Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 506
Filtrar
1.
Int J Mol Sci ; 25(17)2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39273123

RESUMEN

Notch signaling is a conserved pathway crucial for nervous system development. Disruptions in this pathway are linked to neurodevelopmental disorders, neurodegenerative diseases, and brain tumors. Hairy/E(spl) (HES) genes, major downstream targets of Notch, are commonly used as markers for Notch activation. However, these genes can be activated, inhibited, or function independently of Notch signaling, and their response to Notch disruption varies across tissues and developmental stages. MIB1/Mib1 is an E3 ubiquitin ligase that enables Notch receptor activation by processing ligands like Delta and Serrate. We investigated Notch signaling disruption using the zebrafish Mib1 mutant line, mib1ta52b, focusing on changes in the expression of Hairy/E(spl) (her) genes. Our findings reveal significant variability in her gene expression across different neural cell types, regions, and developmental stages following Notch disruption. This variability questions the reliability of Hairy/E(spl) genes as universal markers for Notch activation, as their response is highly context-dependent. This study highlights the complex and context-specific nature of Notch signaling regulation. It underscores the need for a nuanced approach when using Hairy/E(spl) genes as markers for Notch activity. Additionally, it provides new insights into Mib1's role in Notch signaling, contributing to a better understanding of its involvement in Notch signaling-related disorders.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Receptores Notch , Transducción de Señal , Proteínas de Pez Cebra , Pez Cebra , Animales , Pez Cebra/genética , Pez Cebra/metabolismo , Receptores Notch/metabolismo , Receptores Notch/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Neurogénesis/genética
2.
Adv Healthc Mater ; : e2401419, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39252653

RESUMEN

Stem cell transplantation has demonstrated efficacy in treating neurological disorders by generating functional cells and secreting beneficial factors. However, challenges remain for current cell suspension injection therapy, including uncontrollable cell distribution, the potential for tumor formation, and limited ability to treat spatial defects. Therefore, implants with programmable cell development, tailored 3D structure, and functionalized biomaterials have the potential to both control cell distribution and reduce or heal spatial defects. Here, a biomimetic material system comprising gelatin, alginate, and fibrinogen has been developed for neural progenitor cell constructs using 3D printing. The resulting constructs exhibit excellent formability, stability, and developmental functions in vitro, as well as biocompatibility and integration into the hippocampus in vivo. The controllability, reproducibility, and material composition of the constructs show potential for use in personalized stem cell-based therapies for defective neurological disorders, neural development research, disease modeling, and organoid-derived intelligent systems.

3.
Mol Cells ; : 100111, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39265797

RESUMEN

Microtubules are core components of the neuronal cytoskeleton, providing structural support for the complex cytoarchitecture of neurons and serving as tracks for long-distance transport. The properties and functions of neuronal microtubules are controlled by tubulin isoforms and a variety of post-translational modifications (PTMs), collectively known as the 'tubulin code'. The tubulin code exerts direct control over the intrinsic properties of neuronal microtubules and regulates the repertoire of proteins that read the code, which in turn, has a significant impact on microtubule stability and dynamics. Here, we review progress in the understanding of the tubulin code in the nervous system, with a particular focus on tubulin PTMs that have been proposed as potential contributors to the development and maintenance of the mammalian nervous system. Furthermore, we also discuss the potential links between disruptions in the tubulin code and neurological disorders, including neurodevelopmental abnormalities and neurodegenerative diseases.

4.
Genome Biol ; 25(1): 221, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39143563

RESUMEN

BACKGROUND: Increasing evidence suggests that a substantial proportion of disease-associated mutations occur in enhancers, regions of non-coding DNA essential to gene regulation. Understanding the structures and mechanisms of the regulatory programs this variation affects can shed light on the apparatuses of human diseases. RESULTS: We collect epigenetic and gene expression datasets from seven early time points during neural differentiation. Focusing on this model system, we construct networks of enhancer-promoter interactions, each at an individual stage of neural induction. These networks serve as the base for a rich series of analyses, through which we demonstrate their temporal dynamics and enrichment for various disease-associated variants. We apply the Girvan-Newman clustering algorithm to these networks to reveal biologically relevant substructures of regulation. Additionally, we demonstrate methods to validate predicted enhancer-promoter interactions using transcription factor overexpression and massively parallel reporter assays. CONCLUSIONS: Our findings suggest a generalizable framework for exploring gene regulatory programs and their dynamics across developmental processes; this includes a comprehensive approach to studying the effects of disease-associated variation on transcriptional networks. The techniques applied to our networks have been published alongside our findings as a computational tool, E-P-INAnalyzer. Our procedure can be utilized across different cellular contexts and disorders.


Asunto(s)
Elementos de Facilitación Genéticos , Redes Reguladoras de Genes , Regiones Promotoras Genéticas , Humanos , Neurogénesis/genética , Diferenciación Celular , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Modelos Genéticos , Neuronas/metabolismo
5.
Cell Rep ; 43(9): 114666, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39182224

RESUMEN

The exon junction complex (EJC), nucleated by EIF4A3, is indispensable for mRNA fate and function throughout eukaryotes. We discover that EIF4A3 directly controls microtubules, independent of RNA, which is critical for neural wiring. While neuronal survival in the developing mouse cerebral cortex depends upon an intact EJC, axonal tract development requires only Eif4a3. Using human cortical organoids, we show that EIF4A3 disease mutations also impair neuronal growth, highlighting conserved functions relevant for neurodevelopmental pathology. Live imaging of growing neurons shows that EIF4A3 is essential for microtubule dynamics. Employing biochemistry and competition experiments, we demonstrate that EIF4A3 directly binds to microtubules, mutually exclusive of the EJC. Finally, in vitro reconstitution assays and rescue experiments demonstrate that EIF4A3 is sufficient to promote microtubule polymerization and that EIF4A3-microtubule association is a major contributor to axon growth. This reveals a fundamental mechanism by which neurons re-utilize core gene expression machinery to directly control the cytoskeleton.

6.
Cell ; 187(17): 4690-4712.e30, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39142281

RESUMEN

Electrical excitability-the ability to fire and propagate action potentials-is a signature feature of neurons. How neurons become excitable during development and whether excitability is an intrinsic property of neurons remain unclear. Here, we demonstrate that Schwann cells, the most abundant glia in the peripheral nervous system, promote somatosensory neuron excitability during development. We find that Schwann cells secrete prostaglandin E2, which is necessary and sufficient to induce developing somatosensory neurons to express normal levels of genes required for neuronal function, including voltage-gated sodium channels, and to fire action potential trains. Inactivating this signaling pathway in Schwann cells impairs somatosensory neuron maturation, causing multimodal sensory defects that persist into adulthood. Collectively, our studies uncover a neurodevelopmental role for prostaglandin E2 distinct from its established role in inflammation, revealing a cell non-autonomous mechanism by which glia regulate neuronal excitability to enable the development of normal sensory functions.


Asunto(s)
Potenciales de Acción , Dinoprostona , Células de Schwann , Células Receptoras Sensoriales , Animales , Células de Schwann/metabolismo , Dinoprostona/metabolismo , Ratones , Células Receptoras Sensoriales/metabolismo , Transducción de Señal
7.
bioRxiv ; 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39185162

RESUMEN

Nutritional fluctuations that occur early in life dictate metabolic adaptations that will affect susceptibility to weight gain and obesity later in life. The postnatal period in mice represents a time of dynamic changes in hypothalamic development and maternal consumption of a high fat diet during the lactation period (MHFD) changes the composition of milk and leads to enhanced susceptibility to obesity in offspring. Agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus (ARH) react to changes in multiple metabolic signals and distribute neuroendocrine information to other brain regions, such as the paraventricular hypothalamic nucleus (PVH), which is known to integrate a variety of signals that regulate body weight. Development of neural projections from AgRP neurons to the PVH occurs during the lactation period and these projections are reduced in MHFD offspring, but underlying developmental mechanisms remain largely unknown. Microglia are the resident immune cells of the central nervous system and are involved in refinement of neural connections and modulation of synaptic transmission. Because high fat diet exposure causes activation of microglia in adults, a similar activation may occur in offspring exposed to MHFD and play a role in sculpting hypothalamic feeding circuitry. Genetically targeted axonal labeling and immunohistochemistry were used to visualize AgRP axons and microglia in postnatal mice derived from MHFD dams and morphological changes quantified. The results demonstrate regionally localized changes to microglial morphology in the PVH of MHFD offspring that suggest enhanced surveillance activity and are temporally restricted to the period when AgRP neurons innervate the PVH. In addition, axon labeling experiments confirm a significant decrease in AgRP innervation of the PVH in MHFD offspring and provide direct evidence of synaptic pruning of AgRP inputs to the PVH. Microglial depletion with the Colony-stimulating factor 1 receptor inhibitor PLX5622 determined that the decrease in AgRP innervation observed in MHFD offspring is dependent on microglia, and that microglia are required for weight gain that emerges as early as weaning in offspring of MHFD dams. However, these changes do not appear to be dependent on the degree of microglial mediated synaptic pruning. Together, these findings suggest that microglia are activated by exposure to MHFD and interact directly with AgRP axons during development to permanently alter their density, with implications for developmental programming of metabolic phenotype.

8.
Cell Rep ; 43(8): 114615, 2024 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-39133615

RESUMEN

In vertebrate retina, individual neurons of the same type are distributed regularly across the tissue in a pattern known as a mosaic. Establishment of mosaics during development requires cell-cell repulsion among homotypic neurons, but the mechanisms underlying this repulsion remain unknown. Here, we show that two mouse retinal cell types, OFF and ON starburst amacrine cells, establish mosaic spacing by using their dendritic arbors to repel neighboring homotypic somata. Using transgenic tools and single-cell labeling, we identify a developmental period when starburst somata are contacted by neighboring starburst dendrites; these serve to exclude somata from settling within the neighbor's dendritic territory. Dendrite-soma exclusion is mediated by MEGF10, a cell-surface molecule required for starburst mosaic patterning. Our results implicate dendrite-soma exclusion as a key mechanism underlying starburst mosaic spacing and raise the possibility that this could be a general mechanism for mosaic patterning across many cell types and species.


Asunto(s)
Dendritas , Animales , Dendritas/metabolismo , Ratones , Células Amacrinas/metabolismo , Células Amacrinas/citología , Retina/citología , Retina/metabolismo , Mosaicismo , Neuronas Retinianas/citología , Neuronas Retinianas/metabolismo , Ratones Transgénicos , Ratones Endogámicos C57BL
9.
Ecotoxicol Environ Saf ; 283: 116862, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39128450

RESUMEN

Estrogens and estrogenic chemicals are endocrine-disrupting chemicals (EDCs). The potential toxicity of EDCs to humans and aquatic organisms has become increasingly concerning. However, at present, the potential toxic mechanisms of EDCs on neural and vascular development are still being fully investigated. During the study, we utilized zebrafish to assess the developmental neural and vascular toxicity of different estrogens. The results indicated that zebrafish treated with different estrogens, especially E2, exhibit developmental malformations, including increased mortality, decreased body length, decreased heart rate, aberrant swimming behavior, and increased developmental malformations, including spinal curvature (SC), yolk edema (YE) and pericaidial edema (PE), in a dose-dependent manner with 72 h-treated. Further morphological evaluation revealed that E2 exposure significantly induced motor neural abnormalities in zebrafish embryos. In addition, treated with these three estrogens also impaired the vascular development in the early stage of zebrafish embryos. Mechanistically, the identification of downstream factors revealed that several key neural and vascular development-related genes, including syn2a, gfap, gap43, shha, kdr, flt1 and flt4, were transcriptionally downregulated after estrogen exposure in zebrafish, suggesting that estrogen exposure might cause neural and vascular toxicity by interfering the mRNA levels of genes relevant to neural and vascular development.


Asunto(s)
Disruptores Endocrinos , Estrógenos , Contaminantes Químicos del Agua , Pez Cebra , Animales , Disruptores Endocrinos/toxicidad , Estrógenos/toxicidad , Contaminantes Químicos del Agua/toxicidad , Embrión no Mamífero/efectos de los fármacos , Estradiol/toxicidad
10.
Trends Neurosci ; 47(9): 667-668, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39142912

RESUMEN

The maturation of cerebral cortical networks during early life involves a major reorganization of long-range axonal connections. In a recent study, Bragg-Gonzalo, Aguilera, et al. discovered that in mice, the interhemispheric connections sent by S1L4 callosal projection neurons are pruned via the tight control of their ipsilateral synaptic integration, which relies on the early activity of specific interneurons.


Asunto(s)
Corteza Cerebral , Inhibición Neural , Animales , Inhibición Neural/fisiología , Corteza Cerebral/fisiología , Humanos , Neuronas/fisiología , Vías Nerviosas/fisiología , Cuerpo Calloso/fisiología , Interneuronas/fisiología , Red Nerviosa/fisiología , Ratones
11.
Dev Biol ; 515: 178-185, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39021074

RESUMEN

The somatosensory system detects peripheral stimuli that are translated into behaviors necessary for survival. Fishes and amphibians possess two somatosensory systems in the trunk: the primary somatosensory system, formed by the Rohon-Beard neurons, and the secondary somatosensory system, formed by the neural crest cell-derived neurons of the Dorsal Root Ganglia. Rohon-Beard neurons have been characterized as a transient population that mostly disappears during the first days of life and is functionally replaced by the Dorsal Root Ganglia. Here, I follow Rohon-Beard neurons in vivo and show that the entire repertoire remains present in zebrafish from 1-day post-fertilization until the juvenile stage, 15-days post-fertilization. These data indicate that zebrafish retain two complete somatosensory systems until at least a developmental stage when the animals display complex behavioral repertoires.


Asunto(s)
Pez Cebra , Animales , Pez Cebra/embriología , Ganglios Espinales/embriología , Neuronas/fisiología , Cresta Neural/citología , Cresta Neural/embriología , Cresta Neural/fisiología
12.
Acta Histochem Cytochem ; 57(3): 89-100, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38988694

RESUMEN

Neural stem/progenitor cells (NSPCs) in specific brain regions require precisely regulated metabolite production during critical development periods. Purines-vital components of DNA, RNA, and energy carriers like ATP and GTP-are crucial metabolites in brain development. Purine levels are tightly controlled through two pathways: de novo synthesis and salvage synthesis. Enzymes driving de novo pathway are assembled into a large multienzyme complex termed the "purinosome." Here, we review purine metabolism and purinosomes as spatiotemporal regulators of neural development. Notably, around postnatal day 0 (P0) during mouse cortical development, purine synthesis transitions from the de novo pathway to the salvage pathway. Inhibiting the de novo pathway affects mTORC1 pathway and leads to specific forebrain malformations. In this review, we also explore the importance of protein-protein interactions of a newly identified NSPC protein-NACHT and WD repeat domain-containing 1 (Nwd1)-in purinosome formation. Reduced Nwd1 expression disrupts purinosome formation, impacting NSPC proliferation and neuronal migration, resulting in periventricular heterotopia. Nwd1 interacts directly with phosphoribosylaminoimidazole-succinocarboxamide synthetase (PAICS), an enzyme involved in de novo purine synthesis. We anticipate this review will be valuable for researchers investigating neural development, purine metabolism, and protein-protein interactions.

13.
Dev Neurobiol ; 84(3): 203-216, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38830696

RESUMEN

Formation of the corpus callosum (CC), anterior commissure (AC), and postoptic commissure (POC), connecting the left and right cerebral hemispheres, is crucial for cerebral functioning. Collapsin response mediator protein 2 (CRMP2) has been suggested to be associated with the mechanisms governing this formation, based on knockout studies in mice and knockdown/knockout studies in zebrafish. Previously, we reported two cases of non-synonymous CRMP2 variants with S14R and R565C substitutions. Among the, the R565C substitution (p.R565C) was caused by the novel CRMP2 mutation c.1693C > T, and the patient presented with intellectual disability accompanied by CC hypoplasia. In this study, we demonstrate that crmp2 mRNA could rescue AC and POC formation in crmp2-knockdown zebrafish, whereas the mRNA with the R566C mutation could not. Zebrafish CRMP2 R566C corresponds to human CRMP2 R565C. Further experiments with transfected cultured cells indicated that CRMP2 with the R566C mutation could not bind to kinesin light chain 1 (KLC1). Knockdown of klc1a in zebrafish resulted in defective AC and POC formation, revealing a genetic interaction with crmp2. These findings suggest that the CRMP2 R566C mutant fails to bind to KLC1, preventing axonal elongation and leading to defective AC and POC formation in zebrafish and CC formation defects in humans. Our study highlights the importance of the interaction between CRMP2 and KLC1 in the formation of the forebrain commissures, revealing a novel mechanism associated with CRMP2 mutations underlying human neurodevelopmental abnormalities.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular , Proteínas del Tejido Nervioso , Proteínas de Pez Cebra , Pez Cebra , Animales , Humanos , Animales Modificados Genéticamente , Cuerpo Calloso/metabolismo , Embrión no Mamífero , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Cinesinas/metabolismo , Cinesinas/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Prosencéfalo/metabolismo , Proteínas de Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética
14.
Cell Rep ; 43(6): 114339, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38852158

RESUMEN

Quiescent adult neural stem cells (NSCs) in the mammalian brain arise from proliferating NSCs during development. Beyond acquisition of quiescence, an adult NSC hallmark, little is known about the process, milestones, and mechanisms underlying the transition of developmental NSCs to an adult NSC state. Here, we performed targeted single-cell RNA-seq analysis to reveal the molecular cascade underlying NSC development in the early postnatal mouse dentate gyrus. We identified two sequential steps, first a transition to quiescence followed by further maturation, each of which involved distinct changes in metabolic gene expression. Direct metabolic analysis uncovered distinct milestones, including an autophagy burst before NSC quiescence acquisition and cellular reactive oxygen species level elevation along NSC maturation. Functionally, autophagy is important for the NSC transition to quiescence during early postnatal development. Together, our study reveals a multi-step process with defined milestones underlying establishment of the adult NSC pool in the mammalian brain.


Asunto(s)
Autofagia , Hipocampo , Células-Madre Neurales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Animales , Ratones , Hipocampo/metabolismo , Hipocampo/citología , Neurogénesis , Giro Dentado/metabolismo , Giro Dentado/citología , Giro Dentado/crecimiento & desarrollo , Diferenciación Celular , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Células Madre Adultas/metabolismo , Células Madre Adultas/citología , Análisis de la Célula Individual , Proliferación Celular
15.
bioRxiv ; 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38826254

RESUMEN

Background: Increasing evidence suggests that a substantial proportion of disease-associated mutations occur in enhancers, regions of non-coding DNA essential to gene regulation. Understanding the structures and mechanisms of regulatory programs this variation affects can shed light on the apparatuses of human diseases. Results: We collected epigenetic and gene expression datasets from seven early time points during neural differentiation. Focusing on this model system, we constructed networks of enhancer-promoter interactions, each at an individual stage of neural induction. These networks served as the base for a rich series of analyses, through which we demonstrated their temporal dynamics and enrichment for various disease-associated variants. We applied the Girvan-Newman clustering algorithm to these networks to reveal biologically relevant substructures of regulation. Additionally, we demonstrated methods to validate predicted enhancer-promoter interactions using transcription factor overexpression and massively parallel reporter assays. Conclusions: Our findings suggest a generalizable framework for exploring gene regulatory programs and their dynamics across developmental processes. This includes a comprehensive approach to studying the effects of disease-associated variation on transcriptional networks. The techniques applied to our networks have been published alongside our findings as a computational tool, E-P-INAnalyzer. Our procedure can be utilized across different cellular contexts and disorders.

16.
Nutrients ; 16(11)2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38892717

RESUMEN

The placenta is the largest fetal organ, which connects the mother to the fetus and supports most aspects of organogenesis through the transport of nutrients and gases. However, further studies are needed to assess placental pathology as a reliable predictor of long-term physical growth or neural development in newborns. The Consensus Statement of the Amsterdam Placental Workshop Group (APWGCS) on the sampling and definition of placental lesions has resulted in diagnostic uniformity in describing the most common pathological lesions of the placenta and contributed to the international standardization of descriptions of placental pathology. In this narrative review, we reclassified descriptions of placental pathology from previously published papers according to the APWGCS criteria and comparatively assessed the relationship with infantile physical and/or neural development. After reclassification and reevaluation, placental pathology of maternal vascular malperfusion, one of the APWGCS criteria, emerged as a promising candidate as a universal predictor of negative infantile neurodevelopmental outcomes, not only in term and preterm deliveries but also in high-risk groups of very low birthweight newborns. However, there are few studies that examined placental pathology according to the full categories of APWGCS and also included low-risk general infants. It is necessary to incorporate the assessment of placental pathology utilizing APWGCS in the design of future birth cohort studies as well as in follow-up investigations of high-risk infants.


Asunto(s)
Consenso , Placenta , Humanos , Embarazo , Femenino , Placenta/patología , Recién Nacido , Enfermedades Placentarias/patología , Enfermedades Placentarias/diagnóstico , Desarrollo Infantil , Lactante , Países Bajos
17.
G3 (Bethesda) ; 14(9)2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-38869246

RESUMEN

This study investigated the dominant blue eyes (DBE) trait linked to hearing impairment and variable white spotting in Maine Coon cats. Fifty-eight animals descending from 2 different DBE lineages, the Dutch and the Topaz lines, were sampled. They comprised 48 cats from the Dutch bloodline, including 9 green-eyed and 31 blue-eyed cats, with some individuals exhibiting signs of deafness, and 8 stillborn kittens. Samples from the Topaz lineage included 10 blue-eyed animals. A brainstem auditory evoked response test revealed a reduced to absent response to auditory stimuli and absent physiological waveforms in all of the 8 examined DBE animals. We sequenced the genome of 2 affected cats from the Dutch line and searched for variants in 19 candidate genes for the human Waardenburg syndrome and pigmentary disorders. This search yielded 9 private protein-changing candidate variants in the genes PAX3, EDN3, KIT, OCA2, SLC24A5, HERC2, and TYRP1. The genotype-phenotype cosegregation was observed for the PAX3 variant within all animals from the Dutch lineage. The mutant allele was absent from 461 control genomes and 241 additionally genotyped green-eyed Maine Coons. We considered the PAX3 variant as the most plausible candidate-a heterozygous nonsense single base pair substitution in exon 6 of PAX3 (NC_051841.1:g.205,787,310G>A, XM_019838731.3:c.937C>T, XP_019694290.1:p.Gln313*), predicted to result in a premature stop codon. PAX3 variants cause auditory-pigmentary syndrome in humans, horses, and mice. Together with the comparative data from other species, our findings strongly suggest PAX3:c.937C>T (OMIA:001688-9685) as the most likely candidate variant for the DBE, deafness, and minimal white spotting in the Maine Coon Dutch line. Finally, we propose the designation of DBERE (Rociri Elvis Dominant Blue Eyes) allele in the domestic cat.


Asunto(s)
Haploinsuficiencia , Pérdida Auditiva , Factor de Transcripción PAX3 , Síndrome de Waardenburg , Animales , Gatos , Síndrome de Waardenburg/genética , Síndrome de Waardenburg/veterinaria , Factor de Transcripción PAX3/genética , Pérdida Auditiva/genética , Pérdida Auditiva/veterinaria , Humanos , Color del Ojo/genética , Masculino , Fenotipo , Femenino , Alelos
18.
Trends Neurosci ; 47(7): 491-505, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38897852

RESUMEN

While many core biological processes are conserved across species, the human brain has evolved with unique capacities. Current understanding of the neurobiological mechanisms that endow human traits as well as associated vulnerabilities remains limited. However, emerging data have illuminated species divergence in DNA elements and genome organization, in molecular, morphological, and functional features of conserved neural cell types, as well as temporal differences in brain development. Here, we summarize recent data on unique features of the human brain and their complex implications for the study and treatment of brain diseases. We also consider key outstanding questions in the field and discuss the technologies and foundational knowledge that will be required to accelerate understanding of human neurobiology.


Asunto(s)
Encéfalo , Humanos , Encéfalo/fisiología , Animales , Genómica/métodos , Evolución Biológica
19.
Neurosci Biobehav Rev ; 163: 105744, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38825259

RESUMEN

Every species' brain, body and behavior is shaped by the contingencies of their evolutionary history; these exert pressures that change their developmental trajectories. There is, however, another set of contingencies that shape us and other animals: those that occur during a lifetime. In this perspective piece, we show how these two histories are intertwined by focusing on the individual. We suggest that organisms--their brains and behaviors--are not solely the developmental products of genes and neural circuitry but individual centers of action unfolding in time. To unpack this idea, we first emphasize the importance of variation and the central role of the individual in biology. We then go over "errors in time" that we often make when comparing development across species. Next, we reveal how an individual's development is a process rather than a product by presenting a set of case studies. These show developmental trajectories as emerging in the contexts of the "the actual now" and "the presence of the past". Our consideration reveals that individuals are slippery-they are never static; they are a set of on-going, creative activities. In light of this, it seems that taking individual development seriously is essential if we aspire to make meaningful comparisons of neural circuits and behavior within and across species.


Asunto(s)
Evolución Biológica , Encéfalo , Encéfalo/fisiología , Encéfalo/crecimiento & desarrollo , Animales , Humanos
20.
Front Neural Circuits ; 18: 1409680, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38860141

RESUMEN

The brain constructs spatially organized sensory maps to represent sensory information. The formation of sensory maps has traditionally been thought to depend on synchronous neuronal activity. However, recent evidence from the olfactory system suggests that cell type-specific temporal patterns of spontaneous activity play an instructive role in shaping the olfactory glomerular map. These findings challenge traditional views and highlight the importance of investigating the spatiotemporal dynamics of neural activity to understand the development of complex neural circuits. This review discusses the implications of new findings in the olfactory system and outlines future research directions.


Asunto(s)
Vías Olfatorias , Animales , Vías Olfatorias/fisiología , Vías Olfatorias/citología , Humanos , Red Nerviosa/fisiología , Neuronas/fisiología , Bulbo Olfatorio/fisiología , Bulbo Olfatorio/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA