Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.845
Filter
1.
Mol Brain ; 17(1): 24, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762724

ABSTRACT

CD11c-positive (CD11c+) microglia have attracted considerable attention because of their potential implications in central nervous system (CNS) development, homeostasis, and disease. However, the spatiotemporal dynamics of the proportion of CD11c+ microglia in individual CNS regions are poorly understood. Here, we investigated the proportion of CD11c+ microglia in six CNS regions (forebrain, olfactory bulb, diencephalon/midbrain, cerebellum, pons/medulla, and spinal cord) from the developmental to adult stages by flow cytometry and immunohistochemical analyses using a CD11c reporter transgenic mouse line, Itgax-Venus. We found that the proportion of CD11c+ microglia in total microglia varied between CNS regions during postnatal development. Specifically, the proportion was high in the olfactory bulb and cerebellum at postnatal day P(4) and P7, respectively, and approximately half of the total microglia were CD11c+. The proportion declined sharply in all regions to P14, and the low percentage persisted over P56. In the spinal cord, the proportion of CD11c+ microglia was also high at P4 and declined to P14, but increased again at P21 and thereafter. Interestingly, the distribution pattern of CD11c+ microglia in the spinal cord markedly changed from gray matter at P4 to white matter at P21. Collectively, our findings reveal the differences in the spatiotemporal dynamics of the proportion of CD11c+ microglia among CNS regions from early development to adult stages in normal mice. These findings improve our understanding of the nature of microglial heterogeneity and its dynamics in the CNS.


Subject(s)
Brain , Mice, Transgenic , Microglia , Spinal Cord , Animals , Microglia/metabolism , Microglia/cytology , Spinal Cord/growth & development , Brain/growth & development , Brain/cytology , Spatio-Temporal Analysis , Aging , CD11c Antigen/metabolism , Mice, Inbred C57BL , Mice , Animals, Newborn
2.
Genes (Basel) ; 15(4)2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38674338

ABSTRACT

Microribonucleic acids (miRNAs) comprising miR-23a/b clusters, specifically miR-23a and miR-27a, are recognized for their divergent roles in myelination within the central nervous system. However, cluster-specific miRNA functions remain controversial as miRNAs within the same cluster have been suggested to function complementarily. This study aims to clarify the role of miR-23a/b clusters in myelination using mice with a miR-23a/b cluster deletion (KO mice), specifically in myelin expressing proteolipid protein (PLP). Inducible conditional KO mice were generated by crossing miR-23a/b clusterflox/flox mice with PlpCre-ERT2 mice; the offspring were injected with tamoxifen at 10 days or 10 weeks of age to induce a myelin-specific miR-23a/b cluster deletion. Evaluation was performed at 10 weeks or 12 months of age and compared with control mice that were not treated with tamoxifen. KO mice exhibit impaired motor function and hypoplastic myelin sheaths in the brain and spinal cord at 10 weeks and 12 months of age. Simultaneously, significant decreases in myelin basic protein (MBP) and PLP expression occur in KO mice. The percentages of oligodendrocyte precursors and mature oligodendrocytes are consistent between the KO and control mice. However, the proportion of oligodendrocytes expressing MBP is significantly lower in KO mice. Moreover, changes in protein expression occur in KO mice, with increased leucine zipper-like transcriptional regulator 1 expression, decreased R-RAS expression, and decreased phosphorylation of extracellular signal-regulated kinases. These findings highlight the significant influence of miR-23a/b clusters on myelination during postnatal growth and aging.


Subject(s)
Aging , MicroRNAs , Myelin Sheath , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mice , Myelin Sheath/metabolism , Myelin Sheath/genetics , Aging/genetics , Central Nervous System/metabolism , Central Nervous System/growth & development , Mice, Knockout , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Spinal Cord/metabolism , Spinal Cord/growth & development , Myelin Basic Protein/metabolism , Myelin Basic Protein/genetics , Oligodendroglia/metabolism , Brain/metabolism , Brain/growth & development
3.
Int J Mol Sci ; 23(24)2022 Dec 16.
Article in English | MEDLINE | ID: mdl-36555713

ABSTRACT

Although rodents have been widely used for experimental models of spinal cord diseases, the details of the growth curves of their spinal canal and spinal cord, as well as the molecular mechanism of the growth of adult rat spinal cords remain unavailable. They are particularly important when conducting the experiments of cervical spondylotic myelopathy (CSM), since the disease condition depends on the size of the spinal canal and the spinal cord. Thus, the purposes of the present study were to obtain accurate growth curves for the spinal canal and spinal cord in rats; to define the appropriate age in weeks for their use as a CSM model; and to propose a molecular mechanism of the growth of the adult spinal cord in rats. CT myelography was performed on Lewis rats from 4 weeks to 40 weeks of age. The vertical growth of the spinal canal at C5 reached a plateau after 20 and 12 weeks, and at T8 after 20 and 16 weeks, in males and females, respectively. The vertical growth of the C5 and T8 spinal cord reached a plateau after 24 weeks in both sexes. The vertical space available for the cord (SAC) of C5 and T8 did not significantly change after 8 weeks in either sex. Western blot analyses showed that VEGFA, FGF2, and BDNF were highly expressed in the cervical spinal cords of 4-week-old rats, and that the expression of these growth factors declined as rats grew. These findings indicate that the spinal canal and the spinal cord in rats continue to grow even after sexual maturation and that rats need to be at least 8 weeks of age for use in experimental models of CSM. The present study, in conjunction with recent evidence, proposes the hypothetical model that the growth of rat spinal cord after the postnatal period is mediated at least in part by differentiation of neural progenitor cells and that their differentiation potency is maintained by VEGFA, FGF2, and BDNF.


Subject(s)
Sexual Maturation , Spinal Canal , Spinal Cord , Animals , Female , Male , Rats , Brain-Derived Neurotrophic Factor/genetics , Fibroblast Growth Factor 2 , Magnetic Resonance Imaging , Rats, Inbred Lew , Spinal Canal/growth & development , Spinal Cord/growth & development , Spinal Cord Compression , Spinal Cord Diseases
4.
Eur Rev Med Pharmacol Sci ; 26(17): 6273-6282, 2022 09.
Article in English | MEDLINE | ID: mdl-36111928

ABSTRACT

OBJECTIVE: L1  cell adhesion molecule (L1CAM) is a glycoprotein characterized by three components: an extracellular region, a transmembrane segment, and a cytoplasmic tail. L1CAM is expressed in multiple human cells, including neurons. The neural cell adhesion molecule L1 has been implicated in a variety of neurologic processes, including neuritogenesis and cerebellar cell migration. The presence of L1CAM on the surface of nerve cells allows the adhesion of neurons among them. Furthermore, when it is bound to itself or to other proteins, L1-CAM induces signals inside the cell. The aim of this work was to study L1CAM expression in the human spinal cord during development, at different gestational ages, through immunohistochemistry. MATERIALS AND METHODS: Immunohistochemical analysis for L1CAM was performed in five human spinal cord samples, including three embryos and two fetuses of different gestational ages, ranging from 8 to 12 weeks. RESULTS: L1CAM expression was detected in all 5 spinal cords examined in this study. The adhesion molecule was found in the vast majority of cells. The highest levels of immunoreactivity for L1CAM were detected at the periphery of the developing organs, in the spinal cord zones occupied by sensory and motor fibers. In the alar and basal columns, immunoreactivity for L1CAM was characterized by a reticular pattern, being mainly expressed in axons. Strong reactivity of L1CAM was also found in extracellular vesicles. This extracellular localization might indicate the ability of L1CAM to mediate the transduction of extracellular signals that support axon outgrowth. CONCLUSIONS: The high reactivity of L1cam in the axons of developing neurons in the fetal spinal cord confirms previous studies on the ability of L1CAM to promote axon sprouting and branching in the developing nervous system. In this work, a new actor is reported to have a role in the complex field of human spinal cord development: L1CAM, whose expression is highly found in the developing neuronal and glial precursors.


Subject(s)
Extracellular Vesicles , Neural Cell Adhesion Molecule L1 , Spinal Cord , Axons/metabolism , Embryo, Mammalian , Extracellular Vesicles/metabolism , Humans , Infant , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Spinal Cord/embryology , Spinal Cord/growth & development , Spinal Cord/metabolism
5.
Nat Commun ; 13(1): 958, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177643

ABSTRACT

How a single neuronal population diversifies into subtypes with distinct synaptic targets is a fundamental topic in neuroscience whose underlying mechanisms are unclear. Here, we show that the histone H3-lysine 27 demethylase Kdm6b regulates the diversification of motor neurons to distinct subtypes innervating different muscle targets during spinal cord development. In mouse embryonic motor neurons, Kdm6b promotes the medial motor column (MMC) and hypaxial motor column (HMC) fates while inhibiting the lateral motor column (LMC) and preganglionic motor column (PGC) identities. Our single-cell RNA-sequencing analyses reveal the heterogeneity of PGC, LMC, and MMC motor neurons. Further, our single-cell RNA-sequencing data, combined with mouse model studies, demonstrates that Kdm6b acquires cell fate specificity together with the transcription factor complex Isl1-Lhx3. Our study provides mechanistic insight into the gene regulatory network regulating neuronal cell-type diversification and defines a regulatory role of Kdm6b in the generation of motor neuron subtypes in the mouse spinal cord.


Subject(s)
Jumonji Domain-Containing Histone Demethylases/metabolism , Motor Neurons/physiology , Neurogenesis/genetics , Spinal Cord/growth & development , Animals , Cell Differentiation/genetics , DNA Demethylation , Embryo, Mammalian , Female , Gene Knockout Techniques , HEK293 Cells , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , LIM-Homeodomain Proteins/metabolism , Mice , Mice, Transgenic , RNA-Seq , Single-Cell Analysis , Spinal Cord/cytology , Transcription Factors/metabolism
6.
J Neurosurg Pediatr ; 29(1): 100-105, 2022 Jan 01.
Article in English | MEDLINE | ID: mdl-34653991

ABSTRACT

OBJECTIVE: This study aimed to explore the migration process of the conus medullaris (CM) in early infancy using infant MRI and to evaluate the application of MRI for locating the infant CM level. METHODS: The authors retrospectively analyzed the CM level on the lumbosacral MR images of 26 term infants aged < 3 months who were classified into three groups according to age. The authors numbered the CM level in each patient and analyzed the range and average of the CM level of the cohort. The authors studied the linear correlation between CM level and postnatal days with linear regression analysis, 1-way ANOVA, and the least significant difference test. RESULTS: The CM level ranged from the superior border of the L1 vertebra to the top third of the L3 vertebra. About 96.2% of infants had CM higher than the superior border of the L3 vertebra. On average, CM was located between the L1-2 intervertebral disc and the inferior border of the L2 vertebra (mean ± SD score 1.64 ± 1.14). The three groups had no significant statistical difference in CM level (F = 1.071 and p = 0.359; groups 1 and 2, p = 0.408; groups 1 and 3, p = 0.170; groups 2 and 3, p = 0.755). CM level had no linear regression correlation with postnatal days within the first month (r2 = 0.061, F = 0.654, p = 0.438) or within the first 3 months (r2 = 0.002, F = 0.056, p = 0.816). CONCLUSIONS: The CM level reaches the normal adult level by birth in term infants and does not ascend during childhood. On average, the CM was between the L1-2 intervertebral disc and the inferior border of the L2 vertebra in term infants. Considering the possibility of physiologically low-lying CM, the authors agree that normal CM is located above the L3 level in term infants and CM at the L3 level could be equivocal and should be investigated with other clinical data. The study data suggest that MRI is an accurate and valuable method for determining the CM level in term infants.


Subject(s)
Spinal Cord/anatomy & histology , Spinal Cord/growth & development , Female , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Reference Values , Retrospective Studies
7.
Exp Neurol ; 348: 113944, 2022 02.
Article in English | MEDLINE | ID: mdl-34896115

ABSTRACT

Fibroblast growth factor binding protein 3 (Fgfbp3) have been known to be crucial for the process of neural proliferation, differentiation, migration, and adhesion. However, the specific role and the molecular mechanisms of fgfbp3 in regulating the development of motor neurons remain unclear. In this study, we have investigated the function of fgfbp3 in morphogenesis and regeneration of motor neuron in zebrafish. Firstly, we found that fgfbp3 was localized in the motor neurons and loss of fgfbp3 caused the significant decrease of the length and branching number of the motor neuron axons, which could be partially rescued by fgfbp3 mRNA injection. Moreover, the fgfbp3 knockdown (KD) embryos demonstrated similar defects of motor neurons as identified in fgfbp3 knockout (KO) embryos. Furthermore, we revealed that the locomotion and startle response of fgfbp3 KO embryos were significantly restricted, which were partially rescued by the fgfbp3 overexpression. In addition, fgfbp3 KO remarkably compromised axonal regeneration of motor neurons after injury. Lastly, the malformation of motor neurons in fgfbp3 KO embryos was rescued by overexpressing drd1b or neurod6a, respectively, which were screened by transcriptome sequencing. Taken together, our results provide strong cellular and molecular evidence that fgfbp3 is crucial for the axonal morphogenesis and regeneration of motor neurons in zebrafish.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/genetics , Motor Neurons/metabolism , Nerve Regeneration/physiology , Neurogenesis/physiology , Spinal Cord/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Carrier Proteins/antagonists & inhibitors , Gene Knockout Techniques/methods , Reflex, Startle/physiology , Spinal Cord/growth & development , Swimming/physiology , Zebrafish
8.
Biochem Biophys Res Commun ; 587: 85-91, 2022 01 08.
Article in English | MEDLINE | ID: mdl-34864550

ABSTRACT

One of the major challenges of modern neurobiology concerns the inability of the adult mammalian central nervous system (CNS) to regenerate and repair itself after injury. It is still unclear why the ability to regenerate CNS is lost during evolution and development and why it becomes very limited in adult mammals. A convenient model to study cellular and molecular basis of this loss is neonatal opossum (Monodelphis domestica). Opossums are marsupials that are born very immature with the unique possibility to successfully regenerate postnatal spinal cord after injury in the first two weeks of their life, after which this ability abbruptly stops. Using comparative proteomic approach we identified the proteins that are differentially distributed in opossum spinal tissue that can and cannot regenerate after injury, among which stand out the proteins related to neurodegenerative diseases (NDD), such as Huntington, Parkinson and Alzheimer's disease, previously detected by comparative transcriptomics on the analog tissue. The different distribution of the selected proteins detected by comparative proteomics was further confirmed by Western blot (WB), and the changes in the expression of related genes were analysed by quantitative reverse transcription PCR (qRT-PCR). Furthermore, we explored the cellular localization of the selected proteins using immunofluorescent microscopy. To our knowledge, this is the first report on proteins differentially present in developing, non-injured mammalian spinal cord tissue with different regenerative capacities. The results of this study indicate that the proteins known to have an important role in the pathophysiology of neurodegeneration in aged CNS, could also have an important phyisological role during CNS postnatal development and in neuroregeneration process.


Subject(s)
Gene Expression Regulation, Developmental , Monodelphis/genetics , Nerve Regeneration/genetics , Nerve Tissue Proteins/genetics , Spinal Cord/metabolism , Transcriptome , Animals , Animals, Newborn , Female , Gene Expression Profiling , Gene Ontology , Male , Molecular Sequence Annotation , Monodelphis/growth & development , Monodelphis/metabolism , Nerve Tissue Proteins/classification , Nerve Tissue Proteins/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Proteomics/methods , Spinal Cord/growth & development , Spinal Cord Injuries/genetics , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/pathology , Time Factors
9.
J Chem Neuroanat ; 118: 102041, 2021 12.
Article in English | MEDLINE | ID: mdl-34774721

ABSTRACT

Spinal cord injury in fish produces fibrous scar, but spontaneous axonal regeneration beyond the scar sometimes occurs. A previous study revealed that regenerating axons enter the scar through tubular structures with laminin, and that an increased number of axons within the tube is coincident with enlargement of the tube diameter and reduction of the fibrous scar area. The present study investigated the expression of matrix metalloproteinases (MMPs) that might play a role in the degradation of the extracellular matrix in fibrous scar tissue and in the remodeling of tubular structures. Spinal hemisection produced fibrous scar tissue in the lesion center, surrounded by nervous tissue. Two weeks after spinal lesioning, MMP-9 was expressed in some regenerating axons in the fibrous scar tissue. MMP-14 was expressed in the regenerating axons, as well as in glial processes in the fibrous scar tissue. MMP-2 was suggested to be expressed in mast cells in the fibrous scar. The mast cells were in contact with fibroblasts, and in close proximity to the basement membrane of tubular structures surrounding the regenerating axons. The present findings suggest that several MMPs are involved in axon regenerating processes following spinal cord injury in goldfish. MMP-9 and MMP-14 expressed in the regenerating axons might degrade extracellular matrix and support axonal growth deep into the fibrous scar tissue. MMP-14 expressed in glial cells and MMP-2 expressed in mast cells might also provide a beneficial environment for axonal regeneration, leading to successful motor recovery.


Subject(s)
Axons/physiology , Goldfish/physiology , Matrix Metalloproteinases/biosynthesis , Nerve Regeneration/physiology , Spinal Cord/growth & development , Spinal Cord/metabolism , Animals , Basement Membrane/metabolism , Cicatrix/pathology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts , Mast Cells , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Recovery of Function , Spinal Cord Injuries/metabolism
10.
Mol Brain ; 14(1): 154, 2021 10 06.
Article in English | MEDLINE | ID: mdl-34615523

ABSTRACT

Studies on the development of central nervous system (CNS) primarily rely on the use of specific molecular markers for different types of neural cells. S100B is widely being used as a specific marker for astrocytes in the CNS. However, the specificity of its expression in astrocyte lineage has not been systematically investigated and thus has remained a lingering issue. In this study, we provide several lines of molecular and genetic evidences that S100B is expressed in both protoplasmic astrocytes and myelinating oligodendrocytes. In the developing spinal cord, S100B is first expressed in the ventral neuroepithelial cells, and later in ALDH1L1+/GS+ astrocytes in the gray matter. Meanwhile, nearly all the S100B+ cells in the white matter are SOX10+/MYRF+ oligodendrocytes. Consistent with this observation, S100B expression is selectively lost in the white matter in Olig2-null mutants in which oligodendrocyte progenitor cells (OPCs) are not produced, and dramatically reduced in Myrf-conditional knockout mutants in which OPCs fail to differentiate. Similar expression patterns of S100B are observed in the developing forebrain. Based on these molecular and genetic studies, we conclude that S100B is not a specific marker for astrocyte lineage; instead, it marks protoplasmic astrocytes in the gray matter and differentiating oligodendrocytes.


Subject(s)
Astrocytes/metabolism , Gray Matter/cytology , Oligodendroglia/metabolism , Prosencephalon/growth & development , S100 Calcium Binding Protein beta Subunit/biosynthesis , Spinal Cord/growth & development , Animals , Biomarkers , Brain/growth & development , Cell Lineage , Cytoplasm/metabolism , Glial Fibrillary Acidic Protein/analysis , Glutamate-Ammonia Ligase/analysis , Mice , Myelin Sheath/physiology , Neurons/metabolism , Organ Specificity , Oxidoreductases Acting on CH-NH Group Donors/analysis , Prosencephalon/cytology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , S100 Calcium Binding Protein beta Subunit/genetics , SOXE Transcription Factors/analysis , Spinal Cord/cytology
11.
Nat Commun ; 12(1): 5722, 2021 09 29.
Article in English | MEDLINE | ID: mdl-34588430

ABSTRACT

Single-cell RNA sequencing data can unveil the molecular diversity of cell types. Cell type atlases of the mouse spinal cord have been published in recent years but have not been integrated together. Here, we generate an atlas of spinal cell types based on single-cell transcriptomic data, unifying the available datasets into a common reference framework. We report a hierarchical structure of postnatal cell type relationships, with location providing the highest level of organization, then neurotransmitter status, family, and finally, dozens of refined populations. We validate a combinatorial marker code for each neuronal cell type and map their spatial distributions in the adult spinal cord. We also show complex lineage relationships among postnatal cell types. Additionally, we develop an open-source cell type classifier, SeqSeek, to facilitate the standardization of cell type identification. This work provides an integrated view of spinal cell types, their gene expression signatures, and their molecular organization.


Subject(s)
Neurons/classification , Spinal Cord/cytology , Transcriptome , Animals , Atlases as Topic , Cell Nucleus/genetics , Datasets as Topic , Mice , Neurons/cytology , RNA-Seq , Single-Cell Analysis , Spatial Analysis , Spinal Cord/growth & development
12.
Nat Commun ; 12(1): 4857, 2021 08 11.
Article in English | MEDLINE | ID: mdl-34381039

ABSTRACT

Physical exercise stimulates adult neurogenesis, yet the underlying mechanisms remain poorly understood. A fundamental component of the innate neuroregenerative capacity of zebrafish is the proliferative and neurogenic ability of the neural stem/progenitor cells. Here, we show that in the intact spinal cord, this plasticity response can be activated by physical exercise by demonstrating that the cholinergic neurotransmission from spinal locomotor neurons activates spinal neural stem/progenitor cells, leading to neurogenesis in the adult zebrafish. We also show that GABA acts in a non-synaptic fashion to maintain neural stem/progenitor cell quiescence in the spinal cord and that training-induced activation of neurogenesis requires a reduction of GABAA receptors. Furthermore, both pharmacological stimulation of cholinergic receptors, as well as interference with GABAergic signaling, promote functional recovery after spinal cord injury. Our findings provide a model for locomotor networks' activity-dependent neurogenesis during homeostasis and regeneration in the adult zebrafish spinal cord.


Subject(s)
Locomotion , Neuroglia/metabolism , Neurons/metabolism , Spinal Cord/growth & development , Animals , Interneurons/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurogenesis , Physical Conditioning, Animal , Receptors, Cholinergic/metabolism , Receptors, GABA-A/metabolism , Recovery of Function , Spinal Cord/cytology , Spinal Cord/physiology , Synaptic Transmission , Zebrafish , gamma-Aminobutyric Acid/metabolism
13.
PLoS One ; 16(8): e0256207, 2021.
Article in English | MEDLINE | ID: mdl-34403440

ABSTRACT

Thyroid hormones are messengers that bind to specific nuclear receptors and regulate a wide range of physiological processes in the early stages of vertebrate embryonic development, including neurodevelopment and myelogenesis. We here tested the effects of reduced T3 availability upon the myelination process by treating zebrafish embryos with low concentrations of iopanoic acid (IOP) to block T4 to T3 conversion. Black Gold II staining showed that T3 deficiency reduced the myelin density in the forebrain, midbrain, hindbrain and the spinal cord at 3 and 7 dpf. These observations were confirmed in 3 dpf mbp:egfp transgenic zebrafish, showing that the administration of IOP reduced the fluorescent signal in the brain. T3 rescue treatment restored brain myelination and reversed the changes in myelin-related gene expression induced by IOP exposure. NG2 immunostaining revealed that T3 deficiency reduced the amount of oligodendrocyte precursor cells in 3 dpf IOP-treated larvae. Altogether, the present results show that inhibition of T4 to T3 conversion results in hypomyelination, suggesting that THs are part of the key signaling molecules that control the timing of oligodendrocyte differentiation and myelin synthesis from very early stages of brain development.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Larva/genetics , Myelin Sheath/genetics , Thyroxine/deficiency , Triiodothyronine/deficiency , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Antigens/genetics , Antigens/metabolism , Embryo, Nonmammalian , Embryonic Development , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Iopanoic Acid/pharmacology , Larva/cytology , Larva/drug effects , Larva/growth & development , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/growth & development , Mesencephalon/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Oligodendrocyte Transcription Factor 2/genetics , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/growth & development , Prosencephalon/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Rhombencephalon/cytology , Rhombencephalon/drug effects , Rhombencephalon/growth & development , Rhombencephalon/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/growth & development , Spinal Cord/metabolism , Triiodothyronine/pharmacology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Neurochem Res ; 46(11): 2776-2782, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34228233

ABSTRACT

Neurons and glial cells in the central nervous system (CNS) are generated from neuroepithelial cells in the ventricular zone that surrounds the embryonic neural tube. The proliferation and distinct differentiation of neural precursors occurs at certain stages and are regulated by a series of transcription factors leading to the generation of neuronal and glial cell subtypes. In this manuscript, we review the effects of the Olig family, namely, members Olig1, Olig2 and Olig3, on the distinct differentiation of glial and neuronal cells in the developing spinal cord and injured neural tissue.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Nerve Regeneration/physiology , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Spinal Cord/metabolism , Animals , Astrocytes/metabolism , Humans , Neuroglia/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Spinal Cord/embryology , Spinal Cord/growth & development
15.
Prenat Diagn ; 41(8): 989-997, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34128545

ABSTRACT

OBJECTIVE: Before evaluating spinal pathology, it is essential to have knowledge of the normal spinal development at different gestational ages. This study aims to characterize normal spinal growth in human fetuses during the second and third trimesters. METHODS: Postmortem 3.0 T magnetic resonance imaging (MRI) was performed on 55 fetuses at 17-42 gestational weeks by using three-dimensional T2-weighted sequences. Morphological changes and quantitative measurements of the fetal spine were assessed. The correlation between centrum ossification center volume (COCV) and gestational age was investigated. RESULTS: The cervical, thoracic, and lumbar COCVs showed a positive relationship with gestational age (p < 0.05). No gender differences were found in the volumetric development of the cervical, thoracic, and lumbar centrum ossification centers (COCs). The average volumetric growth rate per COC was larger in the lumbar spine than in the cervical and thoracic spine. The L1-L5 COCVs also showed a linear positive relationship with gestational age. CONSULTS: Postmortem 3.0 T MRI clearly demonstrated spinal changes in external contour and internal structure with gestational age. These findings expand our understanding of the early growth pattern of the human spine and could be further used to assess the developmental conditions of the fetal spine.


Subject(s)
Fetal Development/physiology , Spinal Cord/growth & development , Adult , China , Female , Gestational Age , Humans , Magnetic Resonance Imaging/methods , Magnetic Resonance Imaging/statistics & numerical data , Male , Pregnancy , Spinal Cord/physiopathology
16.
Dev Cell ; 56(11): 1617-1630.e6, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34033756

ABSTRACT

Central nervous system injury re-initiates neurogenesis in anamniotes (amphibians and fishes), but not in mammals. Activation of the innate immune system promotes regenerative neurogenesis, but it is fundamentally unknown whether this is indirect through the activation of known developmental signaling pathways or whether immune cells directly signal to progenitor cells using mechanisms that are unique to regeneration. Using single-cell RNA-seq of progenitor cells and macrophages, as well as cell-type-specific manipulations, we provide evidence for a direct signaling axis from specific lesion-activated macrophages to spinal progenitor cells to promote regenerative neurogenesis in zebrafish. Mechanistically, TNFa from pro-regenerative macrophages induces Tnfrsf1a-mediated AP-1 activity in progenitors to increase regeneration-promoting expression of hdac1 and neurogenesis. This establishes the principle that macrophages directly communicate to spinal progenitor cells via non-developmental signals after injury, providing potential targets for future interventions in the regeneration-deficient spinal cord of mammals.


Subject(s)
Histone Deacetylase 1/genetics , Neurogenesis/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Regeneration/genetics , Spinal Cord/growth & development , Zebrafish Proteins/genetics , Animals , Cell Lineage/genetics , Gene Expression Regulation, Developmental/genetics , Macrophages/cytology , Macrophages/metabolism , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis , Spinal Cord/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factor AP-1/genetics , Zebrafish/genetics
17.
Mol Syst Biol ; 17(5): e9902, 2021 05.
Article in English | MEDLINE | ID: mdl-34031978

ABSTRACT

Ultradian oscillations of HES Transcription Factors (TFs) at the single-cell level enable cell state transitions. However, the tissue-level organisation of HES5 dynamics in neurogenesis is unknown. Here, we analyse the expression of HES5 ex vivo in the developing mouse ventral spinal cord and identify microclusters of 4-6 cells with positively correlated HES5 level and ultradian dynamics. These microclusters are spatially periodic along the dorsoventral axis and temporally dynamic, alternating between high and low expression with a supra-ultradian persistence time. We show that Notch signalling is required for temporal dynamics but not the spatial periodicity of HES5. Few Neurogenin 2 cells are observed per cluster, irrespective of high or low state, suggesting that the microcluster organisation of HES5 enables the stable selection of differentiating cells. Computational modelling predicts that different cell coupling strengths underlie the HES5 spatial patterns and rate of differentiation, which is consistent with comparison between the motoneuron and interneuron progenitor domains. Our work shows a previously unrecognised spatiotemporal organisation of neurogenesis, emergent at the tissue level from the synthesis of single-cell dynamics.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , Single-Cell Analysis/methods , Spinal Cord/growth & development , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Communication , Computational Biology , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Mice , Neurogenesis , Receptors, Notch/metabolism , Repressor Proteins/genetics , Signal Transduction , Spatio-Temporal Analysis , Spinal Cord/metabolism , Ultradian Rhythm
18.
PLoS Genet ; 17(4): e1009515, 2021 04.
Article in English | MEDLINE | ID: mdl-33914736

ABSTRACT

Zebrafish exhibit robust regeneration following spinal cord injury, promoted by macrophages that control post-injury inflammation. However, the mechanistic basis of how macrophages regulate regeneration is poorly understood. To address this gap in understanding, we conducted a rapid in vivo phenotypic screen for macrophage-related genes that promote regeneration after spinal injury. We used acute injection of synthetic RNA Oligo CRISPR guide RNAs (sCrRNAs) that were pre-screened for high activity in vivo. Pre-screening of over 350 sCrRNAs allowed us to rapidly identify highly active sCrRNAs (up to half, abbreviated as haCRs) and to effectively target 30 potentially macrophage-related genes. Disruption of 10 of these genes impaired axonal regeneration following spinal cord injury. We selected 5 genes for further analysis and generated stable mutants using haCRs. Four of these mutants (tgfb1a, tgfb3, tnfa, sparc) retained the acute haCR phenotype, validating the approach. Mechanistically, tgfb1a haCR-injected and stable mutant zebrafish fail to resolve post-injury inflammation, indicated by prolonged presence of neutrophils and increased levels of il1b expression. Inhibition of Il-1ß rescues the impaired axon regeneration in the tgfb1a mutant. Hence, our rapid and scalable screening approach has identified functional regulators of spinal cord regeneration, but can be applied to any biological function of interest.


Subject(s)
RNA, Guide, Kinetoplastida/genetics , Regeneration/genetics , Spinal Cord Regeneration/genetics , Transforming Growth Factor beta1/genetics , Zebrafish Proteins/genetics , Animals , Axons/metabolism , Axons/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Disease Models, Animal , Macrophages/metabolism , Osteonectin/genetics , Recovery of Function/genetics , Spinal Cord/growth & development , Spinal Cord/pathology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/therapy , Spinal Cord Regeneration/physiology , Transforming Growth Factor beta3/genetics , Zebrafish/genetics , Zebrafish/growth & development
19.
Development ; 148(6)2021 03 23.
Article in English | MEDLINE | ID: mdl-33658223

ABSTRACT

The anteroposterior axial identity of motor neurons (MNs) determines their functionality and vulnerability to neurodegeneration. Thus, it is a crucial parameter in the design of strategies aiming to produce MNs from human pluripotent stem cells (hPSCs) for regenerative medicine/disease modelling applications. However, the in vitro generation of posterior MNs corresponding to the thoracic/lumbosacral spinal cord has been challenging. Although the induction of cells resembling neuromesodermal progenitors (NMPs), the bona fide precursors of the spinal cord, offers a promising solution, the progressive specification of posterior MNs from these cells is not well defined. Here, we determine the signals guiding the transition of human NMP-like cells toward thoracic ventral spinal cord neurectoderm. We show that combined WNT-FGF activities drive a posterior dorsal pre-/early neural state, whereas suppression of TGFß-BMP signalling pathways promotes a ventral identity and neural commitment. Based on these results, we define an optimised protocol for the generation of thoracic MNs that can efficiently integrate within the neural tube of chick embryos. We expect that our findings will facilitate the comparison of hPSC-derived spinal cord cells of distinct axial identities.


Subject(s)
Cell Differentiation/genetics , Mesoderm/growth & development , Neural Stem Cells/metabolism , Spinal Cord/growth & development , Animals , Body Patterning/genetics , Bone Morphogenetic Proteins/genetics , Cell Lineage/genetics , Chick Embryo , Fibroblast Growth Factors/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Mesoderm/metabolism , Motor Neurons/metabolism , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Signal Transduction/genetics , Spinal Cord/metabolism , Transforming Growth Factor beta/genetics , Wnt Proteins/genetics
20.
J Neurosci ; 41(17): 3808-3821, 2021 04 28.
Article in English | MEDLINE | ID: mdl-33727334

ABSTRACT

To assemble the functional circuits of the nervous system, the neuronal axonal growth cones must be precisely guided to their proper targets, which can be achieved through cell-surface guidance receptor activation by ligand binding in the periphery. We investigated the function of paxillin, a focal adhesion protein, as an essential growth cone guidance intermediary in the context of spinal lateral motor column (LMC) motor axon trajectory selection in the limb mesenchyme. Using in situ mRNA detection, we first show paxillin expression in LMC neurons of chick and mouse embryos at the time of spinal motor axon extension into the limb. Paxillin loss-of-function and gain-of-function using in ovo electroporation in chick LMC neurons, of either sex, perturbed LMC axon trajectory selection, demonstrating an essential role of paxillin in motor axon guidance. In addition, a neuron-specific paxillin deletion in mice led to LMC axon trajectory selection errors. We also show that knocking down paxillin attenuates the growth preference of LMC neurites against ephrins in vitro, and erythropoietin-producing human hepatocellular (Eph)-mediated retargeting of LMC axons in vivo, suggesting paxillin involvement in Eph-mediated LMC motor axon guidance. Finally, both paxillin knockdown and ectopic expression of a nonphosphorylable paxillin mutant attenuated the retargeting of LMC axons caused by Src overexpression, implicating paxillin as a Src target in Eph signal relay in this context. In summary, our findings demonstrate that paxillin is required for motor axon guidance and suggest its essential role in the ephrin-Eph signaling pathway resulting in motor axon trajectory selection.SIGNIFICANCE STATEMENT During the development of neural circuits, precise connections need to be established among neurons or between neurons and their muscle targets. A protein family found in neurons, Eph, is essential at different stages of neural circuit formation, including nerve outgrowth and pathfinding, and is proposed to mediate the onset and progression of several neurodegenerative diseases, such as Alzheimer's disease. To investigate how Ephs relay their signals to mediate nerve growth, we investigated the function of a molecule called paxillin and found it important for the development of spinal nerve growth toward their muscle targets, suggesting its role as an effector of Eph signals. Our work could thus provide new information on how neuromuscular connectivity is properly established during embryonic development.


Subject(s)
Axons/physiology , Paxillin/physiology , Spinal Cord/growth & development , Animals , Axon Guidance/physiology , Chick Embryo , Electroporation , Ephrins/physiology , Female , Gene Knockdown Techniques , Genes, src/genetics , Humans , Male , Mice , MicroRNAs/genetics , Motor Neurons/physiology , Mutation/genetics , Neurites/physiology , Spinal Cord/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...