Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 226
Filter
1.
Oxid Med Cell Longev ; 2021: 6219715, 2021.
Article in English | MEDLINE | ID: mdl-34765084

ABSTRACT

BACKGROUND: Mesenchymal stem cell-derived exosomes (MSC-exos) have been recognized as a promising therapeutic strategy for neonatal hypoxic-ischemic brain damage (HIBD). Recently, microglial pyroptosis was shown to play a vital role in the progression of neonatal HIBD. However, whether MSC-exos improve HIBD by regulating microglial pyroptosis remains unknown. METHODS: Exosomes were isolated from human umbilical cord mesenchymal stem cells (huMSCs) and identified by transmission electron microscopy (TEM), western blot, and nanoparticle tracking analysis (NTA). BV-2 cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to induce microglial ischemia/reperfusion (I/R) in vitro. CCK-8, ELISA, western blot, and Hoechst 33342/PI double staining were performed to detect the pyroptosis of BV-2 cells. Conditioned medium (CM) from BV-2 cells exposed to different treatments was used to investigate its effect on neuronal injury. Moreover, 3-methyladenine (3-MA) and mitochondrial division inhibitor-1 (mdi-1) were used to verify the involvement of mitophagy in the protection of MSC-exos against OGD/R-induced pyroptosis in BV-2 cells. Finally, FOXO3a siRNA was used to investigate the involvement of FOXO3a in MSC-exo-induced mitophagy and pyroptosis inhibition. RESULTS: Exosomes from huMSCs were successfully extracted. In OGD/R-exposed BV-2 cells, MSC-exos increased cell viability and decreased the expression of NLRP3, cleaved caspase-1, and GSDMD-N as well as the release of IL-1ß and IL-18. Compared with CM from OGD/R-exposed BV-2 cells treated with PBS, CM from OGD/R-exposed BV-2 cells treated with MSC-exos significantly increased the viability of SH-SY5Y cells and decreased LDH release. MSC-exos also increased the expression of TOM20 and COX IV in OGD/R-exposed BV-2 cells. Additionally, 3-MA and mdi-1 attenuated the inhibition of pyroptosis with MSC-exo treatment. Furthermore, FOXO3a siRNA partially abolished the neuroprotective effect of MSC-exos and attenuated mitophagy and pyroptosis inhibition induced by MSC-exo treatment. CONCLUSIONS: Our findings demonstrated that MSC-exos increased FOXO3a expression to enhance mitophagy, therefore protecting microglia from I/R-induced pyroptosis and alleviating subsequent neuronal injury.


Subject(s)
Exosomes/physiology , Forkhead Box Protein O3/metabolism , Microglia/cytology , Mitophagy , Neuroblastoma/prevention & control , Pyroptosis , Reperfusion Injury/prevention & control , Glucose/deficiency , Humans , Hypoxia/physiopathology , Mesenchymal Stem Cells/cytology , Microglia/metabolism , Microglia/pathology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Reperfusion Injury/etiology , Reperfusion Injury/pathology , Umbilical Cord/cytology
2.
Exp Cell Res ; 405(1): 112635, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34051241

ABSTRACT

Exosomes have been shown to have therapeutic potential for cerebral ischemic diseases. In this study, we investigated the neuroprotective effects of normoxic and hypoxic bone marrow mesenchymal stromal cells-derived exosomes (N-BM-MSCs-Exo and H-BM-MSCs-Exo, respectively) on oxygen-glucose deprivation (OGD) injury in mouse neuroblastoma N2a cells and rat primary cortical neurons. The proportions of dead cells in N2a and primary cortical neurons after OGD injury were significantly increased, and N-BM-MSCs-Exo (40 µg/ml) could reduce the ratios, noteworthily, the protective effects of H-BM-MSCs-Exo (40 µg/ml) were more potent. Western blotting analysis indicated that N-BM-MSCs-Exo decreased the expression of NLRP3, ASC, Caspase-1, GSDMD-N, cleaved IL-1ß and IL-18 in N2a cells. However, H-BM-MSCs-Exo (40 µg/ml) was more powerful in inhibiting the expression of these proteins in comparison with N-BM-MSCs-Exo. Similar results were obtained in primary cortical neurons. Immunofluorescence assays showed that after N-BM-MSCs-Exo and H-BM-MSCs-Exo treatment, the co-localization of NLRP3, ASC, Caspase-1 and the GSDMD translocation from the nucleus to the cytoplasm and membrane after OGD injury were reduced in N2a cells and primary cortical neurons, and H-BM-MSCs-Exo had a more obvious effect. In addition, N-BM-MSCs-Exo and H-BM-MSCs-Exo significantly reduced lactate dehydrogenase (LDH) release and the IL-18 levels in cell culture medium in N2a cells and primary cortical neurons. Once again H-BM-MSCs-Exo induced these effects more potently than N-BM-MSCs-Exo. All of these results demonstrated that N-BM-MSCs-Exo and H-BM-MSCs-Exo have significant neuroprotective effects against NLRP3 inflammasome-mediated pyroptosis. H-BM-MSCs-Exo has a more pronounced protective effect than N-BM-MSCs-Exo and may be used to ameliorate the progression of cerebral ischemia and hypoxia injury in patients.


Subject(s)
Exosomes/physiology , Hypoxia/physiopathology , Mesenchymal Stem Cells/cytology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroblastoma/prevention & control , Neurons/cytology , Pyroptosis , Animals , Cerebral Cortex/cytology , Cerebral Cortex/immunology , Cerebral Cortex/injuries , Cerebral Cortex/metabolism , Glucose/deficiency , Inflammasomes/physiology , Male , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function , Signal Transduction
3.
Exp Mol Pathol ; 120: 104622, 2021 06.
Article in English | MEDLINE | ID: mdl-33684392

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by neuronal and synaptic loss. The cytoplasmic tail of amyloid precursor protein (APP) undergoes sequential cleavage at a specific intracellular caspase site to generate the cytoplasmic terminal 31 (CT31) fragment. The APP-CT31 fragment is a potent inducer of apoptosis. The cytotoxicity of APP-CT31 in SH-SY5Y cells was evaluated by the lactate dehydrogenase (LDH) assay. TUNEL staining was used to detect apoptotic signals in SH-SY5Y cells and primary cortical neurons. The expression of apoptosis-related proteins, such as p53, PUMA (p53 up-regulated modulator of apoptosis), and cleaved was investigated by immunofluorescence analysis and Western blotting. In this study, we investigated the neuroprotective effect of neuregulin 1 (NRG1) against cytotoxicity induced by APP-CT31. Our data showed that CT31 induced cytotoxicity and apoptosis in SH-SY5Y cells and primary cortical neurons. NRG1 attenuated the neurotoxicity induced by the expression of APP-CT31. We also showed that APP-CT31 altered the expression of p53 and cleaved caspase 3. However, treatment with NRG1 rescued the APP-CT31-induced upregulation of p53 and cleaved caspase 3 expression. The protective effect of NRG1 was abrogated by inhibition of the ErbB4 receptor and Akt. These results indicate an important role of ErbB4/Akt signaling in NRG1-mediated neuroprotection, suggesting that endogenous NRG1/ErbB4 signaling represents a valuable therapeutic target in AD.


Subject(s)
Amyloid beta-Protein Precursor/adverse effects , Neuregulin-1/metabolism , Neuroblastoma/prevention & control , Neuroprotective Agents/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-4/metabolism , Apoptosis , Cell Proliferation , Humans , Neuregulin-1/genetics , Neuroblastoma/etiology , Neuroblastoma/pathology , Protein Domains , Proto-Oncogene Proteins c-akt/genetics , Receptor, ErbB-4/genetics , Tumor Cells, Cultured
4.
RNA Biol ; 18(11): 1546-1554, 2021 11.
Article in English | MEDLINE | ID: mdl-33427561

ABSTRACT

Pathological changes involving TDP-43 protein ('TDP-43 proteinopathy') are typical for several neurodegenerative diseases, including frontotemporal lobar degeneration (FTLD). FTLD-TDP cases are characterized by increased binding of TDP-43 to an abundant lncRNA, NEAT1, in the cortex. However it is unclear whether enhanced TDP-43-NEAT1 interaction represents a protective mechanism. We show that accumulation of human TDP-43 leads to upregulation of the constitutive NEAT1 isoform, NEAT1_1, in cultured cells and in the brains of transgenic mice. Further, we demonstrate that overexpression of NEAT1_1 ameliorates TDP-43 toxicity in Drosophila and yeast models of TDP-43 proteinopathy. Thus, NEAT1_1 upregulation may be protective in TDP-43 proteinopathies affecting the brain. Approaches to boost NEAT1_1 expression in the CNS may prove useful in the treatment of these conditions.


Subject(s)
Amyotrophic Lateral Sclerosis/prevention & control , Brain/metabolism , DNA-Binding Proteins/toxicity , Frontotemporal Dementia/prevention & control , Neuroblastoma/prevention & control , RNA, Long Noncoding/genetics , TDP-43 Proteinopathies/prevention & control , Amyotrophic Lateral Sclerosis/etiology , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Brain/pathology , Disease Models, Animal , Drosophila melanogaster , Frontotemporal Dementia/etiology , Frontotemporal Dementia/metabolism , Frontotemporal Dementia/pathology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA, Long Noncoding/administration & dosage , Saccharomyces cerevisiae , TDP-43 Proteinopathies/etiology , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology
5.
Life Sci ; 269: 119062, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33476635

ABSTRACT

AIMS: Cyclin-dependent kinase 5 (CDK5) is a potential target for the treatment of cerebral ischemia. CDK5 is one of the upstream regulators for Dynamin-related protein 1 (Drp1) phosphorylation. This study intends to discuss whether CDK5 inhibition conferring neuroprotection in cerebral ischemia through regulating Drp1 phosphorylation. MATERIALS AND METHODS: Mouse neuroblastoma N2a cells and N1E-115 cells were cultured and subjected to oxygen-glucose deprivation/reperfusion (OGDR). N2a cells and N1E-115 cells were treated with Roscovitine, a pharmacological inhibitor of CDK5, or transfected with CDK5 siRNA to knock down CDK5 expression. N2a cells were transfected with different plasmids (Drp1-Myc, the dephosphorylation-mimic mutant Drp1S616A-Myc and the phosphorylation-mimic mutant Drp1S616D-Myc). The expression of CDK5 and its activator p35, Drp1 and phosphorylated Drp1 on S616 was determined by western blot. The morphology of mitochondria was detected by immunofluorescence staining and the proportion of N2a cells with apoptosis was detected by flow cytometry analysis. KEY FINDINGS: Expression of CDK5, p35 and phosphorylated Drp1 on S616 was strongly upregulated after 4 h and 12 h reperfusion following 4 h oxygen-glucose deprivation (OGD) at protein level. CDK5 inhibition by pre-treated with Roscovitine or transfection with CDK5 siRNA significantly ameliorated OGDR induced mitochondrial fragmentation and apoptosis. Overexpression of the phosphorylation-mimic mutant Drp1S616D abrogated the protective effect of CDK5 inhibition against OGDR induced mitochondrial fragmentation and apoptosis. SIGNIFICANCE: Our data indicate that the neuroprotective effect of CDK5 inhibition against OGDR induced neuronal damage is Drp1S616 phosphorylation dependent. A better understanding of the neuroprotective mechanisms of CDK5 inhibition in cerebral ischemia will help to develop safe and efficacious drugs targeting CDK5 signaling for clinical use.


Subject(s)
Apoptosis , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Dynamins/metabolism , Glucose/deficiency , Mitochondria/pathology , Neuroblastoma/prevention & control , Oxygen/metabolism , Animals , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Dynamins/genetics , Mice , Mitochondria/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroprotective Agents , Phosphorylation , Reperfusion Injury/complications , Signal Transduction , Tumor Cells, Cultured
6.
Int J Mol Sci ; 21(21)2020 Oct 28.
Article in English | MEDLINE | ID: mdl-33126773

ABSTRACT

Pulsed electromagnetic fields (PEMFs) are emerging as an innovative, non-invasive therapeutic option in different pathological conditions of the central nervous system, including cerebral ischemia. This study aimed to investigate the mechanism of action of PEMFs in an in vitro model of human astrocytes, which play a key role in the events that occur following ischemia. 1321N1 cells were exposed to PEMFs or hypoxic conditions and the release of relevant neurotrophic and angiogenic factors, such as VEGF, EPO, and TGF-ß1, was evaluated by means of ELISA or AlphaLISA assays. The involvement of the transcription factor HIF-1α was studied by using the specific inhibitor chetomin and its expression was measured by flow cytometry. PEMF exposure induced a time-dependent, HIF-1α-independent release of VEGF from 1321N1 cells. Astrocyte conditioned medium derived from PEMF-exposed astrocytes significantly reduced the oxygen-glucose deprivation-induced cell proliferation and viability decrease in the neuron-like cells SH-SY5Y. These findings contribute to our understanding of PEMFs action in neuropathological conditions and further corroborate their therapeutic potential in cerebral ischemia.


Subject(s)
Astrocytes/cytology , Electromagnetic Fields , Glucose/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neuroblastoma/prevention & control , Oxygen/metabolism , Vascular Endothelial Growth Factor A/metabolism , Astrocytes/metabolism , Astrocytes/radiation effects , Cell Hypoxia , Cell Survival , Cells, Cultured , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protective Agents , Signal Transduction , Vascular Endothelial Growth Factor A/genetics
7.
Cell Cycle ; 19(18): 2367-2372, 2020 09.
Article in English | MEDLINE | ID: mdl-32809919

ABSTRACT

Recent studies have revealed that long non-coding RNAs (lncRNAs) play critical roles in the tumorigenesis and proliferation of human cancer. Several polymorphisms of lncRNAs have been found to be involved in the risk of neuroblastoma (NB). However, studies on the relationship between polymorphisms in lncRNA exons and NB are infrequent. We evaluated the association between rs11752942 A > G polymorphism in lnc-RNA-uc003opf.1 exon and neuroblastoma susceptibility by performing a hospital-based study with 275 patients and 531 controls. Odds ratios (ORs) and 95% confidence intervals (CIs) assessed by using logistic regression models were used to determine the strength of the association. We found that the rs11752942 G allele is significantly associated with decreased neuroblastoma risk (AG vs. AA: adjusted OR = 0.72, 95% CI = 0.53-0.98, P = 0.038; and AG/GG vs. AA: adjusted OR = 0.74, 95% CI = 0.55-0.99, P = 0.045) after adjusting for age and gender. This association was more prominent in females, subjects with tumor in the mediastinum or early-stage. Furthermore, the expression quantitative trait locus analysis indicated that rs11752942 G was associated with decreased expression of its neighboring gene LRFN2 mRNA. These results indicate that lncRNA-uc003opf.1 may be a novel potentially functional lncRNA that may be used as a predictive marker, for it might contribute to decreased neuroblastoma risk.


Subject(s)
Neuroblastoma/genetics , Polymorphism, Single Nucleotide , RNA, Long Noncoding/genetics , Age Factors , Asian People/genetics , China , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Infant , Male , Neuroblastoma/diagnosis , Neuroblastoma/ethnology , Neuroblastoma/prevention & control , Protective Factors , Quantitative Trait Loci , Retrospective Studies , Risk Assessment , Risk Factors , Sex Factors
8.
IUBMB Life ; 72(9): 2045-2055, 2020 09.
Article in English | MEDLINE | ID: mdl-32687247

ABSTRACT

Parkinson's disease (PD) is one of the most common neurodegenerative disorders. Long non-coding RNAs have important regulatory values in various human diseases. Non-coding RNA Activated by DNA Damage (NORAD) was reported to regulate PD progression in vitro, but its functional mechanism is fully unknown. We used 1-methyl-4-phenylpyridinium (MPP+ ) to establish the cell-based PD model. NORAD, microRNA-204-5p (miR-204-5p), and solute carrier family 5 member 3 (SLC5A3) levels were quantified using the quantitative real-time polymerase chain reaction. Cell viability and apoptosis were determined by Cell Counting Kit-8 and flow cytometry, respectively. The protein levels were analyzed via western blot. Cytotoxicity was assessed by the released lactate dehydrogenase level in cell supernatant. Oxidative stress and inflammation were measured by the standard indicators. Dual-luciferase reporter and RNA immunoprecipitation assays were performed for intergenic combination. First, we found that NORAD was obviously reduced in MPP+ -treated neuroblastoma cells and lightened the MPP+ -induced cytotoxicity, oxidative stress, and inflammatory response. Then, NORAD was shown to be a miR-204-5p sponge and avoided the injury induced by MPP+ in neuroblastoma cells via targeting miR-204-5p. SLC5A3 was a miR-204-5p target and could be regulated by NORAD/miR-204-5p axis. SLC5A3 knockdown assuaged the anti-miR-204-5p-induced protection for neuroblastoma cells from MPP+ . Altogether, NORAD played a neuroprotective role against the progression of MPP+ -induced PD model in neuroblastoma cells relying on the miR-204-5p/SLC5A3 axis. This study afforded the clear elaboration on the PD pathomechanism concerning NORAD.


Subject(s)
Gene Expression Regulation , Heat-Shock Proteins/metabolism , MicroRNAs/genetics , Neuroblastoma/prevention & control , Oxidative Stress , Parkinson Disease/prevention & control , RNA, Long Noncoding/genetics , Symporters/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Animals , Apoptosis , Cell Proliferation , Cell Survival , Heat-Shock Proteins/genetics , Humans , Mice , Neuroblastoma/chemically induced , Neuroblastoma/pathology , Parkinson Disease/etiology , Parkinson Disease/pathology , Signal Transduction , Symporters/genetics , Tumor Cells, Cultured
9.
Nutrients ; 12(3)2020 Feb 29.
Article in English | MEDLINE | ID: mdl-32121454

ABSTRACT

Environmental stress, exhaustive industrialization and the use of chemicals in our daily lives contribute to increasing incidence of cancer and other pathologies. Although the cancer treatment has revolutionized in last 2-3 decades, shortcomings such as (i) extremely high cost of treatment, (ii) poor availability of drugs, (iii) severe side effects and (iv) emergence of drug resistance have prioritized the need of developing alternate natural, economic and welfare (NEW) therapeutics reagents. Identification and characterization of such anti-stress NEW drugs that not only limit the growth of cancer cells but also reprogram them to perform their specific functions are highly desired. We recruited rat glioma- and human neuroblastoma-based assays to explore such activities of resveratrol, a naturally occurring stilbenoid. We demonstrate that nontoxic doses of resveratrol protect cells against a variety of stresses that are largely involved in age-related brain pathologies. These included oxidative, DNA damage, metal toxicity, heat, hypoxia, and protein aggregation stresses. Furthermore, it caused differentiation of cells to functional astrocytes and neurons as characterized by the upregulation of their specific protein markers. These findings endorse multiple bioactivities of resveratrol and encourage them to be tested for their benefits in animal models and humans.


Subject(s)
Antioxidants/pharmacology , Cellular Reprogramming/drug effects , Glioma/prevention & control , Neuroblastoma/prevention & control , Resveratrol/pharmacology , Aging/physiology , Animals , Antioxidants/therapeutic use , Astrocytes/drug effects , Astrocytes/physiology , Brain/cytology , Brain/drug effects , Brain/pathology , Cell Line, Tumor , DNA Damage/drug effects , Dose-Response Relationship, Drug , Environmental Exposure/adverse effects , Glioma/etiology , Glioma/pathology , Humans , Neuroblastoma/etiology , Neuroblastoma/pathology , Neurons/drug effects , Neurons/physiology , Oxidative Stress/drug effects , Rats , Resveratrol/therapeutic use
10.
Cell Biol Int ; 44(2): 630-636, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31743528

ABSTRACT

Alzheimer's disease (AD) is a chronic progressive neurodegenerative disorder. However, its pathogenetic mechanism is still poorly understood. An increasing number of studies have evidenced the important role of long noncoding RNAs (lncRNAs) in AD. The aim of the current study was to investigate the effect and molecular mechanism of the lncRNA X-inactive specific transcript (XIST) in AD. Bilateral common carotid artery occlusion (2VO) was used to induce an AD model in mice. Hydrogen peroxide (H2 O2 ) was used to induce an AD model in N2a cells. The lncRNA XIST, miR-124, and BACE1 messenger RNA expression levels were detected by a real-time polymerase chain reaction. The BACE1 protein expression level was detected by western blot and immunofluorescence assay. The Aß1-42 expression level was detected using an enzyme-linked immunosorbent assay kit. The expression level of lncRNA XIST was significantly upregulated in AD models, both in vivo and in vitro. Silencing of lncRNA XIST negatively regulated miR-124 and positively regulated BACE1 expression in N2a cells, which is attenuated by cotransfection of anti-miR-124 oligodeoxyribonucleotide (AMO-124). Silencing of lncRNA XIST reversed the effect of H2 O2 on miR-124, BACE1, and Aß1-42 expression in N2a cells, which was reversed by cotransfection of AMO-124. Silencing of lncRNA XIST attenuated AD-related BACE1 alteration through miR-124. LncRNA XIST may be a new potential target for the treatment of AD.


Subject(s)
Alzheimer Disease/prevention & control , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Gene Expression Regulation , Gene Silencing , MicroRNAs/genetics , Neuroblastoma/prevention & control , RNA, Long Noncoding/antagonists & inhibitors , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Animals , Apoptosis , Aspartic Acid Endopeptidases/genetics , Cell Proliferation , Disease Models, Animal , Mice , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA, Long Noncoding/genetics , Tumor Cells, Cultured
11.
J Steroid Biochem Mol Biol ; 192: 105384, 2019 09.
Article in English | MEDLINE | ID: mdl-31175966

ABSTRACT

Insulin-like growth factor-1 (IGF-1), an endogenous peptide, exerts important role in brain development, neurogenesis and neuroprotection. There are accumulating evidence for the interaction of IGF-1 and 17ß-estradiol systems. IGF-1/IGF-1 receptor (IGF-1R) signaling has been reported to regulate G-protein estrogen receptor (GPER) expression in cancer cells. Whether GPER is involved in the neuroprotective effect of IGF-1 against MPTP/MPP+-induced dopaminergic neuronal injury remains unclear. We showed that IGF-1 could improve MPTP-induced motor deficits and ameliorate the decreased contents of DA and its metabolites in striatum as well as the loss of TH-IR neurons in the substantia nigra (SN). IGF-1 pretreatment also reversed the changes of Bcl-2 and Bax protein expressions in SN in MPTP mice. These effects were abolished by IGF-1 receptor (IGF-1R) antagonist JB-1 or GPER antagonist G15 except the inhibitory effect of G15 on Bax protein expression. Moreover, IGF-1 pretreatment enhanced cell survival against MPP+-induced neurotoxicity in SH-SY5Y cells. IGF-1 exerted anti-apoptotic effects by restoring MPP+-induced changes of Bcl-2 and Bax protein expressions as well as mitochondria membrane potential. Co-treatment with JB-1 or G15 could block these effects. Furthermore, IGF-1 regulated the protein expression of GPER through activation of phosphatidylinositol 3-kinase (PI3-K) and mitogen-activated protein kinase (MAPK) signaling pathways. Overall, we show for the first time that GPER may contribute to the neuroprotective effects of IGF-1 against MPTP/MPP+-induced dopaminergic neuronal injury.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Dopaminergic Neurons/drug effects , Insulin-Like Growth Factor I/pharmacology , Neuroblastoma/prevention & control , Parkinson Disease/prevention & control , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroprotective Agents/pharmacology , Neurotoxins/adverse effects , Parkinson Disease/etiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured
12.
Life Sci ; 224: 95-108, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30905782

ABSTRACT

AIMS: Parkinson's disease (PD) is a common neurodegenerative disease typically associated with the accumulation of α-synuclein. Autophagy impairment is thought to be involved in the dopaminergic neurodegeneration in PD. We investigate the effect of Apelin-36 on the activated phosphatidylinositol 3-kinase (PI3K)/protein kinase B(Akt)/the mammalian target of rapamycin (mTOR) autophagy pathway in 1-methyl-4-phenylpyridinium (MPP+)-treated SH-SY5Y cells, which is involved in the cytoprotective effect of Apelin-36. MAIN METHODS: SH-SY5Y cells were treated with 1-Methyl-4-phenylpyridine (MPP+) with or without Apelin-36. The cell viability, apoptotic ratio, the form of autophagic vacuoles, the expression of tyrosine hydroxylase (TH), α-synuclein, phosphorylation of PI3K, AKT, mTOR, microtubule-associated protein 1 Light Chain 3 II/I (LC3II/I) and p62 were detected to investigate the neuroprotective effect of Apelin-36. KEY FINDINGS: The results indicate that Apelin-36 significantly improved the cell viability and decreased the apoptosis in MPP+-treated SH-SY5Y cells. The decreased expression of tyrosine hydroxylase (TH) induced by MPP+ was significantly increased by Apelin36 pretreatment. Moreover, Apelin36 significantly increased the autophagic vacuoles. The ratio of LC3II/I was significantly increased by Apelin36, as well as the decreased p62 expression. In addition, the activated PI3K/AKT/mTOR pathway induced by MPP+ was significantly inhibited by Apelin36. Additionally, Apelin36 significantly decreased the α-synuclein expression. Furthermore, the cytoprotective effect of Apelin-36 was weakened by pretreatment with Insulin-like Growth Factor-1 (IGF-1), an activator of PI3K/Akt, and MHY1485, an mTOR activator. SIGNIFICANCE: Our results demonstrated that Apelin-36 protects against MPP+-induced cytotoxicity through PI3K/Akt/mTOR autophagy pathway in PD model in vitro, which provides a new theoretical basis for the treatment of PD.


Subject(s)
1-Methyl-4-phenylpyridinium/adverse effects , Apelin/metabolism , Neuroblastoma/prevention & control , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Apelin/genetics , Apoptosis , Autophagy , Humans , Neuroblastoma/chemically induced , Neuroblastoma/metabolism , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinase/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction , TOR Serine-Threonine Kinases/genetics , Tumor Cells, Cultured
13.
Mol Pharm ; 16(4): 1456-1466, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30821469

ABSTRACT

The macrolide caniferolide A was isolated from extracts of a culture of the marine-derived actinomycete Streptomyces caniferus, and its ability to ameliorate Alzheimer's disease (AD) hallmarks was determined. The compound reduced neuroinflammatory markers in BV2 microglial cells activated with lipopolysaccharide (LPS), being able to block NFκB-p65 translocation to the nucleus and to activate the Nrf2 pathway. It also produced a decrease in pro-inflammatory cytokines (IL-1ß, IL-6, and TNF-α), reactive oxygen species (ROS) and nitric oxide release and inhibited iNOS, JNK, and p38 activities. Moreover, the compound blocked BACE1 activity and attenuated Aß-activation of microglia by drastically diminishing ROS levels. The phosphorylated state of the tau protein was evaluated in SH-SY5Y tau441 cells. Caniferolide A reduced Thr212 and Ser214 phosphorylation by targeting p38 and JNK MAPK kinases. On the other side, the antioxidant properties of the macrolide were determined in an oxidative stress model with SH-SY5Y cells treated with H2O2. The compound diminished ROS levels and increased cell viability and GSH content by activating the nuclear factor Nrf2. Finally, the neuroprotective ability of the compound was confirmed in two trans-well coculture systems with activated BV2 cells (both with LPS and Aß) and wild type and transfected SH-SY5Y cells. The addition of caniferolide A to microglial cells produced a significant increase in the survival of neuroblastoma in both cases. These results indicate that the compound is able to target many pathological markers of AD, suggesting that caniferolide A could be an interesting drug lead for a polypharmacological approach to the illness.


Subject(s)
Amyloid beta-Peptides/metabolism , Inflammation/prevention & control , Macrolides/pharmacology , Neuroblastoma/prevention & control , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Streptomyces/chemistry , tau Proteins/metabolism , Animals , In Vitro Techniques , Inflammation/metabolism , Inflammation/pathology , Macrolides/chemistry , Neuroblastoma/metabolism , Neuroblastoma/pathology , Reactive Oxygen Species/metabolism
15.
Clin Cancer Res ; 25(9): 2915-2924, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30617136

ABSTRACT

PURPOSE: A delay in encountering the cognate antigen while in the circulation, and the suboptimal costimulation received at the tumor site are key reasons for the limited activity of chimeric antigen receptor-redirected T cells (CAR-T) in solid tumors. We have explored the benefits of incorporating the IL15 cytokine within the CAR cassette to provide both a survival signal before antigen encounter, and an additional cytokine signaling at the tumor site using a neuroblastoma tumor model. EXPERIMENTAL DESIGN: We optimized the construct for the CAR specific for the NB-antigen GD2 without (GD2.CAR) or with IL15 (GD2.CAR.15). We then compared the expansion, phenotype, and antitumor activity of T cells transduced with these constructs against an array of neuroblastoma cell lines in vitro and in vivo using a xenogeneic metastatic model of neuroblastoma. RESULTS: We observed that optimized GD2.CAR.15-Ts have reduced expression of the PD-1 receptor, are enriched in stem cell-like cells, and have superior antitumor activity upon repetitive tumor exposures in vitro and in vivo as compared with GD2.CAR-Ts. Tumor rechallenge experiments in vivo further highlighted the role of IL15 in promoting enhanced CAR-T antitumor activity and survival, both in the peripheral blood and tissues. Finally, the inclusion of the inducible caspase-9 gene (iC9) safety switch warranted effective on demand elimination of the engineered GD2.CAR.15-Ts. CONCLUSIONS: Our results guide new therapeutic options for GD2.CAR-Ts in patients with neuroblastoma, and CAR-T development for a broad range of solid tumors.


Subject(s)
Gangliosides/immunology , Immunotherapy/methods , Interleukin-15/immunology , Neuroblastoma/prevention & control , Receptors, Chimeric Antigen/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Apoptosis , Cell Proliferation , Female , Humans , Interleukin-15/genetics , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neuroblastoma/genetics , Neuroblastoma/immunology , Neuroblastoma/pathology , Protein Engineering , Receptors, Chimeric Antigen/genetics , T-Lymphocytes, Cytotoxic/transplantation , Transduction, Genetic , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Cancer Res ; 79(6): 1151-1164, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30541743

ABSTRACT

In neuroblastoma, the interplay between immune cells of the tumor microenvironment and cancer cells contributes to immune escape mechanisms and drug resistance. In this study, we show that natural killer (NK) cell-derived exosomes carrying the tumor suppressor microRNA (miR)-186 exhibit cytotoxicity against MYCN-amplified neuroblastoma cell lines. The cytotoxic potential of these exosomes was partly dependent upon expression of miR-186. miR-186 was downregulated in high-risk neuroblastoma patients, and its low expression represented a poor prognostic factor that directly correlated with NK activation markers (i.e., NKG2D and DNAM-1). Expression of MYCN, AURKA, TGFBR1, and TGFBR2 was directly inhibited by miR-186. Targeted delivery of miR-186 to MYCN-amplified neuroblastoma or NK cells resulted in inhibition of neuroblastoma tumorigenic potential and prevented the TGFß1-dependent inhibition of NK cells. Altogether, these data support the investigation of a miR-186-containing nanoparticle formulation to prevent tumor growth and TGFß1-dependent immune escape in high-risk neuroblastoma patients as well as the inclusion of ex vivo-derived NK exosomes as a potential therapeutic option alongside NK cell-based immunotherapy.Significance: These findings highlight the therapeutic potential of NK cell-derived exosomes containing the tumor suppressor miR-186 that inhibits growth, spreading, and TGFß-dependent immune escape mechanisms in neuroblastoma.


Subject(s)
Exosomes/metabolism , Killer Cells, Natural/immunology , MicroRNAs/genetics , Neuroblastoma/prevention & control , Tumor Microenvironment/immunology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Exosomes/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred NOD , Mice, SCID , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Cancer Res ; 79(4): 830-840, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30584073

ABSTRACT

Amplification of MYCN plays a pivotal role in multiple types of tumors and correlates with poor prognosis in high-risk neuroblastoma. Despite recent advances in the treatment of neuroblastoma, no approaches directly target the master oncogene MYCN. Difficulties in targeting the MYCN protein inspired us to develop a new gene-level-inhibitory strategy using a sequence-specific gene regulator. Here, we generated a MYCN-targeting pyrrole-imidazole (PI) polyamide, MYCN-A3, which directly binds to and alkylates DNA at homing motifs within the MYCN transcript. Pharmacologic suppression of MYCN inhibited the proliferation of cancer cells harboring MYCN amplification compared with MYCN nonamplified cancer cells. In neuroblastoma xenograft mouse models, MYCN-A3 specifically downregulated MYCN expression and suppressed tumor progression with no detectable adverse effects and resulted in prolonged overall survival. Moreover, treatment with MYCN-A3, but not MYCN nontargeting PI polyamide, precipitated a copy number reduction of MYCN in neuroblastoma cells with MYCN amplification. These findings suggest that directly targeting MYCN with MYCN-A3 is a novel therapeutic approach to reduce copy number of the MYCN gene for MYCN-amplified neuroblastoma. SIGNIFICANCE: This study presents a novel approach to drugging an amplified oncogene by showing that targeting gene amplification of MYCN suppresses MYCN expression and neuroblastoma growth.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Gene Amplification/drug effects , Gene Expression Regulation, Neoplastic/drug effects , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Neuroblastoma/prevention & control , Nylons/pharmacology , Alkylation , Animals , Antineoplastic Agents, Alkylating/chemistry , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Female , Humans , Imidazoles/chemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Neuroblastoma/pathology , Nylons/chemistry , Pyrroles/chemistry , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Oxid Med Cell Longev ; 2018: 9595741, 2018.
Article in English | MEDLINE | ID: mdl-30510632

ABSTRACT

Misfolded tau proteins induce accumulation of free radicals and promote neuroinflammation by activating microglia-releasing proinflammatory cytokines, leading to neuronal cell death. Traditional Chinese herbal medicines (CHMs) have been widely used in clinical practice to treat neurodegenerative diseases associated with oxidative stress and neuroinflammation. This study examined the neuroprotection effects of formulated CHMs Bai-Shao (made of Paeonia lactiflora), Gan-Cao (made of Glycyrrhiza uralensis), and Shaoyao Gancao Tang (SG-Tang, made of P. lactiflora and G. uralensis at 1 : 1 ratio) in cell model of tauopathy. Our results showed that SG-Tang displayed a greater antioxidative and antiaggregation effect than Bai-Shao and Gan-Cao and a stronger anti-inflammatory activity than Bai-Shao but similar to Gan-Cao. In inducible 293/SH-SY5Y cells expressing proaggregant human tau repeat domain (ΔK280 tauRD), SG-Tang reduced tau misfolding and reactive oxygen species (ROS) level in ΔK280 tauRD 293 cells and promoted neurite outgrowth in ΔK280 tauRD SH-SY5Y cells. Furthermore, SG-Tang displayed anti-inflammatory effects by reducing nitric oxide (NO) production in mouse BV-2 microglia and increased cell viability of ΔK280 tauRD-expressing SH-SY5Y cells inflamed by BV-2 conditioned medium. To uncover the neuroprotective mechanisms of SG-Tang, apoptosis protein array analysis of inflamed tau expressing SH-SY5Y cells was conducted and the suppression of proapoptotic proteins was confirmed. In conclusion, SG-Tang displays neuroprotection by exerting antioxidative and anti-inflammatory activities to suppress neuronal apoptosis in human tau cell models. The study results lay the base for future applications of SG-Tang on tau animal models to validate its effect of reducing tau misfolding and potential disease modification.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Drug Compounding , Drugs, Chinese Herbal/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Tauopathies/prevention & control , Animals , Apoptosis/drug effects , Cells, Cultured , HEK293 Cells , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neuroblastoma/prevention & control , Neurons/metabolism , Neurons/pathology , Oxidation-Reduction , Tauopathies/metabolism , Tauopathies/pathology
19.
Mutat Res Genet Toxicol Environ Mutagen ; 836(Pt B): 62-71, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30442347

ABSTRACT

In this work, we aim to provide evidence for the protective effect of a copper chelator, neocuproine (NeoCu), against the oxidative stress in NSC34 cells, which inhibits biomolecule oxidation and cell death. Results obtained with the comet assay allowed to determine the increase in oxidized purines and pyrimidines by H2O2 exposure, and their changes after the addition of NeoCu. We also observed a higher ATP7b activity in nuclei and a higher Cu concentration inside the cells, proving that the NeoCu acts directly in DNA to promote cell recovery in oxidative stress conditions, also observed in Reactive Oxygen Species (ROS) detection assay by Flow Cytometry. Based on these results, we propose that NeoCu is a promising drug for the protection of motor neuron cells during oxidative stress caused by neurodegenerative diseases in this system.


Subject(s)
Chelating Agents/pharmacology , DNA Damage , Hydrogen Peroxide/toxicity , Neuroblastoma/prevention & control , Oxidative Stress/drug effects , Phenanthrolines/pharmacology , Protective Agents/pharmacology , Copper/metabolism , Copper-Transporting ATPases/metabolism , Humans , Neuroblastoma/pathology , Oxidants/toxicity , Reactive Oxygen Species/metabolism , Tumor Cells, Cultured
20.
Sci Rep ; 8(1): 6379, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686306

ABSTRACT

Necroptosis is an essential pathophysiological process in cerebral ischemia-related diseases. Therefore, targeting necroptosis may prevent cell death and provide a much-needed therapy. Ansiomycin is an inhibitor of protein synthesis which can also activate c-Jun N-terminal kinases. The present study demonstrated that anisomycin attenuated necroptosis by upregulating CHIP (carboxyl terminus of Hsc70-interacting protein) leading to the reduced levels of receptor-interacting protein kinase 1 (RIPK1) and receptor-interacting protein kinase 3 (RIPK3) proteins in two in vitro models of cerebral ischemia. Further exploration in this research revealed that losing neither the co-chaperone nor the ubiquitin E3 ligase function of CHIP could abolish its ability to reduce necroptosis. Collectively, this study identifies a novel means of preventing necroptosis in two in vitro models of cerebral ischemia injury through activating the expression of CHIP, and it may provide a potential target for the further study of the disease.


Subject(s)
Anisomycin/pharmacology , Apoptosis , Glucose/deficiency , Necrosis , Neuroblastoma/prevention & control , Ubiquitin-Protein Ligases/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Cell Hypoxia , Female , Gene Expression Regulation, Enzymologic , Mice , Neuroblastoma/etiology , Neuroblastoma/pathology , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...