Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Development ; 151(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38655654

RESUMEN

Foxg1 masters telencephalic development via a pleiotropic control over its progression. Expressed within the central nervous system (CNS), L1 retrotransposons are implicated in progression of its histogenesis and tuning of its genomic plasticity. Foxg1 represses gene transcription, and L1 elements share putative Foxg1-binding motifs, suggesting the former might limit telencephalic expression (and activity) of the latter. We tested such a prediction, in vivo as well as in engineered primary neural cultures, using loss- and gain-of-function approaches. We found that Foxg1-dependent, transcriptional L1 repression specifically occurs in neopallial neuronogenic progenitors and post-mitotic neurons, where it is supported by specific changes in the L1 epigenetic landscape. Unexpectedly, we discovered that Foxg1 physically interacts with L1-mRNA and positively regulates neonatal neopallium L1-DNA content, antagonizing the retrotranscription-suppressing activity exerted by Mov10 and Ddx39a helicases. To the best of our knowledge, Foxg1 represents the first CNS patterning gene acting as a bimodal retrotransposon modulator, limiting transcription of L1 elements and promoting their amplification, within a specific domain of the developing mouse brain.


Asunto(s)
Factores de Transcripción Forkhead , Regulación del Desarrollo de la Expresión Génica , Neocórtex , Proteínas del Tejido Nervioso , ARN Mensajero , Animales , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Ratones , Neocórtex/metabolismo , Neocórtex/embriología , Neocórtex/crecimiento & desarrollo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , ARN Mensajero/metabolismo , ARN Mensajero/genética , Retroelementos/genética , ADN/metabolismo , ADN/genética , Neuronas/metabolismo
2.
BMC Biol ; 22(1): 180, 2024 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-39183266

RESUMEN

BACKGROUND: Mainly known as a transcription factor patterning the rostral brain and governing its histogenesis, FOXG1 has been also detected outside the nucleus; however, biological meaning of that has been only partially clarified. RESULTS: Prompted by FOXG1 expression in cytoplasm of pallial neurons, we investigated its implication in translational control. We documented the impact of FOXG1 on ribosomal recruitment of Grin1-mRNA, encoding for the main subunit of NMDA receptor. Next, we showed that FOXG1 increases GRIN1 protein level by enhancing the translation of its mRNA, while not increasing its stability. Molecular mechanisms underlying this activity included FOXG1 interaction with EIF4E and, possibly, Grin1-mRNA. Besides, we found that, within murine neocortical cultures, de novo synthesis of GRIN1 undergoes a prominent and reversible, homeostatic regulation and FOXG1 is instrumental to that. Finally, by integrated analysis of multiple omic data, we inferred that FOXG1 is implicated in translational control of hundreds of neuronal genes, modulating ribosome engagement and progression. In a few selected cases, we experimentally verified such inference. CONCLUSIONS: These findings point to FOXG1 as a key effector, potentially crucial to multi-scale temporal tuning of neocortical pyramid activity, an issue with profound physiological and neuropathological implications.


Asunto(s)
Factores de Transcripción Forkhead , Neocórtex , Proteínas del Tejido Nervioso , Neuronas , Receptores de N-Metil-D-Aspartato , Animales , Femenino , Masculino , Ratones , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Neocórtex/metabolismo , Neocórtex/embriología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Biosíntesis de Proteínas/genética , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo
3.
Cancer Sci ; 115(8): 2565-2577, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38932521

RESUMEN

Cisplatin (CDDP) is a commonly used chemotherapeutic for osteosarcoma (OS) patients, and drug resistance remains as a major hurdle to undermine the treatment outcome. Here, we investigated the potential involvement of FoxG1 and BNIP3 in CDDP resistance of OS cells. FoxG1 and BNIP3 expression levels were detected in the CDDP-sensitive and CDDP-resistant OS tumors and cell lines. Mitophagy was observed through transmission electron microscope analysis. The sensitivity to CDDP in OS cells upon FoxG1 overexpression was examined in cell and animal models. We found that FoxG1 and BNIP3 showed significant downregulation in the CDDP-resistant OS tumor samples and cell lines. CDDP-resistant OS tumor specimens and cells displayed impaired mitophagy. FoxG1 overexpression promoted BNIP3 expression, enhanced mitophagy in CDDP-resistant OS cells, and resensitized the resistant cells to CDDP treatment in vitro and in vivo. Our data highlighted the role of the FoxG1/BNIP3 axis in regulating mitophagy and dictating CDDP resistance in OS cells, suggesting targeting FoxG1/BNIP3-dependent mitophagy as a potential strategy to overcome CDDP resistance in OS.


Asunto(s)
Neoplasias Óseas , Cisplatino , Resistencia a Antineoplásicos , Factores de Transcripción Forkhead , Proteínas de la Membrana , Mitofagia , Proteínas del Tejido Nervioso , Osteosarcoma , Proteínas Proto-Oncogénicas , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/metabolismo , Osteosarcoma/patología , Osteosarcoma/genética , Mitofagia/efectos de los fármacos , Cisplatino/farmacología , Humanos , Resistencia a Antineoplásicos/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Animales , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Línea Celular Tumoral , Ratones , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias Óseas/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Femenino , Masculino , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos
4.
Dev Growth Differ ; 66(3): 219-234, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38378191

RESUMEN

The vertebrate telencephalic lobes consist of the pallium (dorsal) and subpallium (ventral). The subpallium gives rise to the basal ganglia, encompassing the pallidum and striatum. The development of this region is believed to depend on Foxg1/Foxg1a functions in both mice and zebrafish. This study aims to elucidate the genetic regulatory network controlled by foxg1a in subpallium development using zebrafish as a model. The expression gradient of foxg1a within the developing telencephalon was examined semi-quantitatively in initial investigations. Utilizing the CRISPR/Cas9 technique, we subsequently established a foxg1a mutant line and observed the resultant phenotypes. Morphological assessment revealed that foxg1a mutants exhibit a thin telencephalon together with a misshapen preoptic area (POA). Notably, accumulation of apoptotic cells was identified in this region. In mutants at 24 h postfertilization, the expression of pallium markers expanded ventrally, while that of subpallium markers was markedly suppressed. Concurrently, the expression of fgf8a, vax2, and six3b was shifted ventrally, causing anomalous expression in regions typical of POA formation in wild-type embryos. Consequently, the foxg1a mutation led to expansion of the pallium and disrupted the subpallium and POA. This highlights a pivotal role of foxg1a in directing the dorsoventral patterning of the telencephalon, particularly in subpallium differentiation, mirroring observations in mice. Additionally, reduced expression of neural progenitor maintenance genes was detected in mutants, suggesting the necessity of foxg1a in preserving neural progenitors. Collectively, these findings underscore evolutionarily conserved functions of foxg1 in the development of the subpallium in vertebrate embryos.


Asunto(s)
Factores de Transcripción Forkhead , Redes Reguladoras de Genes , Pez Cebra , Animales , Corteza Cerebral/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Telencéfalo/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
5.
Cell Mol Neurobiol ; 44(1): 35, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38630150

RESUMEN

An increasing body of research suggests that promoting microglial autophagy hinders the neuroinflammation initiated though the NLRP3 inflammasome activation in Alzheimer's disease (AD). The function of FoxG1, a crucial transcription factor involved in cell survival by regulating mitochondrial function, remains unknown during the AD process and neuroinflammation occurs. In the present study, we firstly found that Aß peptides induced AD-like neuroinflammation upregulation and downregulated the level of autophagy. Following low-dose Aß25-35 stimulation, FoxG1 expression and autophagy exhibited a gradual increase. Nevertheless, with high-concentration Aß25-35 treatment, progressive decrease in FoxG1 expression and autophagy levels as the concentration of Aß25-35 escalated. In addition, FoxG1 has a positive effect on cell viability and autophagy in the nervous system. In parallel with the Aß25-35 stimulation, we employed siRNA to decrease the expression of FoxG1 in N2A cells. A substantial reduction in autophagy level (Beclin1, LC3II, SQSTM1/P62) and a notable growth in inflammatory response (NLRP3, TNF-α, and IL-6) were observed. In addition, we found FoxG1 overexpression owned the effect on the activation of AMPK/mTOR autophagy pathway and siRNA-FoxG1 successfully abolished this effect. Lastly, FoxG1 suppressed the NLRP3 inflammasome and enhanced the cognitive function in AD-like mouse model induced by Aß25-35. Confirmed by cellular and animal experiments, FoxG1 suppressed NLRP3-mediated neuroinflammation, which was strongly linked to autophagy regulated by AMPK/mTOR. Taken together, FoxG1 may be a critical node in the pathologic progression of AD and has the potential to serve as therapeutic target.


Asunto(s)
Enfermedad de Alzheimer , Factores de Transcripción Forkhead , Inflamasomas , Animales , Ratones , Enfermedad de Alzheimer/tratamiento farmacológico , Proteínas Quinasas Activadas por AMP , Autofagia , Enfermedades Neuroinflamatorias , Proteína con Dominio Pirina 3 de la Familia NLR , ARN Interferente Pequeño , Factores de Transcripción Forkhead/antagonistas & inhibidores
6.
Cereb Cortex ; 33(6): 3107-3123, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35818636

RESUMEN

Sizes of neuronal, astroglial and oligodendroglial complements forming the neonatal cerebral cortex largely depend on rates at which pallial stem cells give rise to lineage-committed progenitors and the latter ones progress to mature cell types. Here, we investigated the spatial articulation of pallial stem cells' (SCs) commitment to astrogenesis as well as the progression of committed astroglial progenitors (APs) to differentiated astrocytes, by clonal and kinetic profiling of pallial precursors. We found that caudal-medial (CM) SCs are more prone to astrogenesis than rostro-lateral (RL) ones, while RL-committed APs are more keen to proliferate than CM ones. Next, we assessed the control of these phenomena by 2 key transcription factor genes mastering regionalization of the early cortical primordium, Emx2 and Foxg1, via lentiviral somatic transgenesis, epistasis assays, and ad hoc rescue assays. We demonstrated that preferential CM SCs progression to astrogenesis is promoted by Emx2, mainly via Couptf1, Nfia, and Sox9 upregulation, while Foxg1 antagonizes such progression to some extent, likely via repression of Zbtb20. Finally, we showed that Foxg1 and Emx2 may be implicated-asymmetrically and antithetically-in shaping distinctive proliferative/differentiative behaviors displayed by APs in hippocampus and neocortex.


Asunto(s)
Neocórtex , Neurogénesis , Humanos , Recién Nacido , Astrocitos/metabolismo , Astrocitos/fisiología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Factor de Transcripción COUP I/genética , Regulación del Desarrollo de la Expresión Génica/genética , Hipocampo/metabolismo , Hipocampo/fisiología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Neocórtex/metabolismo , Neurogénesis/genética , Neurogénesis/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Oligodendroglía/metabolismo , Oligodendroglía/fisiología
7.
Cereb Cortex ; 33(19): 10272-10285, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37566909

RESUMEN

The cortical plate (CP) is composed of excitatory and inhibitory neurons, the latter of which originate in the ganglionic eminences. From their origin in the ventral telencephalon, maturing postmitotic interneurons migrate during embryonic development over some distance to reach their final destination in the CP. The histone methyltransferase Disruptor of Telomeric Silencing 1-like (DOT1L) is necessary for proper CP development and layer distribution of glutamatergic neurons. However, its specific role on cortical interneuron development has not yet been explored. Here, we demonstrate that DOT1L affects interneuron development in a cell autonomous manner. Deletion of Dot1l in Nkx2.1-expressing interneuron precursor cells results in an overall reduction and altered distribution of GABAergic interneurons in the CP from postnatal day 0 onwards. We observed an altered proportion of GABAergic interneurons in the cortex, with a significant decrease in parvalbumin-expressing interneurons. Moreover, a decreased number of mitotic cells at the embryonic day E14.5 was observed upon Dot1l deletion. Altogether, our results indicate that reduced numbers of cortical interneurons upon DOT1L deletion result from premature cell cycle exit, but effects on postmitotic differentiation, maturation, and migration are likely at play as well.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Interneuronas , Parvalbúminas , Telencéfalo , Diferenciación Celular/fisiología , Interneuronas/citología , Interneuronas/metabolismo , Parvalbúminas/genética , Parvalbúminas/metabolismo , Telencéfalo/citología , Animales , Ratones , N-Metiltransferasa de Histona-Lisina/genética
8.
Int J Mol Sci ; 25(19)2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39409184

RESUMEN

FOXG1 syndrome is a rare neurodevelopmental disorder of the telencephalon, for which there is no cure. Underlying heterozygous pathogenic variants in the Forkhead Box G1 (FOXG1) gene with resulting impaired or loss of FOXG1 function lead to severe neurological impairments. Here, we report a patient with a de novo pathogenic single nucleotide deletion c.946del (p.Leu316Cysfs*10) of the FOXG1 gene that causes a premature protein truncation. To study this variant in vivo, we generated and characterized Foxg1 c946del mice that recapitulate hallmarks of the human disorder. Accordingly, heterozygous Foxg1 c946del mice display neurological symptoms with aberrant neuronal networks and increased seizure susceptibility. Gene expression profiling identified increased oligodendrocyte- and myelination-related gene clusters. Specifically, we showed that expression of the c946del mutant and of other pathogenic FOXG1 variants correlated with overexpression of proteolipid protein 1 (Plp1), a gene linked to white matter disorders. Postnatal administration of Plp1-targeting antisense oligonucleotides (ASOs) in Foxg1 c946del mice improved neurological deficits. Our data suggest Plp1 as a new target for therapeutic strategies mitigating disease phenotypes in FOXG1 syndrome patients.


Asunto(s)
Factores de Transcripción Forkhead , Proteínas del Tejido Nervioso , Oligonucleótidos Antisentido , Animales , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Ratones , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/genética , Proteína Proteolipídica de la Mielina/genética , Masculino , Modelos Animales de Enfermedad , Femenino , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/tratamiento farmacológico
9.
Am J Med Genet B Neuropsychiatr Genet ; 195(6): e32970, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38459409

RESUMEN

Since 2008, FOXG1 haploinsufficiency has been linked to a severe neurodevelopmental phenotype resembling Rett syndrome but with earlier onset. Most patients are unable to sit, walk, or speak. For years, FOXG1 sequencing was only prescribed in such severe cases, limiting insight into the full clinical spectrum associated with this gene. Next-generation sequencing (NGS) now enables unbiased diagnostics. Through the European Reference Network for Rare Malformation Syndromes, Intellectual and Other Neurodevelopmental Disorders, we gathered data from patients with heterozygous FOXG1 variants presenting a mild phenotype, defined as able to speak and walk independently. We also reviewed data from three previously reported patients meeting our criteria. We identified five new patients with pathogenic FOXG1 missense variants, primarily in the forkhead domain, showing varying nonspecific intellectual disability and developmental delay. These features are not typical of congenital Rett syndrome and were rarely associated with microcephaly and epilepsy. Our findings are consistent with a previous genotype-phenotype analysis by Mitter et al. suggesting the delineation of five different FOXG1 genotype groups. Milder phenotypes were associated with missense variants in the forkhead domain. This information may facilitate prognostic assessments in children carrying a FOXG1 variant and improve the interpretation of new variants identified with genomic sequencing.


Asunto(s)
Factores de Transcripción Forkhead , Discapacidad Intelectual , Proteínas del Tejido Nervioso , Fenotipo , Síndrome de Rett , Humanos , Factores de Transcripción Forkhead/genética , Síndrome de Rett/genética , Proteínas del Tejido Nervioso/genética , Femenino , Masculino , Niño , Preescolar , Discapacidad Intelectual/genética , Desarrollo del Lenguaje , Estudios de Asociación Genética/métodos , Mutación Missense/genética , Discapacidades del Desarrollo/genética , Lactante , Adolescente , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Haploinsuficiencia/genética
10.
Int J Mol Sci ; 24(18)2023 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-37762220

RESUMEN

FOXG1 (forkhead box G1) syndrome is a neurodevelopmental disorder caused by variants in the Foxg1 gene that affect brain structure and function. Individuals affected by FOXG1 syndrome frequently exhibit delayed myelination in neuroimaging studies, which may impair the rapid conduction of nerve impulses. To date, the specific effects of FOXG1 on oligodendrocyte lineage progression and myelination during early postnatal development remain unclear. Here, we investigated the effects of Foxg1 deficiency on myelin development in the mouse brain by conditional deletion of Foxg1 in neural progenitors using NestinCreER;Foxg1fl/fl mice and tamoxifen induction at postnatal day 0 (P0). We found that Foxg1 deficiency resulted in a transient delay in myelination, evidenced by decreased myelin formation within the first two weeks after birth, but ultimately recovered to the control levels by P30. We also found that Foxg1 deletion prevented the timely attenuation of platelet-derived growth factor receptor alpha (PDGFRα) signaling and reduced the cell cycle exit of oligodendrocyte precursor cells (OPCs), leading to their excessive proliferation and delayed maturation. Additionally, Foxg1 deletion increased the expression of Hes5, a myelin formation inhibitor, as well as Olig2 and Sox10, two promoters of OPC differentiation. Our results reveal the important role of Foxg1 in myelin development and provide new clues for further exploring the pathological mechanisms of FOXG1 syndrome.


Asunto(s)
Encéfalo , Síndrome de Rett , Animales , Ratones , Potenciales de Acción , Ciclo Celular , Diferenciación Celular/genética , Proteínas del Tejido Nervioso/genética , Factores de Transcripción Forkhead
11.
Dev Biol ; 472: 18-29, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33428890

RESUMEN

The vertebrate eye anlage grows out of the brain and folds into bilayered optic cups. The eye is patterned along multiple axes, precisely controlled by genetic programs, to delineate neural retina, pigment epithelium, and optic stalk tissues. Pax genes encode developmental regulators of key morphogenetic events, with Pax2 being essential for interpreting inductive signals, including in the eye. PAX2 mutations cause ocular coloboma, when the ventral optic fissure fails to close. Previous studies established that Pax2 is necessary for fissure closure and to maintain the neural retina -- glial optic stalk boundary. Using a Pax2GFP/+ knock-in allele we discovered that the mutant optic nerve head (ONH) lacks molecular boundaries with the retina and RPE, rendering the ONH larger than normal. This was preceded by ventronasal cup mispatterning, a burst of overproliferation and followed by optic cup apoptosis. Our findings support the hypothesis that ONH cells are tripotential, requiring Pax2 to remain committed to glial fates. This work extends current models of ocular development, contributes to broader understanding of tissue boundary formation and informs the underlying mechanisms of human coloboma.


Asunto(s)
Ojo/embriología , Ojo/metabolismo , Disco Óptico/embriología , Factor de Transcripción PAX2/genética , Factor de Transcripción PAX2/metabolismo , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Proliferación Celular/genética , Coloboma/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Masculino , Ratones , Ratones Endogámicos C57BL , Disco Óptico/anomalías , Disco Óptico/citología , Retina/embriología , Células Madre/metabolismo
12.
Biochem Biophys Res Commun ; 623: 96-103, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35878429

RESUMEN

The transcription factor FOXG1 plays an important role in inner ear development; however, the cis-regulatory mechanisms controlling the inner-ear-specific expression of FOXG1 are poorly understood. In this study, we aimed to identify the element that specifically regulates FoxG1 expression in the otic vesicle, which develops into the inner ear, through comparative genome analysis between vertebrate species and chromatin immunoprecipitation. The cis-regulatory element (E2) identified showed high evolutionary conservation among vertebrates in the genomic DNA of FoxG1 spanning approximately 3 Mbp. We identified core sequences important for the activity of the otic-vesicle-specific enhancer through in vitro and in vivo reporter assays for various E2 enhancer mutants and determined the consensus sequence for SOX DNA binding. In addition, SoxE, a subfamily of the Sox family, was simultaneously expressed in the otic vesicles of developing embryos and showed a similar protein expression pattern as that of FoxG1. Furthermore, SOXE transcription factors induced specific transcriptional activity through the FoxG1 Otic enhancer (E2b). These findings suggest that the interaction between the otic enhancer of FoxG1 and SOXE transcription factor, in which the otic expression of FoxG1 is evolutionarily well-conserved, is important during early development of the inner ear, a sensory organ important for survival in nature.


Asunto(s)
Oído Interno , Factores de Transcripción SOXE , Animales , ADN/metabolismo , Oído Interno/metabolismo , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción SOXE/genética , Factores de Transcripción/metabolismo
13.
Development ; 146(4)2019 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-30745428

RESUMEN

The self-renewal activity of neural stem cells (NSCs) has been suggested to decrease with aging, resulting in age-dependent declines in brain function, such as presbyopia and memory loss. The molecular mechanisms underlying decreases in NSC proliferation with age need to be elucidated in more detail to develop treatments that promote brain function. We have previously reported that the expression of esophageal cancer-related gene 4 (Ecrg4) was upregulated in aged NSCs, whereas its overexpression decreased NSC proliferation, suggesting a functional relationship between Ecrg4 and NSC aging. Using Ecrg4-deficient mice in which the Ecrg4 locus was replaced with the lacZ gene, we here show that Ecrg4 deficiency recovered the age-dependent decline in NSC proliferation and enhanced spatial learning and memory in the Morris water-maze paradigm. We demonstrate that the proliferation of Ecrg4-deficient NSCs was partly maintained by the increased expression of Foxg1. Collectively, these results determine Ecrg4 as a NSC aging factor.


Asunto(s)
Envejecimiento , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Animales , Proliferación Celular , Replicación del ADN , Femenino , Hipocampo/metabolismo , Masculino , Aprendizaje por Laberinto , Memoria , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regulación hacia Arriba
14.
Inflamm Res ; 71(3): 309-320, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35190853

RESUMEN

OBJECTIVE: Hair cells in the inner ear have been demonstrated to be sensitive to the ototoxicity from some beneficial pharmaceutical drugs. This study aimed to explore the role of protein arginine methyltransferase 6 (PRMT6) in the process of neomycin-induced hearing loss and the underlying mechanism. METHODS: The neomycin-induced hearing loss mouse model and hair cell injury in vitro model were established. We took advantage of the HEI-OC1 cell line to evaluate PRMT6 expression in neomycin-induced hair cells, and the effect of PRMT6 on mitochondrial function and FoxG1 arginine methylation. Apoptotic cells were assessed and apoptotic marker cleaved caspase-3 level was detected. Reactive oxygen species (ROS) level and mitochondrial membrane potential (MMP) were subsequently measured. RESULT: The result showed that PRMT6 was significantly upregulated in neomycin-induced HEI-OC-1 cells, and PRMT6 silencing prevented MMP loss, reduced ROS production, as well as decreased cell apoptosis under neomycin treatment. Further results showed that FoxG1 was downregulated in neomycin-induced HEI-OC-1 cells, and PRMT6 promoted the FoxG1-mediated luciferase activity, while PRMT6 silencing reversed this effect. Mechanistic experiments revealed that PRMT6 silencing reduced the arginine methylation level of FoxG1 protein. In vivo, neomycin-induced upregulation of hearing thresholds and increased cell apoptosis, whereas PRMT6 inhibitor partly reversed these effects. CONCLUSION: Our findings suggested that inhibition of PRMT6 reduced neomycin-induced inner ear hair cell injury through the restraint of FoxG1 arginine methylation.


Asunto(s)
Arginina , Neomicina , Animales , Apoptosis , Arginina/metabolismo , Arginina/farmacología , Factores de Transcripción Forkhead/genética , Cabello/metabolismo , Células Ciliadas Auditivas/metabolismo , Metilación , Ratones , Neomicina/metabolismo , Neomicina/toxicidad , Proteínas del Tejido Nervioso/metabolismo
15.
Int J Mol Sci ; 23(2)2022 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-35055139

RESUMEN

Development of the central nervous system (CNS) depends on accurate spatiotemporal control of signaling pathways and transcriptional programs. Forkhead Box G1 (FOXG1) is one of the master regulators that play fundamental roles in forebrain development; from the timing of neurogenesis, to the patterning of the cerebral cortex. Mutations in the FOXG1 gene cause a rare neurodevelopmental disorder called FOXG1 syndrome, also known as congenital form of Rett syndrome. Patients presenting with FOXG1 syndrome manifest a spectrum of phenotypes, ranging from severe cognitive dysfunction and microcephaly to social withdrawal and communication deficits, with varying severities. To develop and improve therapeutic interventions, there has been considerable progress towards unravelling the multi-faceted functions of FOXG1 in the neurodevelopment and pathogenesis of FOXG1 syndrome. Moreover, recent advances in genome editing and stem cell technologies, as well as the increased yield of information from high throughput omics, have opened promising and important new avenues in FOXG1 research. In this review, we provide a summary of the clinical features and emerging molecular mechanisms underlying FOXG1 syndrome, and explore disease-modelling approaches in animals and human-based systems, to highlight the prospects of research and possible clinical interventions.


Asunto(s)
Factores de Transcripción Forkhead/genética , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Síndrome de Rett/genética , Animales , Modelos Animales de Enfermedad , Humanos , Mutación , Fenotipo
16.
Int J Mol Sci ; 23(23)2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36499306

RESUMEN

Strategies to enhance hippocampal precursor cells efficiently differentiate into neurons could be crucial for structural repair after neurodegenerative damage. FOXG1 has been shown to play an important role in pattern formation, cell proliferation, and cell specification during embryonic and early postnatal neurogenesis. Thus far, the role of FOXG1 in adult hippocampal neurogenesis is largely unknown. Utilizing CAG-loxp-stop-loxp-Foxg1-IRES-EGFP (Foxg1fl/fl), a specific mouse line combined with CreAAV infusion, we successfully forced FOXG1 overexpressed in the hippocampal dentate gyrus (DG) of the genotype mice. Thereafter, we explored the function of FOXG1 on neuronal lineage progression and hippocampal neurogenesis in adult mice. By inhibiting p21cip1 expression, FOXG1-regulated activities enable the expansion of the precursor cell population. Besides, FOXG1 induced quiescent radial-glia like type I neural progenitor, giving rise to intermediate progenitor cells, neuroblasts in the hippocampal DG. Through increasing the length of G1 phase, FOXG1 promoted lineage-committed cells to exit the cell cycle and differentiate into mature neurons. The present results suggest that FOXG1 likely promotes neuronal lineage progression and thereby contributes to adult hippocampal neurogenesis. Elevating FOXG1 levels either pharmacologically or through other means could present a therapeutic strategy for disease related with neuronal loss.


Asunto(s)
Células-Madre Neurales , Neurogénesis , Ratones , Animales , Neurogénesis/genética , Hipocampo/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Proliferación Celular , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo
17.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163265

RESUMEN

FOXG1 is an ancient transcription factor gene mastering telencephalic development. A number of distinct structural FOXG1 mutations lead to the "FOXG1 syndrome", a complex and heterogeneous neuropathological entity, for which no cure is presently available. Reconstruction of primary neurodevelopmental/physiological anomalies evoked by these mutations is an obvious pre-requisite for future, precision therapy of such syndrome. Here, as a proof-of-principle, we functionally scored three FOXG1 neuropathogenic alleles, FOXG1G224S, FOXG1W308X, and FOXG1N232S, against their healthy counterpart. Specifically, we delivered transgenes encoding for them to dedicated preparations of murine pallial precursors and quantified their impact on selected neurodevelopmental and physiological processes mastered by Foxg1: pallial stem cell fate choice, proliferation of neural committed progenitors, neuronal architecture, neuronal activity, and their molecular correlates. Briefly, we found that FOXG1G224S and FOXG1W308X generally performed as a gain- and a loss-of-function-allele, respectively, while FOXG1N232S acted as a mild loss-of-function-allele or phenocopied FOXG1WT. These results provide valuable hints about processes misregulated in patients heterozygous for these mutations, to be re-addressed more stringently in patient iPSC-derivative neuro-organoids. Moreover, they suggest that murine pallial cultures may be employed for fast multidimensional profiling of novel, human neuropathogenic FOXG1 alleles, namely a step propedeutic to timely delivery of therapeutic precision treatments.


Asunto(s)
Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas del Tejido Nervioso/genética , Trastornos del Neurodesarrollo/genética , Alelos , Animales , Encéfalo/metabolismo , Corteza Cerebral/metabolismo , Factores de Transcripción Forkhead/metabolismo , Expresión Génica/genética , Frecuencia de los Genes/genética , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neurogénesis , Neuronas/metabolismo , Cultivo Primario de Células , Prueba de Estudio Conceptual
18.
Development ; 145(1)2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29229772

RESUMEN

During forebrain development, a telencephalic organizer called the cortical hem is crucial for inducing hippocampal fate in adjacent cortical neuroepithelium. How the hem is restricted to its medial position is therefore a fundamental patterning issue. Here, we demonstrate that Foxg1-Lhx2 interactions are crucial for the formation of the hem. Loss of either gene causes a region of the cortical neuroepithelium to transform into hem. We show that FOXG1 regulates Lhx2 expression in the cortical primordium. In the absence of Foxg1, the presence of Lhx2 is sufficient to suppress hem fate, and hippocampal markers appear selectively in Lhx2-expressing regions. FOXG1 also restricts the temporal window in which loss of Lhx2 results in a transformation of cortical primordium into hem. Therefore, Foxg1 and Lhx2 form a genetic hierarchy in the spatiotemporal regulation of cortical hem specification and positioning, and together ensure the normal development of this hippocampal organizer.


Asunto(s)
Factores de Transcripción Forkhead/biosíntesis , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/embriología , Proteínas con Homeodominio LIM/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Telencéfalo/embriología , Factores de Transcripción/biosíntesis , Animales , Factores de Transcripción Forkhead/genética , Proteínas con Homeodominio LIM/genética , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Factores de Transcripción/genética
19.
Cereb Cortex ; 30(9): 5147-5165, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32383447

RESUMEN

Foxg1 is an ancient transcription factor gene orchestrating a number of neurodevelopmental processes taking place in the rostral brain. In this study, we investigated its impact on neocortical activity. We found that mice overexpressing Foxg1 in neocortical pyramidal cells displayed an electroencephalography (EEG) with increased spike frequency and were more prone to kainic acid (KA)-induced seizures. Consistently, primary cultures of neocortical neurons gain-of-function for Foxg1 were hyperactive and hypersynchronized. That reflected an unbalanced expression of key genes encoding for ion channels, gamma aminobutyric acid and glutamate receptors, and was likely exacerbated by a pronounced interneuron depletion. We also detected a transient Foxg1 upregulation ignited in turn by neuronal activity and mediated by immediate early genes. Based on this, we propose that even small changes of Foxg1 levels may result in a profound impact on pyramidal cell activity, an issue relevant to neuronal physiology and neurological aberrancies associated to FOXG1 copy number variations.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Neocórtex/fisiología , Proteínas del Tejido Nervioso/metabolismo , Células Piramidales/metabolismo , Animales , Variaciones en el Número de Copia de ADN , Electroencefalografía , Factores de Transcripción Forkhead/genética , Ratones , Proteínas del Tejido Nervioso/genética , Convulsiones/genética , Convulsiones/metabolismo , Regulación hacia Arriba
20.
Cell Mol Life Sci ; 77(7): 1401-1419, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31485717

RESUMEN

Foxg1 is one of the forkhead box genes that are involved in morphogenesis, cell fate determination, and proliferation, and Foxg1 was previously reported to be required for morphogenesis of the mammalian inner ear. However, Foxg1 knock-out mice die at birth, and thus the role of Foxg1 in regulating hair cell (HC) regeneration after birth remains unclear. Here we used Sox2CreER/+ Foxg1loxp/loxp mice and Lgr5-EGFPCreER/+ Foxg1loxp/loxp mice to conditionally knock down Foxg1 specifically in Sox2+ SCs and Lgr5+ progenitors, respectively, in neonatal mice. We found that Foxg1 conditional knockdown (cKD) in Sox2+ SCs and Lgr5+ progenitors at postnatal day (P)1 both led to large numbers of extra HCs, especially extra inner HCs (IHCs) at P7, and these extra IHCs with normal hair bundles and synapses could survive at least to P30. The EdU assay failed to detect any EdU+ SCs, while the SC number was significantly decreased in Foxg1 cKD mice, and lineage tracing data showed that much more tdTomato+ HCs originated from Sox2+ SCs in Foxg1 cKD mice compared to the control mice. Moreover, the sphere-forming assay showed that Foxg1 cKD in Lgr5+ progenitors did not significantly change their sphere-forming ability. All these results suggest that Foxg1 cKD promotes HC regeneration and leads to large numbers of extra HCs probably by inducing direct trans-differentiation of SCs and progenitors to HCs. Real-time qPCR showed that cell cycle and Notch signaling pathways were significantly down-regulated in Foxg1 cKD mice cochlear SCs. Together, this study provides new evidence for the role of Foxg1 in regulating HC regeneration from SCs and progenitors in the neonatal mouse cochlea.


Asunto(s)
Transdiferenciación Celular , Cóclea/citología , Factores de Transcripción Forkhead/deficiencia , Células Ciliadas Auditivas/citología , Células Laberínticas de Soporte/citología , Proteínas del Tejido Nervioso/deficiencia , Animales , Animales Recién Nacidos , Recuento de Células , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Cóclea/inervación , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Ciliadas Auditivas/ultraestructura , Células Laberínticas de Soporte/ultraestructura , Mecanotransducción Celular , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal/genética , Células Madre/metabolismo , Sinapsis/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA