Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Nanobiotechnology ; 22(1): 296, 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38811964

RESUMEN

BACKGROUND: Combination therapy involving immune checkpoint blockade (ICB) and other drugs is a potential strategy for converting immune-cold tumors into immune-hot tumors to benefit from immunotherapy. To achieve drug synergy, we developed a homologous cancer cell membrane vesicle (CM)-coated metal-organic framework (MOF) nanodelivery platform for the codelivery of a TLR7/8 agonist with an epigenetic inhibitor. METHODS: A novel biomimetic codelivery system (MCM@UN) was constructed by MOF nanoparticles UiO-66 loading with a bromodomain-containing protein 4 (BRD4) inhibitor and then coated with the membrane vesicles of homologous cancer cells that embedding the 18 C lipid tail of 3M-052 (M). The antitumor immune ability and tumor suppressive effect of MCM@UN were evaluated in a mouse model of triple-negative breast cancer (TNBC) and in vitro. The tumor immune microenvironment was analyzed by multicolor immunofluorescence staining. RESULTS: In vitro and in vivo data showed that MCM@UN specifically targeted to TNBC cells and was superior to the free drug in terms of tumor growth inhibition and antitumor immune activity. In terms of mechanism, MCM@UN blocked BRD4 and PD-L1 to prompt dying tumor cells to disintegrate and expose tumor antigens. The disintegrated tumor cells released damage-associated molecular patterns (DAMPs), recruited dendritic cells (DCs) to efficiently activate CD8+ T cells to mediate effective and long-lasting antitumor immunity. In addition, TLR7/8 agonist on MCM@UN enhanced lymphocytes infiltration and immunogenic cell death and decreased regulatory T-cells (Tregs). On clinical specimens, we found that mature DCs infiltrating tumor tissues of TNBC patients were negatively correlated with the expression of BRD4, which was consistent with the result in animal model. CONCLUSION: MCM@UN specifically targeted to TNBC cells and remodeled tumor immune microenvironment to inhibit malignant behaviors of TNBC.


Asunto(s)
Receptor Toll-Like 7 , Receptor Toll-Like 8 , Neoplasias de la Mama Triple Negativas , Microambiente Tumoral , Animales , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas , Ratones , Femenino , Humanos , Línea Celular Tumoral , Microambiente Tumoral/efectos de los fármacos , Nanopartículas/química , Factores de Transcripción/metabolismo , Ratones Endogámicos BALB C , Proteínas de Ciclo Celular/metabolismo , Inmunoterapia/métodos , Epigénesis Genética/efectos de los fármacos , Proteínas que Contienen Bromodominio
2.
J Nanobiotechnology ; 21(1): 170, 2023 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237294

RESUMEN

BACKGROUND: Sepsis is a syndrome of physiological, pathological and biochemical abnormalities caused by infection. Although the mortality rate is lower than before, many survivors have persistent infection, which means sepsis calls for new treatment. After infection, inflammatory mediators were largely released into the blood, leading to multiple organ dysfunction. Therefore, anti-infection and anti-inflammation are critical issues in sepsis management. RESULTS: Here, we successfully constructed a novel nanometer drug loading system for sepsis management, FZ/MER-AgMOF@Bm. The nanoparticles were modified with LPS-treated bone marrow mesenchymal stem cell (BMSC) membrane, and silver metal organic framework (AgMOF) was used as the nanocore for loading FPS-ZM1 and meropenem which was delivery to the infectious microenvironments (IMEs) to exert dual anti-inflammatory and antibacterial effects. FZ/MER-AgMOF@Bm effectively alleviated excessive inflammatory response and eliminated bacteria. FZ/MER-AgMOF@Bm also played an anti-inflammatory role by promoting the polarization of macrophages to M2. When sepsis induced by cecal ligation and puncture (CLP) challenged mice was treated, FZ/MER-AgMOF@Bm could not only reduce the levels of pro-inflammatory factors and lung injury, but also help to improve hypothermia caused by septic shock and prolong survival time. CONCLUSIONS: Together, the nanoparticles played a role in combined anti-inflammatory and antimicrobial properties, alleviating cytokine storm and protecting vital organ functions, could be a potential new strategy for sepsis management.


Asunto(s)
Nanopartículas , Sepsis , Ratones , Animales , Macrófagos/metabolismo , Antibacterianos/uso terapéutico , Sepsis/tratamiento farmacológico , Membrana Celular/metabolismo , Modelos Animales de Enfermedad
3.
Nanoscale ; 15(21): 9457-9476, 2023 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-37161583

RESUMEN

A synergistic combination of treatment with immunogenic cell death (ICD) inducers and immunoadjuvants may be a practical way to boost the anticancer response and successfully induce an immune response. The use of HR@UCNPs/CpG-Apt/DOX, new biomimetic drug delivery nanoparticles generated to combat breast cancer, is reported here as a unique strategy to produce immunogenicity and boost cancer immunotherapy. HR@UCNPs/CpG-Apt/DOX (HR-UCAD) consists of two parts. The core is composed of an immunoadjuvant CpG (a toll-like receptor 9 agonist) fused with a dendritic cell-specific aptamer sequence (CpG-Apt) to decorate upconversion nanoparticles (UCNPs) with the successful intercalation of doxorubicin (DOX) into the consecutive base pairs of Apt-CpG to construct an immune nanodrug UCNPs@CpG-Apt/DOX. The targeting molecule hyaluronic acid (HA) was inserted into a red blood cell membrane (RBCm) to form the shell (HR). HR-UCAD possessed a strong capacity to specifically induce ICD. Following DOX-induced ICD of cancer cells, sufficient exposure to tumor antigens and UCNPs@CpG-Apt (UCA) activated the tumor-specific immune response and reversed the immunosuppressive tumor microenvironment. In addition, HR-UCAD has good biocompatibility and increases the active tumor-targeting effect. Furthermore, HR-UCAD exhibits excellent near-infrared upconversion luminescence emission at 804 nm under irradiation with a 980 nm laser, which has great potential in biomedical imaging. Thus, the RBCm-camouflaged drug delivery system is a promising targeted chemotherapy and immunotherapy nanocomplex that could be used for effective targeted breast cancer treatment.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Nanopartículas , Humanos , Femenino , Membrana Eritrocítica , Antineoplásicos/farmacología , Doxorrubicina , Neoplasias de la Mama/tratamiento farmacológico , Inmunoterapia , Adyuvantes Inmunológicos , ADN , Línea Celular Tumoral , Microambiente Tumoral
4.
J Nanobiotechnology ; 21(1): 89, 2023 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-36918874

RESUMEN

Strategies to overcome toxicity and drug resistance caused by chemotherapeutic drugs for targeted therapy against hepatocellular carcinoma (HCC) are urgently needed. Previous studies revealed that high oxidored-nitro domain-containing protein 1(NOR1) expression in HCC was associated with cisplatin (DDP) resistance. Herein, a novel dual-targeting nanocarrier system AR-NADR was generated for the treatment of DDP resistance in HCC. The core of the nanocarrier system is the metal-organic frameworks (MOF) modified with nuclear location sequence (NLS), which loading with DDP and NOR1 shRNA (R). The shell is an A54 peptide inserted into the erythrocyte membrane (AR). Our results show that AR-NADR efficiently internalized by tumor cells due to its specific binding to the A54 receptors that are abundantly expressed on the surface of HCC cells and NLS peptide-mediated nuclear entry. Additionally, DDP is more likely to be released due to the degradation of Ag-MOF in the acidic tumor microenvironment. Moreover, by acting as a vector for gene delivery, AR-NADR effectively inhibits tumor drug resistance by suppressing the expression of NOR1, which induces intracellular DDP accumulation and makes cells sensitive to DDP. Finally, the anti-HCC efficacy and mechanisms of AR-NADR were systematically elucidated by a HepG2/DDP cell model as well as a tumor model. Therefore, AR-NADR constitutes a key strategy to achieve excellent gene silencing and antitumor efficacy, which provides effective gene therapy and precise treatment strategies for cisplatin resistance in HCC.


Asunto(s)
Antineoplásicos , Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Carcinoma Hepatocelular/metabolismo , Biomimética , Neoplasias Hepáticas/patología , Resistencia a Antineoplásicos , Línea Celular Tumoral , Antineoplásicos/uso terapéutico , Microambiente Tumoral
5.
Front Chem ; 9: 522708, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33796503

RESUMEN

Zirconia nanoparticles (ZrO2 NPs) are commonly used in the field of biomedical materials, but their antitumor activity and mechanism is unclear. Herein, we evaluated the anti-tumor activity of ZrO2 NPs and explored the anti-tumor mechanism. The results of in vitro and in vivo experiments showed that the level of intracellular reactive oxygen species (ROS) in HeLa cells was elevated after ZrO2 NPs treatment. Transmission electron microscopy (TEM) showed that after treatment with ZrO2 NPs, the mitochondria of HeLa cells were swollen, accompanied with the induction of autophagic vacuoles. In addition, flow cytometry analysis showed that the apoptotic rate of HeLa cells increased significantly by Annexin staining after treatment with ZrO2 NPs, and the mitochondrial membrane potential (MMP) was reduced significantly. The proliferation of HeLa cells decreased as indicated by reduced Ki-67 labeling. In contrast, TUNEL-positive cells in tumor tissues increased after treatment with ZrO2 NPs, which is accompanied by increased expression of mitochondrial apoptotic proteins including Bax, Caspase-3, Caspase-9, and Cytochrome C (Cyt C) and increased expression of autophagy-related proteins including Atg5, Atg12, Beclin-1, and LC3-II. Treating HeLa cells with N-acetyl-L-cysteine (NAC) significantly reduced ROS, rate of apoptosis, MMP, and in vivo anti-tumor activity. In addition, apoptosis- and autophagy-related protein expressions were also suppressed. Based on these observations, we conclude that ZrO2 NPs induce HeLa cell death through ROS mediated mitochondrial apoptosis and autophagy.

6.
Exp Ther Med ; 21(4): 355, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33732328

RESUMEN

Currently, bone marrow transplantation remains the basic treatment for various hematological tumors and irradiation is one of the most important pretreatment methods. However, irradiation pretreatment may result in damage to bone mesenchymal stem cells (BMSCs). The present study aimed to investigate the effect of circular RNA-016901 (circ-016901) on the injury of irradiation-induced BMSCs and the underlying mechanism. The expression levels of circ-016901, microRNA-1249-5p (miR-1249-5p) and homeodomain interacting protein kinase 2 (HIPK2) in irradiation-induced mouse BMSCs at various irradiation doses were detected via reverse transcription-quantitative PCR (RT-qPCR). The effect of circ-016901 on cell proliferation was examined using Cell Counting Kit-8 assays following silencing or overexpression of circ-016901. Cell apoptosis was detected by flow cytometry and caspase-3/7 activity. The expression of autophagy-related markers, including Beclin-1 and LC3-II/I, was detected at the mRNA and protein levels by RT-qPCR and western blotting, respectively. Irradiation treatment upregulated the expression of circ-016901 and HIPK2 and downregulated miR-1249-5p expression. The expression levels of LC3-II/I and Beclin-1 in BMSCs were downregulated in a dose-dependent manner. Silencing of circ-016901 promoted proliferation of irradiation-induced BMSCs and attenuated irradiation-induced apoptosis. Moreover, silencing of circ-016901 elevated the expressions of LC3-II/I and Beclin-1 in irradiation-induced BMSCs. Similar results were obtained with miR-1249-5p overexpression and HIPK2 silencing. These results demonstrated that circ-016901 silencing attenuated injury in irradiation-induced mouse BMSCs by regulating the miR-1249-5p/HIPK2 axis, providing a novel target for future research on the mechanism of radiation resistance in BMSCs.

7.
Oncol Res ; 28(5): 553-557, 2020 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-33349308

RESUMEN

Colorectal cancer (CRC) is one of the most common malignancies in the world, with a high incidence and a high mortality. However, the pathogenesis of CRC carcinogenesis is still unexplored. In this study, we investigated the role of miR-107 in the regulation of CRC cell proliferation and apoptosis. First, the expression of miR-107 was observed to be aberrantly increased in human CRC tumor tissues and cell lines when compared to the colonic control tissues and colon epithelial cells. Further study showed that the proliferative and apoptotic capacities of human CRC SW480 and LoVo cells were aberrantly regulated by miR-107. The proliferation of SW480 and LoVo cells was remarkably enhanced by the miR-107 mimic but suppressed by the miR-107 inhibitor when compared to the negative control. On the contrary, the apoptotic rate of both SW480 and LoVo cells was significantly inhibited by miR-107 overexpression but increased by miR-107 inhibition. In addition, we identified prostate apoptosis response-4 (Par4) as a direct target of miR-107 with a potential binding site on the 3-UTR of mRNA, as evaluated by bioinformatics prediction and luciferase reporter assay. Par4 expression levels were significantly inhibited by the miR-107 mimic but upregulated by the miR-107 inhibitor in both SW480 and LoVo cells. Compared to the control, the increase in Par4 expression significantly inhibited the induction role of miR-107 in the proliferation of SW480 and LoVo cells, and the apoptotic rate of cells repressed by the miR-107 mimic was also reversed by Par4 overexpression. In summary, our results demonstrated that miR-107 exerts a positive role in the survival of CRC cells by directly targeting Par4. This might reveal a novel understanding about human CRC pathogenesis.

8.
Front Chem ; 8: 565, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32766207

RESUMEN

Jolkinolide B (JB) is a bioactive compound isolated from a Chinese herbal medicine that exerts antitumor activity. However, the anti-lymphoma effect of JB and its mechanism are yet to be revealed. Because free JB has poor pharmacokinetics and weak antitumor efficacy, we opted to use black phosphorus quantum dot (BPQD) nanomaterials as a drug loading platform to synthesize a nano-traditional Chinese medicine (nano-TCM) called BPQDs@JB. Compared with free JB, Raji cells administrated with BPQDs@JB exhibited the cell viability of 19.85 ± 1.02%, and the production of intracellular reactive oxygen species (ROS) was promoted. Likewise, BPQDs@JB was capable of rising the apoptosis rate of Raji cells to 34.98 ± 1.76%. In nude mice transplanted tumor model administrated with BPQDs@JB, the tumor tissue sections administrated with BPQDS@JB achieved a conspicuous red fluorescence, demonstrating the presence of most ROS production in the BPQDS@JB. TUNEL achieved a number of positive (brown) nuclei in vivo, revealing that BPQDS@JB could significantly induce tumor tissue apoptosis. As revealed from the mentioned results, BPQDs@JB can generate considerable ROS and interfere with the redox state to inhibit tumor. In brief, BPQDs@JB may be adopted as a treatment option for lymphoma.

9.
Front Chem ; 8: 377, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32457875

RESUMEN

Zirconia nanoparticles (ZrO2 NPs) are widely applied in the field of biomedicine. In this study, we constructed a nanoplatform of ZrO2 NPs coated with a platelet membrane (PLTm), named PLT@ZrO2. PLTm nanovesicles camouflage ZrO2 NPs, prevent nanoparticles from being cleared by macrophage, and target tumor sites. Compared to ZrO2 alone, PLT@ZrO2 is better at inhibiting the invasion and metastasis of Hela cells in vitro and in vivo. In vitro, PLT@ZrO2 inhibited the growth and proliferation of Hela cells. Scratch-wound healing recovery assay demonstrated that PLT@ZrO2 inhibited Hela cells migration. Transwell migration and invasion assays showed that PLT@ZrO2 inhibited Hela cells migration and invasion. In vivo, PLT@ZrO2 inhibited the tumor growth of Xenograft mice and inhibited the lung and liver metastasis of Hela cells. Immunofluorescence and Western blotting results showed that anti-metastasis protein (E-cadherin) was upregulated and pro-metastasis proteins (N-cadherin, Smad4, Vimentin, E-cadherin,ß-catenin, Fibronectin, Snail, Slug, MMP2, Smad2) were down-regulated. Our study indicated that PLT@ZrO2 significantly inhibits tumor growth, invasion, and metastasis.

10.
ACS Appl Mater Interfaces ; 12(20): 22687-22701, 2020 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-32330381

RESUMEN

Toxicity and drug resistance caused by chemotherapeutic drugs have become bottlenecks in treating tumors. The delivery of anticancer drugs based on nanocarriers is regarded as an ideal way to solve the aforementioned problems. In this study, a new antilymphoma nanodrug CD20 aptamer-RBCm@Ag-MOFs/PFK15 (A-RAMP) is designed and constructed, and it consists of two parts: (1) metal-organic frameworks Ag-MOFs (AM) loaded with tumor aerobic glycolysis inhibitor PFK15 (P), forming a core part (AMP); (2) targeted molecule CD20 aptamer (A) is inserted into the red blood cell membrane (RBCm) to form the shell part (A-R). A-RAMP under the guidance of CD20 aptamer actively targets B-cell lymphoma both in vitro and in vivo. As a result, A-RAMP not only significantly inhibits the effect on tumor growth but also shows no obvious side effects on the treated nude mice, indicating that A-RAMP can accurately target tumor cells, reprogram aerobic glycolysis, and exert synergistic antitumor effect by Ag+ and PFK 15. Furthermore, the antitumor mechanism of A-RAMP in vivo by apoptotic pathway and targeting metabonomics are explored. These results suggest that A-RAMP has a promising application prospect as an smart, safe, effective, and synergistic antilymphoma agent.


Asunto(s)
Antineoplásicos/uso terapéutico , Portadores de Fármacos/química , Glucólisis/efectos de los fármacos , Linfoma de Células B/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Nanocompuestos/química , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Aptámeros de Nucleótidos/química , Secuencia de Bases , Materiales Biomiméticos/química , Membrana Eritrocítica/química , Humanos , Células K562 , Estructuras Metalorgánicas/química , Ratones , Ratones Endogámicos BALB C , Piridinas/uso terapéutico , Quinolinas/uso terapéutico , Células RAW 264.7 , Plata/química , Plata/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Onco Targets Ther ; 13: 473-486, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32021291

RESUMEN

BACKGROUND: Isatin derivatives have extensive biological activities, such as antitumor. IF203, a novel isatin derivative, has not previously been reported to have antitumor activity. METHODS: Acid phosphatase assays (APAs) and Ki-67 immunohistochemistry were used to detect the proliferation of HepG2 cells. Transmission electron microscope (TEM) was applied to detect ultrastructural changes. Flow cytometry (FCM) was used to detect cell cycle, apoptosis, reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) of HepG2 cells in vitro. TUNEL, MMP and ROS immunofluorescence assays were applied to assess apoptosis, MMP, and ROS of HepG2 cells in vivo. Western Blotting was applied to assess the levels of apoptosis- and autophagy-related proteins. RESULTS: In this study, in vivo and in vitro experiments showed that IF203 possesses antitumor activity. The results of APAs and Ki-67 immunohistochemistry demonstrated that IF203 could inhibit the proliferation of HepG2 cells. Cell cycle assays, downregulation of Cyclin B1 and Cdc2, and upregulation of P53 suggested that IF203 could lead to G2/M cell cycle arrest. In addition, ultrastructural changes, apoptosis assays, TUNEL immunofluorescence results, upregulated expression of Bax, and downregulated expression of Bcl-2 suggest that IF203 can induce apoptosis in HepG2 cells. After IF203 treatment, intracellular ROS levels increased, MMP decreased, JC-1 green fluorescence was enhanced, and the levels of Caspase-9, Caspase-3 and Cytochrome C expression were upregulated, suggesting that IF203 could induce apoptosis of HepG2 cells through the mitochondrial apoptosis pathway. Moreover, characteristic apoptotic ultrastructural changes were accompanied by the appearance of many autophagy bubbles and upregulation of Atg5, Atg12, ULK1, Beclin-1 and LC3-II proteins, suggesting that IF203 could induce autophagy in HepG2 cells. CONCLUSION: This study showed that IF203 leads to the death of HepG2 cells through cell cycle arrest, apoptotic induction, and autophagy promotion.

12.
Front Oncol ; 9: 1346, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31867274

RESUMEN

Background: Nasopharyngeal carcinoma (NPC) is a malignant nasopharyngeal disease with a complicated etiology that occurs mostly in southern China. Intestinal flora imbalance is believed to be associated with a variety of organ malignancies. Current studies revealed that the destruction of intestinal flora is associated with NPC, and many studies have shown that intestinal flora can be used as a biomarker for many cancers and to predict cancer. Methods: To compare the differences in intestinal flora compositions and biological functions among 8 patients with familial NPC (NPC_F), 24 patients with sporadic NPC (NPC_S), and 27 healthy controls (NOR), we compared the intestinal flora DNA sequencing and hematological testing results between every two groups using bioinformatic methods. Results: Compared to the NOR group, the intestinal flora structures of the patients in the NPC_F and NPC_S groups showed significant changes. In NPC_F, Clostridium ramosum, Citrobacter spp., Veillonella spp., and Prevotella spp. were significantly increased, and Akkermansia muciniphila and Roseburia spp. were significantly reduced. In NPC_S, C. ramosum, Veillonella parvula, Veillonella dispar, and Klebsiella spp. were significantly increased, and Bifidobacterium adolescentis was significantly reduced. A beta diversity analysis showed significant difference compared NPC_F with NOR based on Bray Curtis (P = 0.012) and Unweighted UniFrac (P = 0.0045) index, respectively. The areas under the ROC curves plotted were all 1. Additionally, the concentrations of 5-hydroxytryptamine (5-HT) in NPC_F and NPC_S were significantly higher than those of NOR. C. ramosum was positively correlated with 5-HT (rcm: 0.85, P < 0.001). A functional analysis of the intestinal flora showed that NPC_F was associated with Neurodegenerative Diseases (P = 0.023) and that NPC_S was associated with Neurodegenerative Diseases (P = 0.045) as well. Conclusion: We found that NPC was associated with structural imbalances in the intestinal flora, with C. ramosum that promoted the elevation of 5-HT and opportunistic pathogens being significantly increased, while probiotics significantly decreased. C. ramosum can be used as a novel biomarker and disease prediction models should be established for NPC. The new biomarkers and disease prediction models may be used for disease risk prediction and the screening of high-risk populations, as well as for the early noninvasive diagnosis of NPC.

13.
Cell Death Discov ; 5: 121, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31372241

RESUMEN

Hepatocellular carcinoma (HCC) is one of the most common malignant tumors and a leading cause of cancer-related deaths worldwide. Emerging studies have shown that circular RNAs (circRNAs) are differentially expressed in HCC and play an important role in HCC pathogenesis and metastasis. However, the mechanism of circRNA in the chemoresistance of HCC remains unclear. In this study, we aimed to investigate the role of circRNA in cisplatin resistance of HCC. We identified a novel circRNA circRNA_101505 that was decreased in cisplatin-resistant HCC tissues and cell lines, and associated with a poor survival outcome. Gain-of-function investigations showed that overexpression of circRNA_101505 suppressed cancer cell growth in vivo and in vitro, and enhanced cisplatin toxicity in HCC cells. Mechanistic studies found that circRNA_101505 could sensitize HCC cells to cisplatin by sponging miR-103, and thereby promoting oxidored-nitro domain-containing protein 1 (NOR1) expression. In conclusion, the significant inhibitory effects indicate circRNA_101505 to be a potential therapeutic target for HCC treatment. Our findings provide significant evidence to further elucidate the therapeutic use of circRNA in HCC.

14.
Mikrochim Acta ; 186(5): 314, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31041529

RESUMEN

Alpha-fetoprotein (AFP) is a reliable clinical marker of hepatocellular carcinoma (HCC). A highly sensitive fluorometric aptamer nanoprobe is described for AFP detection. It is based on fluorescence resonance energy transfer (FRET) between AFP aptamer labelled with 5-carboxyfluorescein (FAM) and palladium nanoparticles (PdNPs). The PdNPs quench the green fluorescence of the FAM-AFP aptamer via interactions between nitrogen functional groups of the AFP aptamer and PdNPs. When AFP was introduced into the FAM-AFP aptamer-PdNPs FRET system, the AFP aptamer preferentially combines with AFP. This results in a conformational change and weakens the interaction between the aptamer and the PdNPs. Thus, the fluorescence of FAM recovers. The fluorescence recovery of FAM increases linearly in the 5.0-150 ng·mL-1 AFP concentration range and has a 1.4 ng·mL-1 detection limit. The assay was applied to the analysis of spiked diluted human serum. The recovery values ranged from 98.3 to 112.9%, with relative standard deviations of <1.1%. This biosensing strategy provides a reliable and ultrasensitive protocol for the quantification of biomarkers with relevant antigens and aptamers. Graphical abstract Schematic presentation of a fluorometric aptamer nanoprobe for AFP assay based on fluorescence resonance energy transfer (FRET) between AFP aptamer labelled with 5-carboxyfluorescein (FAM) and palladium nanoparticles (PdNPs).


Asunto(s)
Aptámeros de Nucleótidos/química , Técnicas Biosensibles/métodos , Fluoresceínas/química , Nanopartículas del Metal/química , Nanoestructuras/química , Paladio/química , alfa-Fetoproteínas/análisis , Animales , Aptámeros de Nucleótidos/genética , Secuencia de Bases , Estudios de Factibilidad , Transferencia Resonante de Energía de Fluorescencia , Fluorometría , Humanos , Concentración de Iones de Hidrógeno
15.
Bosn J Basic Med Sci ; 19(2): 155-163, 2019 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-30640591

RESUMEN

Hematopoietic stem cell transplantation is commonly used in patients with certain hematological or bone marrow tumors. Total body irradiation combined with chemotherapy is part of the preconditioning protocol that was the most commonly used before hematopoietic stem cell transplantation. However, total body irradiation preconditioning damages other normal cells in bone marrow. Therefore, exploring the mechanism of radiation resistance in bone marrow mesenchymal stem cells is of great significance for recovering the hematopoietic function after cell transplantation. This study aimed to demonstrate the miR-29b adsorption of circRNA_014511 and explore the effect of circRNA_014511 on radiosensitivity of bone marrow mesenchymal stem cells. In this study, circRNA_014511 overexpression vector was constructed and transfected into bone marrow mesenchymal stem cells, miR-29b-2-5p and P53 were found to be decreased, which could be reversed by miR29b-mimics. Dual luciferase reporter assay confirmed the binding of circRNA_014511 and mmu-miR-29b-2-5p. Flow cytometry analysis showed the apoptosis rate of bone marrow mesenchymal stem cells overexpressing circRNA_014511 was significantly decreased. In the circRNA_014511 transfection group, after cells were subjected to 6Gy irradiation, G2 phase arrest appeared, the expression of P21 and GADD45A was significantly decreased, and cyclin B1 was significantly increased. Colony formation assay showed the survival fraction of circRNA_014511 overexpression cells after irradiation was significantly higher than control group, and the radiosensitivity was decreased. In conclusion,our findings demonstrated that circRNA_014511 could inhibit the expression of P53 by binding miR-29b-2-5p, and decrease the radiosensitivity of bone marrow mesenchymal stem cells by affecting cell cycle and cell apoptosis.


Asunto(s)
Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de la radiación , MicroARNs/metabolismo , ARN Circular/genética , Proteína p53 Supresora de Tumor/metabolismo , Adsorción , Animales , Apoptosis , Células de la Médula Ósea/citología , Ciclo Celular/efectos de la radiación , Proteínas de Ciclo Celular/metabolismo , Ciclina B1/metabolismo , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Ratones , Unión Proteica , Tolerancia a Radiación
16.
Oncol Rep ; 41(3): 1487-1496, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30592293

RESUMEN

Our previous study demonstrated that bromodomain­containing protein 7 (BRD7) inhibits cell proliferation and tumor growth, restoring the expression of B­cell lymphoma 2 antagonist/killer (Bak) sensitized breast cancer cells to paclitaxel. However, the association between BRD7 and paclitaxel sensitization, as well as BRD7 and Bak in breast cancer remains unknown. In the present study, immunochemical staining was performed to measure the expression of BRD7 and Bak in breast cancer tissues. Cell Counting Kit­8 assay, flow cytometry and tumor xenograft procedures were performed to evaluate the biological role of BRD7 and Bak in breast cancer cells. Western blotting, reverse transcription­quantitative polymerase chain reaction, chromatin immunoprecipitation and luciferase reporter assays were also performed. BRD7 was positively correlated with Bak levels in breast cancer tissues, and the survival rate of patients with low Bak and BRD7 expression was significantly lower than that of patients with high Bak and BRD7 expression. In addition, BRD7 activated Bak promoter activity and induced Bak expression in an indirect manner. Furthermore, ectopic expression of BRD7 inhibited cell proliferation, tumor growth and sensitized cancer cells to paclitaxel, while knockdown of Bak abolished BRD7­mediated inhibitory effects on cell proliferation and paclitaxel sensitization in breast cancer cells whether in vitro and in vivo. The results demonstrated that BRD7 inhibits cell proliferation and sensitizes breast cancer cells to paclitaxel by activating Bak; they also provide promising targets for the diagnosis and treatment of breast cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Neoplasias de la Mama Masculina/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Cromosómicas no Histona/metabolismo , Paclitaxel/farmacología , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Antineoplásicos Fitogénicos/uso terapéutico , Apoptosis/efectos de los fármacos , Mama/patología , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Neoplasias de la Mama Masculina/mortalidad , Neoplasias de la Mama Masculina/patología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Células MCF-7 , Masculino , Persona de Mediana Edad , Paclitaxel/uso terapéutico , Regiones Promotoras Genéticas/genética , Tasa de Supervivencia , Proteína Destructora del Antagonista Homólogo bcl-2/genética
17.
Toxicol Sci ; 167(1): 269-281, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30239936

RESUMEN

Graphene is a two-dimensional crystal that is stripped from pristine graphite and made of single layer of carbon atoms. Containing numerous functional groups, graphene derivatives (GDs) could be easily modified and have aroused great attention for potential applications in biomedicine. However, pristine graphene and graphene oxide (GO) could arouse cell and animal toxicity. To screen GDs with high biocompatibility applied for biomedicine, general comparison was performed about the toxicities of six GDs with diverse types of surface modification, size, and redox state, including GO, reduced GO (rGO), graphene quantum dot (GQD), aminated GQD (GQD-NH2), carboxyl GQD (GQD-COOH), and graphene oxide quantum dot (GOQD). In contrast, it was found that large particle size, oxidation state, high concentration, and long exposure time were unfavorable factors affecting the cell viability. We further explored the mechanism of different toxicity, which could be contribute to cell membrane destruction by sharpened edges of GDs (LDH release, hemolysis), ROS production, immuno-inflammatory responses, and activation of apoptotic pathways (IKK/IκBα/NF-κB and BAX/BCL-2). Overall, our combined data primarily explored the related biochemical and molecular mechanism underlying the biological behaviors and toxicity of GDs, and we also identified GQD, GQD-NH2, GQD-COOH, and GOQD could be safely used for biomedical application as drug carriers.


Asunto(s)
Materiales Biocompatibles/toxicidad , Portadores de Fármacos/toxicidad , Grafito/toxicidad , Hepatocitos/efectos de los fármacos , Puntos Cuánticos/toxicidad , Aminación , Apoptosis/efectos de los fármacos , Materiales Biocompatibles/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Portadores de Fármacos/química , Grafito/química , Hepatocitos/metabolismo , Humanos , Oxidación-Reducción , Tamaño de la Partícula , Puntos Cuánticos/química , Especies Reactivas de Oxígeno/metabolismo , Propiedades de Superficie
18.
Leuk Res ; 70: 67-73, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29886398

RESUMEN

The development of circular RNA (circRNA) microarrays has facilitated studies on the role of circRNAs in regulating gene expression through a circRNA-miRNA-mRNA network. In our study, a microarray was used to detect the expression profiles of circRNAs in mouse bone marrow stromal cell (MSCs) after total body irradiation (TBI) in an X-ray field. Forty mice were randomly distributed into two groups, the control group and the TBI group, with 20 mice in each. Using circRNA microarray data, we compared the circRNA expression profiles in the MSCs between the two groups. Randomly chosen differentially expressed circRNAs were validated as potential TBI biomarkers. Overall, 148 circRNAs were significantly upregulated while the other 133 were downregulated between the TBI group and the sham group. Among them, two upregulated circRNAs (mmu_circRNA_011235 and mmu_circRNA_016901) and three downregulated circRNAs (mmu_circRNA_008488, mmu_circRNA_000551 and mmu_circRNA_005365) exhibited trends comparable to microarray data in the validation experiments. The miRNA support vector regression method was used to predict potential target miRNAs for the circRNAs while ClueGO was used for functional analysis of predicted miRNA target genes. This is the first study to investigate the differential circRNA expression profile in TBI and the associated functions. Significantly upregulated or downregulated circRNAs may represent novel biomarkers and novel therapeutic targets for the clinical management of radiation injury with TBI.


Asunto(s)
Regulación de la Expresión Génica/efectos de la radiación , Células Madre Mesenquimatosas/metabolismo , ARN , Transcriptoma , Irradiación Corporal Total , Animales , Biomarcadores , Biología Computacional/métodos , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Ratones , MicroARNs , ARN Circular , ARN Mensajero , Reproducibilidad de los Resultados
19.
Oncol Res ; 25(6): 967-974, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27938501

RESUMEN

Colorectal cancer (CRC) is one of the most common malignancies in the world, with a high incidence and a high mortality. However, the pathogenesis of CRC carcinogenesis is still unexplored. In this study, we investigated the role of miR-107 in the regulation of CRC cell proliferation and apoptosis. First, the expression of miR-107 was observed to be aberrantly increased in human CRC tumor tissues and cell lines when compared to the colonic control tissues and colon epithelial cells. Further study showed that the proliferative and apoptotic capacities of human CRC SW480 and LoVo cells were aberrantly regulated by miR-107. The proliferation of SW480 and LoVo cells was remarkably enhanced by the miR-107 mimic but suppressed by the miR-107 inhibitor when compared to the negative control. On the contrary, the apoptotic rate of both SW480 and LoVo cells was significantly inhibited by miR-107 overexpression but increased by miR-107 inhibition. In addition, we identified prostate apoptosis response-4 (Par4) as a direct target of miR-107 with a potential binding site on the 3'-UTR of mRNA, as evaluated by bioinformatics prediction and luciferase reporter assay. Par4 expression levels were significantly inhibited by the miR-107 mimic but upregulated by the miR-107 inhibitor in both SW480 and LoVo cells. Compared to the control, the increase in Par4 expression significantly inhibited the induction role of miR-107 in the proliferation of SW480 and LoVo cells, and the apoptotic rate of cells repressed by the miR-107 mimic was also reversed by Par4 overexpression. In summary, our results demonstrated that miR-107 exerts a positive role in the survival of CRC cells by directly targeting Par4. This might reveal a novel understanding about human CRC pathogenesis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , MicroARNs/genética , Regiones no Traducidas 3' , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Neoplasias Colorrectales/mortalidad , Regulación Neoplásica de la Expresión Génica , Humanos
20.
Oncol Lett ; 11(5): 3003-3008, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27123053

RESUMEN

Hepatocarcinogenesis is a stepwise process during which multiple genes are altered. Understanding the molecular mechanisms that induce hepatocarcinogenesis may improve the screening, prevention and treatment of patients with hepatocellular carcinoma (HCC). In recent years, the oxidored-nitro domain-containing protein 1 (NOR1) gene has been identified to have an important role in the development of HCC in vitro experiments. The current study aimed to examine the expression of NOR1 mRNA and protein expression in specimens of normal liver, hepatitis, cirrhosis and HCC, together representing the process of HCC development. Furthermore, the association between NOR1 expression and clinicopathological parameters of HCC patients was analyzed. Tissue microarrays containing the specimens of human normal liver, hepatitis, cirrhosis and HCC were purchased, and in situ hybridization and immunohistochemistry were used to detect the expression of NOR1 mRNA and protein expression, respectively. It was revealed that the positive rate of NOR1 protein and mRNA expression in the specimens of hepatitis and cirrhosis were not significantly different from that in the normal liver samples. However, the specimens of HCC exhibited an increased positive rate of NOR1 protein and mRNA expression in comparison with the normal liver samples. In addition, a higher positive rate of NOR1 protein expression was observed in HCC patients with a poor pathological differentiation grade and high tumor node metastasis (TNM) stage. In conclusion, the present study provides evidence, for the first time, of the increased expression of NOR1 in human HCC tissues, and its correlation with the pathological stage and TNM status. These findings indicate that NOR1 may be involved in the progression of HCC and it could be employed as a predictive biomarker in HCC development.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA