Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 2023 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-37433968

RESUMEN

The development of Alzheimer's disease (AD) involves central and peripheral immune deregulation. Gene identification and studies of AD genetic variants of peripheral immune components may aid understanding of peripheral-central immune crosstalk and facilitate new opportunities for therapeutic intervention. In this study, we have identified in a Flanders-Belgian family a novel variant p.E317D in the Toll-like receptor 9 gene (TLR9), co-segregating with EOAD in an autosomal dominant manner. In human, TLR9 is an essential innate and adaptive immune component predominantly expressed in peripheral immune cells. The p.E317D variant caused 50% reduction in TLR9 activation in the NF-κB luciferase assay suggesting that p.E317D is a loss-of-function mutation. Cytokine profiling of human PBMCs upon TLR9 activation revealed a predominantly anti-inflammatory response in contrast to the inflammatory responses from TLR7/8 activation. The cytokines released upon TLR9 activation suppressed inflammation and promoted phagocytosis of Aß42 oligomers in human iPSC-derived microglia. Transcriptome analysis identified upregulation of AXL, RUBICON and associated signaling pathways, which may underline the effects of TLR9 signaling-induced cytokines in regulating the inflammatory status and phagocytic property of microglia. Our data suggest a protective role of TLR9 signaling in AD pathogenesis, and we propose that TLR9 loss-of-function may disrupt a peripheral-central immune crosstalk that promotes dampening of inflammation and clearance of toxic protein species, leading to the build-up of neuroinflammation and pathogenic protein aggregates in AD development.

2.
Allergy ; 76(6): 1825-1835, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33150583

RESUMEN

BACKGROUND: Atabecestat is an orally administered BACE inhibitor developed to treat Alzheimer's disease. Elevations in hepatic enzymes were detected in a number of in trial patients, which resulted in termination of the drug development programme. Immunohistochemical characterization of liver tissue from an index case of atabecestat-mediated liver injury revealed an infiltration of T-lymphocytes in areas of hepatocellular damage. This coupled with the fact that liver injury had a delayed onset suggests that the adaptive immune system may be involved in the pathogenesis. The aim of this study was to generate and characterize atabecestat(metabolite)-responsive T-cell clones from patients with liver injury. METHODS: Peripheral blood mononuclear cells were cultured with atabecestat and its metabolites (diaminothiazine [DIAT], N-acetyl DIAT & epoxide) and cloning was attempted in a number of patients. Atabecestat(metabolite)-responsive clones were analysed in terms of T-cell phenotype, function, pathways of T-cell activation and cross-reactivity with structurally related compounds. RESULTS: CD4+ T-cell clones activated with the DIAT metabolite were detected in 5 out of 8 patients (up to 4.5% cloning efficiency). Lower numbers of CD4+ and CD8+ clones displayed reactivity against atabecestat. Clones proliferated and secreted IFN-γ, IL-13 and cytolytic molecules following atabecestat or DIAT stimulation. Certain atabecestat and DIAT-responsive clones cross-reacted with N-acetyl DIAT; however, no cross-reactivity was observed between atabecestat and DIAT. CD4+ clones were activated through a direct, reversible compound-HLA class II interaction with no requirement for protein processing. CONCLUSION: The detection of atabecestat metabolite-responsive T-cell clones activated via a pharmacological interactions pathway in patients with liver injury is indicative of an immune-based mechanism for the observed hepatic enzyme elevations.


Asunto(s)
Preparaciones Farmacéuticas , Linfocitos T , Linfocitos T CD4-Positivos , Células Clonales , Humanos , Leucocitos Mononucleares , Hígado , Activación de Linfocitos , Piridinas , Tiazinas
3.
Int J Mol Sci ; 17(4): 584, 2016 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-27092500

RESUMEN

Accurate prediction of the potential hepatotoxic nature of new pharmaceuticals remains highly challenging. Therefore, novel in vitro models with improved external validity are needed to investigate hepatic metabolism and timely identify any toxicity of drugs in humans. In this study, we examined the effects of diclofenac, as a model substance with a known risk of hepatotoxicity in vivo, in a dynamic multi-compartment bioreactor using primary human liver cells. Biotransformation pathways of the drug and possible effects on metabolic activities, morphology and cell transcriptome were evaluated. Formation rates of diclofenac metabolites were relatively stable over the application period of seven days in bioreactors exposed to 300 µM diclofenac (300 µM bioreactors (300 µM BR)), while in bioreactors exposed to 1000 µM diclofenac (1000 µM BR) metabolite concentrations declined drastically. The biochemical data showed a significant decrease in lactate production and for the higher dose a significant increase in ammonia secretion, indicating a dose-dependent effect of diclofenac application. The microarray analyses performed revealed a stable hepatic phenotype of the cells over time and the observed transcriptional changes were in line with functional readouts of the system. In conclusion, the data highlight the suitability of the bioreactor technology for studying the hepatotoxicity of drugs in vitro.


Asunto(s)
Antiinflamatorios no Esteroideos/toxicidad , Reactores Biológicos , Técnicas de Cultivo de Célula/instrumentación , Diclofenaco/toxicidad , Hepatocitos/efectos de los fármacos , Pruebas de Toxicidad/instrumentación , Antiinflamatorios no Esteroideos/metabolismo , Células Cultivadas , Diclofenaco/metabolismo , Diseño de Equipo , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Transcriptoma
4.
Mol Neurodegener ; 19(1): 37, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38654375

RESUMEN

BACKGROUND: Microglia play important roles in maintaining brain homeostasis and neurodegeneration. The discovery of genetic variants in genes predominately or exclusively expressed in myeloid cells, such as Apolipoprotein E (APOE) and triggering receptor expressed on myeloid cells 2 (TREM2), as the strongest risk factors for Alzheimer's disease (AD) highlights the importance of microglial biology in the brain. The sequence, structure and function of several microglial proteins are poorly conserved across species, which has hampered the development of strategies aiming to modulate the expression of specific microglial genes. One way to target APOE and TREM2 is to modulate their expression using antisense oligonucleotides (ASOs). METHODS: In this study, we identified, produced, and tested novel, selective and potent ASOs for human APOE and TREM2. We used a combination of in vitro iPSC-microglia models, as well as microglial xenotransplanted mice to provide proof of activity in human microglial in vivo. RESULTS: We proved their efficacy in human iPSC microglia in vitro, as well as their pharmacological activity in vivo in a xenografted microglia model. We demonstrate ASOs targeting human microglia can modify their transcriptional profile and their response to amyloid-ß plaques in vivo in a model of AD. CONCLUSIONS: This study is the first proof-of-concept that human microglial can be modulated using ASOs in a dose-dependent manner to manipulate microglia phenotypes and response to neurodegeneration in vivo.


Asunto(s)
Enfermedad de Alzheimer , Microglía , Oligonucleótidos Antisentido , Microglía/metabolismo , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/genética , Humanos , Oligonucleótidos Antisentido/farmacología , Animales , Ratones , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/genética , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/genética , Células Madre Pluripotentes Inducidas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Modelos Animales de Enfermedad
5.
Nucleic Acids Res ; 39(12): e79, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21486749

RESUMEN

Cost-effective oligonucleotide genotyping arrays like the Affymetrix SNP 6.0 are still the predominant technique to measure DNA copy number variations (CNVs). However, CNV detection methods for microarrays overestimate both the number and the size of CNV regions and, consequently, suffer from a high false discovery rate (FDR). A high FDR means that many CNVs are wrongly detected and therefore not associated with a disease in a clinical study, though correction for multiple testing takes them into account and thereby decreases the study's discovery power. For controlling the FDR, we propose a probabilistic latent variable model, 'cn.FARMS', which is optimized by a Bayesian maximum a posteriori approach. cn.FARMS controls the FDR through the information gain of the posterior over the prior. The prior represents the null hypothesis of copy number 2 for all samples from which the posterior can only deviate by strong and consistent signals in the data. On HapMap data, cn.FARMS clearly outperformed the two most prevalent methods with respect to sensitivity and FDR. The software cn.FARMS is publicly available as a R package at http://www.bioinf.jku.at/software/cnfarms/cnfarms.html.


Asunto(s)
Variaciones en el Número de Copia de ADN , Modelos Estadísticos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Programas Informáticos , Algoritmos , Alelos , Biología Computacional , Polimorfismo de Nucleótido Simple
6.
Arthritis Rheumatol ; 75(5): 673-684, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36409582

RESUMEN

OBJECTIVE: CD4+ T cells are implicated in rheumatoid arthritis (RA) pathology from the strong association between RA and certain HLA class II gene variants. This study was undertaken to examine the synovial T cell receptor (TCR) repertoire, T cell phenotypes, and T cell specificities in small joints of RA patients at time of diagnosis before therapeutic intervention. METHODS: Sixteen patients, of whom 11 patients were anti-citrullinated protein antibody (ACPA)-positive and 5 patients were ACPA-, underwent ultrasound-guided synovial biopsy of a small joint (n = 13) or arthroscopic synovial biopsy of a large joint (n = 3), followed by direct sorting of single T cells for paired sequencing of the αß TCR together with flow cytometry analysis. TCRs from expanded CD4+ T cell clones of 4 patients carrying an HLA-DRB1*04:01 allele were artificially reexpressed to study antigen specificity. RESULTS: T cell analysis demonstrated CD4+ dominance and the presence of peripheral helper T-like cells in both patient groups. We identified >4,000 unique TCR sequences, as well as 225 clonal expansions. Additionally, T cells with double α-chains were a recurring feature. We identified a biased gene usage of the Vß chain segment TRBV20-1 in CD4+ cells from ACPA+ patients. In vitro stimulation of T cell lines expressing selected TCRs with an extensive panel of citrullinated and viral peptides identified several different virus-specific TCRs (e.g., human cytomegalovirus and human herpesvirus 2). Still, the majority of clones remained orphans with unknown specificity. CONCLUSION: Minimally invasive biopsies of the RA synovium allow for single-cell TCR sequencing and phenotyping. Clonally expanded, viral-reactive T cells account for part of the diverse CD4+ T cell repertoire. TRBV20-1 bias in ACPA+ patients suggests recognition of common antigens.


Asunto(s)
Artritis Reumatoide , Humanos , Membrana Sinovial/patología , Linfocitos T CD4-Positivos , Receptores de Antígenos de Linfocitos T/genética , Cadenas HLA-DRB1/genética
7.
Eur J Pharmacol ; 931: 175189, 2022 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-35987255

RESUMEN

BACKGROUND: Functional network activity is a characteristic for neuronal cells, and the complexity of the network activity represents the necessary substrate to support complex brain functions. Drugs that drastically increase the neuronal network activity may have a potential higher risk for seizures in human. Although there has been some recent considerable progress made using cultures from different types of human-induced pluripotent stem cell (hiPSC) derived neurons, one of the primary limitations is the lack of - or very low - network activity. METHOD: In the present study, we investigated whether the limited neuronal network activity in commercial hiPSC-neurons (CNS.4U®) is capable of detecting drug-induced potential seizure risks. Therefore, we compared the hiPSC-results to those in rat primary neurons with known high neuronal network activity in vitro. RESULTS: Gene expression and electrical activity from in vitro developing neuronal networks were assessed at multiple time-points. Transcriptomes of 7, 28, and 50 days in vitro were analyzed and compared to those from human brain tissues. Data from measurements of electrical activity using multielectrode arrays (MEAs) indicate that neuronal networks matured gradually over time, albeit in hiPSC this developed slower than rat primary cultures. The response of neuronal networks to neuronal active reference drugs modulating glutamatergic, acetylcholinergic and GABAergic pathways could be detected in both hiPSC-neurons and rat primary neurons. However, in comparison, GABAergic responses were limited in hiPSC-neurons. CONCLUSION: Overall, despite a slower network development and lower network activity, CNS.4U® hiPSC-neurons can be used to detect drug induced changes in neuronal network activity, as shown by well-known seizurogenic drugs (affecting e.g., the Glycine receptor and Na+ channel). However, lower sensitivity to GABA antagonists has been observed.


Asunto(s)
Células Madre Pluripotentes Inducidas , Animales , Diferenciación Celular , Células Cultivadas , Humanos , Neuronas/metabolismo , Ratas , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Transmisión Sináptica
8.
Acta Neuropathol Commun ; 9(1): 150, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34503586

RESUMEN

Dystrophic neuronal processes harboring neuritic plaque (NP) tau pathology are found in association with Aß plaques in Alzheimer's disease (AD) brain. Microglia are also in proximity to these plaques and microglial gene variants are known risk factors in AD, including loss-of-function variants of TREM2. We have further investigated the role of Aß plaque-associated microglia in 5XFAD mice in which NP tau pathology forms after intracerebral injection of AD brain-derived pathologic tau (AD-tau), focusing on the consequences of reduced TREM2 expression and microglial depletion after treatment with the colony-stimulating factor 1 (CSFR1) inhibitor, PLX3397. Young 5XFAD mice treated with PLX3397 had a large reduction of brain microglia, including cortical plaque-associated microglia, with a significant reduction of Aß plaque burden in the cortex. A corresponding decrease in cortical APP-positive dystrophic processes and NP tau pathology were observed after intracerebral AD-tau injection in the PLX3397-treated 5XFAD mice. Consistent with prior reports, 5XFAD × TREM2-/- mice showed a significant reduction of plaque-associated microglial, whereas 5XFAD × TREM2+/- mice had significantly more plaque-associated microglia than 5XFAD × TREM2-/- mice. Nonetheless, AD-tau injected 5XFAD × TREM2+/- mice showed greatly increased AT8-positive NP tau relative to 5XFAD × TREM2+/+ mice. Expression profiling revealed that 5XFAD × TREM2+/- mice had a disease-associated microglial (DAM) gene expression profile in the brain that was generally intermediate between 5XFAD × TREM2+/+ and 5XFAD × TREM2-/- mice. Microarray analysis revealed significant differences in cortical and hippocampal gene expression between AD-tau injected 5XFAD × TREM2+/- and 5XFAD × TREM2-/- mice, including pathways linked to microglial function. These data suggest there is not a simple correlation between the extent of microglia plaque interaction and plaque-associated neuritic damage. Moreover, the differences in gene expression and microglial phenotype between TREM2+/- and TREM2-/- mice suggest that the former may better model the single copy TREM2 variants associated with AD risk.


Asunto(s)
Glicoproteínas de Membrana/deficiencia , Microglía/metabolismo , Placa Amiloide/metabolismo , Receptores Inmunológicos/deficiencia , Proteínas tau/toxicidad , Animales , Femenino , Masculino , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Placa Amiloide/inducido químicamente , Placa Amiloide/genética , Receptores Inmunológicos/genética , Proteínas tau/administración & dosificación
9.
Genes Chromosomes Cancer ; 47(11): 957-64, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18663747

RESUMEN

SNP arrays offer the opportunity to get a genome-wide view on copy number alterations and are increasingly used in oncology. DNA from formalin-fixed paraffin-embedded material (FFPE) is partially degraded which limits the application of those technologies for retrospective studies. We present the use of Affymetrix GeneChip SNP6.0 for identification of copy number alterations in fresh frozen (FF) and matched FFPE samples. Fifteen pairs of adenocarcinomas with both frozen and FFPE embedded material were analyzed. We present an optimization of the sample preparation and show the importance of correcting the measured intensities for fragment length and GC-content when using FFPE samples. The absence of GC content correction results in a chromosome specific "wave pattern" which may lead to the misclassification of genomic regions as being altered. The highest concordance between FFPE and matched FF were found in samples with the highest call rates. Nineteen of the 23 high level amplifications (83%) seen using FF samples were also detected in the corresponding FFPE material. For limiting the rate of "false positive" alterations, we have chosen a conservative False Discovery Rate (FDR). We observed better results using SNP probes than CNV probes for copy number analysis of FFPE material. This is the first report on the detection of copy number alterations in FFPE samples using Affymetrix GeneChip SNP6.0.


Asunto(s)
Dosificación de Gen , Genoma Humano , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Polimorfismo de Nucleótido Simple , ADN de Neoplasias/análisis , Formaldehído/química , Humanos , Adhesión en Parafina/métodos
10.
Eur J Pharmacol ; 858: 172474, 2019 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-31238068

RESUMEN

The Kv7 family of voltage-dependent non-inactivating potassium channels is composed of five members, of which four are expressed in the CNS. Kv7.2, 7.3 and 7.5 are responsible for the M-current, which plays a critical role in the regulation of neuronal excitability. Stimulation of M1 muscarinic acetylcholine receptor, M1 receptor, increases neuronal excitability by suppressing the M-current generated by the Kv7 channel family. The M-current modulation via M1 receptor is well-described in in vitro assays using cell lines and in native rodent tissue. However, this mechanism was not yet reported in human induced pluripotent stem cells (hiPSC) derived neurons. In the present study, we investigated the effects of both agonists and antagonists of Kv7.2/7.3 channel and M1 receptor in hiPSC derived neurons and in primary rat cortical neuronal cells. The role of M1 receptors in the modulation of neuronal excitability could be demonstrated in both rat primary and hiPSC neurons. The M1 receptors agonist, xanomeline, increased neuronal excitability in both rat cortical and the hiPSC neuronal cells. Furthermore, M1 receptor agonist-induced neuronal excitability in vitro was reduced by an agonist of Kv7.2/7.3 in both neuronal cells. These results show that hiPSC derived neurons recreate the modulation of the M-current by the muscarinic receptor in hiPSC neurons similarly to rat native neurons. Thus, hiPSC neurons could be a useful human-based cell assay for characterization of drugs that affect neuronal excitability and/or induce seizure activity by modulation of M1 receptors or inhibition of Kv7 channels.


Asunto(s)
Fenómenos Electrofisiológicos , Células Madre Pluripotentes Inducidas/citología , Canal de Potasio KCNQ2/metabolismo , Canal de Potasio KCNQ3/metabolismo , Neuronas/citología , Receptor Muscarínico M1/metabolismo , Animales , Fenómenos Electrofisiológicos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Canal de Potasio KCNQ2/agonistas , Canal de Potasio KCNQ2/antagonistas & inhibidores , Canal de Potasio KCNQ2/genética , Canal de Potasio KCNQ3/agonistas , Canal de Potasio KCNQ3/antagonistas & inhibidores , Canal de Potasio KCNQ3/genética , Antagonistas Muscarínicos/farmacología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Receptor Muscarínico M1/agonistas , Receptor Muscarínico M1/antagonistas & inhibidores
11.
Bioinformatics ; 23(22): 3032-8, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17893087

RESUMEN

Probe-level data from Affymetrix GeneChips can be summarized in many ways to produce probe-set level gene expression measures (GEMs). Disturbingly, the different approaches not only generate quite different measures but they could also yield very different analysis results. Here, we explore the question of how much the analysis results really do differ, first at the gene level, then at the biological process level. We demonstrate that, even though the gene level results may not necessarily match each other particularly well, as long as there is reasonably strong differentiation between the groups in the data, the various GEMs do in fact produce results that are similar to one another at the biological process level. Not only that the results are biologically relevant. As the extent of differentiation drops, the degree of concurrence weakens, although the biological relevance of findings at the biological process level may yet remain.


Asunto(s)
Algoritmos , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Stat Appl Genet Mol Biol ; 6: Article26, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-18052909

RESUMEN

Dose-response studies are commonly used in experiments in pharmaceutical research in order to investigate the dependence of the response on dose, i.e., a trend of the response level toxicity with respect to dose. In this paper we focus on dose-response experiments within a microarray setting in which several microarrays are available for a sequence of increasing dose levels. A gene is called differentially expressed if there is a monotonic trend (with respect to dose) in the gene expression. We review several testing procedures which can be used in order to test equality among the gene expression means against ordered alternatives with respect to dose, namely Williams' (Williams 1971 and 1972), Marcus' (Marcus 1976), global likelihood ratio test (Bartholomew 1961, Barlow et al. 1972, and Robertson et al. 1988), and M (Hu et al. 2005) statistics. Additionally we introduce a modification to the standard error of the M statistic. We compare the performance of these five test statistics. Moreover, we discuss the issue of one-sided versus two-sided testing procedures. False Discovery Rate (Benjamni and Hochberg 1995, Ge et al. 2003), and resampling-based Familywise Error Rate (Westfall and Young 1993) are used to handle the multiple testing issue. The methods above are applied to a data set with 4 doses (3 arrays per dose) and 16,998 genes. Results on the number of significant genes from each statistic are discussed. A simulation study is conducted to investigate the power of each statistic. A R library IsoGene implementing the methods is available from the first author.


Asunto(s)
Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Biblioteca de Genes , Humanos , Funciones de Verosimilitud , Pruebas Psicológicas , Reproducibilidad de los Resultados
13.
Assay Drug Dev Technol ; 16(3): 162-176, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29658791

RESUMEN

By adding biological information, beyond the chemical properties and desired effect of a compound, uncharted compound areas and connections can be explored. In this study, we add transcriptional information for 31K compounds of Janssen's primary screening deck, using the HT L1000 platform and assess (a) the transcriptional connection score for generating compound similarities, (b) machine learning algorithms for generating target activity predictions, and (c) the scaffold hopping potential of the resulting hits. We demonstrate that the transcriptional connection score is best computed from the significant genes only and should be interpreted within its confidence interval for which we provide the stats. These guidelines help to reduce noise, increase reproducibility, and enable the separation of specific and promiscuous compounds. The added value of machine learning is demonstrated for the NR3C1 and HSP90 targets. Support Vector Machine models yielded balanced accuracy values ≥80% when the expression values from DDIT4 & SERPINE1 and TMEM97 & SPR were used to predict the NR3C1 and HSP90 activity, respectively. Combining both models resulted in 22 new and confirmed HSP90-independent NR3C1 inhibitors, providing two scaffolds (i.e., pyrimidine and pyrazolo-pyrimidine), which could potentially be of interest in the treatment of depression (i.e., inhibiting the glucocorticoid receptor (i.e., NR3C1), while leaving its chaperone, HSP90, unaffected). As such, the initial hit rate increased by a factor 300, as less, but more specific chemistry could be screened, based on the upfront computed activity predictions.


Asunto(s)
Proteínas HSP90 de Choque Térmico/genética , Ensayos Analíticos de Alto Rendimiento , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores de Glucocorticoides/genética , Transcriptoma , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Receptores de Glucocorticoides/metabolismo , Máquina de Vectores de Soporte
14.
Stem Cell Reports ; 11(2): 363-379, 2018 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-30057263

RESUMEN

Tauopathies such as frontotemporal dementia (FTD) remain incurable to date, partially due to the lack of translational in vitro disease models. The MAPT gene, encoding the microtubule-associated protein tau, has been shown to play an important role in FTD pathogenesis. Therefore, we used zinc finger nucleases to introduce two MAPT mutations into healthy donor induced pluripotent stem cells (iPSCs). The IVS10+16 mutation increases the expression of 4R tau, while the P301S mutation is pro-aggregant. Whole-transcriptome analysis of MAPT IVS10+16 neurons reveals neuronal subtype differences, reduced neural progenitor proliferation potential, and aberrant WNT/SHH signaling. Notably, these neurodevelopmental phenotypes could be recapitulated in neurons from patients carrying the MAPT IVS10+16 mutation. Moreover, the additional pro-aggregant P301S mutation revealed additional phenotypes, such as an increased calcium burst frequency, reduced lysosomal acidity, tau oligomerization, and neurodegeneration. This series of iPSCs could serve as a platform to unravel a potential link between pathogenic 4R tau and FTD.

15.
Br J Pharmacol ; 174(21): 3766-3779, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28094846

RESUMEN

BACKGROUND AND PURPOSE: In the pharmaceutical industry risk assessments of chronic cardiac safety liabilities are mostly performed during late stages of preclinical drug development using in vivo animal models. Here, we explored the potential of human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) to detect chronic cardiac risks such as drug-induced cardiomyocyte toxicity. EXPERIMENTAL APPROACH: Video microscopy-based motion field imaging was applied to evaluate the chronic effect (over 72 h) of cardiotoxic drugs on the contractile motion of hiPS-CMs. In parallel, the release of cardiac troponin I (cTnI), heart fatty acid binding protein (FABP3) and N-terminal pro-brain natriuretic peptide (NT-proBNP) was analysed from cell medium, and transcriptional profiling of hiPS-CMs was done at the end of the experiment. KEY RESULTS: Different cardiotoxic drugs altered the contractile motion properties of hiPS-CMs together with increasing the release of cardiac biomarkers. FABP3 and cTnI were shown to be potential surrogates to predict cardiotoxicity in hiPS-CMs, whereas NT-proBNP seemed to be a less valuable biomarker. Furthermore, drug-induced cardiotoxicity produced by chronic exposure of hiPS-CMs to arsenic trioxide, doxorubicin or panobinostat was associated with different profiles of changes in contractile parameters, biomarker release and transcriptional expression. CONCLUSION AND IMPLICATIONS: We have shown that a parallel assessment of motion field imaging-derived contractile properties, release of biomarkers and transcriptional changes can detect diverse mechanisms of chronic drug-induced cardiac liabilities in hiPS-CMs. Hence, hiPS-CMs could potentially improve and accelerate cardiovascular de-risking of compounds at earlier stages of drug discovery. LINKED ARTICLES: This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.


Asunto(s)
Antineoplásicos/toxicidad , Cardiotoxicidad/etiología , Células Madre Pluripotentes Inducidas/citología , Miocitos Cardíacos/efectos de los fármacos , Trióxido de Arsénico , Arsenicales , Biomarcadores/metabolismo , Cardiotoxicidad/fisiopatología , Células Cultivadas , Doxorrubicina/toxicidad , Evaluación Preclínica de Medicamentos/métodos , Humanos , Ácidos Hidroxámicos/toxicidad , Indoles/toxicidad , Microscopía por Video , Contracción Muscular/efectos de los fármacos , Miocitos Cardíacos/patología , Óxidos/toxicidad , Panobinostat
16.
Exp Neurol ; 291: 106-119, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28189729

RESUMEN

Slc17a5-/- mice represent an animal model for the infantile form of sialic acid storage disease (SASD). We analyzed genetic and histological time-course expression of myelin and oligodendrocyte (OL) lineage markers in different parts of the CNS, and related this to postnatal neurobehavioral development in these mice. Sialin-deficient mice display a distinct spatiotemporal pattern of sialic acid storage, CNS hypomyelination and leukoencephalopathy. Whereas few genes are differentially expressed in the perinatal stage (p0), microarray analysis revealed increased differential gene expression in later postnatal stages (p10-p18). This included progressive upregulation of neuroinflammatory genes, as well as continuous down-regulation of genes that encode myelin constituents and typical OL lineage markers. Age-related histopathological analysis indicates that initial myelination occurs normally in hindbrain regions, but progression to more frontal areas is affected in Slc17a5-/- mice. This course of progressive leukoencephalopathy and CNS hypomyelination delays neurobehavioral development in sialin-deficient mice. Slc17a5-/- mice successfully achieve early neurobehavioral milestones, but exhibit progressive delay of later-stage sensory and motor milestones. The present findings may contribute to further understanding of the processes of CNS myelination as well as help to develop therapeutic strategies for SASD and other myelination disorders.


Asunto(s)
Encéfalo/patología , Regulación del Desarrollo de la Expresión Génica/genética , Leucoencefalopatías , Trastornos Mentales/etiología , Transportadores de Anión Orgánico/deficiencia , Enfermedad por Almacenamiento de Ácido Siálico , Simportadores/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Discapacidades del Desarrollo/etiología , Discapacidades del Desarrollo/genética , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Filamentos Intermedios/metabolismo , Leucoencefalopatías/complicaciones , Leucoencefalopatías/etiología , Leucoencefalopatías/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Transportadores de Anión Orgánico/genética , Enfermedad por Almacenamiento de Ácido Siálico/complicaciones , Enfermedad por Almacenamiento de Ácido Siálico/genética , Enfermedad por Almacenamiento de Ácido Siálico/patología , Simportadores/genética
17.
Assay Drug Dev Technol ; 14(4): 252-60, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27187605

RESUMEN

The NIH-funded LINCS program has been initiated to generate a library of integrated, network-based, cellular signatures (LINCS). A novel high-throughput gene-expression profiling assay known as L1000 was the main technology used to generate more than a million transcriptional profiles. The profiles are based on the treatment of 14 cell lines with one of many perturbation agents of interest at a single concentration for 6 and 24 hours duration. In this study, we focus on the chemical compound treatments within the LINCS data set. The experimental variables available include number of replicates, cell lines, and time points. Our study reveals that compound characterization based on three cell lines at two time points results in more genes being affected than six cell lines at a single time point. Based on the available LINCS data, we conclude that the most optimal experimental design to characterize a large set of compounds is to test them in duplicate in three different cell lines. Our conclusions are constrained by the fact that the compounds were profiled at a single, relative high concentration, and the longer time point is likely to result in phenotypic rather than mechanistic effects being recorded.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Biblioteca de Genes , Transcripción Genética/genética , Transcriptoma/genética , Células A549 , Antineoplásicos/farmacología , Bases de Datos Genéticas , Células HT29 , Células Hep G2 , Humanos , Células MCF-7 , Transcripción Genética/efectos de los fármacos , Transcriptoma/efectos de los fármacos
18.
Stem Cells Transl Med ; 5(5): 602-12, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27034410

RESUMEN

UNLABELLED: Histone deacetylase (HDAC) inhibitors possess therapeutic potential to reverse aberrant epigenetic changes associated with cancers, neurological diseases, and immune disorders. Unfortunately, clinical studies with some HDAC inhibitors displayed delayed cardiac adverse effects, such as atrial fibrillation and ventricular tachycardia. However, the underlying molecular mechanism(s) of HDAC inhibitor-mediated cardiotoxicity remains poorly understood and is difficult to detect in the early stages of preclinical drug development because of a delayed onset of effects. In the present study, we show for the first time in human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) that HDAC inhibitors (dacinostat, panobinostat, vorinostat, entinostat, and tubastatin-a) induce delayed dose-related cardiac dysfunction at therapeutic concentrations associated with cardiac adverse effects in humans. HDAC inhibitor-mediated delayed effects on the beating properties of hiPS-CMs developed after 12 hours by decreasing the beat rate, shortening the field potential duration, and inducing arrhythmic behavior under form of sustained contractions and fibrillation-like patterns. Transcriptional changes that are common between the cardiotoxic HDAC inhibitors but different from noncardiotoxic treatments identified cardiac-specific genes and pathways related to structural and functional changes in cardiomyocytes. Combining the functional data with epigenetic changes in hiPS-CMs allowed us to identify molecular targets that might explain HDAC inhibitor-mediated cardiac adverse effects in humans. Therefore, hiPS-CMs represent a valuable translational model to assess HDAC inhibitor-mediated cardiotoxicity and support identification of better HDAC inhibitors with an improved benefit-risk profile. SIGNIFICANCE: Histone deacetylase (HDAC) inhibitors are a promising class of drugs to treat certain cancers, autoimmune, and neurodegenerative diseases. However, treated patients can experience various cardiac adverse events such as hearth rhythm disorders. This study found that human induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) can predict cardiac adverse events in patients caused by HDAC inhibitors. Furthermore, transcriptional changes at the level of gene expression supported the effects on the beating properties of hiPS-CMs and highlight targets that might cause these cardiac adverse effects. hiPS-CMs represent a valuable translational model to assess HDAC inhibitor-mediated cardiotoxicity and to support development of safer HDAC inhibitors.


Asunto(s)
Cardiopatías/inducido químicamente , Inhibidores de Histona Desacetilasas/toxicidad , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Potenciales de Acción , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/enzimología , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Epigénesis Genética/efectos de los fármacos , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Genotipo , Cardiopatías/enzimología , Cardiopatías/genética , Cardiopatías/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/enzimología , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/enzimología , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Medición de Riesgo , Factores de Tiempo
19.
Biosci Rep ; 35(6)2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26431963

RESUMEN

Maternal embryonic leucine zipper kinase (MELK), a serine/threonine protein kinase, has oncogenic properties and is overexpressed in many cancer cells. The oncogenic function of MELK is attributed to its capacity to disable critical cell-cycle checkpoints and reduce replication stress. Most functional studies have relied on the use of siRNA/shRNA-mediated gene silencing. In the present study, we have explored the biological function of MELK using MELK-T1, a novel and selective small-molecule inhibitor. Strikingly, MELK-T1 triggered a rapid and proteasome-dependent degradation of the MELK protein. Treatment of MCF-7 (Michigan Cancer Foundation-7) breast adenocarcinoma cells with MELK-T1 induced the accumulation of stalled replication forks and double-strand breaks that culminated in a replicative senescence phenotype. This phenotype correlated with a rapid and long-lasting ataxia telangiectasia-mutated (ATM) activation and phosphorylation of checkpoint kinase 2 (CHK2). Furthermore, MELK-T1 induced a strong phosphorylation of p53 (cellular tumour antigen p53), a prolonged up-regulation of p21 (cyclin-dependent kinase inhibitor 1) and a down-regulation of FOXM1 (Forkhead Box M1) target genes. Our data indicate that MELK is a key stimulator of proliferation by its ability to increase the threshold for DNA-damage tolerance (DDT). Thus, targeting MELK by the inhibition of both its catalytic activity and its protein stability might sensitize tumours to DNA-damaging agents or radiation therapy by lowering the DNA-damage threshold.


Asunto(s)
Azepinas/administración & dosificación , Benzamidas/administración & dosificación , Neoplasias de la Mama/genética , Daño del ADN/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Proteínas Serina-Treonina Quinasas/biosíntesis , Apoptosis/efectos de los fármacos , Proteínas de la Ataxia Telangiectasia Mutada/biosíntesis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Neoplasias de la Mama/radioterapia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Proteína Forkhead Box M1 , Factores de Transcripción Forkhead/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Células MCF-7 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética
20.
Clin Cancer Res ; 21(14): 3327-39, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25805801

RESUMEN

PURPOSE: Activating ALK mutations are present in almost 10% of primary neuroblastomas and mark patients for treatment with small-molecule ALK inhibitors in clinical trials. However, recent studies have shown that multiple mechanisms drive resistance to these molecular therapies. We anticipated that detailed mapping of the oncogenic ALK-driven signaling in neuroblastoma can aid to identify potential fragile nodes as additional targets for combination therapies. EXPERIMENTAL DESIGN: To achieve this goal, transcriptome profiling was performed in neuroblastoma cell lines with the ALK(F1174L) or ALK(R1275Q) hotspot mutations, ALK amplification, or wild-type ALK following pharmacologic inhibition of ALK using four different compounds. Next, we performed cross-species genomic analyses to identify commonly transcriptionally perturbed genes in MYCN/ALK(F1174L) double transgenic versus MYCN transgenic mouse tumors as compared with the mutant ALK-driven transcriptome in human neuroblastomas. RESULTS: A 77-gene ALK signature was established and successfully validated in primary neuroblastoma samples, in a neuroblastoma cell line with ALK(F1174L) and ALK(R1275Q) regulable overexpression constructs and in other ALKomas. In addition to the previously established PI3K/AKT/mTOR, MAPK/ERK, and MYC/MYCN signaling branches, we identified that mutant ALK drives a strong upregulation of MAPK negative feedback regulators and upregulates RET and RET-driven sympathetic neuronal markers of the cholinergic lineage. CONCLUSIONS: We provide important novel insights into the transcriptional consequences and the complexity of mutant ALK signaling in this aggressive pediatric tumor. The negative feedback loop of MAPK pathway inhibitors may affect novel ALK inhibition therapies, whereas mutant ALK induced RET signaling can offer novel opportunities for testing ALK-RET oriented molecular combination therapies.


Asunto(s)
Fosfatasa Alcalina/genética , Resistencia a Antineoplásicos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Terapia Molecular Dirigida/métodos , Neuroblastoma/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Retroalimentación Fisiológica , Humanos , Ratones , Ratones Transgénicos , Neuroblastoma/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Transcriptoma , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA