Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Poult Sci ; 102(10): 102995, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37566970

ABSTRACT

Pseudomonas aeruginosa (P. aeruginosa) is a serious zoonotic pathogen threaten the poultry industry causing severe economic losses therefor, this study aimed to isolation, phenotypic, molecular identification of P. aeruginosa from different avian sources (chickens, turkey, pigeons, table eggs, and dead in shell chicken embryos), from different Egyptian governorates (Giza, Qalubia, Beheira, El-Minya, and Al-Sharqia) with applying of antibiotic sensitivity test on all P. aeruginosa isolates. Highly resistant isolates (n = 49) were subjected to molecular identification of P. aeruginosa with detection of resistant genes including carbapenemase-encoding genes blaKPC, blaOXA-48, and blaNDM. On the base of molecular results, a highly resistant P. aeruginosa strain was tested for its pathogenicity on day old specific pathogen free (SPF) chicks. Also, in vitro experiment was adopted to evaluate the efficacy of silver nanoparticles (Ag-NPs) against highly antibiotic-resistant P. aeruginosa strains. The overall isolation percentage was from all examined samples were 36.2% (571/1,576) representing 45.2% (532/1,176) from different birds' tissues and 39/400 (9.7%) from total egg samples. Some of isolated strains showed multidrug resistance (MDR) against kanamycin, amoxicillin, amoxicillin-clavulanic acid, neomycin, chloramphenicol, vancomycin, cefotaxime clavulanic acid, lincomycin-spectinomycin, co-trimoxazole, cefoxitin, gentamycin, and doxycycline. These MDR strains were also molecularly positive for ESBL and carbapenemase-encoding genes. MDR strain showed high pathogenicity with histopathological alterations in different organs in challenged birds. Main histopathological lesions were necrosis of hepatocytes, renal tubular epithelium, and heart muscle bundles. The MDR strain showed in vitro sensitivity to Ag-NPs. In conclusion, MDR P. aeruginosa is a serious pathogen causing high morbidity, mortality, and pathological tissue alterations. Ag NPs revealed a promising in vitro antimicrobial sensitivity against MDR P. aeruginosa and further in vivo studies were recommended.


Subject(s)
Metal Nanoparticles , Pseudomonas Infections , Chick Embryo , Animals , Pseudomonas aeruginosa , Silver/pharmacology , Chickens , Virulence , Drug Resistance, Multiple, Bacterial/genetics , Pseudomonas Infections/veterinary , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Microbial Sensitivity Tests/veterinary
2.
Int J Biol Macromol ; 240: 124339, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37028626

ABSTRACT

This study was designed to synthesize a functionalized chitosan by coupling the amine groups of chitosan with 2,4,6-Trimethoxybenzaldehyde, producing a chitosan Schiff base (Cs-TMB). The development of Cs-TMB was verified employing FT-IR, 1H NMR, the electronic spectrum, and elemental analysis. Antioxidant assays exhibited significant ameliorations of Cs-TMB, reporting scavenging activities of 69.67 ± 3.48 % and 39.65 ± 1.98 % for ABTS•+ and DPPH, respectively, while native chitosan showed scavenging ratios of 22.69 ± 1.13 % and 8.24 ± 0.4.1 % toward ABTS•+ and DPPH, respectively. Besides, Cs-TMB exerted significant antibacterial activity up to 90 % with remarkable bactericidal capacity against virulent gram-negative and gram-positive bacteria compared to the original chitosan. Furthermore, Cs-TMB exhibited a safe profile against normal fibroblast cells (HFB4). Interestingly, flow cytometric analysis showed that Cs-TMB demonstrated prominent anticancer properties of 52.35 ± 2.99 % against human skin cancer cells (A375), compared to 10.66 ± 0.55 % for Cs-treated cells. Moreover, Python and PyMOL in-house scripts were used to predict the interaction of Cs-TMB with the adenosine A1 receptor and visualized as a protein-ligand system submerged in a lipid membrane. Overall, these findings accentuate that Cs-TMB could be a favorable representative for wound dressing formulations and skin cancer treatment.


Subject(s)
Chitosan , Skin Neoplasms , Humans , Antioxidants/pharmacology , Antioxidants/chemistry , Chitosan/pharmacology , Chitosan/chemistry , Spectroscopy, Fourier Transform Infrared , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry
3.
Environ Sci Pollut Res Int ; 29(42): 63294-63303, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35449334

ABSTRACT

For the past few years, the environmental safety problems of radioactive nuclides caused wide public concern. In this work, the dodecyl trimethyl ammonium bromide-modified silicon dioxide composite (DTAB/SiO2) was synthesized for the elimination of uranium. The dodecyl trimethyl ammonium bromide can decorate the surface of the silicon dioxide and change its surface topography, which can offer more active sites and functional groups for the combination of U(VI). The removal capacity of U(VI) on DTAB/SiO2 reached 78.1 mg/g, which was greater than that of the silicon dioxide nanopowder. In the adsorption process, the surface oxygen-containing functional groups formed surface complexation with uranium. The results may provide helpful content to eliminate U(VI) and expand the application of surfactant in radioactive nuclide cleanup.


Subject(s)
Uranium , Adsorption , Bromides , Oxygen , Quaternary Ammonium Compounds , Silicon Dioxide/chemistry , Surface-Active Agents , Uranium/chemistry
4.
J Mater Chem B ; 10(9): 1453-1462, 2022 03 02.
Article in English | MEDLINE | ID: mdl-35188170

ABSTRACT

Exploration of versatile nanoplatforms within one single nanostructure for multidisciplinary treatment modalities, especially achieving a synergistic therapeutic efficacy of combinational gene/photothermal cancer therapy is still a great challenge in biomedicine and nanotechnology. In this study, a unique photothermal nanocarrier has successfully been designed and developed for a combination of gene therapy (GT) and photothermal therapy (PTT) of cancer cells. Surface-engineered iron oxides (α-Fe2O3) nanoparticles (NPs) with poly(3,4-ethylenedioxythiophene) (PEDOT) polymer coatings are synthesized using a one-pot in situ oxidative polymerization method. The results show that the as-prepared α-Fe2O3@PEDOT core-shell NPs with a uniform particle size exhibit positively charged surfaces, facilitating efficient siRNA Bcl-2 (B-cell lymphoma-2) uptake for delivery to breast cancer cells. More importantly, α-Fe2O3@PEDOT core-shell NPs not only display good biocompatibility and water dispersibility but also strong optical absorption enhancement in the Vis-NIR region as compared to α-Fe2O3 NPs. The obtained α-Fe2O3@PEDOT core-shell NPs show an efficient photothermal conversion efficacy (η = 54.3%) and photostability under NIR laser irradiation. As a result, both in vitro and in vivo biological studies on two types of breast cancer cells/tumors treated with α-Fe2O3@PEDOT-siRNA nanocomplexes demonstrate high cancer cell apoptosis and tumor inhibition induced by synergistic GT/PTT therapy under mild conditions compared to an individual GT or PTT alone. Taken together, this is the first example of the use of an α-Fe2O3@PEDOT core-shell nanoagent as a siRNA delivery nanocarrier for highly effective gene/photothermal combination anticancer therapy.


Subject(s)
Breast Neoplasms , Ferric Compounds/chemistry , Phototherapy , Breast Neoplasms/therapy , Bridged Bicyclo Compounds, Heterocyclic , Cell Line, Tumor , Female , Humans , Nanostructures , Phototherapy/methods , Polymers , RNA, Small Interfering/pharmacology
5.
Cancer Cell Int ; 20: 123, 2020.
Article in English | MEDLINE | ID: mdl-32322170

ABSTRACT

BACKGROUND: Kinesin superfamily (KIFs) has a long-reported significant influence on the initiation, development, and progress of breast cancer. However, the prognostic value of whole family members was poorly done. Our study intends to demonstrate the value of kinesin superfamily members as prognostic biomarkers as well as a therapeutic target of breast cancer. METHODS: Comprehensive bioinformatics analyses were done using data from TCGA, GEO, METABRIC, and GTEx. LASSO regression was done to select tumor-related members. Nomogram was constructed to predict the overall survival (OS) of breast cancer patients. Expression profiles were testified by quantitative RT-PCR and immunohistochemistry. Transcription factor, GO and KEGG enrichments were done to explore regulatory mechanism and functions. RESULTS: A total of 20 differentially expressed KIFs were identified between breast cancer and normal tissue with 4 (KIF17, KIF26A, KIF7, KIFC3) downregulated and 16 (KIF10, KIF11, KIF14, KIF15, KIF18A, KIF18B, KIF20A, KIF20B, KIF22, KIF23, KIF24, KIF26B, KIF2C, KIF3B, KIF4A, KIFC1) overexpressed. Among which, 11 overexpressed KIFs (KIF10, KIF11, KIF14, KIF15, KIF18A, KIF18B, KIF20A, KIF23, KIF2C, KIF4A, KIFC1) significantly correlated with worse OS, relapse-free survival (RFS) and distant metastasis-free survival (DMFS) of breast cancer. A 6-KIFs-based risk score (KIF10, KIF15, KIF18A, KIF18B, KIF20A, KIF4A) was generated by LASSO regression with a nomogram validated an accurate predictive efficacy. Both mRNA and protein expression of KIFs are experimentally demonstrated upregulated in breast cancer patients. Msh Homeobox 1 (MSX1) was identified as transcription factors of KIFs in breast cancer. GO and KEGG enrichments revealed functions and pathways affected in breast cancer. CONCLUSION: Overexpression of tumor-related KIFs correlate with worse outcomes of breast cancer patients and can work as potential prognostic biomarkers.

6.
Am J Cancer Res ; 9(3): 529-545, 2019.
Article in English | MEDLINE | ID: mdl-30949408

ABSTRACT

Hypoxia plays an extensive role in the development of the tumor microenvironment (TME), particularly in mediating immunosuppression. Respiratory hyperoxia therapy has the potential to improve the effects of conventional cancer therapies via molecular mechanisms mediating antitumor immunity. Here, we investigated whether hyperoxia therapy can restore tumor immunity and inhibit lung metastases in a mouse model of triple-negative breast cancer (TNBC) by treating a 4T1 mammary carcinoma mouse model with normoxia (21% oxygen) or hyperoxia (60% oxygen) therapy, after tumor development. Using flow cytometry analysis, we observed significant organ-specific expansion of myeloid-derived suppressor cells (MDSCs) and protein expression upregulation of the programmed death-ligand 1 (PD-L1) in the hypoxic TME of 4T1 tumor-bearing mice maintained under normoxia conditions, with the TME converting to a T-cell immune-suppressive state as early as the premetastatic phase. Markedly, hyperoxia treatments ameliorated hypoxia levels in the lung TME and decreased the proportion of MDSCs and the expression of PD-L1 in both the primary tumor and in the metastatic lung, when compared to animals treated with respiratory normoxia therapy. In addition, the number of lung metastatic nodes fell from 90 per lung in the normoxic treated group to 13 per lung in the hyperoxic treated group (P < 0.05), with the latter having limited hyperoxia effects on primary tumor growth (mammary glands). Notably, hyperoxia therapy was characterized by the differential recruitment of CD4+ and CD8+ T-cells. Thus, our study confirms that hyperoxia therapy may be used to overcome TME immunosuppression and control the extend of lung metastases in TNBC. Importantly, changes in immunosuppressive MDSCs frequency and PD-L1 expression levels may serve as biomarkers of hypoxia levels in cancer affected tissues that can benefit from hyperoxia treatments.

7.
J Biomater Appl ; 33(2): 216-226, 2018 08.
Article in English | MEDLINE | ID: mdl-30096997

ABSTRACT

Gene therapy with herpes simplex virus thymidine kinase gene (HSV-TK), which is also known as "suicide" gene therapy, is effective in various tumor models. The lack of a safe and efficient gene delivery system has become a major obstacle to "suicide" gene therapy. In this study, the cytotoxicity and transfection efficiency of graphene oxide-hydroxyapatite (GO-Hap) were analyzed by MTS and flow cytometry, respectively. A series of assays were performed to evaluate the effects of GO-HAp/p-HRE/ERE-Sur-TK combined with ganciclovir treatment on growth of human breast normal and cancer cells. The results showed that GO-HAp nanocomposites effectively transfected cells with minimum toxicity. GO-HAp/p-HRE/ERE-Sur-TK combined with ganciclovir treatment inhibited the proliferation and induced cell apoptosis in cancer cells, while the cytotoxic effects are tolerable in normal breast cells. We conclude that the GO-HAp nanocomposites have significant potential as a gene delivery vector for cancer therapy.


Subject(s)
Breast Neoplasms/therapy , Durapatite/chemistry , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Graphite/chemistry , Simplexvirus/enzymology , Thymidine Kinase/genetics , Antiviral Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Ganciclovir/pharmacology , Gene Transfer Techniques , Genes, Transgenic, Suicide , Genes, Viral , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Humans , Nanocomposites/chemistry , Simplexvirus/genetics , Transfection/methods
8.
Dalton Trans ; 47(33): 11705-11712, 2018 Aug 21.
Article in English | MEDLINE | ID: mdl-30106076

ABSTRACT

Metal-organic framework (MOF)-based materials possess numerous attractive characteristics; however, the application of MOF-based photocatalysts in the area of visible-light photocatalytic H2 evolution is still in its infancy. Herein, we develop a series of novel UiO-66-NH2-based composites with embedded NiO nanoparticles via solvothermal treatment and subsequent calcination. Their characterizations demonstrate intimate lattice-level contacts between UiO-66-NH2 photocatalysts and NiO nanoparticles. By optimizing each component, even without noble metal loading, the U6N-NiO-2 sample (the weight ratio of NiO to U6N-NiO-2 is theoretically calculated to be ca. 10 wt%) with 15 mg eosin Y as a sensitizer causes an enhanced H2 generation rate of 2561.32 µmol h-1 g-1 under visible-light irradiation using TEOA as a sacrificial reagent; furthermore, its corresponding quantum efficiency is as high as 6.4% at 420 nm. The H2 evolution activity of U6N-NiO-2 is about 5 times higher than that of the UiO-66-NH2 photocatalyst (denoted as U6N) and 23 times higher than that of U6N-NiO-2 without sensitizer. It is demonstrated that the high efficiency originates from the visible-light generated electrons of eosin Y and UiO-66-NH2, the efficient separation of carriers by the cascaded band structure and more negative CB of NiO as well as the good dispersion of NiO nanoparticles on the octahedral skeleton. This study provides new insights for the design of MOF-based materials without noble metal loading for visible-light photocatalytic H2 evolution.

9.
Eur J Pharmacol ; 819: 58-67, 2018 Jan 15.
Article in English | MEDLINE | ID: mdl-29183837

ABSTRACT

Previous studies have shown that melatonin can protect cells against rotenone-induced cell death. Yet, the mechanism involved in this protection requires further research. In this study, we aimed to further investigate the effects of melatonin on inhibiting rotenone-induced SH-SY5Y cells and the underlying molecular mechanisms. Human neuroblastoma SH-SY5Y cells were treated with 0.3 or 1µM rotenone for 6 or 12h. Cell viability was measured with an MTS assay, the mitochondrial membrane potential was determined with a Rhodamine 123 staining assay, and the protein expression levels of the markers of autophagy, including cytochrome C release (Cyt C), light chain 3B (LC3 B) and Dynamin-Related Protein 1 (Drp1) were analyzed by western blotting. The co-localization of Drp1 and TOM20 proteins in the mitochondria of SH-SY5Y cells was measured by immunofluorescence coupled with confocal microscopy and the overexpression of the Drp1 gene was then conducted. The viability and expression levels of Cyt C and LC3 B in rotenone and melatonin + rotenone-treated Drp1-overexpressed SH-SY5Y cells were analyzed with MTS and western blotting, respectively. We found that rotenone effectively induced SH-SY5Y cell death by causing mitochondrial dysfunction and increasing Cyt C expression. Drp1 expression and its regulation of mitochondrial translocation mediated the rotenone-induced cell death and melatonin inhibited this process. Overexpression of Drp1 protein attenuated melatonin's inhibition of rotenone-induced SH-SY5Y cell death. In conclusion, melatonin effectively inhibits rotenone-induced neuronal cell death via the regulation of Drp1 expression.


Subject(s)
Down-Regulation/drug effects , GTP Phosphohydrolases/metabolism , Melatonin/pharmacology , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins/metabolism , Rotenone/antagonists & inhibitors , Rotenone/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cytochromes c/metabolism , Cytoprotection/drug effects , Dose-Response Relationship, Drug , Dynamins , GTP Phosphohydrolases/genetics , Humans , Microtubule-Associated Proteins/genetics , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Neurons/cytology , Neurons/drug effects , Protein Transport/drug effects
10.
Int J Oncol ; 49(6): 2442-2452, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27748911

ABSTRACT

Evidence suggests that RING1 and YY1 binding protein (RYBP) functions as a tumor suppressor. However, its role in breast cancer remains unclear. In the present study, the expression of RYBP was assessed in breast cancer patients and cell lines. Disease-free survival durations of breast cancer patients with high RYBP expression were determined based on the ATCG dataset. The effects of RYBP overexpression on cell growth, migration and invasive potency were also assessed. Nude mouse xenograft and lung metastasis models were also used to confirm the role of RYBP. The involvement of SRRM3 in RYBP-mediated breast cancer suppression was explored using SRRM3 siRNA. The potential relationship between RYBP, SRRM3, and REST-003 was examined by qPCR. The results showed that RYBP was downregulated in breast cancer patients and in several breast cancer cell lines. Breast cancer patients with high expression levels of RYBP displayed better disease-free survival. Overexpression of RYBP in MDA-MB-231 and SK-BR-3 cells significantly decreased cell proliferation, migration, and invasion ability, and increased the proportion of cells arrested in S-phase compared with the negative control cells. Additionally, upregulation of proliferation-related cell cycle proteins (cyclin A and cyclin B1) and E-cadherin, and downregulation of snail were observed in RYBP-overexpressing cells. Overexpression of RYBP reduced tumor volume and weight as well as metastatic foci in the lungs of nude mice. SRRM3 knockdown by siRNA, which is downregulated after RYBP overexpression, suppressed cell growth and metastasis in MDA-MB-231 and SK-BR-3 cells. Furthermore, qPCR analysis revealed that REST-003 ncRNA was downregulated in cells overexpressing RYBP and in SRRM3-inhibited cells. Moreover, cell invasion ability and growth were increased after SRRM3 upregulation in RYBP-overexpressing cells, but they were decreased following si-REST-003 transfection. In conclusion, overexpression of RYBP suppresses breast cancer growth and metastasis both in vitro and in vivo. SRRM3 and REST-003, which are downregulated in cells overexpressing RYBP, may be involved in RYBP-mediated breast cancer progression.


Subject(s)
Breast Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Proteins/genetics , Repressor Proteins/genetics , Animals , Breast Neoplasms/genetics , Cadherins/biosynthesis , Cell Line, Tumor , Cyclin A1/biosynthesis , Cyclin B1/biosynthesis , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Polycomb Repressive Complex 1/metabolism , RNA Interference , RNA, Small Interfering/genetics , Snail Family Transcription Factors/biosynthesis , Xenograft Model Antitumor Assays , YY1 Transcription Factor/metabolism
11.
J Transl Med ; 14: 54, 2016 Feb 17.
Article in English | MEDLINE | ID: mdl-26883017

ABSTRACT

BACKGROUND: Chromosome 14 open reading frame 166 (C14orf166) is upregulated in various tumors, but its role in breast cancer has not been reported. METHODS: Quantitative real-time PCR and western blot were used to determine C14orf166 expression in normal breast epithelial cells (NBEC), breast cancer cells, and four matched pairs of breast cancer tissues and adjacent noncancerous tissues. Using immunohistochemistry, we determined C14orf166 expression in paraffin-embedded tissues from 121 breast cancer patients. Statistical analyses were performed to examine the associations among C14or166 expression, clinicopathological parameters and prognosis outcome of breast cancer. MTT and colony formation assay were used to determine the effect of C14orf166 on cell proliferation by overexpression or knockdown of C14orf166 level. RESULTS: C14orf166 was upregulated in breast cancer cell lines and tissues compared with the normal cells and adjacent normal breast tissues, high C14orf166 expression was positively with advancing clinical stage. The correlation analysis between C14orf166 expression and clinicopathological characteristics suggested C14orf166 expression was significantly correlated with clinical stages, T classification, N classification and PR expression, Kaplan-Meier curves with log rank tests showed patients with low C14orf166 expression had better survival, Cox-regression analysis suggested C14orf166 was an unfavorable prognostic factor for breast cancer patients. C14orf166 overexpression promoted breast cancer cell proliferation, whereas knockdown of C14orf166 inhibited this effect. Further analysis found C14orf166 overexpression inhibited cell cycle inhibitors P21 and P27 expression, and increased the levels of Cyclin D1 and phosphorylation of Rb, suggesting C14orf166 contributed to cell proliferation by regulating G1/S transition. CONCLUSION: Our findings suggested C14orf166 could be a novel prognostic biomarker of breast cancer, it also contributes to cell proliferation by regulating G1/S transition.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease Progression , Trans-Activators/metabolism , Adult , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Statistics, Nonparametric , Trans-Activators/genetics , Treatment Outcome , Up-Regulation
12.
Oncol Lett ; 10(3): 1870-1874, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26622767

ABSTRACT

The transcriptional factor Osterix is specifically expressed in bone tissues to regulate the differentiation and maturation of osteoblasts. Recent studies have also identified the expression of Osterix in a number of cancer tissues, such as kidney and lung cancers. However, the association of Osterix with the metastasis of breast cancers has never been reported. The present study, for the first time, provides evidence supporting the involvement of Osterix in breast cancer metastasis. Western blotting was employed to investigate the expression of Osterix in a number of human breast cancer cell lines with different metastatic features. Gain-of-function and loss-of-function experiments were performed in MCF7 cells (low level of metastasis) and MDA-MB-361 cells (high level of metastasis). The expression of several metastasis-associated genes was analyzed by western blotting and quantitative polymerase chain reaction. A firefly luciferase-based reporter gene assay was conducted in order to study whether Osterix regulated the promoter activities of the MMP2 and MMP9 genes, which play critical roles in cancer metastasis. The results showed that Osterix was highly expressed in the MDA-MB-231 and MDA-MB-361 cells, but was not detectable in the MCF7 cells. The overexpression of Osterix in the MCF7 cells promoted the expression of VEGF, MMP9 and ß-catenin, while downregulating the expression of E-cadherin. In addition, suppression of Osterix expression in the MDA-MB-361 cells reversed the alteration of VEGF, MMP9, ß-catenin and E-cadherin expression. A reporter gene assay suggested that Osterix activated MMP2 and MMP9 promoter activity. In conclusion, Osterix is involved in the metastasis of human breast cancer and may be a target for the efficient treatment of human breast cancers.

13.
Int J Nanomedicine ; 10: 4255-66, 2015.
Article in English | MEDLINE | ID: mdl-26170662

ABSTRACT

BACKGROUND: Small interfering RNA (siRNA)-mediated gene therapy is a promising strategy to temporarily inhibit the expression of genes involved in development of breast cancer. The lack of a safe and efficient gene delivery system has become a major hurdle for siRNA-mediated gene therapy in breast cancer. Our previous studies have demonstrated that inorganic amorphous calcium carbonate (ACC) hybrid nanospheres functionalized with CaIP6 (ACC/CaIP6) nanoparticles are an efficient nucleic acid delivery tool. The present study aimed to evaluate the safety and efficiency of ACC/CaIP6 in delivering siRNA targeting AKT1 (siAKT1) for the treatment of breast cancer. METHODS: The cytotoxicity of the ACC/CaIP6 nanoparticles was evaluated using a tetrazolium assay. The transfection efficiency and intracellular distribution of ACC/siAKT1 were analyzed by flow cytometry and confocal laser scanning microscopy, respectively. A series of in vitro and in vivo assays was performed to evaluate the effects of ACC/CaIP6/siAKT1 on growth of breast cancer cells. RESULTS: ACC/CaIP6 nanoparticles effectively transfected cells with little or no toxicity. AKT1 knockdown by ACC/CaIP6/siAKT1 inhibited cell cycle progression and promoted apoptosis of MCF-7 cells. Intratumoral injection of ACC/CaIP6/siAKT1 significantly suppressed the growth of breast cancer in mice. CONCLUSION: ACC/CaIP6 nanoparticles are a safe and efficient method of delivering siRNA for gene therapy in breast cancer.


Subject(s)
Antineoplastic Agents , Calcium Carbonate/chemistry , Inositol Phosphates/chemistry , Nanocomposites/chemistry , Proto-Oncogene Proteins c-akt/genetics , RNA, Small Interfering , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Breast Neoplasms/chemistry , Cell Proliferation/drug effects , Humans , MCF-7 Cells , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , RNA, Small Interfering/pharmacology , Xenograft Model Antitumor Assays
14.
J Nanobiotechnology ; 12: 37, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25266303

ABSTRACT

BACKGROUND: Inhalation of chemotherapeutic drugs directly into the lungs augments the drug exposure to lung cancers. The inhalation of free drugs however results in over exposure and causes severe adverse effect to normal cells. In the present study, epidermal growth factor (EGF)-modified gelatin nanoparticles (EGNP) was developed to administer doxorubicin (DOX) to lung cancers. RESULTS: The EGNP released DOX in a sustained manner and effectively internalized in EGFR overexpressing A549 and H226 lung cancer cells via a receptor-mediated endocytosis. In vitro cytotoxicity assay showed that EGNP effectively inhibited the growth of A549 and H226 cells in a dose-dependent manner. In vivo biocompatibility study showed that both GNP and EGNP did not activate the inflammatory response and had a low propensity to cause immune response. Additionally, EGNP maintained a high therapeutic concentration in lungs throughout up to 24 h comparing to that of free drug and GNP, implying the effect of ligand-targeted tumor delivery. Mice treated with EGNP remarkably suppressed the tumor growth (~90% tumor inhibition) with 100% mice survival rate. Furthermore, inhalation of EGNP resulted in elevated levels of cleaved caspase-3 (apoptotic marker), while MMP-9 level significantly reduced comparing to that of control group. CONCLUSIONS: Overall, results suggest that EGF surface-modified nanocarriers could be delivered to lungs via inhalation and controlled delivery of drugs in the lungs will greatly improve the therapeutic options in lung cancer therapy. This ligand-targeted nanoparticulate system could be promising for the lung cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Lung Neoplasms/drug therapy , Nanoparticles/administration & dosage , Neoplasm Metastasis/drug therapy , Animals , Caspase 3/metabolism , Cell Line, Tumor , Doxorubicin/administration & dosage , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays/methods
15.
J Mater Chem B ; 2(18): 2714-2724, 2014 May 14.
Article in English | MEDLINE | ID: mdl-32261437

ABSTRACT

To overcome drug resistance, the combination of two or more therapeutic strategies with different mechanisms has received much attention in recent years. In this study, a common approach has been used to process curcumin and Zn2+ into colloidal dispersions known as "nanoparticles", which are cheap and easy to prepare with high reproducibility. This novel vehicle has good biocompatibility and high cellular uptake for simultaneously delivering the curcumin drug and siRNA into tumor cells. Complexation of Zn2+ with curcumin enhances the aqueous solubility of the hydrophobic drug curcumin and further improves the cellular uptake and bioavailability. The acid-labile coordination Zn(ii)-O bond in Zn(ii)-curcumin drug nanoparticles (Zn(ii)-Cur NPs) can respond to tumor intracellular acidic pH environments to release curcumin, and promoting acid-triggered intracellular drug release. The positively charged Zn(ii)-Cur NPs can efficiently deliver siRNA into human bladder cancer cells, protect siRNA against enzymatic degradation, and facilitate the escape of loaded siRNA from the endosome into the cytoplasm, which successfully downregulates the targeted EIF5A2 oncogene and consequently inhibits cancer cell growth in vitro and in vivo. Proliferation and migration of cancer cells are inhibited by silencing the expression of EIF5A2 and increasing the ratio of pro-apoptotic BAX to anti-apoptotic BCL-2. In vitro and in vivo experiments have demonstrated that bifunctional Zn(ii)-Cur NPs/siEIF5A2 can combine chemotherapy with gene therapy to afford higher therapeutic efficacy than the individual therapeutic protocols.

16.
World J Surg Oncol ; 11: 161, 2013 Jul 18.
Article in English | MEDLINE | ID: mdl-23866030

ABSTRACT

BACKGROUND: Although many studies have indicated that high-mobility group box 1 protein (HMGB1) is associated with oncogenesis and a worse prognosis, the prognostic value of HMGB1 in gastric cancer (GC) remains unclear. In the present work, we aimed to evaluate the role of HMGB1 in GC and examined whether aberrant expression of both HMGB1 and vascular endothelial growth factor C (VEGF-C) increased the malignant potential of GC. METHODS: A total of 166 GC patients and 32 normal subjects were enrolled. HMGB1 and VEGF-C expression was detected by tissue microarrays (TMAs) and immunohistochemical staining. The correlation between HMGB1 and VEGF-C expression and their relationships with clinicopathological GC variables were examined. Univariate and multivariate analyses were performed using the Cox proportional hazard model to predict the factors related to the patients' overall survival rates. RESULTS: HMGB1 and VEGF-C expression were observed in 81 (48.80%) and 88 (53.01%) tumors, respectively, significantly higher than the rates among the corresponding controls. In addition, HMGB1 and VEGF-C expression were positively correlated (R2 = 0.972). HMGB1 expression was also closely associated with tumor size, pT stage, nodal status, metastasis status, TNM stage, and poor prognosis. Multivariate survival analysis indicated that patients with HMGB1 and VEGF-C coexpression had the worst prognoses and survival rates (hazard ratio, 2.78; log rank P<0.001). CONCLUSIONS: HMGB1 is commonly expressed in GC. Combined evaluation of HMGB1 and VEGF-C may serve as a valuable independent prognostic factor for GC patients.


Subject(s)
Adenocarcinoma/mortality , HMGB1 Protein/metabolism , Neoplasm Recurrence, Local/mortality , Stomach Neoplasms/mortality , Vascular Endothelial Growth Factor C/metabolism , Adenocarcinoma/secondary , Adenocarcinoma/surgery , Biomarkers, Tumor/metabolism , Case-Control Studies , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Neoplasm Staging , Prognosis , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Survival Rate , Tissue Array Analysis
17.
Biomaterials ; 34(4): 1246-54, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23127333

ABSTRACT

Previously, we reported that inorganic amorphous calcium carbonate (ACC) hybrid nanospheres functionalized with Ca(II)-IP6 compound (CaIP6) is a promising gene vector in vitro. Here, nonviral gene carrier, ACC/CaIP6 nanocomposite particles (NPACC/CaIP6), was evaluated for efficient in vitro and in vivo delivery of small interfering RNA (siRNA) targeting human Amplified in breast cancer 1 (AIB1). The nanoparticle is capable of forming ACC/CaIP6 nanoparticle-siRNA complexes and transferring siRNA into targeted cells with high transfection efficiency. Meanwhile the ACC/CaIP6 nanoparticle-siRNA complexes have no obvious cytotoxicity for human bladder cancer T24 cells. Furthermore, NPACC/CaIP6 effectively protected the encapsulated siRNA from degradation, AIB1 knockdown mediated by ACC/CaIP6/siRNA complexes transfection resulted in cells proliferation inhibition, apoptosis induction and cell cycle arrest in vitro. NPACC/CaIP6 exhibited well tissues penetrability in localized siRNA delivering, intratumoral injection of NPACC/CaIP6/siAIB1 could attenuate tumor growth and downregulation of PI3K/Akt signaling pathway in vivo. We conclude that ACC/CaIP6 nanoparticle is a promising system for effective delivery of siRNA for cancer gene therapy.


Subject(s)
Calcium Carbonate/chemistry , Nanocapsules/administration & dosage , RNA, Small Interfering/administration & dosage , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/therapy , Animals , Cell Line, Tumor , Female , Humans , Mice , Nanocapsules/chemistry , RNA, Small Interfering/chemistry , Treatment Outcome
18.
Int J Nanomedicine ; 7: 1061-7, 2012.
Article in English | MEDLINE | ID: mdl-22403488

ABSTRACT

Nanoparticles have an enormous potential for development in biomedical applications, such as gene or drug delivery. We developed and characterized aminopropyltriethoxysilane-functionalized silicon dioxide nanoparticles (APTES-SiNPs) for gene therapy. Lipofectamine(®) 2000, a commonly used agent, served as a contrast. We showed that APTES-SiNPs had a gene transfection efficiency almost equal to that of Lipofectamine 2000, but with lower cytotoxicity. Thus, these novel APTES-SiNPs can achieve highly efficient transfection of plasmid DNA, and to some extent reduce cytotoxicity, which might overcome the critical drawbacks in vivo of conventional carriers, such as viral vectors, organic polymers, and liposomes, and seem to be a promising nonviral gene therapy vector.


Subject(s)
DNA/chemistry , Genetic Vectors/chemistry , Nanoparticles/chemistry , Silanes/chemistry , Silicon Dioxide/chemistry , Transfection/methods , Cell Survival/drug effects , Cells, Cultured , DNA/pharmacokinetics , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Genetic Vectors/pharmacokinetics , Genetic Vectors/pharmacology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lipids/chemistry , Lipids/pharmacokinetics , Lipids/pharmacology , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Particle Size , Propylamines , Silanes/pharmacology
19.
Mol Med Rep ; 5(3): 610-8, 2012 03.
Article in English | MEDLINE | ID: mdl-22159329

ABSTRACT

Marine fungal metabolite 1386A is a newly identified small molecular compound extracted from the mangrove fungus 1386A in the South China Sea. Preliminary experiments have demonstrated its amazing cytotoxity to cancer cells, while the mechanism remains poorly understood. microRNAs (miRNAs) are a newly identified class of small regulatory RNAs which play an important role in gene regulation at the post-transcriptional level. They usually function as oncogenes or tumor suppressors and are related to drug sensitivity and resistance. We aimed to test the hypothesis that the potential antineoplastic compound, 1386A, alters the miRNA profile in MCF-7 and whether its unknown mechanism may be predicted by analysis of the altered miRNA profile. Cell proliferation was analyzed by MTT assay. The alteration of the miRNA expression profile of MCF-7 cells was investigated using advanced microarray technology. Silico analysis using TargetScan was used to predict the putative targeted transcripts encoding the dysregulated miRNAs. 1386A inhibited MCF-7 cell proliferation in a time- and dose-dependent manner (the IC50 value at 48 h was 17.1 µmol/l). 1386A (17.1 µmol/l) significantly altered the global miRNA expression profile of the MCF-7 cells at 48 h. Forty-five miRNAs were differentially expressed in MCF-7 cells. Target prediction suggested that these miRNAs potentially target many oncogenes and tumor-suppressor genes associated with cancer development, progression and metastasis. The promising antineoplastic compound marine fungal metabolite 1386A alters the miRNA profiles of MCF-7 breast cancer cells. Analyzing the alteration of the miRNA profile caused by this potential antineoplastic compound may help to predict the unknown mechanism of 1386A.


Subject(s)
Antineoplastic Agents/pharmacology , Fungi/metabolism , Gene Expression Regulation, Neoplastic/drug effects , MicroRNAs/metabolism , Antineoplastic Agents/chemistry , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Microarray Analysis
20.
Arterioscler Thromb Vasc Biol ; 31(9): 2044-53, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21817107

ABSTRACT

OBJECTIVE: The goal of this study was to determine the expression signature and the potential role of microRNAs in human arteries with arteriosclerosis obliterans (ASO). METHODS AND RESULTS: The expression profiles of microRNAs in human arteries with ASO and in normal control arteries were determined by quantitative reverse transcription-polymerase chain reaction array. Among the 617 detected microRNAs, multiple microRNAs were aberrantly expressed in arteries with ASO. Some of these dysregulated microRNAs were further verified by quantitative reverse transcription-polymerase chain reaction. Among them, microRNA-21 (miR-21) was mainly located in arterial smooth muscle cells (ASMCs) and was increased by more than 7-fold in ASO that was related to hypoxia inducible factor 1-α. In cultured human ASMCs, cell proliferation and migration were significantly decreased by inhibition of miR-21. 3'-Untranslated region luciferase assay confirmed that tropomyosin 1 was a target of miR-21 that was involved in miR-21-mediated cellular effects, such as cell shape modulation. CONCLUSION: The results suggest that miR-21 is able to regulate ASMC function by targeting tropomyosin 1. The hypoxia inducible factor-1 α/miR-21/tropomyosin 1 pathway may play a critical role in the pathogenesis of ASO. These findings might provide a new therapeutic target for human ASO.


Subject(s)
Arteriosclerosis Obliterans/etiology , Lower Extremity/physiopathology , MicroRNAs/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Tropomyosin/physiology , Actins/chemistry , Arteriosclerosis Obliterans/genetics , Arteriosclerosis Obliterans/physiopathology , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , MicroRNAs/analysis , Muscle, Smooth, Vascular/cytology , Tropomyosin/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...