Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 566
1.
PLoS One ; 16(11): e0260609, 2021.
Article En | MEDLINE | ID: mdl-34843603

Cell counting is a frequent task in medical research studies. However, it is often performed manually; thus, it is time-consuming and prone to human error. Even so, cell counting automation can be challenging to achieve, especially when dealing with crowded scenes and overlapping cells, assuming different shapes and sizes. In this paper, we introduce a deep learning-based cell detection and quantification methodology to automate the cell counting process in the zebrafish xenograft cancer model, an innovative technique for studying tumor biology and for personalizing medicine. First, we implemented a fine-tuned architecture based on the Faster R-CNN using the Inception ResNet V2 feature extractor. Second, we performed several adjustments to optimize the process, paying attention to constraints such as the presence of overlapped cells, the high number of objects to detect, the heterogeneity of the cells' size and shape, and the small size of the data set. This method resulted in a median error of approximately 1% of the total number of cell units. These results demonstrate the potential of our novel approach for quantifying cells in poorly labeled images. Compared to traditional Faster R-CNN, our method improved the average precision from 71% to 85% on the studied data set.


Cell Count/methods , Deep Learning , Image Processing, Computer-Assisted/methods , Neoplasms, Experimental/diagnosis , Animals , Heterografts , Humans , Neoplasm Transplantation , Neoplasms/diagnosis , Neoplasms/pathology , Neoplasms, Experimental/pathology , Zebrafish
2.
PLoS One ; 16(3): e0248531, 2021.
Article En | MEDLINE | ID: mdl-33705492

Mapping of sentinel lymph nodes (SLNs) can enable less invasive surgery. However, mapping is challenging for cancers of difficult-to-access visceral organs, such as the gallbladder, because the standard method using radioisotopes (RIs) requires preoperative tracer injection. Indocyanine green (ICG) and superparamagnetic iron oxide (SPIO) have also been used as alternative tracers. In this study, we modified a previously reported magnetic probe for laparoscopic use and evaluated the feasibility of detecting SLNs of the gallbladder using a laparoscopic dual tracer method by injecting ICG and SPIO into five swine and one cancer-bearing swine. The laparoscopic probe identified SPIO nanoparticles in the nodes of 4/5 swine in situ, the magnetic field counts were 2.5-15.9 µT, and fluorescence was detected in SLNs in all five swine. ICG showed a visual lymph flow map, and SPIO more accurately identified each SLN with a measurable magnetic field quite similar to the RI. We then developed an advanced gallbladder cancer model with lymph node metastasis using recombination activating gene 2-knockout swine. We identified an SLN in the laparoscopic investigation, and the magnetic field count was 3.5 µT. The SLN was histologically determined to be one of the two metastatic lymph nodes. In conclusion, detecting the SLNs of gallbladder cancer in situ using a dual tracer laparoscopic technique with ICG and SPIO was feasible in a swine model.


Gallbladder Neoplasms , Indocyanine Green , Laparoscopy , Magnetic Iron Oxide Nanoparticles , Neoplasms, Experimental , Sentinel Lymph Node Biopsy , Animals , Cell Line, Tumor , Gallbladder Neoplasms/diagnosis , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/surgery , Humans , Indocyanine Green/pharmacokinetics , Indocyanine Green/pharmacology , Lymphatic Metastasis , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/pathology , Neoplasms, Experimental/surgery , Sentinel Lymph Node/pathology , Sentinel Lymph Node/surgery , Swine
3.
Biol Chem ; 402(6): 739-748, 2021 05 26.
Article En | MEDLINE | ID: mdl-33711213

Signal transducer and activator of transcription 3 (STAT3) is a widely-reported oncogene in many human cancers, but its role in the peritoneal metastasis of gastric cancer (GC) has yet to be studied. The expression level of STAT3 in GC patient tissues was assessed. Stable shRNA knockdown (KD) of STAT3 was established in GC cell line AGS, followed by examination of its effect on AGC cell viability and proliferation, xenograft tumor growth, metastatic potential, mesothelial-to-mesenchymal transition (MMT)-related properties and peritoneal metastasis in a mouse model. The specific STAT3 inhibitor BP1-102 was also employed to verify findings from STAT3 KD experiments. Expression of activated STAT3 was upregulated in GC patient tumor tissues, and further elevated among patients diagnosed with peritoneal metastasis. STAT3 deactivation suppressed viability and proliferation of GC cells in vitro, as well as GC tumorigenesis in vivo. Furthermore, the metastatic properties and production of MMT-inducing factors of GC cells in vitro were also dependent on STAT3 activation. Importantly, STAT3 KD significantly compromised peritoneal metastasis of GC in vivo. STAT3 activation contributes to peritoneal metastasis of GC by promoting MMT, warranting further investigation to explore its potential for GC treatment, in particular among peritoneal metastasis patients.


STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Animals , Cell Adhesion , Epithelial-Mesenchymal Transition , Humans , Mice , Mice, Nude , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/metabolism , STAT3 Transcription Factor/genetics , Stomach Neoplasms/diagnosis , Tumor Cells, Cultured
4.
J Clin Invest ; 131(4)2021 02 15.
Article En | MEDLINE | ID: mdl-33332285

To clarify the function of cyclin A2 in colon homeostasis and colorectal cancer (CRC), we generated mice deficient for cyclin A2 in colonic epithelial cells (CECs). Colons of these mice displayed architectural changes in the mucosa and signs of inflammation, as well as increased proliferation of CECs associated with the appearance of low- and high-grade dysplasias. The main initial events triggering those alterations in cyclin A2-deficient CECs appeared to be abnormal mitoses and DNA damage. Cyclin A2 deletion in CECs promoted the development of dysplasia and adenocarcinomas in a murine colitis-associated cancer model. We next explored the status of cyclin A2 expression in clinical CRC samples at the mRNA and protein levels and found higher expression in tumors of patients with stage 1 or 2 CRC compared with those of patients with stage 3 or 4 CRC. A meta-analysis of 11 transcriptome data sets comprising 2239 primary CRC tumors revealed different expression levels of CCNA2 (the mRNA coding for cyclin A2) among the CRC tumor subtypes, with the highest expression detected in consensus molecular subtype 1 (CMS1) and the lowest in CMS4 tumors. Moreover, we found high expression of CCNA2 to be a new, independent prognosis factor for CRC tumors.


Colon/metabolism , Colorectal Neoplasms/metabolism , Cyclin A2/metabolism , Homeostasis , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Animals , Colon/pathology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclin A2/genetics , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Staging , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Prognosis
5.
Mol Oncol ; 15(2): 579-595, 2021 02.
Article En | MEDLINE | ID: mdl-33159833

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive cancer. Most patients present with advanced disease at diagnosis, which only permits palliative chemotherapeutic treatments. RNA dysregulation is a hallmark of most human cancers, including PDAC. To test the impact of RNA processing dysregulation on PDAC pathology, we performed a bioinformatics analysis to identify RNA-binding proteins (RBPs) associated with prognosis. Among the 12 RBPs associated with progression-free survival, we focused on MEX3A because it was recently shown to mark an intestinal stem cell population that is refractory to chemotherapeutic treatments, a typical feature of PDAC. Increased expression of MEX3A was correlated with higher disease stage in PDAC patients and with tumor development in a mouse model of PDAC. Depletion of MEX3A in PDAC cells enhanced sensitivity to chemotherapeutic treatment with gemcitabine, whereas its expression was increased in PDAC cells selected upon chronic exposure to the drug. RNA-sequencing analyses highlighted hundreds of genes whose expression is sensitive to MEX3A expression, with significant enrichment in cell cycle genes. MEX3A binds to its target mRNAs, like cyclin-dependent kinase 6 (CDK6), and promotes their stability. Accordingly, knockdown of MEX3A caused a significant reduction in PDAC cell proliferation and in progression to the S phase of the cell cycle. These findings uncover a novel role for MEX3A in the acquisition and maintenance of chemoresistance by PDAC cells, suggesting that it may represent a novel therapeutic target for PDAC.


Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/metabolism , Neoplasms, Experimental/metabolism , Pancreatic Neoplasms/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Animals , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Cell Line, Tumor , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Humans , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Phosphoproteins/genetics , Prognosis , RNA-Binding Proteins/genetics , Gemcitabine
6.
Int J Med Sci ; 17(16): 2505-2510, 2020.
Article En | MEDLINE | ID: mdl-33029093

Background and purpose: The discovery of chemical substances with carcinogenic properties has allowed the development of several experimental models of colorectal cancer (CRC). Classically, experimental models of CRC in mice have been evaluated through clinical or serial euthanasia. The present study aims to investigate the role of low endoscopy in the analysis of carcinogenesis induced by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG). Methods: Thirty C57BL6 mice were divided into two groups: a control group with fifteen animals that underwent rectal instillation of saline solution on day 0 and a carcinogen group with fifteen animals that underwent a 100 mg/kg MNNG rectal instillation on day 0. In both groups, low endoscopies were performed on weeks 4 and 8. We used a validated endoscopic scoring system to evaluate the severity of colitis and colorectal tumor. Euthanasia was carried out at week 12. Results: We observed higher inflammation scores (p <0.001) and a higher number of tumors (p <0.05) in the MNNG group than the control group, both at weeks 4 and 8. A worsening of inflammation scores from the first to the second endoscopy was also noticeable in the MNNG group. There were no bowel perforations related to the procedure, and there was one death in the control group. Conclusion: Low endoscopy in experimental animals allows safe macroscopic evaluation of colorectal carcinogenesis without the need for euthanasia.


Methylnitronitrosoguanidine/toxicity , Neoplasms, Experimental/chemically induced , Rectal Neoplasms/chemically induced , Administration, Rectal , Animals , Carcinogenesis/chemically induced , Carcinogenesis/pathology , Colonoscopy/methods , Female , Humans , Methylnitronitrosoguanidine/administration & dosage , Mice , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/pathology , Rectal Neoplasms/diagnosis , Rectal Neoplasms/pathology , Rectum/diagnostic imaging , Rectum/drug effects , Rectum/pathology
7.
ACS Appl Mater Interfaces ; 12(43): 48371-48379, 2020 Oct 28.
Article En | MEDLINE | ID: mdl-33078608

Ovarian cancer survival and the recurrence rate are drastically affected by the amount of tumor that can be surgically removed prior to chemotherapy. Surgeons are currently limited to visual inspection, making smaller tumors difficult to be removed surgically. Enhancing the surgeon's ability to selectively remove cancerous tissue would have a positive effect on a patient's prognosis. One approach to aid in surgical tumor removal involves using targeted fluorescent probes to selectively label cancerous tissue. To date, there has been a trade-off in balancing two requirements for the surgeon: the ability to see maximal tumors and the ability to identify these tumors by eye while performing the surgery. The ability to see maximal tumors has been prioritized and this has led to the use of fluorophores activated by near-infrared (NIR) light as NIR penetrates most deeply in this surgical setting, but the light emitted by traditional NIR fluorophores is invisible to the naked eye. This has necessitated the use of specialty detectors and monitors that the surgeon must consult while performing the surgery. In this study, we develop nanoparticles that selectively label ovarian tumors and are activated by NIR light but emit visible light. This potentially allows for maximal tumor observation and real-time detection by eye during surgery. We designed two generations of up-converting nanoparticles that emit green light when illuminated with NIR light. These particles specifically label ovarian tumors most likely via tumor-associated macrophages, which are prominent in the tumor microenvironment. Our results demonstrate that this approach is a viable means of visualizing tumors during surgery without the need for complicated, expensive, and bulky detection equipment. Continued improvement and experimentation could expand our approach into a much needed surgical technique to aid ovarian tumor removal.


Nanoparticles/chemistry , Ovarian Neoplasms/surgery , Animals , Female , Humans , Mice , Mice, Nude , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/surgery , Ovarian Neoplasms/diagnosis , Particle Size , Surface Properties
8.
ACS Appl Mater Interfaces ; 12(42): 47233-47244, 2020 Oct 21.
Article En | MEDLINE | ID: mdl-32970405

Complex experimental design is a common problem in the preparation of theranostic nanoparticles, resulting in poor reaction control, expensive production cost, and low experiment success rate. The present study aims to develop PEGylated bismuth (PEG-Bi) nanoparticles with a precisely controlled one-pot approach, which contains only methoxy[(poly(ethylene glycol)]trimethoxy-silane (PEG-silane) and bismuth oxide (Bi2O3). A targeted pyrolysis of PEG-silane was achieved to realize its roles as both the reduction and PEGylation agents. The unwanted methoxy groups of PEG-silane were selectively pyrolyzed to form reductive agents, while the useful PEG-chain was fully preserved to enhance the biocompatibility of Bi nanoparticles. Moreover, Bi2O3 not only acted as the raw material of the Bi source but also presented a self-promotion in the production of Bi nanoparticles via catalyzing the pyrolysis of PEG-silane. The reaction mechanism was systematically validated with different methods such as nuclear magnetic resonance spectroscopy. The PEG-Bi nanoparticles showed better compatibility and photothermal conversion than those prepared by the complex multiple step approaches in literature studies. In addition, the PEG-Bi nanoparticles possessed prominent performance in X-ray computed tomography imaging and photothermal cancer therapy in vivo. The present study highlights the art of precise reaction control in the synthesis of PEGylated nanoparticles for biomedical applications.


Bismuth/pharmacology , Nanoparticles/chemistry , Photothermal Therapy , Animals , Bismuth/administration & dosage , Bismuth/chemistry , Cell Line, Tumor , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Molecular Structure , Nanoparticles/administration & dosage , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Particle Size , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/chemistry , Pyrolysis/drug effects , RAW 264.7 Cells , Surface Properties , Tomography, X-Ray Computed
9.
Anal Chem ; 92(12): 8010-8014, 2020 06 16.
Article En | MEDLINE | ID: mdl-32441506

In vivo monitoring of reactive oxygen species (ROS) in tumors during treatment with anticancer therapy is important for understanding the mechanism of action and in the design of new anticancer drugs. In this work, a platinized nanoelectrode is placed into a single cell for detection of the ROS signal, and drug-induced ROS production is then recorded. The main advantages of this method are the short incubation time with the drug and its high sensitivity which allows the detection of low intracellular ROS concentrations. We have shown that our new method can measure the ROS response to chemotherapy in tumor-bearing mice in real-time. ROS levels were measured in vivo inside the tumor at different depths in response to doxorubicin. This work provides an effective new approach for the measurement of intracellular ROS by platinized nanoelectrodes.


Antineoplastic Agents/pharmacology , Biosensing Techniques , Doxorubicin/pharmacology , Electrochemical Techniques , Reactive Oxygen Species/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Mice , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , PC-3 Cells , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism
10.
Anal Chem ; 92(9): 6349-6357, 2020 05 05.
Article En | MEDLINE | ID: mdl-32275820

For a more comprehensive characterization of molecular heterogeneities of matter, multimodal mass spectrometry imaging must be developed to take advantage of the complementarity of information available through different ionization mechanisms. We report the design, implementation, and performance validation of a laser desorption imaging interface composed of add-on components that adapt a commercial Desorption Electrospray Ionization Mass Spectrometry (DESI-MS) imaging interface for dual imaging of Picosecond Infrared Laser Mass Spectrometry (PIRL-MS) with DESI-MS. The interface utilizes hardware elements and data analysis pipelines already established for DESI-MS imaging, and was further validated in cancer margin assessments using human medulloblastoma cancers. The PIRL-MS images were robust and reproducible across multiple experimental runs on independently prepared xenograft tumors, and could be segmented into cancer and healthy regions in concordance with pathology using a variety of supervised and unsupervised clustering methods. The spectral quality and complexity obtained with this interface were examined with infiltrating and noninfiltrating tumors, and were comparable to other mass spectrometry analysis interfaces. The average PIRL-MS spectra from spatially resolved images could be used for robust cancer m/z model building to classify medulloblastoma cancer from healthy tissue without any misclassifications, an observation that held true over close to 70 sampling data points. While the unsupervised spectral analysis methods suggested a slight suppression of signal in the phospholipid range compared to the hand-held configuration, these changes were insufficient to hamper utility in cancer margin assessment with spatially resolved data obtained with our interface. Dual PIRL-MS and DESI-MS imaging of consecutive sections, as suggested by multivariate loading plots, revealed highly complementary molecular information with m/z values identifiable with one desorption method sufficient to reveal cancer regions being absent in another, further emphasizing the need for effective hardware and software interfaces for dual mass spectrometry imaging.


Cerebellar Neoplasms/diagnosis , Medulloblastoma/diagnosis , Animals , Humans , Mice , Neoplasms, Experimental/diagnosis , Printing, Three-Dimensional , Spectrometry, Mass, Electrospray Ionization
11.
Nanoscale ; 12(3): 1886-1893, 2020 Jan 23.
Article En | MEDLINE | ID: mdl-31904049

The aggregation of superparamagnetic iron oxide (SPIO) nanoparticles (NPs) can greatly enhance magnetic resonance imaging (MRI) T2-weighted imaging and near-infrared (NIR) absorption in experiments. In this study, an Ac-Arg-Val-Arg-Arg-Cys(StBu)-Lys-CBT probe was designed and coupled with monodispersed carboxyl-decorated SPIO NPs to form SPIO@1NPs, which use it for intracellular self-aggregation. In vitro experiments showed that the self-aggregation of SPIO@1NPs was induced by a condensation reaction mediated by the enzyme furin in furin-overexpressing tumor cells. Moreover, the NPs in the aggregated state showed significantly higher MR r2 values and photothermal conversion efficiency than the NPs in the monodisperse state. Then, the in vivo SPIO@1NP self-aggregation in tumors can facilitate accurate MRI T2 imaging-guided photothermal therapy for effectively killing cancer cells. We believe that this basic technique, based on tumor-specific enzyme-instructed intracellular self-aggregation of NPs, could be useful for the rational synthesis of other inorganic NPs for use in the fields of tumor diagnosis and treatment.


Furin/metabolism , Hyperthermia, Induced , Magnetic Resonance Imaging , Magnetite Nanoparticles , Neoplasm Proteins/metabolism , Neoplasms, Experimental , Phototherapy , Animals , Cell Line, Tumor , Female , Humans , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/therapy
12.
Eur Rev Med Pharmacol Sci ; 24(1): 164-173, 2020 01.
Article En | MEDLINE | ID: mdl-31957829

OBJECTIVE: To explore the roles and underlying mechanism of LINC00963 in esophageal squamous cell carcinoma (ESCC) progression. PATIENTS AND METHODS: Quantitative Real Time-PCR (qRT-PCR) was used to detect LINC00963 expression in ESCC tissues. EdU, colony formation, and transwell invasion assays were used to detect the proliferation and metastasis ability of ESCC, respectively. The correlation between LINC00963 and miR-214-5p in ESCC was confirmed by a Luciferase reporter and RIP assays. RESULTS: LINC00963 expression was significantly increased in ESCC tissues and correlated with advanced TNM stage, metastasis, and poor prognosis. The knockdown of LINC00963 expression reduced ESCC cells proliferation, invasion in vitro, and reduced tumor growth in vivo. In mechanism, LINC00963 served as a sponge for miR-214-5p in ESCC progression. In addition, miR-214-5p could bind to RAB14 and regulate its expression. CONCLUSIONS: LINC00963 might promote ESCC cells proliferation and invasion via regulating the miR-214-5p/RAB14 axis and it might serve as a therapeutic target for ESCC treatment.


Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Proliferation , Cells, Cultured , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/diagnosis , Esophageal Squamous Cell Carcinoma/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , RNA, Long Noncoding/genetics , rab GTP-Binding Proteins/genetics
13.
Angew Chem Int Ed Engl ; 59(12): 4678-4683, 2020 03 16.
Article En | MEDLINE | ID: mdl-31840357

We report a new approach to monitor drug release from nanocarriers via a paclitaxel-methylene blue conjugate (PTX-MB) with redox activity. This construct is in a photoacoustically silent reduced state inside poly(lactic-co-glycolic acid) (PLGA) nanoparticles (PTX-MB@PLGA NPs). During release, PTX-MB is spontaneously oxidized to produce a concentration-dependent photoacoustic signal. An in vitro drug-release study showed an initial burst release (25 %) between 0-24 h and a sustained release between 24-120 h with a cumulative release of 40.6 % and a 670-fold increase in photoacoustic signal. An in vivo murine drug release showed a photoacoustic signal enhancement of up to 649 % after 10 hours. PTX-MB@PLGA NPs showed an IC50 of 78 µg mL-1 and 44.7±4.8 % decrease of tumor burden in an orthotopic model of colon cancer via luciferase-positive CT26 cells.


Antineoplastic Agents, Phytogenic/pharmacology , Colonic Neoplasms/drug therapy , Fluorescent Dyes/chemistry , Methylene Blue/chemistry , Nanoparticles/chemistry , Paclitaxel/pharmacology , Photoacoustic Techniques , Animals , Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/diagnosis , Drug Carriers/chemistry , Drug Liberation , Drug Screening Assays, Antitumor , Fluorescent Dyes/chemical synthesis , Methylene Blue/chemical synthesis , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Oxidation-Reduction , Paclitaxel/chemical synthesis , Paclitaxel/chemistry , Particle Size , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Surface Properties
14.
Eur Rev Med Pharmacol Sci ; 23(19): 8449-8455, 2019 Oct.
Article En | MEDLINE | ID: mdl-31646575

OBJECTIVE: The importance of circular RNAs in malignant tumors increases the attention of researchers. The role of circ_0067934 in glioma remains unclear. Our study aims to uncover how circ_0067934 functions in glioma development. PATIENTS AND METHODS: Real Time-quantitative Polymerase Chain Reaction (RT-qPCR) was utilized to determine the level of circ_0067934 in glioma tissues. Circ_0067934 was knocked down in glioma cells. Cell migrated and invaded ability was detected through functional assay in vitro and in vivo. Further mechanism assays were performed to explore the potential targets of circ_0067934. RESULTS: The circ_0067934 was highly expressed in glioma tissues compared with adjacent samples. The expression of circ_0067934 was upregulated in glioma cell lines. The cell migrated and invaded ability of glioma cells was inhibited after circ_0067934 was knocked down. Besides, CSF1 expression was decreased via knockdown of circ_0067934. Furthermore, tumor metastasis was inhibited after circ_0067934 was knocked down in nude mice. CONCLUSIONS: The circ_0067934 could suppress cell migration and invasion of glioma by upregulating CSF1.


Brain Neoplasms/metabolism , Glioma/metabolism , Macrophage Colony-Stimulating Factor/metabolism , RNA, Circular/metabolism , Animals , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Cell Movement , Cells, Cultured , Glioma/diagnosis , Glioma/genetics , Humans , Macrophage Colony-Stimulating Factor/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , RNA, Circular/genetics
15.
ACS Appl Mater Interfaces ; 11(43): 39624-39632, 2019 Oct 30.
Article En | MEDLINE | ID: mdl-31573175

To advance anti-tumor efficiency and lessen the adverse effect caused by nanodrug residues in the body, a smart nanoagent system is developed and successfully used in intracellular ATP imaging and in vivo chemo-photothermal synergetic therapy. The nanoagent system is facilely prepared using a DNA complex to modify gold nanoparticles (AuNPs). The DNA complex is formed by three oligonucleotides (ATP aptamer, rC-DNA, and rG-DNA). The CG-rich structure in a ternary DNA complex could be exploited for payload of chemotherapeutic medicine doxorubicin (DOX), thus making efficient DOX transport into the tumor site possible. In tumor cells, especially in acidic organelles (e.g., endosome and lysosome), DOX could be rapidly released via the dual stimuli of overexpressed ATP and pH. What is more, the specific recognition of a fluorescently labeled aptamer strand to ATP can achieve the intracellular ATP imaging. pH-controlled reversible folding and unfolding of intermolecular i-motif formed by C-rich strands can lead to intracellular in situ assembly of AuNP aggregates with high photothermal conversion efficiency and promote relatively facile renal clearance of AuNPs through the disassociation of the aggregates in extracellular environments. Experiments in vivo and vitro present feasibility for a synergetic chemo-photothermal therapy. Such an in situ reversible assembly strategy of a chemo-photothermal agent also presents a new paradigm for a smart and highly efficient disease treatment with reduced side effects.


Adenosine Triphosphate/metabolism , Doxorubicin , Gold , Hyperthermia, Induced , Metal Nanoparticles , Molecular Imaging , Neoplasms, Experimental , Phototherapy , Animals , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacokinetics , Delayed-Action Preparations/pharmacology , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , HeLa Cells , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , Xenograft Model Antitumor Assays
16.
Biomater Sci ; 7(11): 4769-4781, 2019 Nov 01.
Article En | MEDLINE | ID: mdl-31509113

808 nm near-infrared (NIR) light-induced biological theranostics is gradually becoming a popular method for cancer treatment. Meanwhile, mild synthetic methods to prepare medicines and gentle treatment conditions for cancer patients are becoming increasingly important to oncotherapy. Herein, tiny AgBiS2 nanodots were synthesized via a simple method, and for the first time, discovered to produce a photodynamic therapy (PDT) effect for cancer treatment under 808 nm laser irradiation, which was characterized by both chemical probe and intracellular reactive oxygen species (ROS) detection. Subsequently, because tumor cells have more mitochondria than normal cells to generate more energy to maintain rapid growth for population expansion, and triphenylphosphonium (TPP) can transport tiny nanoparticles into the mitochondria, the as-synthesized AgBiS2 nanodots were combined with TPP in a facile route. In our design, the AgBiS2 nanodots exhibit photothermal properties and TPP can enhance the photothermal properties of the AgBiS2 nanodots to a certain extent, which make the AgBiS2-TPP nanocomposite applicable in photothermal therapy (PTT). Furthermore, the AgBiS2-TPP nanocomposite showed a remarkable computed tomography (CT) imaging performance for tumor diagnosis. The facile synthetic strategy, satisfactory anticancer effect, CT imaging and mitochondrial targeting of the AgBiS2-TPP nanocomposite demonstrate its high potential in the anticancer field.


Antineoplastic Agents/pharmacology , Nanocomposites/chemistry , Organophosphorus Compounds/pharmacology , Photochemotherapy , Photosensitizing Agents/pharmacology , Silver Compounds/pharmacology , Sulfides/pharmacology , Tomography, X-Ray Computed , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Female , HeLa Cells , Humans , Infrared Rays , Mice , Mice, Inbred Strains , Mitochondria/drug effects , Molecular Structure , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Organophosphorus Compounds/chemistry , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/chemistry , Reactive Oxygen Species/analysis , Reactive Oxygen Species/metabolism , Silver Compounds/chemistry , Sulfides/chemistry
17.
Cell Commun Signal ; 17(1): 108, 2019 08 28.
Article En | MEDLINE | ID: mdl-31455353

BACKGROUND: Glioma is the most common and primary brain tumors in adults. Despite the available multimodal therapies, glioma patients appear to have a poor prognosis. The Hedgehog (Hh) signaling is involved in tumorigenesis and emerged as a promising target for brain tumors. Glabrescione B (GlaB) has been recently identified as the first direct inhibitor of Gli1, the downstream effector of the pathway. METHODS: We established the overexpression of Gli1 in murine glioma cells (GL261) and GlaB effect on cell viability. We used 1H-nuclear magnetic resonance (NMR) metabolomic approach to obtain informative metabolic snapshots of GL261 cells acquired at different time points during GlaB treatment. The activation of AMP activated protein Kinase (AMPK) induced by GlaB was established by western blot. After the orthotopic GL261 cells injection in the right striatum of C57BL6 mice and the intranasal (IN) GlaB/mPEG5kDa-Cholane treatment, the tumor growth was evaluated. The High Performance Liquid Chromatography (HPLC) combined with Mass Spectrometry (MS) was used to quantify GlaB in brain extracts of treated mice. RESULTS: We found that GlaB affected the growth of murine glioma cells both in vitro and in vivo animal model. Using an untargeted 1H-NMR metabolomic approach, we found that GlaB stimulated the glycolytic metabolism in glioma, increasing lactate production. The high glycolytic rate could in part support the cytotoxic effects of GlaB, since the simultaneous blockade of lactate efflux with α-cyano-4-hydroxycinnamic acid (ACCA) affected glioma cell growth. According to the metabolomic data, we found that GlaB increased the phosphorylation of AMPK, a cellular energy sensor involved in the anabolic-to-catabolic transition. CONCLUSIONS: Our results indicate that GlaB inhibits glioma cell growth and exacerbates Warburg effect, increasing lactate production. In addition, the simultaneous blockade of Gli1 and lactate efflux amplifies the anti-tumor effect in vivo, providing new potential therapeutic strategy for this brain tumor.


Chromones/pharmacology , Glioma/drug therapy , Glioma/metabolism , Metabolomics , Animals , Cell Proliferation/drug effects , Glioma/diagnosis , Glycolysis/drug effects , Humans , Male , Mice , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Proton Magnetic Resonance Spectroscopy , Signal Transduction/drug effects , Tumor Cells, Cultured
18.
Acta Biomater ; 96: 491-504, 2019 09 15.
Article En | MEDLINE | ID: mdl-31302299

The combination of multi-targeting magnetic nanoprobes and multi-targeting strategies has potential to facilitate magnetic resonance imaging (MRI) and magnetic induction hyperthermia of the tumor. Although the thermo-agents based on magnetic iron oxide nanoparticles (MION) have been successfully used in the form of intratumoral injection in clinical cure of glioblastoma, the tumor-targeted thermotherapy by intravenous administration remains challenging. Herein, we constructed a c(RGDyK)- and d-glucosamine-grafted bispecific molecular nanoprobe (Fe3O4@RGD@GLU) with a magnetic iron oxide core of size 22.17 nm and a biocompatible shell of DSPE-PEG2000, which can specially target the tumor vessel and cancer cells. The selection of c(RGDyK) could make the nanoprobe enter the neovascularization endotheliocyte through αvß3-mediated endocytosis, which drastically reduced the dependence on the enhanced permeability and retention (EPR) effect in tumor. This dual-ligand nanoprobe exhibited strong magnetic properties and favorable biocompatibility. In vitro studies confirmed the anti-phagocytosis ability against macrophages and the specific targeting capability of Fe3O4@RGD@GLU. Then, the imaging effect and anti-tumor efficacy were compared using different targeting strategies with untargeted nanoprobes, dual-targeted nanoprobes, and magnetic targeting combined with dual-targeted nanoprobes. Moreover, the combination strategy of magnetic targeting and active targeting promoted the penetration depth of nanoprobes in addition to the increased accumulation in tumor tissue. Thus, the dual-targeted magnetic nanoprobe together with the combined targeting strategy could be a promising method in tumor imaging and hyperthermia through in vivo delivery of theranostic agents. STATEMENT OF SIGNIFICANCE: Magnetic induction hyperthermia based on iron oxide nanoparticles has been used in clinic for adjuvant treatment of recurrent glioblastoma. Nonetheless, this application is limited to intratumoral injection, and tumor-targeted hyperthermia by intravenous injection remains challenging. In this study, we developed a multi-targeted strategy by combining magnetic targeting with active targeting of dual-ligand magnetic nanoprobes. This combination mode acquired optimum contrast imaging effect through MRI and tumor-suppressive effect through hyperthermia under an alternating current magnetic field. The design of the nanoprobe was suitable for targeting most tumor lesions, which enabled it to be an effective theranostic agent with extensive uses. This study showed significant enhancement of the penetration depth and accumulation of nanoprobes in the tumor tissue for efficient imaging and hyperthermia.


Contrast Media , Hyperthermia, Induced , Magnetic Resonance Imaging , Magnetite Nanoparticles , Neoplasms, Experimental , Animals , Cell Line, Tumor , Contrast Media/chemistry , Contrast Media/pharmacology , Female , Human Umbilical Vein Endothelial Cells , Humans , Magnetite Nanoparticles/chemistry , Magnetite Nanoparticles/therapeutic use , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/therapy , RAW 264.7 Cells
19.
Eur Rev Med Pharmacol Sci ; 23(13): 5802-5814, 2019 Jul.
Article En | MEDLINE | ID: mdl-31298332

OBJECTIVE: The aberrant expression of long noncoding RNAs (lncRNAs) is involved in the molecular regulation of non-small cell lung cancer (NSCLC). This study aims to investigate the biological interaction of lnc-FAM201A and its downstream factors and their impacts on the radiotherapy response of NSCLC. PATIENTS AND METHODS: Quantitative Polymerase Chain Reaction (qPCR) was used to determine the expression of FAM201A in NSCLC tissues. The Chi-square tests explored the association between FAM201A level and the poor clinicopathological characteristics (including radioresistance) of NSCLC. Univariate and multivariate Cox proportional hazards regression analyses were used to evaluate various prognostic factors for overall survival (OS). The effect of FAM201A on OS was tested by the log-rank test. A549/SK-MES-1 cell lines transfected with short hairpin RNA (shRNA) were used to verify the promoting effects of FAM201A on radiotherapy resistance in vitro and in vivo. Cell apoptosis (analyzed by flow cytometry), cell proliferation (determined by Cell Counting Kit-8), and mice xenograft models were performed to confirm the results. The downstream targets of FAM201A were predicted by bioinformatics tools. Additionally, the Dual-luciferase reporter assay, qPCR, and Western blotting were performed to confirm their interaction. RESULTS: FAM201A was significantly upregulated in tissues obtained from NSCLC patients resistant to radiotherapy. Increased FAM201A expression was strongly associated with radioresistance and inferior survival in NSCLC, as demonstrated by clinical data. The silence of FAM201A could inhibit cell proliferation and further cell apoptosis of NSCLC cells under X-ray irradiation both in vitro and in vivo. Moreover, by competitively targeting miR-370, FAM201A elevated the epidermal growth factor receptor (EGFR) and the hypoxia-inducible factor 1alpha (HIF-1α) levels. After FAM201A knockdown, EGFR and HIF-1α were repressed with enhanced radiosensitivity. CONCLUSIONS: The interference of FAM201A impairs its suppression of miR-370, resulting in the upregulation of EGFR and HIF-1α and enhancement of radiosensitivity in NSCLC patients. Collectively, our results indicated that this regulatory axis might serve as a potential therapeutic target to increase the sensitivity of radiotherapy in NSCLC patients.


Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , A549 Cells , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Proliferation , Disease Models, Animal , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/radiotherapy , Male , Mice , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/radiotherapy , RNA, Long Noncoding/genetics , Radiation Tolerance/genetics , Regression Analysis , Tumor Cells, Cultured
20.
Eur Rev Med Pharmacol Sci ; 23(13): 5730-5740, 2019 Jul.
Article En | MEDLINE | ID: mdl-31298352

OBJECTIVE: Colorectal carcinoma (CRC) is one of the most common factors for tumor-associated mortalities globally. In recent years, microRNAs (miRNAs) have been identified as novel therapeutic biomarkers for cancer treatment. The purpose of the current study was to unravel the clinical significance and underlying molecular mechanisms of miR-760 in CRC progression. PATIENTS AND METHODS: Fifty-four pairs of CRC tissue samples and adjacent para-carcinoma tissue samples were collected from CRC patients who underwent surgical resection. We measured miR-760 expressions in CRC using quantitative Real-time polymerase chain reaction (qRT-PCR) analysis. MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assays and transwell assays were performed to determine the functions of miR-760 in CRC cell proliferation, invasion and migration. Dual-luciferase reporter assays and Western blots were used to investigate the underlying molecular mechanisms. Moreover, the association between miR-760 expressions and clinicopathological features was analyzed. RESULTS: In this study, the results showed that the down-regulated miR-760 expressions were related to the poor prognosis and malignant clinicopathologic features of CRC patients. Furthermore, functional assays revealed that miR-760 restoration obviously suppressed CRC cell proliferation, migration and invasion through modulating phosphatidylinositol 3-kinase/ protein kinase B (PI3K/AKT) pathway and epithelial-mesenchymal transition (EMT). FOXA1 was also considered as a functional target of miR-760 in CRC cells. Furthermore, miR-760 up-regulation also significantly repressed tumorigenesis in vivo. CONCLUSIONS: These results suggested that miR-760 exerted cancer-suppressive functions in CRC, providing a therapeutic strategy for CRC treatment.


Biomarkers, Tumor/metabolism , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Hepatocyte Nuclear Factor 3-alpha/antagonists & inhibitors , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Cells, Cultured , Colorectal Neoplasms/diagnosis , Female , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/metabolism , Signal Transduction
...