Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
PNAS Nexus ; 2(6): pgad170, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37346271

RESUMO

The expanding field of precision gene editing using CRISPR/Cas9 has demonstrated its potential as a transformative technology in the treatment of various diseases. However, whether this genome-editing tool could be used to modify neural circuits in the central nervous system (CNS), which are implicated in complex behavioral traits, remains uncertain. In this study, we demonstrate the feasibility of noninvasive, intranasal delivery of adeno-associated virus serotype 9 (AAV9) vectors containing CRISPR/Cas9 cargo within the CNS resulting in modification of the HTR2A receptor gene. In vitro, exposure to primary mouse cortical neurons to AAV9 vectors targeting the HT2RA gene led to a concentration-dependent decrease in spontaneous electrical activity following multielectrode array (MEA) analysis. In vivo, at 5 weeks postintranasal delivery in mice, analysis of brain samples revealed single base pair deletions and nonsense mutations, leading to an 8.46-fold reduction in mRNA expression and a corresponding 68% decrease in the 5HT-2A receptor staining. Our findings also demonstrate a significant decrease in anxiety-like behavior in treated mice. This study constitutes the first successful demonstration of a noninvasive CRISPR/Cas9 delivery platform, capable of bypassing the blood-brain barrier and enabling modulation of neuronal 5HT-2A receptor pathways. The results of this study targeting the HTR2A gene provide a foundation for the development of innovative therapeutic strategies for a broad range of neurological disorders, including anxiety, depression, attentional deficits, and cognitive dysfunction.

2.
EClinicalMedicine ; 38: 101028, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34308321

RESUMO

BACKGROUND: The negative impact of continued school closures during the height of the COVID-19 pandemic warrants the establishment of cost-effective strategies for surveillance and screening to safely reopen and monitor for potential in-school transmission. Here, we present a novel approach to increase the availability of repetitive and routine COVID-19 testing that may ultimately reduce the overall viral burden in the community. METHODS: We implemented a testing program using the SalivaClear࣪ pooled surveillance method that included students, faculty and staff from K-12 schools (student age range 5-18 years) and universities (student age range >18 years) across the country (Mirimus Clinical Labs, Brooklyn, NY). The data analysis was performed using descriptive statistics, kappa agreement, and outlier detection analysis. FINDINGS: From August 27, 2020 until January 13, 2021, 253,406 saliva specimens were self-collected from students, faculty and staff from 93 K-12 schools and 18 universities. Pool sizes of up to 24 samples were tested over a 20-week period. Pooled testing did not significantly alter the sensitivity of the molecular assay in terms of both qualitative (100% detection rate on both pooled and individual samples) and quantitative (comparable cycle threshold (Ct) values between pooled and individual samples) measures. The detection of SARS-CoV-2 in saliva was comparable to the nasopharyngeal swab. Pooling samples substantially reduced the costs associated with PCR testing and allowed schools to rapidly assess transmission and adjust prevention protocols as necessary. In one instance, in-school transmission of the virus was determined within the main office and led to review and revision of heating, ventilating and air-conditioning systems. INTERPRETATION: By establishing low-cost, weekly testing of students and faculty, pooled saliva analysis for the presence of SARS-CoV-2 enabled schools to determine whether transmission had occurred, make data-driven decisions, and adjust safety protocols. We provide strong evidence that pooled testing may be a fundamental component to the reopening of schools by minimizing the risk of in-school transmission among students and faculty. FUNDING: Skoll Foundation generously provided funding to Mobilizing Foundation and Mirimus for these studies.

3.
J Neurooncol ; 147(3): 557-566, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32193690

RESUMO

INTRODUCTION: To improve the standard treatment paradigm for glioblastoma (GBM), efforts have been made to explore the efficacy of epigenetic agents as chemosensitizers. Recent data suggest possible synergy between decitabine (DAC), a DNA hypomethylating agent, and temozolomide (TMZ) in GBM, but the mechanism remains unclear. The objective of this study was to determine the effects of DAC on TMZ sensitization in a consecutively derived set of primary GBM cultures, with a focus on mismatch repair (MMR) proteins. METHODS: Half maximal inhibitory concentrations (IC50) of TMZ were calculated in eleven consecutive patient-derived GBM cell lines before and after preconditioning with DAC. MMR protein expression changes were determined by quantitative immunoblots and qPCR arrays. Single-molecule real-time (SMRT) sequencing of bisulfite (BS)-converted PCR amplicons of the MLH1 promoter was performed to determine methylation status. RESULTS: TMZ IC50 significantly changed in 6 of 11 GBM lines of varying MGMT promoter methylation status in response to DAC preconditioning. Knockdown of MLH1 after preconditioning reversed TMZ sensitization. SMRT-BS sequencing of the MLH1 promoter region revealed higher levels of baseline methylation at proximal CpGs in desensitized lines compared to sensitized lines. CONCLUSIONS: DAC enhances TMZ cytotoxicity in a subset of GBM cell lines, comprising lines both MGMT methylated and unmethylated tumors. This effect may be driven by levels of MLH1 via E2F1 transcription factor binding. Using unbiased long-range next-generation bisulfite-sequencing, we identified a region of the proximal MLH1 promoter with differential methylation patterns that has potential utility as a clinical biomarker for TMZ sensitization.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Encefálicas/genética , Decitabina/administração & dosagem , Epigênese Genética/efeitos dos fármacos , Glioblastoma/genética , Proteína 1 Homóloga a MutL/metabolismo , Temozolomida/administração & dosagem , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Glioblastoma/metabolismo , Humanos , Concentração Inibidora 50
4.
Oncotarget ; 9(2): 2603-2621, 2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29416795

RESUMO

BACKGROUND: Gliosarcoma is a rare variant of glioblastoma (GBM) that exhibits frequent mutations in TP53 and can develop in a secondary fashion after chemoradiation of a primary GBM. Whether temozolomide (TMZ)-induced mutagenesis of the TP53 DNA-binding domain (DBD) can drive the pathogenesis of gliosarcoma is unclear. METHODS: We identified a case of a primary GBM that rapidly progressed into secondary gliosarcoma shortly after chemoradiation was initiated. Bulk tumor was collected and gliomasphere cultures derived from both the pre- and post-treatment tumors. We performed targeted DNA sequencing and transcriptome analyses of the specimens to understand their phylogenetic relationship and identify differentially expressed gene pathways. Gliomaspheres from the primary GBM were treated with TMZ and then analyzed to compare patterns of mutagenesis in vivo and ex vivo. RESULTS: The pre- and post-treatment tumors shared EGFR, CDKN2A, and PTEN mutations, but only the secondary gliosarcoma exhibited TP53 DBD missense mutations. Two mutations, R110C, and R175H, were identified, each in distinct clones. Both were base transitions characteristic of TMZ mutagenesis. Gene expression analysis identified increased JAK-STAT signaling in the gliosarcoma, together with reduced expression of microRNAs known to regulate epithelial-mesenchymal transition. Ex vivo treatment of the GBM spheres with TMZ generated numerous variants in cancer driver genes, including TP53 and CDH1, which were mutated in the post-treatment tumor. CONCLUSIONS: TMZ-induced TP53 gain-of-function mutations can have a driving role in secondary gliosarcoma pathogenesis. Analysis of variants identified in ex vivo TMZ-treated gliomaspheres may have utility in predicting GBM evolutionary trajectories in vivo during standard chemoradiation.

5.
Cancer Res ; 77(20): 5518-5529, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-28855212

RESUMO

Glioblastoma multiforme (GBM) remains a mainly incurable disease in desperate need of more effective treatments. In this study, we develop evidence that the mitotic spindle checkpoint molecule BUB1B may offer a predictive marker for aggressiveness and effective drug response. A subset of GBM tumor isolates requires BUB1B to suppress lethal kinetochore-microtubule attachment defects. Using gene expression data from GBM stem-like cells, astrocytes, and neural progenitor cells that are sensitive or resistant to BUB1B inhibition, we created a computational framework to predict sensitivity to BUB1B inhibition. Applying this framework to tumor expression data from patients, we stratified tumors into BUB1B-sensitive (BUB1BS) or BUB1B-resistant (BUB1BR) subtypes. Through this effort, we found that BUB1BS patients have a significantly worse prognosis regardless of tumor development subtype (i.e., classical, mesenchymal, neural, proneural). Functional genomic profiling of BUB1BR versus BUB1BS isolates revealed a differential reliance of genes enriched in the BUB1BS classifier, including those involved in mitotic cell cycle, microtubule organization, and chromosome segregation. By comparing drug sensitivity profiles, we predicted BUB1BS cells to be more sensitive to type I and II topoisomerase inhibitors, Raf inhibitors, and other drugs, and experimentally validated some of these predictions. Taken together, the results show that our BUB1BR/S classification of GBM tumors can predict clinical course and sensitivity to drug treatment. Cancer Res; 77(20); 5518-29. ©2017 AACR.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/enzimologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Glioblastoma/tratamento farmacológico , Glioblastoma/enzimologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Antineoplásicos Fitogênicos/farmacologia , Biomarcadores Tumorais/antagonistas & inibidores , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral/metabolismo , Etoposídeo/farmacologia , Perfilação da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Irinotecano , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/genética
6.
Stem Cell Reports ; 8(5): 1421-1429, 2017 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-28434940

RESUMO

Characterization of non-neoplastic and malignant human stem cell populations in their native state can provide new insights into gliomagenesis. Here we developed a purification strategy to directly isolate EGFR+/- populations from human germinal matrix (GM) and adult subventricular zone autopsy tissues, and from de novo glioblastoma (GBM) resections, enriching for cells capable of binding EGF ligand (LBEGFR+), and uniquely compared their functional and molecular properties. LBEGFR+ populations in both GM and GBM encompassed all sphere-forming cells and displayed proliferative stem cell properties in vitro. In xenografts, LBEGFR+ GBM cells showed robust tumor initiation and progression to high-grade, infiltrative gliomas. Whole-transcriptome sequencing analysis confirmed enrichment of proliferative pathways in both developing and neoplastic freshly isolated EGFR+ populations, and identified both unique and shared sets of genes. The ability to prospectively isolate stem cell populations using native ligand-binding capacity opens new doors onto understanding both normal human development and tumor cell biology.


Assuntos
Neoplasias Encefálicas/patologia , Proliferação de Células , Glioblastoma/patologia , Células-Tronco Neoplásicas/fisiologia , Células-Tronco Neurais/fisiologia , Animais , Neoplasias Encefálicas/metabolismo , Separação Celular/métodos , Células Cultivadas , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Humanos , Masculino , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/transplante , Células-Tronco Neurais/metabolismo , Cultura Primária de Células/métodos , Transcriptoma , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Oncotarget ; 6(9): 7293-304, 2015 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-25762646

RESUMO

Invasive growth is a major determinant of the high lethality of malignant gliomas. Plexin-B2, an axon guidance receptor important for mediating neural progenitor cell migration during development, is upregulated in gliomas, but its function therein remains poorly understood. Combining bioinformatic analyses, immunoblotting and immunohistochemistry of patient samples, we demonstrate that Plexin-B2 is consistently upregulated in all types of human gliomas and that its expression levels correlate with glioma grade and poor survival. Activation of Plexin-B2 by Sema4C ligand in glioblastoma cells induced actin-based cytoskeletal dynamics and invasive migration in vitro. This proinvasive effect was associated with activation of the cell motility mediators RhoA and Rac1. Furthermore, costimulation of Plexin-B2 and the receptor tyrosine kinase Met led to synergistic Met phosphorylation. In intracranial glioblastoma transplants, Plexin-B2 knockdown hindered invasive growth and perivascular spreading, and resulted in decreased tumor vascularity. Our results demonstrate that Plexin-B2 promotes glioma invasion and vascularization, and they identify Plexin-B2 as a potential novel prognostic marker for glioma malignancy. Targeting the Plexin-B2 pathway may represent a novel therapeutic approach to curtail invasive growth of glioblastoma.


Assuntos
Neoplasias Encefálicas/patologia , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Biomarcadores Tumorais , Linhagem Celular Tumoral , Movimento Celular , Biologia Computacional , GTP Fosfo-Hidrolases/metabolismo , Perfilação da Expressão Gênica , Glioblastoma , Humanos , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Fosforilação , Semaforinas/metabolismo , Regulação para Cima , Quinases Associadas a rho/metabolismo
8.
Nat Commun ; 6: 5903, 2015 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-25582907

RESUMO

Mice have been a long-standing model for human biology and disease. Here we characterize, by RNA sequencing, the transcriptional profiles of a large and heterogeneous collection of mouse tissues, augmenting the mouse transcriptome with thousands of novel transcript candidates. Comparison with transcriptome profiles in human cell lines reveals substantial conservation of transcriptional programmes, and uncovers a distinct class of genes with levels of expression that have been constrained early in vertebrate evolution. This core set of genes captures a substantial fraction of the transcriptional output of mammalian cells, and participates in basic functional and structural housekeeping processes common to all cell types. Perturbation of these constrained genes is associated with significant phenotypes including embryonic lethality and cancer. Evolutionary constraint in gene expression levels is not reflected in the conservation of the genomic sequences, but is associated with conserved epigenetic marking, as well as with characteristic post-transcriptional regulatory programme, in which sub-cellular localization and alternative splicing play comparatively large roles.


Assuntos
Evolução Molecular , Regulação da Expressão Gênica , Transcriptoma , Processamento Alternativo , Animais , Evolução Biológica , Linhagem Celular , Epigênese Genética , Perfilação da Expressão Gênica , Biblioteca Gênica , Genoma , Histonas/química , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Genéticos , Oligonucleotídeos Antissenso , Fenótipo , Análise de Sequência de RNA
9.
Nat Commun ; 5: 5799, 2014 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-25517576

RESUMO

The misrepair of DNA double-strand breaks in close spatial proximity within the nucleus can result in chromosomal rearrangements that are important in the pathogenesis of haematopoietic and solid malignancies. It is unknown why certain epigenetic states, such as those found in stem or progenitor cells, appear to facilitate neoplastic transformation. Here we show that altering the transcriptional state of human astrocytes alters patterns of DNA damage repair from ionizing radiation at a gene locus-specific and genome-wide level. Astrocytes induced into a reactive state exhibit increased DNA repair, compared with non-reactive cells, in actively transcribed chromatin after irradiation. In mapping these repair sites, we identify misrepair events and repair hotspots that are unique to each state. The precise characterization of genomic regions susceptible to mutation in specific transcriptional states provides new opportunities for addressing clonal evolution in solid cancers, in particular those where double-strand break induction is a cornerstone of clinical intervention.


Assuntos
Astrócitos/efeitos da radiação , Transformação Celular Neoplásica/efeitos da radiação , Reparo do DNA , DNA/metabolismo , Transcrição Gênica , Adulto , Animais , Astrócitos/citologia , Astrócitos/metabolismo , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Córtex Cerebral/efeitos da radiação , Cromatina/química , Cromatina/metabolismo , Cromatina/efeitos da radiação , Evolução Clonal , DNA/química , Quebras de DNA de Cadeia Dupla , Feto , Raios gama , Expressão Gênica , Genoma Humano , Histonas/genética , Histonas/metabolismo , Humanos , Macaca fascicularis , Mutação , Cultura Primária de Células
10.
Cell ; 148(1-2): 84-98, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22265404

RESUMO

Higher-order chromosomal organization for transcription regulation is poorly understood in eukaryotes. Using genome-wide Chromatin Interaction Analysis with Paired-End-Tag sequencing (ChIA-PET), we mapped long-range chromatin interactions associated with RNA polymerase II in human cells and uncovered widespread promoter-centered intragenic, extragenic, and intergenic interactions. These interactions further aggregated into higher-order clusters, wherein proximal and distal genes were engaged through promoter-promoter interactions. Most genes with promoter-promoter interactions were active and transcribed cooperatively, and some interacting promoters could influence each other implying combinatorial complexity of transcriptional controls. Comparative analyses of different cell lines showed that cell-specific chromatin interactions could provide structural frameworks for cell-specific transcription, and suggested significant enrichment of enhancer-promoter interactions for cell-specific functions. Furthermore, genetically-identified disease-associated noncoding elements were found to be spatially engaged with corresponding genes through long-range interactions. Overall, our study provides insights into transcription regulation by three-dimensional chromatin interactions for both housekeeping and cell-specific genes in human cells.


Assuntos
Cromatina/metabolismo , Regulação da Expressão Gênica , Regiões Promotoras Genéticas , RNA Polimerase II/metabolismo , Transcrição Gênica , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Elementos Facilitadores Genéticos , Estudo de Associação Genômica Ampla , Humanos
11.
Nat Methods ; 7(7): 528-34, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20543846

RESUMO

Large-scale sequencing projects have revealed an unexpected complexity in the origins, structures and functions of mammalian transcripts. Many loci are known to produce overlapping coding and noncoding RNAs with capped 5' ends that vary in size. Methods to identify the 5' ends of transcripts will facilitate the discovery of new promoters and 5' ends derived from secondary capping events. Such methods often require high input amounts of RNA not obtainable from highly refined samples such as tissue microdissections and subcellular fractions. Therefore, we developed nano-cap analysis of gene expression (nanoCAGE), a method that captures the 5' ends of transcripts from as little as 10 ng of total RNA, and CAGEscan, a mate-pair adaptation of nanoCAGE that captures the transcript 5' ends linked to a downstream region. Both of these methods allow further annotation-agnostic studies of the complex human transcriptome.


Assuntos
Perfilação da Expressão Gênica , Regulação da Expressão Gênica/fisiologia , Nanotecnologia/métodos , Regiões Promotoras Genéticas/fisiologia , RNA/metabolismo , Genoma Humano , Humanos , RNA/genética
12.
Nat Neurosci ; 10(12): 1578-86, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17982450

RESUMO

In humans and many other animals, memory consolidation occurs through multiple temporal phases and usually involves more than one neuroanatomical brain system. Genetic dissection of Pavlovian olfactory learning in Drosophila melanogaster has revealed multiple memory phases, but the predominant view holds that all memory phases occur in mushroom body neurons. Here, we demonstrate an acute requirement for NMDA receptors (NMDARs) outside of the mushroom body during long-term memory (LTM) consolidation. Targeted dsRNA-mediated silencing of Nmdar1 and Nmdar2 (also known as dNR1 or dNR2, respectively) in cholinergic R4m-subtype large-field neurons of the ellipsoid body specifically disrupted LTM consolidation, but not retrieval. Similar silencing of functional NMDARs in the mushroom body disrupted an earlier memory phase, leaving LTM intact. Our results clearly establish an anatomical site outside of the mushroom body involved with LTM consolidation, thus revealing both a distributed brain system subserving olfactory memory formation and the existence of a system-level memory consolidation in Drosophila.


Assuntos
Aprendizagem por Associação/fisiologia , Encéfalo/citologia , Memória/fisiologia , Neurônios/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Análise de Variância , Animais , Animais Geneticamente Modificados , Aprendizagem por Associação/efeitos dos fármacos , Comportamento Animal , Condicionamento Clássico , Drosophila , Proteínas de Drosophila/genética , Aprendizagem em Labirinto , Memória/efeitos dos fármacos , Corpos Pedunculados/efeitos dos fármacos , Corpos Pedunculados/fisiologia , Odorantes , RNA de Cadeia Dupla/farmacologia , Receptores de N-Metil-D-Aspartato/genética , Fatores de Tempo
13.
Genome ; 50(8): 767-77, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17893736

RESUMO

The mammalian CECR2 protein contains a highly conserved bromodomain and forms a chromatin-remodelling complex with the ISWI homologue SNF2L. Mutation of the mouse CECR2 homologue results in a neural tube defect. Here we describe the characterization of the Drosophila melanogaster homologue of CECR2. Originally annotated as 2 genes, dikar and CG32394 now appear to encode both a long dikar/CG32394 transcript homologous to CECR2 and a truncated transcript missing the bromodomain. This truncated transcript may be specific to Diptera, as it is predicted from the genomic sequences of several other dipteran species but it is not predicted in the honey bee, Apis mellifera, and it is not found in mammals. Five different P element-mediated 5' deletions of the Drosophila dikar gene were generated. All mutants were homozygous-viable and the 3 mutants examined further displayed continued, albeit aberrant, transcription of dikar/CG32394. In a previous study, a dikar insertion mutation was associated with long-term memory deficits. However, the 2 deletion mutants tested here showed normal long-term memory, suggesting that the memory deficit associated with the dikar P element insertion is not due to disruption of dikar. No genetic interaction was seen between Iswi and dikar mutations. This study therefore suggests that the lack of a visible phenotype in dikar mutants is due to compensation by a second gene, possibly acf1.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Cromatina/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Deleção de Genes , Peptídeos e Proteínas de Sinalização Intercelular/genética , Animais , Cromatina/metabolismo , Cruzamentos Genéticos , DNA/genética , DNA/isolamento & purificação , DNA Complementar , Proteínas de Drosophila/metabolismo , Homozigoto , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Filogenia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sequência de DNA , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA