Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Colloid Interface Sci ; 616: 81-92, 2022 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-35189506

RESUMO

Although photodynamic therapy (PDT) has been extensively studied as an established modality of cancer treatment, it still suffers from a few clinical limitations, such as skin phototoxicity and tumor hypoxia. To circumvent these hurdles, hollow silica mesoporous nanoparticles (HMSNs) loaded with photosensitizers were employed as the nanoplatform to construct multifunctional nanoparticles (NPs). Specifically, an ultra-uniform polydopamine (PDA) shell was highly controlled grown around HMSNs by photogenerated outwards-diffused 1O2, followed by conjugation of folic acid-poly(ethylene glycol) and chelation of Fe2+ ions. Thanks to the optimal thickness of light-absorbing PDA shell, the multifunctional NPs exhibited not only negligible skin phototoxicity but also efficient 1O2 generation and photothermal (PT)-enhanced •OH generation upon respective photoirradiation. Anti-tumor therapy was then performed on both 4 T1 tumor cells and tumor-bearing mice by the combination of 638 nm PDT and 808 nm PT-enhanced chemodynamic therapy (CDT). As a result, high therapeutic efficacy was achieved compared to single-modality therapy, with a cell inhibitory rate of 86% and tumor growth inhibition of 70.4% respectively. More interestingly, tumor metastasis was effectively inhibited by the synergetic treatment. These results convincingly demonstrate that our multifunctional NPs are very promising skin-safe PDT agents combined with CDT for efficient tumor therapy.


Assuntos
Nanopartículas , Neoplasias , Fotoquimioterapia , Animais , Linhagem Celular Tumoral , Indóis/farmacologia , Indóis/uso terapêutico , Camundongos , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Polímeros/uso terapêutico , Dióxido de Silício/uso terapêutico
2.
BMC Gastroenterol ; 21(1): 188, 2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33892648

RESUMO

BACKGROUND: Currently, there are no circulating diagnostic biomarkers for gastric neuroendocrine neoplasms (g-NENs). In previous studies, we found that miRNA-202-3p is overexpressed in the tumour tissue of type 1 g-NEN. We speculated that miRNA-202-3p is also likely to be highly expressed in circulating blood. METHODS: A total of 27 patients with type 1 g-NEN and 27 age- and sex-matched control participants were enrolled in this study. The miRNA-202-3p levels in serum obtained from the participants were measured by qRT-PCR. The expression level of miRNA-202-3p in the samples was calculated by comparison with a standard curve. RESULTS: The clinical characteristics of the patients were similar to those of the patient samples in previous reports. Expression of miRNA-202-3p was significantly higher in the patient group (3.84 × 107 copies/nl) than in the control group (0.635 × 107 copies/nl). The area under the ROC curve (AUC) was 0.878 (95% CI: 0.788-0.968), and the optimal cut-off point was approximately 1.12 × 107 copies/nl. The sensitivity and specificity were 88.9% and 77.8%, respectively. CONCLUSION: This study suggests that miRNA-202-3p is potentially useful as a biomarker of type 1 g-NEN; further investigation and verification should be performed in future research.


Assuntos
MicroRNAs/sangue , Tumores Neuroendócrinos , Neoplasias Gástricas , Biomarcadores , Biomarcadores Tumorais/genética , Perfilação da Expressão Gênica , Humanos , Tumores Neuroendócrinos/diagnóstico , Tumores Neuroendócrinos/genética , Curva ROC , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética
3.
Zhonghua Yi Xue Za Zhi ; 91(8): 554-9, 2011 Mar 01.
Artigo em Chinês | MEDLINE | ID: mdl-21418860

RESUMO

OBJECTIVE: To investigate the effect of inhibiting the expression of human suppressor of morphogenesis in genitalia-1 (hSMG-1) on chemosensitivity in human lung cancer H1299 cells. METHODS: Specialized small interference RNAs (siRNAs) of hSMG-1 were transfected into H1299 cells. The knockdown effect was evaluated by quantitative real-time reverse transcription-polymerase chain reaction (RT-PCR) and immunofluorescence. After the administration of anti-cancer drugs, the cell viable rate was determined by cell counting kit (CCK-8) and apoptotic rate was measured by Annexin V-FITC PI double staining on flow cytometry. The apoptotic related activated caspase 3 and caspase 9 were determined by colorimetric assay. RESULTS: The expression of hSMG-1 mRNA was significantly inhibited by hSMG-1 siRNA. And the inhibition rate of (73.8 ± 10.3)% was obtained (P < 0.01). The knockdown effect was further confirmed by immunofluorescence. The inhibition of hSMG-1 enhanced the sensitivity of H1299 cells to gemcitabine and cisplatin. The survival rates significantly decreased when the hSMG-1 siRNA transfected cells were treated with 10.0 mg/L gemcitabine and 10.0 mg/L cisplatin for 48 h respectively (0.51 ± 0.02 vs 0.69 ± 0.01, P < 0.01 and 0.34 ± 0.03 vs 0.48 ± 0.01, P < 0.01;all compared with control siRNA group). Annexin V-FITC PI double staining showed that, under the treatment of anti-cancer drugs, the apoptotic rate of H1299 cells was significantly increased by hSMG-1 knockdown [gemcitabine, (20.9 ± 3.4)% vs (12.0 ± 2.7)%, P < 0.05; cisplatin, (10.2 ± 1.8)% vs (4.5 ± 2.0)%, P < 0.05; all compared with control siRNA group]. Further study showed the inhibition of hSMG-1 up-regulated the activated caspase 3 and caspase 9 in the treated H1299 cells (gemcitabine, 0.1 mg/L, 48 h, caspase 3: 14.4 ± 3.8 vs 2.3 ± 0.4, P < 0.01; caspase 9: 15.5 ± 2.4 vs 4.2 ± 0.9, P < 0.01; cisplatin, 1.0 mg/L, 48 h, caspase 3: 18.8 ± 3.0 vs 6.5 ± 1.5, P < 0.01; caspase 9: 20.3 ± 4.2 vs 8.1 ± 2.0, P < 0.05; all compared with control siRNA group). CONCLUSION: The inhibition of hSMG-1 significantly enhances the sensitivity of human lung cancer H1299 cells to gemcitabine and cisplatin through an induction of more apoptotic cells. And the activation of mitochondrial apoptotic pathway is involved.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/genética , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Serina-Treonina Quinases , Interferência de RNA , RNA Interferente Pequeno/genética
4.
Oncol Rep ; 25(4): 953-62, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21234522

RESUMO

Current treatment modalities for melanoma do not offer satisfactory efficacy. We have developed a new, minimally invasive hyperthermia technology based on radio-frequency hyperthermia. Herein, we investigated the feasibility of using a nickel-copper thermoseed for inductive hyperthermia at a relatively high temperature (46-55 ˚C). In vitro, the thermoseed showed good thermal effects and effective killing of B16/F10 melanoma cells. Temperatures of 53.1 ± 0.5 ˚C were achieved for a single thermoseed and 56.5 ± 0.5 ˚C for two in parallel (spacing 5 mm). No B16/F10 melanoma cells survived with heating time longer than 20 min in the parallel thermoseed group. Magnetic fields or thermoseeds alone did not affect the survival rate of B16/F10 cells (P>0.05). In vivo, B16/F10 melanoma cells were subcutaneously injected into the right axilla of C57BL/6 mice. After the tumors grew to ~11-13 mm, two thermoseeds (spacing 5 mm) were implanted into the tumors and the mice were subjected to an alternating magnetic field (100-250 kHz, 15 kA/m) to induce hyperthermia. The temperature at the center of the tumor reached 46 ˚C at 5 min and plateaued at 50 ˚C. Thermoseed treatment produced large necrotic areas, inhibited tumor growth in 60% (6 of 10) of animals and prolonged survival time (P<0.05). Thus, with further optimization and testing, high-temperature thermoseed inductive hyperthermia may have therapeutic potential for melanoma.


Assuntos
Hipertermia Induzida/instrumentação , Magnetoterapia , Melanoma Experimental/terapia , Animais , Cobre , Estudos de Viabilidade , Temperatura Alta , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Níquel , Aço Inoxidável , Taxa de Sobrevida , Células Tumorais Cultivadas
5.
Pancreas ; 40(1): 55-60, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20871480

RESUMO

OBJECTIVES: Human primary microRNA-155/B-cell integration cluster (BIC) transcript is the precursor of microRNA-155. The overexpression of them has been widely observed in the progression of various types of tumors. Our objective was to investigate the effect of anticancer agents on the expression of BIC and possible signal pathways that involved in. METHODS: Quantitative real-time reverse transcriptase polymerase chain reaction was used to measure the expression of BIC. Chemical inhibitors against c-Jun N-terminal kinase 1, mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2, protein kinase C, checkpoint kinase 1, and phosphatidylinositol 3 kinase (PI3K) were used for the evaluation of involved signal pathways. RNA interference was used to knock down the expression of ataxia-telangiectasia mutated, ataxia-telangiectasia and Rad3 related, and suppressor of morphogenesis in genitalia-1 (SMG-1), and Western blot was carried out to evaluate the knockdown effect. RESULTS: B-cell integration cluster expression was induced by a representative anti-pancreatic cancer drug, gemcitabine, in human pancreatic cancer PANC-1 cells. The mitogen-activated protein kinase/extracellular signal-regulated kinase kinase 1/2 and c-Jun N-terminal kinase inhibitors, but not the checkpoint kinase 1 and protein kinase C inhibitors, suppressed the up-regulation of BIC. B-cell integration cluster up-regulation was also significantly inhibited by the PI3K inhibitor wortmannin. RNA interference studies showed that wortmannin-sensitive SMG-1 but not ataxia-telangiectasia mutated or ataxia-telangiectasia and Rad3 related was involved in the up-regulation. CONCLUSIONS: Our results show that multiple pathways can be involved in the up-regulation of BIC. Furthermore, we demonstrate for the first time that PI3K SMG-1 is required for gemcitabine-induced up-regulation of BIC transcript.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Desoxicitidina/análogos & derivados , MicroRNAs/genética , Neoplasias Pancreáticas/tratamento farmacológico , Fosfatidilinositol 3-Quinases/fisiologia , Androstadienos/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia , Proteínas de Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/fisiologia , Desoxicitidina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/fisiologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Regulação para Cima , Wortmanina , Gencitabina
6.
Int J Oncol ; 37(3): 583-93, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20664927

RESUMO

The Family with sequence similarity 107 (FAM107) possesses an N-terminal domain of unknown function (DUF1151) that is highly conserved beyond species. In human, FAM107A termed TU3A/DRR1 has been reported as a candidate tumor suppressor gene which expression is downregulated in several types of cancer, however no studies have investigated the other family protein, FAM107B. In the present study, we designated FAM107B as heat shock-inducible tumor small protein (HITS) and studied its expression and functional properties in cancer. HITS is an 18-kDa nuclear protein expressed in a variety of tissues including stomach, colon, lung and lymphoid organs. In human gastric and colorectal cancers and a mouse model of colon cancer, its expression in tumor cells was much lower than normal epithelial cells, while expression pattern and intensity varied among different histological types of cancer. In functional analysis in vitro, forced expression of this protein suppresses the cellular responses to growth factors. Furthermore, HITS gene carries the promoter region providing heat shock transcription factor (HSF) binding sites and amplifying the transcription of HITS by heat shock or hyperthermia treatment both in vitro and in vivo. Thus HITS would be a potential tumor suppressor gene similar to TU3A containing heat responding elements, which contrasts with previously described oncogenic activities of other heat shock proteins such as HSP70 and HSP90.


Assuntos
Proteínas de Choque Térmico/biossíntese , Resposta ao Choque Térmico , Neoplasias/metabolismo , Animais , Modelos Animais de Doenças , Células HEK293 , Células HeLa , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Imuno-Histoquímica , Células Jurkat , Masculino , Camundongos , Camundongos Endogâmicos ICR , Neoplasias/genética , Transfecção
7.
Zhonghua Yi Xue Za Zhi ; 90(2): 123-7, 2010 Jan 12.
Artigo em Chinês | MEDLINE | ID: mdl-20356498

RESUMO

OBJECTIVE: To investigate the effect of anti-cancer drugs on the expression of B-cell integration cluster (BIC) RNA/miRNA-155 in human pancreatic cancer PANC-1 cells. METHODS: PANC-1 cells were treated with different concentrations of anti-cancer drugs. Total RNA of the treated cells were harvested and the expression levels of BIC RNA and mature miR-155 were quantified by using Taqman FAM/MGB probes on a real-time PCR system. Relative quantification was carried out using the DeltaDeltaCt method. A PI3K-related kinases inhibitor was used to determine whether these kinases were involved in the regulation of BIC RNA. RESULTS: The expression of BIC RNA was strongly induced by anti-cancer drugs. When PANC-1 cells were treated by gemcitabine with concentrations of 1.0, 2.5, 5.0, 10.0 mg/L for 48 h and 72 h, the level of BIC RNA (48 h: 37.1 +/- 4.1, 29.0 +/- 5.7, 21.0 +/- 7.6, 40.4 +/- 9.0, 72 h: 27.7 +/- 3.1, 43.1 +/- 1.2, 31.8 +/- 5.4, 23.1 +/- 1.4) were significantly higher than that of the control (48 h: 1.6 +/- 1.1, 72 h: 1.0 +/- 0.1, all P < 0.05). 5-FU (10 mg/L, 48 h) and bleomycin (100 mg/L, 48 h) also induced BIC RNA up-regulation (5.2 +/- 1.1 vs 1.7 +/- 0.7, 11.5 +/- 0.7 vs 1.7 +/- 0.7, both P < 0.05). When PANC-1 cells treated with 1.0, 2.5, 5.0, 10.0 mg/L gemcitabine for 72 h, the level of miR-155 (2.21 +/- 0.40, 1.86 +/- 0.03, 2.47 +/- 0.04, 3.24 +/- 0.05) also higher than that of the control (1.11 +/- 0.09, P < 0.05), while no change was observed when the cells only treated for 48 h. Further study showed gemcitabine-induced BIC RNA up-regulation was inhibited by wortmannin, a specific PI3K inhibitor, the expression levels of BIC RNA of 1 micromol/L wortmannin + 5 mg/L gemcitabine group and 10 micromol/L wortmannin + 5 mg/L gemcitabine group were 5.34 +/- 1.11 and 1.26 +/- 0.07, lower than that of 5 mg/L gemcitabine group (11.82 +/- 3.11, P < 0.05). CONCLUSIONS: BIC RNA is strongly induced by anti-cancer drugs in PANC-1 cells and the levels of miR-155 also slightly increase. PI3K pathway is involved in gemcitabine-induced BIC RNA up-regulation.


Assuntos
Antineoplásicos/farmacologia , MicroRNAs/genética , Neoplasias Pancreáticas/genética , Antineoplásicos/uso terapêutico , Linfócitos B , Linhagem Celular Tumoral , Expressão Gênica , Humanos , Família Multigênica , Neoplasias Pancreáticas/tratamento farmacológico
8.
Biomed Mater ; 1(4): 198-201, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18458406

RESUMO

The magnetite (Fe(3)O(4)) nanoparticles were prepared by coprecipitation of Fe(3+) and Fe(2+) with an aqueous NaOH solution. The Fe(3)O(4)/polyaniline (PANI) magnetic composite nanoparticles with a core-shell structure with a diameter of 30-50 nm were prepared via an in situ polymerization of aniline in an aqueous solution containing the Fe(3)O(4) magnetic fluid. The inductive heat property of Fe(3)O(4)/PANI composite nanoparticles in an alternating current (ac) magnetic field was investigated. The potential of Fe(3)O(4)/PANI nanoparticles was evaluated for localized hyperthermia treatment of cancers. The saturation magnetization, M(s), and coercivity, H(c), are 50.05 emu g(-1) and 137 Oe for Fe(3)O(4) nanoparticles and 26.34 emu g(-1) and 0 Oe for Fe(3)O(4)/PANI composite nanoparticles, respectively. Exposed in the ac magnetic field for 29 min, the temperatures of physiological saline suspensions containing Fe(3)O(4) nanoparticles or Fe(3)O(4)/PANI composite nanoparticles are 63.6 degrees C and 52.4 degrees C, respectively. The Fe(3)O(4)/PANI composite nanoparticles would be useful as good thermoseeds for localized hyperthermia treatment of cancers.


Assuntos
Compostos Férricos/química , Compostos Férricos/uso terapêutico , Hipotermia Induzida/instrumentação , Hipotermia Induzida/métodos , Teste de Materiais , Nanopartículas/química , Nanopartículas/uso terapêutico , Campos Eletromagnéticos , Compostos Férricos/efeitos da radiação , Temperatura Alta , Nanopartículas/efeitos da radiação
9.
Ai Zheng ; 24(9): 1148-50, 2005 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-16159444

RESUMO

BACKGROUND & OBJECTIVE: Ferromagnet has thermal effect in alternating magnetic field (AMF). Magnetically mediating hyperthermia is to localize magnetic substance within tumor tissue under AMF to promote the targeting ability and heat distribution of hyperthermia. This study was to develop a new method of showing the thermal effect of iron oxide Fe3O4 nanoparticles in AMF in vitro. METHODS: Melted polyethelene glycol (PEG) was dropped on a slide to form crystal monolayer after cooling, and was covered with a bipartite blood coverslip. Certain amount of Fe3O4 powder (average diameter, 10 nm) was added in the gap of the blood coverslip to form a strip wandering on the slide with the width of 0.5-1.0 mm. This apparatus, named analog vessel bed, was then alternated in a 4.6 mT AMF at 100-250 kHz for 15 min. The changes of PEG crystal images were observed under micropolariscope. The thermal range of the heated Fe3O4 powder was affirmed according to melting status and melting point value of PEG crystal. RESULTS: In AMF, 5 mg of Fe3O4 could rise the temperature to above 45 degrees C-50 degrees C, which exceeded the required hyperthermic temperature 43 degree C; 15 mg of Fe3O4 could rise the temperature to no more than 59 degrees C-61 degrees C; while 25 mg of Fe3O4 could be heated up to 59 degrees C-61 degrees C. The PEG melting area was enlarged with the increasing Fe3O4 quantity or decreasing PEG melting point. CONCLUSION: The analog vessel bed apparatus could display the thermal effect of Fe3O4 nanoparticles in AMF, and would be helpful for further studies on effect of magnetically mediated hyperthermia on cancer cells.


Assuntos
Campos Eletromagnéticos , Compostos Férricos , Temperatura Alta , Hipertermia Induzida/métodos , Nanopartículas , Polietilenoglicóis , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...