Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood Cells Mol Dis ; 94: 102651, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35051688

RESUMEN

BACKGROUND: To develop and validate a predictive model to determinate patients at increased risk to suffer from recurrence following a first provoked deep vein thrombosis (VTE). METHODS: Predictive variables, i.e. male sex [1 point], inherited thrombophilia (IT) status (none [0 points], single [1 point], combined variants [2 points]), blood group non-0, and age at first VTE onset were included into a risk assessment model, which was derived in 511 patients and then validated in 509 independent subjects. RESULTS: VTE recurrence risk score (maximum 4 points, range 0-3) was below two for patients scored as low-risk (LRS) and ≥2 for patients at high-risk (HRS). Within a median time of 3 years after withdrawal of anticoagulation (AC) recurrence rate in LRG (derivation) was 11.8% versus 26.0% in HRS (p < 0.001). In the validation cohort within 2.2 years the recurrence rate was 9.8% in LRS versus 30.1% in HRS (p < 0.001). In multivariable analysis adjusted for age at first VTE and blood group the recurrent risk in HRS was significantly increased compared with the LRS (derivation: hazard/95% confidence interval: 3.7/1.75-7.91; validation: 4.7/2.24-9.81; combined 5.2/1.92-13.9). Model specificity (sensitivity) was 79.0% (52.0%) in the derivation cohort compared with 78.0% (43.0%) in the validation group. In conclusion, in the prediction model presented here the risk of VTE recurrence was associated with male gender and combined ITs. Based on the negative predictive value calculated the model may identify patients with a first provoked VTE not being at risk for recurrence.


Asunto(s)
Antígenos de Grupos Sanguíneos , Trombofilia , Tromboembolia Venosa , Trombosis de la Vena , Adolescente , Anticoagulantes/efectos adversos , Humanos , Masculino , Recurrencia , Factores de Riesgo , Trombofilia/complicaciones , Tromboembolia Venosa/diagnóstico , Tromboembolia Venosa/epidemiología , Tromboembolia Venosa/etiología , Adulto Joven
2.
Int J Mol Sci ; 22(6)2021 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-33806835

RESUMEN

Chromodomain helicase domain 8 (CHD8) is one of the most frequently mutated and most penetrant genes in the autism spectrum disorder (ASD). Individuals with CHD8 mutations show leading symptoms of autism, macrocephaly, and facial dysmorphisms. The molecular and cellular mechanisms underpinning the early onset and development of these symptoms are still poorly understood and prevent timely and more efficient therapies of patients. Progress in this area will require an understanding of "when, why and how cells deviate from their normal trajectories". High-throughput single-cell RNA sequencing (sc-RNAseq) directly quantifies information-bearing RNA molecules that enact each cell's biological identity. Here, we discuss recent insights from sc-RNAseq of CRISPR/Cas9-editing of Chd8/CHD8 during mouse neocorticogenesis and human cerebral organoids. Given that the deregulation of the balance between excitation and inhibition (E/I balance) in cortical and subcortical circuits is thought to represent a major etiopathogenetic mechanism in ASD, we focus on the question of whether, and to what degree, results from current sc-RNAseq studies support this hypothesis. Beyond that, we discuss the pros and cons of these approaches and further steps to be taken to harvest the full potential of these transformative techniques.


Asunto(s)
Trastorno Autístico/etiología , Trastorno Autístico/metabolismo , Proteínas de Unión al ADN/genética , Susceptibilidad a Enfermedades , Factores de Transcripción/genética , Transcriptoma , Animales , Trastorno Autístico/psicología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Mutación , Neurogénesis , Análisis de la Célula Individual/métodos , Factores de Transcripción/metabolismo
3.
Int J Mol Sci ; 20(19)2019 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-31581684

RESUMEN

Progress in iPSC-based cellular systems provides new insights into human brain development and early neurodevelopmental deviations in psychiatric disorders. Among these, studies on schizophrenia (SCZ) take a prominent role owing to its high heritability and multifarious evidence that it evolves from a genetically induced vulnerability in brain development. Recent iPSC studies on patients with SCZ indicate that functional impairments of neural progenitor cells (NPCs) in monolayer culture extend to brain organoids by disrupting neocorticogenesis in an in vitro model. In addition, the formation of hippocampal circuit-like structures in vitro is impaired in patients with SCZ as is the case for glia development. Intriguingly, chimeric-mice experiments show altered oligodendrocyte and astrocyte development in vivo that highlights the importance of cell-cell interactions in the pathogenesis of early-onset SCZ. Likewise, cortical imbalances in excitatory-inhibitory signaling may result from a cell-autonomous defect in cortical interneuron (cIN) development. Overall, these findings indicate that genetic risk in SCZ impacts neocorticogenesis, hippocampal circuit formation, and the development of distinct glial and neuronal subtypes. In light of this remarkable progress, we discuss current limitations and further steps necessary to harvest the full potential of iPSC-based investigations on psychiatric disorders.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Trastornos Mentales/etiología , Trastornos Mentales/metabolismo , Modelos Biológicos , Animales , Hipocampo/embriología , Hipocampo/metabolismo , Hipocampo/fisiopatología , Humanos , Células-Madre Neurales/metabolismo , Neurogénesis
4.
J Cell Mol Med ; 22(2): 982-998, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29210175

RESUMEN

Infectious agents such as lipopolysaccharides (LPS) challenge the functional properties of the alveolar-capillary barrier (ACB) in the lung. In this study, we analyse the site-specific effects of LPS on the ACB and reveal the effects on the individual cell types and the ACB as a functional unit. Monocultures of H441 epithelial cells and co-cultures of H441 with endothelial cells cultured on Transwells® were treated with LPS from the apical or basolateral compartment. Barrier properties were analysed by the transepithelial electrical resistance (TEER), by transport assays, and immunostaining and assessment of tight junctional molecules at protein level. Furthermore, pro-inflammatory cytokines and immune-modulatory molecules were evaluated by ELISA and semiquantitative real-time PCR. Liquid chromatography-mass spectrometry-based proteomics (LS-MS) was used to identify proteins and effector molecules secreted by endothelial cells in response to LPS. In co-cultures treated with LPS from the basolateral compartment, we noticed a significant reduction of TEER, increased permeability and induction of pro-inflammatory cytokines. Conversely, apical treatment did not affect the barrier. No changes were noticed in H441 monoculture upon LPS treatment. However, LPS resulted in an increased expression of pro-inflammatory cytokines such as IL-6 in OEC and in turn induced the reduction of TEER and an increase in SP-A expression in H441 monoculture, and H441/OEC co-cultures after LPS treatment from basolateral compartment. LS-MS-based proteomics revealed factors associated with LPS-mediated lung injury such as ICAM-1, VCAM-1, Angiopoietin 2, complement factors and cathepsin S, emphasizing the role of epithelial-endothelial crosstalk in the ACB in ALI/ARDS.


Asunto(s)
Capilares/fisiopatología , Células Endoteliales/patología , Lipopolisacáridos/toxicidad , Alveolos Pulmonares/fisiopatología , Adulto , Capilares/efectos de los fármacos , Caveolina 1/metabolismo , Línea Celular , Permeabilidad de la Membrana Celular/efectos de los fármacos , Técnicas de Cocultivo , Impedancia Eléctrica , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/patología , Fosforilación/efectos de los fármacos , Proteómica , Alveolos Pulmonares/efectos de los fármacos , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Proteínas de Uniones Estrechas/metabolismo , Proteína de la Zonula Occludens-1/metabolismo
5.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L32-L53, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28860142

RESUMEN

The biological and immune-protective properties of surfactant-derived phospholipids and phospholipid subfractions in the context of neonatal inflammatory lung disease are widely unknown. Using a porcine neonatal triple-hit acute respiratory distress syndrome (ARDS) model (repeated airway lavage, overventilation, and LPS instillation into airways), we assessed whether the supplementation of surfactant (S; poractant alfa) with inositol derivatives [inositol 1,2,6-trisphosphate (IP3) or phosphatidylinositol 3,5-bisphosphate (PIP2)] or phosphatidylglycerol subfractions [16:0/18:1-palmitoyloleoyl-phosphatidylglycerol (POPG) or 18:1/18:1-dioleoyl-phosphatidylglycerol (DOPG)] would result in improved clinical parameters and sought to characterize changes in key inflammatory pathways behind these improvements. Within 72 h of mechanical ventilation, the oxygenation index (S+IP3, S+PIP2, and S+POPG), the ventilation efficiency index (S+IP3 and S+POPG), the compliance (S+IP3 and S+POPG) and resistance (S+POPG) of the respiratory system, and the extravascular lung water index (S+IP3 and S+POPG) significantly improved compared with S treatment alone. The inositol derivatives (mainly S+IP3) exerted their actions by suppressing acid sphingomyelinase activity and dependent ceramide production, linked with the suppression of the inflammasome nucleotide-binding domain, leucine-rich repeat-containing protein-3 (NLRP3)-apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC)-caspase-1 complex, and the profibrotic response represented by the cytokines transforming growth factor-ß1 and IFN-γ, matrix metalloproteinase (MMP)-1/8, and elastin. In addition, IκB kinase activity was significantly reduced. S+POPG and S+DOPG treatment inhibited polymorphonuclear leukocyte activity (MMP-8 and myeloperoxidase) and the production of interleukin-6, maintained alveolar-capillary barrier functions, and reduced alveolar epithelial cell apoptosis, all of which resulted in reduced pulmonary edema. S+DOPG also limited the profibrotic response. We conclude that highly concentrated inositol derivatives and phosphatidylglycerol subfractions in surfactant preparations mitigate key inflammatory pathways in inflammatory lung disease and that their clinical application may be of interest for future treatment of the acute exudative phase of neonatal ARDS.


Asunto(s)
Modelos Animales de Enfermedad , Inositol/farmacología , Fosfatidilgliceroles/farmacología , Edema Pulmonar/tratamiento farmacológico , Surfactantes Pulmonares/farmacología , Síndrome de Dificultad Respiratoria del Recién Nacido/tratamiento farmacológico , Animales , Animales Recién Nacidos , Apoptosis , Líquido del Lavado Bronquioalveolar , Citocinas/genética , Citocinas/metabolismo , Femenino , Humanos , Masculino , FN-kappa B/genética , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Intercambio Gaseoso Pulmonar , Distribución Aleatoria , Respiración Artificial , Síndrome de Dificultad Respiratoria del Recién Nacido/metabolismo , Síndrome de Dificultad Respiratoria del Recién Nacido/patología , Porcinos , Investigación Biomédica Traslacional , Complejo Vitamínico B/farmacología
6.
Int J Mol Sci ; 19(12)2018 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-30513688

RESUMEN

Childhood-onset schizophrenia (COS) is a rare psychiatric disorder characterized by earlier onset, more severe course, and poorer outcome relative to adult-onset schizophrenia (AOS). Even though, clinical, neuroimaging, and genetic studies support that COS is continuous to AOS. Early neurodevelopmental deviations in COS are thought to be significantly mediated through poorly understood genetic risk factors that may also predispose to long-term outcome. In this review, we discuss findings from induced pluripotent stem cells (iPSCs) that allow the generation of disease-relevant cell types from early brain development. Because iPSCs capture each donor's genotype, case/control studies can uncover molecular and cellular underpinnings of COS. Indeed, recent studies identified alterations in neural progenitor and neuronal cell function, comprising dendrites, synapses, electrical activity, glutamate signaling, and miRNA expression. Interestingly, transcriptional signatures of iPSC-derived cells from patients with COS showed concordance with postmortem brain samples from SCZ, indicating that changes in vitro may recapitulate changes from the diseased brain. Considering this progress, we discuss also current caveats from the field of iPSC-based disease modeling and how to proceed from basic studies to improved diagnosis and treatment of COS.


Asunto(s)
Células Madre Pluripotentes Inducidas/metabolismo , Esquizofrenia Infantil/genética , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Variaciones en el Número de Copia de ADN/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , MicroARNs/genética
7.
Int J Mol Sci ; 19(3)2018 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-29517996

RESUMEN

Bipolar disease (BD) is one of the major public health burdens worldwide and more people are affected every year. Comprehensive genetic studies have associated thousands of single nucleotide polymorphisms (SNPs) with BD risk; yet, very little is known about their functional roles. Induced pluripotent stem cells (iPSCs) are powerful tools for investigating the relationship between genotype and phenotype in disease-relevant tissues and cell types. Neural cells generated from BD-specific iPSCs are thought to capture associated genetic risk factors, known and unknown, and to allow the analysis of their effects on cellular and molecular phenotypes. Interestingly, an increasing number of studies on BD-derived iPSCs report distinct alterations in neural patterning, postmitotic calcium signaling, and neuronal excitability. Importantly, these alterations are partly normalized by lithium, a first line treatment in BD. In light of these exciting findings, we discuss current challenges to the field of iPSC-based disease modelling and future steps to be taken in order to fully exploit the potential of this approach for the investigation of BD and the development of new therapies.


Asunto(s)
Trastorno Bipolar/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Trastorno Bipolar/genética , Señalización del Calcio , Diferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/citología , Potenciales de la Membrana , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/fisiología , Cultivo Primario de Células/métodos
8.
Int J Mol Sci ; 18(8)2017 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-28777307

RESUMEN

Brain development is guided by the interactions between the genetic blueprint and the environment. Epigenetic mechanisms, especially DNA methylation, can mediate these interactions and may also trigger long-lasting adaptations in developmental programs that increase the risk of major depressive disorders (MDD) and schizophrenia (SCZ). Early life adversity is a major risk factor for MDD/SCZ and can trigger persistent genome-wide changes in DNA methylation at genes important to early, but also to mature, brain function, including neural proliferation, differentiation, and synaptic plasticity, among others. Moreover, genetic variations controlling dynamic DNA methylation in early life are thought to influence later epigenomic changes in SCZ. This finding corroborates the high genetic load and a neurodevelopmental origin of SCZ and shows that epigenetic responses to the environment are, at least in part, genetically controlled. Interestingly, genetic variants influencing DNA methylation are also enriched in risk variants from genome-wide association studies (GWAS) on SCZ supporting a role in neurodevelopment. Overall, epigenomic responses to early life adversity appear to be controlled to different degrees by genetics in MDD/SCZ, even though the potential reversibility of epigenomic processes may offer new hope for timely therapeutic interventions in MDD/SCZ.


Asunto(s)
Trastorno Depresivo Mayor/genética , Epigenómica , Acontecimientos que Cambian la Vida , Esquizofrenia/genética , Conducta , Epigénesis Genética , Humanos
9.
Stem Cells ; 31(8): 1621-32, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23630160

RESUMEN

Cell-fate decisions and differentiation of embryonic and adult neural stem cells (NSC) are tightly controlled by lineage-restricted and temporal factors that interact with cell-intrinsic programs and extracellular signals through multiple regulatory loops. Imprinted genes are important players in neurodevelopment and mental health although their molecular and cellular functions remain poorly understood. Here, we show that the paternally expressed transcriptional regulator Zac1 (zinc finger protein regulating apoptosis and cell cycle arrest) is transiently induced during astroglial and neuronal differentiation of embryonic and adult NSC lines. Thereby, Zac1 transactivates Socs3 (suppressor of cytokine signaling 3), a potent inhibitor of prodifferentiative Jak/Stat3 signaling, in a lineage-specific manner to prevent precocious astroglial differentiation. In vivo, Zac1 and Socs3 colocalize in the neocortical ventricular zone during incipient astrogliogenesis. Zac1 overexpression in primary NSCs delays astroglial differentiation whereas knockdown of Zac1 or Socs3 facilitates formation of astroglial cells. This negative feedback loop is unrelated to Zac1's cell cycle arrest function and specific to the Jak/Stat3 pathway. Hence, reinstating Jak/Stat3 signaling in the presence of increased Zac1 expression allows for timely astroglial differentiation. Overall, we suggest that the imprinted gene Zac1 curtails astroglial differentiation of NSCs in the developing and adult brain.


Asunto(s)
Astrocitos/citología , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/metabolismo , Células-Madre Neurales/citología , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Factores de Transcripción/biosíntesis , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Animales , Apoptosis/fisiología , Astrocitos/metabolismo , Proteínas de Ciclo Celular/genética , Diferenciación Celular/fisiología , Metilación de ADN , Regulación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Quinasas Janus/metabolismo , Ratones , Plásmidos/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Factores de Transcripción/genética , Activación Transcripcional , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
bioRxiv ; 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-38260577

RESUMEN

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.

11.
Curr Psychiatry Rep ; 14(2): 138-49, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22307409

RESUMEN

DNA methylation and chromatin modifications regulate gene expression and contribute to changes in brain transcriptomes underlying neurodevelopmental and psychiatric disorders. Clinical genetics and preclinical animal models highlight the crucial importance of the correct establishment of epigenetic marks during sensitive windows of development for normal brain function. On the same side of the coin, some of the concerned factors also appear engaged in the programming of experience-dependent long-term effects on mental health following exposure to relevant early-life events. Delineating the particular role of genetic variations in these players could provide new insights into the molecular basis of vulnerability and resilience and advance tailored therapies.


Asunto(s)
Epigénesis Genética/genética , Trastornos Mentales/genética , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Cromatina/metabolismo , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Variación Genética , Histonas/metabolismo , Humanos , Ratones , Modelos Biológicos
12.
Eur J Pharm Sci ; 175: 106216, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35618202

RESUMEN

A strong inflammatory immune response drives the lung pathology in neonatal acute respiratory distress syndrome (nARDS). Anti-inflammatory therapy is therefore a promising strategy for improved treatment of nARDS. We demonstrate a new function of the anionic phospholipids POPG, DOPG, and PIP2 as inhibitors of IL-1ß release by LPS and ATP-induced inflammasome activation in human monocyte-derived and lung macrophages. Curosurf® surfactant was enriched with POPG, DOPG, PIP2 and the head-group derivative IP3, biophysically characterized and applicability was evaluated in a piglet model of nARDS. The composition of pulmonary surfactant from piglets was determined by shotgun lipidomics screens. After 72 h of nARDS, levels of POPG, DOPG, and PIP2 were enhanced in the respective treatment groups. Otherwise, we did not observe changes of individual lipid species in any of the groups. Surfactant proteins were not affected, with the exception of the IP3 treated group. Our data show that POPG, DOPG, and PIP2 are potent inhibitors of inflammasome activation; their enrichment in a surfactant preparation did not induce any negative effects on lipid profile and reduced biophysical function in vitro was mainly observed for PIP2. These results encourage to rethink the current strategies of improving surfactant preparations by inclusion of anionic lipids as potent anti-inflammatory immune regulators.


Asunto(s)
Surfactantes Pulmonares , Síndrome de Dificultad Respiratoria , Animales , Antiinflamatorios/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Inflamasomas/metabolismo , Lipidómica , Pulmón/metabolismo , Fosfolípidos/farmacología , Surfactantes Pulmonares/metabolismo , Surfactantes Pulmonares/farmacología , Tensoactivos , Porcinos
13.
Stress ; 14(6): 581-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21854166

RESUMEN

Stress during early life can impact the developing brain and increase vulnerability to mood disorders later in life. Here, we argue that epigenetic mechanisms can mediate the gene-environment dialogue in early life and give rise to persistent epigenetic programming of adult physiology eventually resulting in disease. Early life stress in mice leads to epigenetic marking of the arginine vasopressin (AVP) gene underpinning sustained expression and increased hypothalamic-pituitary-adrenal axis activity. This epigenetic memory is laid down in the parvocellular neurons of the paraventricular nucleus and involves Ca(2+)/calmodulin kinase-mediated phosphorylation of the methyl-CpG binding domain protein MeCP2 leading to dissociation from its DNA-binding site and derepression of the AVP gene. The reduced occupancy of MeCP2 during this early stage of life facilitates the development of hypomethylation at the AVP enhancer, which sustains derepression throughout later life and thereby serves to hardwire early life experiences. The sequential order of these events may represent a critical time window for the preventive therapy of severe trauma.


Asunto(s)
Arginina Vasopresina/genética , Epigénesis Genética , Sistema Hipotálamo-Hipofisario/fisiopatología , Proteína 2 de Unión a Metil-CpG/metabolismo , Sistema Hipófiso-Suprarrenal/fisiopatología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Metilación de ADN , Sistema Hipotálamo-Hipofisario/fisiología , Privación Materna , Ratones , Núcleo Hipotalámico Paraventricular/fisiología , Ratas
14.
Pediatr Crit Care Med ; 12(6): e376-82, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21499172

RESUMEN

OBJECTIVE: A device for the application of continuous positive airway pressure to switch injected breathing gas to the outlet during expiration, known as Infant Flow, claims to reduce work of breathing and peak pressure change. So far the Infant Flow system has been investigated in lung models with tidal volumes of not <12 mL. However, premature neonates below 1000 g of weight generate a tidal volume of approximately 4 mL only. The aim of this study was to compare work of breathing and peak pressure change of the Infant Flow and another system that uses nasal prongs, Baby Flow, with conventional continuous positive airway pressure delivered by a pharyngeal tube. DESIGN: Laboratory investigation, basic research. SETTING: University research laboratory. MODEL: A piston pump simulating the spontaneous breathing of premature neonates was connected without leak to three different continuous positive airway pressure devices (pharyngeal tube, Baby Flow, and Infant Flow) and with a produced leak to the systems using nasal prongs (Baby Flow and Infant Flow). INTERVENTION: The pressures of the airway and continuous positive airway pressure systems and airway flow were recorded. Peak pressure change and work of breathing were determined for all systems and settings. Percentages of reduction of peak pressure change and work of breathing in relation to the continuous positive airway pressure delivered by pharyngeal tube were calculated. MEASUREMENTS AND MAIN RESULTS: The switching of injected breathing gas to the outlet during expiration of Infant Flow systems require a tidal volume of at least 5 mL. It was possible to decrease peak pressure change and work of breathing: Baby Flow system at a tidal volume of 4 mL (Inspiration: peak pressure change 82%, work of breathing 80%; Expiration: peak pressure change: 68%, work of breathing: 61%) and at a tidal volume of 8 mL (Inspiration: peak pressure change 75%, work of breathing 73%; Expiration: peak pressure change: 67%, work of breathing: 57%). Infant Flow system at tidal volume of 4 mL (Inspiration: peak pressure change 50%, work of breathing 55%; Expiration: peak pressure change: 46%, work of breathing: 43%) and at a tidal volume of 8 mL (Inspiration: peak pressure change 47%, work of breathing 46%; Expiration: peak pressure change: 24%, work of breathing: 23%), related to the continuous positive airway pressure delivered by pharyngeal tube without leak.Even under conditions of leak peak pressure change and work of breathing could be reduced: Baby Flow system at a tidal volume of 4 mL (Inspiration: peak pressure change 59%, work of breathing 64%; Expiration: peak pressure change: 68%, work of breathing: 59%) and at a tidal volume of 8 mL (Inspiration: peak pressure change 45%, work of breathing 43%; Expiration: peak pressure change: 54%, work of breathing: 53%). Infant Flow system at a tidal volume of 4 mL (Inspiration: peak pressure change 49%, work of breathing 53%; Expiration: peak pressure change: 44%, work of breathing: 40%) and at a tidal volume of 8 mL (Inspiration: peak pressure change 48%, work of breathing 43%; Expiration: peak pressure change: 36%, work of breathing: 40%), related to the continuous positive airway pressure delivered by pharyngeal tube without leak. CONCLUSION: Peak pressure change and work of breathing were decreased by Baby Flow and Infant Flow systems, even under conditions of leak.


Asunto(s)
Presión de las Vías Aéreas Positiva Contínua/normas , Pulmón , Modelos Biológicos , Nacimiento Prematuro , Trabajo Respiratorio/fisiología , Presión de las Vías Aéreas Positiva Contínua/instrumentación , Humanos , Recién Nacido , Masculino , Intercambio Gaseoso Pulmonar
15.
J Clin Med ; 10(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33477995

RESUMEN

Chromodomain Helicase DNA-binding 8 (CHD8) is a high confidence risk factor for autism spectrum disorders (ASDs) and the genetic cause of a distinct neurodevelopmental syndrome with the core symptoms of autism, macrocephaly, and facial dysmorphism. The role of CHD8 is well-characterized at the structural, biochemical, and transcriptional level. By contrast, much less is understood regarding how mutations in CHD8 underpin altered brain function and mental disease. Studies on various model organisms have been proven critical to tackle this challenge. Here, we scrutinize recent advances in this field with a focus on phenotypes in transgenic animal models and highlight key findings on neurodevelopment, neuronal connectivity, neurotransmission, synaptic and homeostatic plasticity, and habituation. Against this backdrop, we further discuss how to improve future animal studies, both in terms of technical issues and with respect to the sex-specific effects of Chd8 mutations for neuronal and higher-systems level function. We also consider outstanding questions in the field including 'humanized' mice models, therapeutic interventions, and how the use of pluripotent stem cell-derived cerebral organoids might help to address differences in neurodevelopment trajectories between model organisms and humans.

16.
Cells ; 9(2)2020 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-32033412

RESUMEN

Genome-wide association studies (GWAS) have identified an increasing number of genetic variants that significantly associate with psychiatric disorders. Despite this wealth of information, our knowledge of which variants causally contribute to disease, how they interact, and even more so of the functions they regulate, is still poor. The availability of embryonic stem cells (ESCs) and the advent of patient-specific induced pluripotent stem cells (iPSCs) has opened new opportunities to investigate genetic risk variants in living disease-relevant cells. Here, we analyze how this progress has contributed to the analysis of causal relationships between genetic risk variants and neuronal phenotypes, especially in schizophrenia (SCZ) and bipolar disorder (BD). Studies on rare, highly penetrant risk variants have originally led the field, until more recently when the development of (epi-) genetic editing techniques spurred studies on cause-effect relationships between common low risk variants and their associated neuronal phenotypes. This reorientation not only offers new insights, but also raises issues on interpretability. Concluding, we consider potential caveats and upcoming developments in the field of ESC/iPSC-based modeling of causality in psychiatric disorders.


Asunto(s)
Células Madre Embrionarias Humanas/patología , Células Madre Pluripotentes Inducidas/patología , Trastornos Mentales/patología , Modelos Biológicos , Causalidad , Estudio de Asociación del Genoma Completo , Humanos
17.
J Hypertens ; 27(2): 314-21, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19155788

RESUMEN

OBJECTIVES: ZAC1 (zinc finger protein regulating apoptosis and cell cycle arrest) is a member of the new subfamily of zinc-finger transcription factors, designated as PLAG (pleomorphic adenoma gene) family. The ZAC1 gene is maternally imprinted and is linked to developmental disorders such as growth retardation and transient neonatal diabetes mellitus. We wanted to assess whether the genetic variability of the ZAC1 gene was associated with anthropometric (weight, BMI, waist-to-hip ratio) or biochemical (plasma lipid, insulin, glucose levels, blood pressure level) phenotypes. METHODS: We selected 37 independent SNPs (single nucleotide polymorphisms) or tagSNPs in the ZAC1 locus from the literature and several databases and, based on the linkage disequilibrium map, identified 27 independent SNPs. Those 27 SNPs were genotyped in a French population-based sample (n = 1155). Associations with a P value lower than 0.0019 (Bonferroni correction) were considered significant. RESULTS: We found that women carrying the T allele of rs9403542 had lower waist-to-hip ratio (P = 0.0006) than women with the CC genotype. Also, men bearing the T allele of rs13218225 had lower systolic (P = 3.6 x 10(-5)) and diastolic (P = 4.1 x 10(-4)) blood pressure than GG men. As a consequence, the adjusted (for age, smoking habit, alcohol consumption, physical activity level and BMI) odds ratio (95% confidence interval) of hypertension for T allele carrier men was 0.55 [0.35-0.86], P = 0.009. We genotyped two other independent samples (MONICA Toulouse, n = 1130 and MONICA Strasbourg, n = 1048) for rs9403542 and rs13218225 but we could not confirm these associations. CONCLUSION: We found no evidence that polymorphisms in ZAC1 might influence anthropometric, biochemical or clinical parameters in French individuals.


Asunto(s)
Proteínas de Ciclo Celular/genética , Diabetes Mellitus/genética , Hiperlipidemias/genética , Hipertensión/genética , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Adulto , Anciano , Femenino , Francia , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Fenotipo , Polimorfismo de Nucleótido Simple , Población Blanca
18.
Mol Cell Biol ; 26(14): 5544-57, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16809786

RESUMEN

Zac is a C(2)H(2) zinc finger protein that regulates apoptosis and cell cycle arrest through DNA binding and transactivation. The coactivator proteins p300/CBP enhance transactivation through their histone acetyltransferase (HAT) activity by modulating chromatin structure. Here, we show that p300 increases Zac transactivation in a strictly HAT-dependent manner. Whereas the classic recruitment model proposes that coactivation simply depends on the capacity of the activator to recruit the coactivator, we demonstrate that coordinated binding of Zac zinc fingers and C terminus to p300 regulates HAT function by increasing histone and acetyl coenzyme A affinities and catalytic activity. This concerted regulation of HAT function is mediated via the KIX and CH3 domains of p300 in an interdependent manner. Interestingly, Zac zinc fingers 6 and 7 simultaneously play key roles in DNA binding and p300 regulation. Our findings demonstrate, for the first time, that C(2)H(2) zinc fingers can link DNA binding to HAT signaling and suggest a dynamic role for DNA-binding proteins in the enzymatic control of transcription.


Asunto(s)
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , ADN/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Histona Acetiltransferasas/metabolismo , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Proteínas de Ciclo Celular/genética , ADN/genética , Células HeLa , Humanos , Cinética , Células LLC-PK1 , Sustancias Macromoleculares , Unión Proteica , Estructura Terciaria de Proteína , Porcinos , Factores de Transcripción/genética , Activación Transcripcional , Proteínas Supresoras de Tumor/genética , Dedos de Zinc
19.
Front Genet ; 10: 682, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31396263

RESUMEN

The nucleosome remodeling and deacetylase (NuRD) complex presents one of the major chromatin remodeling complexes in mammalian cells. Here, we discuss current evidence for NuRD's role as an important epigenetic regulator of gene expression in neural stem cell (NSC) and neural progenitor cell (NPC) fate decisions in brain development. With the formation of the cerebellar and cerebral cortex, NuRD facilitates experience-dependent cerebellar plasticity and regulates additionally cerebral subtype specification and connectivity in postmitotic neurons. Consistent with these properties, genetic variation in NuRD's subunits emerges as important risk factor in common polygenic forms of neurodevelopmental disorders (NDDs) and neurodevelopment-related psychiatric disorders such as schizophrenia (SCZ) and bipolar disorder (BD). Overall, these findings highlight the critical role of NuRD in chromatin regulation in brain development and in mental health and disease.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA