Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
J Drug Target ; 29(3): 336-348, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33115283

RESUMO

Insulin resistance promotes the occurrence of liver cancer and decreases its chemosensitivity. Rosiglitazone (ROSI), a thiazolidinedione insulin sensitiser, could be used for diabetes with insulin resistance and has been reported to show anticancer effects on human malignant cells. In this paper, we investigated the combination of ROSI and chemotherapeutics on the growth and metastasis of insulin-resistant hepatoma. In vitro assay, ROSI significantly enhanced the inhibitory effects of adriamycin (ADR) on the proliferation, autophagy and migration of insulin-resistant hepatoma HepG2/IR cells via downregulation of EGFR/ERK and AKT/mTOR signalling pathway. In addition, ROSI promoted the apoptosis of HepG2/IR cells induced by ADR. In vivo assay, high fat and glucose diet and streptozotocin (STZ) induced insulin resistance in mice by increasing the body weight, fasting blood glucose (FBG) level, oral glucose tolerance, fasting insulin level and insulin resistance index. Both the growth of mouse liver cancer hepatoma H22 cells and serum FBG level in insulin resistant mice were significantly inhibited by combination of ROSI and ADR. Thus, ROSI and ADR in combination showed a stronger anti-tumour effect in insulin resistant hepatoma cells accompanying with glucose reduction and might represent an effective therapeutic strategy for liver cancer accompanied with insulin resistant diabetes.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Doxorrubicina/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Rosiglitazona/farmacologia , Animais , Animais não Endogâmicos , Antibióticos Antineoplásicos/administração & dosagem , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Glicemia/efeitos dos fármacos , Carcinoma Hepatocelular/patologia , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Doxorrubicina/administração & dosagem , Quimioterapia Combinada , Células Hep G2 , Células Endoteliais da Veia Umbilical Humana , Humanos , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/farmacologia , Resistência à Insulina , Neoplasias Hepáticas/patologia , Masculino , Camundongos , Rosiglitazona/administração & dosagem
2.
Int J Nanomedicine ; 15: 8875-8892, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33209022

RESUMO

PURPOSE: Vascular endothelial growth factor receptor 2 (VEGFR-2) and neuropilin-1 (NRP-1) are two prominent synergistic receptors overexpressed on new blood vessels in glioma and may be promising targets for antiglioma therapy. The aim of this study was to design a dual receptor targeting and blood-brain barrier (BBB) penetrating peptide-modified polyethyleneimine (PEI) nanocomplex that can efficiently deliver the angiogenesis-inhibiting secretory endostatin gene (pVAXI-En) to treat glioma. MATERIALS AND METHODS: We first constructed the tandem peptide TAT-AT7 by conjugating AT7 to TAT and evaluated its binding affinity to VEGFR-2 and NRP-1, vasculature-targeting ability and BBB crossing capacity. Then, TAT-AT7-modified PEI polymer (PPTA) was synthesized, and a pVAXI-En-loaded PPTA nanocomplex (PPTA/pVAXI-En) was prepared. The physicochemical properties, cytotoxicity, transfection efficiency, capacities to cross the BBB and BTB (blood-tumor barrier) and glioma-targeting properties of PPTA/pVAXI-En were investigated. Moreover, the in vivo anti-angiogenic behaviors and anti-glioma effects of PPTA/pVAXI-En were evaluated in nude mice. RESULTS: The binding affinity of TAT-AT7 to VEGFR-2 and NRP-1 was approximately 3 to 10 times greater than that of AT7 or TAT. The cellular uptake of TAT-AT7 in endothelial cells was 5-fold and 119-fold greater than that of TAT and AT7 alone, respectively. TAT-AT7 also displayed remarkable efficiency in penetrating the BBB and glioma tissue in vivo. PPTA/pVAXI-En exhibited lower cytotoxicity, stronger BBB and BTB traversing abilities, higher selective glioma targeting and better gene transfection efficiency than PEI/pVAXI-En. More importantly, PPTA/pVAXI-En significantly suppressed the tube formation and migration of endothelial cells, inhibited glioma growth, and reduced the microvasculature in orthotopic U87 glioma-bearing nude mice. CONCLUSION: Our study demonstrates that PPTA/pVAXI-En can be exploited as an efficient dual-targeting nanocomplex to cross the BBB and BTB, and hence it represents a feasible and promising nonviral gene delivery system for effective glioma therapy.


Assuntos
Barreira Hematoencefálica/metabolismo , Endostatinas/genética , Glioma/patologia , Nanoestruturas/química , Neuropilina-1/metabolismo , Peptídeos/química , Polietilenoimina/química , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Portadores de Fármacos/química , Endostatinas/química , Terapia Genética , Glioma/genética , Glioma/terapia , Humanos , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Permeabilidade , Transfecção
3.
Biomed Pharmacother ; 120: 109478, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31568987

RESUMO

Resistance to chemotherapeutic agents is a major cause of treatment failure in patients with oral cancer. Proton pump inhibitors (PPIs), essentially H+-K+-ATPase inhibitors which are currently used in the treatment of acid related diseases, have demonstrated promising antitumor and chemo-sensitizing efficacy. The main purpose of the present study was to investigate whether pantoprazole (PPZ, one of PPIs) could increase the sensitivity of chemoresistant oral epidermoid carcinoma cells (KB/V) to vincristine (VCR) and elucidate the underlying action mechanism. Results showed that combination treatment of PPZ and VCR synergistically inhibited the proliferation of KB/V cells in vitro and in vivo. Furthermore, administration of PPZ and VCR not only induce apoptosis and G2/M phase arrest in KB/V cells but also suppress the migration and invasion of KB/V cells. The mechanism underlying synergistic anti-tumor effect of PPZ and VCR was related to the inhibition of the function and expression of P-glycoprotein (P-gp) and the down-regulation of EGFR/MAPK and PI3K/Akt/mTOR signaling pathways in KB/V cells. Additionally, we observed that PPZ treatment induced an increase in lysosomal pH and inhibited the activity of lysosomal enzyme acid phosphatase in KB/V cells, which could functionally reduce the sequestration of VCR in lysosomes and sensitized KB/V cells to VCR. In conclusion, our study demonstrated that PPZ could be included in new combined therapy of human oral cancer (especially on VCR-resistant therapy) together with VCR.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma de Células Escamosas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Bucais/tratamento farmacológico , Pantoprazol/farmacologia , Vincristina/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Células KB , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Bucais/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
4.
Exp Ther Med ; 17(3): 2279-2283, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30783486

RESUMO

Expression levels of interleukin-17 (IL-17) and IL-34 was investigated to analyze the influencing factors for prognosis in patients with lupus nephritis (LN). Clinical data of 45 patients (LN group) treated and diagnosed with LN via renal biopsy in Yanan University Affiliated Hospital from October 2010 to October 2012 and 50 healthy subjects (control group) were analyzed retrospectively. Levels of serum IL-17 and IL-34 were detected via enzyme-linked immunosorbent assay. Correlations of serum IL-17 and IL-34 with urinary protein in LN patients were analyzed via Pearson correlation analysis. Univariate survival analysis was performed using the Kaplan-Meier method, and multivariate analysis was performed for LN prognosis using the Cox proportional hazards model. Levels of serum IL-34 and IL-17 in patients in LN group were significantly higher than those in control group (P<0.001). Serum IL-17 and IL-34 in LN patients were positively correlated with urinary protein (r= 0.436 and 0.714, P<0.05). Adverse factors affecting the prognosis of 45 LN patients including age, hemoglobin, platelet, blood uric acid, urinary protein, IL-17 and IL-34, showing statistically significant differences (P<0.05). Age, hemoglobin, blood uric acid, urinary protein, IL-17 and IL-34 were independent risk factors for poor prognosis of LN (P<0.05). The inflammatory factors IL-17 and IL-34 are highly expressed in the serum of LN patients. Levels of serum IL-17 and IL-34 in LN patients have positive correlations with urinary protein. Results of univariate and multivariate Cox regression analyses reveal that age, hemoglobin, blood uric acid, urinary protein, IL-17 and IL-34 are independent risk factors for poor prognosis of LN. IL-17 and IL-34 can therefore serve as effective indexes for clinical diagnosis, treatment and prognosis of LN.

5.
Medicine (Baltimore) ; 98(2): e14106, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30633223

RESUMO

BACKGROUND: This study aimed to evaluate the effectiveness of neuromuscular electrical stimulation (NMES) therapy for chronic urinary retention (CUR) following traumatic brain injury (TBI). METHODS: This 2-arm randomized controlled trial (RCT) enrolled 86 eligible patients with CUR following TBI. All included patients were randomly allocated to a treatment group (n = 43) or a sham group (n = 43). The administration of NMES or sham NMES, as intervention, was performed for an 8-week period treatment, and 4-week period follow-up. In addition, all subjects were required to undergo indwelling urinary catheter throughout the study period. The primary outcome was assessed by the post-voiding residual urine volume (PV-VRU). The secondary outcomes were evaluated by the voided volume, maximum urinary flow rate (Qmax), and quality of life, as assessed by Barthel Index (BI) scale. In addition, adverse events were also recorded during the study period. All primary and secondary outcomes were measured at baseline, at the end of 8-week treatment, and 4-week follow-up. RESULTS: At the end of 8-week treatment, the patients in the treatment group did not achieve better outcomes in PV-VRU (P = .66), voided volume (P = .59), Qmax (P = .53), and BI scores (P = .67), than patients in the control group. At the end of 4-week follow-up, there were also no significant differences regarding the PV-VRU (P = .42), voided volume (P = .71), Qmax (P = .24), and BI scores (P = .75) between 2 groups. No adverse events occurred in either group. CONCLUSIONS: In summary, the findings of this study showed that NMES therapy may not benefit patients with CUR following TBI.


Assuntos
Lesões Encefálicas Traumáticas/complicações , Terapia por Estimulação Elétrica/métodos , Retenção Urinária/terapia , Adolescente , Adulto , Idoso , Doença Crônica , Método Duplo-Cego , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Resultado do Tratamento , Retenção Urinária/etiologia , Adulto Jovem
6.
Mol Pharm ; 15(11): 4898-4911, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30223653

RESUMO

Sambutoxin, a representative derivative of 4-hydroxy-2-pyridone, was isolated from Hericium alpestre for the first time in this study. The possible correlation between the sambutoxin-induced suppression of tumor growth and its influence on cell-cycle arrest and apoptosis was investigated. The effects of sambutoxin on reactive oxygen species (ROS) production, DNA damage, mitochondrial transmembrane potential, cell apoptosis, and the expression of related proteins were evaluated. An in vitro cell viability study demonstrated that sambutoxin could inhibit the proliferation of various cancer cells. Treatment with sambutoxin induced the production of ROS, which caused DNA damage. Furthermore, the subsequent sambutoxin-induced activation of ATM and Chk2 resulted in G2/M arrest, accompanied by decreased expression of cdc25C, cdc2, and cyclin B1. Sambutoxin induced apoptosis by activating the mitochondrial apoptosis pathway through an increased Bax/Bcl-2 ratio, loss of mitochondrial membrane potential (ΔΨm), cytochrome (Cyt) c release, caspase-9 and caspase-3 activation, and poly(ADP-ribose) polymerase (PARP) degradation. The ROS elevation induced the sustained phosphorylation of c-Jun N-terminal kinase (JNK), while SP600125, a JNK inhibitor, nearly completely reversed sambutoxin-induced apoptosis. Accordingly, an in vivo study showed that sambutoxin exhibited potential antitumor activity in a BALB/c nude mouse xenograft model without significant systemic toxicity. Moreover, the expression changes in proteins related to the G2/M phase, DNA damage, and apoptosis in vivo were consistent with those in vitro. Importantly, sambutoxin has remarkable antiproliferative effects and is a promising anticarcinogen candidate for cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Micotoxinas/farmacologia , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Basidiomycota/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Micotoxinas/química , Micotoxinas/isolamento & purificação , Micotoxinas/uso terapêutico , Neoplasias/patologia , Piridinas/química , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Chem Biol Interact ; 294: 18-27, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30125547

RESUMO

Non-small cell lung cancer (NSCLC) is known as highly metastatic disease because it is difficult to diagnose at early stage. More than 60% of NSCLC patients' overexpress receptor tyrosine kinase (RTK) such as EGFR that has been proved to display resistance to receptor tyrosine kinase inhibitor (TKI) through PI3K signaling, while single PI3K inhibitors increase RTK expression as feedback. So, to select the proper targeted agent or target an assortment of molecular subsets, such as EGFR mutations for different subgroups of patients with NSCLC is urgent. Compound BENC-511, a potent PI3K inhibitor, had effects on inhibiting cancer cell survival and delaying tumor growth, but the effects and mechanisms on cancer metastasis are not clear. Methods of Scratch assay, Transwell system, experimental metastasis mice models, plasmid transfection, quantitative real-time PCR and Western blot were used. Results showed that BENC-511 could significantly inhibit lung cancer cells invasion and metastasis both in vitro and in vivo. And it not only inhibited PI3K/Akt signal pathway, but also directly suppressed phosphorylation of EGFR and nuclear translocation of ß-catenin. Moreover, our study firstly reported BENC-511 seemed more sensitive to NSCLC cells that highly expressed Zinc-finger E-box binding protein 1 (ZEB1), one of the epithelial-mesenchymal transition (EMT) inducer, and knockdown of ZEB1 could improve the effects of this compound. These findings suggested that BENC-511 should be a promising lead molecule for anti-metastasis therapy by targeting ß-catenin/ZEB1 regulatory loop and serve as a therapeutic agent to inhibit metastasis of NSCLC.


Assuntos
Benzopiranos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais/efeitos dos fármacos , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo , beta Catenina/metabolismo , Animais , Benzopiranos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Melanoma Experimental/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transplante Homólogo , Homeobox 1 de Ligação a E-box em Dedo de Zinco/antagonistas & inibidores , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética
8.
Neuroreport ; 29(14): 1174-1179, 2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-29985831

RESUMO

The neurotoxicity of aggregated amyloid ß (Aß) has been implicated as a critical cause in the pathogenesis of Alzheimer's disease. In a previous work, we have shown that low-molecular-weight chondroitin sulfate (LMWCS), a derivative of chondroitin sulfate, protected the SH-SY5Y neuroblastoma cells from Aß25-35-induced neurotoxicity, decreased intracellular reactive oxygen species level and inhibited the cell apoptosis. However, the underlying mechanism of the antioxidative effect of LMWCS in the SH-SY5Y cells has not been well explored. In the present study, the SH-SY5Y cells were cultured and exposed to 30 µM Aß25-35 in the absence or presence of LMWCS (50, 100 and 200 µg/ml). Results indicate that incubation of cells with LMWCS before Aß25-35 exposure increased superoxide dismutase, glutathione peroxidase and Na/K-ATPase activities and decreased the malondialdehyde content. In addition, LMWCS inhibited the imbalance of Bcl-2 and Bax and decreased caspase-3 and caspase-9 expressions. LMWCS antagonizes Aß25-35-induced neurotoxicity by attenuating oxidative stress, and our results suggest that LMWCS might be used as a potential compound for Alzheimer's disease prevention.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Sulfatos de Condroitina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sulfatos de Condroitina/metabolismo , Humanos , Peso Molecular , Neuroblastoma/tratamento farmacológico , Neuroblastoma/patologia , Fármacos Neuroprotetores/farmacologia
9.
Int J Biol Macromol ; 107(Pt A): 349-362, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28870748

RESUMO

Previous studies demonstrated that SIP-SII, a sulfated derivative of SIP that is isolated from the ink of Sepiella maindroni, showed significant inhibition of tumor growth and metastasis. In this study, the effects of SIP-SII on the migration, invasion and molecular mechanism in ovarian cancer cell line, SKOV-3 cells, were investigated. The flow cytometry, confocal microscope observation, western blot and RT-PCR results indicated that SIP-SII located on cell membrane and inhibited the expression and activation of epidermal growth factor receptor (EGFR). Moreover, the binding capacity of SIP-SII with EGFR was confirmed by surface plasmon resonance (SPR) analysis and co-localization of EGFR and SIP-SII. Accordingly, SIP-SII was proved to attenuate the EGF-induced EGFR phosphorylation and migration by western blot and wound healing assay, respectively. Additionally, SIP-SII inhibited p38/MAPK and PI3K/Akt/mTOR signaling pathways in SKOV-3 cells significantly. What is more, SIP-SII showed amplified inhibitory activity on migration, invasion, and MMP-2 expression in combination with p38-specific inhibitor, PI3K-specific inhibitor or mTOR-specific inhibitor in SKOV-3 cells. Therefore, the mechanism that SIP-SII suppressed EGFR-mediated p38/MAPK and PI3K/Akt/mTOR signaling pathways to inhibit migration and invasion of SKOV-3 cells was demonstrated. These findings suggested that SIP-SII might be used as a potential inhibitor against tumor metastasis.


Assuntos
Proliferação de Células/efeitos dos fármacos , Decapodiformes/química , Neoplasias Ovarianas/tratamento farmacológico , Polissacarídeos/química , Animais , Extratos Celulares/química , Linhagem Celular Tumoral , Receptores ErbB/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/genética , Polissacarídeos/isolamento & purificação , Polissacarídeos/farmacologia , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais/efeitos dos fármacos , Sulfatos/química , Serina-Treonina Quinases TOR/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
10.
Mol Carcinog ; 57(1): 44-56, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28833603

RESUMO

Hepatocellular carcinoma (HCC) has poor prognosis due to the advanced disease stages by the time it is diagnosed, high recurrence rates and metastasis. In the present study, we investigated the effects of metformin (a safe anti-diabetic drug) and curcumin (a turmeric polyphenol extracted from rhizome of Curcuma longa Linn.) on proliferation, apoptosis, invasion, metastasis, and angiogenesis of HCC in vitro and in vivo. It was found that co-treatment of metformin and curcumin could not only induce tumor cells into apoptosis through activating the mitochondria pathways, but also suppress the invasion, metastasis of HCC cells and angiogenesis of HUVECs. These effects were associated with downregulation of the expression of MMP2/9, VEGF, and VEGFR-2, up-regulation of PTEN, P53 and suppression of PI3K/Akt/mTOR/NF-κB and EGFR/STAT3 signaling. Co-administration of metformin and curcumin significantly inhibited HCC tumor growth than administration with metformin or curcumin alone in a xenograft mouse model. Thus, metformin and curcumin in combination showed a better anti-tumor effects in hepatoma cells than either metformin or curcumin presence alone and might represent an effective therapeutic strategy for HCC treatment.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Neovascularização Patológica/prevenção & controle , Animais , Carcinoma Hepatocelular/irrigação sanguínea , Carcinoma Hepatocelular/patologia , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Curcumina/administração & dosagem , Feminino , Células Hep G2 , Humanos , Neoplasias Hepáticas/irrigação sanguínea , Neoplasias Hepáticas/patologia , Metformina/administração & dosagem , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Transdução de Sinais/efeitos dos fármacos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Int J Biochem Cell Biol ; 93: 1-11, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29074436

RESUMO

Multidrug resistance (MDR) is one of major obstacles to effective chemotherapeutic treatment of cancer. This study showed that DHPAC, 2-(6-ethoxy-3-(3-ethoxyphenylamino) -1-methyl-1,4-dihydroindeno[1,2-c]pyrazol-7-yloxy) acetamide, a novel compound that binds to the same site on microtubules as colchicine, has high anti-tumour activity in vincristine-resistant oral epidermoid carcinoma (KB/V) cells. It found that the presence of DHPAC strongly inhibited KB/V cell growth in vivo and in mice xenograft. The inhibitory effect of DHPAC is much stronger than that by colchicine in these KB/V cells (IC50: 64.4nM and 458.0nM respectively). Treatment of the cells with DHPAC induced cell apoptosis by reducing mitochondrial membrane potential and altered the expression of several apoptosis-related proteins such as Bcl-2, Bax, Caspase-9, Cytochrome c and PARP. DHPAC treatment also caused cell rest in G2/M phase by regulating of the expression of a number of cell cycle-related proteins (e.g. Cyclin B1, Cdc2, Cdc25b, Cdc25c, RSK2). Furthermore, DHPAC presence inhibits PTEN phosphorylation and PTEN/Akt/NF-κB signalling. Thus, DHPAC has potent anti-cancer activity in MDR tumuors and may be a potential therapeutic agent for the treatment of vincristine-resistant human oral epidermoid carcinoma.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Bucais/tratamento farmacológico , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Antineoplásicos/química , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Humanos , Células K562 , Células MCF-7 , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neoplasias Bucais/metabolismo , Neoplasias Bucais/patologia , Proteínas de Neoplasias/metabolismo , Vincristina
12.
Mol Nutr Food Res ; 61(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28421659

RESUMO

SCOPE: Although the previous trials of inflammation have indicated that morin hydrate (MO) hold considerable promise, understanding the distinct mechanism of MO against inflammation remains a challenge. METHODS AND RESULTS: This study investigated the effect of MO in atherosclerosis in ApoE-/- mice and underlying cell signaling of MO effect in inflammation in human umbilical vein endothelial cells (HUVECs). Administration of MO significantly reduced serum lipid level, inflammatory cytokines (TNF-α and ICAM-1), and atherosclerotic plaque formation in vivo. MO presence attenuated the expression of TNF-α-induced inflammatory cytokines (ICAM-1, COX-2, and MMP-9), and remarkably enhanced microtubule associated protein 1 light chain 3 beta 2 (MAP1LC3B2) expression and sequestosome 1 (SQSTM1/p62) degradation in HUVECs. These MO effects were significantly prevented by the presence of autophagic inhibitors, 3-methyladenine (3-MA), or chloroquine (CQ), as well as siRNA suppression of ATG5 and BECN1. MO increased intracellular cAMP levels and activated cAMP-PKA-AMPK-SIRT1 signaling in vivo and in vitro. These changes resulted in increased expression of autophagy-related protein MAP1LC3B2 and decreased secretion of inflammatory cytokines (ICAM-1, COX-2, and MMP-9). CONCLUSION: Our results suggest that anti-AS and anti-inflammatory effects of MO are largely associated with its induction of autophagy through stimulation of cAMP-PKA-AMPK-SIRT1 signaling pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Aterosclerose/tratamento farmacológico , AMP Cíclico/fisiologia , Flavonoides/farmacologia , Transdução de Sinais/fisiologia , Animais , Autofagia/efeitos dos fármacos , Células Cultivadas , Flavonoides/uso terapêutico , Lipídeos/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/farmacologia
13.
Braz. J. Pharm. Sci. (Online) ; 53(4): e17081, 2017. tab, graf, ilus
Artigo em Inglês | LILACS | ID: biblio-889411

RESUMO

ABSTRACT Heparin-SOD conjugate (Hep-SOD) was prepared by modifying Cu,Zn-SOD with heparin. An acute radiation-induced mouse injury model was constructed to study the radiation protection effects of Hep-SOD conjugate. Fifty-six mice were randomly divided into seven groups: (I) normal control group; (II) irradiated control group; (III) positive control group (amifostine group, 300 mg/kg); (IV) SOD group (35000 U/kg); (V) high dosage of Hep-SOD group (70000 U/kg); (VI) medium dosage of Hep-SOD group (35000 U/kg); (VII) low dosage of Hep-SOD group (17500 U/kg). Drugs were intraperitoneally injected into each mouse 1 h before radiation except for the normal control group. All the irradiated groups were irradiated with 6 Gy. Organ indices, haematopoietic function indices, peripheral blood cells, liver function test, oxidative stress state and pathological observation were detected to study the effects of Hep-SOD on irradiated mice. Results showed that bone marrow suppression of irradiated mice could be reduced when treated by Hep-SOD before radiation. Oxidative stress detection and pathological observation of the liver and intestine showed that the damage caused by radiation was relieved when mice were treated with Hep-SOD before radiation. This study shows a new direction to prevent organisms from the damage caused by radiation.


Assuntos
Animais , Masculino , Ratos , Superóxido Dismutase , Heparina , Liberação Nociva de Radioativos , Radiação/classificação , Anormalidades Induzidas por Radiação , Estresse Oxidativo/efeitos da radiação
14.
Crit Care Med ; 44(11): e1090-e1096, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27760056

RESUMO

OBJECTIVES: To evaluate the resuscitative efficacy and the effect on reperfusion injury of two site-specific PEGylated human serum albumins modified with linear or branched PEG20kDa, compared with saline, 8% human serum albumin and 25% human serum albumin, in a hemorrhagic shock model. SETTING: Laboratory. SUBJECTS: Male Wistar rats. DESIGN: Prospective study. INTERVENTIONS: Rats were bled to hemorrhagic hypovolemic shock and resuscitated with different resuscitation fluids. MEASUREMENTS AND MAIN RESULTS: The mean arterial pressure and blood gas variables were measured. Hemorheology analysis was performed to evaluate the influence of resuscitation on RBCs and blood viscosity. The microvascular state was indirectly characterized in terms of monocyte chemotactic protein-1 and endothelial nitric oxide synthase that related to shear stress and vasodilation, respectively. The levels of inflammation-related factors and apoptosis-related proteins were used to evaluate the reperfusion injury in lungs. The results showed that PEGylated human serum albumin could improve the level of mean arterial pressure and blood gas variables more effectively at the end of resuscitation. poly(ethylene glycol) modification was able to increase the viscosity of human serum albumin to the level of effectively enhancing the expression of monocyte chemotactic protein-1 and endothelial nitric oxide synthase, which could promote microvascular perfusion. The hyperosmotic resuscitative agents including both 25% human serum albumin and PEGylated human serum albumins could greatly attenuate lung injury. No significant therapeutic advantages but some disadvantages were found for Y shaped poly(ethylene glycol) modification over linear poly(ethylene glycol) modification, such as causing the decrease of erythrocyte deformability. CONCLUSIONS: Linear high molecular weight site-specific PEGylated human serum albumin is recommended to be used as a hyperosmotic resuscitative agent.


Assuntos
Ressuscitação/métodos , Albumina Sérica/farmacologia , Choque Hemorrágico/tratamento farmacológico , Animais , Velocidade do Fluxo Sanguíneo/efeitos dos fármacos , Gasometria , Pressão Sanguínea/efeitos dos fármacos , Quimiocina CCL2/sangue , Citocinas/metabolismo , Pulmão/metabolismo , Lesão Pulmonar/prevenção & controle , Masculino , Óxido Nítrico Sintase Tipo III/metabolismo , Polietilenoglicóis/química , Estudos Prospectivos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos Wistar , Traumatismo por Reperfusão/prevenção & controle , Albumina Sérica/química , Proteína X Associada a bcl-2/metabolismo
15.
Biochim Biophys Acta ; 1860(11 Pt A): 2589-2597, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27491336

RESUMO

METHODS: 8-week-age male ApoE(-/-) mice were fed with the atherogenic diet together with or without tested compounds (rosuvastatin calcium, α-LNA-LMWCS, LMWCS and α-LNA) for 16 weeks. When the animals were killed, blood plasma was isolated to test the level of TC, LDL-C, TNF-α, IL-6 and CRP by biochemistry analysis and ELISA method. The whole aorta and aortic root sections were also collected to study atherogenesis level and reveal the possible mechanism by histological examination, real-time PCR and Western blot analysis. RESULTS: The level of TC, LDL-C, TNF-α, IL-6 and CRP in plasma in H-LNA-LMWCS group were significantly lower than those of the control group (rosuvastatin calcium). Plaques in H-LNA-LMWCS group showed higher content of smooth muscle cells, lower content of lipid and macrophages, and lower mRNA levels of TNF-α, IL-6, CRP, MCP-1, VCAM-1 and ICAM-1 than those in the control group. In addition, α-LNA-LMWCS could reduce the nuclear translocation of NF-κB, inhibit expressions of p-ERK1/2, p-p38, MCP-1, VCAM-1 and ICAM-1 in mice aorta. CONCLUSION: α-LNA-LMWCS exhibited anti-atherosclerosis effect through regulating the lipid metabolism and diminishing the synthesis of pro-inflammatory cytokines. The possible mechanism may be that α-LNA-LMWCS could influence MAPK/ NF-κB related signal pathway. GENERAL SIGNIFICANCE: The results may provide significant suggestions for the application of α-LNA-LMWCS in anti-atherosclerosis.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/tratamento farmacológico , Sulfatos de Condroitina/uso terapêutico , Animais , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Aterosclerose/genética , Quimiocina CCL2/metabolismo , Sulfatos de Condroitina/administração & dosagem , Sulfatos de Condroitina/farmacologia , Citocinas/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Ácidos Linolênicos/química , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , NF-kappa B/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo
16.
J Med Chem ; 59(5): 2151-62, 2016 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-26821136

RESUMO

Fluorescent ligands are gaining popularity as tools to aid GPCR research. Nonetheless, in vivo application of such tools is hampered due to their short excitation wavelengths in the visible range and lack of fluorogenic switch. Here we report the discovery of fluorescent ligands (3a-f) for α1-adrenergic receptors (α1-ARs) by conjugating the environment-sensitive fluorophore cyane 5 (Cy5) with the quinazoline pharmacophore. Among them, the conjugated compound 3a, with acylated piperazine and the shortest carbon chain spacer, exhibited potent binding and remarkable changes in fluorescence (10-fold) upon binding to α1-AR. Furthermore, it could be employed to selectively and specifically label α1-ARs with no washing procedures in single cells, prostate tissue slices, intact tumor xenografts and organs in living mice. Especially, the slice imaging results gave direct and visual evidence that there is a close relationship between α1-ARs and pathological prostate. It is anticipated that our fluorescent tools will find broad applications in the study of α1-AR pharmacology and physiology to aid development of novel therapeutics.


Assuntos
Carbocianinas/química , Descoberta de Drogas , Fluorescência , Corantes Fluorescentes/química , Quinazolinas/química , Receptores Adrenérgicos alfa 1/química , Animais , Sítios de Ligação , Células CHO , Linhagem Celular Tumoral , Cricetulus , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Ligantes , Camundongos , Estrutura Molecular , Neoplasias Experimentais/química , Neoplasias Experimentais/metabolismo , Piperazina , Piperazinas/química , Quinazolinas/síntese química , Relação Estrutura-Atividade
17.
Photochem Photobiol Sci ; 15(2): 244-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26789132

RESUMO

The luciferase reporter gene assay system is broadly applied in various biomedical aspects, including signaling pathway dissection, transcriptional activity analysis, and genetic toxicity testing. It significantly improves the experimental accuracy and reduces the experimental error by the addition of an internal control. In the current research, we discovered some specific ions that could selectively inhibit firefly luciferase while having a negligible effect on renilla luciferase in vitro in the dual-reporter gene assay. We showed that these ionic compounds had a high potential of being utilized as quench-and-activate reagents in the dual-reporter assay. Furthermore, results from kinetic studies on ion-mediated quenching effects indicated that different ions have distinct inhibition modes. Our study is anticipated to guide a more affordable design of quench-and-activate reagents in biomedicine and pharmaceutical analysis.


Assuntos
Vaga-Lumes/enzimologia , Íons/metabolismo , Luciferases de Vaga-Lume/metabolismo , Luciferases de Renilla/metabolismo , Substâncias Luminescentes/metabolismo , Renilla/enzimologia , Animais , Ensaios Enzimáticos , Vaga-Lumes/genética , Genes Reporter , Luciferases de Vaga-Lume/antagonistas & inibidores , Luciferases de Vaga-Lume/genética , Luciferases de Renilla/antagonistas & inibidores , Luciferases de Renilla/genética , Luminescência , Renilla/genética
18.
Carbohydr Polym ; 129: 50-4, 2015 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-26050887

RESUMO

Our previous studies demonstrated that SIP-S had anti-metastatic activity and inhibited the growth of metastatic foci. Here we report the anti-tumor and immunoregulatory potential of SIP-S. SIP-S could significantly inhibit tumor growth in S180-bearing mice, and the inhibition rates was 43.7% at 30 mg/kg d. Besides, SIP-S could improve the thymus and spleen indices of S180-bearing mice and the mice treated with CTX. The combination of SIP-S (15 mg/kg d) with CTX (12.5 mg/kg d) showed higher anti-tumor potency than CTX (25 mg/kg d) alone. These results indicated that SIP-S had immunoenhancing and anticancer activity, and the immunoenhancing activity might be one mechanism for its anti-tumor activity. Flow cytometry results showed that SIP-S could induce tumor cells apoptosis. Western blot analysis indicated that SIP-S could upregulate the expression of pro-apoptotic proteins, caspase-3, -8, -9 and Bax, and downregulate the expression of anti-apoptotic protein PARP-1 in tumor cells in a dose-dependent manner. In summary, SIP-S has anti-tumor activity, which may be associated with its immunostimulating and pro-apoptotic activity.


Assuntos
Antineoplásicos/uso terapêutico , Metástase Neoplásica/tratamento farmacológico , Polissacarídeos/uso terapêutico , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclofosfamida/farmacologia , Masculino , Camundongos , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Polissacarídeos/farmacologia , Baço/efeitos dos fármacos , Baço/metabolismo , Timo/efeitos dos fármacos , Timo/metabolismo
19.
Biochim Biophys Acta ; 1850(6): 1140-9, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25662072

RESUMO

BACKGROUND: Endostatin, a specific inhibitor of endothelial cell proliferation and angiogenesis, has been proved to have effects on ocular neovascular diseases by intraocular injection. In order to increase its permeability to ocular barriers and make it effective on fundus oculi angiogenesis diseases via non-invasive administration (eye drops), endostatin was fused to Tat PTD via a genetic engineering method. METHODS: Most of the Tat PTD- endostatin was expressed as inclusion bodies in Escherichia coli, so pure and active Tat PTD-endostatin was prepared by a series of operations, including inclusion body denaturation, refolding and chromatography. The anti-angiogenesis activity of Tat PTD-endostatin was investigated by cell proliferation experiments and chick embryo chorioallantoic membrane assay. In addition, its translocating ability and concrete entry mechanism into cells were also investigated by fluorescence microscope and flow cytometry. The penetrating ability to ocular barriers was also studied by immunohistochemistry. A mouse choroidal neovascularization model was established to investigate the pharmacodynamics of Tat PTD-endostatin. RESULTS: The obtained Tat PTD-endostatin had excellent anti-angiogenesis activity and was superior to Es in cellular translocating. Macropinocytosis may be the dominant route of entry of Tat PTD-endostatin into cells. Tat PTD-endostatin could cross ocular barriers and arrive at the retina after eye-drop administration. In addition, it displayed inhibitory effects on choroidal neovascularization via eye drops. CONCLUSIONS: Tat PTD-endostatin possessed excellent ocular penetrating ability and anti-angiogenesis effects. GENERAL SIGNIFICANCE: Tat PTD is a promising ocular delivery tool, and Tat PTD-endostatin is a potential drug for curing fundus oculi angiogenesis diseases.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Membrana Corioalantoide/irrigação sanguínea , Neovascularização de Coroide/prevenção & controle , Endostatinas/administração & dosagem , Células Endoteliais/efeitos dos fármacos , Olho/irrigação sanguínea , Neovascularização Fisiológica/efeitos dos fármacos , Administração Oftálmica , Inibidores da Angiogênese/metabolismo , Animais , Transporte Biológico , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Embrião de Galinha , Neovascularização de Coroide/metabolismo , Neovascularização de Coroide/patologia , Neovascularização de Coroide/fisiopatologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Endostatinas/metabolismo , Células Endoteliais/metabolismo , Olho/metabolismo , Injeções Intravítreas , Masculino , Camundongos Endogâmicos C57BL , Soluções Oftálmicas , Permeabilidade , Pinocitose , Proteínas Recombinantes de Fusão/administração & dosagem , Fatores de Tempo
20.
Int J Mol Sci ; 16(1): 2252-68, 2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-25608657

RESUMO

TIPP is a novel thymic immunosuppressive pentapeptide originally obtained from calf thymic immunosuppressive extract. The present study aimed to investigate the inhibitory activity of TIPP on IgE-mediated activation of RBL-2H3 cells. Release of ß-hexosaminidase and histamine, intracellular calcium, membrane ruffling, mRNA levels of cytokines, cyclooxygenase-2 (COX-2) expression, and activation of mitogen-activated protein kinases (MAP kinases) and NF-κB were determined by colorimetric assay, fluorescence spectrophotometer, confocal fluorescence microscope, quantification PCR, and Western blot, respectively. The results showed that TIPP significantly inhibited the degranulation in IgE-antigen complex-stimulated RBL-2H3 cells without cytotoxicity. TIPP significantly suppressed the increase of intracellular calcium and the rearrangement of F-actin, attenuated the transcription of pro-inflammatory cytokines (IL-3, -4, -6, -13, TNF-α, and monocyte chemotactic protein-1 (MCP-1)), and decreased the expression of COX-2. Western blot analysis showed that TIPP had an inhibitory activity on the phosphorylation of extracellular signal-regulated protein kinase 1/2 (ERK1/2) and ERK kinase 1/2 (MEK1/2), and inhibited the activation of NF-κB. The data suggested that TIPP effectively suppressed IgE-mediated activation of RBL-2H3 cells via blocking MEK/ERK and NF-κB signaling pathways.


Assuntos
Imunoglobulina E/metabolismo , Imunossupressores/farmacologia , Mastócitos/metabolismo , Oligopeptídeos/farmacologia , Tetra-Hidroisoquinolinas/farmacologia , Timo/imunologia , Animais , Complexo Antígeno-Anticorpo/metabolismo , Cálcio/metabolismo , Morte Celular/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Linhagem Celular , Estruturas da Membrana Celular/efeitos dos fármacos , Estruturas da Membrana Celular/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Ciclo-Oxigenase 2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Histamina/metabolismo , Mediadores da Inflamação/metabolismo , Mastócitos/efeitos dos fármacos , Mastócitos/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Oligopeptídeos/química , Transporte Proteico/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Tetra-Hidroisoquinolinas/química , Timo/efeitos dos fármacos , beta-N-Acetil-Hexosaminidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA