Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
1.
Molecules ; 29(11)2024 May 23.
Article in English | MEDLINE | ID: mdl-38893336

ABSTRACT

Glioblastoma, the most aggressive and challenging brain tumor, is a key focus in neuro-oncology due to its rapid growth and poor prognosis. The C6 glioma cell line is often used as a glioblastoma model due to its close simulation of human glioma characteristics, including rapid expansion and invasiveness. Alongside, herbal medicine, particularly Artemisia spp., is gaining attention for its anticancer potential, offering mechanisms like apoptosis induction, cell cycle arrest, and the inhibition of angiogenesis. In this study, we optimized extraction conditions of polyphenols from Artemisia annua L. and Artemisia vulgaris L. herbs and investigated their anticancer effects in silico and in vitro. Molecular docking of the main phenolic compounds of A. annua and A. vulgaris and potential target proteins, including programmed cell death (apoptosis) pathway proteins proapoptotic Bax (PDB ID 6EB6), anti-apoptotic Bcl-2 (PDB ID G5M), and the necroptosis pathway protein (PDB ID 7MON), mixed lineage kinase domain-like protein (MLKL), in complex with receptor-interacting serine/threonine-protein kinase 3 (RIPK3), revealed the high probability of their interactions, highlighting the possible influence of chlorogenic acid in modulating necroptosis processes. The cell viability of rat C6 glioma cell line was assessed using a nuclear fluorescent double-staining assay with Hoechst 33342 and propidium iodide. The extracts from A. annua and A. vulgaris have demonstrated anticancer activity in the glioblastoma model, with the synergistic effects of their combined compounds surpassing the efficacy of any single compound. Our results suggest the potential of these extracts as a basis for developing more effective glioblastoma treatments, emphasizing the importance of further research into their mechanisms of action and therapeutic applications.


Subject(s)
Apoptosis , Artemisia annua , Glioblastoma , Molecular Docking Simulation , Plant Extracts , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Plant Extracts/pharmacology , Plant Extracts/chemistry , Artemisia annua/chemistry , Cell Line, Tumor , Humans , Apoptosis/drug effects , Artemisia/chemistry , Rats , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Computer Simulation , Cell Survival/drug effects , Animals , Cell Proliferation/drug effects
2.
Metabolites ; 13(7)2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37512530

ABSTRACT

Acidification of cancerous tissue induced pharmacologically may slow tumor growth and can be detected using magnetic resonance imaging. Numerous studies have shown that pharmacologically inhibiting specific transporters, such as the Na+/H+ exchanger 1 (NHE1), can alter glycolitic metabolism and affect tumor acidosis. The sodium proton exchanger inhibitor Cariporide can acidify U87MG gliomas in mice. This study aimed to determine whether Cariporide could acidify C6 glioma tumors in rats with an intact immune system. C6 glioma cells were implanted in the right brain hemisphere of ten rats. Chemical exchange saturation transfer (CEST) MRI (9.4T) was acquired on days 7-8 and 14-15 after implantation to measure in vivo tissue intracellular pH (pHi) within the tumors and on the contralateral side. pHi was basic relative to contralateral tissue at both time points assessed using the amine and amide concentration-independent detection (AACID) value. On day 14-15, measurements were made before and up to 160 min after Cariporide injection (N = 6). Twenty minutes after drug injection, the average AACID value in the tumor significantly increased by ∼6.4% compared to pre-injection, corresponding to 0.31 ± 0.20 lower pHi, while in contralateral tissue, AACID value increased significantly by ∼4.3% compared to pre-injection, corresponding to 0.22 ± 0.19 lower pHi. Control rats without tumors showed no changes following injection of Cariporide dissolved in 10% or 1% DMSO and diluted in PBS. This study demonstrates the sensitivity of CEST-based pH-weighted imaging for monitoring the response of tumors to pharmacologically induced acidification.

3.
World Neurosurg ; 168: e595-e606, 2022 12.
Article in English | MEDLINE | ID: mdl-36280046

ABSTRACT

BACKGROUND: Surgical resection is a key method for glioma treatment. This inherently invasive procedure alters the tumor microenvironment of glioma cells that cannot be removed by surgery. However, few studies have focused on the impact of this microenvironment change on the growth of glioma cells. METHODS: The authors preconstructed a surgical brain injury model, and then C6 glioma cells were transplanted. HE staining was used to observe the general morphology of tumor cells, and immunohistochemistry of MMP-2, MMP-9, GFAP, and CD31 was used to evaluate the invasiveness of glioma cells and activation of astrocytes and calculate microvessel density. In vitro, primary rat astrocytes were exposed to different temperature gradients. The supernatant was made into conditioned medium for culturing C6 glioma cells. The scratch test and transwell test were used to evaluate the migration and invasion of tumor cells. RESULTS: GFAP expression was stronger in surgical brain injury rats, C6 cells implanted in these rats showed stronger expression of MMP-2 and MMP-9, and CD31 was expressed in more microvessels. Astrocytes exposed to high temperatures of 40°C and 43°C expressed stronger GFAP, and C6 cells cultured in their supernatants had stronger scratch healing ability and the ability to cross transwell chambers. CONCLUSIONS: The microenvironment changes caused by surgical brain injury will enhance the migration and invasion of glioma cells and increase the microvessel density in the tumor. This effect may be related to the activation of astrocytes caused by the thermal injury of bipolar coagulation during surgery.


Subject(s)
Brain Injuries , Brain Neoplasms , Glioma , Rats , Animals , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Brain Neoplasms/pathology , Astrocytes/metabolism , Glioma/pathology , Brain Injuries/metabolism , Cell Line, Tumor , Cell Movement , Tumor Microenvironment
4.
J Microencapsul ; 39(2): 136-144, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35313794

ABSTRACT

AIM: Docetaxel (DTX) loaded bio-compatible PLGA-PEG encapsulated zinc ferrite nanoparticles (ZFNP) formulation was developed and evaluated against C6 glioma cells. METHODS: The ZFNP were characterised using XRD, FE-SEM, TEM, etc. A series of drug formulations were fabricated by conjugating hydrothermally synthesised ZFNP with DTX in a PLGA-PEG matrix and optimised for drug loading. FTIR and DLS analysis of the formulation along with in vitro drug release, cytotoxicity, cellular uptake, and haemolytic effect were evaluated. RESULTS: Spherical, monodisperse, crystalline ZFNP with an average size of ∼28 nm were formed. The optimised formulation showed a hydrodynamic diameter of ∼147 nm, a surface charge of -34.8 mV, a drug loading of 6.9% (w/w) with prolonged drug release properties, and higher toxicity in C6 glioma cells compared to free DTX along with good internalisation and negligible haemolysis. CONCLUSION: The results indicate ZFNP could be effectively used as nanodrug carrier for delivery of docetaxel to glioma cells.


Subject(s)
Antineoplastic Agents , Glioma , Nanoparticles , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Survival , Docetaxel/pharmacology , Drug Carriers/chemistry , Excipients , Ferric Compounds , Glioma/drug therapy , Humans , In Vitro Techniques , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Particle Size , Taxoids/pharmacology , Zinc/pharmacology
5.
Stem Cell Rev Rep ; 18(4): 1495-1509, 2022 04.
Article in English | MEDLINE | ID: mdl-34403074

ABSTRACT

Many studies have shown that mesenchymal stromal cells (MSCs) and their secreted factors may modulate the biology of tumor cells. However, how these interactions happen in vivo remains unclear. In the present study, we investigated the effects of rat adipose-derived stromal cells (ADSCs) and their conditioned medium (ADSC-CM) in glioma tumor growth and malignancy in vivo. Our results showed that when we co-injected C6 cells plus ADSCs into the rat brains, the tumors generated were larger and the animals exhibited shorter survival, when compared with tumors of the animals that received only C6 cells or C6 cells pre-treated with ADSC-CM. We further showed that the animals that received C6 plus ADSC did not present enhanced expression of CD73 (a gene highly expressed in ADSCs), indicating that the tumor volume observed in these animals was not a mere consequence of the higher density of cells administered in this group. Finally, we showed that the animals that received C6 + ADSC presented tumors with larger necrosis areas and greater infiltration of immune cells. These results indicate that the immunoregulatory properties of ADSCs and its contribution to tumor stroma can support tumor growth leading to larger zones of necrosis, recruitment of immune cells, thus facilitating tumor progression. Our data provide new insights into the way by which ADSCs and tumor cells interact and highlight the importance of understanding the fate and roles of MSCs in tumor sites in vivo, as well as their intricate crosstalk with cancer cells.


Subject(s)
Glioblastoma , Adipose Tissue/metabolism , Animals , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Glioblastoma/genetics , Glioblastoma/therapy , Necrosis , Rats , Stromal Cells/metabolism
6.
Anticancer Agents Med Chem ; 22(9): 1780-1792, 2022.
Article in English | MEDLINE | ID: mdl-34238172

ABSTRACT

BACKGROUND: Benzimidazole derivatives bearing pyridyl/pyrimidinyl piperazine moiety has attracted attention in medicinal chemistry and modern drug discovery since it exhibited a variety of biological activities, including anticancer activity. OBJECTIVE: In this study, we designed and synthesized novel 1-[2-oxo-2-(4-substituted phenyl)ethyl]benzimidazol-2- yl)methyl 4-(2-pyridyl/pyrimidin-2-yl)piperazine-1-carbodithioate derivatives (2a-m). We also investigated their anticancer activities against A549 lung adenocarcinoma and C6 rat glioma cell lines. We further studied the selectivity of the compounds against the NIH/3T3 mouse embryonic fibroblast cell line. Cholinesterase inhibition effects of these compounds were also investigated to measure the relationship between anticancer activity and cholinesterases. METHODS: The cytotoxic activities of these acquired thirteen final compounds were screened using MTT assay on A549, C6, and NIH/3T3 cell lines. Cell proliferation ELISA, BRDU (colorimetric) assay was used to measure the proliferation in replicative cells in which DNA synthesis occurs. Flow cytometric analysis was used to measure apoptotic cell percentages, caspase 3 activity, and mitochondrial membrane depolarised cell percentages. RESULTS: Compounds 2e, 2f, and 2k were shown to be the most active antitumor agents with selective cytotoxicities (the results for A549 were 76.58±6.43, 55.13±5.75, and 32.94±3.02 µM, respectively; and for C6 they were 86.48±3.60, 97.12±30.21, and 59.29±3.95 µM, respectively), high DNA synthesis inhibition rates and high apoptotic cell percentages on both cell lines. CONCLUSION: The results showed that compounds 2e, 2f, and 2k have potential anticancer activity against A549 and C6 cell lines.


Subject(s)
Antineoplastic Agents , Fibroblasts , Animals , Antineoplastic Agents/chemistry , Apoptosis , Benzimidazoles/pharmacology , Cell Line, Tumor , Cell Proliferation , DNA , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fibroblasts/metabolism , Humans , Mice , NIH 3T3 Cells , Piperazine/pharmacology , Rats , Structure-Activity Relationship
7.
Acta Chim Slov ; 68(4): 970-982, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34918770

ABSTRACT

Effective treatment of glioma still stands as a challenge in medical science. The work aims for the fabrication and evaluation of lipid based nanostructures for improved delivery of lomustine to brain tumor cells. Experimental formulations (LNLs) were developed by modified lipid layer hydration technique and evaluated for different in vitro characteristics like particle size analysis, surface charge, surface morphology, internal structure, in vitro drug loading, drug release profile etc. Anticancer potential of selected LNLs was tested in vitro on C6 glioma cell line. Electron microscopic study depicted a size of less than 50 nm for the selected LNLs along 8.8% drug loading with a sustained drug release tendency over 48 h study period. Confocal microscopy revealed extensive internalization of the selected LNL in C6 cells. LNLs were found more cytotoxic than free drug and blank nanocarriers as depicted from MTT assay. The selected LNL showed improved pharmacokinetic profile both in blood and brain in the experimental mice models along with negligible hemolysis in mice blood cells. Further studies are warranted for the future translation of LNLs at clinics.


Subject(s)
Antineoplastic Agents/chemistry , Lipids/chemistry , Lomustine/chemistry , Nanostructures/chemistry , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Drug Liberation , Female , Glioma/metabolism , Glioma/pathology , Half-Life , Hemolysis/drug effects , Kinetics , Male , Mice , Nanostructures/toxicity
8.
Biomedicines ; 9(11)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34829821

ABSTRACT

Despite multimodal approaches for the treatment of multiforme glioblastoma (GBM) advances in outcome have been very modest indicating the necessity of novel diagnostic and therapeutic strategies. Currently, mesenchymal stem cells (MSCs) represent a promising platform for cell-based cancer therapies because of their tumor-tropism, low immunogenicity, easy accessibility, isolation procedure, and culturing. In the present study, we assessed the tumor-tropism and biodistribution of the superparamagnetic iron oxide nanoparticle (SPION)-labeled MSCs in the orthotopic model of C6 glioblastoma in Wistar rats. As shown in in vitro studies employing confocal microscopy, high-content quantitative image cytometer, and xCelligence system MSCs exhibit a high migratory capacity towards C6 glioblastoma cells. Intravenous administration of SPION-labeled MSCs in vivo resulted in intratumoral accumulation of the tagged cells in the tumor tissues that in turn significantly enhanced the contrast of the tumor when high-field magnetic resonance imaging was performed. Subsequent biodistribution studies employing highly sensitive nonlinear magnetic response measurements (NLR-M2) supported by histological analysis confirm the retention of MSCs in the glioblastoma. In conclusion, MSCs due to their tumor-tropism could be employed as a drug-delivery platform for future theranostic approaches.

9.
J Biochem Mol Toxicol ; 35(9): e22857, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34338399

ABSTRACT

Gliomas are a type of brain cancer that occurs in the supporting glial cells of the brain. It is highly malignant and accounts for 80% of brain tumors with high mortality and morbidity. Phytomedicines are potent alternatives for allopathic drugs which cause side effects. They have been used from ancient times by traditional Chinese, Ayurveda, and Siddha medicine. Arubtin is a glycoside phytochemical extracted from plants and belongs to the family of Ericaceae. Arbutin possesses various pharmacological properties such as anti-inflammatory, antioxidant, antitumor, and so on. Hence in the present study, we analyzed the anticancer potency of arbutin against rat C6 glioma cells. Rat C6 glioma cells were procured from American Type Culture Collection and the cells were cultured in Roswell Park Memorial Institute-1640 medium. To assess the cytotoxicity effect of the arbutin against C6 glioma cells, an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide test was performed with different doses from 10 to 60 µM. Arbutin effectively induced apoptosis in the cells and the IC50 dose was obtained at 30 µM. For further studies, we selected the 30 µM IC50 dose and a higher dose of 40 µM. Reactive oxygen species (ROS) generated were analyzed with DCFDA/H2DCFDA stain and the destruction of mitochondrial membrane permeability which is the initiator of apoptosis was analyzed with a cationic stain Rhodamine 123. Dual staining with acridine orange and ethidium bromide was performed to assess the viable and dead cells. Cell adhesion properties of glioma cells were analyzed with Matrigel assay. The apoptotic, inflammatory, and phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) signaling molecules were analyzed with quantitative polymerase chain reaction (qPCR) analysis to confirm the anticancer effect of arbutin. Arbutin generated excessive ROS and disrupted the mitochondrial membrane, which induced apoptosis in cells, it also inhibited the cell adhesion property of C6 glioma cells. qPCR analysis clearly indicates arbutin increases the apoptotic genes and decreased the inflammatory and PI3K/mTOR signaling molecules. Overall, our results authentically confirm that arbutin can be a potent alternative for treating glioma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Arbutin/pharmacology , Glioma , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Glioma/drug therapy , Glioma/metabolism , Glioma/pathology , Rats
10.
Diagnostics (Basel) ; 11(7)2021 Jul 19.
Article in English | MEDLINE | ID: mdl-34359378

ABSTRACT

Tumor blood flow (TBF) is related to drug delivery and hypoxia, both of which can impact the efficacy of anti-cancer therapies. Although integrin αvß3 expression is related to tumor angiogenesis, it remains unclear whether the degree of angiogenesis affects TBF. This study aimed to evaluate the expression of integrin αvß3 in mouse tumor models using [68Ga]Ga-DOTA-c(RGDfK) peptide positron emission tomography (PET) and immunohistochemical staining. PET studies were conducted using mouse C6 glioma models and MIA PaCa-2 (n = 6 each). The [68Ga]Ga-DOTA-c(RGDfK) peptide was injected via the tail vein (2.17 ± 0.28 MBq), and 10 min static PET scans were performed. Immunohistochemical analysis was conducted using an integrin αVß3 antibody. [68Ga]Ga-DOTA-c(RGDfK) peptide PET revealed higher uptake of the radiotracer in C6 gliomas than in MIA PaCa-2 tumors. The mean standardized uptake value was significantly higher in C6 gliomas (0.35 ± 0.058) than in MIA PaCa-2 tumors (0.17 ± 0.045). Histological analysis revealed intense integrin αVß3 expression in the C6 gliomas, whereas the MIA PaCa-2 tumors had low expression levels. This study showed that the expression of integrin αvß3 can be differentiated by the [68Ga]Ga-DOTA-c(RGDfK) peptide, suggesting the potential applicability of this peptide in the evaluation of the relationship between angiogenesis and TBF.

11.
Heliyon ; 7(2): e06352, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33718649

ABSTRACT

Glioblastoma is a severe cancer with extremely poor survival. Its treatment typically involves a combination of surgery, chemotherapy, and radiation therapy. However, glioma stem-like cells (GSCs)-a subpopulation of tumor-propagating glioblastoma cells-cause post-treatment recurrence and are a major factor in the poor prognosis of the disease. GSCs have higher proliferation than non-GSCs and are more resistant to invasive chemotherapy and radiotherapy. In this study, we subjected GSCs to nutrient starvation (deprived of glucose, glutamine, and calcium) to determine whether cell death can be triggered as a potential strategy to improve treatment outcomes. Flow cytometry revealed that 35.1%, 96.1%, and 99.9% of starved GSCs underwent apoptosis on days 1, 3, and 5, respectively, along with nearly 100% autophagy on all three days. Western blots detected cleaved caspase-3 (an apoptosis marker) and phospho-beclin 1, LC 3B-I, LC 3B-II (autophagy markers) in C6 GSCs after nutrient starvation for 1, 3, 4, and 5 days. Transmission electron microscopic observation of GSC ultrastructure after starvation treatment revealed that compared with control GSCs, starved cells had more pyknotic nuclei, membrane bleb, swollen endoplasmic reticulum, degenerative mitochondria, lipid droplets, and microvilli loss. Thus, nutrient starvation stresses cells by increasing free radicals. Cell stress opens more channels between mitochondria and endoplasmic reticulum. This study demonstrated that nutrient starvation decreases proliferation by approximately 81%, while increasing apoptosis (99.9%) and autophagy (94.6%) in C6 GSCs by the fifth day. Nutrient starvation of GSCs may, therefore, be an effective therapeutic strategy that can trigger apoptotic and autophagic metabolic reprogramming in cancer cells.

12.
Mol Med Rep ; 23(4)2021 04.
Article in English | MEDLINE | ID: mdl-33576438

ABSTRACT

Malignant tumors of the central nervous system (CNS) are among the types of cancer with the poorest prognosis and glioma is the commonest primary CNS tumor. A mitochondrial DNA (mtDNA)­depleted cell line C6ρ0 was generated from C6 glioma cells after long­term exposure to ethidium bromide and 2',3'­dideoxycytidine in order to determine the effect of mtDNA damage on cell proliferation and pathological changes in glioma cells. Single cell clones were isolated and identified after 42 days of incubation. Repopulated cybrids were formed when the clonal C6ρ0 cells were fused with rat platelets and no difference was observed in their growth in a selective medium without uridine and pyruvate compared with the growth of the parent C6 cells. Disruption of mtDNA resulted in changes in mitochondrial morphology, decreased cell proliferation, reduced intracellular reactive oxygen species and intracellular ATP, along with decreased mtDNA and mitochondrial membrane potential in C6ρ0 cells compared with the C6 cells. Taken together, C6ρ0 cells without mtDNA were established for the first time and their characteristics were compared with parent cells. This C6ρ0 cell line could be used to explore the contribution of mitochondrial dysfunction and mtDNA mutations in the pathogenesis of glioma.


Subject(s)
Brain Neoplasms/genetics , DNA, Mitochondrial/genetics , Glioma/genetics , Mitochondria/genetics , Adenosine Triphosphate/metabolism , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , DNA, Mitochondrial/metabolism , Glioma/metabolism , Glioma/pathology , Intracellular Space/metabolism , Membrane Potential, Mitochondrial/genetics , Membrane Potential, Mitochondrial/physiology , Mitochondria/metabolism , Rats , Reactive Oxygen Species/metabolism
13.
Neurotox Res ; 39(2): 327-334, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33196952

ABSTRACT

Since proline metabolism has been implicated to play an underlying role in apoptotic signaling and cancer, and hyperprolinemic patients present susceptibility to tumors development, this study investigated the effect of proline on cell death, cell cycle, antioxidant enzymes activities, and immunocontent/activity of proteins involved in cell death/survival signaling pathways in C6 glioma cells. C6 cells were incubated with proline (0-5 mM) for 1 h, 24 h, 48 h, 72 h, or 7 days. Proline in high concentrations slightly decreased LDH release, and no cytotoxic effect was seen by Annexin-PI staining. Superoxide dismutase and catalase activities were increased by proline (1 mM) after 72 h, suggesting an increase in reactive species levels. Acetylcholinesterase activity was inhibited by proline at 1, 3, and 5 mM. The cell cycle progression was not altered. Results from Western blot analyses showed that proline at 1 mM after 72 h increased p-NF-ĸB and decreased acetylcholinesterase immunocontent but did not altered AKT, p-AKT, GSK3ß, and p-GSK3ß. Taken together, the data suggest that high proline levels seems to favor the signaling pathways towards cell proliferation, since acetylcholinesterase, which may act as tumor suppressor, is inhibited by proline. Also, p-NF-κB is increased by proline treatment and its activation is related to tumor cell proliferation and cellular response to oxidants. Proline also induced oxidative stress, but it appears to be insufficient to induce a significant change in cell cycle progression. These data may be related, at least in part, to the increased susceptibility to tumor development in hyperprolinemic individuals.


Subject(s)
Cell Cycle/drug effects , Cell Death/drug effects , Glioblastoma/metabolism , Oxidative Stress/drug effects , Proline/administration & dosage , Proline/metabolism , Animals , Cell Line, Tumor , Rats , Signal Transduction
14.
Nat Prod Res ; 35(23): 5489-5492, 2021 Dec.
Article in English | MEDLINE | ID: mdl-32608261

ABSTRACT

A HP20 resin-based unique method was adopted to get an active fraction of the hydroalcoholic extract of G. glabra roots. The fraction showed potent cytotoxicity against cancer cell line and was further subjected to detailed phytochemical investigation to obtain ten biomarkers. The isolated compounds were also tested for the cytotoxicity against the C6 glioma cell line in vitro using MTT assay. Among the isolated compounds, glycyrrhetic acid (1), glabrol (6), and glabridin (9) exhibited significant cytotoxicity. The compounds showed a dose-dependent decrease in cell viability. The active compounds were subjected to molecular docking study against topoisomerase I and topoisomerase II to support the mechanism of antitumor activity.


Subject(s)
Glioma , Glycyrrhiza , Glioma/drug therapy , Humans , Molecular Docking Simulation , Plant Extracts/pharmacology , Plant Roots
15.
Cell Transplant ; 29: 963689720964383, 2020.
Article in English | MEDLINE | ID: mdl-33356508

ABSTRACT

The optic pathway glioma (OPG) is a slow-growing brain tumor that arises along the optic nerve or its downstream connections and causing vision to gradually worsen with time. This tumor forms in children with a genetic condition called neurofibromatosis type 1 (NF1), causing tumors to grow on nerves. In normal conditions, glial cells are there to support and protect nerve cells but, in NF1-OPG, glial cells have a genetic defect and grow out of control forming a tumor called a glioma. There are no rat models of NF1-OPG that can be used to explore various treatment options, and mouse models make interventional studies difficult due to their small eye size. We have created a model in which to study the progression of tumor growth in the optic nerve and establish the anatomical and functional consequences of the model and determine its suitability to serve as a surrogate for human disease. C6 rat glioma cells were injected into the optic nerve of Long-Evans rats and allowed to proliferate for 2 weeks. The eye clearly showed proptosis and lens opacity was observed, likely due to increased intraocular pressure caused by growing tumors. Hematoxylin-eosin staining showed marked cellularity, with hyperchromatism and pleomorphism. There was prominent area of necrosis with neoplastic cells palisading around the penumbra. Immunostaining with markers such as S100, ß-tubulin III, Foxp3, CD45, Vimentin, and Ki67 confirmed low-grade tumor formation, with a mild immune response. Our results show the utility of a surgically induced rat model of OPG that may be used for exploring various treatment options for NF1 ocular tumors.


Subject(s)
Glioma/metabolism , Optic Nerve/metabolism , Retinal Diseases/metabolism , Cell Line, Tumor , Flow Cytometry , Forkhead Transcription Factors/metabolism , Glioma/genetics , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , Neurofibromin 1/metabolism , Optic Nerve/pathology , Retinal Diseases/genetics , Retinal Diseases/pathology , Tubulin/metabolism , Vimentin/metabolism
16.
Asia Ocean J Nucl Med Biol ; 8(2): 102-108, 2020.
Article in English | MEDLINE | ID: mdl-32714997

ABSTRACT

OBJECTIVES: L-4-borono-2-18F-fluoro-phenylalanine (L-[18F]FBPA), a substrate of L-type amino acid transporter 1 (LAT1), is a tumor-specific probe used in positron emission tomography (PET). On the other hand, it has not been examined whether another isomer D-[18F]FBPA accumulates specifically in the tumor. Here, we compared the accumulation of D-[18F]FBPA in C6 glioma and inflammation to evaluate the performance of D-[18F]FBPA as a tumor-specific probe. METHODS: HEK293-LAT1 and HEK293-LAT2 cells were tested for [14C]-leucine or [14C]-alanine transport, and IC50 values of L- and D-FBPA were evaluated in both cell types. PET was conducted in rat xenograft model of C6 glioma with LAT1 expression and model of turpentine oil-induced subcutaneous inflammation (n=10 for both models). The concentrations of D-[18F]FBPA were compared between glioma and inflammatory lesion using standardized uptake value (SUV). RESULTS: In contrast to L-FBPA, which inhibited substrate uptake in both HEK293-LAT1 and -LAT2 cells, D-FBPA showed no inhibitory effect on both cells, suggesting low transporter selectivity of D-[18F]FBPA against LAT1 and LAT2. Static PET analysis showed low accumulation of D-[18F]FBPA in C6 glioma and inflammatory lesion (SUVmax=0.80±0.16, 0.56±0.09, respectively). Although there was a statistical difference in SUVmax between these tissues, it was difficult to distinguish glioma from inflammation on the PET image due to its low uptake level. Therefore, it was suggested that D-[18F]FBPA is not a suitable tumor-specific probe for oncology PET in contrast to L-[18F]FBPA. CONCLUSION: This study demonstrated that D-[18F]FBPA is not a LAT1-specific PET probe and shows low uptake in C6 glioma, indicating its unsuitability as a tumor diagnosis PET probe.

17.
J Drug Target ; 28(7-8): 789-801, 2020.
Article in English | MEDLINE | ID: mdl-32242754

ABSTRACT

Ligands are an important part of targeted drug delivery systems. Optimised lignads not only improve the target efficiency, but also enhance therapeutical effect of drugs. In our research, five sugar molecules (Mannose, Galactose, Glucose, Malt disaccharide, and Maltotriose) conjugated PEG600-DSPE were synthesised, of which polysaccharides were first discovered by us as sugar ligands to modify liposomes, which interacts with over expressive GLUT on cancer cells. DiO was encapsulated as fluorescent probe to evaluate their cellular uptake abilities of targeting C6 glioma cells, and the distribution in different visceral organs of rats. The results demonstrated that Malt disaccharide and Glucose-PEG600-DSPE had the strong efficiency of cellular uptake by C6 glioma cells. The distribution and accumulation of liposomes showed that different sugars modified liposomes could target different visceral organs in rats. It has provided a novel idea for ligand selectivity and optimisation of nanocarriers for tumour targeted therapy.


Subject(s)
Drug Delivery Systems/methods , Hexoses/chemistry , Liposomes/chemistry , Polyethylene Glycols/chemistry , Animals , Cell Line, Tumor , Cell Survival , Ligands , Nanoparticles , Rats , Rats, Sprague-Dawley , Surface Properties
18.
Pharm Dev Technol ; 25(6): 757-766, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32192406

ABSTRACT

Quercetin (Qu) is a natural flavonoid present in many commonly consumed food items. The dietary phytochemical quercetin prevents tumor proliferation and is a potent therapeutic cancer agent. The purpose of this study was to synthesize and characterize quercetin-loaded poly(lactic-co-glycolic acid) nanoparticles (Qu1NP, Qu2NP, and Qu3NP) with different size and encapsulation properties and to evaluate their in vitro activity on C6 glioma cells. Nanoparticles were synthesized by single emulsion solvent evaporation method. Then, particle size, zeta potential, polydispersity index and encapsulation efficiency of nanoparticles were determined. Particle size of Qu1NP, Qu2NP, and Qu3NPs were determined as 215.2 ± 6.2, 282.3 ± 7.9, and 584.5 ± 15.2 nm respectively. Treating C6 glioma cells with all nanoparticle formulations effectively inhibited the cell proliferation. Qu1NPs were showed the lowest IC50 value in 48 h with 29.9 µg/ml and achieved higher cellular uptake among other nanoparticles and Qu. Additionally, 48-h treatment with Qu1NPs significantly decreased MDA level (14.90 nmol/µg protein) on C6 glioma cells which is related to reduced oxidative stress in cells. Findings of this study revealed that quercetin's cellular uptake and anti-oxidant activity is improved by small-sized Qu1NPs in C6 glioma cells.


Subject(s)
Antioxidants/toxicity , Cytotoxins/toxicity , Glioma/metabolism , Nanoparticles/metabolism , Nanoparticles/toxicity , Quercetin/toxicity , Animals , Antioxidants/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cytotoxins/pharmacokinetics , Glioma/drug therapy , Particle Size , Quercetin/pharmacokinetics , Rats
19.
J Inorg Biochem ; 207: 111053, 2020 06.
Article in English | MEDLINE | ID: mdl-32193050

ABSTRACT

A set of new copper(II) complexes containing the Schiff base ligand derived from pyridine-2-carboxaldehyde and 5,6-diamino-1,3-dimethyluracil (6-amino-1,3-dimethyl-5-[(pyridin-2-ylmethylidene)-amino]-pyrimidine-2,4(1H,3H)-dione) with several anions (Cl-, Br-, I-, ClO4-, NO3-) and, two of them with 1,10-phenanthroline, were synthesized and characterized by means of elemental analysis, FT-IR, and single-crystal X-ray diffraction methods. Their ability to act as antitumor agents against C6 glioma cells has been also explored. These complexes contain copper a redox active metal essential for the regulation of cellular pathways that are fundamental for brain function. The antiproliferative activity of the complexes and their effect on cell cycle, apoptosis profile, bioenergetic behavior, intracellular reactive oxygen species (ROS) production, autophagy and enzyme antioxidant defense systems (superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx) activities) were analyzed in C6 glioma cells. Although the compounds show limited antiproliferative activity, they are able to modify S-phase of cell cycle and induce G2/M phase arrest. Also, copper(II) complexes promote apoptosis and, in a lesser extent, autophagy, being both processes modulated by ROS generation, due to their property to affect the enzyme antioxidant defense systems, mainly SOD and CAT but not GPx.


Subject(s)
Aldehydes/chemistry , Cell Proliferation/drug effects , Coordination Complexes/chemistry , Copper/chemistry , Pyridines/chemistry , Schiff Bases/chemistry , Uracil/analogs & derivatives , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/chemistry , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Coordination Complexes/pharmacology , Copper/pharmacology , Crystallography, X-Ray , Glioma/metabolism , Humans , Oxidation-Reduction/drug effects , Phenanthrolines/chemistry , Rats , Reactive Oxygen Species , Schiff Bases/pharmacology , Spectroscopy, Fourier Transform Infrared , Uracil/chemistry
20.
Brain Behav Immun ; 87: 645-659, 2020 07.
Article in English | MEDLINE | ID: mdl-32097763

ABSTRACT

Glioblastoma is a kind of malignant tumour and originates from the central nervous system. In the last century, some researchers and clinician have noticed that the psychosocial and neurocognitive functioning of patients with malignant gliomas can be impaired. Many clinical studies have demonstrated that part of patients, adults or children, diagnosed with glioblastoma will suffer from cognitive deficiency during their clinical course, especially in long-term survivors. Many nanoparticles (NPs) can inhibit the biological functions of tumours by modulating tumour-associated inflammation, which provokes angiogenesis and tumour growth. As one of the best antiviral nanoparticles (AVNPs), AVNP2 is the 2nd generation of AVNP2 that have been conjugated to graphite-graphene for improving physiochemical performance and reducing toxicity. AVNP2 inactivates viruses, such as the H1N1 and H5N1influenza viruses and even the SARS coronavirus, while it inhibits bacteria, such as MRSA and E. coli. As antimicrobials, nanoparticles are considered to be one of the vectors for the administration of therapeutic compounds. Yet, little is known about their potential functionalities and toxicities to the neurotoxic effects of cancer. Herein, we explored the functionality of AVNP2 on inhibiting C6 in glioma-bearing rats. The novel object-recognition test and open-field test showed that AVNP2 significantly improved the neuro-behaviour affected by C6 glioma. AVNP2 also alleviated the decline of long-term potentiation (LTP) and the decreased density of dendritic spines in the CA1 region induced by C6. Western blot assay and immunofluorescence staining showed that the expressions of synaptic-related proteins (PSD-95 and SYP) were increased, and these findings were in accordance with the results mentioned above. It revealed that the sizes of tumours in C6 glioma-bearing rats were smaller after treatment with AVNP2. The decreased expression of inflammatory factors (IL-1ß, IL-6 and TNF-α) by Western blotting assay and ELISA, angiogenesis protein (VEGF) by Western blotting assay and other related proteins (BDNF, NF-ĸB, iNOS and COX-2) by Western blotting assay in peri-tumour tissue indicated that AVNP2 could control tumour-associated inflammation, thus efficiently ameliorating the local inflammatory condition and, to some extent, inhibiting angiogenesis in C6-bearing rats. In conclusion, our results suggested that AVNP2 could have an effect on the peri-tumor environment, obviously restraining the growth progress of gliomas, and eventually improving cognitive levels in C6-bearing rats.


Subject(s)
Antiviral Agents/therapeutic use , Brain Neoplasms/complications , Brain Neoplasms/psychology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/prevention & control , Encephalitis/etiology , Encephalitis/prevention & control , Glioma/complications , Glioma/psychology , Nanoparticles/therapeutic use , Animals , Behavior, Animal , Body Weight/drug effects , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/pathology , Cognitive Dysfunction/psychology , Cytokines/biosynthesis , Dendritic Spines/drug effects , Dendritic Spines/pathology , Humans , Long-Term Potentiation/drug effects , Male , Neoplasm Transplantation , Rats , Rats, Sprague-Dawley , Recognition, Psychology/drug effects , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...