Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Aging (Albany NY) ; 16(10): 8697-8716, 2024 05 09.
Article in English | MEDLINE | ID: mdl-38738989

ABSTRACT

BACKGROUND: It is unknown what variables contribute to the formation and multiplication of low-grade gliomas (LGG). An emerging process of cell death is called cuproptosis. Our research aims to increase therapeutic options and gain a better understanding of the role that cuproptosis-related genes play in the physical characteristics of low-grade gliomas. METHODS: The TCGA database was utilized to find cuproptosis genes that may be used to develop LGG risk model. Cox analysis in three different formats: univariate, multivariate, and LASSO. The gene signature's independent predictive ability was assessed using ROC curves and Cox regression analysis based on overall survival. Use of CGGA data and nomogram model for external validation Immunohistochemistry, gene mutation, and functional enrichment analysis are also employed to clarify risk models' involvement. Next, we analyzed changes in the immunological microenvironment in the risk model and forecasted possible chemotherapeutic drugs to target each group. Finally, we validated the protein expression levels of cuproptosis-related genes using LGG and adjacent normal tissues in a small self-case-control study. RESULTS: This study developed a glioma predictive model based on five cuproptosis-associated genes. Compared to the high-risk group, the low-risk group's OS was significantly longer. The ROC curves showed high genetic signature performance in both groups. The signature-based categorisation was also linked to clinical characteristics and molecular subgroups. The prognosis of individuals with grade 2 or 3 glioma is also influenced by our risk model. Immunological testing revealed that the high-risk group had more immune cells and immunological function. The risk model also predicted immunotherapy and chemotherapy medication results. Also, this study confirmed that the expression of cuproptosis-related genes by Western blot. CONCLUSION: We developed a prediction model for LGG patients using genes associated with cuproptosis. With acceptable prediction performance, this risk model may effectively stratify the prognosis of glioma patients.


Subject(s)
Brain Neoplasms , Glioma , Humans , Glioma/genetics , Glioma/mortality , Glioma/pathology , Prognosis , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Nomograms , Neoplasm Grading , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Female , Male , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism
2.
Exp Neurol ; 370: 114568, 2023 12.
Article in English | MEDLINE | ID: mdl-37820939

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) is an important cause of disability and death. TBI leads to multiple forms of nerve cell death including ferroptosis due to iron-dependent lipid peroxidation. Anacardic acid (AA) is a natural component extracted from cashew nut shells, which has been reported to have neuroprotective effects in traumatic brain injury. We investigated whether AA has an anti-ferroptosis effect in TBI. METHODS: We used the Feeney free-fall impact method to construct a TBI model to investigate the effect of AA on ferroptosis caused by TBI, in which Ferrostatin-1 (Fer-1), a ferroptosis inhibitor, served as a positive control group. We first identified the therapeutic effect of AA on TBI through modified neurological severity score (mNSS) and determined the appropriate concentration. Secondly, we investigated the effect of AA on the expression level of the key protein of ferroptosis by Western blotting and immunohistochemistry. Then the effect of AA on nerve tissue injury and nerve function improvement was verified. Finally, enzym-linked immunosorbent assay (ELISA) was used to verify that AA could reduce inflammation after TBI. RESULTS: We found the intensely inhibitory effect of AA on ferroptosis, which is in parallel with the results obtained after Fer-1 treatment. In addition, AA and Fer-1 mitigated TBI-mediated tissue defects, destruction of the blood-brain barrier, and neurodegeneration. Novel object recognition (NOR), mNSS and water maze test showed that AA could significantly reduce the impairment of neural function and behavioral cognitive ability caused by TBI. Finally, we also demonstrated that AA has not only an anti-ferroptosis effect, but also an anti-inflammation effect. CONCLUSIONS: AA can reduce the neurological impairment and behavioral cognitive impairment caused by TBI through the dual effect of anti-ferroptosis and anti-inflammation.


Subject(s)
Brain Injuries, Traumatic , Ferroptosis , Humans , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/metabolism , Anacardic Acids/therapeutic use , Anti-Inflammatory Agents/therapeutic use
3.
Front Genet ; 12: 698831, 2021.
Article in English | MEDLINE | ID: mdl-34721518

ABSTRACT

Glioma is considered one of the most lethal brain tumors, as the aggressive blood vessel formation leads to high morbidity and mortality rates. However, the mechanisms underlying the initiation and progression of glioma remain unclear. Here, we aimed to reveal the role of circTLK1 in glioma development. Our results revealed that circTLK1 is highly expressed in glioma tumor tissues and glioma cell lines. We then conducted a series of experiments that showed that circTLK1 was involved in the progression of gliomas. Mechanistically, investigation of the factors downstream of circTLK1 revealed that circTLK1 activated JAK/STAT signaling in glioma cells. Furthermore, AGO2-RIP, RNA-pull down, and luciferase reporter gene assays led to the identification of the novel circTLK1/miR-452-5p/SSR1 axis. Moreover, we investigated the upstream regulator of circTLK1 and found that circTLK1 expression in glioma cells could be regulated by the transcriptional factor PBX2. Taken together, our findings show that circTLK1 mediated by PBX2 activates JAK/STAT signaling to promote glioma progression through the miR-452-5p/SSR1 pathway. These results provide new insights into glioma diagnosis and therapy.

4.
Biomater Sci ; 9(4): 1466, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33570070

ABSTRACT

Correction for 'Injectable postoperative enzyme-responsive hydrogels for reversing temozolomide resistance and reducing local recurrence after glioma operation' by Zongren Zhao et al., Biomater. Sci., 2020, 8, 5306-5316, DOI: 10.1039/D0BM00338G.

5.
Int J Biol Macromol ; 176: 145-156, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33571591

ABSTRACT

Karyopherins mediate the macromolecular transport between the cytoplasm and the nucleus and participate in cancer progression. However, the role and mechanism of importin-11 (IPO11), a member of the karyopherin family, in glioma progression remain undefined. Effects of IPO11 on glioma progression were detected using CCK-8, colony formation assay, flow cytometry analysis, caspase-3 activity assay, and Transwell invasion assay. Western blot analysis was used to detect the expression of active caspase-3, active caspase-7, active caspase-9, N-cadherin, Vimentin, E-cadherin, ß-catenin, and c-Myc. The activity of Wnt/ß-catenin pathway was evaluated by the T-cell factor/lymphoid enhancer factor (TCF/LEF) transcription factor reporter assay. Results showed that IPO11 knockdown inhibited proliferation and reduced colony number in glioma cells. IPO11 silencing promoted the apoptotic rate, increased expression levels of active caspase-3, caspase-7, and caspase-9, and enhanced caspase-3 activity. Moreover, IPO11 silencing inhibited glioma cell invasion by suppressing epithelial-to-mesenchymal transition (EMT). Mechanistically, IPO11 knockdown inactivated the Wnt/ß-catenin pathway. ß-Catenin overexpression abolished the effects of IPO11 silencing on the proliferation, apoptosis, and invasion in glioma cells. Furthermore, IPO11 silencing blocked the malignant phenotypes and repressed the Wnt/ß-catenin pathway in vivo. In conclusion, IPO11 knockdown suppressed the malignant phenotypes of glioma cells by inactivating the Wnt/ß-catenin pathway.


Subject(s)
Brain Neoplasms/metabolism , Carcinogens/metabolism , Glioma/metabolism , beta Karyopherins/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Carcinogens/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Female , Gene Knockdown Techniques , Glioma/genetics , Glioma/pathology , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Phenotype , Transcriptome , Up-Regulation , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/genetics
6.
ACS Appl Mater Interfaces ; 12(47): 52319-52328, 2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33166112

ABSTRACT

Glioma is the most prevalent type of malignant brain tumor and is usually very aggressive. Because of the high invasiveness and aggressive proliferative growth of glioma, it is difficult to resect completely or cure with surgery. Residual glioma cells are a primary cause of postoperative recurrence. Herein, we describe a hypoxia-responsive lipid polymer nanoparticle (LN) for fluorescence-guided surgery, chemotherapy, photodynamic therapy (PDT), and photothermal therapy (PTT) combination multitherapy strategies targeting glioma. The hypoxia-responsive LN [LN (DOX + ICG)] contains a hypoxia-responsive component poly(nitroimidazole)25 [P-(Nis)25], the glioma-targeting peptide angiopep-2 (A2), indocyanine green (ICG), and doxorubicin (DOX). LN (DOX + ICG) comprises four distinct functional components: (1) A2: A2 modified nanoparticles effectively target gliomas, enhancing drug concentration in gliomas; (2) P-(Nis)25: (i) the hydrophobic component of LN (DOX + ICG) with hypoxia responsive ability to encapsulate DOX and ICG; (ii) allows rapid release of DOX from LN (DOX + ICG) after 808 nm laser irradiation; (3) ICG: (i) ICG allows imaging-guided surgery, combining PDT and PTT therapies; (ii) upon irradiation with an 808 nm laser, ICG creates a hypoxic environment; (4) DOX inhibits glioma growth. This work demonstrates that LN (DOX + ICG) might provide a novel clinical approach to preventing post-surgical recurrence of glioma.


Subject(s)
Doxorubicin/chemistry , Lipids/chemistry , Nanoparticles/chemistry , Polymers/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Antibiotics, Antineoplastic/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Female , Glioma/diagnostic imaging , Glioma/drug therapy , Glioma/pathology , Humans , Indocyanine Green/chemistry , Indocyanine Green/pharmacology , Indocyanine Green/therapeutic use , Infrared Rays , Mice , Mice, Inbred ICR , Peptides/chemistry , Peptides/therapeutic use , Photochemotherapy , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photothermal Therapy , Transplantation, Heterologous
7.
Biomater Sci ; 8(15): 4370, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32638709

ABSTRACT

Correction for 'Injectable postoperative enzyme-responsive hydrogels for reversing temozolomide resistance and reducing local recurrence after glioma operation' by Zongren Zhao et al., Biomater. Sci., 2020, DOI: .

8.
Int J Nanomedicine ; 15: 3347-3362, 2020.
Article in English | MEDLINE | ID: mdl-32494134

ABSTRACT

INTRODUCTION: Temozolomide (TMZ) is the first-line chemotherapeutic option to treat glioma; however, its efficacy and clinical application are limited by its drug resistance properties. Polo-like kinase 1 (PLK1)-targeted therapy causes G2/M arrest and increases the sensitivity of glioma to TMZ. Therefore, to limit TMZ resistance in glioma, an angiopep-2 (A2)-modified polymeric micelle (A2PEC) embedded with TMZ and a small interfering RNA (siRNA) targeting PLK1 (siPLK1) was developed (TMZ-A2PEC/siPLK). MATERIALS AND METHODS: TMZ was encapsulated by A2-PEG-PEI-PCL (A2PEC) through the hydrophobic interaction, and siPLK1 was complexed with the TMZ-A2PEC through electrostatic interaction. Then, an angiopep-2 (A2) modified polymeric micelle (A2PEC) embedding TMZ and siRNA targeting polo-like kinase 1 (siPLK1) was developed (TMZ-A2PEC/siPLK). RESULTS: In vitro experiments indicated that TMZ-A2PEC/siPLK effectively enhanced the cellular uptake of TMZ and siPLK1 and resulted in significant cell apoptosis and cytotoxicity of glioma cells. In vivo experiments showed that glioma growth was inhibited, and the survival time of the animals was prolonged remarkably after TMZ-A2PEC/siPLK1 was injected via their tail vein. DISCUSSION: The results demonstrate that the combination of TMZ and siPLK1 in A2PEC could enhance the efficacy of TMZ in treating glioma.


Subject(s)
Cell Cycle Proteins/metabolism , Drug Delivery Systems , Glioma/drug therapy , Nanoparticles/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/administration & dosage , Temozolomide/administration & dosage , Temozolomide/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Resistance, Neoplasm/drug effects , Endocytosis/drug effects , Endosomes/drug effects , Endosomes/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Silencing/drug effects , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Temozolomide/pharmacology , Tissue Distribution/drug effects , Treatment Outcome , Polo-Like Kinase 1
9.
Biomater Sci ; 8(19): 5306-5316, 2020 Sep 30.
Article in English | MEDLINE | ID: mdl-32573615

ABSTRACT

Glioma is the most aggressive primary malignant brain tumor. The eradication of the gliomas by performing neurosurgery has not been successful due to the diffuse nature of malignant gliomas. Temozolomide (TMZ) is the first-line agent in treating gliomas after surgery, and its therapeutic efficacy is limited mainly due to the high activity levels of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT) in glioma cells. Herein, we used an injectable matrix metalloproteinase (MMP) enzyme responsive hydrogel that loaded TMZ and O6-benzylamine (BG) (MGMT inhibitor) for eradicating residual TMZ-resistant gliomas after surgery. The hydrogels exhibited three features: (1) TMZ and BG could be encapsulated within the hydrophobic lamellae of the hydrogel to form Tm (TMZ + BG) hydrogels; (2) The hydrogels could release TMZ and BG in response to the high concentration of MMP enzymes after glioma surgery; (3) The hydrogels could increase local TMZ concentration and reduce side effects of BG. In vivo, the Tm (TMZ + BG) hydrogels inhibited the MGMT expression and sensitized TMZ-resistant glioma cells to TMZ. Moreover, the Tm (TMZ + BG) hydrogels effectively reduced the recurrence of TMZ-resistant glioma after surgery and significantly enhanced the efficiency of TMZ to inhibit glioma growth. Together, these data suggest that an MMP-responsive hydrogel is a promising localized drug delivery method to inhibit TMZ-resistant glioma recurrence after surgery.


Subject(s)
Dacarbazine , Glioma , Antineoplastic Agents, Alkylating/therapeutic use , Cell Line, Tumor , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Resistance, Neoplasm , Glioma/drug therapy , Humans , Hydrogels/pharmacology , Temozolomide/pharmacology , Temozolomide/therapeutic use
11.
J Mol Med (Berl) ; 97(11): 1575-1588, 2019 11.
Article in English | MEDLINE | ID: mdl-31673738

ABSTRACT

Glioblastoma is one of the most aggressive types of brain tumor. Epidermal growth factor receptors (EGFRs) are overexpressed in glioma, and EGFR amplifications and mutations lead to rapid proliferation and invasion. EGFR-targeted therapy might be an effective treatment for glioma. Gefitinib (Ge) is an EGFR tyrosine kinase inhibitor (TKI), and Golgi phosphoprotein 3 (GOLPH3) expression is associated with worse glioma prognosis. Downregulation of GOLPH3 could promote EGFR degradation. Here, an angiopep-2 (A2)-modified cationic lipid-poly (lactic-co-glycolic acid) (PLGA) nanoparticle (A2-N) was developed that can release Ge and GOLPH3 siRNA (siGOLPH3) upon entering glioma cells and therefore acts as a combinatorial anti-tumor therapy. The in vitro and in vivo studies proved that A2-N/Ge/siGOLPH3 successfully crossed the blood-brain barrier (BBB) and targeted glioma. Released siGOLPH3 effectively silenced GOLPH3 mRNA expression and further promoted EGFR and p-EGFR degradation. Released Ge also markedly inhibited EGFR signaling. This combined EGFR-targeted action achieved remarkable anti-glioma effects and could be a safe and effective treatment for glioma. KEY MESSAGES: Angiopep-2-modified cationic lipid polymer can penetrate the BBB. Gefitinib can inhibit EGFR signaling and block the autophosphorylation of critical tyrosine residues on EGFR. GOLPH3 siRNA can be transfected into glioma and downregulate GLOPH3 expression. A2-N/Ge/siGOLPH3 can inhibit glioma growth.


Subject(s)
ErbB Receptors/metabolism , Gefitinib/therapeutic use , Glioma/drug therapy , Glioma/metabolism , Membrane Proteins/metabolism , Nanoparticles/chemistry , Phosphoproteins/metabolism , RNA, Small Interfering/metabolism , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Male , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphoproteins/genetics , RNA, Small Interfering/genetics
12.
J Biomed Nanotechnol ; 15(9): 1982-1993, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31387684

ABSTRACT

Radiotherapy (RT) has become one of the most effective treatments for malignant tumor. Intra-tumoral hypoxia is recognized as a chief reason that induces resistance to radiation. Moreover, the toxicities of RT to normal tissues limits the usage of high doses of radiation to eliminate cancer cells. Therefore, developing an effective radiosensitizer is critical for improving the curative effects of RT. In the present study, we developed angiopep-2 (A2) modified hypoxic lipid radiosensitizer (HLR) coated gold nanoparticles (GNPs) (referred to as A2-HRGNPs) to increase the RT sensitivity of tumors. The A2-HRGNPs are comprised of the following two functional components: (1) HLR enhances the RT sensitivity on hypoxic tumor cells; (2) alkylthiol modified GNPs (DGNPs) increase radiation effects by a dose enhancing effect in RT. Our findings suggest that the synergistic radiosensitizing effects of A2-HRGNPs can significantly enhance radiosensitization effects and thus, inhibit tumor growth in vivo.


Subject(s)
Metal Nanoparticles , Neoplasms , Gold , Humans , Hypoxia , Lipids , Radiation-Sensitizing Agents
13.
Drug Deliv ; 26(1): 34-44, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30744436

ABSTRACT

The addition of temozolomide (TMZ) to radiotherapy (RT) improves survival of patients with glioblastoma (GBM). However, TMZ + RT causes excess toxicity in patients. In this study, we prepared angiopep-2 (A2) modified lipid-poly (hypoxic radiosensitized polyprodrug) nanoparticles for TMZ delivery (A2-P(MIs)25/TMZ) to achieve synergistic effects against glioma. This A2-P(MIs)25/TMZ display highly promising advantages: (1) a hydrophobic P-(MIs)25 core where poorly water-soluble TMZ can be encapsulated; (2) nitro groups of the hydrophobic P-(MIs)25 core that are converted into hydrophilic amino groups (P(NH2s)25) under low oxygen conditions to mimic the oxygen-increased sensitization to RT; (3) a lipid monolayer at the interface of the core and the shell to modify the A2 (a specific ligand for low-density lipoprotein receptor-related protein-1 (LRP-1), which are expressed in the blood-brain barrier (BBB) and human glioma cells), thereby enhancing the drug encapsulation efficiency in glioma. These nanoparticles appear as a promising and robust nanoplatforms for TMZ and hypoxic cell radiosensitization delivery.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Glioma/therapy , Nanoparticles/administration & dosage , Peptides/administration & dosage , Radiation-Sensitizing Agents/administration & dosage , Temozolomide/administration & dosage , Animals , Antineoplastic Agents, Alkylating/chemical synthesis , Antineoplastic Agents, Alkylating/metabolism , Cell Line, Tumor , Combined Modality Therapy/methods , Drug Delivery Systems/methods , Glioma/metabolism , Humans , Male , Mice , Mice, Inbred ICR , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/metabolism , Peptides/chemical synthesis , Peptides/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemical synthesis , Polylactic Acid-Polyglycolic Acid Copolymer/metabolism , Prodrugs/administration & dosage , Prodrugs/chemistry , Prodrugs/metabolism , Radiation-Sensitizing Agents/chemical synthesis , Radiation-Sensitizing Agents/metabolism , Radiotherapy/methods , Temozolomide/chemical synthesis , Temozolomide/metabolism , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...