a mouse xenograft model. C2 is a promising lead as a potential CDT anticancer agent.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Animales , Ratones , Línea Celular Tumoral , Cobre/farmacología , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Estrés Oxidativo , Autofagia , Peróxido de Hidrógeno , Glutatión/metabolismo
3.
J Med Chem ; 65(6): 5134-5148, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35255688

RESUMEN

Twelve new complexes Cu(L1)2-Cu(L12)2 were designed and synthesized to improve their chemotherapeutic properties. They showed considerable antiproliferative activity against T24 cancer cells but lower cytotoxicity to human normal cells HL-7702 and WI-38. A mechanism study indicated that Cu(L4)2 and Cu(L10)2 were reduced to Fenton-like Cu+ by glutathione depletion, and the resulting Cu+ catalyzed the generation of highly toxic hydroxyl radicals from excess H2O2. Simultaneously, Cu(L4)2 and Cu(L10)2 could decrease the catalase activity to restrain H2O2 transfer to H2O for enhanced chemodynamic therapy (CDT). These induced mitochondrial dysfunctions and endoplasmic reticulum stress to induce T24 cell apoptosis. In addition, Cu(L4)2 and Cu(L10)2 inhibited autophagy flux to promote cell apoptosis. Cu(L4)2 and Cu(L10)2 demonstrated strong tumor inhibition ability in the T24 xenograft model. Moreover, Cu(L10)2 showed higher antitumor activity and a better safety profile than the CDT agent Cu1. Cu(L10)2 exhibited excellent pharmacokinetic properties. Collectively, Cu(L4)2 and Cu(L10)2 could be developed as potential CDT candidates for cancer treatment.


Asunto(s)
Antineoplásicos , Hidroxiquinolinas , Neoplasias , Quinolinas , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Autofagia , Línea Celular Tumoral , Cobre , Glutatión , Humanos , Peróxido de Hidrógeno/farmacología , Neoplasias/tratamiento farmacológico , Bases de Schiff/farmacología
4.
Dalton Trans ; 51(4): 1333-1343, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-34989734

RESUMEN

Three ruthenium(III) complexes with pyrazolopyrimidine [Ru(Ln)(H2O)Cl3] (1-3, n = 1-3) were prepared and characterized. These Ru(III) compounds show strong cytotoxicity against six cancer cell lines and low toxicity to normal human liver cells. Particularly, they exhibited stronger cytotoxicity to SK-OV-3 cells than cisplatin. Mechanism studies revealed that complex 1 inhibited tumor cell invasion and suppressed cell proliferation, induced apoptosis by elevating the levels of intracellular ROS (reactive oxygen species) and free calcium (Ca2+), and reduced mitochondrial membrane potential (ΔΨ). It also activated the caspase cascade, accompanied with upregulation of cytochrome c, Bax, p53, Apaf-1 and downregulation of Bcl-2. Moreover, complex 1 caused cell cycle arrest at S phase by inhibiting the expression of CDC 25, cyclin A2 and CDK 2 proteins, and induced DNA damage by interacting with DNA and inhibiting the topoisomerase I enzyme. Complex 1 exhibited efficient in vivo anticancer activity in a model of SK-OV-3 tumor xenograft.


Asunto(s)
Antineoplásicos/farmacología , Complejos de Coordinación/uso terapéutico , Piridinas/uso terapéutico , Compuestos de Rutenio/uso terapéutico , Animales , Antineoplásicos/química , Apoptosis , Bencimidazoles , Calcio , Caspasa 3/genética , Caspasa 3/metabolismo , Caspasa 9/genética , Caspasa 9/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/química , Daño del ADN , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Membranas Mitocondriales/efectos de los fármacos , Piridinas/química , Especies Reactivas de Oxígeno , Compuestos de Rutenio/química , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Eur J Med Chem ; 223: 113636, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34175540

RESUMEN

Three Cu(II) complexes of quinoline derivatives as cancer chemodynamic therapy agents were synthesized and characterized. These complexes were heavily taken up by cells and reacted with cellular glutathione (GSH) to reduce Cu2+ to Fenton-like Cu+, which catalyzed endogenous H2O2 to produce the highly toxic hydroxyl radicals (•OH) to kill cancer cells. Cu1 and Cu2 initiated CAT activity declines, mitochondrial membrane potential and ATP concentration decrease, mitochondrial Ca2+ overload and ER stress response, leading to cell cycle arrest in sub-G1 and cancer cell caspase-dependent apoptosis. On account of the high GSH and H2O2 specific properties of the tumor microenvironment, Cu1 and Cu2 exhibited higher in vitro anticancer activity and lower toxicity to normal cells. Cu1 and Cu2 efficiently inhibited tumor growth in the SK-OV-3 xenograft mouse model without obvious systemic toxicity.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Complejos de Coordinación/química , Cobre/química , Estrés del Retículo Endoplásmico/efectos de los fármacos , Quinolinas/química , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Complejos de Coordinación/síntesis química , Complejos de Coordinación/metabolismo , Complejos de Coordinación/farmacología , Cristalografía por Rayos X , Ensayos de Selección de Medicamentos Antitumorales , Glutatión/química , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Conformación Molecular , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Distribución Tisular , Trasplante Heterólogo
6.
J Inorg Biochem ; 201: 110820, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31518871

RESUMEN

Seven Cu(II) complexes with 5-pyridin-2-yl-[1,3]dioxolo[4,5-g]isoquinoline derivatives as ligands: [Cu2(L1)2Cl4] (1), [Cu(L2)Cl2] (2), [Cu(L1)(NO3)2] (3), [Cu(L2)(NO3)2] (4), [Cu(L3)Cl2] (5), [Cu(L3)Br2] (6) and [Cu(L3)(NO3)2] (7){L1=9-nitro-5-pyridin-2-yl-[1,3]dioxolo[4,5-g]isoquinoline, L2=4-nitro-5-pyridin-2-yl-[1,3]dioxolo[4,5-g]isoquinoline, L3=9-bromo-5-pyridin-2-yl-[1,3]dioxolo[4,5-g]isoquinoline}, were synthesized and characterized. Their in vitro anticancer activities against T-24, MGC-80-3, HeLa, Hep-G2, A549 and SK-OV-3 were evaluated. Compared with their corresponding ligands, most of these complexes exhibited enhanced anticancer activities in contrast to their corresponding ligands and copper salt. Among them, complexes 1 and 3 displayed selective cytotoxicity to HeLa cells comparing with normal liver cell HL-7702, with IC50 values of 5.03 ±â€¯1.20 µM and 10.05 ±â€¯0.52 µM, respectively. Complexes 1 and 3 inhibited telomerase activity by interacting with c-myc promoter elements, and therefore exerted their antitumor activity. Furthermore, complexes 1 and 3 could trigger cell apoptosis via disruption of mitochondrial pathway through notably increased reactive oxygen species (ROS) levels, loss of mitochondrial membrane potential (Δψm), increase of the cytochrome c and apaf-1, decrease of bcl-2, and activation of caspases 3/9. Complexes 1 and 3 exhibited enhanced cytotoxicity, presenting synergetic effect after the ligands coordinated to copper(II) center.


Asunto(s)
Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Complejos de Coordinación/síntesis química , Cobre/química , Inhibidores Enzimáticos/síntesis química , Compuestos Organometálicos/síntesis química , Quinolinas/química , Apoptosis/efectos de los fármacos , Complejos de Coordinación/farmacología , Inhibidores Enzimáticos/farmacología , Células HeLa , Células Hep G2 , Humanos , Mitocondrias/efectos de los fármacos , Compuestos Organometálicos/farmacología , Piridinas/química , Telomerasa/antagonistas & inhibidores
7.
Spectrochim Acta A Mol Biomol Spectrosc ; 209: 141-149, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30384019

RESUMEN

A fluorescent sensor L based on a pyrazolopyrimidine core simultaneously detects Cu2+ and Ni2+ ions by photoluminescence quenching, even in the presence of other metal cations. Sensor L possesses high association constants of 5.24 × 103 M-1 and 2.85 × 104 M-1 and low detection limits of 0.043 µM and 0.038 µM for Cu2+ and Ni2+, respectively. The binding stoichiometry ratios of L to Cu2+ or Ni2+ is 1:1 as determined by Benesi-Hildebrand and Job's plots, and by crystal structures. DFT calculations on L-Cu2+ indicated reduced electron donation from the coordinated pyrazolopyridine to the fused pyrimidine and pendant phenyl group which, together with a smaller HOMO-LUMO orbital gap could favour non-radiative decay and explain the observed fluorescence quenching. Sensor L possessed low cytotoxicity and good imaging characteristics for Cu2+ and Ni2+ in living cells, suggesting potential applications for detecting Cu2+ and Ni2+in vivo.


Asunto(s)
Cobre/análisis , Fluorescencia , Colorantes Fluorescentes/química , Níquel/análisis , Pirazoles/química , Pirimidinas/química , Neoplasias de la Vejiga Urinaria/metabolismo , Supervivencia Celular , Humanos , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/patología
8.
Sci Rep ; 7: 46056, 2017 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-28436418

RESUMEN

Three new oxoaporphine Co(II), Ni(II) and Zn(II) complexes 1-3 have been synthesized and fully characterized. 1-3 have similar mononuclear structures with the metal and ligand ratio of 1:2. 1-3 exhibited higher cytotoxicity than the OD ligand and cisplatin against HepG2, T-24, BEL-7404, MGC80-3 and SK-OV-3/DDP cells, with IC50 value of 0.23-4.31 µM. Interestingly, 0.5 µM 1-3 significantly caused HepG2 arrest at S-phase, which was associated with the up-regulation of p53, p21, p27, Chk1 and Chk2 proteins, and decrease in cyclin A, CDK2, Cdc25A, PCNA proteins. In addition, 1-3 induced HepG2 apoptosis via a caspase-dependent mitochondrion pathway as evidenced by p53 activation, ROS production, Bax up-regulation and Bcl-2 down-regulation, mitochondrial dysfunction, cytochrome c release, caspase activation and PARP cleavage. Furthermore, 3 inhibited tumor growth in HepG2 xenograft model, and displayed more safety profile in vivo than cisplatin.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Aporfinas/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Complejos de Coordinación/farmacología , Metales/farmacología , Mitocondrias/metabolismo , Fase S/efectos de los fármacos , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Aporfinas/síntesis química , Aporfinas/química , Calcio/metabolismo , Caspasas/metabolismo , Forma de la Célula/efectos de los fármacos , Complejos de Coordinación/síntesis química , Complejos de Coordinación/química , Citocromos c/metabolismo , Daño del ADN , ADN-Topoisomerasas de Tipo II/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Células Hep G2 , Humanos , Ligandos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Mitocondrias/efectos de los fármacos , Modelos Biológicos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Carga Tumoral/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Dalton Trans ; 44(25): 11408-19, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26017376

RESUMEN

Complexes of yttrium(III) and dysprosium(III) with the traditional Chinese medicine active ingredient oxoglaucine (OG), namely [Y(OG)2(NO3)3]·CH3OH (1) and [Dy(OG)2(NO3)3]·H2O (2), were synthesized and characterized by elemental analysis, IR, ESI-MS, (1)H and (13)C NMR as well as single-crystal X-ray diffraction analysis. In vitro the complexes exhibited higher anticancer activity than the free ligand OG against the tested cancer cell lines. Among the tested cell lines, HepG2 is the most sensitive to the complexes. Complex 2 can trigger DNA damage in HepG2 cells, resulting in cell cycle arrest in the S phase and leading to cell apoptosis. The S phase cell-cycle arrest is caused via the ATM (ataxia-telangiectasia mutated)-Chk2-Cdc25A pathway. Chk2 is phosphorylated and activated in an ATM-dependent manner. It, in turn, phosphorylates Cdc25A phosphatise on serine124, causing the inactivation of Cdc25A in ubiquitin-mediated proteolytic degradation. The cyclin-Cdk complexes of the S phase could also be inhibited by limited supply of cyclins A and E. This irreversible cell cycle arrest process ultimately induces mitochondria-involved apoptotic cell death via the activation of Bcl-2 protein. Complex e2 ffectively inhibited tumour growth in the BEL-7402 xenograft mouse model and exhibited higher safety in vivo than cisplatin.


Asunto(s)
Antineoplásicos , Apomorfina/análogos & derivados , Complejos de Coordinación , Disprosio , Inhibidores de Topoisomerasa , Itrio , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apomorfina/química , Apomorfina/farmacología , Apomorfina/uso terapéutico , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Complejos de Coordinación/uso terapéutico , ADN/metabolismo , Daño del ADN , Disprosio/química , Disprosio/farmacología , Disprosio/uso terapéutico , Humanos , Medicina Tradicional China , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Fase S/efectos de los fármacos , Solubilidad , Inhibidores de Topoisomerasa/química , Inhibidores de Topoisomerasa/farmacología , Inhibidores de Topoisomerasa/uso terapéutico , Carga Tumoral/efectos de los fármacos , Agua/química , Difracción de Rayos X , Itrio/química , Itrio/farmacología , Itrio/uso terapéutico
10.
Eur J Med Chem ; 89: 77-87, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25462228

RESUMEN

A new platinum(II) complex of [Pt(II)(L) (pn)]Cl·2H2O (1) (pn = 1,3-propanediamine) with 2-(4-methoxy-phenyl)imidazo [4,5-f]-[1,10]phenanthrolin (H-L) was synthesized and characterized. In complex 1, the platinum adopts a four-coordinated square planar geometry. Complex 1 exhibited selective cytotoxicity against NCI-H460, BEL-7402, SK-OV-3, SK-OV-3/DDP and HeLa cell lines with IC50 values in the micromolar range (9.7-35.8 µM), but low cytotoxicity toward normal human liver HL-7702 cells. Complex 1 caused HeLa cell cycle arrest at S phase and it induced HeLa apoptosis by the activation of caspase-3/9. Various experiments showed that complex 1 preferred to bind with G-quadruplex in c-myc. Taken together, we found that complex 1 exerted its antitumor activity mainly via inhibiting telomerase by interaction with c-myc quadruplex and activation of caspase-3/9.


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Compuestos Organoplatinos/farmacología , Fenantrolinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Imidazoles/química , Estructura Molecular , Compuestos Organoplatinos/síntesis química , Compuestos Organoplatinos/química , Fenantrolinas/química , Relación Estructura-Actividad , Células Tumorales Cultivadas
11.
Zhonghua Wei Chang Wai Ke Za Zhi ; 9(4): 342-4, 2006 Jul.
Artículo en Chino | MEDLINE | ID: mdl-16886120

RESUMEN

OBJECTIVE: To explore the relationship between the changes of trace elements and lymphatic metastasis in gastric carcinoma. METHODS: Trace elements including Fe, Mg, Mn, Ca, Cu, Zn, Se were measured in primary gastric carcinoma and regional lymph nodes from 40 patients with gastric carcinoma, and compared among the primary tumor, metastatic, and non-metastatic nodes. RESULTS: There were no significant differences in the contents of Fe, Mg, Mn and Ca among primary gastric tumors, regional lymph nodes with or without metastasis (P=0.372 - 0.741, P > 005), and no significant differences in the contents of all 7 trace elements between primary tumors and metastatic lymph nodes (P=0.15 - 0.59, P > 005). Compared with metastatic lymph nodes, the contents of Zn, Se significantly decreased, while Cu and Cu/Zn significantly increased (P=0.001 - 0.009, P< 0.01) in non-metastatic lymph nodes. The content of Zn in N2 positive lymph nodes was significant lower than that in N1 positive nodes (P=0.027). There were no significant difference in the contents of all 7 elements between intestinal type and diffuse type (P=0.149 - 0.758, P > 0.05). CONCLUSIONS: Lymphatic metastasis of gastric cancer is concomitant with the changes of trace elements, and the changes of Zn, Cu, Se may be related with lymphatic metastasis.


Asunto(s)
Ganglios Linfáticos/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Oligoelementos/metabolismo , Adulto , Anciano , Femenino , Humanos , Metástasis Linfática , Masculino , Persona de Mediana Edad
12.
Int J Mol Med ; 11(4): 479-84, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12632101

RESUMEN

To explore the relationship between the reactive oxygen species (ROS) and apoptosis in esophageal carcinoma cells (SHEE85) induced by arsenic trioxide (As2O3), we focused on changes of apoptosis, ROS, and antioxidants. Apoptosis of SHEE85 was confirmed by means of DNA fragmentation stained by Hoechst 33342, Sub-G1 cells scored by flow cytometry and ultrastructure of cells by electron microscopy. To evaluate the level of ROS, the chemiluminescent method was used for measuring the production of superoxide anion (O(-)*2). Lipid peroxide (malondialdehyde, MDA), superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) were measured respectively by the photometry method. In the cells treated with As2O3 at a concentration of 5.0 micromol/l for 2-24 h, the content of cellular O(-)*2 and MDA was increased, but SOD and GSH-Px were significantly lower in the process of apoptosis in SHEE85. As2O3 at concentration of 0.5 micromol/l did not cause cell apoptosis but promoted cell proliferation. These results suggest that As2O3 at a high dosage (5 micromol/l) causes cell apoptosis and at a low dosage (0.5 micromol/l) causes cell proliferation. The essential mechanisms of cell apoptosis induced by As2O3 may be related to the increase of ROS and decrease of anti-oxidation. ROS and antioxidants participate in the apoptotic pathway of esophageal carcinoma cells.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Neoplasias Esofágicas/tratamiento farmacológico , Óxidos/farmacología , Antioxidantes/metabolismo , Trióxido de Arsénico , Ciclo Celular/efectos de los fármacos , Fragmentación del ADN/efectos de los fármacos , Neoplasias Esofágicas/ultraestructura , Humanos , Microscopía Electrónica , Especies Reactivas de Oxígeno/metabolismo
13.
Int J Mol Med ; 9(4): 385-90, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11891533

RESUMEN

In order to explore the early apoptotic signal messengers and to search the apoptotic pathway, the morphological and functional changes of mitochondria were examined, and nitric oxide (NO) and calcium ions (Ca2+) were measured in the course of apoptosis in esophageal carcinoma cells induced by As2O3. The esophageal carcinoma cell line SHEEC1, established by HPV in synergy with TPA in our laboratory, were cultured with serum-free medium in a culture flask, 24-well plate and small culture chambers, and added with As2O3 at 1, 3, 5 micromol/l. After 0, 2, 4, 8, 12, 24 h of drug adding the NO were measured from extracellular cultured medium and the SHEEC1 cells were collected from flasks for electron microscopic examination. Fluorescent intensity (FI) of rhodamine 123 (Rho123) labeled cells was detected using laser confocal scanning microscope (LCSM) for evaluation of mitochondrial membrane potential. Intracellular Ca2+ of cells in small culture chambers labeled with Fluo-3 dye were measured using LCSM over time. After adding As2O3, SHEEC1 cells revealed characteristic morphological and functional changes of mitochondria such as hyperplasia, swelling and disruption, accompanying decrease of transmembrane potential (FI of Rho123 decreased). The Ca2+ level increased at once after adding As2O3 and the NO concentration increased step by step till 24 h, then apoptotic morphology of cells occurred. The results suggest that by inducement of As2O3 increasing Ca2+ and NO, the apoptotic signal messengers, will initiate the mitochondria-dependent apoptotic pathway.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Arsenicales/farmacología , Calcio/metabolismo , Mitocondrias/metabolismo , Óxido Nítrico/metabolismo , Óxidos/farmacología , Apoptosis/fisiología , Trióxido de Arsénico , Neoplasias Esofágicas/tratamiento farmacológico , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/ultraestructura , Humanos , Potenciales de la Membrana/fisiología , Mitocondrias/ultraestructura
14.
World J Gastroenterol ; 8(1): 40-3, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11833068

RESUMEN

AIM: To Quantitatively analyze the nitric oxide (NO) and Ca2+ in apoptosis of esophageal carcinoma cells induced by arsenic trioxide (As2O3). METHODS: The cell line SHEEC1, a malignant esophageal epithelial cell induced by HPV in synergy with TPA in our laboratory, was cultured in a serum-free medium and treated with As2O3. Before and after administration of As2O3, NO production in cultured medium was detected quantitatively using the Griess Colorimetric method. Intracellular Ca2+ was labeled by using the fluorescent dye Fluo3-AM and detected under confocal laser scanning microscope (CLSM), which was able to acquire data in real-time enabling Ca2+ dynamics of individual cells in vitro. The apoptotic cells were examined under electron microscopy. RESULTS: Intracellular concentration of Ca2+ increased from 1.00 units to 1.09-1.38 units of fluorescent intensity at As2O3 treatment and NO products subsequently released from As2O3-treated cells increased from 0.98-1.00 x10(-2)micromol x L(-1) up to 1.48-1.52 x10(-2)micromol x L(-1) and maintained in a high level continuously. Finally apoptosis of cells occurred,chromatin being agglutinated, cells shrunk,nuclei became round and mitochondria swelled. CONCLUSION: Ca2+ and NO increased with cell damage and apoptosis in cells treated by As2O3. The Ca2+ is an initial messenger to the apoptotic pathway. To investigate Ca2+ and NO will be a new direction for studying the apoptotic signaling messenger of the esophageal carcinoma cells induced by As2O3.


Asunto(s)
Apoptosis/fisiología , Arsenitos/farmacología , Calcio/metabolismo , Neoplasias Esofágicas , Óxido Nítrico/metabolismo , Teratógenos/farmacología , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Trióxido de Arsénico , Arsenicales/farmacología , Humanos , Microscopía Electrónica , Óxidos/farmacología , Células Tumorales Cultivadas/efectos de los fármacos , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/ultraestructura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA