Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
J Cell Sci ; 134(8)2021 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-33758077

RESUMO

Autophagy is considered to be an important switch for facilitating normal to malignant cell transformation during colorectal cancer development. Consistent with other reports, we found that the membrane receptor Neuropilin1 (NRP1) is greatly upregulated in colon cancer cells that underwent autophagy upon glucose deprivation. However, the mechanism underlying NRP1 regulation of autophagy is unknown. We found that knockdown of NRP1 inhibits autophagy and largely upregulates the expression of aldo-keto reductase family 1 B10 (AKR1B10). Moreover, we demonstrated that AKR1B10 interacts with and inhibits the nuclear importation of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and then subsequently represses autophagy. Interestingly, we also found that an NADPH-dependent reduction reaction could be induced when AKR1B10 interacts with GAPDH, and the reductase activity of AKR1B10 is important for its repression of autophagy. Together, our findings unravel a novel mechanism of NRP1 in regulating autophagy through AKR1B10.


Assuntos
Aldeído Redutase , Neoplasias do Colo , Aldeído Redutase/genética , Aldo-Ceto Redutases , Autofagia , Neoplasias do Colo/genética , Glucose , Gliceraldeído-3-Fosfato Desidrogenases , Humanos
2.
Mol Pharm ; 19(7): 2607-2619, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35485954

RESUMO

Chlorin e6-C-15-ethyl ester (LS-HB), a newly identified photosensitizer, was isolated from chlorin e6. The mechanism of tumor cell death induced by photodynamic therapy with LS-HB (LS-HB-PDT) is still unknown. Here, we investigated the photophysical properties of LS-HB, evaluated the antitumor effect on melanoma in vitro and in vivo, and explored its possible mechanisms. LS-HB not only has an optimal spectral band of red wavelength (660 nm) for photosensitization but also has favorable photostability. More importantly, LS-HB-PDT elicited a potent dose-dependent phototoxic effect in vitro. We discovered that LS-HB located in the mitochondria of B16F10 cells was able to generate excess reactive oxygen species, which subsequently resulted in mitochondrial membrane potential loss and induced apoptosis via caspase-9 and caspase-3 pathways. Moreover, PDT with LS-HB markedly inhibited the growth of melanoma in vivo. Therefore, LS-HB is expected to be an effective potential photosensitizer in antitumor therapy.


Assuntos
Melanoma , Fotoquimioterapia , Porfirinas , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Melanoma/tratamento farmacológico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/farmacologia , Porfirinas/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo
3.
Mol Pharm ; 19(5): 1356-1367, 2022 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-35420039

RESUMO

Selectively inducing tumor thrombosis and subsequent necrosis is a novel and promising antitumor strategy. We have previously designed a targeting procoagulant protein, called tTF-EG3287, which is a fusion of a truncated tissue factor (tTF) with EG3287, a short peptide against the neuropilin-1 (NRP1) binding site of vascular endothelial growth factor-A 165 (VEGF-A 165). However, off-target effects and high-dose requirements limit the further use of tTF-EG3287 in antitumor therapy. Therefore, we encapsulated tTF-EG3287 into poly(2-ethyl-2-oxazoline)-distearoyl phosphatidyl ethanolamine (PEOz-DSPE)-modified liposomes to construct pH-responsive liposomes as a novel vascular embolization agent, called tTF-EG3287@Liposomes. The liposomes had an average particle size of about 100 nm and showed considerable drug-loading capacity, encapsulation efficiency, and biocompatibility. Under the stimulation of acidic microenvironments (pH 6.5), the lipid membrane of tTF-EG3287@Liposomes collapsed, and the cumulative drug release rate within 72 h was 83 ± 1.26%. When administered to a mouse model of hepatocellular carcinoma (HCC), tTF-EG3287@Liposomes showed prolonged retention and enhanced accumulation in the tumor as well as a superior antitumor effec, compared with tTF-EG3287. This study demonstrates the potential of tTF-EG3287@Liposomes as a novel embolic agent for solid tumors and provides a new strategy for tumor-targeted infarction therapy.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/tratamento farmacológico , Linhagem Celular Tumoral , Concentração de Íons de Hidrogênio , Lipossomos/química , Neoplasias Hepáticas/tratamento farmacológico , Camundongos , Tromboplastina , Microambiente Tumoral , Fator A de Crescimento do Endotélio Vascular
4.
Microb Pathog ; 130: 213-218, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30862559

RESUMO

OBJECTIVES: The host immune response could be an imperative factor in the pathogenesis of neurosyphilis, but the role of T lymphocyte subsets remains unclear. In the present study, we assessed the CD4+ T and CD8+ T cell subsets in the peripheral blood of patients with HIV-negative symptomatic neurosyphilis and then explored the clinical application value of neurosyphilis. METHODS: In total, 24 patients with HIV-negative symptomatic neurosyphilis and 22 patients with syphilis/non-neurosyphilis were included in this study and cerebrospinal fluid (CSF) and blood samples were obtained. Th1, Th2, Th17, Th9, CD8+IFN-γ+, CD8+IL-4+, CD8+IL-9+, and CD8+IL-17 + cells were identified by flow cytometry. RESULTS: The levels of CD8+IFN-γ+ were significantly increased in the peripheral blood of neurosyphilis patients compared to that in syphilis/non-neurosyphilis patients, but it was opposite to Th2, Th9, CD8+IL-4+, CD8+IL-9+, and CD8+IL-17 + cells. Dendritic cells (DCs) of neurosyphilis matured by T. pallidum induced the development of a combination of IFN-γ-producing Th1 cells. The number of CD8+IL-17 + cells was significantly correlated with the CSF RPR and CSF TPPA levels. ROC curve analysis revealed that the number of CD8+IFN-γ+ cells could be a potential biomarker for neurosyphilis from non-neurosyphilis/syphilis. CONCLUSIONS: High expression of CD8+IFN-γ+ cells and low expression of CD8+IL-17 + cells in patients with symptomatic neurosyphilis, which explains the pathogenesis of symptomatic neurosyphilis, meanwhile CD8+IFN-γ+ cells may be a better indicator in classifying symptomatic neurosyphilis from non-neurosyphilis/syphilis among patients without HIV infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Neurossífilis/patologia , Subpopulações de Linfócitos T/imunologia , Adulto , Idoso , Células Sanguíneas , Líquido Cefalorraquidiano/citologia , Feminino , Citometria de Fluxo , Humanos , Masculino , Pessoa de Meia-Idade
5.
Anticancer Drugs ; 30(5): 441-450, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30807552

RESUMO

In recent decades, selectively inducing tumor vascular thrombosis, followed by necrosis of tumor tissues has been a promising and potential anticancer strategy. In this report, we prepared a kind of vascular targeting drug that consists of anti-neuropilin-1 monoclonal antibody (anti-NRP-1 mAb) and truncated tissue factor (tTF). Anti-NRP-1 mAb could guide tTF to the surface of tumor vascular endothelial cells and lead to subsequent vascular embolization. This vascular targeting drug, which is also one of the antibody drug conjugates, was generated using a coupling method with water-soluble 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide and N-hydroxysulfosuccimide. Afterwards, in-vitro and in-vivo assays were performed to characterize its potential coagulation ability and antitumor activity. In-vitro experiments indicated that tTF-anti-NRP-1 monoclonal antibody (tTF-mAb) retained both the targeting activity of anti-NRP-1 mAb and the procoagulant activity of tTF. Live imaging system was used to assess its biodistribution and tumor-binding capability, which also yielded promising results. Furthermore, in-vivo studies showed that tTF-mAb was capable of significantly inducing tumor vascular thrombosis and inhibiting tumor growth in nude mice bearing subcutaneous xenografts, and histopathologic changes were rarely observed in normal organs.


Assuntos
Anticorpos Monoclonais/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Neuropilina-1/imunologia , Tromboplastina/imunologia , Trombose/prevenção & controle , Animais , Anticorpos Monoclonais/biossíntese , Anticorpos Monoclonais/imunologia , Apoptose , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Proliferação de Células , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Trombose/imunologia , Trombose/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Clin Lab Anal ; 33(4): e22850, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30758083

RESUMO

BACKGROUND: Neuropilins (Nrps) are a new type of broad-spectrum tumor marker. Currently, a method for accurate simultaneous quantification of Nrps is not available. We aimed to develop a bead-based and duplexed flow cytometric assay that could be used for accurate and simultaneous quantification of Nrp1 and Nrp2 for scientific research or clinical diagnosis. METHODS: We coupled anti-human Nrp1-11# mAb and anti-human Nrp2-C3 mAb to magnetic beads 18# and 25#, respectively. Capturing antibodies and detecting antibodies were then combined to detect Nrps by a bead-based Luminex assay, which was subsequently applied to quantify Nrps in clinical serum samples. RESULTS: The results showed that the detection value of Nrps ranged from 10 to 100 000 pg/mL for Nrp1 and from 25 to 100 000 pg/mL for Nrp2. The detection sensitivity reached 10 pg/mL for Nrp1 and 24.8 pg/mL for Nrp2. Intra-assay variances ranged from 1.0% to 2.6% for Nrp1 and from 2.9% to 4.0% for Nrp2, and interassay variances ranged from 1.5% to 6.4% for Nrp1 and from 4.2% to 8.1% for Nrp2. The Nrp1 and Nrp2 recoveries were 96.6%-103.6% and 95.6%-102.3%, respectively. Irrelevant antigens had no interference in the paired-detection system, and the mean fluorescence intensity (MFI) values were stable for months. CONCLUSION: A bead-based, duplexed flow cytometric assay (xMAP® technology) was developed to detect Nrp1 and Nrp2. The assay provided rapid, high-throughput results and was much more sensitive, specific, reproducible, and stable than existing assays. In addition, this assay could be applied in early-stage cancer screening, tumor malignancy analysis, and prognosis assessment.


Assuntos
Imunoensaio/métodos , Neuropilina-1/sangue , Neuropilina-2/sangue , Anticorpos Monoclonais , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/imunologia , Biotinilação , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoensaio/instrumentação , Neoplasias/sangue , Neuropilina-1/imunologia , Neuropilina-2/imunologia , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
7.
Int J Cancer ; 143(3): 635-644, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29457830

RESUMO

Glioblastoma multiforme (GBM) is the most lethal brain malignancy which involves multi-gene abnormality. Unfortunately, effective therapy against GBM remains lacking. Previously, we found that NRP-1 and its downstream NRP-1/GIPC1 pathway played an important role in GBM. In our study, we further investigated the upstream signaling of NRP-1 to understand how it is regulated. First, we identified that hsa-miR-124-3p was miRNA differentially expressed in GBM and in normal brain tissues by high-throughput sequencing. Then, by dual luciferase reporter gene, we found miR-124-3p can specially bind to the 3'UTR region of the NRP-1 thus suppresses its expression. Moreover, miR-124-3p overexpression significantly inhibited GBM cell proliferation, migration and tumor angiogenesis which resulted in GBM apoptosis and cell cycle arrest, putatively via NRP-1 mediated PI3K/Akt/NFκB pathways activation in GBM cells. Meanwhile, miR-124-3p overexpression also suppressed tumor growth and reduced tumor angiogenesis when targeted by NRP-1 in a PDX model. Furthermore, NRP-1 mAb exerted synergistic inhibitory effects with miR-124-3p overexpression in GBM. Thus, we discovered that miR-124-3p acts as the upstream suppressor of NRP-1 which promotes GBM cell development and growth by PI3K/Akt/NFκB pathway. The miR-124-3p/NRP-1/GIPC1 pathway as a new pathway has a vital role in GBM, and it could be considered as the potential target for malignant gliomas in future.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Neovascularização Patológica/genética , Neuropilina-1/genética , Interferência de RNA , Regiões 3' não Traduzidas , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose/genética , Encéfalo/metabolismo , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Perfilação da Expressão Gênica , Glioblastoma/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , MicroRNAs/genética , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Proteína Oncogênica v-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Transcriptoma
8.
J Nanobiotechnology ; 16(1): 90, 2018 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-30424776

RESUMO

BACKGROUND: In the last decade, the biosynthesis of metal nanoparticles using organisms have received more and more considerations. However, the complex composition of organisms adds up to a great barrier for the characterization of biomolecules involved in the synthesis process and their biological mechanisms. RESULTS: In this research, we biosynthesized a kind of flower-shaped Au nanoclusters (Au NCs) using one definite component-epigallocatechin gallate (EGCG), which was the main biomolecules of green tea polyphenols. Possessing good stability for 6 weeks and a size of 50 nm, the Au NCs might be a successful candidate for drug delivery. Hence, both methotrexate (MTX) and doxorubicin (DOX) were conjugated to the Au NCs through a bridge of cysteine (Cys). The introduction of MTX provided good targeting property for the Au NCs, and the conjugation of DOX provided good synergistic effect. Then, a novel kind of dual-drug loaded, tumor-targeted and highly efficient drug delivery system (Au-Cys-MTX/DOX NCs) for combination therapy was successfully prepared. The TEM of HeLa cells incubated with Au-Cys-MTX/DOX NCs indicated that the Au-Cys-MTX/DOX NCs could indeed enter and kill cancer cells. The Au-Cys-MTX/DOX NCs also possessed good targeting effect to the FA-receptors-overpressed cancer cells both in vitro and in vivo. Importantly, the Au-Cys-MTX/DOX NCs resulted in an excellent anticancer activity in vivo with negligible side effects. CONCLUSIONS: These results suggest that the biosynthesized Au-Cys-MTX/DOX NCs could be a potential carrier with highly efficient anticancer properties for tumor-targeted drug delivery.


Assuntos
Antineoplásicos/química , Catequina/análogos & derivados , Portadores de Fármacos/química , Ouro/química , Nanopartículas Metálicas/química , Animais , Antineoplásicos/farmacologia , Catequina/química , Sobrevivência Celular/efeitos dos fármacos , Preparações de Ação Retardada , Doxorrubicina/química , Doxorrubicina/farmacologia , Liberação Controlada de Fármacos , Células HeLa , Células Hep G2 , Humanos , Metotrexato/química , Metotrexato/farmacologia , Camundongos Endogâmicos BALB C , Tamanho da Partícula , Propriedades de Superfície
9.
Tumour Biol ; 37(10): 13777-13788, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27481513

RESUMO

Glioma occurs due to multi-gene abnormalities. Neuropilin-1 (NRP-1), as a transmembrane protein, involves in glioma proliferation, invasion, and migration, as well as tumor angiogenesis. The cytoplasmic protein, GAIP/RGS19-interacting protein (GIPC1), could regulate the clathrin-vesicles trafficking and recycling. Here, we show that NRP-1 co-localizes and co-immunoprecipitates with GIPC1, and the C-terminal SEA-COOH motif of NRP-1 interacts specially with the named from three proteins: PSD-95 (a 95 kDa protein involved in signaling at the post-synaptic density), DLG (the Drosophila melanogaster Discs Large protein) and ZO-1 (the zonula occludens 1 protein involved in maintenance of epithelial polarity) (PDZ) domain of GIPC1 in glioma cells. Knockdown of GIPC1 by small interfering RNA (siRNA) significantly reduces the proliferation and invasion of glioma cells in vitro and increases its apoptosis. Furthermore, si-GIPC1 prevents the action of adaptor proteins adaptor protein, phosphotyrosine interaction, PH domain and leucine zipper containing 1 (APPL1) and p130Cas and inhibits the downstream kirsten rat sarcoma viral oncogene homolog (KRAS)-ERK signaling pathway. This study demonstrated that NRP-1/GIPC1 pathway plays a vital role in glioma progression, and it is a potential important target for multi-gene combined therapeutics.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Glioma/patologia , Neuropilina-1/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Western Blotting , Movimento Celular , Proliferação de Células , Progressão da Doença , Citometria de Fluxo , Imunofluorescência , Glioma/metabolismo , Humanos , Técnicas Imunoenzimáticas , Imunoprecipitação , Invasividade Neoplásica , Ligação Proteica , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais , Células Tumorais Cultivadas
10.
Anticancer Drugs ; 25(6): 663-72, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24583771

RESUMO

Neuropilin-1 (NRP-1) is a nontyrosine kinase coreceptor for semaphorin 3A and the vascular endothelial growth factor involved in tumor angiogenesis, growth, and metastasis and is regarded as a promising target for cancer therapy. In the present study, we investigated the effects of an anti-NRP-1 monoclonal antibody (mAb) that we generated for MCF7 breast cancer cellular adhesion studies. MTT, colony formation, and adhesion assays showed that our anti-NRP-1 mAb dose-dependently inhibited MCF7 proliferation and fibronectin adhesion, leading to a rounded cellular morphology. Further, rhodamine phalloidin stain revealed that fibronectin-dependent formation of actin stress fibers was inhibited by anti-NRP-1 mAb. Immunoprecipitation and western blot showed that anti-NRP-1 mAb treatment inhibited the formation of NRP-1-α5ß1 integrin complexes and suppressed the phosphorylation of focal adhesion kinase and p130cas in MCF7 cells. These findings contribute to further understanding the NRP-1 function in cell adhesion and tumor metastasis. Moreover, our anti-NRP-1 mAb is a prospective drug candidate for tumor treatment.


Assuntos
Anticorpos Monoclonais/farmacologia , Neoplasias da Mama/patologia , Adesão Celular/efeitos dos fármacos , Proteína Substrato Associada a Crk/metabolismo , Fibronectinas/fisiologia , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Neuropilina-1/metabolismo , Actinas/metabolismo , Anticorpos Monoclonais/isolamento & purificação , Feminino , Humanos , Células MCF-7 , Neuropilina-1/imunologia , Transdução de Sinais
11.
Lasers Med Sci ; 28(6): 1541-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23377406

RESUMO

Photodynamic therapy (PDT) has been clinically used for liver cancer. The pharmacokinetics of a photosensitizer needs to be monitored so that PDT can be performed at the most favorable time and with the proper dose to increase the cure rate. As mTHPC is a fluorescent compound, we investigate its pharmacokinetics, distribution, and elimination in the rat orthotropic liver cancer model in order to confirm an optimal treatment opportunity of liver cancer PDT. After intravenous administration at a single dose of 300 µg/kg, mTHPC was extracted from tissue homogenates or plasma. Then, mTHPC concentrations were assessed by fluorescence spectroscopy and the data were processed with PK-GRAPH pharmacokinetic procedure. The plasma concentration-time profile of mTHPC showed a short distribution half-life (T½α = 0.082 h) and a relatively longer elimination half-life (T½ß = 28.23 h), which quite fitted with a two-compartment model. The results of mTHPC tissue distributions showed that the highest drug accumulation was in tumor tissue, and successively decreased in liver, heart, spleen, muscle, and skin tissues. The drug distribution ratio of tumor to normal tissue reached the peak at 24 h after mTHPC administration. mTHPC was eliminated at a suitable rate in rat orthotropic liver cancer model, and there was no long-term accumulation of mTHPC in rat tissues. For PDT of orthotropic liver cancer, 24 h after mTHPC intravenous injection may be the optimal treatment time point, which might provide higher clinical efficacy and reduce side effects.


Assuntos
Neoplasias Hepáticas Experimentais/tratamento farmacológico , Mesoporfirinas/uso terapêutico , Fotoquimioterapia , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Carcinoma 256 de Walker/tratamento farmacológico , Carcinoma 256 de Walker/metabolismo , Linhagem Celular Tumoral , Meia-Vida , Injeções Intravenosas , Neoplasias Hepáticas Experimentais/metabolismo , Masculino , Mesoporfirinas/administração & dosagem , Mesoporfirinas/farmacocinética , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/farmacocinética , Ratos , Ratos Wistar , Espectrometria de Fluorescência , Distribuição Tecidual
12.
ScientificWorldJournal ; 2013: 637086, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23861656

RESUMO

PURPOSE: Truncated tissue factor (tTF) fusion protein targeting tumor vasculature can induce tumor vascular thrombosis and necrosis. Here, we generated (RGD)3-tTF in which three arginine-glycine-aspartic (RGD) targeting integrin α(v)ß3 and tTF induce blood coagulation in tumor vessels. METHODS: The bioactivities of (RGD)3-tTF including coagulation activity, FX activation, and binding with integrin α(v)ß3 were performed. The fluorescent labeled (RGD)3-tTF was intravenously injected into tumor-bearing mice and traced in vivo. The tumor growth, volume, blood vessel thrombosis, tumor necrosis, and survival time of mice treated with (RGD)3-tTF were evaluated. RESULTS: The clotting time and FX activation of (RGD)3-tTF were similar to that of TF (P > 0.05) but different with that of RGD (P < 0.05). (RGD)3-tTF presented a higher binding with α(v)ß3 than that of RGD and TF at the concentration of 0.2 µmol/L (P < 0.05). (RGD)3-tTF could specifically assemble in tumor and be effective in reducing tumor growth by selectively inducing tumor blood vessels thrombosis and tumor necrosis which were absent in mice treated with RGD or TF. The survival time of mice treated with (RGD)3-tTF was higher than that of mice treated with TF or RGD (P < 0.05). CONCLUSION: (RGD)3-tTF may be a promising strategy for the treatment of colorectal cancer.


Assuntos
Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Integrina alfaVbeta3/metabolismo , Neovascularização Patológica/tratamento farmacológico , Neovascularização Patológica/metabolismo , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico , Inibidores da Angiogênese/uso terapêutico , Animais , Linhagem Celular Tumoral , Neoplasias Colorretais/irrigação sanguínea , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Resultado do Tratamento
13.
Adv Healthc Mater ; 12(8): e2202418, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36459700

RESUMO

The hypoxic character of tumors and the poor targeting ability of photosensitizers often limit the efficacy of photodynamic therapy (PDT). In recent years, the discovery of metal nanoenzymes and nanocarriers has improved PDT. Thereby, to improve the effective utilization of photosensitizers and oxygen (O2 ) in tumors, herein, a nanosystem (LS-HB@HvCeO2 -NRP1 mAb, LHCN1) is reported, in which a hollow virus-like cerium oxide (HvCeO2 ) is surface-decorated with tumor-targeting neuropilin-1 monoclonal antibody (NRP1 mAb), and loaded with a photosensitizer (chlorin e6-C-15-ethyl ester, LS-HB). In vitro and in vivo experiments demonstrate that LHCN1 can efficiently accumulate within the tumor sites via the targeting guidance of NRP1 mAb and is then rapidly endocytosed into cells. Furthermore, HvCeO2 with catalase-mimetic activity can decompose the endogenous hydrogen peroxide (H2 O2 ) to promote O2 via the valence transformation between Ce4+ and Ce3+ , relieving tumor hypoxia and improving the PDT efficacy. Upon near-infrared laser irradiation, LS-HB produces large amounts of cytotoxic reactive oxygen species. Moreover, LHCN1 is used in fluorescence/photoacoustic multimodal imaging for in vivo drug localization, and its use in PDT evidently helps inhibit tumor growth with no apparent toxicity to normal tissues. Thus, LHCN1 may provide a promising strategy for precise tumor-specific diagnosis and treatment.


Assuntos
Nanopartículas , Fotoquimioterapia , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fotoquimioterapia/métodos , Vírus Oncogênicos , Linhagem Celular Tumoral , Peróxido de Hidrogênio , Oxigênio
14.
Oncol Res ; 32(2): 361-371, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38186571

RESUMO

The high mortality rate associated with gastric cancer (GC) has resulted in an urgent need to identify novel therapeutic targets for GC. This study aimed to investigate whether GAIP interacting protein, C terminus 1 (GIPC1) represents a therapeutic target and its regulating mechanism in GC. GIPC1 expression was elevated in GC tissues, liver metastasis tissues, and lymph node metastases. GIPC1 knockdown or GIPC1 blocking peptide blocked the platelet-derived growth factor receptor (PDGFR)/PI3K/AKT signaling pathway, and inhibited the proliferation and migration of GC cells. Conversely, GIPC1 overexpression markedly activated the PDGFR/PI3K/AKT signaling pathway, and promoted GC cell proliferation and migration. Furthermore, platelet-derived growth factor subunit BB (PDGF-BB) cytokines and the AKT inhibitor attenuated the effect of differential GIPC1 expression. Moreover, GIPC1 silencing decreased tumor growth and migration in BALB/c nude mice, while GIPC1 overexpression had contrasting effects. Taken together, our findings suggest that GIPC1 functions as an oncogene in GC and plays a central role in regulating cell proliferation and migration via the PDGFR/PI3K/AKT signaling pathway.


Assuntos
Neoplasias Gástricas , Humanos , Animais , Camundongos , Neoplasias Gástricas/genética , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Camundongos Nus , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal
15.
Adv Sci (Weinh) ; 10(30): e2303872, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37661565

RESUMO

The lethality and chemotherapy resistance of pancreatic cancer necessitates the urgent development of innovative strategies to improve patient outcomes. To address this issue, we designed a novel drug delivery system named GDMCN2,which uses iron-based metal organic framework (Fe-MOF) nanocages encased in a covalent organic framework (COF) and modified with the pancreatic cancer-specific antibody, NRP2. After being targeted into tumor cells, GDMCN2 gradually release the sonosensitizer sinoporphyrin sodium (DVDMS) and chemotherapeutic gemcitabine (GEM) and simultaneously generated reactive oxygen species (ROS) under ultrasound (US) irradiation. This system can overcome gemcitabine resistance in pancreatic cancer and reduce its toxicity to non-targeted cells and tissues. In a mechanistic cascade, the release of ROS activates the mitochondrial transition pore (MPTP), leading to the release of Ca2+ and induction of endoplasmic reticulum (ER) stress. Therefore, microtubule-associated protein 1A/1B-light chain 3 (LC3) is activated, promoting lysosomal autophagy. This process also induces autophagy-dependent ferroptosis, aided by the upregulation of Nuclear Receptor Coactivator 4 (NCOA4). This mechanism increases the sensitivity of pancreatic cancer cells to chemotherapeutic drugs and increases mitochondrial and DNA damage. The findings demonstrate the potential of GDMCN2 nanocages as a new avenue for the development of cancer therapeutics.


Assuntos
Ferroptose , Estruturas Metalorgânicas , Neoplasias Pancreáticas , Humanos , Estruturas Metalorgânicas/metabolismo , Linhagem Celular Tumoral , Espécies Reativas de Oxigênio/metabolismo , Apoptose , Anticorpos Monoclonais/uso terapêutico , Autofagia , Gencitabina , Neoplasias Pancreáticas/tratamento farmacológico , Retículo Endoplasmático/metabolismo , Neoplasias Pancreáticas
16.
Mol Cell Biochem ; 368(1-2): 69-76, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22644486

RESUMO

Metformin acts as an energy regulator by activating 5'-adenosine monophosphate-activated protein kinase (AMPK), which is a key player in the regulation of energy homeostasis, but it is uncertain whether AMPK is its direct target. This study aims to investigate the possible interaction between metformin and AMPK. First, we verified that metformin can promote AMPK activation and induce ACC inactivation in human HepG2 cells using western blot. Then we predicted that metformin may interact with the γ subunit of AMPK by molecular docking analysis. The fluorescence spectrum and ForteBio assays indicated that metformin has a stronger binding ability to the γ subunit of AMPK than to α subunit. In addition, interaction of metformin with γ-AMPK resulted in a decrease in the α-helicity determined by CD spectra, but relatively little change was seen with α-AMPK. These results demonstrate that metformin may interact with AMPK through binding to the γ subunit.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Acetil-CoA Carboxilase/metabolismo , Hipoglicemiantes/farmacocinética , Metformina/farmacocinética , Ativação Enzimática/efeitos dos fármacos , Células Hep G2 , Humanos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Ligação Proteica/efeitos dos fármacos
17.
Zhonghua Zhong Liu Za Zhi ; 34(4): 249-53, 2012 Apr.
Artigo em Zh | MEDLINE | ID: mdl-22781034

RESUMO

OBJECTIVE: To investigate the inhibitory effects of humanized monoclonal antibody-3 (huTNT-3) mediated truncated tissue factor (tTF) on the H(22) hepatoma-bearing mice, and to explore its mechanisms. METHODS: The coagulation activity of the huTNT-3/tTF fusion protein was detected by clotting assay and clotting factor X (FX) activation test in vitro. Mouse hepatoma cell line H(22) cells were inoculated subcutaneously into mice to establish the mouse models of hepatoma. The mice were randomly divided into two groups to be injected once with huTNT-3/tTF fusion protein or tTF protein labeled with rhodamine B isothiocyanate (RBITC), respectively. The localization of huTNT-3/tTF fusion protein in the mouse hepatoma tissue was analyzed by confocal laser scanning microscopy 24 hour after the injection. Fifteen mice were randomly divided into three groups to be injected with the huTNT-3/tTF fusion protein, tTF protein or phosphate buffered saline (PBS) once, respectively. The tumor size was measured every two days to calculate the tumor volume. Ten days after the injection the mice were sacrificed. Samples of the tumor, heart, livers, spleen, lung, kidney and brains of the mice were taken for histopathological examination. RESULTS: Both the huTNT-3/tTF fusion protein and tTF protein effectively promoted blood coagulation. Under the conditions of Ca(2+), the coagulation time in the 1.5, 3, 6 µmol/L huTNT-3/tTF groups was (12.90 ± 0.60) min, (10.39 ± 0.40) min and(8.15 ± 0.24) min, respectively, and the coagulation time of the 1.5, 3, 6 µmol/L tTF groups was (14.23 ± 0.46) min, (12.10 ± 0.49) min and (9.83 ± 0.52) min, respectively, the difference between the two groups was not significant (F = 0.145, P = 0.705). The huTNT-3/tTF fusion protein was similar to the tTF protein in the ability of activating FX (t = 0.101, P > 0.05). The confocal laser scanning microscopic analysis showed that RBITC-fluorescence labeled huTNT-3/tTF fusion protein was enriched in the hepatoma tissue. The tumor volume of the huTNT-3/tTF fusion protein group was significantly lower than that of the tTF and PBS groups (both P < 0.001), however, there was not significant difference between the tTF and PBS groups (t = -0.616, P > 0.05). The survival time of the huTNT-3/tTF group was (25.5 ± 2.5) d, significantly longer than that of the PBS group (17.3 ± 1.9) d and the tTF group (18.6 ± 1.9) d, (both P < 0.05). CONCLUSION: The huTNT-3/tTF fusion protein retains the coagulation ability and has the capability of targeting to tumor vasculature, and induces thrombosis in the tumor vessels, thus to suppress the growth of hepatoma in the mice.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Tromboplastina/uso terapêutico , Animais , Coagulação Sanguínea , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Fator X/metabolismo , Neoplasias Hepáticas/sangue , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Transplante de Neoplasias , Distribuição Aleatória , Carga Tumoral
18.
Yao Xue Xue Bao ; 47(11): 1483-8, 2012 Nov.
Artigo em Zh | MEDLINE | ID: mdl-23387081

RESUMO

To investigate the influence of bear bile on rat hepatocarcinoma induced by diethylnitrosamine (DEN), a total of 40 rats were randomly divided into 4 groups: normal control group, model group, and two bear bile treatment groups. The rat liver cancer model was induced by breeding with water containing 100 mg x L(-1) DEN for 14 weeks. The rats of the bear bile groups received bear bile powder (200 or 400 mg x kg(-1)) orally 5 times per week for 18 weeks. The general condition and the body weight of rats were examined every day. After 18 weeks the activities of serum alanine transaminase (ALT), aspartate transaminase (AST) and total bilirubin (TBIL) were detected. Meanwhile, the pathological changes of liver tissues were observed after H&E staining. The expression of proliferative cell nuclear antigen (PCNA) and a-smooth muscle actin (alpha-SMA) in liver tissue were detected by immunohistochemical method. After 4 weeks the body weights of rats in normal group were significantly more than that in other groups (P < 0.05); and that in the two bile groups was significantly more than that in the model group. Compared with normal group, the level of serum glutamic-pyruvic transaminase and total bilirubin increased significantly in other groups; compared with model group, these two indexes decreased significantly in two bile groups. Hepatocellular carcinoma occurred in all rats except for normal group; there were classic cirrhosis and cancer in model group while there were mild cirrhosis and high differentiation in two bile groups. There were almost no expressions of PCNA and alpha-SMA in normal group while there were high expressions in model group; the two bile groups had some expressions but were inferior to the model group, and alpha-SMA reduced markedly. It indicated that bear bile restrained the development of liver cancer during DEN inducing rat hepatocarcinoma, which may be related to its depressing hepatic stellate cell activation and relieving hepatic lesion and cirrhosis.


Assuntos
Antineoplásicos/farmacologia , Bile/química , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas Experimentais/patologia , Ursidae , Actinas/metabolismo , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Bilirrubina/sangue , Peso Corporal/efeitos dos fármacos , Carcinoma Hepatocelular/sangue , Carcinoma Hepatocelular/induzido quimicamente , Dietilnitrosamina , Fígado/metabolismo , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Neoplasias Hepáticas Experimentais/sangue , Neoplasias Hepáticas Experimentais/induzido quimicamente , Masculino , Pós/farmacologia , Antígeno Nuclear de Célula em Proliferação/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley
19.
Adv Healthc Mater ; 11(16): e2102770, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35575205

RESUMO

The self-delivery of sonosensitizers and immunomodulators to tumor areas, which is highly recommended for enhancing sonodynamic immunotherapy, remains a challenge. Herein, a self-delivering nanodrug (HB-NLG8189, drug loading: ≈100 wt%) is developed by the small-molecule self-assembly of "HB" (a new clinical photosensitizer) and NLG8189 (indoleamine-(2,3)-dioxygenase (IDO) pathway inhibitor) for sonodynamic-augmented immunotherapy; this preparation method ensures the absence of excipient-related toxicity and immunogenicity. To evade immune recognition and prolong the circulation time, the HB-NLG8189 nanodrugs are camouflaged using macrophage cell membranes (MPCMs). The constructed HB-NLG8189@MPCM nanodrugs show an ability to preferentially accumulate within tumors. Upon ultrasound triggering, the HB-NLG8189@MPCM is able to generate reactive oxygen species efficiently for robust sonodynamic therapy; it induces immunogenic cell death, initiates an antitumor immune response to activate tumor-specific effector T cells, and promotes the secretion of inflammatory cytokines. The concomitant delivery of NLG8189 reverses the immunosuppressive tumor microenvironment by restraining IDO-1 activation and the intratumoral infiltration of regulatory T cells. Sonodynamic-augmented immunotherapy with HB-NLG8189@MPCM significantly inhibits the growth of both primary and distant tumors with little systemic toxicity. The biomimetic self-delivery nanodrug provides a promising paradigm for improving sonodynamic immunotherapy.


Assuntos
Nanopartículas , Neoplasias , Linhagem Celular Tumoral , Humanos , Fatores Imunológicos/uso terapêutico , Imunoterapia , Indolamina-Pirrol 2,3,-Dioxigenase , Macrófagos/patologia , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Microambiente Tumoral
20.
Adv Healthc Mater ; 11(6): e2102060, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34894092

RESUMO

The anti-tumor efficacy of single photodynamic therapy (PDT) and radiotherapy (RT) has been greatly affected by inadequate tumor uptake of photo/radiation sensitizers, limited laser penetration depth, and radiation sickness caused by high doses of X-rays. Here, the authors report a biomimetic coronavirus-inspired hollow mesoporous gadolinium/bismuth nanocarrier loaded with a new NIR photosensitizer HB (termed as HB@VHMBi-Gd) for magnetic resonance imaging (MRI)-guided synergistic photodynamic-RT. HB@VHMBi-Gd displayed a faster cellular uptake rate than the conventional spherical HMBi-Gd loaded with HB (HB@SHMBi-Gd) because of rough surface-enhanced adhesion. After intravenous injection, HB@VHMBi-Gd is efficiently delivered to the tumor and rapidly invades the tumor cells by surface spikes. Interestingly, lysosomal acidity can trigger the degradation of VHMBi-Gd to produce ultrasmall nanoparticles to amplify the X-ray attenuation ability and enhance MRI contrast and radiosensitization. Under laser and X-ray irradiation, HB@VHMBi-Gd significantly enhances 1 O2 generation from HB to induce activation of caspase 9/3 and inhibition of C-myc, while enhancing hydroxyl radical generation from Bi2 O3 to induce intense DNA breakage. By synergistically inducing cell apoptosis by distinct reactive oxygen species (ROS), HB@VHMBi-Gd exhibits superior anticancer efficacy with ≈90% tumor inhibition. They envision that biomimetic virus-inspired hollow hybrid metal nanoparticles can provide a promising strategy for imaging-guided synergistic photodynamic-RT.


Assuntos
Nanopartículas Metálicas , Nanopartículas , Fotoquimioterapia , Bismuto , Linhagem Celular Tumoral , Gadolínio , Imageamento por Ressonância Magnética , Fotoquimioterapia/métodos , Nanomedicina Teranóstica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA