Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Cell Death Dis ; 15(4): 294, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664379

ABSTRACT

Although many important advances have been made in the treatment of nasopharyngeal carcinoma (NPC) in recent years, local recurrence and distant metastasis remain the main factors affecting NPC prognosis. Biomarkers for predicting the prognosis of NPC need to be urgently identified. Here, we used whole-exon sequencing (WES) to determine whether PICK1 mutations are associated with the prognosis of NPC. Functionally, PICK1 inhibits the proliferation and metastasis of NPC cells both in vivo and in vitro. Mechanistically, PICK1 inhibited the expression of proteins related to the Wnt/ß-catenin signaling pathway. PICK1 restrained the nuclear accumulation of ß-catenin and accelerated the degradation of ß-catenin through the ubiquitin-proteasome pathway. The reduced PICK1 levels were significantly associated with poor patient prognosis. Hence, our study findings reveal the mechanism by which PICK1 inactivates the Wnt/ß-catenin signaling pathway, thereby inhibiting the progression of NPC. They support PICK1 as a potential tumor suppressor and prognostic marker for NPC.


Subject(s)
Biomarkers, Tumor , Carrier Proteins , Cell Proliferation , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Nuclear Proteins , Wnt Signaling Pathway , Humans , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/metabolism , Prognosis , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Cell Line, Tumor , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/metabolism , Animals , Carrier Proteins/metabolism , Carrier Proteins/genetics , beta Catenin/metabolism , Mice, Nude , Male , Female , Mice , Gene Expression Regulation, Neoplastic , Cell Movement , Mutation/genetics
2.
Life Sci ; 321: 121594, 2023 May 15.
Article in English | MEDLINE | ID: mdl-36934971

ABSTRACT

AIMS: Hypoxia is an important feature of nasopharyngeal carcinoma (NPC). "Protein interacting with PRKCA 1" (PICK1) is commonly downregulated in human malignancies and is functionally related to poor prognosis. However, there is a limited understanding of the upstream mechanisms regulating PICK1 currently. MAIN METHODS: PICK1 and HIF-1α expression levels were analyzed by Immunohistochemistry (IHC), western blotting, and quantitative real-time PCR assay. Protein stability and ubiquitin assays were used to investigate PICK1 protein degradation. Immunofluorescence and co-immunoprecipitation assays were used to demonstrate the interaction between RBCK1 and PICK1. Gene knockdown by siRNA transfection was used to investigate the role of HIF-1α and RBCK1 in hypoxia-induced PICK1 degradation. Cell Counting Kit-8 (CCK-8), 5-Ethynyl-2'-deoxyuridine (EdU) assays and subcutaneous xenograft nude models were used to explore the roles of RBCK1 and PICK1 in NPC cell proliferation. KEY FINDINGS: PICK1 expression in NPC tissue was negatively relative to that of HIF-1α. HIF-1α downregulated PICK1 expression by facilitating its ubiquitination by the E3 ligases RANBP2-type and C3HC4-type zinc finger containing 1 (RBCK1), thereby enhancing proteasome-mediated PICK1 degradation. RBCK1 knockdown inhibited NPC cell proliferation, which was ameliorated by double knockdown of RBCK1/PICK1. SIGNIFICANCE: These data provide evidence for an NPC cell adaptation mechanism to hypoxia, where HIF-1α regulates RBCK1, which targets PICK1 for degradation to promote cell proliferation.


Subject(s)
Nasopharyngeal Neoplasms , Ubiquitin-Protein Ligases , Humans , Nasopharyngeal Carcinoma , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Cell Proliferation , Adaptor Proteins, Signal Transducing/metabolism , Nasopharyngeal Neoplasms/genetics , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Cell Line, Tumor , Carrier Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism
3.
Theranostics ; 12(7): 3420-3437, 2022.
Article in English | MEDLINE | ID: mdl-35547775

ABSTRACT

Rationale: Development of intelligent radiosensitization nanoplatforms for imaging-guided tumor radiotherapy (RT) remains challenging. We report here the construction of an intelligent nanoplatform based on poly(N-vinylcaprolactam) (PVCL) nanogels (NGs) co-loaded with gold (Au) and manganese dioxide (MnO2) nanoparticles (NPs) for dual-mode computed tomography (CT)/magnetic resonance (MR) imaging-guided "full-process" sensitized RT of tumors. Methods: PVCL NGs were synthesized via precipitation polymerization and in situ loaded with Au and MnO2 NPs. The created PVCL-Au-MnO2 NGs were well characterized and systematically examined in their cytotoxicity, cellular uptake, intracellular oxygen and ·OH production, and cell cycle arrest in vitro, evaluated to disclose their RT sensitization effects of cancer cells and a tumor model, and assessed to validate their dual-mode CT/MR imaging potential, pharmacokinetics, biodistribution, and biosafety in vivo. Results: The formed PVCL-Au-MnO2 NGs with a size of 121.5 nm and good stability can efficiently generate reactive oxygen species through a Fenton-like reaction to result in cell cycle distribution toward highly radiosensitive G2/M phase prior to X-ray irradiation, sensitize the RT of cancer cells under X-ray through the loaded Au NPs to induce the significant DNA damage, and further prevent DNA-repairing process after RT through the continuous production of O2 catalyzed by MnO2 in the hybrid NGs to relieve the tumor hypoxia. Likewise, the in vivo tumor RT can also be guided through dual mode CT/MR imaging due to the Au NPs and Mn(II) transformed from MnO2 NPs. Conclusion: Our study suggests an intelligent PVCL-based theranostic NG platform that can achieve "full-process" sensitized tumor RT under the guidance of dual-mode CT/MR imaging.


Subject(s)
Manganese Compounds , Nanoparticles , Cell Line, Tumor , Magnetic Resonance Imaging , Nanogels , Oxides , Polymers , Tissue Distribution , Tomography, X-Ray Computed
4.
Front Cell Dev Biol ; 10: 892575, 2022.
Article in English | MEDLINE | ID: mdl-35557942

ABSTRACT

Radiation-induced rectal injury is a common side effect of radiotherapy. Hypoxia often occurs after radiotherapy. This study aimed to explore the bystander effect of hypoxia on radiation-induced rectal injury. In vivo, apoptosis increased nearby the highly hypoxic area in the rectal tissues in the mouse models of radiation-induced rectal injury, indicating the potential involvement of hypoxia. In vitro, flow cytometry and Western blotting showed that both hypoxia and hypoxic human intestinal epithelial crypt (HIEC) cell supernatant promoted apoptosis in normoxic HIEC cells. The pro-apoptotic effect of extracellular vesicles (EVs) derived from hypoxic HIEC cell to normoxic HIEC cells was then determined. MiR-122-5p was chosen for further studies through a microRNA (miRNA) microarray assay and apoptosis was alleviated in cells receiving miR-122-5p inhibiting hypoxic EVs. Together, our study demonstrated that the miR-122-5p containing-EVs derived from hypoxic HIEC cells promoted apoptosis in normoxic HIEC cells. Hypoxic EV-derived miR-122-5p plays a critical pathologic role in radiation-induced rectal injury and may be a potential therapeutic target.

5.
Biomed Pharmacother ; 151: 113114, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35594704

ABSTRACT

Radiation therapy offers limited clinical benefits for patients with pancreatic cancer, partly as a result of the predominantly immunosuppressive microenvironment characteristic of this specific type of cancer. A large number of abnormal blood vessels and high-density fibrous matrices in pancreatic cancer will lead to hypoxia within tumor tissue and hinder immune cell infiltration. We used low-dose X-ray irradiation, also known as low-dose radiation therapy (LDRT), to normalize the blood vessels in pancreatic cancer, while simultaneously administering an inhibitor of focal adhesion kinase (FAK) to reduce pancreatic cancer fibrosis. We found that this treatment successfully reduced pancreatic cancer hypoxia, increased immune cell infiltration, and increased sensitivity to radiation therapy for pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Tumor Microenvironment , X-Ray Therapy , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/radiotherapy , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Focal Adhesion Protein-Tyrosine Kinases/therapeutic use , Humans , Hypoxia , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/radiotherapy , Tumor Microenvironment/immunology , X-Ray Therapy/methods , Pancreatic Neoplasms
6.
Biochem Biophys Res Commun ; 554: 49-55, 2021 05 21.
Article in English | MEDLINE | ID: mdl-33774279

ABSTRACT

Radiation-induced rectal injury is one of the common side effects of pelvic radiation therapy. This study aimed to explore the role of nuclear factor erythroid 2-related factor 2 (Nrf2) in this process. In vivo, knockout (KO) of Nrf2 led to aggravated radiation-induced histological changes in the rectums. In vitro, interference or overexpression of Nrf2 resulted in enhanced or reduced radiosensitivity in human intestinal epithelial crypts (HIEC) cells, respectively. A potential relationship between Nrf2 and necroptosis was identified using RNA sequencing (RNA-seq) and western blotting (WB), which showed that necroptosis-related proteins were negatively correlated with Nrf2. Upon treatment with necrostatin-1 (Nec-1), the increased radiosensitivity, decreased cell viability, increased γH2AX foci formation, and decreased mitochondrial membrane potential (MMP) in Nrf2-interfered HIEC cells were alleviated. A significant recovery in morphological alterations was also observed in Nrf2 KO mice administered with Nec-1. Taken together, our results highlight the important protective effect of Nrf2 in radiation-induced rectal injury through the inhibition of necroptosis, and the physiological significance of necroptosis in radiation-induced rectal injury.


Subject(s)
NF-E2-Related Factor 2/metabolism , Radiation Injuries/metabolism , Radiation Injuries/pathology , Rectum/radiation effects , Animals , Apoptosis/radiation effects , Cell Line , Cell Survival/radiation effects , DNA Damage , Disease Models, Animal , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/genetics , Necroptosis , Radiation Tolerance , Rectum/metabolism , Rectum/pathology
7.
Theranostics ; 11(4): 1721-1731, 2021.
Article in English | MEDLINE | ID: mdl-33408777

ABSTRACT

Development of a powerful sensitization system to alleviate radioresistance for enhanced tumor radiotherapy (RT) remains to be explored. Herein, we present a unique dual-mode endogenous and exogenous nanosensitizer based on dendrimer-entrapped gold nanoparticles (Au DENPs) to realize enhanced tumor RT. Methods: Generation 5 poly(amidoamine) dendrimers partially modified with 1,3-propanesultone were used for templated synthesis of Au NPs, and the created zwitterionic Au DENPs were adopted for serum-enhanced delivery of siRNA to lead to the knockdown of hypoxia-inducible factor-1α (HIF-1α) protein and downstream genes to relieve tumor invasion. The Au DENPs/siRNA polyplexes were also used for dual-mode endogenous and exogenous sensitization of tumor RT in vivo. Results: Due to the dual-mode endogenous sensitization through HIF-1α gene silencing and the exogenous sensitization through the existing Au component, enhanced RT of cancer cells in vitro and a tumor model in vivo can be realized, which was confirmed by enhanced cytotoxic reactive oxygen species (ROS) generation in vitro and double-strand DNA damage verified from the γ-H2AX protein expression in tumor cells in vivo. By integrating the advantages of HIF-1α gene silencing-induced downregulation of downstream genes and the dual-mode sensitization-enhanced RT, simultaneous inhibition of primary tumors and metastasis can be readily realized. Conclusions: The developed zwitterionic Au DENPs may be used as a promising platform for dual-mode endogenously and exogenously sensitized RT of other tumor types.


Subject(s)
Dendrimers/chemistry , Gold/chemistry , Lung Neoplasms/radiotherapy , Metal Nanoparticles/administration & dosage , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/administration & dosage , Animals , Apoptosis , Cell Proliferation , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Metal Nanoparticles/chemistry , Mice , Neoplasm Invasiveness , Radiation-Sensitizing Agents/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Oncol Lett ; 20(3): 2356-2368, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32782553

ABSTRACT

Glycoprotein non-metastatic melanoma protein B (GPNMB), a transmembrane glycoprotein, has been reported to be involved in tumor progression, but its prognostic value for glioma and the mechanistic effects on glioma progression have not been clearly explored. The present study aimed to investigate the prognostic role of GPNMB in glioma and the potential mechanisms of how GPNMB mediates glioma progression. Differentially expressed genes between the four highest and four lowest GPNMB expression samples in the GSE53733 dataset were first determined. Gene ontology, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and Gene set enrichment analysis results demonstrated that the significantly enriched pathways in samples with high GPNMB expression compared with those with low GPNMB expression were associated with hypoxia, angiogenesis, migration and invasion. Pearson correlation analysis was conducted to investigate the correlations between GPNMB expression and the markers of hypoxia, angiogenesis, migration and invasion in GSE53733, which were further validated using another mRNA microarray dataset from the Chinese Glioma Genome Atlas (CGGA). In addition, using the CGGA dataset, high GPNMB expression was demonstrated to be significantly associated with advanced WHO grade and short survival time in patients with glioma. Of note, based on the immunohistochemical staining of the tissue microarrays, Kaplan-Meier analysis with the Renyi test and a Cox proportional hazards model were used to validate the unfavorable prognostic role of high GPNMB expression in glioma. In conclusion, high GPNMB expression may be associated with high tumor grade and unfavorable prognosis in glioma. GPNMB expression was demonstrated to correlate with the markers of hypoxia, angiogenesis, migration and invasion, which may be potential mechanisms through which GPNMB mediates glioma progression.

9.
Toxicol Appl Pharmacol ; 399: 115054, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32422326

ABSTRACT

Radiation-induced rectal injury is a major side-effect observed in patients with pelvic malignancies who receive radiotherapy. MicroRNA (miRNA), involved in many cellular biological processes, can be disturbed by ionizing radiation (IR). In this study, we have investigated the function of microRNA-122-5p (miR-122-5p) in radiation-induced rectal injury. MiR-122-5p levels in the serum of rectal cancer patients or in the rectal tissues of C57BL/6 mice before and after IR were detected by quantitative real-time PCR (qRT-PCR). We found that the miR-122-5p levels were significantly up-regulated in patients' serum or in mice rectal tissues after IR. Elevation of miR-122-5p levels sensitized human intestinal epithelial crypt (HIEC) cells to IR both in vitro and in vivo. MiR-122-5p mimic was transfected to HIEC cells and the downstream targets were predicted by bioinformatic analysis. Two putative target sites of miR-122-5p in the 3'UTR of the cell cycle and apoptosis regulator 1 (CCAR1) mRNA were found and verified by luciferase reporter assay. Overexpression of miR-122-5p or silencing CCAR1 combined with IR significantly inhibited cell survival, enhanced radiosensitivity, and increased cell apoptosis compared to that in the negative control group in vitro. In vivo injection of miR-122-5p antagomir after IR significantly alleviated radiation-induced rectal injury in mice. These results suggest that miR-122-5p aggravates radiation-induced rectal injury through targeting CCAR1.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Cell Cycle Proteins/genetics , MicroRNAs/genetics , Radiation Injuries/genetics , Radiation Tolerance/genetics , Rectum/radiation effects , 3' Untranslated Regions/genetics , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Line , Cell Proliferation/genetics , Cell Survival/genetics , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , RNA, Messenger/genetics , Up-Regulation/genetics
10.
J Cancer ; 11(8): 2022-2031, 2020.
Article in English | MEDLINE | ID: mdl-32127930

ABSTRACT

Macrophages play a critical role in the initiation and progression in various human solid tumors; however, their role and transformation in pancreatic ductal adenocarcinoma (PDAC) were still illusive. Here, immunohistochemistry was used to determine CD206 (specific marker of M2 macrophage) and PKM2 expression in PDAC tissues. Statistical analysis, such as Pearson χ2 test, Spearman's rank test, Kaplan-Meier and COX regression assay were used to evaluate their roles on PDAC prognosis. Data showed that both CD206 and PKM2 were elevated and responsible for a poor prognosis for PDAC. In addition, we showed that the two factors were positively correlated; co-overexpression of the two factors conferred the worst prognosis and functioned as an independent prognostic factor for the disease. Our data showed that M2 macrophage infiltration was correlated with PKM2 expression in PDAC cells. The two markers exerted synergistic effect on PDAC progression. Our results suggested dual-target inhibition M2 macrophage polarization and PKM2 expression of cancer cells might be novel approaches to treat PDAC.

11.
Radiat Res ; 193(2): 119-129, 2020 02.
Article in English | MEDLINE | ID: mdl-31841081

ABSTRACT

The goal of this work was to elucidate the mechanisms of bystander effects outside the localized irradiation field and their potential hematological toxicity. In this study, an in vitro multicellular co-culture system was used to investigate the intercellular commutation and related signaling pathways between either irradiated A549 cells or Beas-2B cells and bystander lymphoblast TK6 cells with or without macrophage U937 cells as an intermediator. Results showed that the proliferation ability of bystander TK6 cells was inhibited after co-culture with A549 cells irradiated with γ rays rather than carbon ions. When macrophages were contained in the co-culture system, the cell viability damage to the bystander TK6 cells were further enhanced. However, the proliferation inhibition of bystander TK6 cells after co-culture with irradiated Beas-2B cells was observed only when intermediator macrophages existed in the cell co-culture system. More serious cell injury was detected after carbon-ion irradiation compared with γ-ray irradiation. The p53-relevant apoptosis pathway was activated in both irradiated A549 and Beas-2B cells, each to a different extent. When the p53 pathway of irradiated cells was inhibited by PFT-α, PFTµ or p53 siRNA, the bystander damage to TK6 cells were clearly alleviated. In conclusion, the bystander lymphoblast damage was induced in different cells using different LET radiations. An amplified bystander response was modulated by the intermediator macrophage. The underlying molecular mechanisms of these bystander effects were dependent on the activation of p53 and its relevant apoptosis pathway in the irradiated cells. These results suggest that the bystander and macrophage-mediated bystander effects contribute to the common acute side effect of lymphocytopenia after local irradiation.


Subject(s)
Bystander Effect/radiation effects , Macrophages/cytology , Macrophages/radiation effects , Tumor Suppressor Protein p53/metabolism , A549 Cells , Cell Survival/radiation effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Intracellular Space/metabolism , Intracellular Space/radiation effects , Linear Energy Transfer , RNA, Small Interfering/genetics , Signal Transduction/radiation effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
12.
Radiat Res ; 193(1): 63-72, 2020 01.
Article in English | MEDLINE | ID: mdl-31714866

ABSTRACT

It is well known that mitochondria and the endoplasmic reticulum (ER) play important roles in radiation response, but their functions in radiation-induced bystander effect (RIBE) are largely unclear. In this study, we found that when a small portion of cells in a population of human lung fibroblast MRC-5 cells were precisely irradiated through either the nuclei or cytoplasm with counted microbeam protons, the yield of micronuclei (MN) and the levels of intracellular reactive oxygen species (ROS) in nonirradiated cells neighboring irradiated cells were significantly increased. Mito/ER-tracker staining demonstrated that the mitochondria were clearly activated after nuclear irradiation and ER mass approached a higher level after cytoplasmic irradiation. Moreover, the radiation-induced ROS was diminished by rotenone, an inhibitor of mitochondria activation, but it was not influenced by siRNA interference of BiP, an ER regulation protein. While for nuclear irradiation, rotenone-enhanced radiation-induced ER expression, and BiP siRNA eliminated radiation-induced activation of mitochondria, these phenomena were not observed for cytoplasmic irradiation. Bystander MN was reduced by rotenone but enhanced by BiP siRNA. When the cells were treated with both rotenone and BiP siRNA, the MN yield was reduced for nuclear irradiation but was enhanced for cytoplasmic irradiation. Our results suggest that the organelles of mitochondria and ER have different roles in RIBE with respect to nuclear and cytoplasmic irradiation, and the function of ER is a prerequisite for mitochondrial activation.


Subject(s)
Bystander Effect/radiation effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/radiation effects , Mitochondria/metabolism , Mitochondria/radiation effects , Protons/adverse effects , Cell Line , Fibroblasts/cytology , Fibroblasts/radiation effects , Humans , Reactive Oxygen Species/metabolism
13.
Medicine (Baltimore) ; 98(31): e16684, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31374051

ABSTRACT

The epidemiological and prognostic data focusing on clear cell renal cell carcinoma (ccRCC) are rarely presented. This study was aimed to define the frequency, incidence, and survival outcomes of ccRCC in the United States.The Surveillance, Epidemiology, and End Results (SEER) database was searched for patients with ccRCC from 1973 to 2014. Two patient cohorts were utilized: patient cohorts of SEER 18 registries and 9 registries. Overall survival was determined with Kaplan-Meier method and compared across groups with log-rank test.The incidence rate of ccRCC increased with advancing age, peaked in individuals aged 60 to 79 years, and declined in individuals aged ≥80 years. The incidence rate of ccRCC was significantly higher in males than females (1.94: 1, P < .0001), in Whites than Blacks or others (1:0.79:0.91, P < .0001). The incidence rate of ccRCC with right side as primary origin was slightly but significantly higher than that with left side as primary origin (1:0.96, P = .0006). The incidence rate of ccRCC in Grade II was higher than other grades. Generally, the incidence rates of ccRCC in most circumstances started to surge in the middle 1990s. Survival outcomes of ccRCC worsened with advancing age at diagnosis, tumor grade, and stage. A better prognosis was observed in females than males, in Whites than Blacks, and in individuals diagnosed in 2006 to 2014 than 1973 to 2005.To the best of our knowledge, the present study firstly presented long-term and updated epidemiological and prognostic data concerning ccRCC in the United States. Significant differences in incidence rates and survival outcomes stratified by different variables were identified.


Subject(s)
Carcinoma, Renal Cell/mortality , Kidney Neoplasms/mortality , Adult , Age Distribution , Aged , Aged, 80 and over , Female , Humans , Incidence , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Retrospective Studies , SEER Program , Sex Distribution , United States/epidemiology , Young Adult
14.
Theranostics ; 9(18): 5214-5226, 2019.
Article in English | MEDLINE | ID: mdl-31410211

ABSTRACT

Multifunctional nanomaterials that have integrated diagnostic and therapeutic functions and low toxicity, and can enhance treatment efficacy through combination therapy have drawn tremendous amounts of attention. Herein, a newly developed multifunctional theranostic agent is reported, which is PEGylated W-doped TiO2 (WTO) nanoparticles (NPs) synthesized via a facile organic route, and the results demonstrated strong absorbance of these WTO NPs in the second near-infrared (NIR-II) window due to successful doping with W. These PEGylated WTO NPs can absorb both NIR-II laser and ionizing radiation, rendering them well suited for dual-modal computed tomography/NIR-II photoacoustic imaging and synergistic NIR-II photothermal/radiotherapy of tumors. In addition, the long-term in vivo studies indicated that these PEGylated WTO NPs had no obvious toxicity on mice in vivo, and they can be cleared after a 30-day period. In summary, this multifunctional theranostic agent can absorb both NIR-II laser and ionizing radiation with negligible toxicity and rapid clearance, therefore it has great promise for applications in imaging and therapeutics in biomedicine.


Subject(s)
Hyperthermia, Induced , Nanoparticles/chemistry , Neoplasms/diagnostic imaging , Neoplasms/therapy , Photoacoustic Techniques , Titanium/chemistry , Tomography, X-Ray Computed , Tungsten/chemistry , Absorption, Radiation , Animals , Cell Line, Tumor , Diagnostic Imaging , Infrared Rays , Mice, Nude , Neoplasms/pathology , Temperature
15.
Cell Oncol (Dordr) ; 42(5): 679-690, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31325096

ABSTRACT

BACKGROUND: Apatinib is a tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor-2 (VEGFR2), and has shown encouraging therapeutic effects in various malignant tumors. As yet, however, the role of apatinib in ovarian cancer has remained unknown. Here, we sought to elucidate the role of apatinib in the in vitro and in vivo viability and proliferation of ovarian cancer cells, as well as in glucose metabolism in these cells. METHODS: The effects of apatinib on ovarian cancer cell viability and proliferation were assessed using Cell Counting Kit-8 (CCK-8) and colony formation assays, respectively. The expression of VEGFR2/AKT1/SOX5/GLUT4 pathway proteins was assessed using Western blotting, and glucose uptake and lactate production assays were used to detect glycolysis in ovarian cancer cells. SOX5 was exogenously over-expressed and silenced in ovarian cancer cells using expression vector and shRNA-based methods, respectively. RNA expression analyses were performed using RNA-seq and gene-chip-based methods. GLUT4 promoter activity was assessed using a dual-luciferase reporter assay. The expression of p-VEGFR2 (Tyr1175), p-AKT1 (Ser473), p-GSK3ß (Ser9), SOX5 and GLUT4 in xenograft tissues was assessed using immunohistochemistry (IHC). RESULTS: We found that apatinib inhibited the in vitro and in vivo viability and proliferation in Hey and OVCA433 ovarian cancer cells in a dose-dependent and time-dependent manner. We also found that apatinib effectively suppressed glucose uptake and lactate production by blocking the expression of GLUT4 in these cells. In addition, we found that SOX5 predominantly rescued the inhibitory effect of apatinib on GLUT4 expression by activating its promoter. Finally, we found that apatinib regulated the expression of SOX5 by suppressing the VEGFR2/AKT1/GSK3ß signaling pathway. CONCLUSIONS: From our results, we conclude that apatinib suppresses the in vitro and in vivo viability and proliferation of ovarian cancer cells, as well as glycolysis by inhibiting the VEGFR2/AKT1/GSK3ß/SOX5/GLUT4 signaling pathway. Apatinib may serve as a promising drug for the treatment of ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Glycolysis/drug effects , Ovarian Neoplasms/metabolism , Pyridines/pharmacology , SOXD Transcription Factors/metabolism , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Glucose Transporter Type 4/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/therapeutic use , SOXD Transcription Factors/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/metabolism
16.
Int J Radiat Biol ; 95(8): 1051-1057, 2019 08.
Article in English | MEDLINE | ID: mdl-30964366

ABSTRACT

Purpose: To investigate the relationship between autophagy and radiation damage of human hepatoma cells and to explore the role of reactive oxygen species (ROS). Materials and methods: HepG2 cells were exposed to X-rays, then the protein expressions of microtubule-associated protein 1 light chain 3 (LC3) and poly ADP-ribose polymerase-1 (PARP-1) were measured by Western blot assay, the formation of autophagosomes was detected by an autophagy detection kit, the intracellular ROS level was measured by flow cytometer, and DNA damage was evaluated by the incidence of micronuclei (MN). A CCK-8 kit was used to measure the proliferation ability of irradiated cells with or without N-acetyl-l-cysteine (NAC) treatment. In some experiments, the hepatoma cells were transferred with LC3 siRNA or PARP-1 siRNA before irradiation. Results: The protein expressions of LC3 and PARP-1 and the inductions of autophagosomes and intracellular ROS were increased in the irradiated HepG2 cells. Pretreatment of cells with NAC relieved the irradiation-induced inhibition of cell proliferation. When HepG2 cells were transfected with the LC3 siRNA, the over-expression of PARP-1 was diminished in the irradiated cells. Compared with the control group, the inhibitions of LC3 and PARP-1 increased ROS level in the irradiated HepG2 cells and hence sensitized radiation responses of both proliferation inhibition and MN induction. Conclusion: Autophagy upregulates the expression of PARP-1 and relieves radiation damage by reducing the generation of ROS.


Subject(s)
Autophagy/physiology , Carcinoma, Hepatocellular/radiotherapy , Liver Neoplasms/radiotherapy , Poly (ADP-Ribose) Polymerase-1/physiology , Radiation Injuries/prevention & control , Reactive Oxygen Species/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/radiation effects , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Microtubule-Associated Proteins/physiology , Oxidative Stress
17.
Life Sci ; 222: 228-234, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30858123

ABSTRACT

AIMS: In comparison with a low linear energy transfer (LET) radiation, a high-LET radiation induces more complex DNA damage. This study wonders whether radiation-induced bystander effect (RIBE) is dependent of LET. MATERIALS AND METHODS: Chinese hamster ovary CHO-9 cells and its subline EM-C11 cells (SSB repair deficient) and XR-C1 cells (DSB repair deficient) were irradiated by γ-rays, α-particles, or carbon ions with different LETs of 13, 30 and 70 keV/µm. Cell proliferation, cell death, DNA damage, cell cycle distribution and some protein expressions were measured with the cell counting kit-8 (CCK-8), colony formation, micronuclei (MN), flow cytometry and western blot, respectively. KEY FINDINGS: A series of cell responses were induced by these radiations in a LET-dependent manner, including proliferation inhibition, cell death, MN induction, G2/M phase arrest and the expression of γH2AX protein. These cell injuries were also depended on DNA repair capacity, and XR-C1 cells were the most sensitive to each radiation. Furthermore, when the cells were treated with the conditioned medium (CM) collected from irradiated CHO-9 cells, the MN induction and cell death response in the bystander cells of EM-C11 or XR-C1 increased along with LET of irradiation, and the bystander damage was easier to be induced in EM-C11 and XR-C1 cells than that in CHO-9 cells. SIGNIFICANCE: Both cellular DNA repair capacity and the LET value of radiation could deeply influence damage extents of not only the irradiated cells but also the bystander cells.


Subject(s)
Bystander Effect/radiation effects , DNA Damage/radiation effects , Linear Energy Transfer/radiation effects , Animals , Bystander Effect/physiology , CHO Cells , Cricetinae , Cricetulus , DNA Damage/physiology , Linear Energy Transfer/physiology
18.
Toxicol Appl Pharmacol ; 360: 131-140, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30292832

ABSTRACT

Radiation-induced rectal injury is closely related with radiotherapy efficiency. Here, we investigated the effect of focal adhesion kinase (FAK) in radiation-induced rectal injury. Peripheral blood samples of patients with rectal cancer were collected prior to radiotherapy. Differentially expressed genes and copy number variations (CNVs) were analyzed by microarray analysis. The CTCAE v3.0 toxicity grades were used to assess acute rectal injury. The radiosensitivity of human intestinal epithelial crypt (HIEC) cells were assayed by colony formation, mitochondrial membrane potential, flow cytometry and western blotting. The rectums of C57BL/6 mice were X-irradiated locally with a single dose of 15 Gy. The effect of FAK on radiation-induced injury was investigated by hematoxylin-eosin (H&E) staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL), immunohistochemistry (IHC) and quantitative real-time PCR (qRT-PCR). FAK mRNA level was inversely correlated with rectal injury severity in patient samples. A CNV amplification located on chromosome 8 was closely related with FAK. Further functional assays revealed increased levels of γH2AX expression and apoptosis-related proteins in FAK-silenced HIEC cells. The ratio of TUNEL, cl-caspase-3, cyto-c and bax/bcl-2 expression in the rectum mucosa treated with a FAK inhibitor increased significantly. These results demonstrated that FAK reduced radiation-induced rectal injury by decreasing apoptosis.


Subject(s)
Apoptosis/physiology , Focal Adhesion Kinase 1/metabolism , Radiation Injuries/metabolism , Rectum/metabolism , Animals , Caspase 3/metabolism , Cell Line , DNA Copy Number Variations/physiology , Female , Histones/metabolism , Humans , In Situ Nick-End Labeling/methods , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/metabolism , Radiation Tolerance/physiology , bcl-2-Associated X Protein/metabolism
19.
Int J Clin Exp Pathol ; 11(7): 3567-3574, 2018.
Article in English | MEDLINE | ID: mdl-31949735

ABSTRACT

Previous studies have showed that bile acids (BAs) play essential roles in the progression of various human cancers, and the G-protein coupled bile acid receptor-1 (Gpbar-1, or TGR5), a receptor of BAs, has been reported to connect BAs with cancers. However, little is known about the prognostic role of TGR5 in pancreatic cancer. In this study, we found that the expression of TGR5 was significantly higher in the cancerous tissues than the adjacent normal tissues by immunohistochemical staining (81.6% vs. 36.8%). Meanwhile, TGR5 was positively correlated with lymph node metastasis (P=0.021) and advanced stage (P=0.011). Finally, univariate analysis showed that patients with high TGR5 expression (P<0.001), lymph node metastasis (P=0.002) and advanced tumor stage (P=0.008) had decreased overall survival, and Cox proportional hazards regression analysis confirmed that TGR5 expression was an independent predictor of the overall survival of patients with pancreatic cancer (P=0.019). Our findings suggested that TGR5 might serve as an important predictor of poor survival in pancreatic cancer.

20.
Am J Clin Oncol ; 41(7): 619-625, 2018 07.
Article in English | MEDLINE | ID: mdl-28263232

ABSTRACT

OBJECTIVES: The Intergroup 0116 study has demonstrated a significant survival benefit for completely resected (R0) gastric cancer patients treated with a fluorouracil/leucovorin chemoradiotherapy regimen. However, this regimen is also toxic and less effective in terms of distant disease control. Therefore, a more efficacious and safer regimen is urgently needed. METHODS: Patients with R0 resected gastric carcinoma received up to two 21-day cycles of postoperative adjuvant preradiation and postradiation DCF chemotherapy (docetaxel 37.5 mg/m on days 1 and 8, cisplatin 25 mg/m on days 1 to 3, and a continuous infusion of fluorouracil 750 mg/m on days 1 to 5), respectively. Chemoradiotherapy between preradiation and postradiation chemotherapy was initiated on day 43 and consisted of intensity-modulated radiotherapy (45 Gy) plus concurrent docetaxel 20 mg/m weekly for 5 weeks. RESULTS: A total of 55 patients were evaluated and 76% (42) of patients completed the prescribed therapy. With a median follow-up of 61 months, the 3- and 5-year progression-free survival rates were 67% (95% confidence interval [CI], 54%-80%) and 59% (95% CI, 46%-72%), respectively; and the 3- and 5-year overall survival rates were 72% (95% CI, 60%-84%) and 61% (95% CI, 48%-74%), respectively. The most common grade 3 or greater toxicity, during the chemotherapy phase, was neutropenia (24%). Common grade 3/4 toxicities during concurrent chemoradiotherapy were nausea (32%), vomiting (26%), fatigue (15%), and anorexia (19%). CONCLUSIONS: These results demonstrate that this adjuvant regimen is active with an acceptable toxicity profile. A randomized phase 3 trial comparing the Intergroup 0116 chemoradiotherapy regimen with this regimen is underway.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemoradiotherapy, Adjuvant/mortality , Chemoradiotherapy/mortality , Stomach Neoplasms/therapy , Adult , Aged , Cisplatin/administration & dosage , Docetaxel/administration & dosage , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/pathology , Survival Rate , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...