Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Oncol ; 18(3): 517-527, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37507199

RESUMO

TWIST1 (TW) is a pro-oncogenic basic helix-loop-helix (bHLH) transcription factor and promotes the hallmark features of malignancy (e.g., cell invasion, cancer cell stemness, and treatment resistance), which contribute to poor prognoses of glioblastoma (GBM). We previously reported that specific TW dimerization motifs regulate unique cellular phenotypes in GBM. For example, the TW:E12 heterodimer increases periostin (POSTN) expression and promotes cell invasion. TW dimer-specific transcriptional regulation requires binding to the regulatory E-box consensus sequences, but alternative bHLH dimers that balance TW dimer activity in regulating pro-oncogenic TW target genes are unknown. We leveraged the ENCODE DNase I hypersensitivity data to identify E-box sites and tethered TW:E12 and TW:TW proteins to validate dimer binding to E-boxes in vitro. Subsequently, TW knockdown revealed a novel TCF4:TCF12 bHLH dimer occupying the same TW E-box site that, when expressed as a tethered TCF4:TCF12 dimer, markedly repressed POSTN expression and extended animal survival. These observations support TCF4:TCF12 as a novel dimer with tumor-suppressor activity in GBM that functions in part through displacement of and/or competitive inhibition of pro-oncogenic TW dimers at E-box sites.


Assuntos
Glioblastoma , Animais , Glioblastoma/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica , Dimerização
2.
bioRxiv ; 2023 Dec 07.
Artigo em Inglês | MEDLINE | ID: mdl-38076912

RESUMO

We report a highly significant correlation in brain proteome changes between Alzheimers disease (AD) and CRND8 APP695NL/F transgenic mice. However, integrating protein changes observed in the CRND8 mice with co-expression networks derived from human AD, reveals both conserved and divergent module changes. For the most highly conserved module (M42, matrisome) we find many proteins accumulate in plaques, cerebrovascular amyloid (CAA), dystrophic processes, or a combination thereof. Overexpression of two M42 proteins, midkine (Mdk) and pleiotrophin (PTN), in CRND8 mice brains leads to increased accumulation of A ß ; in plaques and in CAA; further, recombinant MDK and PTN enhance A ß ; aggregation into amyloid. Multiple M42 proteins, annotated as heparan sulfate binding proteins, bind to fibrillar A ß 42 and a non-human amyloid fibril in vitro. Supporting this binding data, MDK and PTN co-accumulate with transthyretin (TTR) amyloid in the heart and islet amyloid polypeptide (IAPP) amyloid in the pancreas. Our findings establish several critical insights. Proteomic changes in modules observed in human AD brains define an A ß ; amyloid responsome that is well conserved from mouse model to human. Further, distinct amyloid structures may serve as scaffolds, facilitating the co-accumulation of proteins with signaling functions. We hypothesize that this co-accumulation may contribute to downstream pathological sequalae. Overall, this contextualized understanding of proteomic changes and their interplay with amyloid deposition provides valuable insights into the complexity of AD pathogenesis and potential biomarkers and therapeutic targets.

3.
Alzheimers Res Ther ; 14(1): 104, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35897046

RESUMO

BACKGROUND: The S209F variant of Abelson Interactor Protein 3 (ABI3) increases risk for Alzheimer's disease (AD), but little is known about its function in relation to AD pathogenesis. METHODS: Here, we use a mouse model that is deficient in Abi3 locus to study how the loss of function of Abi3 impacts two cardinal neuropathological hallmarks of AD-amyloid ß plaques and tau pathology. Our study employs extensive neuropathological and transcriptomic characterization using transgenic mouse models and adeno-associated virus-mediated gene targeting strategies. RESULTS: Analysis of bulk RNAseq data confirmed age-progressive increase in Abi3 levels in rodent models of AD-type amyloidosis and upregulation in AD patients relative to healthy controls. Using RNAscope in situ hybridization, we localized the cellular distribution of Abi3 in mouse and human brains, finding that Abi3 is expressed in both microglial and non-microglial cells. Next, we evaluated Abi3-/- mice and document that both Abi3 and its overlapping gene, Gngt2, are disrupted in these mice. Using multiple transcriptomic datasets, we show that expression of Abi3 and Gngt2 are tightly correlated in rodent models of AD and human brains, suggesting a tight co-expression relationship. RNAseq of the Abi3-Gngt2-/- mice revealed upregulation of Trem2, Plcg2, and Tyrobp, concomitant with induction of an AD-associated neurodegenerative signature, even in the absence of AD-typical neuropathology. In APP mice, loss of Abi3-Gngt2 resulted in a gene dose- and age-dependent reduction in Aß deposition. Additionally, in Abi3-Gngt2-/- mice, expression of a pro-aggregant form of human tau exacerbated tauopathy and astrocytosis. Further, using in vitro culture assays, we show that the AD-associated S209F mutation alters the extent of ABI3 phosphorylation. CONCLUSIONS: These data provide an important experimental framework for understanding the role of Abi3-Gngt2 function and early inflammatory gliosis in AD. Our studies also demonstrate that inflammatory gliosis could have opposing effects on amyloid and tau pathology, highlighting the unpredictability of targeting immune pathways in AD.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Doença de Alzheimer , Amiloidose , Subunidades gama da Proteína de Ligação ao GTP , Animais , Humanos , Camundongos , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Amiloidose/genética , Encéfalo/metabolismo , Modelos Animais de Doenças , Gliose/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/genética , Glicoproteínas de Membrana/metabolismo , Camundongos Transgênicos , Placa Amiloide/patologia , Receptores Imunológicos/metabolismo , Proteínas tau/genética , Proteínas tau/metabolismo
4.
Mol Neurodegener ; 16(1): 32, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33957936

RESUMO

INTRODUCTION: Passive immunotherapies targeting Aß continue to be evaluated as Alzheimer's disease (AD) therapeutics, but there remains debate over the mechanisms by which these immunotherapies work. Besides the amount of preexisting Aß deposition and the type of deposit (compact or diffuse), there is little data concerning what factors, independent of those intrinsic to the antibody, might influence efficacy. Here we (i) explored how constitutive priming of the underlying innate activation states by Il10 and Il6 might influence passive Aß immunotherapy and (ii) evaluated transcriptomic data generated in the AMP-AD initiative to inform how these two cytokines and their receptors' mRNA levels are altered in human AD and an APP mouse model. METHODS: rAAV2/1 encoding EGFP, Il6 or Il10 were delivered by somatic brain transgenesis to neonatal (P0) TgCRND8 APP mice. Then, at 2 months of age, the mice were treated bi-weekly with a high-affinity anti-Aß1-16 mAb5 monoclonal antibody or control mouse IgG until 6 months of age. rAAV mediated transgene expression, amyloid accumulation, Aß levels and gliosis were assessed. Extensive transcriptomic data was used to evaluate the mRNA expression levels of IL10 and IL6 and their receptors in the postmortem human AD temporal cortex and in the brains of TgCRND8 mice, the later at multiple ages. RESULTS: Priming TgCRND8 mice with Il10 increases Aß loads and blocks efficacy of subsequent mAb5 passive immunotherapy, whereas priming with Il6 priming reduces Aß loads by itself and subsequent Aß immunotherapy shows only a slightly additive effect. Transcriptomic data shows that (i) there are significant increases in the mRNA levels of Il6 and Il10 receptors in the TgCRND8 mouse model and temporal cortex of humans with AD and (ii) there is a great deal of variance in individual mouse brain and the human temporal cortex of these interleukins and their receptors. CONCLUSIONS: The underlying immune activation state can markedly affect the efficacy of passive Aß immunotherapy. These results have important implications for ongoing human AD immunotherapy trials, as they indicate that underlying immune activation states within the brain, which may be highly variable, may influence the ability for passive immunotherapy to alter Aß deposition.


Assuntos
Doença de Alzheimer/imunologia , Peptídeos beta-Amiloides/antagonistas & inibidores , Anticorpos Monoclonais/farmacologia , Imunidade Inata/efeitos dos fármacos , Imunização Passiva/métodos , Animais , Humanos , Interleucina-10/imunologia , Interleucina-6/imunologia , Camundongos , Camundongos Transgênicos
5.
Alzheimers Dement ; 17(6): 984-1004, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33480174

RESUMO

Intron retention (IR) has been implicated in the pathogenesis of complex diseases such as cancers; its association with Alzheimer's disease (AD) remains unexplored. We performed genome-wide analysis of IR through integrating genetic, transcriptomic, and proteomic data of AD subjects and mouse models from the Accelerating Medicines Partnership-Alzheimer's Disease project. We identified 4535 and 4086 IR events in 2173 human and 1736 mouse genes, respectively. Quantitation of IR enabled the identification of differentially expressed genes that conventional exon-level approaches did not reveal. There were significant correlations of intron expression within innate immune genes, like HMBOX1, with AD in humans. Peptides with a high probability of translation from intron-retained mRNAs were identified using mass spectrometry. Further, we established AD-specific intron expression Quantitative Trait Loci, and identified splicing-related genes that may regulate IR. Our analysis provides a novel resource for the search for new AD biomarkers and pathological mechanisms.


Assuntos
Doença de Alzheimer , Autopsia , Encéfalo/patologia , Modelos Animais de Doenças , Genômica , Íntrons/genética , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Animais , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Proteômica , Locos de Características Quantitativas , Transcriptoma
6.
BMC Genomics ; 21(1): 128, 2020 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-32028886

RESUMO

BACKGROUND: Intron retention (IR) has been traditionally overlooked as 'noise' and received negligible attention in the field of gene expression analysis. In recent years, IR has become an emerging field for interrogating transcriptomes because it has been recognized to carry out important biological functions such as gene expression regulation and it has been found to be associated with complex diseases such as cancers. However, methods for detecting IR today are limited. Thus, there is a need to develop novel methods to improve IR detection. RESULTS: Here we present iREAD (intron REtention Analysis and Detector), a tool to detect IR events genome-wide from high-throughput RNA-seq data. The command line interface for iREAD is implemented in Python. iREAD takes as input a BAM file, representing the transcriptome, and a text file containing the intron coordinates of a genome. It then 1) counts all reads that overlap intron regions, 2) detects IR events by analyzing the features of reads such as depth and distribution patterns, and 3) outputs a list of retained introns into a tab-delimited text file. iREAD provides significant added value in detecting IR compared with output from IRFinder with a higher AUC on all datasets tested. Both methods showed low false positive rates and high false negative rates in different regimes, indicating that use together is generally beneficial. The output from iREAD can be directly used for further exploratory analysis such as differential intron expression and functional enrichment. The software is freely available at https://github.com/genemine/iread. CONCLUSION: Being complementary to existing tools, iREAD provides a new and generic tool to interrogate poly-A enriched transcriptomic data of intron regions. Intron retention analysis provides a complementary approach for understanding transcriptome.


Assuntos
Íntrons , RNA-Seq , Software , Algoritmos , Animais , Humanos , Camundongos
7.
Neuron ; 99(1): 64-82.e7, 2018 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-29937276

RESUMO

Investigators have long suspected that pathogenic microbes might contribute to the onset and progression of Alzheimer's disease (AD) although definitive evidence has not been presented. Whether such findings represent a causal contribution, or reflect opportunistic passengers of neurodegeneration, is also difficult to resolve. We constructed multiscale networks of the late-onset AD-associated virome, integrating genomic, transcriptomic, proteomic, and histopathological data across four brain regions from human post-mortem tissue. We observed increased human herpesvirus 6A (HHV-6A) and human herpesvirus 7 (HHV-7) from subjects with AD compared with controls. These results were replicated in two additional, independent and geographically dispersed cohorts. We observed regulatory relationships linking viral abundance and modulators of APP metabolism, including induction of APBB2, APPBP2, BIN1, BACE1, CLU, PICALM, and PSEN1 by HHV-6A. This study elucidates networks linking molecular, clinical, and neuropathological features with viral activity and is consistent with viral activity constituting a general feature of AD.


Assuntos
Doença de Alzheimer/virologia , Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/virologia , Encefalite Viral/virologia , Herpesvirus Humano 6 , Herpesvirus Humano 7 , Infecções por Roseolovirus/virologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Secretases da Proteína Precursora do Amiloide/genética , Animais , Ácido Aspártico Endopeptidases/genética , Encéfalo/metabolismo , Encéfalo/patologia , Estudos de Casos e Controles , Clusterina/genética , Estudos de Coortes , Encefalite Viral/genética , Encefalite Viral/metabolismo , Encefalite Viral/patologia , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Genômica , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , MicroRNAs/genética , Microbiota , Proteínas Monoméricas de Montagem de Clatrina/genética , Proteínas Nucleares/genética , Presenilina-1/genética , Proteômica , Infecções por Roseolovirus/genética , Infecções por Roseolovirus/metabolismo , Infecções por Roseolovirus/patologia , Proteínas Supressoras de Tumor/genética , Carga Viral
8.
Mol Oncol ; 12(7): 1188-1202, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29754406

RESUMO

TWIST1 (TW) is a bHLH transcription factor (TF) and master regulator of the epithelial-to-mesenchymal transition (EMT). In vitro, TW promotes mesenchymal change, invasion, and self-renewal in glioblastoma (GBM) cells. However, the potential therapeutic relevance of TW has not been established through loss-of-function studies in human GBM cell xenograft models. The effects of TW loss of function (gene editing and knockdown) on inhibition of tumorigenicity of U87MG and GBM4 glioma stem cells were tested in orthotopic xenograft models and conditional knockdown in established flank xenograft tumors. RNAseq and the analysis of tumors investigated putative TW-associated mechanisms. Multiple bioinformatic tools revealed significant alteration of ECM, membrane receptors, signaling transduction kinases, and cytoskeleton dynamics leading to identification of PI3K/AKT signaling. We experimentally show alteration of AKT activity and periostin (POSTN) expression in vivo and/or in vitro. For the first time, we show that effect of TW knockout inhibits AKT activity in U87MG cells in vivo independent of PTEN mutation. The clinical relevance of TW and candidate mechanisms was established by analysis of the TCGA and ENCODE databases. TW expression was associated with decreased patient survival and LASSO regression analysis identified POSTN as one of top targets of TW in human GBM. While we previously demonstrated the role of TW in promoting EMT and invasion of glioma cells, these studies provide direct experimental evidence supporting protumorigenic role of TW independent of invasion in vivo and the therapeutic relevance of targeting TW in human GBM. Further, the role of TW driving POSTN expression and AKT signaling suggests actionable targets, which could be leveraged to mitigate the oncogenic effects of TW in GBM.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Neoplasias Encefálicas/genética , Carcinogênese/genética , Carcinogênese/patologia , Moléculas de Adesão Celular/metabolismo , Linhagem Celular Tumoral , Edição de Genes , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/genética , Humanos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas Nucleares/genética , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Análise de Sobrevida , Proteína 1 Relacionada a Twist/genética
9.
Cell Syst ; 4(5): 516-529.e7, 2017 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-28365151

RESUMO

We present a systems strategy that facilitated the development of a molecular signature for glioblastoma (GBM), composed of 33 cell-surface transmembrane proteins. This molecular signature, GBMSig, was developed through the integration of cell-surface proteomics and transcriptomics from patient tumors in the REMBRANDT (n = 228) and TCGA datasets (n = 547) and can separate GBM patients from control individuals with a Matthew's correlation coefficient value of 0.87 in a lock-down test. Functionally, 17/33 GBMSig proteins are associated with transforming growth factor ß signaling pathways, including CD47, SLC16A1, HMOX1, and MRC2. Knockdown of these genes impaired GBM invasion, reflecting their role in disease-perturbed changes in GBM. ELISA assays for a subset of GBMSig (CD44, VCAM1, HMOX1, and BIGH3) on 84 plasma specimens from multiple clinical sites revealed a high degree of separation of GBM patients from healthy control individuals (area under the curve is 0.98 in receiver operating characteristic). In addition, a classifier based on these four proteins differentiated the blood of pre- and post-tumor resections, demonstrating potential clinical value as biomarkers.


Assuntos
Perfilação da Expressão Gênica/métodos , Glioblastoma/metabolismo , Proteínas de Membrana/metabolismo , Biomarcadores Tumorais , Neoplasias Encefálicas/genética , Diferenciação Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Proliferação de Células , Biologia Computacional/métodos , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/genética , Humanos , Proteínas de Membrana/genética , Proteômica/métodos , Biologia de Sistemas/métodos , Transcriptoma/genética , Fator de Crescimento Transformador beta/metabolismo
10.
PLoS One ; 12(3): e0172884, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28264064

RESUMO

Ion channels and transporters have increasingly recognized roles in cancer progression through the regulation of cell proliferation, migration, and death. Glioblastoma stem-like cells (GSCs) are a source of tumor formation and recurrence in glioblastoma multiforme, a highly aggressive brain cancer, suggesting that ion channel expression may be perturbed in this population. However, little is known about the expression and functional relevance of ion channels that may contribute to GSC malignancy. Using RNA sequencing, we assessed the enrichment of ion channels in GSC isolates and non-tumor neural cell types. We identified a unique set of GSC-enriched ion channels using differential expression analysis that is also associated with distinct gene mutation signatures. In support of potential clinical relevance, expression of selected GSC-enriched ion channels evaluated in human glioblastoma databases of The Cancer Genome Atlas and Ivy Glioblastoma Atlas Project correlated with patient survival times. Finally, genetic knockdown as well as pharmacological inhibition of individual or classes of GSC-enriched ion channels constrained growth of GSCs compared to normal neural stem cells. This first-in-kind global examination characterizes ion channels enriched in GSCs and explores their potential clinical relevance to glioblastoma molecular subtypes, gene mutations, survival outcomes, regional tumor expression, and experimental responses to loss-of-function. Together, the data support the potential biological and therapeutic impact of ion channels on GSC malignancy and provide strong rationale for further examination of their mechanistic and therapeutic importance.


Assuntos
Neoplasias Encefálicas/genética , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Canais Iônicos/genética , Células-Tronco Neoplásicas/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/terapia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Análise por Conglomerados , Junções Comunicantes/genética , Junções Comunicantes/metabolismo , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/terapia , Humanos , Canais Iônicos/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Prognóstico , Transdução de Sinais , Análise de Sobrevida , Transcriptoma , Resultado do Tratamento
11.
Immunity ; 43(5): 933-44, 2015 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-26588779

RESUMO

Mutations in ADAR, which encodes the ADAR1 RNA-editing enzyme, cause Aicardi-Goutières syndrome (AGS), a severe autoimmune disease associated with an aberrant type I interferon response. How ADAR1 prevents autoimmunity remains incompletely defined. Here, we demonstrate that ADAR1 is a specific and essential negative regulator of the MDA5-MAVS RNA sensing pathway. Moreover, we uncovered a MDA5-MAVS-independent function for ADAR1 in the development of multiple organs. We showed that the p150 isoform of ADAR1 uniquely regulated the MDA5 pathway, whereas both the p150 and p110 isoforms contributed to development. Abrupt deletion of ADAR1 in adult mice revealed that both of these functions were required throughout life. Our findings delineate genetically separable roles for both ADAR1 isoforms in vivo, with implications for the human diseases caused by ADAR mutations.


Assuntos
Adenosina Desaminase/metabolismo , Autoimunidade/fisiologia , RNA Helicases DEAD-box/metabolismo , Isoformas de Proteínas/metabolismo , Edição de RNA/fisiologia , RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Doenças Autoimunes do Sistema Nervoso/metabolismo , Células HEK293 , Humanos , Interferon Tipo I/metabolismo , Helicase IFIH1 Induzida por Interferon , Camundongos , Malformações do Sistema Nervoso/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/fisiologia
12.
Neuron ; 85(3): 519-33, 2015 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-25619653

RESUMO

Anti-inflammatory strategies are proposed to have beneficial effects in Alzheimer's disease. To explore how anti-inflammatory cytokine signaling affects Aß pathology, we investigated the effects of adeno-associated virus (AAV2/1)-mediated expression of Interleukin (IL)-10 in the brains of APP transgenic mouse models. IL-10 expression resulted in increased Aß accumulation and impaired memory in APP mice. A focused transcriptome analysis revealed changes consistent with enhanced IL-10 signaling and increased ApoE expression in IL-10-expressing APP mice. ApoE protein was selectively increased in the plaque-associated insoluble cellular fraction, likely because of direct interaction with aggregated Aß in the IL-10-expressing APP mice. Ex vivo studies also show that IL-10 and ApoE can individually impair glial Aß phagocytosis. Our observations that IL-10 has an unexpected negative effect on Aß proteostasis and cognition in APP mouse models demonstrate the complex interplay between innate immunity and proteostasis in neurodegenerative diseases, an interaction we call immunoproteostasis.


Assuntos
Precursor de Proteína beta-Amiloide , Transtornos Cognitivos/metabolismo , Imunoproteínas/biossíntese , Interleucina-10/biossíntese , Placa Amiloide/metabolismo , Deficiências na Proteostase/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animais , Animais Recém-Nascidos , Células Cultivadas , Transtornos Cognitivos/imunologia , Células HEK293 , Humanos , Camundongos , Camundongos Transgênicos , Placa Amiloide/imunologia , Deficiências na Proteostase/imunologia
13.
Mol Cell Proteomics ; 13(10): 2618-31, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24997998

RESUMO

Glioblastoma multiforme is a highly invasive and aggressive brain tumor with an invariably poor prognosis. The overexpression of epidermal growth factor receptor (EGFR) is a primary influencer of invasion and proliferation in tumor cells and the constitutively active EGFRvIII mutant, found in 30-65% of Glioblastoma multiforme, confers more aggressive invasion. To better understand how EGFR contributes to tumor aggressiveness, we investigated the effect of EGFR on the secreted levels of 65 rationally selected proteins involved in invasion. We employed selected reaction monitoring targeted mass spectrometry using stable isotope labeled internal peptide standards to quantity proteins in the secretome from five GBM (U87) isogenic cell lines in which EGFR, EGFRvIII, and/or PTEN were expressed. Our results show that cell lines with EGFR overexpression and constitutive EGFRvIII expression differ remarkably in the expression profiles for both secreted and intracellular signaling proteins, and alterations in EGFR signaling result in reproducible changes in concentrations of secreted proteins. Furthermore, the EGFRvIII-expressing mutant cell line secretes the majority of the selected invasion-promoting proteins at higher levels than other cell lines tested. Additionally, the intracellular and extracellular protein measurements indicate elevated oxidative stress in the EGFRvIII-expressing cell line. In conclusion, the results of our study demonstrate that EGFR signaling has a significant effect on the levels of secreted invasion-promoting proteins, likely contributing to the aggressiveness of Glioblastoma multiforme. Further characterization of these proteins may provide candidates for new therapeutic strategies and targets as well as biomarkers for this aggressive disease.


Assuntos
Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Glioblastoma/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteômica/métodos , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais
14.
PLoS One ; 8(10): e76694, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24146911

RESUMO

Astrocytoma is the most common glioma, accounting for half of all primary brain and spinal cord tumors. Late detection and the aggressive nature of high-grade astrocytomas contribute to high mortality rates. Though many studies identify candidate biomarkers using high-throughput transcriptomic profiling to stratify grades and subtypes, few have resulted in clinically actionable results. This shortcoming can be attributed, in part, to pronounced lab effects that reduce signature robustness and varied individual gene expression among patients with the same tumor. We addressed these issues by uniformly preprocessing publicly available transcriptomic data, comprising 306 tumor samples from three astrocytoma grades (Grade 2, 3, and 4) and 30 non-tumor samples (normal brain as control tissues). Utilizing Differential Rank Conservation (DIRAC), a network-based classification approach, we examined the global and individual patterns of network regulation across tumor grades. Additionally, we applied gene-based approaches to identify genes whose expression changed consistently with increasing tumor grade and evaluated their robustness across multiple studies using statistical sampling. Applying DIRAC, we observed a global trend of greater network dysregulation with increasing tumor aggressiveness. Individual networks displaying greater differences in regulation between adjacent grades play well-known roles in calcium/PKC, EGF, and transcription signaling. Interestingly, many of the 90 individual genes found to monotonically increase or decrease with astrocytoma grade are implicated in cancer-affected processes such as calcium signaling, mitochondrial metabolism, and apoptosis. The fact that specific genes monotonically increase or decrease with increasing astrocytoma grade may reflect shared oncogenic mechanisms among phenotypically similar tumors. This work presents statistically significant results that enable better characterization of different human astrocytoma grades and hopefully can contribute towards improvements in diagnosis and therapy choices. Our results also identify a number of testable hypotheses relating to astrocytoma etiology that may prove helpful in developing much-needed biomarkers for earlier disease detection.


Assuntos
Astrocitoma/genética , Astrocitoma/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Perfilação da Expressão Gênica , Heterogeneidade Genética , Algoritmos , Carcinogênese/genética , Carcinogênese/patologia , Bases de Dados Genéticas , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Genes Neoplásicos , Humanos , Gradação de Tumores , Invasividade Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos
15.
PLoS Comput Biol ; 9(7): e1003148, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23935471

RESUMO

We utilized abundant transcriptomic data for the primary classes of brain cancers to study the feasibility of separating all of these diseases simultaneously based on molecular data alone. These signatures were based on a new method reported herein--Identification of Structured Signatures and Classifiers (ISSAC)--that resulted in a brain cancer marker panel of 44 unique genes. Many of these genes have established relevance to the brain cancers examined herein, with others having known roles in cancer biology. Analyses on large-scale data from multiple sources must deal with significant challenges associated with heterogeneity between different published studies, for it was observed that the variation among individual studies often had a larger effect on the transcriptome than did phenotype differences, as is typical. For this reason, we restricted ourselves to studying only cases where we had at least two independent studies performed for each phenotype, and also reprocessed all the raw data from the studies using a unified pre-processing pipeline. We found that learning signatures across multiple datasets greatly enhanced reproducibility and accuracy in predictive performance on truly independent validation sets, even when keeping the size of the training set the same. This was most likely due to the meta-signature encompassing more of the heterogeneity across different sources and conditions, while amplifying signal from the repeated global characteristics of the phenotype. When molecular signatures of brain cancers were constructed from all currently available microarray data, 90% phenotype prediction accuracy, or the accuracy of identifying a particular brain cancer from the background of all phenotypes, was found. Looking forward, we discuss our approach in the context of the eventual development of organ-specific molecular signatures from peripheral fluids such as the blood.


Assuntos
Neoplasias Encefálicas/genética , Transcriptoma , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Biologia Computacional , Humanos , Reprodutibilidade dos Testes
16.
Mol Endocrinol ; 25(10): 1699-709, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21868451

RESUMO

The estrogen receptor α (ERα) is a master regulator of gene expression and works along with cooperating transcription factors in mediating the actions of the hormone estradiol (E2) in ER-positive tissues and breast tumors. Here, we report that expression of paired-like homeodomain transcription factor (PITX1), a tumor suppressor and member of the homeobox family of transcription factors, is robustly up-regulated by E2 in several ERα-positive breast cancer cell lines via ERα-dependent interaction between the proximal promoter and an enhancer region 5' upstream of the PITX1 gene. Overexpression of PITX1 selectively inhibited the transcriptional activity of ERα and ERß, while enhancing the activities of the glucocorticoid receptor and progesterone receptor. Reduction of PITX1 by small interfering RNA enhanced ERα-dependent transcriptional regulation of a subset of ERα target genes. The consensus PITX1 binding motif was found to be present in 28% of genome-wide ERα binding sites and was in close proximity to estrogen response elements in a subset of ERα binding sites, and E2 treatment enhanced PITX1 as well as ERα recruitment to these binding sites. These studies identify PITX1 as a new ERα transcriptional target that acts as a repressor to coordinate and fine tune target-specific, ERα-mediated transcriptional activity in human breast cancer cells.


Assuntos
Neoplasias da Mama/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Fatores de Transcrição Box Pareados/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Cromossomos Humanos/metabolismo , Elementos Facilitadores Genéticos/genética , Feminino , Genoma Humano/genética , Humanos , Conformação de Ácido Nucleico , Fatores de Transcrição Box Pareados/química , Fatores de Transcrição Box Pareados/genética , Ligação Proteica/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
17.
Discov Med ; 10(55): 531-42, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21189224

RESUMO

The search for improved molecular cancer diagnostics is a challenge for which systems approaches show great promise. As is becoming increasingly clear, cancer is a perpetually-evolving, highly multi-factorial disease. With next generation sequencing providing an ever-increasing amount of high-throughput data, the need for analytical tools that can provide meaningful context is critical. Systems approaches have demonstrated an ability to separate meaningful signal from noise that arises from population heterogeneity, heterogeneity within and across tumors, and multiple sources of technical variation when sufficient sample sizes are obtained and standardized measurement technologies are used. The ability to develop clinically useful molecular cancer diagnostics will be predicated on advancements on two major fronts: 1) more comprehensive and accurate measurements of multiple endpoints, and 2) more sophisticated analytical tools that synthesize high-throughput data into meaningful reflections of cellular states. To this end, systems approaches that have integrated transcriptomic data onto biomolecular networks have shown promise in their ability to classify tumor subtypes, predict clinical progression, and inform treatment options. Ultimately, the success of systems approaches will be measured by their ability to develop molecular cancer diagnostics through distilling complex, systems-wide information into actionable information in the clinic.


Assuntos
Neoplasias/diagnóstico , Patologia Molecular/métodos , Humanos
18.
Mol Endocrinol ; 20(3): 491-502, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16306086

RESUMO

Estrogenic hormones are classically thought to exert their effects by binding to nuclear estrogen receptors and altering target gene transcription, but estrogens can also have nongenomic effects through rapid activation of membrane-initiated kinase cascades. The development of ligands that selectively activate only the nongenomic pathways would provide useful tools to investigate the significance of these pathways. We have prepared large, abiotic, nondegradable poly(amido)amine dendrimer macromolecules that are conjugated to multiple estrogen molecules through chemically robust linkages. Because of their charge and size, these estrogen-dendrimer conjugates (EDCs) remain outside the nucleus. They stimulate ERK, Shc, and Src phosphorylation in MCF-7 breast cancer cells at low concentrations, yet they are very ineffective in stimulating transcription of endogenous estrogen target genes, being approximately 10,000-fold less potent than estradiol in genomic actions. In contrast to estradiol, EDC was not effective in stimulating breast cancer cell proliferation. Because these EDC ligands activate nongenomic activity at concentrations at which they do not alter the transcription of estrogen target genes, they should be useful in studying extranuclear initiated pathways of estrogen action in a variety of target cells.


Assuntos
Estrogênios/metabolismo , Estrogênios/farmacologia , Poliaminas/farmacologia , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ligação Competitiva , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Membrana Celular/metabolismo , Proliferação de Células , Citoplasma/metabolismo , Dendrímeros , Relação Dose-Resposta a Droga , Estradiol/metabolismo , Estradiol/farmacologia , Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Genoma/efeitos dos fármacos , Humanos , Fosforilação , Poliaminas/química , Poliaminas/metabolismo , Receptores de Estrogênio/metabolismo , Rodaminas/química , Proteínas Adaptadoras da Sinalização Shc , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Células Tumorais Cultivadas , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
19.
Proc Natl Acad Sci U S A ; 102(37): 13153-7, 2005 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-16141343

RESUMO

The nuclear receptor corepressor N-CoR plays a crucial role in the repressive activity of diverse transcription factors, yet little is known about what regulates its cellular level. We have found that estrogen markedly down-regulates N-CoR protein levels in estrogen receptor (ER)-positive breast cancer cells without affecting N-CoR mRNA levels, whereas levels of the related corepressor SMRT are unaffected. This effect is attributable to estrogen up-regulation of the ubiquitin ligase Siah2, which is a rapid and primary transcriptional response mediated by the ER, and precedes the loss of N-CoR. Treatment with proteasomal inhibitor or with small interfering RNA against Siah2 prevented the down-regulation of N-CoR by estrogen. Furthermore, the expression of 24-hydroxylase, a gene repressed by unliganded vitamin D receptor through its interaction with N-CoR, was up-regulated by estrogen and required Siah2. Our results illustrate a mechanism by which the estrogen-ER complex markedly reduces the level of N-CoR through a process involving the up-regulation of Siah2 and the subsequent targeting of N-CoR for proteasomal degradation. These findings reveal that, although estrogen directly regulates the transcription of many genes, by regulating a gene such as Siah2 it can exert profound "secondary" effects on cellular activity through mechanisms such as targeting regulatory proteins for degradation. This estrogen-evoked down-regulation of N-CoR could have a global derepressive effect on genes whose repression depends on N-CoR and thereby have broad impact on the activity of transcription factors and nuclear receptors whose actions involve N-CoR.


Assuntos
Regulação para Baixo , Estrogênios/fisiologia , Proteínas Nucleares/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Linhagem Celular Tumoral , Cisteína Endopeptidases/metabolismo , Estrogênios/farmacologia , Feminino , Humanos , Proteínas Nucleares/análise , Proteínas Nucleares/metabolismo , Correpressor 1 de Receptor Nuclear , RNA Mensageiro/análise , Receptores de Estrogênio/metabolismo , Proteínas Repressoras/análise , Proteínas Repressoras/metabolismo , Fatores de Transcrição/fisiologia , Ubiquitina-Proteína Ligases , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA