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

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(21): e2202016119, 2022 05 24.
Article in English | MEDLINE | ID: mdl-35537042

ABSTRACT

Autophagy defects are a risk factor for inflammatory bowel diseases (IBDs) through unknown mechanisms. Whole-body conditional deletion of autophagy-related gene (Atg) Atg7 in adult mice (Atg7Δ/Δ) causes tissue damage and death within 3 mo due to neurodegeneration without substantial effect on intestine. In contrast, we report here that whole-body conditional deletion of other essential Atg genes Atg5 or Fip200/Atg17 in adult mice (Atg5Δ/Δ or Fip200Δ/Δ) caused death within 5 d due to rapid autophagy inhibition, elimination of ileum stem cells, and loss of barrier function. Atg5Δ/Δ mice lost PDGFRα+ mesenchymal cells (PMCs) and Wnt signaling essential for stem cell renewal, which were partially rescued by exogenous Wnt. Matrix-assisted laser desorption ionization coupled to mass spectrometry imaging (MALDI-MSI) of Atg5Δ/Δ ileum revealed depletion of aspartate and nucleotides, consistent with metabolic insufficiency underlying PMC loss. The difference in the autophagy gene knockout phenotypes is likely due to distinct kinetics of autophagy loss, as deletion of Atg5 more gradually extended lifespan phenocopying deletion of Atg7 or Atg12. Thus, autophagy is required for PMC metabolism and ileum stem cell and mammalian survival. Failure to maintain PMCs through autophagy may therefore contribute to IBD.


Subject(s)
Autophagy , Intestines , Receptor, Platelet-Derived Growth Factor alpha , Stem Cells , Animals , Autophagy/genetics , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Autophagy-Related Proteins , Cell Survival , Mice , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Stem Cells/metabolism
2.
BMC Public Health ; 22(1): 297, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35164721

ABSTRACT

BACKGROUND: This study assessed the feasibility and effect of two mobile modes (WhatsApp vs. a specially designed app) in their delivery of updates and assignments to parents. METHODS: Two three-armed, randomized, controlled feasibility studies were conducted. In each trial, four schools with a total of 418 students in grade 5th, mean age 10.1 years, were randomly allocated to the control arm, youth-only arm, or youth & parental component arm. Only the data of those that completed all three assessments (pre, post and 3 months post program) were analyzed: 133 in the first trial and 137 in the second. In the youth-only arm, students participated in an interactive age-tailored prevention program delivered in 10 weekly, 90-min sessions on self-care behaviors, media literacy, self-esteem, and positive body image. The control groups in both studies received three health- and nutrition-related sessions. In the parental arm, in addition to the 'Favoring-Myself-Young's ten sessions program, parents received updates and were requested to complete shared assignments with their children. In the first year, the assignments were sent via WhatsApp, and in the following year via "Favoring Myself" smartphone application. Facilitators were third year undergraduate students. They used a detailed semi-structured guide and received 4-weekly hours of didactic and group dynamic supervision. Mixed-methods assessments were performed using semi-structured interviews with ten parents and five school staff members each year, as well as a computerized self-report questionnaire. RESULTS: Feasibility of parent-adolescent shared assignments in both digital modes was lower than expected. The use of WhatsApp had higher feasibility and uptake than the use of the special application. The addition of the concurrent parental component via WhatsApp was associated with superior improvement in self-esteem and identification of advertisement strategies, compared with the youth-only program. However, adolescents in the youth-only program delivered via the smartphone application demonstrated superior improvement compared to those in the youth and parental component arm. CONCLUSIONS: Although the addition of the concurrent parenting component was praised by the actively participating parents, overall, under the chosen structure and population, it did not prove to add statistically significant value to the youth-only arm. TRIAL REGISTRATIONS: NCT03216018 (12.7.2017) and NCT03540277 (26.4.2018).


Subject(s)
Health Promotion , Parents , Adolescent , Child , Feasibility Studies , Health Promotion/methods , Humans , Parenting , Parents/education , Technology
3.
J Nanobiotechnology ; 14(1): 37, 2016 May 14.
Article in English | MEDLINE | ID: mdl-27179923

ABSTRACT

BACKGROUND: The ability to direct and manipulate neuronal cells has important potential in therapeutics and neural network studies. An emerging approach for remotely guiding cells is by incorporating magnetic nanoparticles (MNPs) into cells and transferring the cells into magnetic sensitive units. Recent developments offer exciting possibilities of magnetic manipulations of MNPs-loaded cells by external magnetic fields. In the present study, we evaluated and characterized uptake properties for optimal loading of cells by MNPs. We examined the interactions between MNPs of different cores and coatings, with primary neurons and neuron-like cells. RESULTS: We found that uncoated-maghemite iron oxide nanoparticles maximally interact and penetrate into cells with no cytotoxic effect. We observed that the cellular uptake of the MNPs depends on the time of incubation and the concentration of nanoparticles in the medium. The morphology patterns of the neuronal cells were not affected by MNPs uptake and neurons remained electrically active. We theoretically modeled magnetic fluxes and demonstrated experimentally the response of MNP-loaded cells to the magnetic fields affecting cell motility. Furthermore, we successfully directed neurite growth orientation along regeneration. CONCLUSIONS: Applying mechanical forces via magnetic mediators is a useful approach for biomedical applications. We have examined several types of MNPs and studied the uptake behavior optimized for magnetic neuronal manipulations.


Subject(s)
Ferric Compounds/pharmacokinetics , Magnetite Nanoparticles/analysis , Neurons/cytology , Animals , Cell Movement/drug effects , Cell Survival/drug effects , Ferric Compounds/analysis , Ferric Compounds/therapeutic use , Ferric Compounds/toxicity , Magnetic Fields , Magnetics/methods , Magnetite Nanoparticles/therapeutic use , Magnetite Nanoparticles/toxicity , Micromanipulation/methods , Nerve Regeneration/drug effects , Neurons/drug effects , Neurons/physiology , PC12 Cells , Rats
4.
Acta Neuropathol ; 128(6): 853-62, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25315281

ABSTRACT

Although telomeres are maintained in most cancers by telomerase activation, a subset of tumors utilize alternative lengthening of telomeres (ALT) to sustain self-renewal capacity. In order to study the prevalence and significance of ALT in childhood brain tumors we screened 517 pediatric brain tumors using the novel C-circle assay. We examined the association of ALT with alterations in genes found to segregate with specific histological phenotypes and with clinical outcome. ALT was detected almost exclusively in malignant tumors (p = 0.001). ALT was highly enriched in primitive neuroectodermal tumors (12 %), choroid plexus carcinomas (23 %) and high-grade gliomas (22 %). Furthermore, in contrast to adult gliomas, pediatric low grade gliomas which progressed to high-grade tumors did not exhibit the ALT phenotype. Somatic but not germline TP53 mutations were highly associated with ALT (p = 1.01 × 10(-8)). Of the other alterations examined, only ATRX point mutations and reduced expression were associated with the ALT phenotype (p = 0.0005). Interestingly, ALT attenuated the poor outcome conferred by TP53 mutations in specific pediatric brain tumors. Due to very poor prognosis, one year overall survival was quantified in malignant gliomas, while in children with choroid plexus carcinoma, five year overall survival was investigated. For children with TP53 mutant malignant gliomas, one year overall survival was 63 ± 12 and 23 ± 10 % for ALT positive and negative tumors, respectively (p = 0.03), while for children with TP53 mutant choroid plexus carcinomas, 5 years overall survival was 67 ± 19 and 27 ± 13 % for ALT positive and negative tumors, respectively (p = 0.07). These observations suggest that the presence of ALT is limited to a specific group of childhood brain cancers which harbor somatic TP53 mutations and may influence the outcome of these patients. Analysis of ALT may contribute to risk stratification and targeted therapies to improve outcome for these children.


Subject(s)
Brain Neoplasms/genetics , Carcinoma/genetics , Choroid Plexus Neoplasms/genetics , Glioma/genetics , Neuroectodermal Tumors, Primitive/genetics , Telomere , Tumor Suppressor Protein p53/genetics , Adolescent , Brain Neoplasms/physiopathology , Carcinoma/physiopathology , Choroid Plexus Neoplasms/physiopathology , Cohort Studies , DNA Helicases/genetics , Glioma/physiopathology , Humans , Kaplan-Meier Estimate , Mutation , Neoplasm Grading , Neuroectodermal Tumors, Primitive/physiopathology , Nuclear Proteins/genetics , Phenotype , Prognosis , Telomere/metabolism , X-linked Nuclear Protein
5.
Lancet Oncol ; 14(6): 534-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23598174

ABSTRACT

BACKGROUND: Identification of robust biomarkers of malignancy and methods to establish disease progression is a major goal in paediatric neuro-oncology. We investigated whether methylation of the TERT promoter can be a biomarker for malignancy and patient outcome in paediatric brain tumours. METHODS: For the discovery cohort, we used samples obtained from patients with paediatric brain tumours and individuals with normal brain tissues stored at the German Cancer Research Center (Heidelberg, Germany). We used methylation arrays for genome-wide assessment of DNA. For the validation cohort, we used samples obtained from several tissues for which full clinical and follow-up data were available from two hospitals in Toronto (ON, Canada). We did methylation analysis using quantitative Sequenom and pyrosequencing of an identified region of the TERT promoter. We assessed TERT expression by real-time PCR. To establish whether the biomarker could be used to assess and predict progression, we analysed methylation in paired samples of tumours that transformed from low to high grade and from localised to metastatic, and in choroid plexus tumours of different grades. Finally, we investigated overall survival in patients with posterior fossa ependymomas in which the identified region was hypermethylated or not. All individuals responsible for assays were masked to the outcome of the patients. FINDINGS: Analysis of 280 samples in the discovery cohort identified one CpG site (cg11625005) in which 78 (99%) of 79 samples from normal brain tissues and low-grade tumours were not hypermethylated, but 145 (72%) of 201 samples from malignant tumours were hypermethylated (>15% methylated; p<0.0001). Analysis of 68 samples in the validation cohort identified a subset of five CpG sites (henceforth, upstream of the transcription start site [UTSS]) that was hypermethylated in all malignant paediatric brain tumours that expressed TERT but not in normal tissues that did not express TERT (p<0.0001). UTSS had a positive predictive value of 1.00 (95% CI 0.95-1.00) and a negative predictive value of 0.95 (0.87-0.99). In two paired samples of paediatric gliomas, UTSS methylation increased during transformation from low to high grade; it also increased in two paired samples that progressed from localised to metastatic disease. Two of eight atypical papillomas that had high UTSS methylation progressed to carcinomas, while the other six assessed did not progress or require additional treatment. 5-year overall survival was 51% (95% CI 31-71) for 25 patients with hypermethylated UTSS posterior fossa ependymomas and 95% (86-100) for 20 with non-hypermethylated tumours (p=0.0008). 5-year progression-free survival was 86% (68-100) for the 25 patients with non-hypermethylated UTSS tumours and 30% (10-50) for those with hypermethylated tumours (p=0.0008). INTERPRETATION: Hypermethylation of the UTSS region in the TERT promoter is associated with TERT expression in cancers. In paediatric brain tumours, UTSS hypermethylation is associated with tumour progression and poor prognosis. This region is easy to amplify, and the assay to establish hypermethylation can be done on most tissues in most clinical laboratories. Therefore the UTSS region is a potentially accessible biomarker for various cancers. FUNDING: The Canadian Institute of Health Research and the Terry Fox Foundation.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , DNA Methylation , Promoter Regions, Genetic , Telomerase/genetics , Age of Onset , Brain Neoplasms/enzymology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , CpG Islands , Disease-Free Survival , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genome-Wide Association Study , Germany , Humans , Kaplan-Meier Estimate , Multivariate Analysis , Neoplasm Grading , Ontario , Phenotype , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Retrospective Studies , Time Factors , Transcription Initiation Site
6.
Cancer Res Commun ; 3(5): 738-754, 2023 05.
Article in English | MEDLINE | ID: mdl-37377903

ABSTRACT

Li-Fraumeni syndrome (LFS) is an autosomal dominant cancer-predisposition disorder. Approximately 70% of individuals who fit the clinical definition of LFS harbor a pathogenic germline variant in the TP53 tumor suppressor gene. However, the remaining 30% of patients lack a TP53 variant and even among variant TP53 carriers, approximately 20% remain cancer-free. Understanding the variable cancer penetrance and phenotypic variability in LFS is critical to developing rational approaches to accurate, early tumor detection and risk-reduction strategies. We leveraged family-based whole-genome sequencing and DNA methylation to evaluate the germline genomes of a large, multi-institutional cohort of patients with LFS (n = 396) with variant (n = 374) or wildtype TP53 (n = 22). We identified alternative cancer-associated genetic aberrations in 8/14 wildtype TP53 carriers who developed cancer. Among variant TP53 carriers, 19/49 who developed cancer harbored a pathogenic variant in another cancer gene. Modifier variants in the WNT signaling pathway were associated with decreased cancer incidence. Furthermore, we leveraged the noncoding genome and methylome to identify inherited epimutations in genes including ASXL1, ETV6, and LEF1 that confer increased cancer risk. Using these epimutations, we built a machine learning model that can predict cancer risk in patients with LFS with an area under the receiver operator characteristic curve (AUROC) of 0.725 (0.633-0.810). Significance: Our study clarifies the genomic basis for the phenotypic variability in LFS and highlights the immense benefits of expanding genetic and epigenetic testing of patients with LFS beyond TP53. More broadly, it necessitates the dissociation of hereditary cancer syndromes as single gene disorders and emphasizes the importance of understanding these diseases in a holistic manner as opposed to through the lens of a single gene.


Subject(s)
Li-Fraumeni Syndrome , Humans , Li-Fraumeni Syndrome/genetics , Tumor Suppressor Protein p53/genetics , Genetic Predisposition to Disease/genetics , Genes, p53 , Germ-Line Mutation/genetics
7.
Biotechnol Bioeng ; 109(7): 1791-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22252990

ABSTRACT

We study the role of nano-scale cues in controlling neuronal growth. We use photolithography to fabricate substrates with repeatable line-pattern ridges of nano-scale heights. We find that neuronal processes, which are of micron size, have strong interactions with ridges even as low as 10 nm. The interaction between the neuronal process and the ridge leads to a deflection of growth direction and a preferred alignment with the ridges. The interaction strength clearly depends on the ridges' height. For 25 nm ridges approximately half of the neuronal processes are modified, while at 100 nm the majority of neurites change their original growth direction post interaction. In addition, the effect on growth correlates with the incoming angle between the neuronal process and the ridge. We underline the adhesion as a key mechanism in directing neuronal growth. Our study highlights the sensitivity of growing neurites to nano-scale cues thus opens a new avenue of research for pre-designed neuronal growth and circuitry.


Subject(s)
Hirudo medicinalis/cytology , Neurons/cytology , Tissue Scaffolds/chemistry , Animals , Cell Proliferation , Cells, Cultured , Nanostructures/chemistry
8.
Nat Commun ; 11(1): 3945, 2020 08 07.
Article in English | MEDLINE | ID: mdl-32770028

ABSTRACT

TP53 missense mutations leading to the expression of mutant p53 oncoproteins are frequent driver events during tumorigenesis. p53 mutants promote tumor growth, metastasis and chemoresistance by affecting fundamental cellular pathways and functions. Here, we demonstrate that p53 mutants modify structure and function of the Golgi apparatus, culminating in the increased release of a pro-malignant secretome by tumor cells and primary fibroblasts from patients with Li-Fraumeni cancer predisposition syndrome. Mechanistically, interacting with the hypoxia responsive factor HIF1α, mutant p53 induces the expression of miR-30d, which in turn causes tubulo-vesiculation of the Golgi apparatus, leading to enhanced vesicular trafficking and secretion. The mut-p53/HIF1α/miR-30d axis potentiates the release of soluble factors and the deposition and remodeling of the ECM, affecting mechano-signaling and stromal cells activation within the tumor microenvironment, thereby enhancing tumor growth and metastatic colonization.


Subject(s)
Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Golgi Apparatus/pathology , Li-Fraumeni Syndrome/genetics , MicroRNAs/metabolism , Tumor Suppressor Protein p53/genetics , Animals , Biopsy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Female , Fibroblasts , Gene Expression Regulation, Neoplastic , Golgi Apparatus/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Li-Fraumeni Syndrome/pathology , Mice , Microtubules/metabolism , Microtubules/pathology , Mutation , Primary Cell Culture , Secretory Vesicles/metabolism , Secretory Vesicles/pathology , Signal Transduction/genetics , Skin/cytology , Skin/pathology , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
9.
Mutat Res ; 578(1-2): 23-32, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-15998523

ABSTRACT

Renal clear-cell carcinoma (RCC) is the predominant form of kidney cancer and is highly refractory to conventional anti-cancer therapies. The status of p53 tumor suppressor gene has been correlated with the efficacy of radio- and chemotherapies, where presence of mutant p53 is associated with reduced responsiveness to treatment. However, p53 itself is rarely mutated in RCC, rather suggesting that the p53 pathway might be compromized in RCC cells. In support of this notion, the transactivation property of normal p53 was shown to be repressed in various transformed kidney epithelial cells via an unknown dominant-negative mechanism. Mutation of the von Hippel-Lindau (VHL) gene causes familial VHL disease, which includes predisposition to RCC. Moreover, biallelic inactivation of VHL has been observed in the vast majority of sporadic RCC. Recently, the expression of pVHL in RCC cells was demonstrated to elevate the expression of p53 by inducing the binding of RNA-stabilizing protein HuR to the 3'untranslated region of p53 mRNA. Contrary to this finding, we report here that the reconstitution of a variety of VHL(-/-) RCC lines including 786-O, RCC4, and A498 or non-RCC cells with wild-type pVHL does not influence the expression of p53 and fails to induce p53-responsive gene p21CIP1/WAF1 or p53-responsive reporters. These results suggest that the expression of p53 in RCC cells is independent of pVHL.


Subject(s)
Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Humans , Molecular Sequence Data , Mutation, Missense , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
10.
J Biomed Opt ; 20(2): 20502, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25652701

ABSTRACT

We irradiated neuroblastoma SH-SY5Y cell line with low-level light-emitting diode (LED) illumination at a visible wavelength of 520 nm (green) and intensity of 100 mW∕cm2. We captured and analyzed the cell morphology before LED treatment, immediately after, and 12 and 24 h after treatment. Our study demonstrated that LED illumination increases the amount of sprouting dendrites in comparison to the control untreated cells. This treatment also resulted in more elongated cells after treatment in comparison to the control cells and higher levels of expression of a differentiation related gene. This result is a good indication that the proposed method could serve in phototherapy treatment for increasing sprouting and enhancing neural network formation.


Subject(s)
Models, Biological , Neurites/radiation effects , Neurons/radiation effects , Phototherapy , Animals , Cell Line, Tumor , Light , Lighting
11.
J Clin Oncol ; 33(9): 1015-22, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25667294

ABSTRACT

PURPOSE: To uncover the genetic events leading to transformation of pediatric low-grade glioma (PLGG) to secondary high-grade glioma (sHGG). PATIENTS AND METHODS: We retrospectively identified patients with sHGG from a population-based cohort of 886 patients with PLGG with long clinical follow-up. Exome sequencing and array CGH were performed on available samples followed by detailed genetic analysis of the entire sHGG cohort. Clinical and outcome data of genetically distinct subgroups were obtained. RESULTS: sHGG was observed in 2.9% of PLGGs (26 of 886 patients). Patients with sHGG had a high frequency of nonsilent somatic mutations compared with patients with primary pediatric high-grade glioma (HGG; median, 25 mutations per exome; P = .0042). Alterations in chromatin-modifying genes and telomere-maintenance pathways were commonly observed, whereas no sHGG harbored the BRAF-KIAA1549 fusion. The most recurrent alterations were BRAF V600E and CDKN2A deletion in 39% and 57% of sHGGs, respectively. Importantly, all BRAF V600E and 80% of CDKN2A alterations could be traced back to their PLGG counterparts. BRAF V600E distinguished sHGG from primary HGG (P = .0023), whereas BRAF and CDKN2A alterations were less commonly observed in PLGG that did not transform (P < .001 and P < .001 respectively). PLGGs with BRAF mutations had longer latency to transformation than wild-type PLGG (median, 6.65 years [range, 3.5 to 20.3 years] v 1.59 years [range, 0.32 to 15.9 years], respectively; P = .0389). Furthermore, 5-year overall survival was 75% ± 15% and 29% ± 12% for children with BRAF mutant and wild-type tumors, respectively (P = .024). CONCLUSION: BRAF V600E mutations and CDKN2A deletions constitute a clinically distinct subtype of sHGG. The prolonged course to transformation for BRAF V600E PLGGs provides an opportunity for surgical interventions, surveillance, and targeted therapies to mitigate the outcome of sHGG.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Cyclin-Dependent Kinase Inhibitor p16/genetics , Gene Deletion , Glioma/genetics , Glioma/secondary , Proto-Oncogene Proteins B-raf/genetics , Adolescent , Cell Transformation, Neoplastic , Child , Child, Preschool , Chromatin/chemistry , Disease Progression , Female , Follow-Up Studies , Humans , Infant , Male , Mutation , Point Mutation , Retrospective Studies , Telomere/ultrastructure , Treatment Outcome
12.
ACS Chem Neurosci ; 5(5): 370-6, 2014 May 21.
Article in English | MEDLINE | ID: mdl-24559496

ABSTRACT

Studying the interaction between neuronal cells and chiral molecules is fundamental for the design of novel biomaterials and drugs. Chirality influences all biological processes that involve intermolecular interaction. One common method used to study cellular interactions with different enantiomeric targets is the use of chiral surfaces. Based on previous studies that demonstrated the importance of cysteine in the nervous system, we studied the effect of L- and D-cysteine on single neuronal growth. L-Cysteine, which normally functions as a neuromodulator or a neuroprotective antioxidant, causes damage at elevated levels, which may occur post trauma. In this study, we grew adult neurons in culture enriched with L- and D-cysteine as free compounds or as self-assembled monolayers of chiral surfaces and examined the effect on the neuronal morphology and adhesion. Notably, we have found that exposure to the L-cysteine enantiomer inhibited, and even prevented, neuronal attachment more severely than exposure to the D-cysteine enantiomer. Atop the L-cysteine surfaces, neuronal growth was reduced and degenerated. Since the cysteine molecules were attached to the surface via the thiol groups, the neuronal membrane was exposed to the molecular chiral site. Thus, our results have demonstrated high neuronal chiral sensitivity, revealing chiral surfaces as indirect regulators of neuronal cells and providing a reference for studying chiral drugs.


Subject(s)
Cysteine/pharmacology , Neurogenesis/drug effects , Neurons/drug effects , Adsorption , Animals , Cells, Cultured , Dose-Response Relationship, Drug , Ganglia/cytology , Leeches , Membrane Potentials/drug effects , Mice , Microscopy, Electron, Scanning , Neurons/ultrastructure , Stereoisomerism , Surface Properties/drug effects , Time Factors
13.
Int J Nanomedicine ; 9 Suppl 1: 23-31, 2014.
Article in English | MEDLINE | ID: mdl-24872701

ABSTRACT

Much effort has been devoted to the design of effective biomaterials for nerve regeneration. Here, we report the novel use of silver nanoparticles (AgNPs) as regenerative agents to promote neuronal growth. We grew neuroblastoma cells on surfaces coated with AgNPs and studied the effect on the development of the neurites during the initiation and the elongation growth phases. We find that the AgNPs function as favorable anchoring sites, and the growth on the AgNP-coated substrates leads to a significantly enhanced neurite outgrowth. Cells grown on substrates coated with AgNPs have initiated three times more neurites than cells grown on uncoated substrates, and two times more than cells grown on substrates sputtered with a plain homogenous layer of silver. The growth of neurites on AgNPs in the elongation phase was enhanced as well. A comparison with substrates coated with gold nanoparticles (AuNPs) and zinc oxide nanoparticles (ZnONPs) demonstrated a clear silver material-driven promoting effect, in addition to the nanotopography. The growth on substrates coated with AgNPs has led to a significantly higher number of initiating neurites when compared to substrates coated with AuNPs or ZnONPs. All nanoparticle-coated substrates affected and promoted the elongation of neurites, with a significant positive maximal effect for the AgNPs. Our results, combined with the well-known antibacterial effect of AgNPs, suggest the use of AgNPs as an attractive nanomaterial - with dual activity - for neuronal repair studies.


Subject(s)
Metal Nanoparticles/chemistry , Nerve Regeneration/drug effects , Neurites/drug effects , Silver/pharmacology , Anti-Bacterial Agents , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Nanomedicine , Silver/chemistry
14.
Cancer Res ; 72(3): 636-44, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22144470

ABSTRACT

Although monoallelic expression (MAE) is a frequent genomic event in normal tissues, its role in tumorigenesis remains unclear. Here we carried out single-nucleotide polymorphism arrays on DNA and RNA from a large cohort of pediatric and adult brain tumor tissues to determine the genome-wide rate of MAE, its role in specific cancer-related genes, and the clinical consequences of MAE in brain tumors. We also used targeted genotyping to examine the role of tumor-related genes in brain tumor development and specifically examined the clinical consequences of MAE at TP53 and IDH1. The genome-wide rate of tumor MAE was higher than in previously described normal tissue and increased with specific tumor grade. Oncogenes, but not tumor suppressors, exhibited significantly higher MAE in high-grade compared with low-grade tumors. This method identified nine novel genes highly associated with MAE. Within cancer-related genes, MAE was gene specific; hTERT was most significantly affected, with a higher frequency of MAE in adult and advanced tumors. Clinically, MAE at TP53 exists only in mutated tumors and increases with tumor aggressiveness. MAE toward the normal allele at IDH1 conferred worse survival even in IDH1 mutated tumors. Taken together, our findings suggest that MAE is tumor and gene specific, frequent in brain tumor subtypes, and may be associated with tumor progression/aggressiveness. Further exploration of MAE at relevant genes may contribute to better understanding of tumor development and determine survival in brain tumor patients.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Isocitrate Dehydrogenase/genetics , Polymorphism, Single Nucleotide/genetics , Tumor Suppressor Protein p53/genetics , Adult , Alleles , Autopsy , Brain Neoplasms/pathology , Cell Line , Cell Line, Tumor , Child , Disease Progression , Genetic Predisposition to Disease/genetics , Genome, Human/genetics , Genome-Wide Association Study/methods , Genotype , Humans , Loss of Heterozygosity , Mutation , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Prognosis , Survival Analysis , Telomerase/genetics
15.
Clin Cancer Res ; 17(14): 4790-8, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21610142

ABSTRACT

PURPOSE: Recent studies have revealed that the majority of pediatric low-grade astrocytomas (PLGA) harbor the BRAF-KIAA1549 (B-K) fusion gene resulting in constitutive activation of the RAS/MAPK pathway. However, the clinical significance of this genetic alteration is yet to be determined. We aimed to test the prognostic role of the B-K fusion in progression of incompletely resected PLGA. EXPERIMENTAL DESIGN: We retrospectively identified 70 consecutive patients with incompletely resected "clinically relevant" PLGA. We added 76 tumors diagnosed at our institution between 1985 and 2010 as controls. We examined BRAF alterations by reverse transcriptase PCR, FISH, and single-nucleotide polymorphism array analysis and correlated that with progression-free survival (PFS). RESULTS: Overall, 60% of tumors were B-K fusion positive. All patients with B-K fused PLGA are still alive. Five-year PFS was 61% ± 8% and 18% ± 8% for fusion positive and negative patients, respectively (P = 0.0004). B-K fusion resulted in similarly significant favorable PFS for patients who received chemotherapy. Multivariate analysis revealed that B-K fusion was the most significant favorable prognostic factor in incompletely resected PLGA and was independent of location, pathology, and age. In vitro, BRAF overexpression resulted in growth arrest associated with DNA damage (γH2AX expression). Five-year PFS was 68% ± 15% and 0% for patients with B-K fused and γH2AX-expressing PLGA versus negative tumors (P = 0.001). CONCLUSION: These data suggest that B-K fusion confers a less aggressive clinical phenotype on PLGA and may explain their tendency to growth arrest. Combined analysis of B-K fusion and γH2AX expression can determine prognosis and may be a powerful tool to tailor therapy for these patients.


Subject(s)
Astrocytoma/diagnosis , Astrocytoma/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Aging/genetics , Astrocytes/metabolism , Astrocytes/pathology , Astrocytoma/mortality , Child , Child, Preschool , Disease Progression , Female , Gene Expression , Gene Expression Regulation, Neoplastic , Histones/genetics , Humans , Infant , Kaplan-Meier Estimate , Male , Neoplasm Staging , Oncogene Proteins, Fusion/metabolism , Treatment Outcome
16.
J Clin Oncol ; 28(8): 1345-50, 2010 Mar 10.
Article in English | MEDLINE | ID: mdl-20142599

ABSTRACT

PURPOSE: Medulloblastoma is the prototype of treatment success in modern pediatric neuro-oncology. Unfortunately, 20% to 30% of tumors recur despite maximal resection and multimodal therapy. Multiple biologic prognostic markers have been investigated to predict recurrences, but controversy remains regarding their clinical utility. Because p53 immunopositivity is an adverse prognostic marker in pediatric medulloblastoma and TP53 mutations are associated with chemotherapy and radiation therapy resistance, we aimed to determine the extent and role of TP53 mutations in pediatric medulloblastoma treatment failure. PATIENTS AND METHODS: One hundred eight of 111 consecutive patients diagnosed with medulloblastoma in our institution from 1995 to 2007 were included. Median follow-up time was 5.3 years in survivors. All samples were immunostained for p53 and erbB-2. Histologic grade and immunostaining were scored by two blinded reviewers. For 49 patients, frozen material was available for TP53 sequencing. The main outcome measures were overall and progression-free survival. RESULTS: Sixteen percent of sequenced medulloblastomas harbored a TP53 mutation. As a screening test, p53 immunohistochemistry was 100% sensitive and 83% specific for a TP53 mutation. Strikingly, all mutated tumors recurred early, and 5-year survival for average-risk patients was 0% for TP53-mutated medulloblastoma compared with 74% +/- 8% for wild-type medulloblastoma (P < .0001). Furthermore, 75% of recurrences in average-risk patients were associated with TP53 mutations. On multivariate analysis, TP53 mutation status was the strongest adverse prognostic factor (hazard ratio = 10.4, P = .003). CONCLUSION: Lack of long-term survival in TP53-mutated medulloblastomas highlights the role of TP53 mutations in medulloblastoma resistance to conventional therapies and the need for alternative treatments, and prospective validation of these findings is needed.


Subject(s)
Biomarkers, Tumor/genetics , Genes, p53/genetics , Medulloblastoma/genetics , Medulloblastoma/mortality , Mutation , Adolescent , Child , Child, Preschool , Follow-Up Studies , Humans , Immunohistochemistry , Infant , Infant, Newborn , Medulloblastoma/pathology , Multivariate Analysis , Ontario/epidemiology , Receptor, ErbB-2/metabolism , Retrospective Studies , Sensitivity and Specificity , Survival Analysis , Treatment Failure , Tumor Suppressor Protein p53/metabolism
17.
J Clin Oncol ; 28(12): 1995-2001, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20308654

ABSTRACT

PURPOSE Choroid plexus carcinomas are pediatric tumors with poor survival rates and a strong, but poorly understood, association with Li-Fraumeni syndrome (LFS). Currently, with lack of biologic predictors, most children are treated with aggressive chemoradiation protocols. PATIENTS AND METHODS We established a multi-institutional tissue and clinical database, which enabled the analysis of specific alterations of the TP53 tumor suppressor and its modifiers in choroid plexus tumors (CPTs). We conducted high-resolution copy-number analysis to correlate these genetic parameters with family history and outcome. Results We studied 64 patients with CPTs. All individuals with germline TP53 mutations fulfilled LFS criteria, whereas all patients not meeting these criteria harbored wild-type TP53 (P < .001). TP53 mutations were found in 50% of choroid plexus carcinomas (CPCs). Additionally, two sequence variants known to confer TP53 dysfunction, TP53 codon72 and MDM2 SNP309, coexisted in the majority of TP53 wild-type CPCs (92%) and not in TP53 mutated CPC (P = .04), which suggests a complementary mechanism of TP53 dysfunction in the absence of a TP53 mutation. High-resolution single nucleotide polymorphism (SNP) array analysis revealed extremely high total structural variation (TSV) in TP53-mutated CPC tumor genomes compared with TP53 wild-type tumors and choroid plexus papillomas (CPPs; P = .006 and .004, respectively). Moreover, high TSV was associated with significant risk of progression (P < .001). Five-year survival rates for patients with TP53-immunopositive and -immunonegative CPCs were 0% and 82 (+/- 9%), respectively (P < .001). Furthermore, 14 of 16 patients with TP53 wild-type CPCs are alive without having received radiation therapy. CONCLUSION Patients with CPC who have low tumor TSV and absence of TP53 dysfunction have a favorable prognosis and can be successfully treated without radiation therapy.


Subject(s)
Carcinoma/genetics , Choroid Plexus Neoplasms/genetics , Germ-Line Mutation , Papilloma, Choroid Plexus/genetics , Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics , Carcinoma/chemistry , Carcinoma/mortality , Carcinoma/therapy , Chi-Square Distribution , Child , Child, Preschool , Choroid Plexus Neoplasms/chemistry , Choroid Plexus Neoplasms/mortality , Choroid Plexus Neoplasms/therapy , Databases as Topic , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Oligonucleotide Array Sequence Analysis , Ontario/epidemiology , Papilloma, Choroid Plexus/chemistry , Papilloma, Choroid Plexus/mortality , Papilloma, Choroid Plexus/therapy , Phenotype , Time Factors , Treatment Outcome , Tumor Suppressor Protein p53/analysis , United States/epidemiology
18.
J Biol Chem ; 280(43): 36118-25, 2005 Oct 28.
Article in English | MEDLINE | ID: mdl-16127171

ABSTRACT

Fanconi anemia (FA) is a genetically heterogeneous disorder characterized by bone marrow failure, cancer predisposition, and increased cellular sensitivity to DNA-cross-linking agents. The products of seven of the nine identified FA genes participate in a protein complex required for monoubiquitination of the FANCD2 protein. Direct interaction of the FANCE protein with both fellow FA complex component FANCC and the downstream FANCD2 protein has been observed in the yeast two-hybrid system. Here, we demonstrate the ability of FANCE to mediate the interaction between FANCC and FANCD2 in the yeast three-hybrid system and confirm the FANCE-mediated association of FANCC with FANCD2 in human cells. A yeast two-hybrid system-based screen was devised to identify randomly mutagenized FANCE proteins capable of interaction with FANCC but not with FANCD2. Exogenous expression of these mutants in an FA-E cell line and subsequent evaluation of FANCD2 monoubiquitination and DNA cross-linker sensitivity indicated a critical role for the FANCE/FANCD2 interaction in maintaining FA pathway integrity. Three-hybrid experiments also demonstrated the ability of FANCE to mediate the interaction between FA core complex components FANCC and FANCF, indicating an additional role for FANCE in complex assembly. Thus, FANCE is shown to be a key mediator of protein interactions both in the architecture of the FA protein complex and in the connection of complex components to the putative downstream targets of complex activity.


Subject(s)
DNA Damage , Fanconi Anemia Complementation Group C Protein/physiology , Fanconi Anemia Complementation Group D2 Protein/physiology , Fanconi Anemia Complementation Group E Protein/physiology , Fanconi Anemia/genetics , Fanconi Anemia/metabolism , Cell Line , Cell Survival , Cross-Linking Reagents/pharmacology , Dose-Response Relationship, Drug , Genes, Reporter , Genetic Vectors , Humans , Immunoprecipitation , Mitomycin/pharmacology , Models, Biological , Models, Genetic , Mutagenesis , Mutagenesis, Site-Directed , Mutation , Open Reading Frames , Protein Binding , Saccharomyces cerevisiae/metabolism , Two-Hybrid System Techniques
19.
J Hum Genet ; 48(12): 668-671, 2003.
Article in English | MEDLINE | ID: mdl-14605947

ABSTRACT

Loss of heterozygosity (LOH) involving chromosome 16q23.4 occurs frequently in breast tumors, which suggests that this region may contain a tumor suppressor gene. Since ZFP276 is located in this region, we have therefore cloned and performed mutation analysis of its coding region in 70 breast tumors. One silent polymorphism and two alterations predicted to result in amino acid changes were detected. Absence of the wild-type allele in tumors carrying the E530D variant suggests a possible role for this change in tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , Genes, Tumor Suppressor , Polymorphism, Genetic , Tumor Suppressor Proteins/genetics , Alleles , Amino Acid Sequence , Blotting, Northern , Breast Neoplasms/pathology , Chromosomes, Human, Pair 16 , Cloning, Molecular , DNA Mutational Analysis , DNA, Complementary/metabolism , Female , Humans , Models, Genetic , Molecular Sequence Data , Nuclear Proteins , Polymorphism, Single-Stranded Conformational , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Zinc Fingers
20.
Hum Mol Genet ; 12(16): 2063-76, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12913077

ABSTRACT

Fanconi Anemia (FA) is an autosomal recessive disorder characterized by cellular hypersensitivity to DNA cross-linking agents. Recent studies suggest that FA proteins share a common pathway with BRCA proteins. To study the in vivo role of the FA group A gene (Fanca), gene-targeting techniques were used to generate Fanca(tm1Hsc) mice in which Fanca exons 1-6 were replaced by a beta-galactosidase reporter construct. Fanca(tm1.1Hsc) mice were generated by Cre-mediated removal of the neomycin cassette in Fanca(tm1Hsc) mice. Fanca(tm1.1Hsc) homozygotes display FA-like phenotypes including growth retardation, microphthalmia and craniofacial malformations that are not found in other Fanca mouse models, and the genetic background affects manifestation of certain phenotypes. Both male and female mice homozygous for Fanca mutation exhibit hypogonadism, and homozygous females demonstrate premature reproductive senescence and an increased incidence of ovarian cysts. We showed that fertility defects in Fanca(tm1.1Hsc) homozygotes might be related to a diminished population of primordial germ cells (PGCs) during migration into the gonadal ridges. We also found a high level of Fanca expression in pachytene spermatocytes. Fanca(tm1Hsc) homozygous males exhibited an elevated frequency of mispaired meiotic chromosomes and increased apoptosis in germ cells, implicating a role for Fanca in meiotic recombination. However, the localization of Rad51, Brca1, Fancd2 and Mlh1 appeared normal on Fanca(tm1Hsc) homozygous meiotic chromosomes. Taken together, our results suggest that the FA pathway plays a role in the maintenance of reproductive germ cells and in meiotic recombination.


Subject(s)
DNA-Binding Proteins , Fanconi Anemia/genetics , Germ Cells/pathology , Meiosis/genetics , Proteins/genetics , Proteins/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Exons , Fanconi Anemia Complementation Group A Protein , Female , Fetal Growth Retardation , Gene Targeting , Infertility, Female/genetics , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microphthalmos/genetics , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL