Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
bioRxiv ; 2024 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-38260577

RESUMO

Schizophrenia (SCZ) is a genetically heterogenous psychiatric disorder of highly polygenic nature. Correlative evidence from genetic studies indicate that the aggregated effects of distinct genetic risk factor combinations found in each patient converge onto common molecular mechanisms. To prove this on a functional level, we employed a reductionistic cellular model system for polygenic risk by differentiating induced pluripotent stem cells (iPSCs) from 104 individuals with high polygenic risk load and controls into cortical glutamatergic neurons (iNs). Multi-omics profiling identified widespread differences in alternative polyadenylation (APA) in the 3' untranslated region of many synaptic transcripts between iNs from SCZ patients and healthy donors. On the cellular level, 3'APA was associated with a reduction in synaptic density of iNs. Importantly, differential APA was largely conserved between postmortem human prefrontal cortex from SCZ patients and healthy donors, and strongly enriched for transcripts related to synapse biology. 3'APA was highly correlated with SCZ polygenic risk and affected genes were significantly enriched for SCZ associated common genetic variation. Integrative functional genomic analysis identified the RNA binding protein and SCZ GWAS risk gene PTBP2 as a critical trans-acting factor mediating 3'APA of synaptic genes in SCZ subjects. Functional characterization of PTBP2 in iNs confirmed its key role in 3'APA of synaptic transcripts and regulation of synapse density. Jointly, our findings show that the aggregated effects of polygenic risk converge on 3'APA as one common molecular mechanism that underlies synaptic impairments in SCZ.

2.
Front Psychiatry ; 14: 1179811, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37215661

RESUMO

Introduction: Treatment of severe mental illness (SMI) symptoms, especially negative symptoms and cognitive dysfunction in schizophrenia, remains a major unmet need. There is good evidence that SMIs have a strong genetic background and are characterized by multiple biological alterations, including disturbed brain circuits and connectivity, dysregulated neuronal excitation-inhibition, disturbed dopaminergic and glutamatergic pathways, and partially dysregulated inflammatory processes. The ways in which the dysregulated signaling pathways are interconnected remains largely unknown, in part because well-characterized clinical studies on comprehensive biomaterial are lacking. Furthermore, the development of drugs to treat SMIs such as schizophrenia is limited by the use of operationalized symptom-based clusters for diagnosis. Methods: In line with the Research Domain Criteria initiative, the Clinical Deep Phenotyping (CDP) study is using a multimodal approach to reveal the neurobiological underpinnings of clinically relevant schizophrenia subgroups by performing broad transdiagnostic clinical characterization with standardized neurocognitive assessments, multimodal neuroimaging, electrophysiological assessments, retinal investigations, and omics-based analyzes of blood and cerebrospinal fluid. Moreover, to bridge the translational gap in biological psychiatry the study includes in vitro investigations on human-induced pluripotent stem cells, which are available from a subset of participants. Results: Here, we report on the feasibility of this multimodal approach, which has been successfully initiated in the first participants in the CDP cohort; to date, the cohort comprises over 194 individuals with SMI and 187 age and gender matched healthy controls. In addition, we describe the applied research modalities and study objectives. Discussion: The identification of cross-diagnostic and diagnosis-specific biotype-informed subgroups of patients and the translational dissection of those subgroups may help to pave the way toward precision medicine with artificial intelligence-supported tailored interventions and treatment. This aim is particularly important in psychiatry, a field where innovation is urgently needed because specific symptom domains, such as negative symptoms and cognitive dysfunction, and treatment-resistant symptoms in general are still difficult to treat.

3.
Cells ; 11(2)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35053357

RESUMO

Oligodendrocytes (OLs) are critical for myelination and are implicated in several brain disorders. Directed differentiation of human-induced OLs (iOLs) from pluripotent stem cells can be achieved by forced expression of different combinations of the transcription factors SOX10 (S), OLIG2 (O), and NKX6.2 (N). Here, we applied quantitative image analysis and single-cell transcriptomics to compare different transcription factor (TF) combinations for their efficacy towards robust OL lineage conversion. Compared with S alone, the combination of SON increases the number of iOLs and generates iOLs with a more complex morphology and higher expression levels of myelin-marker genes. RNA velocity analysis of individual cells reveals that S generates a population of oligodendrocyte-precursor cells (OPCs) that appear to be more immature than those generated by SON and to display distinct molecular properties. Our work highlights that TFs for generating iOPCs or iOLs should be chosen depending on the intended application or research question, and that SON might be beneficial to study more mature iOLs while S might be better suited to investigate iOPC biology.


Assuntos
Diferenciação Celular , Linhagem da Célula , Oligodendroglia/citologia , Oligodendroglia/metabolismo , Fatores de Transcrição/metabolismo , Diferenciação Celular/genética , Linhagem da Célula/genética , Células Cultivadas , Regulação da Expressão Gênica , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Modelos Biológicos , Neurogênese/genética , RNA/metabolismo , Transcriptoma/genética
4.
Anal Biochem ; 412(2): 141-52, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21295005

RESUMO

G protein-coupled receptors (GPCRs) constitute the largest receptor family in mammals and represent important drug targets. Signaling through GPCRs mediates physiological effects that are strongly dependent on the cellular context. Therefore, the availability of assays monitoring GPCR activation applicable in different cell types could help to better understand GPCR functions and to realize the potential of known substances as well as novel ones. Here we introduce a split-TEV (tobacco etch virus) assay to monitor GPCR activation through the stimulation-dependent recruitment of ß-arrestin 2. Inactive N- and C-terminal fragments of the TEV protease are coupled to a GPCR and ß-arrestin 2, respectively. Ligand-dependent interaction of the two fusion proteins leads to functional complementation of the TEV protease, followed by the cleavage of an artificial transcription factor and successive reporter gene activation. The presented split-TEV assay system is highly sensitive and was successfully applied in heterologous cell lines as well as in primary cultured neuronal and glial cells. We show that assay performance strongly depends on the endogenous properties of different cell types. The sensitivity and flexibility make split-TEV assays a valuable tool to analyze GPCR activation in different cell types in a rapid and cost-effective way.


Assuntos
Bioensaio/métodos , Nicotiana/virologia , Vírus de Plantas/genética , Receptores Acoplados a Proteínas G/metabolismo , Animais , Endopeptidases/metabolismo , Humanos , Células PC12 , Ratos , Receptores de Vasopressinas/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Transdução de Sinais
5.
Trends Pharmacol Sci ; 41(5): 318-335, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32223968

RESUMO

Drug discovery campaigns are hampered by substantial attrition rates largely due to a lack of efficacy and safety reasons associated with candidate drugs. This is true in particular for genetically complex diseases, where insufficient knowledge of the modulatory actions of candidate drugs on targets and entire target pathways further adds to the problem of attrition. To better profile compound actions on targets, potential off-targets, and disease-linked pathways, new innovative technologies need to be developed that can elucidate the complex cellular signaling networks in health and disease. Here, we discuss progress in genetically encoded multiparametric assays and mass spectrometry (MS)-based proteomics, which both represent promising toolkits to profile multifactorial actions of drug candidates in disease-relevant cellular systems to promote drug discovery and personalized medicine.


Assuntos
Descoberta de Drogas , Proteômica , Humanos
6.
Sci Rep ; 8(1): 8137, 2018 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-29802268

RESUMO

G protein-coupled receptors (GPCRs) are the largest class of cell surface receptors and are implicated in the physiological regulation of many biological processes. The high diversity of GPCRs and their physiological functions make them primary targets for therapeutic drugs. For the generation of novel compounds, however, selectivity towards a given target is a critical issue in drug development as structural similarities between members of GPCR subfamilies exist. Therefore, the activities of multiple GPCRs that are both closely and distantly related to assess compound selectivity need to be tested simultaneously. Here, we present a cell-based multiplexed GPCR activity assay, termed GPCRprofiler, which uses a ß-arrestin recruitment strategy and combines split TEV protein-protein interaction and EXT-based barcode technologies. This approach enables simultaneous measurements of receptor activities of multiple GPCR-ligand combinations by applying massively parallelized reporter assays. In proof-of-principle experiments covering 19 different GPCRs, both the specificity of endogenous agonists and the polypharmacological effects of two known antipsychotics on GPCR activities were demonstrated. Technically, normalization of barcode reporters across individual assays allows quantitative pharmacological assays in a parallelized manner. In summary, the GPCRprofiler technique constitutes a flexible and scalable approach, which enables simultaneous profiling of compound actions on multiple receptor activities in living cells.


Assuntos
Bioensaio/métodos , Receptores Acoplados a Proteínas G/metabolismo , Animais , Antipsicóticos/farmacologia , Linhagem Celular Tumoral , Células PC12 , Ratos , Transdução de Sinais/efeitos dos fármacos
7.
NPJ Schizophr ; 4(1): 23, 2018 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-30451850

RESUMO

Postmortem studies in patients with schizophrenia (SCZ) have revealed deficits in myelination, abnormalities in myelin gene expression and altered numbers of oligodendrocytes in the brain. However, gaining mechanistic insight into oligodendrocyte (OL) dysfunction and its contribution to SCZ has been challenging because of technical hurdles. The advent of individual patient-derived human-induced pluripotent stem cells (hiPSCs), combined with the generation of in principle any neuronal and glial cell type, including OLs and oligodendrocyte precursor cells (OPCs), holds great potential for understanding the molecular basis of the aetiopathogenesis of genetically complex psychiatric diseases such as SCZ and could pave the way towards personalized medicine. The development of neuronal and glial co-culture systems now appears to enable the in vitro study of SCZ-relevant neurobiological endophenotypes, including OL dysfunction and myelination, with unprecedented construct validity. Nonetheless, the meaningful stratification of patients before the subsequent functional analyses of patient-derived cell systems still represents an important bottleneck. Here, to improve the predictive power of ex vivo disease modelling we propose using hiPSC technology to focus on representatives of patient subgroups stratified for genomic and/or phenomic features and neurobiological cell systems. Therefore, this review will outline the evidence for the involvement of OPCs/OLs in SCZ in the context of their proposed functions, including myelination and axon support, the implications for hiPSC-based cellular disease modelling and potential strategies for patient selection.

8.
Methods Mol Biol ; 1272: 107-18, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25563180

RESUMO

G protein-coupled receptors (GPCRs) modulate cellular signaling, often in a ligand-specific manner. Cellular effects regulated include differentiation, proliferation, hormonal regulation, and neuronal activity. Further, they are involved in many disease-relevant processes, such as cancer and neurodevelopmental diseases, and represent the largest class of drug targets. Therefore, monitoring how GPCRs are regulated in their activity is crucial to understand their role in physiological processes and implications for drug development. Split TEV, a method based on TEV protease fragment complementation, can be used to sensitively assay GPCR activities in living cells. The activity of a given GPCR is monitored through its binding to ß-arrestin. Split TEV reporters provide at minimum a two-step amplification process facilitating a flexible format and a robust readout. For the initial setup, a GPCR of interest and ß-arrestin are fused to the N- and C-terminal fragments of the TEV protease, and occurred interactions are indicated by increased fluorescence or luminescence of TEV cleavage-dependent reporters. The experimental procedure takes 24-72 h to complete, depending on the cell type and complexity of the experimental setup applied.


Assuntos
Bioensaio , Endopeptidases/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Animais , Arrestinas/genética , Arrestinas/metabolismo , Dibenzocicloeptenos , Endopeptidases/genética , Expressão Gênica , Genes Reporter , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Ligantes , Luciferases/genética , Luciferases/metabolismo , Células PC12 , Plasmídeos/química , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , Proteólise , Ratos , Receptor 5-HT1A de Serotonina/genética , Proteínas Recombinantes de Fusão/genética , Serotonina/farmacologia , Transdução de Sinais , Transativadores/genética , Transativadores/metabolismo , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA