Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(17): e108780, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35815410

RESUMEN

Schwann cell precursors (SCPs) are nerve-associated progenitors that can generate myelinating and non-myelinating Schwann cells but also are multipotent like the neural crest cells from which they originate. SCPs are omnipresent along outgrowing peripheral nerves throughout the body of vertebrate embryos. By using single-cell transcriptomics to generate a gene expression atlas of the entire neural crest lineage, we show that early SCPs and late migratory crest cells have similar transcriptional profiles characterised by a multipotent "hub" state containing cells biased towards traditional neural crest fates. SCPs keep diverging from the neural crest after being primed towards terminal Schwann cells and other fates, with different subtypes residing in distinct anatomical locations. Functional experiments using CRISPR-Cas9 loss-of-function further show that knockout of the common "hub" gene Sox8 causes defects in neural crest-derived cells along peripheral nerves by facilitating differentiation of SCPs towards sympathoadrenal fates. Finally, specific tumour populations found in melanoma, neurofibroma and neuroblastoma map to different stages of SCP/Schwann cell development. Overall, SCPs resemble migrating neural crest cells that maintain multipotency and become transcriptionally primed towards distinct lineages.


Asunto(s)
Cresta Neural , Células de Schwann , Diferenciación Celular/fisiología , Neurogénesis/fisiología , Nervios Periféricos , Células de Schwann/metabolismo
2.
Development ; 149(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35245348

RESUMEN

The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the polycomb repressor complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3, a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g. Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that most neuronal subtypes were still generated in Eed mutants. However, we observed an increase in glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, hypocretin and Tac2-Pax6 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, but points to a unique sensitivity of particular neuronal subtypes to a disrupted epigenomic landscape.


Asunto(s)
Desarrollo Embrionario/fisiología , Hipotálamo/fisiología , Neuronas/fisiología , Complejo Represivo Polycomb 2/genética , Proteínas del Grupo Polycomb/genética , Animales , Proliferación Celular/genética , Represión Epigenética/genética , Femenino , Masculino , Ratones , Mutación/genética , Transcriptoma/genética
3.
PLoS Genet ; 17(9): e1009822, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34570766

RESUMEN

Dopamine (DA) neurons of the midbrain are at risk to become affected by mitochondrial damage over time and mitochondrial defects have been frequently reported in Parkinson's disease (PD) patients. However, the causal contribution of adult-onset mitochondrial dysfunction to PD remains uncertain. Here, we developed a mouse model lacking Mitofusin 2 (MFN2), a key regulator of mitochondrial network homeostasis, in adult midbrain DA neurons. The knockout mice develop severe and progressive DA neuron-specific mitochondrial dysfunction resulting in neurodegeneration and parkinsonism. To gain further insights into pathophysiological events, we performed transcriptomic analyses of isolated DA neurons and found that mitochondrial dysfunction triggers an early onset immune response, which precedes mitochondrial swelling, mtDNA depletion, respiratory chain deficiency and cell death. Our experiments show that the immune response is an early pathological event when mitochondrial dysfunction is induced in adult midbrain DA neurons and that neuronal death may be promoted non-cell autonomously by the cross-talk and activation of surrounding glial cells.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Inmunidad , Mesencéfalo/metabolismo , Mitocondrias/metabolismo , Animales , ADN Mitocondrial/genética , Modelos Animales de Enfermedad , Homeostasis , Ratones , Trastornos Parkinsonianos/genética
4.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35805964

RESUMEN

The development of midbrain dopaminergic (DA) neurons requires a fine temporal and spatial regulation of a very specific gene expression program. Here, we report that during mouse brain development, the microRNA (miR-) 204/211 is present at a high level in a subset of DA precursors expressing the transcription factor Lmx1a, an early determinant for DA-commitment, but not in more mature neurons expressing Th or Pitx3. By combining different in vitro model systems of DA differentiation, we show that the levels of Lmx1a influence the expression of miR-204/211. Using published transcriptomic data, we found a significant enrichment of miR-204/211 target genes in midbrain dopaminergic neurons where Lmx1a was selectively deleted at embryonic stages. We further demonstrated that miR-204/211 controls the timing of the DA differentiation by directly downregulating the expression of Nurr1, a late DA differentiation master gene. Thus, our data indicate the Lmx1a-miR-204/211-Nurr1 axis as a key component in the cascade of events that ultimately lead to mature midbrain dopaminergic neurons differentiation and point to miR-204/211 as the molecular switch regulating the timing of Nurr1 expression.


Asunto(s)
Neuronas Dopaminérgicas , Proteínas con Homeodominio LIM , MicroARNs , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares , Animales , Diferenciación Celular/fisiología , Dopamina/metabolismo , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Mesencéfalo/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
5.
Proc Natl Acad Sci U S A ; 114(10): 2735-2740, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28137881

RESUMEN

Individuals with Parkinson's disease (PD) often suffer from comorbid depression. P11 (S100A10), a member of the S100 family of proteins, is expressed widely throughout the body and is involved in major depressive disorder and antidepressant response. Central p11 levels are reduced in postmortem tissue from depressed individuals; however, p11 has not yet been investigated in PD patients with depression or those without depression. We investigated p11 levels in postmortem PD brains and assessed whether peripheral p11 levels correlate with disease severity. Substantia nigra, putamen, and cortical p11 protein levels were assessed in postmortem brain samples from PD patients and matched controls. In a different set of postmortem brains, p11 mRNA expression was measured in dopaminergic cells from the substantia nigra. Both p11 protein and mRNA levels were decreased in PD patients. Peripheral p11 protein levels were investigated in distinct leukocyte populations from PD patients with depression and those without depression. Monocyte, natural killer (NK) cell, and cytotoxic T-cell p11 levels were positively associated with the severity of PD, and NK cell p11 levels were positively associated with depression scores. Given that inflammation plays a role in both PD and depression, it is intriguing that peripheral p11 levels are altered in immune cells in both conditions. Our data provide insight into the pathological alterations occurring centrally and peripherally in PD. Moreover, if replicated in other cohorts, p11 could be an easily accessible biomarker for monitoring the severity of PD, especially in the context of comorbid depression.


Asunto(s)
Anexina A2/genética , Trastorno Depresivo Mayor/genética , Enfermedad de Parkinson/genética , ARN Mensajero/genética , Proteínas S100/genética , Anciano , Anciano de 80 o más Años , Anexina A2/sangre , Autopsia , Encéfalo/metabolismo , Encéfalo/patología , Trastorno Depresivo Mayor/sangre , Trastorno Depresivo Mayor/complicaciones , Trastorno Depresivo Mayor/patología , Femenino , Regulación de la Expresión Génica/genética , Humanos , Células Asesinas Naturales/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Enfermedad de Parkinson/sangre , Enfermedad de Parkinson/complicaciones , Enfermedad de Parkinson/patología , ARN Mensajero/sangre , Proteínas S100/sangre , Índice de Severidad de la Enfermedad , Linfocitos T Citotóxicos/metabolismo
6.
Development ; 143(16): 2877-81, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27531947

RESUMEN

The third 'Stem Cell Niche' meeting, supported by The Novo Nordisk Foundation, was held this year on May 22-26 and brought together 185 selected participants from 24 different countries to Hillerød, Denmark. Diverse aspects of embryonic and adult stem cell biology were discussed, including their respective niches in ageing, disease and regeneration. Many presentations focused on emerging technologies, including single-cell analysis, in vitro organogenesis and direct reprogramming. Here, we summarize the data presented at this exciting and highly enjoyable meeting, where speakers as well as kitchen chefs were applauded at every session.


Asunto(s)
Nicho de Células Madre/fisiología , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Humanos , Análisis de la Célula Individual , Nicho de Células Madre/genética , Transcriptoma/genética
7.
Development ; 143(14): 2616-28, 2016 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-27287799

RESUMEN

Intestinal hormone-producing cells represent the largest endocrine system in the body, but remarkably little is known about enteroendocrine cell type specification in the embryo and adult. We analyzed stage- and cell type-specific deletions of Nkx2.2 and its functional domains in order to characterize its role in the development and maintenance of enteroendocrine cell lineages in the mouse duodenum and colon. Although Nkx2.2 regulates enteroendocrine cell specification in the duodenum at all stages examined, it controls the differentiation of progressively fewer enteroendocrine cell populations when deleted from Ngn3(+) progenitor cells or in the adult duodenum. During embryonic development Nkx2.2 regulates all enteroendocrine cell types, except gastrin and preproglucagon. In developing Ngn3(+) enteroendocrine progenitor cells, Nkx2.2 is not required for the specification of neuropeptide Y and vasoactive intestinal polypeptide, indicating that a subset of these cell populations derive from an Nkx2.2-independent lineage. In adult duodenum, Nkx2.2 becomes dispensable for cholecystokinin and secretin production. In all stages and Nkx2.2 mutant conditions, serotonin-producing enterochromaffin cells were the most severely reduced enteroendocrine lineage in the duodenum and colon. We determined that the transcription factor Lmx1a is expressed in enterochromaffin cells and functions downstream of Nkx2.2. Lmx1a-deficient mice have reduced expression of Tph1, the rate-limiting enzyme for serotonin biosynthesis. These data clarify the function of Nkx2.2 in the specification and homeostatic maintenance of enteroendocrine populations, and identify Lmx1a as a novel enterochromaffin cell marker that is also essential for the production of the serotonin biosynthetic enzyme Tph1.


Asunto(s)
Linaje de la Célula , Células Enterocromafines/citología , Células Enteroendocrinas/citología , Proteínas de Homeodominio/metabolismo , Proteínas con Homeodominio LIM/metabolismo , Serotonina/biosíntesis , Factores de Transcripción/metabolismo , Envejecimiento/metabolismo , Animales , Biomarcadores/metabolismo , Linaje de la Célula/genética , Colon/metabolismo , Duodeno/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/química , Ratones Endogámicos C57BL , Modelos Biológicos , Mutación/genética , Reacción en Cadena de la Polimerasa , Dominios Proteicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ARN , Células Madre/citología , Factores de Transcripción/química , Proteínas de Pez Cebra
8.
Proc Natl Acad Sci U S A ; 113(30): E4387-96, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27407143

RESUMEN

The LIM-homeodomain transcription factors Lmx1a and Lmx1b play critical roles during the development of midbrain dopaminergic progenitors, but their functions in the adult brain remain poorly understood. We show here that sustained expression of Lmx1a and Lmx1b is required for the survival of adult midbrain dopaminergic neurons. Strikingly, inactivation of Lmx1a and Lmx1b recreates cellular features observed in Parkinson's disease. We found that Lmx1a/b control the expression of key genes involved in mitochondrial functions, and their ablation results in impaired respiratory chain activity, increased oxidative stress, and mitochondrial DNA damage. Lmx1a/b deficiency caused axonal pathology characterized by α-synuclein(+) inclusions, followed by a progressive loss of dopaminergic neurons. These results reveal the key role of these transcription factors beyond the early developmental stages and provide mechanistic links between mitochondrial dysfunctions, α-synuclein aggregation, and the survival of dopaminergic neurons.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Proteínas con Homeodominio LIM/genética , Mesencéfalo/metabolismo , Mitocondrias/metabolismo , Factores de Transcripción/genética , Animales , Supervivencia Celular/genética , Daño del ADN , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Proteínas con Homeodominio LIM/deficiencia , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/genética , Estrés Oxidativo , Agregación Patológica de Proteínas , Factores de Transcripción/deficiencia , alfa-Sinucleína/metabolismo
9.
Genes Dev ; 25(19): 2031-40, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21979916

RESUMEN

DNA-dependent protein kinase (DNA-PK) is a central regulator of DNA double-strand break (DSB) repair; however, the identity of relevant DNA-PK substrates has remained elusive. NR4A nuclear orphan receptors function as sequence-specific DNA-binding transcription factors that participate in adaptive and stress-related cell responses. We show here that NR4A proteins interact with the DNA-PK catalytic subunit and, upon exposure to DNA damage, translocate to DSB foci by a mechanism requiring the activity of poly(ADP-ribose) polymerase-1 (PARP-1). At DNA repair foci, NR4A is phosphorylated by DNA-PK and promotes DSB repair. Notably, NR4A transcriptional activity is entirely dispensable in this function, and core components of the DNA repair machinery are not transcriptionally regulated by NR4A. Instead, NR4A functions directly at DNA repair sites by a process that requires phosphorylation by DNA-PK. Furthermore, a severe combined immunodeficiency (SCID)-causing mutation in the human gene encoding the DNA-PK catalytic subunit impairs the interaction and phosphorylation of NR4A at DSBs. Thus, NR4As represent an entirely novel component of DNA damage response and are substrates of DNA-PK in the process of DSB repair.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Línea Celular , Células Cultivadas , Técnicas de Inactivación de Genes , Humanos , Ratones , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Fosforilación , Transporte de Proteínas , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/fisiopatología
10.
Nat Rev Genet ; 13(6): 429-39, 2012 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-22596319

RESUMEN

Various studies have demonstrated that somatic differentiated cells can be reprogrammed into other differentiated states or into pluripotency, thus showing that the differentiated cellular state is not irreversible. These findings have generated intense interest in the process of reprogramming and in mechanisms that govern the pluripotent state. However, the realization that differentiated cells can be triggered to switch to considerably different lineages also emphasizes that we need to understand how the identity of mature cells is normally maintained. Here we review recent studies on how the differentiated state is controlled at the transcriptional level and discuss how new insights have begun to elucidate mechanisms underlying the stable maintenance of mature cell identities.


Asunto(s)
Desdiferenciación Celular/genética , Diferenciación Celular/genética , Transdiferenciación Celular/genética , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Redes Reguladoras de Genes/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Modelos Genéticos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/genética
11.
J Neurosci ; 35(42): 14370-85, 2015 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-26490873

RESUMEN

α-synuclein, a protein enriched in Lewy bodies and highly implicated in neurotoxicity in Parkinson's disease, is distributed both at nerve terminals and in the cell nucleus. Here we show that a nuclear derivative of α-synuclein induces more pronounced changes at the gene expression level in mouse primary dopamine (DA) neurons compared to a derivative that is excluded from the nucleus. Moreover, by RNA sequencing we analyzed the extent of genome-wide effects on gene expression resulting from expression of human α-synuclein in primary mouse DA neurons. The results implicated the transcription factor Nurr1 as a key dysregulated target of α-synuclein toxicity. Forced Nurr1 expression restored the expression of hundreds of dysregulated genes in primary DA neurons expressing α-synuclein, and therefore prompted us to test the possibility that Nurr1 can be pharmacologically targeted by bexarotene, a ligand for the retinoid X receptor that forms heterodimers with Nurr1. Although our data demonstrated that bexarotene was ineffective in neuroprotection in rats in vivo, the results revealed that bexarotene has the capacity to coregulate subsets of Nurr1 target genes including the receptor tyrosine kinase subunit Ret. Moreover, bexarotene was able to restore dysfunctional Ret-dependent neurotrophic signaling in α-synuclein-overexpressing mouse DA neurons. These data highlight the role of the Nurr1-Ret signaling pathway as a target of α-synuclein toxicity and suggest that retinoid X receptor ligands with appropriate pharmacological properties could have therapeutic potential in Parkinson's disease. SIGNIFICANCE STATEMENT: How α-synuclein, a protein enriched in Lewy bodies in Parkinson's disease, is causing neuropathology in dopamine neurons remains unclear. This study elucidated how α-synuclein is influencing gene expression and how Nurr1, a transcription factor known to protect dopamine neurons against α-synuclein toxicity, can counteract these effects. Moreover, given the protective role of Nurr1, this study also investigated how Nurr1 could be pharmacologically targeted via bexarotene, a ligand of Nurr1's heterodimerization partner retinoid X receptor (RXR). The results showed that RXR ligands could increase neurotrophic signaling, but provided a mixed picture of its potential in a Parkinson's disease rat model in vivo. However, this study clearly emphasized Nurr1's neuroprotective role and indicated that other RXR ligands could have therapeutic potential in Parkinson's disease.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Regulación de la Expresión Génica/genética , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Receptores X Retinoide/metabolismo , Transducción de Señal/genética , alfa-Sinucleína/metabolismo , Animales , Bexaroteno , Células Cultivadas , Neuronas Dopaminérgicas/efectos de los fármacos , Embrión de Mamíferos , Femenino , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Mesencéfalo/citología , Ratones , Ratones Transgénicos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Oxidopamina/toxicidad , Ratas , Ratas Sprague-Dawley , Receptores X Retinoide/agonistas , Receptores X Retinoide/genética , Conducta Estereotipada/fisiología , Sinapsinas/genética , Sinapsinas/metabolismo , Tetrahidronaftalenos/farmacología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , alfa-Sinucleína/genética
12.
J Neurosci ; 35(41): 14057-69, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26468205

RESUMEN

Parkinson's disease (PD) is a movement disorder characterized by a progressive loss of nigrostriatal dopaminergic neurons. Restoration of dopamine transmission by l-DOPA relieves symptoms of PD but causes dyskinesia. Trace Amine-Associated Receptor 1 (TAAR1) modulates dopaminergic transmission, but its role in experimental Parkinsonism and l-DOPA responses has been neglected. Here, we report that TAAR1 knock-out (KO) mice show a reduced loss of dopaminergic markers in response to intrastriatal 6-OHDA administration compared with wild-type (WT) littermates. In contrast, the TAAR1 agonist RO5166017 aggravated degeneration induced by intrastriatal 6-OHDA in WT mice. Subchronic l-DOPA treatment of TAAR1 KO mice unilaterally lesioned with 6-OHDA in the medial forebrain bundle resulted in more pronounced rotational behavior and dyskinesia than in their WT counterparts. The enhanced behavioral sensitization to l-DOPA in TAAR1 KO mice was paralleled by increased phosphorylation of striatal GluA1 subunits of AMPA receptors. Conversely, RO5166017 counteracted both l-DOPA-induced rotation and dyskinesia as well as AMPA receptor phosphorylation. Underpinning a role for TAAR1 receptors in modulating glutamate neurotransmission, intrastriatal application of RO5166017 prevented the increase of evoked corticostriatal glutamate release provoked by dopamine deficiency after 6-OHDA-lesions or conditional KO of Nurr1. Finally, inhibition of corticostriatal glutamate release by TAAR1 showed mechanistic similarities to that effected by activation of dopamine D2 receptors. These data unveil a role for TAAR1 in modulating the degeneration of dopaminergic neurons, the behavioral response to l-DOPA, and presynaptic and postsynaptic glutamate neurotransmission in the striatum, supporting their relevance to the pathophysiology and, potentially, management of PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is characterized by a progressive loss of nigrostriatal dopaminergic neurons. Restoration of dopamine transmission by l-DOPA relieves symptoms of PD but causes severe side effects. Trace Amine-Associated Receptor 1 (TAAR1) modulates dopaminergic transmission, but its role in PD and l-DOPA responses has been neglected. Here, we report that TAAR1 potentiates the degeneration of dopaminergic neurons and attenuates the behavioral response to l-DOPA and presynaptic and postsynaptic glutamate neurotransmission in the striatum, supporting the relevance of TAAR1 to the pathophysiology and, potentially, management of PD.


Asunto(s)
Antiparkinsonianos/uso terapéutico , Ácido Glutámico/metabolismo , Levodopa/uso terapéutico , Trastornos Parkinsonianos/tratamiento farmacológico , Receptores Acoplados a Proteínas G/deficiencia , Transmisión Sináptica/genética , Adrenérgicos/toxicidad , Acatisia Inducida por Medicamentos/etiología , Animales , Cocaína/análogos & derivados , Cocaína/farmacocinética , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/patología , Modelos Animales de Enfermedad , Dopamina/metabolismo , Proteínas de Transporte de Dopamina a través de la Membrana Plasmática/metabolismo , Fosfoproteína 32 Regulada por Dopamina y AMPc/metabolismo , Levodopa/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodesoxirribonucleótidos Antisentido/farmacología , Oxidopamina/toxicidad , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/patología , Radiofármacos/farmacocinética , Receptores Acoplados a Proteínas G/genética , Conducta Estereotipada/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Factores de Tiempo , Tirosina 3-Monooxigenasa/metabolismo
13.
Proc Natl Acad Sci U S A ; 110(6): 2360-5, 2013 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-23341612

RESUMEN

Developmental transcription factors important in early neuron specification and differentiation often remain expressed in the adult brain. However, how these transcription factors function to mantain appropriate neuronal identities in adult neurons and how transcription factor dysregulation may contribute to disease remain largely unknown. The transcription factor Nurr1 has been associated with Parkinson's disease and is essential for the development of ventral midbrain dopamine (DA) neurons. We used conditional Nurr1 gene-targeted mice in which Nurr1 is ablated selectively in mature DA neurons by treatment with tamoxifen. We show that Nurr1 ablation results in a progressive pathology associated with reduced striatal DA, impaired motor behaviors, and dystrophic axons and dendrites. We used laser-microdissected DA neurons for RNA extraction and next-generation mRNA sequencing to identify Nurr1-regulated genes. This analysis revealed that Nurr1 functions mainly in transcriptional activation to regulate a battery of genes expressed in DA neurons. Importantly, nuclear-encoded mitochondrial genes were identified as the major functional category of Nurr1-regulated target genes. These studies indicate that Nurr1 has a key function in sustaining high respiratory function in these cells, and that Nurr1 ablation in mice recapitulates early features of Parkinson's disease.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/metabolismo , Animales , Conducta Animal , Núcleo Celular/genética , Dopamina/metabolismo , Neuronas Dopaminérgicas/ultraestructura , Expresión Génica , Genes Mitocondriales , Ratones , Ratones Noqueados , Ratones Transgénicos , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Corteza Visual/metabolismo
14.
Development ; 139(14): 2625-34, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22696295

RESUMEN

The transcription factors Foxa1 and Foxa2 promote the specification of midbrain dopaminergic (mDA) neurons and the floor plate. Whether their role is direct has remained unclear as they also regulate the expression of Shh, which has similar roles. We characterized the Foxa2 cis-regulatory network by chromatin immunoprecipitation followed by high-throughput sequencing of mDA progenitors. This identified 9160 high-quality Foxa2 binding sites associated with 5409 genes, providing mechanistic insights into Foxa2-mediated positive and negative regulatory events. Foxa2 regulates directly and positively key determinants of mDA neurons, including Lmx1a, Lmx1b, Msx1 and Ferd3l, while negatively inhibiting transcription factors expressed in ventrolateral midbrain such as Helt, Tle4, Otx1, Sox1 and Tal2. Furthermore, Foxa2 negatively regulates extrinsic and intrinsic components of the Shh signaling pathway, possibly by binding to the same enhancer regions of co-regulated genes as Gli1. Foxa2 also regulates the expression of floor plate factors that control axon trajectories around the midline of the embryo, thereby contributing to the axon guidance function of the floor plate. Finally, this study identified multiple Foxa2-regulated enhancers that are active in the floor plate of the midbrain or along the length of the embryo in mouse and chick. This work represents the first comprehensive characterization of Foxa2 targets in mDA progenitors and provides a framework for elaborating gene regulatory networks in a functionally important progenitor population.


Asunto(s)
Neuronas Dopaminérgicas/metabolismo , Factor Nuclear 3-beta del Hepatocito/metabolismo , Mesencéfalo/citología , Células Madre/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Inmunoprecipitación de Cromatina , Electroporación , Genotipo , Factor Nuclear 3-beta del Hepatocito/genética , Inmunohistoquímica , Hibridación in Situ , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Factor de Transcripción MSX1/genética , Factor de Transcripción MSX1/metabolismo , Ratones , Ratones Mutantes , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Proteínas Represoras , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
15.
Stem Cells ; 32(3): 609-22, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24549637

RESUMEN

An important goal in stem cell biology is to develop methods for efficient generation of clinically interesting cell types from relevant stem cell populations. This is particularly challenging for different types of neurons of the central nervous system where hundreds of distinct neuronal cell types are generated during embryonic development. We previously used a strategy based on forced transcription factor expression in embryonic stem cell-derived neural progenitors to generate specific types of neurons, including dopamine and serotonin neurons. Here, we extend these studies and show that noradrenergic neurons can also be generated from pluripotent embryonic stem cells by forced expression of the homeobox transcription factor Phox2b under the signaling influence of fibroblast growth factor 8 (FGF8) and bone morphogenetic proteins. In neural progenitors exposed to FGF8 and sonic hedgehog both Phox2b and the related Phox2a instead promoted the generation of neurons with the characteristics of mid- and hindbrain motor neurons. The efficient generation of these neuron types enabled a comprehensive genome-wide gene expression analysis that provided further validation of the identity of generated cells. Moreover, we also demonstrate that the generated cell types are amenable to drug testing in vitro and we show that variants of the differentiation protocols can be applied to cultures of human pluripotent stem cells for the generation of human noradrenergic and visceral motor neurons. Thus, these studies provide a basis for characterization of yet an additional highly clinically relevant neuronal cell type.


Asunto(s)
Neuronas Adrenérgicas/citología , Linaje de la Célula , Células Madre Embrionarias/citología , Neuronas Motoras/citología , Factores de Transcripción/metabolismo , Neuronas Adrenérgicas/metabolismo , Animales , Línea Celular , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Ingeniería Genética , Genoma/genética , Proteínas de Homeodominio/metabolismo , Humanos , Ratones , Neuronas Motoras/metabolismo , Transducción de Señal
16.
Exp Cell Res ; 329(1): 94-100, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25173987

RESUMEN

Cellular systems for DNA repair ensure prompt removal of DNA lesions that threaten the genomic stability of the cell. Transcription factors (TFs) have long been known to facilitate DNA repair via transcriptional regulation of specific target genes encoding key DNA repair proteins. However, recent findings identified TFs as DNA repair components acting directly at the DNA lesions in a transcription-independent fashion. Together this recent progress is consistent with the hypothesis that TFs have acquired the ability to localize DNA lesions and function by facilitating chromatin remodeling at sites of damaged DNA. Here we review these recent findings and discuss how TFs may function in DNA repair.


Asunto(s)
Ensamble y Desensamble de Cromatina , Daño del ADN/genética , Reparación del ADN/genética , Factores de Transcripción/metabolismo , Animales , Humanos
17.
Development ; 138(16): 3399-408, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21752929

RESUMEN

The severe disorders associated with a loss or dysfunction of midbrain dopamine neurons (DNs) have intensified research aimed at deciphering developmental programs controlling midbrain development. The homeodomain proteins Lmx1a and Lmx1b are important for the specification of DNs during embryogenesis, but it is unclear to what degree they may mediate redundant or specific functions. Here, we provide evidence showing that DN progenitors in the ventral midbrain can be subdivided into molecularly distinct medial and lateral domains, and these subgroups show different sensitivity to the loss of Lmx1a and Lmx1b. Lmx1a is specifically required for converting non-neuronal floor-plate cells into neuronal DN progenitors, a process that involves the establishment of Notch signaling in ventral midline cells. On the other hand, lateral DN progenitors that do not appear to originate from the floor plate are selectively ablated in Lmx1b mutants. In addition, we also reveal an unanticipated role for Lmx1b in regulating Phox2a expression and the sequential specification of ocular motor neurons (OMNs) and red nucleus neurons (RNNs) from progenitors located lateral to DNs in the midbrain. Our data therefore establish that Lmx1b influences the differentiation of multiple neuronal subtypes in the ventral midbrain, whereas Lmx1a appears to be exclusively devoted to the differentiation of the DN lineage.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Mesencéfalo/embriología , Mesencéfalo/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis , Linaje de la Célula , Dopamina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas con Homeodominio LIM , Mesencéfalo/citología , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Factores de Transcripción/genética
18.
Elife ; 122024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38587883

RESUMEN

Midbrain dopamine (mDA) neurons comprise diverse cells with unique innervation targets and functions. This is illustrated by the selective sensitivity of mDA neurons of the substantia nigra compacta (SNc) in patients with Parkinson's disease, while those in the ventral tegmental area (VTA) are relatively spared. Here, we used single nuclei RNA sequencing (snRNA-seq) of approximately 70,000 mouse midbrain cells to build a high-resolution atlas of mouse mDA neuron diversity at the molecular level. The results showed that differences between mDA neuron groups could best be understood as a continuum without sharp differences between subtypes. Thus, we assigned mDA neurons to several 'territories' and 'neighborhoods' within a shifting gene expression landscape where boundaries are gradual rather than discrete. Based on the enriched gene expression patterns of these territories and neighborhoods, we were able to localize them in the adult mouse midbrain. Moreover, because the underlying mechanisms for the variable sensitivities of diverse mDA neurons to pathological insults are not well understood, we analyzed surviving neurons after partial 6-hydroxydopamine (6-OHDA) lesions to unravel gene expression patterns that correlate with mDA neuron vulnerability and resilience. Together, this atlas provides a basis for further studies on the neurophysiological role of mDA neurons in health and disease.


Asunto(s)
Ascomicetos , Trastornos Parkinsonianos , Adulto , Humanos , Animales , Ratones , Neuronas Dopaminérgicas , Perfilación de la Expresión Génica , Trastornos Parkinsonianos/genética , Mesencéfalo , Oxidopamina
19.
NPJ Parkinsons Dis ; 10(1): 93, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684669

RESUMEN

Loss-of-function variants in the PRKN gene encoding the ubiquitin E3 ligase PARKIN cause autosomal recessive early-onset Parkinson's disease (PD). Extensive in vitro and in vivo studies have reported that PARKIN is involved in multiple pathways of mitochondrial quality control, including mitochondrial degradation and biogenesis. However, these findings are surrounded by substantial controversy due to conflicting experimental data. In addition, the existing PARKIN-deficient mouse models have failed to faithfully recapitulate PD phenotypes. Therefore, we have investigated the mitochondrial role of PARKIN during ageing and in response to stress by employing a series of conditional Parkin knockout mice. We report that PARKIN loss does not affect oxidative phosphorylation (OXPHOS) capacity and mitochondrial DNA (mtDNA) levels in the brain, heart, and skeletal muscle of aged mice. We also demonstrate that PARKIN deficiency does not exacerbate the brain defects and the pro-inflammatory phenotype observed in mice carrying high levels of mtDNA mutations. To rule out compensatory mechanisms activated during embryonic development of Parkin-deficient mice, we generated a mouse model where loss of PARKIN was induced in adult dopaminergic (DA) neurons. Surprisingly, also these mice did not show motor impairment or neurodegeneration, and no major transcriptional changes were found in isolated midbrain DA neurons. Finally, we report a patient with compound heterozygous PRKN pathogenic variants that lacks PARKIN and has developed PD. The PARKIN deficiency did not impair OXPHOS activities or induce mitochondrial pathology in skeletal muscle from the patient. Altogether, our results argue that PARKIN is dispensable for OXPHOS function in adult mammalian tissues.

20.
J Neurosci ; 32(2): 436-51, 2012 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-22238080

RESUMEN

Prenatal exposure to infection has been linked to increased risk of neurodevelopmental brain disorders, and recent evidence implicates altered dopaminergic development in this association. However, since the relative risk size of prenatal infection appears relatively small with respect to long-term neuropsychiatric outcomes, it is likely that this prenatal insult interacts with other factors in shaping the risk of postnatal brain dysfunctions. In the present study, we show that the neuropathological consequences of prenatal viral-like immune activation are exacerbated in offspring with genetic predisposition to dopaminergic abnormalities induced by mutations in Nurr1, a transcription factor highly essential for normal dopaminergic development. We combined a mouse model of heterozygous genetic deletion of Nurr1 with a model of prenatal immune challenge by the viral mimetic poly(I:C) (polyriboinosinic polyribocytidilic acid). In our gene-environment interaction model, we demonstrate that the combination of the genetic and environmental factors not only exerts additive effects on locomotor hyperactivity and sensorimotor gating deficits, but further produces synergistic effects in the development of impaired attentional shifting and sustained attention. We further demonstrate that the combination of the two factors is necessary to trigger maldevelopment of prefrontal cortical and ventral striatal dopamine systems. Our findings provide evidence for specific gene-environment interactions in the emergence of enduring attentional impairments and neuronal abnormalities pertinent to dopamine-associated brain disorders such as schizophrenia and attention deficit/hyperactivity disorder, and further emphasize a critical role of abnormal dopaminergic development in these etiopathological processes.


Asunto(s)
Trastorno por Déficit de Atención con Hiperactividad/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Trastornos Neurocognitivos/genética , Trastornos Neurocognitivos/inmunología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/genética , Animales , Trastorno por Déficit de Atención con Hiperactividad/genética , Trastorno por Déficit de Atención con Hiperactividad/virología , Enfermedades Autoinmunes del Sistema Nervioso/genética , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Trastornos Neurocognitivos/virología , Miembro 2 del Grupo A de la Subfamilia 4 de Receptores Nucleares/deficiencia , Embarazo , Efectos Tardíos de la Exposición Prenatal , Esquizofrenia/genética , Esquizofrenia/inmunología , Esquizofrenia/virología , Virosis/complicaciones , Virosis/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA