Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 230
Filter
Add more filters

Publication year range
1.
Int J Cancer ; 153(5): 1026-1034, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37246577

ABSTRACT

Children with relapsed/refractory (R/R) neuroblastoma (NB) and medulloblastoma (MB) have poor outcomes. We evaluated the efficacy of nifurtimox (Nfx) in a clinical trial for children with R/R NB and MB. Subjects were divided into three strata: first relapse NB, multiply R/R NB, and R/R MB. All patients received Nfx (30 mg/kg/day divided TID daily), Topotecan (0.75 mg/m2 /dose, days 1-5) and Cyclophosphamide (250 mg/m2 /dose, days 1-5) every 3 weeks. Response was assessed after every two courses using International Neuroblastoma Response Criteria and Response Evaluation Criteria in Solid Tumors (RECIST) criteria. One hundred and twelve eligible patients were enrolled with 110 evaluable for safety and 76 evaluable for response. In stratum 1, there was a 53.9% response rate (CR + PR), and a 69.3% total benefit rate (CR + PR + SD), with an average time on therapy of 165.2 days. In stratum 2, there was a 16.3% response rate, and a 72.1% total benefit rate, and an average time on study of 158.4 days. In stratum 3, there was a 20% response rate and a 65% total benefit rate, an average time on therapy of 105.0 days. The most common side effects included bone marrow suppression and reversible neurologic complications. The combination of Nfx, topotecan and cyclophosphamide was tolerated, and the objective response rate plus SD of 69.8% in these heavily pretreated populations suggests that this combination is an effective option for patients with R/R NB and MB. Although few objective responses were observed, the high percentage of stabilization of disease and prolonged response rate in patients with multiply relapsed disease shows this combination therapy warrants further testing.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Neuroblastoma , Child , Humans , Topotecan/adverse effects , Nifurtimox/therapeutic use , Medulloblastoma/drug therapy , Neoplasm Recurrence, Local/pathology , Neuroblastoma/drug therapy , Neuroblastoma/etiology , Cyclophosphamide , Antineoplastic Combined Chemotherapy Protocols/adverse effects
2.
Pediatr Blood Cancer ; 70(1): e29981, 2023 01.
Article in English | MEDLINE | ID: mdl-36129239

ABSTRACT

BACKGROUND: The purpose of this study is to analyze renal function outcomes in abdominal neuroblastoma patients undergoing proton therapy (PT). PROCEDURE: From 2011 to 2019, two single-institution Institutional Review Board-approved protocols prospectively enrolled neuroblastoma patients for data collection. To assess renal function, serum creatinine (Cr), blood urea nitrogen (BUN), and creatinine clearance (CrCl) before proton therapy (pre-PT) were compared with the values at last follow-up. RESULTS: A total of 30 children with abdominal neuroblastoma with median age 3.5 years (range, 0.9-9.1) at time of PT were included in this study. All patients underwent chemotherapy and resection of primary tumor prior to PT. Two patients required radical nephrectomy. Median follow-up after PT was 35 months. Mean dose to ipsilateral and contralateral kidney was 13.9 and 5.4 Gy, respectively. No patients developed hypertension or renal dysfunction during follow-up. There was no statistically significant change in serum BUN (p = .508), CrCl (p = .280), or eGFR (p = .246) between pre-PT and last follow-up. CONCLUSION: At a median follow-up of almost 3 years, renal toxicity was uncommon after PT. Longer follow-up and larger patient cohort data are needed to further assess impact of PT on renal function in this population.


Subject(s)
Neuroblastoma , Proton Therapy , Child , Humans , Child, Preschool , Protons , Nephrectomy , Neuroblastoma/radiotherapy , Neuroblastoma/etiology , Kidney/physiology , Proton Therapy/adverse effects , Follow-Up Studies
3.
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
4.
BMC Cancer ; 21(1): 393, 2021 Apr 10.
Article in English | MEDLINE | ID: mdl-33838662

ABSTRACT

BACKGROUND: Majority of neuroblastoma patients develop metastatic disease at diagnosis and their prognosis is poor with current therapeutic approach. Major challenges are how to tackle the mechanisms responsible for tumorigenesis and metastasis. Human mesenchymal stem cells (hMSCs) may be actively involved in the constitution of cancer microenvironment. METHODS: An orthotopic neuroblastoma murine model was utilized to mimic the clinical scenario. Human neuroblastoma cell line SK-N-LP was transfected with luciferase gene, which were inoculated with/without hMSCs into the adrenal area of SCID-beige mice. The growth and metastasis of neuroblastoma was observed by using Xenogen IVIS 100 in vivo imaging and evaluating gross tumors ex vivo. The homing of hMSCs towards tumor was analyzed by tracing fluorescence signal tagged on hMSCs using CRI Maestro™ imaging system. RESULTS: hMSCs mixed with neuroblastoma cells significantly accelerated tumor growth and apparently enhanced metastasis of neuroblastoma in vivo. hMSCs could be recruited by primary tumor and also become part of the tumor microenvironment in the metastatic lesion. The metastatic potential was consistently reduced in lung and tumor when hMSCs were pre-treated with stromal cell derived factor-1 (SDF-1) blocker, AMD3100, suggesting that the SDF-1/CXCR4 axis was one of the prime movers in the metastatic process. CONCLUSIONS: hMSCs accelerated and facilitated tumor formation, growth and metastasis. Furthermore, the homing propensity of hMSCs towards both primary tumor and metastatic loci can also provide new therapeutic insights in utilizing bio-engineered hMSCs as vehicles for targeted anti-cancer therapy.


Subject(s)
Cell Communication , Mesenchymal Stem Cells/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Expression , Genes, Reporter , Heterografts , Humans , Mesenchymal Stem Cell Transplantation , Mice , Mice, SCID , Neoplastic Processes , Neuroblastoma/etiology , Receptors, CXCR4/metabolism , Tumor Burden , Tumor Microenvironment
5.
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
6.
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
7.
Curr Treat Options Oncol ; 22(11): 102, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580780

ABSTRACT

OPINION STATEMENT: Neuroblastoma (NB) is a heterogeneous solid tumor of the pediatric population that originates from neural crest cells and affects the developing sympathetic nervous system. It is the most common neuroblastic tumor accounting for approximately 10% of all childhood cancers and 10-15% of pediatric tumor mortalities. The outcomes range from spontaneous tumor regression in low-risk groups to metastasis and death even after multimodal therapy in high-risk groups. Hence, the detection of NB at an early stage improves outcomes and provides a better prognosis for patients. Early detection and prognosis of NB depend on specific molecules termed biomarkers which can be tissue-specific or circulating. Certain biomarkers are employed in the classification of NB into different groups to improve the treatment and prognosis, and others can be used as therapeutic targets. Therefore, novel biomarker discovery is essential for the early detection of NB, predicting the course of the disease, and developing new targeted treatment strategies. In this review, we aim to summarize the literature pertinent to some important biomarkers of NB and discuss the prognostic role of these biomarkers as well as their potential role in targeted therapy.


Subject(s)
Biomarkers, Tumor , Neuroblastoma/diagnosis , Disease Management , Disease Susceptibility , Humans , Molecular Diagnostic Techniques , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Neuroblastoma/etiology , Neuroblastoma/mortality , Neuroblastoma/therapy , Prognosis , Treatment Outcome
8.
J Toxicol Environ Health A ; 84(20): 846-857, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34196262

ABSTRACT

The aim of this study was to examine the potential effects of long-term evolution (LTE) radiofrequency electromagnetic fields (RF-EMF) on cell proliferation using SH-SY5Y neuronal cells. The growth rate and proliferation of SH-SY5Y cells were significantly decreased upon exposure to 1760 MHz RF-EMF at 4 W/kg specific absorption rate (SAR) for 4 hr/day for 4 days. Cell cycle analysis indicated that the cell cycle was delayed in the G0/G1 phase after RF-EMF exposure. However, DNA damage or apoptosis was not involved in the reduced cellular proliferation following RF-EMF exposure because the expression levels of histone H2A.X at Ser139 (γH2AX) were not markedly altered and the apoptotic pathway was not activated. However, SH-SY5Y cells exposed to RF-EMF exhibited a significant elevation in Akt and mTOR phosphorylation levels. In addition, the total amount of p53 and phosphorylated-p53 was significantly increased. Data suggested that Akt/mTOR-mediated cellular senescence led to p53 activation via stimulation of the mTOR pathway in SH-SY5Y cells. The transcriptional activation of p53 led to a rise in expression of cyclin-dependent kinase (CDK) inhibitors p21 and p27. Further, subsequent inhibition of CDK2 and CDK4 produced a fall in phosphorylated retinoblastoma (pRb at Ser807/811), which decreased cell proliferation. Taken together, these data suggest that exposure to RF-EMF might induce Akt/mTOR-mediated cellular senescence, which may delay the cell cycle without triggering DNA damage in SH-SY5Y neuroblastoma cells.


Subject(s)
Cell Proliferation/radiation effects , Cellular Senescence/radiation effects , Electromagnetic Fields/adverse effects , Neuroblastoma/physiopathology , Radio Waves/adverse effects , Cellular Senescence/genetics , Humans , Neuroblastoma/etiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
9.
Proc Natl Acad Sci U S A ; 115(31): E7351-E7360, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30021854

ABSTRACT

Neuroblastoma is a neural crest-derived childhood tumor of the peripheral nervous system in which MycN amplification is a hallmark of poor prognosis. Here we show that MycN is expressed together with phosphorylation-stabilizing factor CIP2A in regions of the neural plate destined to form the CNS, but MycN is excluded from the neighboring neural crest stem cell domain. Interestingly, ectopic expression of MycN or CIP2A in the neural crest domain biases cells toward CNS-like neural stem cells that express Sox2. Consistent with this, some forms of neuroblastoma have been shown to share transcriptional resemblance with CNS neural stem cells. As high MycN/CIP2A levels correlate with poor prognosis, we posit that a MycN/CIP2A-mediated cell-fate bias may reflect a possible mechanism underlying early priming of some aggressive forms of neuroblastoma. In contrast to MycN, its paralogue cMyc is normally expressed in the neural crest stem cell domain and typically is associated with better overall survival in clinical neuroblastoma, perhaps reflecting a more "normal" neural crest-like state. These data suggest that priming for some forms of aggressive neuroblastoma may occur before neural crest emigration from the CNS and well before sympathoadrenal specification.


Subject(s)
Autoantigens/physiology , Membrane Proteins/physiology , N-Myc Proto-Oncogene Protein/physiology , Neural Crest/cytology , Neural Stem Cells/physiology , Neuroblastoma/etiology , Autoantigens/analysis , Cell Movement , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/analysis , N-Myc Proto-Oncogene Protein/analysis , Neuroblastoma/pathology , SOXB1 Transcription Factors/analysis
10.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768913

ABSTRACT

Aggregation of α-synuclein (α-Syn) is implicated in the pathogenesis of Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Therefore, the removal of α-Syn aggregation could lead to the development of many new therapeutic agents for neurodegenerative diseases. In the present study, we succeeded in generating a new α-Syn stably expressing cell line using a piggyBac transposon system to investigate the neuroprotective effect of the flavonoid kaempferol on α-Syn toxicity. We found that kaempferol provided significant protection against α-Syn-related neurotoxicity. Furthermore, kaempferol induced autophagy through an increase in the biogenesis of lysosomes by inducing the expression of transcription factor EB and reducing the accumulation of α-Syn; thus, kaempferol prevented neuronal cell death. Moreover, kaempferol directly blocked the amyloid fibril formation of α-Syn. These results support the therapeutic potential of kaempferol in diseases such as synucleinopathies that are characterized by α-Syn aggregates.


Subject(s)
Amyloid/drug effects , Autophagy , Kaempferols/pharmacology , Neuroblastoma/drug therapy , Neurotoxicity Syndromes/drug therapy , Protective Agents/pharmacology , alpha-Synuclein/toxicity , Amyloid/metabolism , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Mice , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurotoxicity Syndromes/etiology , Neurotoxicity Syndromes/metabolism , Neurotoxicity Syndromes/pathology
11.
Molecules ; 27(1)2021 Dec 24.
Article in English | MEDLINE | ID: mdl-35011335

ABSTRACT

Neuroblastoma (NB) is a type of peripheral sympathetic nervous system cancer that most commonly affects children. It is caused by the improper differentiation of primitive neural crest cells during embryonic development. Although NB occurs for 8% of paediatric cancers, it accounts for 15% of cancer-related deaths. Despite a considerable increase in cytotoxic chemo- and radiotherapy, patients in advanced stages remain virtually incurable. Therefore, there is a desperate necessity for new treatment strategies to be investigated. Accumulating evidence suggested that microRNAs (miRNAs) are a class of non-coding RNAs with 19-25 nucleotides lengths and play a central role in the development of NB carcinogenesis. Fascinatingly, miRNA inhibitors have an antisense property that can inhibit miRNA function and suppress the activity of mature miRNA. However, many studies have addressed miRNA inhibition in the treatment of NB, but their molecular mechanisms and signalling pathways are yet to be analysed. In this study, we impart the current state of knowledge about the role of miRNA inhibition in the aetiology of NB.


Subject(s)
Disease Susceptibility , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neuroblastoma/etiology , Biomarkers, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , RNA Interference , RNA, Messenger/genetics
12.
Genes Chromosomes Cancer ; 59(4): 240-248, 2020 04.
Article in English | MEDLINE | ID: mdl-31749253

ABSTRACT

Whereas neuroblastoma is the most common extracranial solid tumor of childhood, less than 5% of cases occur in adults. Pediatric neuroblastoma shows marked heterogeneity of histology and molecular biology. Information about this tumor in adults is limited, especially regarding molecular biology. We report a series of nine neuroblastoma cases diagnosed in adulthood (18 to 40 years old) with molecular biologic characterization in seven. All tumors were Schwannian stroma-poor, and mostly poorly differentiated. Tumors expressed neural markers including PHOX2B, NB84, synaptophysin, chromogranin, CD56, neuron-specific enolase, and PGP9.5. Two out of six cases expressed ALK and one had the F1174 L mutation reported in childhood neuroblastoma. Fluorescent in situ hybridization (FISH) revealed MYCN amplification in 2/7 cases, chromosome 1p deletion in 1/5 cases and 17q gain in 4/4 cases. One in five cases showed loss of ATRX expression by immunohistochemistry and alternate lengthening of telomeres by FISH. Zero out of five cases showed rearrangement of the TERT gene by FISH, but one case showed high level amplification. In conclusion, the morphology and immunophenotype of adult-onset neuroblastoma are similar to pediatric cases although less differentiated than some childhood tumors. Similarly, molecular genetic alterations in adult-onset neuroblastoma are not unique to this age group. However, 80% of cases tested showed genetic changes that would promote maintenance of telomeres, which is a molecular marker of high risk cases. This may help explain the poor response in adults to pediatric treatment protocols. Additional studies to characterize the biology of this tumor in the adult age group will facilitate the design of more personalized therapeutic approaches.


Subject(s)
Neuroblastoma/diagnosis , Neuroblastoma/etiology , Adolescent , Adult , Age of Onset , Biomarkers, Tumor , Disease Susceptibility , Female , Gene Dosage , Gene Rearrangement , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Neuroblastoma/metabolism , Telomere Homeostasis , Young Adult
13.
J Cell Mol Med ; 24(2): 1160-1168, 2020 01.
Article in English | MEDLINE | ID: mdl-31830377

ABSTRACT

Neuroblastoma (NB), a neuroendocrine tumour, is one of the most prevalent cancers in children. The link between LMO1 polymorphisms and NB has been investigated by several groups, rendering inconclusive results. Here, with this comprehensive systematic review and up-to-date meta-analysis, we aim to distinctively elucidate the possible correlation between LMO1 polymorphisms and NB susceptibility. Eligible studies were systematically researched and identified using PubMed, Web of Science and Scopus databases up to 10 February 2019. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the strength of the associations. Our findings revealed that rs110419 and rs2168101 polymorphisms were significantly associated with a decreased risk of NB in all genetic models. In addition, the rs4758051 variant appeared protective against NB in homozygous, dominant and allele genetic models, whereas the rs10840002 variant markedly decreased the risk of NB in the allele model. In contrast, the rs204938 polymorphism showed a positive association with NB susceptibility in allele genetic models. In summary, our meta-analysis is the first to provide clear evidence of an association between specific polymorphisms of LMO1 and susceptibility to NB. Of note, additional larger well-designed studies would be helpful to further evaluate and confirm this association.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , LIM Domain Proteins/genetics , Neuroblastoma/etiology , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Case-Control Studies , DNA-Binding Proteins/metabolism , Genome-Wide Association Study , Humans , LIM Domain Proteins/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Risk Factors , Transcription Factors/metabolism
14.
J Biol Chem ; 294(26): 10336-10348, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31113867

ABSTRACT

Parkinson's disease (PD) is a major human disease associated with degeneration of the central nervous system. Evidence suggests that several endogenously formed 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-mimicking chemicals that are metabolic conversion products, especially ß-carbolines and isoquinolines, act as neurotoxins that induce PD or enhance progression of the disease. We have demonstrated previously that mitochondrially targeted human cytochrome P450 2D6 (CYP2D6), supported by mitochondrial adrenodoxin and adrenodoxin reductase, can efficiently catalyze the conversion of MPTP to the toxic 1-methyl-4-phenylpyridinium ion. In this study, we show that the mitochondrially targeted CYP2D6 can efficiently catalyze MPTP-mimicking compounds, i.e. 2-methyl-1,2,3,4-tetrahydroisoquinoline, 2-methyl-1,2,3,4-tetrahydro-ß-carboline, and 9-methyl-norharmon, suspected to induce PD in humans. Our results reveal that activity and respiration in mouse brain mitochondrial complex I are significantly affected by these toxins in WT mice but remain unchanged in Cyp2d6 locus knockout mice, indicating a possible role of CYP2D6 in the metabolism of these compounds both in vivo and in vitro These metabolic effects were minimized in the presence of two CYP2D6 inhibitors, quinidine and ajmalicine. Neuro-2a cells stably expressing predominantly mitochondrially targeted CYP2D6 were more sensitive to toxin-mediated respiratory dysfunction and complex I inhibition than cells expressing predominantly endoplasmic reticulum-targeted CYP2D6. Exposure to these toxins also induced the autophagic marker Parkin and the mitochondrial fission marker Dynamin-related protein 1 (Drp1) in differentiated neurons expressing mitochondrial CYP2D6. Our results show that monomethylamines are converted to their toxic cationic form by mitochondrially directed CYP2D6 and result in neuronal degradation in mice.


Subject(s)
Cytochrome P-450 CYP2D6/physiology , Disease Models, Animal , Methylamines/toxicity , Mitochondria/pathology , Neuroblastoma/pathology , Neurons/pathology , Parkinson Disease/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , Animals , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/etiology , Neuroblastoma/metabolism , Neurons/drug effects , Neurons/metabolism , Neurotoxins/toxicity , Parkinson Disease/etiology , Parkinson Disease/metabolism , Tumor Cells, Cultured
15.
Int J Cancer ; 146(2): 553-565, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31173338

ABSTRACT

Tumors are complex networks of constantly interacting elements: tumor cells, stromal cells, immune and stem cells, blood/lympathic vessels, nerve fibers and extracellular matrix components. These elements can influence their microenvironment through mechanical and physical signals to promote tumor cell growth. To get a better understanding of tumor biology, cooperation between multidisciplinary fields is needed. Diverse mathematic computations and algorithms have been designed to find prognostic targets and enhance diagnostic assessment. In this work, we use computational digital tools to study the topology of vitronectin, a glycoprotein of the extracellular matrix. Vitronectin is linked to angiogenesis and migration, two processes closely related to tumor cell spread. Here, we investigate whether the distribution of this molecule in the tumor stroma may confer mechanical properties affecting neuroblastoma aggressiveness. Combining image analysis and graph theory, we analyze different topological features that capture the organizational cues of vitronectin in histopathological images taken from human samples. We find that the Euler number and the branching of territorial vitronectin, two topological features, could allow for a more precise pretreatment risk stratification to guide treatment strategies in neuroblastoma patients. A large amount of recently synthesized VN would create migration tracks, pinpointed by both topological features, for malignant neuroblasts, so that dramatic change in the extracellular matrix would increase tumor aggressiveness and worsen patient outcomes.


Subject(s)
Neuroblastoma/etiology , Neuroblastoma/genetics , Vitronectin/genetics , Algorithms , Cell Proliferation/genetics , Extracellular Matrix/genetics , Extracellular Matrix/pathology , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neuroblastoma/pathology , Prognosis , Risk , Stromal Cells/pathology , Tumor Microenvironment/genetics
16.
Int J Mol Sci ; 21(20)2020 Oct 15.
Article in English | MEDLINE | ID: mdl-33076337

ABSTRACT

Bcl-xL, a member of the Bcl-2 family, is a pro-survival protein involved in apoptosis regulation. We have previously reported the ability of Bcl-xL to form various types of fibers, from native to amyloid conformations. Here, we have mimicked the effect of apoptosis-induced caspase activity on Bcl-xL by limited proteolysis using trypsin. We show that cleaved Bcl-xL (ΔN-Bcl-xL) forms fibers that exhibit the features of amyloid structures (BclxLcf37). Moreover, three monoclonal antibodies (mAbs), produced by mouse immunization and directed against ΔN-Bcl-xL or Bcl-xL fibers, were selected and characterized. Our results show that these mAbs specifically target ΔN-Bcl-xL in amyloid fibers in vitro. Upon metal-stress-induced apoptosis, these mAbs are able to detect the presence of Bcl-xL in amyloid aggregates in neuroblastoma SH-SY5Y cell lines. In conclusion, these specific mAbs directed against amyloidogenic conformations of Bcl-xL constitute promising tools for studying, in vitro and in cellulo, the contribution of Bcl-xL in apoptosis. These mAbs may further help in developing new diagnostics and therapies, considering Bcl-xL as a strategic target for treating brain lesions relevant to stroke and neurodegenerative diseases.


Subject(s)
Amyloid/immunology , Antibodies, Monoclonal/immunology , Neuroblastoma/metabolism , bcl-X Protein/immunology , Amyloid/chemistry , Animals , Apoptosis , Cell Line, Tumor , Humans , Metals, Heavy/toxicity , Mice , Neuroblastoma/etiology , Oxidants/toxicity , Protein Conformation , bcl-X Protein/chemistry
17.
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
18.
Int J Mol Sci ; 21(12)2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32585871

ABSTRACT

The movement disorder Parkinson's disease (PD) is the second most frequently diagnosed neurodegenerative disease, and is associated with aging, the environment, and genetic factors. The intracellular aggregation of α-synuclein and the loss of dopaminergic neurons in the substantia nigra pars compacta are the pathological hallmark of PD. At present, there is no successful treatment for PD. Maackiain (MK) is a flavonoid extracted from dried roots of Sophora flavescens Aiton. MK has emerged as a novel agent for PD treatment that acts by inhibiting monoamine oxidase B. In this study, we assessed the neuroprotective potential of MK in Caenorhabditis elegans and investigated possible mechanism of this neuroprotection in the human SH-SY5Y cell line. We found that MK significantly reduced dopaminergic neuron damage in 6-hydroxydopamine (6-OHDA)-exposed worms of the BZ555 strain, with corresponding improvements in food-sensing behavior and life-span. In transgenic worms of strain NL5901 treated with 0.25 mM MK, the accumulation of α-synuclein was diminished by 27% (p < 0.01) compared with that in untreated worms. Moreover, in worms and the SH-SY5Y cell line, we confirmed that the mechanism of MK-mediated protection against PD pathology may include blocking apoptosis, enhancing the ubiquitin-proteasome system, and augmenting autophagy by increasing PINK1/parkin expression. The use of small interfering RNA to downregulate parkin expression in vivo and in vitro could reverse the benefits of MK in PD models. MK may have considerable therapeutic applications in PD.


Subject(s)
Caenorhabditis elegans/drug effects , Neuroblastoma/drug therapy , Oxidopamine/toxicity , Parkinson Disease/drug therapy , Protein Kinases/metabolism , Pterocarpans/pharmacology , Ubiquitin-Protein Ligases/metabolism , alpha-Synuclein/toxicity , Adrenergic Agents/toxicity , Animals , Apoptosis , Autophagy , Caenorhabditis elegans/growth & development , Disease Models, Animal , Gene Expression Regulation/drug effects , Humans , Neuroblastoma/etiology , Neuroblastoma/pathology , Parkinson Disease/etiology , Parkinson Disease/pathology , Protein Kinases/genetics , Ubiquitin-Protein Ligases/genetics
19.
Molecules ; 25(5)2020 Mar 06.
Article in English | MEDLINE | ID: mdl-32155795

ABSTRACT

Neuroblastoma (NB) is the most frequent extracranial pediatric tumor. Despite the current available multiple therapeutic options, the prognosis for high-risk NB patients remains unsatisfactory and makes the disease a clear unmet medical need. Thus, more tailored therapeutic approaches are warranted to improve both the quality of life and the survival of the patients. Macrophage migration inhibitory factor (MIF) is a pleiotropic cytokine that plays a key role in several diseases, including cancer. Preclinical and clinical studies in NB patients convergently indicate that MIF exerts pro-tumorigenic properties in NB. MIF is upregulated in NB tumor tissues and cell lines and it contributes to NB aggressiveness and immune-escape. To date, there are only a few data about the role of the second member of the MIF family, the MIF homolog d-dopachrome tautomerase (DDT), in NB. Here, we review the preclinical and clinical studies on the role of the MIF family of cytokines in NB and suggest that MIF and possibly DDT inhibitors may be promising novel prognostic and therapeutic targets in NB management.


Subject(s)
Cytokines/genetics , Cytokines/metabolism , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Multigene Family , Neuroblastoma/etiology , Neuroblastoma/metabolism , Animals , Biomarkers, Tumor , Cytokines/antagonists & inhibitors , Disease Management , Disease Susceptibility , Drug Discovery , Drug Repositioning , Humans , Intramolecular Oxidoreductases/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Molecular Targeted Therapy , Neuroblastoma/drug therapy , Neuroblastoma/pathology
20.
Genesis ; 57(10): e23323, 2019 10.
Article in English | MEDLINE | ID: mdl-31218818

ABSTRACT

Neuroblastoma, an embryonal tumor arising from the sympathetic ganglia and adrenal medulla, is among the most intractable pediatric cancers. Although a variety of genetic changes have been identified in neuroblastoma, how they contribute to its pathogenesis remains largely unclear. Recent studies have identified alterations of the anaplastic lymphoma kinase (ALK) gene in neuroblastoma; ALK F1174L (a phenylalanine-to-leucine substitution at codon 1174) represents one of the most frequent of these somatic mutations, and is associated with amplification of the MYCN gene, the most reliable marker for the poor survival. We engineered the mouse Alk locus so that ALK F1174L is expressed by its endogenous promoter and can be induced in a spatiotemporally controlled fashion using Cre-loxP system. Although expression of ALK F1174L resulted in enhanced proliferation of sympathetic ganglion progenitors and increased the size of the sympathetic ganglia, it was insufficient to cause neuroblastoma. However, lethal neuroblastoma frequently developed in mice co-expressing ALK F1174L and MYCN, even in a genetic background where MYCN alone does not cause overt tumors. These data reveal that physiological expression of ALK F1174L significantly potentiates the oncogenic ability of MYCN in vivo. Our conditional mutant mice provide a valuable platform for investigating the pathogenesis of neuroblastoma.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Neoplasms, Experimental/genetics , Neuroblastoma/etiology , Animals , Carcinogenesis/genetics , Female , Ganglia, Sympathetic/growth & development , Genetic Engineering , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains/genetics , Mutagenesis, Insertional , N-Myc Proto-Oncogene Protein/biosynthesis , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/etiology , Neuroblastoma/enzymology , Neuroblastoma/genetics
SELECTION OF CITATIONS
SEARCH DETAIL