Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 567(7749): 535-539, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30867594

RESUMEN

Chemical modifications of histones can mediate diverse DNA-templated processes, including gene transcription1-3. Here we provide evidence for a class of histone post-translational modification, serotonylation of glutamine, which occurs at position 5 (Q5ser) on histone H3 in organisms that produce serotonin (also known as 5-hydroxytryptamine (5-HT)). We demonstrate that tissue transglutaminase 2 can serotonylate histone H3 tri-methylated lysine 4 (H3K4me3)-marked nucleosomes, resulting in the presence of combinatorial H3K4me3Q5ser in vivo. H3K4me3Q5ser displays a ubiquitous pattern of tissue expression in mammals, with enrichment observed in brain and gut, two organ systems responsible for the bulk of 5-HT production. Genome-wide analyses of human serotonergic neurons, developing mouse brain and cultured serotonergic cells indicate that H3K4me3Q5ser nucleosomes are enriched in euchromatin, are sensitive to cellular differentiation and correlate with permissive gene expression, phenomena that are linked to the potentiation of TFIID4-6 interactions with H3K4me3. Cells that ectopically express a H3 mutant that cannot be serotonylated display significantly altered expression of H3K4me3Q5ser-target loci, which leads to deficits in differentiation. Taken together, these data identify a direct role for 5-HT, independent from its contributions to neurotransmission and cellular signalling, in the mediation of permissive gene expression.


Asunto(s)
Regulación de la Expresión Génica , Histonas/química , Histonas/metabolismo , Lisina/metabolismo , Procesamiento Proteico-Postraduccional , Serotonina/metabolismo , Factor de Transcripción TFIID/metabolismo , Animales , Diferenciación Celular , Línea Celular , Femenino , Proteínas de Unión al GTP/metabolismo , Glutamina/química , Glutamina/metabolismo , Humanos , Metilación , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Neuronas Serotoninérgicas/citología , Transglutaminasas/metabolismo
2.
Cell ; 156(5): 1114-1114.e1, 2014 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-24581504
3.
Mol Psychiatry ; 26(7): 2753-2763, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33767349

RESUMEN

The serotonergic system in the human brain modulates several physiological processes, and altered serotonergic neurotransmission has been implicated in the neuropathology of several psychiatric disorders. The study of serotonergic neurotransmission in psychiatry has long been restricted to animal models, but advances in cell reprogramming technology have enabled the generation of serotonergic neurons from patient-induced pluripotent stem cells (iPSCs). While iPSC-derived human serotonergic neurons offer the possibility to study serotonin (5-HT) release and uptake, particularly by 5-HT-modulating drugs such as selective serotonin reuptake inhibitors (SSRIs), a major limitation is the inability to reliably quantify 5-HT secreted from neurons in vitro. Herein, we address this technical gap via a novel sensing technology that couples 5-HT-specific DNA aptamers into nanopores (glass nanopipettes) with orifices of ~10 nm to detect 5-HT in complex neuronal culture medium with higher selectivity, sensitivity, and stability than existing methods. The 5-HT aptamers undergo conformational rearrangement upon target capture and serve as gatekeepers of ionic flux through the nanopipette opening. We generated human serotonergic neurons in vitro and detected secreted 5-HT using aptamer-coated nanopipettes in a low nanomolar range, with the possibility of detecting significantly lower (picomolar) concentrations. Furthermore, as a proof of concept, we treated human serotonergic neurons in vitro with the SSRI citalopram and detected a significant increase in extracellular 5-HT using the aptamer-modified nanopipettes. We demonstrate the utility of such methods for 5-HT detection, raising the possibility of fast quantification of neurotransmitters secreted from patient-derived live neuronal cells.


Asunto(s)
Neuronas Serotoninérgicas , Serotonina , Animales , Encéfalo , Citalopram/farmacología , Humanos , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología
4.
Mol Psychiatry ; 24(6): 808-818, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30903001

RESUMEN

Disrupted serotonergic neurotransmission has long been implicated in major depressive disorder (MDD), for which selective serotonin reuptake inhibitors (SSRIs) are the first line of treatment. However, a significant percentage of patients remain SSRI-resistant and it is unclear whether and how alterations in serotonergic neurons contribute to SSRI resistance in these patients. Induced pluripotent stem cells (iPSCs) facilitate the study of patient-specific neural subtypes that are typically inaccessible in living patients, enabling the discovery of disease-related phenotypes. In our study of a well-characterized cohort of over 800 MDD patients, we generated iPSCs and serotonergic neurons from three extreme SSRI-remitters (R) and SSRI-nonremitters (NR). We studied serotonin (5-HT) biochemistry and observed no significant differences in 5-HT release and reuptake or in genes related to 5-HT biochemistry. NR patient-derived serotonergic neurons exhibited altered neurite growth and morphology downstream of lowered expression of key Protocadherin alpha genes as compared to healthy controls and Rs. Furthermore, knockdown of Protocadherin alpha genes directly regulated iPSC-derived neurite length and morphology. Our results suggest that intrinsic differences in serotonergic neuron morphology and the resulting circuitry may contribute to SSRI resistance in MDD patients.


Asunto(s)
Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Trastorno Depresivo Resistente al Tratamiento/fisiopatología , Serotonina/metabolismo , Adulto , Antidepresivos/uso terapéutico , Estudios de Cohortes , Trastorno Depresivo Mayor/tratamiento farmacológico , Femenino , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Persona de Mediana Edad , Neuronas , Neuronas Serotoninérgicas/fisiología , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Transmisión Sináptica
5.
Mol Psychiatry ; 24(6): 795-807, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30700803

RESUMEN

Selective serotonin reuptake inhibitors (SSRIs) are the most prescribed antidepressants. They regulate serotonergic neurotransmission, but it remains unclear how altered serotonergic neurotransmission may contribute to the SSRI resistance observed in approximately 30% of major depressive disorder (MDD) patients. Patient stratification based on pharmacological responsiveness and the use of patient-derived neurons may make possible the discovery of disease-relevant neural phenotypes. In our study from a large cohort of well-characterized MDD patients, we have generated induced pluripotent stem cells (iPSCs) from SSRI-remitters and SSRI-nonremitters. We studied serotonergic neurotransmission in patient forebrain neurons in vitro and observed that nonremitter patient-derived neurons displayed serotonin-induced hyperactivity downstream of upregulated excitatory serotonergic receptors, in contrast to what is seen in healthy and remitter patient-derived neurons. Our data suggest that postsynaptic forebrain hyperactivity downstream of SSRI treatment may play a role in SSRI resistance in MDD.


Asunto(s)
Trastorno Depresivo Resistente al Tratamiento/tratamiento farmacológico , Trastorno Depresivo Resistente al Tratamiento/fisiopatología , Serotonina/metabolismo , Adulto , Acatisia Inducida por Medicamentos/fisiopatología , Antidepresivos/uso terapéutico , Estudios de Cohortes , Trastorno Depresivo Mayor/tratamiento farmacológico , Femenino , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Persona de Mediana Edad , Neuronas , Agitación Psicomotora/metabolismo , Inhibidores Selectivos de la Recaptación de Serotonina/farmacología , Inhibidores Selectivos de la Recaptación de Serotonina/uso terapéutico , Transmisión Sináptica
6.
Cell Tissue Res ; 371(1): 161-170, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28812143

RESUMEN

Several lines of evidence implicate serotonin in the etiology of multiple psychiatric disorders, especially mood disorders, such as major depressive disorder (MDD) and bipolar disorder (BD). Much of our current understanding of biological mechanisms underlying serotonergic alterations in mood disorders comes from animal studies. Innovation in induced pluripotent stem cell and transdifferentiation technologies for deriving neurons from adult humans has enabled the study of disease-relevant cellular phenotypes in vitro. In this context, human serotonergic neurons can now be generated using three recently published methodologies. In this mini-review, we broadly discuss evidence linking altered serotonergic neurotransmission in MDD and BD and focus on recently published methods for generating human serotonergic neurons in vitro.


Asunto(s)
Trastorno Bipolar/fisiopatología , Trastorno Depresivo Mayor/fisiopatología , Células Madre Pluripotentes Inducidas/fisiología , Modelos Neurológicos , Neuronas Serotoninérgicas/fisiología , Serotonina/fisiología , Adulto , Animales , Técnicas de Cultivo de Célula , Transdiferenciación Celular , Humanos , Ratones , Transmisión Sináptica
7.
Mol Psychiatry ; 22(6): 820-835, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27378147

RESUMEN

Autism spectrum disorders (ASD) are common, complex and heterogeneous neurodevelopmental disorders. Cellular and molecular mechanisms responsible for ASD pathogenesis have been proposed based on genetic studies, brain pathology and imaging, but a major impediment to testing ASD hypotheses is the lack of human cell models. Here, we reprogrammed fibroblasts to generate induced pluripotent stem cells, neural progenitor cells (NPCs) and neurons from ASD individuals with early brain overgrowth and non-ASD controls with normal brain size. ASD-derived NPCs display increased cell proliferation because of dysregulation of a ß-catenin/BRN2 transcriptional cascade. ASD-derived neurons display abnormal neurogenesis and reduced synaptogenesis leading to functional defects in neuronal networks. Interestingly, defects in neuronal networks could be rescued by insulin growth factor 1 (IGF-1), a drug that is currently in clinical trials for ASD. This work demonstrates that selection of ASD subjects based on endophenotypes unraveled biologically relevant pathway disruption and revealed a potential cellular mechanism for the therapeutic effect of IGF-1.


Asunto(s)
Trastorno Autístico/metabolismo , Trastorno Autístico/patología , Técnicas de Cultivo de Tejidos/métodos , Adolescente , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/fisiopatología , Encéfalo/metabolismo , Proliferación Celular/genética , Células Cultivadas , Niño , Preescolar , Femenino , Fibroblastos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Factor I del Crecimiento Similar a la Insulina/uso terapéutico , Masculino , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/metabolismo , Neuronas/fisiología , beta Catenina/metabolismo
8.
Bioessays ; 38(11): 1123-1129, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27716980

RESUMEN

Technologies for deriving human neurons in vitro have transformed our ability to study cellular and molecular components of human neurotransmission. Three groups, including our own, have recently published methods for efficiently generating human serotonergic neurons in vitro. Remarkably, serotonergic neurons derived from each method robustly produce serotonin, express raphe genes, are electrically active, and respond to selective serotonin reuptake inhibitors in vitro. Two of the methods utilize transdifferentiation technology by overexpressing key serotonergic transcription factors. The third and most recent method involves differentiating induced pluripotent stem cells (iPSCs) to serotonergic neurons using developmental patterning cues. In this mini-review, we briefly describe the developmental programs governing serotonergic specification in vivo and how they have been harnessed to achieve serotonergic differentiation in vitro. We discuss the distinct and overlapping features of the recently published methodologies and their value in the context of in vitro disease modeling. Also see the video abstract here.


Asunto(s)
Técnicas de Reprogramación Celular/métodos , Neuronas Serotoninérgicas , Animales , Diferenciación Celular , Transdiferenciación Celular , Humanos , Células Madre Pluripotentes Inducidas/fisiología , Péptidos y Proteínas de Señalización Intercelular , Ratones , Factores de Transcripción
9.
J Neurosci ; 33(3): 1179-89, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23325254

RESUMEN

The molecular mechanisms underlying the generation, maturation, and integration of new granule cells generated throughout life in the mammalian hippocampus remain poorly understood. Small Rho GTPases, such as Cdc42 and Rac1, have been implicated previously in neural stem/progenitor cell (NSPC) proliferation and neuronal maturation during embryonic development. Here we used conditional genetic deletion and virus-based loss-of-function approaches to identify temporally distinct functions for Cdc42 and Rac1 in adult hippocampal neurogenesis. We found that Cdc42 is involved in mouse NSPC proliferation, initial dendritic development, and dendritic spine maturation. In contrast, Rac1 is dispensable for early steps of neuronal development but is important for late steps of dendritic growth and spine maturation. These results establish cell-autonomous and stage-specific functions for the small Rho GTPases Cdc42 and Rac1 in the course of adult hippocampal neurogenesis.


Asunto(s)
Hipocampo/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Linaje de la Célula/fisiología , Movimiento Celular/fisiología , Proliferación Celular , Dendritas/metabolismo , Espinas Dendríticas/metabolismo , Femenino , Ratones , Ratones Transgénicos , Ratas , Ratas Endogámicas F344 , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP rac1/genética
10.
Proc Natl Acad Sci U S A ; 108(14): 5807-12, 2011 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-21436036

RESUMEN

Neural stem cells (NSCs) generate new granule cells throughout life in the mammalian hippocampus. Canonical Wnt signaling regulates the differentiation of NSCs towards the neuronal lineage. Here we identified the prospero-related homeodomain transcription factor Prox1 as a target of ß-catenin-TCF/LEF signaling in vitro and in vivo. Prox1 overexpression enhanced neuronal differentiation whereas shRNA-mediated knockdown of Prox1 impaired the generation of neurons in vitro and within the hippocampal niche. In contrast, Prox1 was not required for survival of adult-generated granule cells after they had matured, suggesting a role for Prox1 in initial granule cell differentiation but not in the maintenance of mature granule cells. The data presented here characterize a molecular pathway from Wnt signaling to a transcriptional target leading to granule cell differentiation within the adult brain and identify a stage-specific function for Prox1 in the process of adult neurogenesis.


Asunto(s)
Diferenciación Celular/fisiología , Hipocampo/crecimiento & desarrollo , Proteínas de Homeodominio/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Transducción de Señal/fisiología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/metabolismo , Animales , Secuencia de Bases , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Hipocampo/citología , Proteínas de Homeodominio/genética , Inmunohistoquímica , Hibridación in Situ , Luciferasas , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Supresoras de Tumor/genética
11.
Neurobiol Stress ; 23: 100530, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36891528

RESUMEN

Hypothalamic-pituitary adrenal (HPA)axis dysregulation has long been implicated in stress-related disorders such as major depression and post-traumatic stress disorder. Glucocorticoids (GCs) are released from the adrenal glands as a result of HPA-axis activation. The release of GCs is implicated with several neurobiological changes that are associated with negative consequences of chronic stress and the onset and course of psychiatric disorders. Investigating the underlying neurobiological effects of GCs may help to better understand the pathophysiology of stress-related psychiatric disorders. GCs impact a plethora of neuronal processes at the genetic, epigenetic, cellular, and molecular levels. Given the scarcity and difficulty in accessing human brain samples, 2D and 3D in vitro neuronal cultures are becoming increasingly useful in studying GC effects. In this review, we provide an overview of in vitro studies investigating the effects of GCs on key neuronal processes such as proliferation and survival of progenitor cells, neurogenesis, synaptic plasticity, neuronal activity, inflammation, genetic vulnerability, and epigenetic alterations. Finally, we discuss the challenges in the field and offer suggestions for improving the use of in vitro models to investigate GC effects.

12.
IBRO Neurosci Rep ; 13: 420-425, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36386600

RESUMEN

Norepinephrine (NE), and specific adrenoceptors, have been reported to influence distinct aspects of adult hippocampal neurogenesis, including latent stem cell activation, progenitor proliferation, and differentiation. These findings are predominantly based on the use of pharmacological approaches in both in vitro and in vivo systems. Here, we sought to assess the consequences of genetic ablation of NE on adult hippocampal neurogenesis, by examining dopamine ß hydroxylase knockout (Dbh -/-) mice, which lack NE from birth. We find that Dbh -/- mice exhibit no difference in adult hippocampal progenitor proliferation and survival. Further, the number of immature newborn neurons, labeled using stage-specific developmental markers within the hippocampal neurogenic niche, was also unaltered in Dbh -/- mice. In contrast, the noradrenergic neurotoxin DSP-4, which had previously been shown to reduce adult hippocampal neurogenesis in rats, also resulted in a decline in hippocampal progenitor proliferation in C57/Bl6N mice. These findings indicate that pharmacological lesioning of noradrenergic afferents in adulthood, but not the complete genetic loss of NE from birth, impairs adult hippocampal neurogenesis in mice.

13.
J Neurosci ; 30(3): 1096-109, 2010 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-20089918

RESUMEN

Slow-onset adaptive changes that arise from sustained antidepressant treatment, such as enhanced adult hippocampal neurogenesis and increased trophic factor expression, play a key role in the behavioral effects of antidepressants. alpha(2)-Adrenoceptors contribute to the modulation of mood and are potential targets for the development of faster acting antidepressants. We investigated the influence of alpha(2)-adrenoceptors on adult hippocampal neurogenesis. Our results indicate that alpha(2)-adrenoceptor agonists, clonidine and guanabenz, decrease adult hippocampal neurogenesis through a selective effect on the proliferation, but not the survival or differentiation, of progenitors. These effects persist in dopamine beta-hydroxylase knock-out (Dbh(-/-)) mice lacking norepinephrine, supporting a role for alpha(2)-heteroceptors on progenitor cells, rather than alpha(2)-autoreceptors on noradrenergic neurons that inhibit norepinephrine release. Adult hippocampal progenitors in vitro express all the alpha(2)-adrenoceptor subtypes, and decreased neurosphere frequency and BrdU incorporation indicate direct effects of alpha(2)-adrenoceptor stimulation on progenitors. Furthermore, coadministration of the alpha(2)-adrenoceptor antagonist yohimbine with the antidepressant imipramine significantly accelerates effects on hippocampal progenitor proliferation, the morphological maturation of newborn neurons, and the increase in expression of brain derived neurotrophic factor and vascular endothelial growth factor implicated in the neurogenic and behavioral effects of antidepressants. Finally, short-duration (7 d) yohimbine and imipramine treatment results in robust behavioral responses in the novelty suppressed feeding test, which normally requires 3 weeks of treatment with classical antidepressants. Our results demonstrate that alpha(2)-adrenoceptors, expressed by progenitor cells, decrease adult hippocampal neurogenesis, while their blockade speeds up antidepressant action, highlighting their importance as targets for faster acting antidepressants.


Asunto(s)
Antagonistas de Receptores Adrenérgicos alfa 2 , Antidepresivos/farmacología , Conducta Animal/efectos de los fármacos , Hipocampo/efectos de los fármacos , Imipramina/farmacología , Neurogénesis/efectos de los fármacos , Agonistas alfa-Adrenérgicos/farmacología , Antagonistas Adrenérgicos alfa/farmacología , Agonistas Adrenérgicos beta/farmacología , Análisis de Varianza , Animales , Animales Recién Nacidos , Antiparkinsonianos/farmacología , Ácido Ascórbico/farmacología , Factor Neurotrófico Derivado del Encéfalo , Bromodesoxiuridina/metabolismo , Células Cultivadas , Dopamina beta-Hidroxilasa/genética , Proteínas de Dominio Doblecortina , Droxidopa/farmacología , Esquema de Medicación , Interacciones Farmacológicas , Embrión de Mamíferos , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Técnicas In Vitro , Isoproterenol/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuropéptidos/metabolismo , Fenilefrina/farmacología , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Wistar , Tiempo de Reacción/efectos de los fármacos , Receptores Adrenérgicos alfa 2/fisiología , Células Madre/efectos de los fármacos , Yohimbina/farmacología
14.
Transl Psychiatry ; 11(1): 608, 2021 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-34848679

RESUMEN

Major depressive disorder (MDD) is a prevalent psychiatric disorder, and exposure to stress is a robust risk factor for MDD. Clinical data and rodent models have indicated the negative impact of chronic exposure to stress-induced hormones like cortisol on brain volume, memory, and cell metabolism. However, the cellular and transcriptomic changes that occur in the brain after prolonged exposure to cortisol are less understood. Furthermore, the astrocyte-specific contribution to cortisol-induced neuropathology remains understudied. Here, we have developed an in vitro model of "chronic stress" using human induced pluripotent stem cell (iPSC)-derived astrocytes treated with cortisol for 7 days. Whole transcriptome sequencing reveals differentially expressed genes (DEGs) uniquely regulated in chronic cortisol compared to acute cortisol treatment. Utilizing this paradigm, we examined the stress response transcriptome of astrocytes generated from MDD patient iPSCs. The MDD-specific DEGs are related to GPCR ligand binding, synaptic signaling, and ion homeostasis. Together, these data highlight the unique role astrocytes play in the central nervous system and present interesting genes for future study into the relationship between chronic stress and MDD.


Asunto(s)
Trastorno Depresivo Mayor , Células Madre Pluripotentes Inducidas , Astrocitos , Humanos , Hidrocortisona , Ligandos , Receptores Acoplados a Proteínas G
15.
Stem Cell Reports ; 16(4): 825-835, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33667413

RESUMEN

Bipolar disorder (BD) is characterized by cyclical mood shifts. Studies indicate that BD patients have a peripheral pro-inflammatory state and alterations in glial populations in the brain. We utilized an in vitro model to study inflammation-related phenotypes of astrocytes derived from induced pluripotent stem cells (iPSCs) generated from BD patients and healthy controls. BD astrocytes showed changes in transcriptome and induced a reduction in neuronal activity when co-cultured with neurons. IL-1ß-stimulated BD astrocytes displayed a unique inflammatory gene expression signature and increased secretion of IL-6. Conditioned medium from stimulated BD astrocytes reduced neuronal activity, and this effect was partially blocked by IL-6 inactivating antibody. Our results suggest that BD astrocytes are functionally less supportive of neuronal excitability and this effect is partially mediated by IL-6. We confirmed higher IL-6 in blood in a distinct cohort of BD patients, highlighting the potential role of astrocyte-mediated inflammatory signaling in BD neuropathology.


Asunto(s)
Astrocitos/patología , Trastorno Bipolar/patología , Inflamación/patología , Neuronas/patología , Técnicas de Cocultivo , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Interleucina-1beta/farmacología , Interleucina-6/metabolismo , Neuroglía/efectos de los fármacos , Neuroglía/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo
16.
Artículo en Inglés | MEDLINE | ID: mdl-31767646

RESUMEN

Brain disorders, from neurodegenerative to psychiatric disorders, are among the most challenging conditions to study because of the intricate nature of the human brain and the limitations of existing model systems in recapitulating all these intricacies. However, innovations in stem cell technologies now allow us to reprogram patient somatic cells to induced pluripotent stem cells (iPSCs), which can then be differentiated to disease-relevant neural and glial cells. iPSCs are a valuable tool to model brain disorders, as they can be derived from patients with known symptom histories, genetics, and drug-response profiles. Here, we discuss the premise and validity of the iPSC-based in vitro model system and highlight key findings from the most commonly studied neurodegenerative and psychiatric disorders.


Asunto(s)
Encefalopatías/metabolismo , Encéfalo/fisiopatología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Encéfalo/fisiología , Diferenciación Celular , Sistema Nervioso Central , Química Farmacéutica/métodos , Diseño de Fármacos , Técnicas Genéticas , Humanos , Modelos Biológicos , Modelos Neurológicos , Enfermedades Neurodegenerativas , Neuroglía/metabolismo , Neuronas/metabolismo , Células Madre Pluripotentes/fisiología , Trasplante de Células Madre
17.
Focus (Am Psychiatr Publ) ; 17(3): 294-307, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32015721

RESUMEN

Reprinted with permission from Open Biol. 8: 180031. The Royal Society.

18.
Antioxid Redox Signal ; 31(4): 275-317, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30585734

RESUMEN

Significance: Our current knowledge of the pathophysiology and molecular mechanisms causing psychiatric disorders is modest, but genetic susceptibility and environmental factors are central to the etiology of these conditions. Autism, schizophrenia, bipolar disorder and major depressive disorder show genetic gene risk overlap and share symptoms and metabolic comorbidities. The identification of such common features may provide insights into the development of these disorders. Recent Advances: Multiple pieces of evidence suggest that brain energy metabolism, mitochondrial functions and redox balance are impaired to various degrees in psychiatric disorders. Since mitochondrial metabolism and redox signaling can integrate genetic and environmental environmental factors affecting the brain, it is possible that they are implicated in the etiology and progression of psychiatric disorders. Critical Issue: Evidence for direct links between cellular mitochondrial dysfunction and disease features are missing. Future Directions: A better understanding of the mitochondrial biology and its intracellular connections to the nuclear genome, the endoplasmic reticulum and signaling pathways, as well as its role in intercellular communication in the organism, is still needed. This review focuses on the findings that implicate mitochondrial dysfunction, the resultant metabolic changes and oxidative stress as important etiological factors in the context of psychiatric disorders. We also propose a model where specific pathophysiologies of psychiatric disorders depend on circuit-specific impairments of mitochondrial dysfunction and redox signaling at specific developmental stages.


Asunto(s)
Trastornos Mentales/metabolismo , Mitocondrias/metabolismo , Animales , Humanos , Oxidación-Reducción
19.
Elife ; 82019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30730291

RESUMEN

Comparative analyses of neuronal phenotypes in closely related species can shed light on neuronal changes occurring during evolution. The study of post-mortem brains of nonhuman primates (NHPs) has been limited and often does not recapitulate important species-specific developmental hallmarks. We utilize induced pluripotent stem cell (iPSC) technology to investigate the development of cortical pyramidal neurons following migration and maturation of cells grafted in the developing mouse cortex. Our results show differential migration patterns in human neural progenitor cells compared to those of chimpanzees and bonobos both in vitro and in vivo, suggesting heterochronic changes in human neurons. The strategy proposed here lays the groundwork for further comparative analyses between humans and NHPs and opens new avenues for understanding the differences in the neural underpinnings of cognition and neurological disease susceptibility between species.


Asunto(s)
Neuronas/citología , Pan paniscus/fisiología , Pan troglodytes/fisiología , Animales , Diferenciación Celular , Línea Celular , Movimiento Celular/genética , Dendritas/metabolismo , Regulación de la Expresión Génica , Humanos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Especificidad de la Especie
20.
Genome Med ; 10(1): 1, 2018 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-29301565

RESUMEN

Our understanding of the neurobiology of psychiatric disorders remains limited, and biomarker-based clinical management is yet to be developed. Induced pluripotent stem cell (iPSC) technology has revolutionized our capacity to generate patient-derived neurons to model psychiatric disorders. Here, we highlight advantages and caveats of iPSC disease modeling and outline strategies for addressing current challenges.


Asunto(s)
Células Madre Pluripotentes Inducidas/patología , Trastornos Mentales/patología , Modelos Biológicos , Neuronas/patología , Animales , Humanos , Trastornos Mentales/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA