Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Bioresour Technol ; 393: 130116, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38016583

RESUMO

Hydrogenotrophic denitrification, an environment-friendly process for organic-free influents, is limited due to poor hydrogen mass transfer efficiency and significant pH fluctuations. In this study, we manipulated the carbon dioxide-to-hydrogen ratio to improve hydrogenotrophic denitrification. When carbon dioxide-to-hydrogen ratio was 1:1 (carbon dioxide, 200 ml: hydrogen, 200 ml), the hydrogen utilization and denitrification rates were 2.4 times and 3.0 times that when carbon dioxide-to-hydrogen ratio was 0:1 (carbon dioxide, 0 ml: hydrogen, 200 ml), respectively. The pH fluctuation decreased from 3.1±0.3 to 0.2±0.1. Furthermore, the hydrogenotrophic denitrification, acetoclastic denitrification, homoacetogenic, and electron transfer activities of the sludge were improved. A high carbon dioxide-to-hydrogen ratio augmented the acid-producing and heterotrophic denitrifying microorganism populations. By maintaining a high carbon dioxide-to-hydrogen ratio, the dominant hydrogenotrophic autotrophic denitrification pathway was transformed into a homoacetogenesis-heterotrophic denitrification pathway, thereby achieving higher hydrogen utilization and denitrification rates.


Assuntos
Dióxido de Carbono , Desnitrificação , Nitratos/metabolismo , Hidrogênio , Reatores Biológicos , Processos Autotróficos , Nitrogênio/metabolismo
2.
Int J Oncol ; 63(6)2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37888731

RESUMO

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that, for the scratch wound assay experiments shown in Fig. 1 on p. 2413, the panels showing the '0 h' experiments for the respective incubations with VEGF or BC001 were apparently identical. The authors were able to re­examine their original data files, and realized that this figure had been inadverently assembled incorrectly. The revised version of Fig. 1, containing the correct data for the '0 h / BC001' panel, is shown below. Note that the revisions made to this figure do not affect the overall conclusions reported in the paper. The authors are grateful to the Editor of International Journal of Oncology for allowing them the opportunity to publish this Corrigendum, and apologize to the readership for any inconvenience caused. [International Journal of Oncology 45: 2411­2420, 2014; DOI: 10.3892/ijo.2014.2690].

3.
J Hazard Mater ; 459: 132235, 2023 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-37562349

RESUMO

Struvite recovered from wastewater contains high concentration of fecal indicator bacteria (FIB), porcine adenoviruses (PAdV) and antibiotic resistance genes (ARGs), becoming potential resources of these microbial hazards. Understanding the precipitation behavior of pathogenic indicators and ARGs with suspended solids (SS) will provide the possible strategy for the control of co-precipitation. In this study, SS was divided into high-density SS (separated by centrifugation) and low-density SS (further separated by filtration), and the role of SS on the co-precipitation of FIB, PAdV and ARGs was investigated. The distribution analysis showed that 35.5-73.0% FIB, 79.6% PAdV and 64.5-94.8% ARGs existed in high-density SS, while the corresponding values were 26.9-64.4%, 11.7% and 3.5-24.3% in low-density SS. During struvite generation, 82.7-96.9% FIB, 75.5% PAdV and 56.3-86.5% ARGs were co-precipitated into struvite. High-density SS contributed 20.7-68.5% FIB, 63.9% PAdV and 38.7-87.2% ARGs co-precipitation, and the corresponding contribution of low-density SS was 31.4-79.2%, 3.9% and 6.2-54.7%. Moreover, the precipitated SS in struvite obviously decreased inactivation efficiency of FIB and ARGs in drying process. These results provide a potential way to control the co-precipitation and inactivation of FIB, PAdV and ARGs in struvite through removing high-density SS prior to struvite recovery.


Assuntos
Fosfatos , Águas Residuárias , Suínos , Animais , Estruvita , Fosfatos/análise , Antibacterianos/farmacologia , Resistência Microbiana a Medicamentos , Genes Bacterianos
4.
Environ Sci Pollut Res Int ; 30(15): 45077-45087, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36701053

RESUMO

Anaerobic conversion rate of phenol to methane was low due to its biological toxicity. In this study, the coupling of granular activated carbon (GAC) and exogenous hydrogen (EH) could enhance greatly methane production of phenol anaerobic digestion, and the metagenomic was firstly used to analyze its potential mechanism. The results indicated that a mass of syntrophic acetate-oxidizing bacteria and hydrogen-utilizing methanogens were enriched on the GAC surface, and SAO-HM pathway has become the dominant pathway. The energy transfer analysis implied that the abundance of adenosine triphosphate (ATP) and nicotinamide adenine dinucleotide (NADH) oxidase increased. Furthermore, direct interspecies electron transfer (DIET) was formed by promoting type IV e-pili between Methanobacterium and Syntrophus, thereby improving the interspecies electron transfer efficiency. The dominant SAO-HM pathway was induced and DIET was formed, which was the internal mechanism of the coupling of GAC and EH to enhance anaerobic biotransformation of phenol.


Assuntos
Microbiota , Fenol , Anaerobiose , Carvão Vegetal , Hidrogênio , Fenóis , Metano/metabolismo , Reatores Biológicos
5.
Int J Nanomedicine ; 18: 209-223, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36660339

RESUMO

Background: Extracellular vesicles (EVs) are considered a promising drug delivery platform. Naïve EVs face numerous issues that limit their applications, such as fast clearance, hepatic accumulations, and a lack of target-specific tropism. We aimed to explore a series of surface engineering approaches to: 1) reduce the non-specific adhesion of EVs, and 2) improve their enrichment in the target tissue. As a proof-of-concept, we investigated the therapeutic potentials of a multi-modal EVs system carrying a tumor-specific nanobody and the immuno-stimulant interleukin-12 (IL12) using in vivo models of hepatocellular carcinoma. Methods: The major cell adhesion molecule on the HEK293-derived EVs, integrin ß1 (ITGB1), was knocked out (KO) by CRISPR/Cas9-mediated gene editing, followed by deglycosylation to generate ITGB1-Deg EVs for the subsequent pharmacokinetic and biodistribution analyses. ITGB1-Deg EVs were further loaded with glypican-3 (GPC3)-specific nanobody (HN3) and mouse single-chain IL12 (mscIL12) to generate ITGB1-mscIL12+HN3+Deg EVs, for evaluation of tumor tropism and therapeutic potential in a mice model of hepatocellular carcinoma. Results: Removal of ITGB1 led to the broad suppression of integrins on the EVs surface, resulting in a decrease in cellular uptake. Deglycosylation of ITGB1- EVs gave rise to inhibition of the EVs uptake by activated RAW264.7 cells. ITGB1 removal did not significantly alter the pharmacokinetic behaviors of HEK293-EVs, whereas the ITGB1-Deg EVs exhibited enhanced systemic exposure with reduced hepatic accumulation. Loading of HN3 conferred the ITGB1-Deg EVs with tumor-specific tropism for both subcutaneous and metastasized tumors in mice. The ITGB1-mscIL12+HN3+Deg EVs activated mouse splenocytes with high potency. Systemic administration of the EVs with the equivalent dose of 1.5µg/kg of exosomal IL12 achieved satisfactory tumor growth inhibition and good tolerability. Conclusion: The combinatorial approach of EVs surface engineering conferred HEK293-EVs with reduced non-specific clearance and enhanced tumor targeting efficacy, which constituted an efficient delivery platform for critical cancer therapeutics like IL12.


Assuntos
Carcinoma Hepatocelular , Vesículas Extracelulares , Neoplasias Hepáticas , Humanos , Animais , Camundongos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Interleucina-12/genética , Células HEK293 , Linhagem Celular Tumoral , Neoplasias Hepáticas/terapia , Neoplasias Hepáticas/metabolismo , Distribuição Tecidual , Vesículas Extracelulares/metabolismo , Glipicanas/metabolismo
6.
Environ Sci Pollut Res Int ; 29(33): 50208-50217, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35226263

RESUMO

Floating photocatalyst is of extensive interest due to easy recovery and efficient light harvest. Support materials largely determine the stability of floating photocatalysts and their synthesis complexity. Thus, finding proper floating supports is very important. Herein, ethylene-vinyl acetate copolymer (EVA) was investigated as a support to prepare floating TiO2/EVA using a simple thermal crosslinking procedure. Multiple characterization analyses demonstrated that TiO2 was anchored onto EVA surface evenly via hydrogen-bond-enhanced physical crosslinking and remained its virgin crystal structure. Photocatalytic experiments showed that the removal efficiency of Rhodamine B (RhB) by floating TiO2/EVA increased by 33.8% as compared to suspended particle TiO2. The h+ and ·O2- played dominant roles in TiO2/EVA-driven RhB degradation. A 30-day stability test demonstrated that TiO2/EVA had a high thermal, pH, and photo- stability. The three-run reuse test proved that TiO2/EVA exhibited satisfactory reusability. This study provides a new option for floating photocatalyst synthesis.


Assuntos
Etilenos , Titânio , Catálise , Titânio/química , Compostos de Vinila
7.
Sci Total Environ ; 819: 152080, 2022 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-34856273

RESUMO

Zero-valent iron (ZVI) can enhance anaerobic digestion, and has great potential to alleviate/eliminate methanogenesis inhibition. Little is known about the feasibility of utilizing ZVI to alleviate methanogenesis inhibition that is caused by typical animal feed additive roxarsone in livestock wastewater. In this study, the role of ZVI on alleviating roxarsone-induced methanogenic inhibition and its mechanisms were investigated. With the increase of roxarsone concentration from 5 to 50 mg/L, the inhibition of methanogenesis increased from 3.0% to 65.7%. This inhibition was alleviated by 80.7% and 57.2% when 1.0 and 10.0 g/L ZVI were added, respectively. Due to ZVI addition, an efficient arsenic immobilization onto ZVI (45.4-85.8%) was achieved mainly through the formation of FeAsO4 precipitate and adsorption by ZVI. Under the function of ZVI, hydrogenotrophic methanogenic activity was obviously restored. The microbial community analysis indicates that the ZVI-regulated alleviation on the methanogenesis inhibition was attributed to the enrichment of Methanobacterium and Methanosarcina. The findings from this study demonstrate that ZVI addition is an effective way for treatment of organoarsenic-contaminated wastewater.


Assuntos
Ferro , Roxarsona , Anaerobiose , Animais , Metano , Esgotos/microbiologia
8.
Anticancer Drugs ; 32(10): 1003-1010, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34145179

RESUMO

Surgical tumor removing is the most common procedure after a confirmed cancer diagnosis with no detected metastasis. Surgery can reduce tumor burden and address pathologic changes caused by local compression of tissues by the tumor. This lowers the chances of tumor cell spreading and creates more favorable conditions for further treatment. However, not all tumor cells can be eliminated through surgery. Even in the early stages of the disease, tumor cells often metastasize and cannot be identified by current detection methods. These tiny, disseminated tumors are often the cause of tumor recurrence. There is currently a lack of effective treatment options that can completely prevent tumor recurrence after surgery. To simulate the actual clinical situation, we selected murine-derived tumor cell lines S180 and Kcc853 to establish a post-transplantation residual tumor model in mice. Surgery was performed on mice inoculated with tumors. Tumor tissue was partially excised to set up the postsurgical residual tumor models. The model simulated the clinical situation where tumor cells were not completely eliminated or there were small tumors that had metastasized before surgery. IL-12 was injected to observe its effect on residual tumors or metastatic microtumors. The administration of IL-12 after surgery can significantly inhibit the growth of residual tumors and metastasis, improve the postoperative tumor-free rate and address the problem of tumor recurrence caused by the growth of residual tumors and micro-metastasis. Therefore, the use of IL-12 antitumor cytokine combined with surgery can effectively inhibit tumor recurrence. Low-dose IL-12 (1-10 ng/kg in humans) can inhibit residual tumor growth.


Assuntos
Antineoplásicos/farmacologia , Interleucina-12/farmacologia , Neoplasias Renais/tratamento farmacológico , Sarcoma/tratamento farmacológico , Animais , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Neoplasias Renais/cirurgia , Camundongos , Camundongos Nus , Sarcoma/cirurgia , Vincristina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Agric Food Chem ; 67(6): 1691-1701, 2019 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-30658530

RESUMO

In recent years, various studies have confirmed the role of natural products as effective cancer prevention and treatment drugs. The present study demonstrated that chitosan oligosaccharide (COS) from shells of shrimp and crab caused an inhibitory effect on the proliferation of human renal carcinoma in vitro and in vivo. First, the in vivo biodistribution of COS was investigated by the synthesis of cyanine-7-labeled COS (COS-Cy7) following tail vein injection. The kidney was found to be a major target organ. Then, the impacts on renal carcinoma cell proliferation, apoptosis, and reactive oxygen species (ROS) production were observed in vitro, and an orthotopic xenograft tumor model was designed to evaluate the antitumor efficacy of COS in vivo. In renal carcinoma cells, COS induced G2/M phase arrest and apoptosis in a ROS-dependent fashion. COS significantly promoted mRNA expression of nuclear factor erythroid 2-related factor (Nrf2) and Nrf2 target genes, such as heme oxygenase 1, modifier subunit of glutamate cysteine ligase, and solute carrier family 7 member 11. Additionally, COS significantly upregulated the protein expression of glucose-regulated protein 78, protein RNA-like endoplasmic reticulum (ER) kinase, eukaryotic initiation factor 2α, activating transcription factor 4, C/EBP homologous protein, and cytochrome c, which justified the activation of the ER stress signaling pathway. In vivo, COS repressed tumor growth and induced apoptosis and ROS accumulation, consistent with the in vitro results. Taken together, COS repressed human renal carcinoma growth and induced apoptosis both in vitro and in vivo, mainly via ROS-dependent ER stress pathways.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Quitosana/administração & dosagem , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Neoplasias Renais/tratamento farmacológico , Oligossacarídeos/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/fisiopatologia , Linhagem Celular Tumoral , Feminino , Humanos , Neoplasias Renais/genética , Neoplasias Renais/metabolismo , Neoplasias Renais/fisiopatologia , Camundongos Nus , Fator 2 Relacionado a NF-E2 , Fator de Transcrição CHOP
10.
Life Sci ; 202: 21-27, 2018 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-29626528

RESUMO

Chitosan oligosaccharides (COS), hydrolyzed products of chitosan, have recently been reported to have various biological activities. Herein, the present study was undertaken to assess the ability of COS to potentiate the antitumor effect of cyclophosphamide (CTX) as well as alleviating the CTX-associated toxicities in vivo, in a residual-tumor; a model which is closer to clinical surgery. Sarcoma 180 (S180) residual-tumor mice were divided into 6 groups (n = 6); including control, CTX, COS 40 mg/kg, COS 80 mg/kg, and combination groups (CTX + COS 40, CTX + COS 80). Animals were killed 18 days post-intraperitoneal administration and the tumors were weighed. The spleens were harvested to determine lymphocytes proliferation and NK cell activities; blood cells were evaluated by flow cytometry, and the expression levels of TNF-α were measured using ELISA. Notably, the combined therapy (CTX + COS80) showed the most effective reduction of the tumor weight, the highest inhibition of tumor growth, and proliferation, when compared with control as well single CTX group. Additionally, COS was able to recover the CTX-induced decreases in the lymphocyte proliferation, splenocyte NK cell activity, TNF-α concentration, and abnormal CD4+/CD8+ T lymphocyte subset. The increase in infiltrating T cells and macrophages best explain the immunostimulatory effect of COS. Results herein highlighted the therapeutic potential of COS as adjuvant treatment during tumor chemotherapy.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Quitosana/farmacologia , Ciclofosfamida/farmacologia , Oligossacarídeos/farmacologia , Sarcoma 180/tratamento farmacológico , Animais , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Injeções Intraperitoneais , Células Matadoras Naturais/efeitos dos fármacos , Camundongos , Sarcoma 180/patologia , Baço/citologia , Baço/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Fator de Necrose Tumoral alfa/biossíntese
11.
Oncol Rep ; 39(6): 2759-2768, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29620221

RESUMO

A novel small molecular compound, 4­ethyl­8­fluoro­hydroxy­9­methoxy­11­methyl­1,12­dihydro­4H­2­oxa­6,12a­diaza­dibenzo[b,h]fluorene­3,13­dione (CT­1042) exhibits potent antitumor activity against many tumor cells in vitro. However, the effects and underlying mechanisms of CT­1042 in non­small cell lung cancer (NSCLC) remain unclear. The present study was designed to determine the anticancer properties and underlying molecular mechanisms of CT­1042 in NCI­H460 NSCLC cells. A thiazolyl blue tetrazolium bromide assay (MTT) was performed to evaluate cell viability and flow cytometry was used to analyze apoptosis, mitochondrial membrane potential (MMP) and cell cycle. Real­time quantitative PCR and western blotting were conducted to determine relative mRNA and protein levels. A tumor xenograft experiment was performed to investigate the effects of CT­1042 on tumor growth in vivo. CT­1042 markedly inhibited the proliferation of twelve cancer cell lines, decreased MMP in subject cells and increased caspase­3 activity. Cell cycle analysis indicated that CT­1042 delayed the cell cycle progression during the G2/M phase in a dose­dependent manner. In addition, CT­1042 induced mitochondrial­mediated apoptosis by activating p53 and Bax, as well as inhibiting Bcl­2 and survivin. Finally, CT­1042 significantly suppressed NCI­H460 xenograft tumor growth in vivo, with low systemic toxicity. Collectively, these results revealed that CT­1042 has significant lung anticancer properties.


Assuntos
Antineoplásicos/administração & dosagem , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Fluorenos/administração & dosagem , Proteínas Inibidoras de Apoptose/genética , Neoplasias Pulmonares/tratamento farmacológico , Proteínas Repressoras/genética , Bibliotecas de Moléculas Pequenas/administração & dosagem , Proteína Supressora de Tumor p53/genética , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Camptotecina/análogos & derivados , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Fluorenos/química , Fluorenos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Proteínas Repressoras/metabolismo , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Survivina , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Cancer Chemother Pharmacol ; 81(5): 853-862, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29532153

RESUMO

PURPOSE: MBRI-001 is a novel synthetic derivative of plinabulin. In this study, our purpose is to investigate the inhibition effects of MBRI-001 on human hepatocellular carcinoma as monotherapy or in combination with sorafenib. METHODS: HCCLM3 and Bel-7402 cell lines were used for activity evaluation in vitro. The anti-proliferative activity of MBRI-001 was assessed by MTT assay. The morphological change of microtubules was determined by immunofluorescence assay. The cell cycle was measured by flow cytometer. The expression of cyclin B1 (CCNB1) was analyzed by RT-qPCR and western blotting assays. The antitumor activities in vivo were evaluated with human HCC xenograft mice model. RESULTS: Our data demonstrated that MBRI-001 had better anti-proliferative activities than that of plinabulin against HCCLM3 and Bel-7402 cell lines. MBRI-001 inhibited the formation of microtubules and induced G2/M arrest with the downregulation of CCNB1. In vivo orthotopic mice model demonstrated that MBRI-001 significantly inhibited the growth of HCCLM3 with the apoptosis and necrosis observed in tumor. The combination treatment of MBRI-001 with sorafenib in subcutaneous mice model exhibited a higher antitumor inhibition rate at 72.0%, in comparison with MBRI-001 or sorafenib as monotherapy at 40.7% or 47.7%, respectively. CONCLUSION: MBRI-001 had better inhibition effects on microtubules and human hepatocellular carcinoma than that of plinabulin. The combination treatment of MBRI-001 and sorafenib exhibited a higher antitumor effect, which could provide a new strategy to treat HCC in the future.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Dicetopiperazinas/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Sorafenibe/farmacologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Dicetopiperazinas/uso terapêutico , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas/patologia , Camundongos Nus , Microtúbulos/efeitos dos fármacos , Sorafenibe/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Cell Death Dis ; 9(2): 143, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29396440

RESUMO

ZAK, a mixed lineage kinase, is often described as a positive or negative regulator of cell growth. We identified it as one of the top hits in our kinome cDNA screen for potent regulators of epithelial mesenchymal transition (EMT). Ectopic expression of ZAK promoted EMT phenotypes and apoptosis resistance in multiple epithelial cell lines, while having different impacts on cell growth in different cell lines. Conversely, depletion of ZAK in aggressive mesenchymal cancer cells reversed EMT phenotypes, increased sensitivity to conventional cytotoxic drugs, and attenuated bone metastasis potential, with little impact on primary tumor growth. Mechanistically, ZAK-mediated EMT is associated with activation of ZEB1 and suppression of epithelial splicing regulatory proteins (ESRPs), which results in a switch in CD44 expression from the epithelial CD44v8-9 isoform to the mesenchymal CD44s isoform. Of note, transcriptomic analysis showed that ZAK overexpression is significantly associated with poor survival in a number of human cancer types. Tissue microarray analysis on breast invasive carcinoma further supported that ZAK overexpression is an independent poor prognostic factor for overall survival in breast cancer. Through combination with ZAK, prognostic accuracy of other common clinicopathological markers in breast cancer is improved by up to 21%. Taken together, these results suggest that promoting EMT is the primary role for ZAK in cancer progression. They also highlight its potential as a biomarker to identify high-risk patients, and suggest its promise as a therapeutic target for inhibiting metastasis and overcoming drug resistance.


Assuntos
Progressão da Doença , Transição Epitelial-Mesenquimal , Neoplasias/enzimologia , Neoplasias/patologia , Proteínas Quinases/metabolismo , Animais , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Feminino , Inativação Gênica , Humanos , MAP Quinase Quinase Quinases , Camundongos Nus , Análise Multivariada , Fenótipo , Prognóstico , Modelos de Riscos Proporcionais , Curva ROC , Fatores de Transcrição/metabolismo
14.
J Food Sci ; 83(2): 535-542, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29350748

RESUMO

Chitosan oligosaccharides (COS), hydrolyzed products of chitosan, was found to display various biological activities. Herein, we assessed the immunostimulatory activity of COS both in in vitro and in vivo studies. In vitro cytotoxicity studies to murine macrophage RAW264.7 revealed that COS is safe even at the maximum tested concentration of 1000 µg/mL. It also stimulates the production of nitric oxide (NO) and tumor necrosis factor (TNF-α) and enhances the phagocytosis in COS-stimulated RAW264.7. We have shown that the COS could significantly (P < 0.05) restore the reduced immune organs indices, phagocytic index, lymphocyte proliferation, natural killer cell activity, and antioxidant enzyme activities in a cyclophosphamide-induced immunosuppressed mice model. COS can also improve the survival rate in irradiation injury mice and significantly (P < 0.05) increased the spleen indices and up-regulates the CD4+/CD8+ ratio in splenocytes. In sum, the aforementioned results suggest that COS might has the potential to be used as an immunostimulatory agent in patients with immune dysfunctions or be a model for functional food development. PRACTICAL APPLICATION: COS might has the potential to be used as an immunostimulatory agent in patients with immune dysfunctions or be a model for functional food development.


Assuntos
Quitosana/administração & dosagem , Ciclofosfamida/efeitos adversos , Fatores Imunológicos/administração & dosagem , Oligossacarídeos/administração & dosagem , Protetores contra Radiação/administração & dosagem , Animais , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Proliferação de Células/efeitos dos fármacos , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Óxido Nítrico/imunologia , Óxido Nítrico Sintase Tipo II/imunologia , Oligossacarídeos/farmacologia , Fagocitose/efeitos dos fármacos , Fator de Necrose Tumoral alfa/imunologia
15.
Chem Biol Interact ; 279: 129-135, 2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29155028

RESUMO

Colon cancer is the third most common cancer, and yet there is a lack of effective therapeutic method with low side effects. Chitosan oligosaccharides (COS) is derived from chitosan after chitin deacetylation, and attracts more interests due to smaller molecular weight and soluble property. Previously, COS, mainly absorbed through intestinal epithelia, has been reported to exhibit many bioactivities, especially its anti-tumor effect. Recent references pay little attention to molecular weight distribution which is crucial for understanding its biological behavior. Here, we studied reducing sugar content and degree of polymerization (DP) of COS. 86.73% reducing sugar exists in COS sample and the content of chitosan fractions with 2-6 is 85.8%. COS suppressed the growth of HCT116 cells in vitro and in vivo, and the inhibition rate of tumor weight in vivo was high up to 58.6%. Moreover, the morphology observation, flow cytometry analysis and mRNA expression were applied to study the apoptosis related mechanism. COS treatment promoted mitosis, late stage apoptosis and S cell cycle arrest in HCT116 cells, and enhanced the mRNA expression of BAK and reduce BCL-2 and BCL-xL. These findings may provide an important clue for clinical applications of COS as anti-tumor drug or pharmaceutic adjuvant in the future.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Quitosana/análogos & derivados , Neoplasias do Colo/tratamento farmacológico , Oligossacarídeos/farmacologia , Animais , Antineoplásicos/química , Quitosana/química , Quitosana/farmacologia , Relação Dose-Resposta a Droga , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Humanos , Camundongos , Camundongos Nus , Oligossacarídeos/química
16.
Oncotarget ; 8(42): 71782-71796, 2017 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-29069746

RESUMO

Cancer drug researchers have been seeking microtubule-inhibiting agents (MIAs) with higher bioactivity and lower toxicity than currently marketed drugs. WX-132-18B, a novel structural compound synthesized at our institute, specifically bound to the colchicine-binding site on tubulin rather than the vinblastine site, and concentration-dependently reduced microtubule content via depolymerization. It exhibited the same cellular phenotypic profiles as the classic MIAs (colchicine, vincristine, and taxol), including inducing cell cycle arrest at the G2/M phase, triggering tumor cell apoptosis, promoting nuclear membrane permeability, reducing mitochondrial membrane potential, and disrupting the redox system balance. Importantly, WX-132-18B displayed more potent in vitro bioactivity (IC50 0.45-0.99 nM) than did the classic MIAs; it inhibited the proliferation of human umbilical vein endothelial cells and seven types of human tumor cells, especially the taxol-resistant breast cancer cells MX-1/T. WX-132-18B also dose-dependently inhibited mice sarcoma, human lung, and gastric cancer xenograft tumors and the formation of tumor blood vessels in mice. In conclusion, WX-132-18B is a novel microtubule-depolymerizing agent that selectively acts on the colchicine-binding site of tubulin and exerts potent in vitro and in vivo anti-tumor effects. These characteristics, along with its anti-angiogenesis and anti-drug resistance properties, make WX-132-18B a promising anti-tumor drug candidate.

17.
J Med Chem ; 60(13): 5586-5598, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28653846

RESUMO

7-Methoxy-4-(2-methylquinazolin-4-yl)-3,4-dihydroquinoxalin-2(1H)-one (2), a promising anticancer lead previously identified by us, inhibited tumor growth by 62% in mice at 1.0 mg/kg without obvious signs of toxicity. Moreover, compound 2 exhibited extremely high antiproliferative activity in the NIH-NCI 60 human tumor cell line panel, with low to sub-nanomolar GI50 values (10-10 M level). It also showed a suitable balance between aqueous solubility and lipophilicity, as well as moderate metabolic stability in vivo. Mechanistic studies using Mayer's hematoxylin and eosin and immunohistochemistry protocols on xenograft tumor tissues showed that 2 inhibited tumor cell proliferation, induced apoptosis, and disrupted tumor vasculature. Moreover, evaluation of new synthetic analogues (6a-6t) of 2 indicated that appropriate 2-substitution on the quinazoline ring could enhance antitumor activity and improve druglike properties. Compound 2 and its analogues with a 4-(2-methylquinazolin-4-yl)-3,4-dihydroquinoxalin-2(1H)-one scaffold thus represent a novel class of tubulin-binding tumor-vascular disrupting agents (tumor-VDAs) that target established blood vessels in tumors.


Assuntos
Inibidores da Angiogênese/química , Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Quinazolinas/química , Quinazolinas/uso terapêutico , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Camundongos Nus , Simulação de Acoplamento Molecular , Neoplasias/irrigação sanguínea , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Quinazolinas/farmacologia , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química , Moduladores de Tubulina/farmacologia , Moduladores de Tubulina/uso terapêutico
18.
Oncol Lett ; 13(6): 4762-4768, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28599477

RESUMO

5-Fluorouracil (5-FU) is one of the most important agents used to treat colorectal cancer. However, the therapeutic effect of 5-FU on colon cancer is limited. SM-1 is a novel type of proapoptotic agent that directly activates procaspase-3 to caspase-3, leading to apoptosis in human cancer cells. The aim of the present study was to evaluate the antitumor effects of 5-FU in combination with SM-1. The human colorectal cancer cell lines HCT116 and LoVo were cultured in the presence of SM-1 and 5-FU. The combination of SM-1 and 5-FU treatment exhibited increased proliferation inhibitory effects compared with 5-FU treatment alone in HCT116 and LoVo cells, as determined using an MTT assay. SM-1 significantly decreased the half-maximal inhibitory concentration of 5-FU from 8.07±0.49 to 2.55±0.41 µmol/l in HCT116 cells, and from 7.90±0.98 to 3.14±0.81 µmol/l in LoVo cells. Similarly, the apoptotic activity was increased to 47.95 and 35.19% in HCT116 and LoVo cells, respectively, as determined using Annexin V/propidium iodide staining and flow cytometry. The combination of SM-1 and 5-FU treatment led to significantly increased caspase-3 activity compared with either compound alone. The reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot analysis revealed the downregulation of B-cell lymphoma 2 and Survivin, and the upregulation of apoptosis regulator Bcl-2-associated X protein and cleaved poly (ADP-ribose) polymerase in HCT116 and LoVo cells. In addition, RT-qPCR identified downregulation of X-linked inhibitor of apoptosis protein mRNA. 5-FU and SM-1 treatment in combination increased tumor proliferation inhibition in HCT116 and LoVo xenograft mouse models of colorectal cancer, compared with SM-1 or 5-FU treatment alone. SM-1 significantly enhanced the antitumor activity of 5-FU in colorectal cancer. These improved effects were due to increased activity of the apoptotic signaling pathway.

19.
J Food Sci ; 81(12): H3035-H3042, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27802366

RESUMO

Chitosan oligosaccharides (COSs) are hydrolysate mixture of chitin and possess various biomedical properties, such as antimicrobial, immunoenhancing, and antitumor effects. Antiproliferation activity of COS and commercially available samples was compared in the terms of A549 and HCT-116 cells. Ten tumor cells were used to estimate cytotoxicity of COS. Although there were some researches on the antitumor effects of COS, we highlighted the in vivo antitumor activities of COS administrated orally and intraperitoneally on S180-bearing/residual mouse. Results turned out that in vitro IC50 values of COS were 48.6 ± 7.0 to 1329.9 ± 93.4 µg/mL against 10 different tumor cell lines. Then, the in vivo experiments proved that the inhibition rate was high up to 58.5%. Significant cell death and necrosis were observed in COS-treated groups by histological analysis. COS stimulated the mRNA expression of tumor necrosis factor alpha. In summary, COS may be considered promising candidate as antitumor functional food or pharmaceutic adjuvant in oncotherapy, especially for patients after surgical resection.


Assuntos
Antineoplásicos/farmacologia , Quitosana/farmacologia , Oligossacarídeos/farmacologia , Células A549 , Administração Oral , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células HCT116 , Humanos , Camundongos , Peso Molecular , Fator de Necrose Tumoral alfa/metabolismo
20.
World J Surg Oncol ; 14(1): 237, 2016 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-27589869

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) is the most frequent cause of cancer deaths worldwide. The targeted therapy had made important progress in recent years, but few potential predictive biomarkers for prognosis of NSCLC patients were identified. Angiopoietin-2 (Ang-2), a cytokine upregulated in tumor endothelial cells and some tumor cells including NSCLC, is a partial agonist and antagonist of angiopoietin-1 (Ang-1). Ang-1 is another ligand for the tyrosine kinase receptor Tie2; it promotes recruitment of pericytes and smooth muscle cells, stabilizing vascular networks by binding to Tie2. Although many studies mainly considered that Ang-2 correlated with progression and prognosis of NSCLC significantly, there are much conflicting and controversial data. Therefore, we conducted a meta-analysis to assess the relationship between Ang-2 and prognosis, a clinical outcome of NSCLC. METHODS: The search was based on major databases from PubMed, Cochrane Library, EMBASE, and CNKI, and 20 eligible publications (range from 2002 to 2015) are included in our meta-analysis with 2011 NSCLC patients in total. These studies illuminated the correlation between the expression of Ang-2 and NSCLC, based on either prognostic factors or clinicopathological features. Pooled calculations were carried out on the odds ratio (OR) and the corresponding 95 % confidence interval (CI) to perform this meta-analysis, and all statistical analyses were carried out by STATA 12.0 and Review Manager 5.3. RESULTS: According to our results, the expression of Ang-2 in NSCLC tissues was significantly higher than that in normal lung tissues, indicating that Ang-2 over-expression may be a predictive marker (pooled OR = 5.09, corresponding 95 % confidence interval (95 % CI) 3.10-8.36, p = 0.000). In addition, our pooled data showed that Ang-2 expression was positively correlated with tumor stages (pooled OR = 3.58, 95 % CI 2.40-5.35, p = 0.000), differentiation (pooled OR = 0.65, 95 % CI 0.45-0.94, p = 0.02), lymphatic invasion (pooled OR = 3.15, 95 % CI 1.97-5.03, p = 0.000), and poor survival (pooled OR = 1.93, 95 % CI 1.47-2.52, p = 0.000) of NSCLC, but seems to have no significant impact on tumor size (pooled OR = 1.09, 95 % CI 0.59-2.00, p = 0.78). CONCLUSIONS: These results demonstrate that Ang-2 expression significantly correlated with poor prognosis for patients with NSCLC.


Assuntos
Angiopoietina-2/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/mortalidade , Angiopoietina-1/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Neoplasias Pulmonares/patologia , Metástase Linfática , Estadiamento de Neoplasias , Taxa de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA