Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.041
Filter
1.
Nature ; 609(7929): 907-910, 2022 09.
Article in English | MEDLINE | ID: mdl-36171373

ABSTRACT

Self-organizing three-dimensional cellular models derived from human pluripotent stem cells or primary tissue have great potential to provide insights into how the human nervous system develops, what makes it unique and how disorders of the nervous system arise, progress and could be treated. Here, to facilitate progress and improve communication with the scientific community and the public, we clarify and provide a basic framework for the nomenclature of human multicellular models of nervous system development and disease, including organoids, assembloids and transplants.


Subject(s)
Consensus , Nervous System , Organoids , Terminology as Topic , Humans , Models, Biological , Nervous System/cytology , Nervous System/pathology , Organoids/cytology , Organoids/pathology , Pluripotent Stem Cells/cytology
2.
Science ; 374(6568): 717-723, 2021 Nov 05.
Article in English | MEDLINE | ID: mdl-34735222

ABSTRACT

The evolutionary origin of metazoan cell types such as neurons and muscles is not known. Using whole-body single-cell RNA sequencing in a sponge, an animal without nervous system and musculature, we identified 18 distinct cell types. These include nitric oxide­sensitive contractile pinacocytes, amoeboid phagocytes, and secretory neuroid cells that reside in close contact with digestive choanocytes that express scaffolding and receptor proteins. Visualizing neuroid cells by correlative x-ray and electron microscopy revealed secretory vesicles and cellular projections enwrapping choanocyte microvilli and cilia. Our data show a communication system that is organized around sponge digestive chambers, using conserved modules that became incorporated into the pre- and postsynapse in the nervous systems of other animals.


Subject(s)
Biological Evolution , Porifera/cytology , Animals , Cell Communication , Cell Surface Extensions/ultrastructure , Cilia/physiology , Cilia/ultrastructure , Digestive System/cytology , Mesoderm/cytology , Nervous System/cytology , Nervous System Physiological Phenomena , Nitric Oxide/metabolism , Porifera/genetics , Porifera/metabolism , RNA-Seq , Secretory Vesicles/ultrastructure , Signal Transduction , Single-Cell Analysis , Transcriptome
3.
Nature ; 600(7887): 93-99, 2021 12.
Article in English | MEDLINE | ID: mdl-34759317

ABSTRACT

In most animals, the majority of the nervous system is generated and assembled into neuronal circuits during embryonic development1. However, during juvenile stages, nervous systems still undergo extensive anatomical and functional changes to eventually form a fully mature nervous system by the adult stage2,3. The molecular changes in post-mitotic neurons across post-embryonic development and the genetic programs that control these temporal transitions are not well understood4,5. Here, using the model system Caenorhabditis elegans, we comprehensively characterized the distinct functional states (locomotor behaviour) and the corresponding distinct molecular states (transcriptome) of the post-mitotic nervous system across temporal transitions during post-embryonic development. We observed pervasive, neuron-type-specific changes in gene expression, many of which are controlled by the developmental upregulation of the conserved heterochronic microRNA LIN-4 and the subsequent promotion of a mature neuronal transcriptional program through the repression of its target, the transcription factor lin-14. The functional relevance of these molecular transitions are exemplified by a temporally regulated target gene of the LIN-14 transcription factor, nlp-45, a neuropeptide-encoding gene, which we find is required for several distinct temporal transitions in exploratory activity during post-embryonic development. Our study provides insights into regulatory strategies that control neuron-type-specific gene batteries to modulate distinct behavioural states across temporal, sexual and environmental dimensions of post-embryonic development.


Subject(s)
Caenorhabditis elegans/growth & development , Mitosis , Nervous System/growth & development , Neurons/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Exploratory Behavior , Female , Gene Expression Regulation, Developmental , Locomotion/genetics , Male , MicroRNAs/genetics , Nervous System/cytology , Nervous System/metabolism , Neuropeptides/genetics , Nuclear Proteins/genetics , Time Factors , Transcriptome
4.
Biomed Res Int ; 2021: 1601565, 2021.
Article in English | MEDLINE | ID: mdl-34604381

ABSTRACT

Trogocytosis is a general biological process that involves one cell physically taking small parts of the membrane and other components from another cell. In trogocytosis, one cell seems to take little "bites" from another cell resulting in multiple outcomes from these cell-cell interactions. Trogocytosis was first described in protozoan parasites, which by taking pieces of host cells, kill them and cause tissue damage. Now, it is known that this process is also performed by cells of the immune system with important consequences such as cell communication and activation, elimination of microbial pathogens, and even control of cancer cells. More recently, trogocytosis has also been reported to occur in cells of the central nervous system and in various cells during development. Some of the molecules involved in phagocytosis also participate in trogocytosis. However, the molecular mechanisms that regulate trogocytosis are still a mystery. Elucidating these mechanisms is becoming a research area of much interest. For example, why neutrophils can engage trogocytosis to kill Trichomonas vaginalis parasites, but neutrophils use phagocytosis to eliminate already death parasites? Thus, trogocytosis is a significant process in normal physiology that multiple cells from different organisms use in various scenarios of health and disease. In this review, we present the basic principles known on the process of trogocytosis and discuss the importance in this process to host-pathogen interactions and to normal functions in the immune and nervous systems.


Subject(s)
Growth and Development , Immunity , Nervous System/cytology , Trogocytosis/immunology , Animals , Bacteria/metabolism , Cell Death , Humans
5.
Int J Mol Sci ; 22(5)2021 Mar 05.
Article in English | MEDLINE | ID: mdl-33807555

ABSTRACT

Although the human brain would be an ideal model for studying human neuropathology, it is difficult to perform in vitro culture of human brain cells from genetically engineered healthy or diseased brain tissue. Therefore, a suitable model for studying the molecular mechanisms responsible for neurological diseases that can appropriately mimic the human brain is needed. Somatic cell nuclear transfer (SCNT) was performed using an established porcine Yucatan EGFP cell line and whole seeding was performed using SCNT blastocysts. Two Yucatan EGFP porcine embryonic stem-like cell (pESLC) lines were established. These pESLC lines were then used to establish an in vitro neuro-organoids. Aggregates were cultured in vitro until 61 or 102 days after neural induction, neural patterning, and neural expansion. The neuro-organoids were sampled at each step and the expression of the dopaminergic neuronal marker (TH) and mature neuronal marker (MAP2) was confirmed by reverse transcription-PCR. Expression of the neural stem cell marker (PAX6), neural precursor markers (S100 and SOX2), and early neural markers (MAP2 and Nestin) were confirmed by immunofluorescence staining. In conclusion, we successfully established neuro-organoids derived from pESLCs in vitro. This protocol can be used as a tool to develop in vitro models for drug development, patient-specific chemotherapy, and human central nervous system disease studies.


Subject(s)
Embryonic Stem Cells/cytology , Organoids/cytology , Animals , Biomarkers/metabolism , Blastocyst/cytology , Blastocyst/metabolism , Cell Line , Embryonic Stem Cells/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Inbred ICR , Nervous System/cytology , Nervous System/metabolism , Nuclear Transfer Techniques , Organoids/metabolism , Swine
6.
Genes (Basel) ; 12(2)2021 02 10.
Article in English | MEDLINE | ID: mdl-33578707

ABSTRACT

Holothurians, or sea cucumbers, belong to the phylum Echinodermata. They show good regenerative abilities. The present review provides an analysis of available data on the molecular aspects of regeneration mechanisms in holothurians. The genes and signaling pathways activated during the asexual reproduction and the formation of the anterior and posterior parts of the body, as well as the molecular mechanisms that provide regeneration of the nervous and digestive systems, are considered here. Damage causes a strong stress response, the signs of which are recorded even at late regeneration stages. In holothurian tissues, the concentrations of reactive oxygen species and antioxidant enzymes increase. Furthermore, the cellular and humoral components of the immune system are activated. Extracellular matrix remodeling and Wnt signaling play a major role in the regeneration in holothurians. All available morphological and molecular data show that the dedifferentiation of specialized cells in the remnant of the organ and the epithelial morphogenesis constitute the basis of regeneration in holothurians. However, depending on the type of damage, the mechanisms of regeneration may differ significantly in the spatial organization of regeneration process, the involvement of different cell types, and the depth of reprogramming of their genome (dedifferentiation or transdifferentiation).


Subject(s)
Digestive System/metabolism , Immune System/metabolism , Nervous System/metabolism , Proteins/genetics , Regeneration/genetics , Sea Cucumbers/genetics , Animals , Antioxidants/metabolism , Digestive System/cytology , Digestive System/growth & development , Digestive System/injuries , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Gene Expression Regulation , Immune System/cytology , Immune System/growth & development , Immune System/injuries , Nervous System/cytology , Nervous System/growth & development , Proteins/metabolism , Reactive Oxygen Species/metabolism , Reproduction, Asexual/genetics , Sea Cucumbers/growth & development , Sea Cucumbers/metabolism , Wnt Signaling Pathway
7.
Dev Biol ; 475: 165-180, 2021 07.
Article in English | MEDLINE | ID: mdl-32017903

ABSTRACT

Complex nervous systems have a modular architecture, whereby reiterative groups of neurons ("modules") that share certain structural and functional properties are integrated into large neural circuits. Neurons develop from proliferating progenitor cells that, based on their location and time of appearance, are defined by certain genetic programs. Given that genes expressed by a given progenitor play a fundamental role in determining the properties of its lineage (i.e., the neurons descended from that progenitor), one efficient developmental strategy would be to have lineages give rise to the structural modules of the mature nervous system. It is clear that this strategy plays an important role in neural development of many invertebrate animals, notably insects, where the availability of genetic techniques has made it possible to analyze the precise relationship between neuronal origin and differentiation since several decades. Similar techniques, developed more recently in the vertebrate field, reveal that functional modules of the mammalian cerebral cortex are also likely products of developmentally defined lineages. We will review studies that relate cell lineage to circuitry and function from a comparative developmental perspective, aiming at enhancing our understanding of neural progenitors and their lineages, and translating findings acquired in different model systems into a common conceptual framework.


Subject(s)
Cell Lineage/physiology , Nerve Net/cytology , Neurons/cytology , Animals , Brain/cytology , Cell Differentiation , Cell Lineage/genetics , Cerebral Cortex/cytology , Gene Expression Regulation, Developmental , Humans , Nerve Net/metabolism , Nerve Net/physiology , Nervous System/cytology , Neurons/metabolism , Neurons/physiology , Stem Cells/cytology
8.
FEBS J ; 288(1): 99-110, 2021 01.
Article in English | MEDLINE | ID: mdl-32307851

ABSTRACT

How evolutionary novelties have arisen is one of the central questions in evolutionary biology. Preexisting gene regulatory networks or signaling pathways have been shown to be co-opted for building novel traits in several organisms. However, the structure of entire gene regulatory networks and evolutionary events of gene co-option for emergence of a novel trait are poorly understood. In this study, to explore the genetic and molecular bases of the novel wing pigmentation pattern of a polka-dotted fruit fly (Drosophila guttifera), we performed de novo genome sequencing and transcriptome analyses. As a result, we comprehensively identified the genes associated with the pigmentation pattern. Furthermore, we revealed that 151 of these associated genes were positively or negatively regulated by wingless, a master regulator of wing pigmentation. Genes for neural development, Wnt signaling, Dpp signaling, and effectors (such as enzymes) for melanin pigmentation were included among these 151 genes. None of the known regulatory genes that regulate pigmentation pattern formation in other fruit fly species were included. Our results suggest that the novel pigmentation pattern of a polka-dotted fruit fly might have emerged through multistep co-options of multiple gene regulatory networks, signaling pathways, and effector genes, rather than recruitment of one large gene circuit.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Neurogenesis/genetics , Pigmentation/genetics , Transcriptome , Wings, Animal/metabolism , Wnt1 Protein/genetics , Animals , Biological Evolution , Drosophila/growth & development , Drosophila/metabolism , Drosophila Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Melanins/genetics , Melanins/metabolism , Nervous System/cytology , Nervous System/growth & development , Nervous System/metabolism , Phenotype , Signal Transduction , Wings, Animal/growth & development , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt1 Protein/metabolism
9.
Cell Mol Life Sci ; 78(5): 1971-1982, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33151389

ABSTRACT

Understanding how an adult brain reaches an appropriate size and cell composition from a pool of progenitors that proliferates and differentiates is a key question in Developmental Neurobiology. Not only the control of final size but also, the proper arrangement of cells of different embryonic origins is fundamental in this process. Each neural progenitor has to produce a precise number of sibling cells that establish clones, and all these clones will come together to form the functional adult nervous system. Lineage cell tracing is a complex and challenging process that aims to reconstruct the offspring that arise from a single progenitor cell. This tracing can be achieved through strategies based on genetically modified organisms, using either genetic tracers, transfected viral vectors or DNA constructs, and even single-cell sequencing. Combining different reporter proteins and the use of transgenic mice revolutionized clonal analysis more than a decade ago and now, the availability of novel genome editing tools and single-cell sequencing techniques has vastly improved the capacity of lineage tracing to decipher progenitor potential. This review brings together the strategies used to study cell lineages in the brain and the role they have played in our understanding of the functional clonal relationships among neural cells. In addition, future perspectives regarding the study of cell heterogeneity and the ontogeny of different cell lineages will also be addressed.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Gene Expression Profiling/methods , Nervous System/metabolism , Neural Stem Cells/metabolism , Animals , CRISPR-Cas Systems/genetics , Gene Editing/methods , High-Throughput Nucleotide Sequencing/methods , Nervous System/cytology , Neural Stem Cells/cytology , Single-Cell Analysis/methods
10.
J Histochem Cytochem ; 69(1): 61-80, 2021 01.
Article in English | MEDLINE | ID: mdl-32936033

ABSTRACT

The orderly development of the nervous system is characterized by phases of cell proliferation and differentiation, neural migration, axonal outgrowth and synapse formation, and stabilization. Each of these processes is a result of the modulation of genetic programs by extracellular cues. In particular, chondroitin sulfate proteoglycans (CSPGs) have been found to be involved in almost every aspect of this well-orchestrated yet delicate process. The evidence of their involvement is complex, often contradictory, and lacking in mechanistic clarity; however, it remains obvious that CSPGs are key cogs in building a functional brain. This review focuses on current knowledge of the role of CSPGs in each of the major stages of neural development with emphasis on areas requiring further investigation.


Subject(s)
Chondroitin Sulfate Proteoglycans/metabolism , Nervous System/growth & development , Neurogenesis , Animals , Brain/cytology , Brain/embryology , Brain/growth & development , Brain/metabolism , Cell Movement , Humans , Nervous System/cytology , Nervous System/embryology , Nervous System/metabolism , Neurons/cytology , Neurons/metabolism , Synapses/metabolism
11.
Nat Commun ; 11(1): 6411, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33339816

ABSTRACT

Over 250 million people suffer from schistosomiasis, a tropical disease caused by parasitic flatworms known as schistosomes. Humans become infected by free-swimming, water-borne larvae, which penetrate the skin. The earliest intra-mammalian stage, called the schistosomulum, undergoes a series of developmental transitions. These changes are critical for the parasite to adapt to its new environment as it navigates through host tissues to reach its niche, where it will grow to reproductive maturity. Unravelling the mechanisms that drive intra-mammalian development requires knowledge of the spatial organisation and transcriptional dynamics of different cell types that comprise the schistomulum body. To fill these important knowledge gaps, we perform single-cell RNA sequencing on two-day old schistosomula of Schistosoma mansoni. We identify likely gene expression profiles for muscle, nervous system, tegument, oesophageal gland, parenchymal/primordial gut cells, and stem cells. In addition, we validate cell markers for all these clusters by in situ hybridisation in schistosomula and adult parasites. Taken together, this study provides a comprehensive cell-type atlas for the early intra-mammalian stage of this devastating metazoan parasite.


Subject(s)
Mammals/parasitology , Parasites/cytology , Parasites/growth & development , Schistosoma mansoni/cytology , Schistosoma mansoni/growth & development , Single-Cell Analysis , Animals , Esophagus/metabolism , Exons/genetics , Gene Expression Regulation , Humans , Muscle Cells/metabolism , Nervous System/cytology , Neurons/cytology , Parasites/genetics , Schistosoma mansoni/genetics , Stem Cells/cytology , Stem Cells/metabolism , Transcription, Genetic
12.
Sci Rep ; 10(1): 18746, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33127972

ABSTRACT

Dendrites, branched structures extending from neuronal cell soma, are specialized for processing information from other neurons. The morphogenesis of dendritic structures is spatiotemporally regulated by well-orchestrated signaling cascades. Dysregulation of these processes impacts the wiring of neuronal circuit and efficacy of neurotransmission, which contribute to the pathogeneses of neurological disorders. While Cdk5 (cyclin-dependent kinase 5) plays a critical role in neuronal dendritic development, its underlying molecular control is not fully understood. In this study, we show that p39, one of the two neuronal Cdk5 activators, is a key regulator of dendritic morphogenesis. Pyramidal neurons deficient in p39 exhibit aberrant dendritic morphology characterized by shorter length and reduced arborization, which is comparable to dendrites in Cdk5-deficient neurons. RNA sequencing analysis shows that the adaptor protein, WDFY1 (WD repeat and FYVE domain-containing 1), acts downstream of Cdk5/p39 to regulate dendritic morphogenesis. While WDFY1 is elevated in p39-deficient neurons, suppressing its expression rescues the impaired dendritic arborization. Further phosphoproteomic analysis suggests that Cdk5/p39 mediates dendritic morphogenesis by modulating various downstream signaling pathways, including PI3K/Akt-, cAMP-, or small GTPase-mediated signaling transduction pathways, thereby regulating cytoskeletal organization, protein synthesis, and protein trafficking.


Subject(s)
Cyclin-Dependent Kinase 5/metabolism , Cytoskeletal Proteins/metabolism , Dendrites/metabolism , Lipid-Linked Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Differentiation/physiology , Cyclic AMP/metabolism , Cyclin-Dependent Kinase 5/genetics , Cytoskeletal Proteins/genetics , HEK293 Cells , Humans , Lipid-Linked Proteins/genetics , Mass Spectrometry , Mice , Monomeric GTP-Binding Proteins/genetics , Monomeric GTP-Binding Proteins/metabolism , Morphogenesis/genetics , Morphogenesis/physiology , Nervous System/cytology , Nervous System/metabolism , Neurons/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology
13.
Invert Neurosci ; 20(4): 19, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33090291

ABSTRACT

Sensory and motor systems in insects with hemimetabolous development must be ready to mediate adaptive behavior directly on hatching from the egg. For the desert locust S. gregaria, cholinergic transmission from antennal sensillae to olfactory or mechanosensory centers in the brain requires that choline acetyltransferase (ChAT) and the vesicular acetylcholine transporter (vAChT) already be present in sensory cells in the first instar. In this study, we used immunolabeling to demonstrate that ChAT and vAChT are both expressed in sensory cells from identifiable sensilla types in the immature antennal nervous system. We observed ChAT expression in dendrites, neurites and somata of putative basiconic-type sensillae at the first instar stage. We also detected vAChT in the sensory axons of these sensillae in a major antennal nerve tract. We then examined whether evidence for cholinergic transmission is present during embryogenesis. Immunolabeling confirms that vAChT is expressed in somata typical of campaniform sensillae, as well as in small sensory cell clusters typically associated with either a large basiconic or coeloconic sensilla, at 99% of embryogenesis. The vAChT is also expressed in the somata of these sensilla types in multiple antennal regions at 90% of embryogenesis, but not at earlier (70%) embryonic stages. Neuromodulators are known to appear late in embryogenesis in neurons of the locust central complex, and the cholinergic system of the antenna may also only reach maturity shortly before hatching.


Subject(s)
Choline O-Acetyltransferase/metabolism , Grasshoppers/cytology , Nervous System/cytology , Sensilla/cytology , Vesicular Acetylcholine Transport Proteins/metabolism , Animals
14.
Molecules ; 25(19)2020 Oct 06.
Article in English | MEDLINE | ID: mdl-33036163

ABSTRACT

Several biocompatible materials have been applied for managing soft tissue lesions; cerium oxide nanoparticles (CNPs, or nanoceria) are among the most promising candidates due to their outstanding properties, including antioxidant, anti-inflammatory, antibacterial, and angiogenic activities. Much attention should be paid to the physical properties of nanoceria, since most of its biological characteristics are directly determined by some of these relevant parameters, including the particle size and shape. Nanoceria, either in bare or functionalized forms, showed the excellent capability of accelerating the healing process of both acute and chronic wounds. The skin, heart, nervous system, and ophthalmic tissues are the main targets of nanoceria-based therapies, and the other soft tissues may also be evaluated in upcoming experimental studies. For the repair and regeneration of soft tissue damage and defects, nanoceria-incorporated film, hydrogel, and nanofibrous scaffolds have been proven to be highly suitable replacements with satisfactory outcomes. Still, some concerns have remained regarding the long-term effects of nanoceria administration for human tissues and organs, such as its clearance from the vital organs. Moreover, looking at the future, it seems necessary to design and develop three-dimensional (3D) printed scaffolds containing nanoceria for possible use in the concepts of personalized medicine.


Subject(s)
Cerium/chemistry , Tissue Engineering/methods , Antioxidants/chemistry , Humans , Nanoparticles/chemistry , Nervous System/cytology , Tissue Scaffolds/chemistry
15.
Nature ; 584(7822): 595-601, 2020 08.
Article in English | MEDLINE | ID: mdl-32814896

ABSTRACT

It is not known at present whether neuronal cell-type diversity-defined by cell-type-specific anatomical, biophysical, functional and molecular signatures-can be reduced to relatively simple molecular descriptors of neuronal identity1. Here we show, through examination of the expression of all of the conserved homeodomain proteins encoded by the Caenorhabditis elegans genome2, that the complete set of 118 neuron classes of C. elegans can be described individually by unique combinations of the expression of homeodomain proteins, thereby providing-to our knowledge-the simplest currently known descriptor of neuronal diversity. Computational and genetic loss-of-function analyses corroborate the notion that homeodomain proteins not only provide unique descriptors of neuron type, but also have a critical role in specifying neuronal identity. We speculate that the pervasive use of homeobox genes in defining unique neuronal identities reflects the evolutionary history of neuronal cell-type specification.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Gene Expression Regulation , Genes, Homeobox , Homeodomain Proteins/metabolism , Neurons/classification , Neurons/metabolism , Animals , Caenorhabditis elegans/metabolism , Genome/genetics , Homeodomain Proteins/genetics , Nervous System/cytology , Nervous System/metabolism , Neurons/cytology
16.
Curr Biol ; 30(18): 3617-3623.e3, 2020 09 21.
Article in English | MEDLINE | ID: mdl-32707065

ABSTRACT

Biological sex in animals is often considered a fixed, individual-level characteristic. However, not all sex-specific features are static: for example, C. elegans males (XO) can sometimes exhibit hermaphrodite (XX)-like feeding behavior [1, 2]. (C. elegans hermaphrodites are somatic females that transiently produce self-sperm.) Essentially all somatic sex differences in C. elegans are governed by the master regulator tra-1, whose activity is controlled by chromosomal sex and is necessary and sufficient to specify the hermaphrodite state [3]. One aspect of this state is high expression of the chemoreceptor odr-10. In hermaphrodites, high odr-10 expression promotes feeding, but in males, low odr-10 expression facilitates exploration [4]. However, males suppress this sex difference in two contexts: juvenile males exhibit high odr-10 expression and food deprivation activates odr-10 in adult males [4-6]. Remarkably, we find that both of these phenomena require tra-1. In juvenile (L3) males, tra-1 is expressed in numerous neurons; this expression diminishes as individuals mature into adulthood, a process that requires conserved regulators of sexual maturation. tra-1 remains expressed in a small number of neurons in adult males, where it likely has a permissive role in odr-10 activation. Thus, the neuronal functions of tra-1 are not limited to hermaphrodites; rather, tra-1 also acts in the male nervous system to transiently suppress a sexual dimorphism, developmentally and in response to nutritional stress. Our results show that the molecular and functional representation of sexual state in C. elegans is neither static nor homogeneous, challenging traditional notions about the nature of biological sex.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Nervous System/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Sex Characteristics , Transcription Factors/metabolism , Animals , Caenorhabditis elegans Proteins/genetics , DNA-Binding Proteins/genetics , Female , Male , Nervous System/cytology , Receptors, Cytoplasmic and Nuclear/genetics , Sex Factors , Transcription Factors/genetics
17.
Nat Commun ; 11(1): 3512, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32665545

ABSTRACT

Regional brain morphology has a complex genetic architecture, consisting of many common polymorphisms with small individual effects. This has proven challenging for genome-wide association studies (GWAS). Due to the distributed nature of genetic signal across brain regions, multivariate analysis of regional measures may enhance discovery of genetic variants. Current multivariate approaches to GWAS are ill-suited for complex, large-scale data of this kind. Here, we introduce the Multivariate Omnibus Statistical Test (MOSTest), with an efficient computational design enabling rapid and reliable inference, and apply it to 171 regional brain morphology measures from 26,502 UK Biobank participants. At the conventional genome-wide significance threshold of α = 5 × 10-8, MOSTest identifies 347 genomic loci associated with regional brain morphology, more than any previous study, improving upon the discovery of established GWAS approaches more than threefold. Our findings implicate more than 5% of all protein-coding genes and provide evidence for gene sets involved in neuron development and differentiation.


Subject(s)
Brain/metabolism , Genome-Wide Association Study/methods , Nervous System/metabolism , Female , Genetic Predisposition to Disease/genetics , Humans , Multivariate Analysis , Nervous System/cytology
18.
Nat Commun ; 11(1): 2123, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358558

ABSTRACT

Mammals differ in their regeneration potential after traumatic injury, which might be caused by species-specific regeneration programs. Here, we compared murine and human Schwann cell (SC) response to injury and developed an ex vivo injury model employing surgery-derived human sural nerves. Transcriptomic and lipid metabolism analysis of murine SCs following injury of sural nerves revealed down-regulation of lipogenic genes and regulator of lipid metabolism, including Pparg (peroxisome proliferator-activated receptor gamma) and S1P (sphingosine-1-phosphate). Human SCs failed to induce similar adaptations following ex vivo nerve injury. Pharmacological PPARg and S1P stimulation in mice resulted in up-regulation of lipid gene expression, suggesting a role in SCs switching towards a myelinating state. Altogether, our results suggest that murine SC switching towards a repair state is accompanied by transcriptome and lipidome adaptations, which are reduced in humans.


Subject(s)
Lipid Metabolism/physiology , Schwann Cells/cytology , Schwann Cells/metabolism , Animals , Female , Humans , Lysophospholipids/metabolism , Male , Mice , Myelin Sheath/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Nervous System/cytology , Nervous System/metabolism , Neuroglia/cytology , Neuroglia/metabolism , PPAR gamma/metabolism , Peripheral Nervous System/cytology , Peripheral Nervous System/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
19.
Nat Commun ; 11(1): 2138, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32358570

ABSTRACT

Signaling molecules that regulate neurodevelopmental processes in the early postnatal subventricular zone (SVZ) are critical for proper brain development yet remain poorly characterized. Here, we report that Endothelin-1 (ET-1), a molecular component of the postnatal SVZ, promotes radial glial cell maintenance and proliferation in an autocrine manner via Notch signaling. Loss of ET-1 signaling increases neurogenesis and reduces oligodendrocyte progenitor cell proliferation (OPC) in the developing SVZ, thereby altering cellular output of the stem cell niche. We also show that ET-1 is required for increased neural stem cell and OPC proliferation in the adult mouse SVZ following demyelination. Lastly, high levels of ET-1 in the SVZ of patients with Cathepsin A-related arteriopathy with strokes and leukoencephalopathy correlate with an increased number of SVZ OPCs, suggesting ET-1's role as a regulator of glial progenitor proliferation may be conserved in humans. ET-1 signaling therefore presents a potential new therapeutic target for promoting SVZ-mediated cellular repair.


Subject(s)
Endothelin-1/metabolism , Nervous System/cytology , Nervous System/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Stem Cell Niche/physiology , Animals , Cell Proliferation/physiology , Endothelin-1/genetics , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction/physiology , Stem Cell Niche/genetics
20.
J Biol Chem ; 295(18): 6120-6137, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32229587

ABSTRACT

The developing nervous system is remarkably sensitive to environmental signals, including disruptive toxins, such as polybrominated diphenyl ethers (PBDEs). PBDEs are an environmentally pervasive class of brominated flame retardants whose neurodevelopmental toxicity mechanisms remain largely unclear. Using dissociated cortical neurons from embryonic Rattus norvegicus, we found here that chronic exposure to 6-OH-BDE-47, one of the most prevalent hydroxylated PBDE metabolites, suppresses both spontaneous and evoked neuronal electrical activity. On the basis of our previous work on mitogen-activated protein kinase (MAPK)/extracellular signal-related kinase (ERK) (MEK) biology and our observation that 6-OH-BDE-47 is structurally similar to kinase inhibitors, we hypothesized that certain hydroxylated PBDEs mediate neurotoxicity, at least in part, by impairing the MEK-ERK axis of MAPK signal transduction. We tested this hypothesis on three experimental platforms: 1) in silico, where modeling ligand-protein docking suggested that 6-OH-BDE-47 is a promiscuous ATP-competitive kinase inhibitor; 2) in vitro in dissociated neurons, where 6-OH-BDE-47 and another specific hydroxylated BDE metabolite similarly impaired phosphorylation of MEK/ERK1/2 and activity-induced transcription of a neuronal immediate early gene; and 3) in vivo in Drosophila melanogaster, where developmental exposures to 6-OH-BDE-47 and a MAPK inhibitor resulted in offspring displaying similarly increased frequency of mushroom-body ß-lobe midline crossing, a metric of axonal guidance. Taken together, our results support that certain ortho-hydroxylated PBDE metabolites are promiscuous kinase inhibitors and can cause disruptions of critical neurodevelopmental processes, including neuronal electrical activity, pre-synaptic functions, MEK-ERK signaling, and axonal guidance.


Subject(s)
Ethers/chemistry , Ethers/pharmacology , Halogenation , Nervous System/growth & development , Neurons/cytology , Neurons/drug effects , Signal Transduction/drug effects , Animals , Drosophila melanogaster , Hydroxylation , Intracellular Space/drug effects , Intracellular Space/metabolism , Nervous System/cytology , Nervous System/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...