Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 11.147
Filtrer
1.
Cell Mol Life Sci ; 81(1): 286, 2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-38970652

RÉSUMÉ

Paralog factors are considered to ensure the robustness of biological processes by providing redundant activity in cells where they are co-expressed. However, the specific contribution of each factor is frequently underestimated. In the developing spinal cord, multiple families of transcription factors successively contribute to differentiate an initially homogenous population of neural progenitors into a myriad of neuronal subsets with distinct molecular, morphological, and functional characteristics. The LIM-homeodomain transcription factors Lhx3, Lhx4, Isl1 and Isl2 promote the segregation and differentiation of spinal motor neurons and V2 interneurons. Based on their high sequence identity and their similar distribution, the Lhx3 and Lhx4 paralogs are considered to contribute similarly to these processes. However, the specific contribution of Lhx4 has never been studied. Here, we provide evidence that Lhx3 and Lhx4 are present in the same cell populations during spinal cord development. Similarly to Lhx3, Lhx4 can form multiproteic complexes with Isl1 or Isl2 and the nuclear LIM interactor NLI. Lhx4 can stimulate a V2-specific enhancer more efficiently than Lhx3 and surpasses Lhx3 in promoting the differentiation of V2a interneurons in chicken embryo electroporation experiments. Finally, Lhx4 inactivation in mice results in alterations of differentiation of the V2a subpopulation, but not of motor neuron production, suggesting that Lhx4 plays unique roles in V2a differentiation that are not compensated by the presence of Lhx3. Thus, Lhx4 could be the major LIM-HD factor involved in V2a interneuron differentiation during spinal cord development and should be considered for in vitro differentiation of spinal neuronal populations.


Sujet(s)
Différenciation cellulaire , Interneurones , Protéines à homéodomaine LIM , Moelle spinale , Facteurs de transcription , Animaux , Protéines à homéodomaine LIM/métabolisme , Protéines à homéodomaine LIM/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Interneurones/métabolisme , Interneurones/cytologie , Moelle spinale/cytologie , Moelle spinale/métabolisme , Moelle spinale/embryologie , Embryon de poulet , Souris , Motoneurones/métabolisme , Motoneurones/cytologie , Humains , Régulation de l'expression des gènes au cours du développement
2.
PLoS One ; 19(6): e0301670, 2024.
Article de Anglais | MEDLINE | ID: mdl-38917070

RÉSUMÉ

The Hedgehog (HH) pathway is crucial for embryonic development, and adult homeostasis. Its dysregulation is implicated in multiple diseases. Existing cellular models used to study HH signal regulation in mammals do not fully recapitulate the complexity of the pathway. Here we show that Spinal Cord Organoids (SCOs) can be applied to quantitively study the activity of the HH pathway. During SCO formation, the specification of different categories of neural progenitors (NPC) depends on the intensity of the HH signal, mirroring the process that occurs during neural tube development. By assessing the number of NPCs within these distinct subgroups, we are able to categorize and quantify the activation level of the HH pathway. We validate this system by measuring the effects of mutating the HH receptor PTCH1 and the impact of HH agonists and antagonists on NPC specification. SCOs represent an accessible and reliable in-vitro tool to quantify HH signaling and investigate the contribution of genetic and chemical cues in the HH pathway regulation.


Sujet(s)
Protéines Hedgehog , Organoïdes , Transduction du signal , Moelle spinale , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Animaux , Organoïdes/métabolisme , Organoïdes/cytologie , Moelle spinale/métabolisme , Moelle spinale/cytologie , Souris , Cellules souches neurales/métabolisme , Cellules souches neurales/cytologie , Récepteur Patched-1/métabolisme , Récepteur Patched-1/génétique
3.
Nature ; 630(8018): 926-934, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38898273

RÉSUMÉ

Krause corpuscles, which were discovered in the 1850s, are specialized sensory structures found within the genitalia and other mucocutaneous tissues1-4. The physiological properties and functions of Krause corpuscles have remained unclear since their discovery. Here we report the anatomical and physiological properties of Krause corpuscles of the mouse clitoris and penis and their roles in sexual behaviour. We observed a high density of Krause corpuscles in the clitoris compared with the penis. Using mouse genetic tools, we identified two distinct somatosensory neuron subtypes that innervate Krause corpuscles of both the clitoris and penis and project to a unique sensory terminal region of the spinal cord. In vivo electrophysiology and calcium imaging experiments showed that both Krause corpuscle afferent types are A-fibre rapid-adapting low-threshold mechanoreceptors, optimally tuned to dynamic, light-touch and mechanical vibrations (40-80 Hz) applied to the clitoris or penis. Functionally, selective optogenetic activation of Krause corpuscle afferent terminals evoked penile erection in male mice and vaginal contraction in female mice, while genetic ablation of Krause corpuscles impaired intromission and ejaculation of males and reduced sexual receptivity of females. Thus, Krause corpuscles of the clitoris and penis are highly sensitive mechanical vibration detectors that mediate sexually dimorphic mating behaviours.


Sujet(s)
Clitoris , Mécanorécepteurs , Pénis , Comportement sexuel chez les animaux , Toucher , Vibration , Animaux , Femelle , Mâle , Souris , Clitoris/innervation , Clitoris/physiologie , Éjaculation/physiologie , Mécanorécepteurs/métabolisme , Mécanorécepteurs/physiologie , Optogénétique , Érection du pénis/physiologie , Pénis/innervation , Pénis/physiologie , Comportement sexuel chez les animaux/physiologie , Moelle spinale/physiologie , Moelle spinale/cytologie , Toucher/physiologie , Vagin/physiologie , Neurones/physiologie
4.
eNeuro ; 11(6)2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38744490

RÉSUMÉ

Oligodendrocytes, the myelin-producing glial cells of the central nervous system (CNS), crucially contribute to myelination and circuit function. An increasing amount of evidence suggests that intracellular calcium (Ca2+) dynamics in oligodendrocytes mediates activity-dependent and activity-independent myelination. Unraveling how myelinating oligodendrocytes orchestrate and integrate Ca2+ signals, particularly in relation to axonal firing, is crucial for gaining insights into their role in the CNS development and function, both in health and disease. In this framework, we used the recombinant adeno-associated virus/Olig001 capsid variant to express the genetically encoded Ca2+ indicator jGCaMP8s, under the control of the myelin basic protein promoter. In our study, this tool exhibits excellent tropism and selectivity for myelinating and mature oligodendrocytes, and it allows monitoring Ca2+ activity in myelin-forming cells, both in isolated primary cultures and organotypic spinal cord explants. By live imaging of myelin Ca2+ events in oligodendrocytes within organ cultures, we observed a rapid decline in the amplitude and duration of Ca2+ events across different in vitro developmental stages. Active myelin sheath remodeling and growth are modulated at the level of myelin-axon interface through Ca2+ signaling, and, during early myelination in organ cultures, this phase is finely tuned by the firing of axon action potentials. In the later stages of myelination, Ca2+ events in mature oligodendrocytes no longer display such a modulation, underscoring the involvement of complex Ca2+ signaling in CNS myelination.


Sujet(s)
Calcium , Dependovirus , Gaine de myéline , Oligodendroglie , Techniques de culture d'organes , Moelle spinale , Animaux , Oligodendroglie/métabolisme , Moelle spinale/métabolisme , Moelle spinale/cytologie , Calcium/métabolisme , Dependovirus/génétique , Gaine de myéline/métabolisme , Signalisation calcique/physiologie , Souris de lignée C57BL , Souris , Cellules cultivées , Femelle , Rats
5.
Proc Natl Acad Sci U S A ; 121(23): e2314213121, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38805282

RÉSUMÉ

The anterolateral system (ALS) is a major ascending pathway from the spinal cord that projects to multiple brain areas and underlies the perception of pain, itch, and skin temperature. Despite its importance, our understanding of this system has been hampered by the considerable functional and molecular diversity of its constituent cells. Here, we use fluorescence-activated cell sorting to isolate ALS neurons belonging to the Phox2a-lineage for single-nucleus RNA sequencing. We reveal five distinct clusters of ALS neurons (ALS1-5) and document their laminar distribution in the spinal cord using in situ hybridization. We identify three clusters of neurons located predominantly in laminae I-III of the dorsal horn (ALS1-3) and two clusters with cell bodies located in deeper laminae (ALS4 and ALS5). Our findings reveal the transcriptional logic that underlies ALS neuronal diversity in the adult mouse and uncover the molecular identity of two previously identified classes of projection neurons. We also show that these molecular signatures can be used to target groups of ALS neurons using retrograde viral tracing. Overall, our findings provide a valuable resource for studying somatosensory biology and targeting subclasses of ALS neurons.


Sujet(s)
Protéines à homéodomaine , Animaux , Souris , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme , Moelle spinale/cytologie , Moelle spinale/métabolisme , Neurones/métabolisme , Séquençage nucléotidique à haut débit , Mâle , Noyau de la cellule/métabolisme , Noyau de la cellule/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
6.
Curr Top Dev Biol ; 159: 168-231, 2024.
Article de Anglais | MEDLINE | ID: mdl-38729676

RÉSUMÉ

The development of the vertebrate spinal cord involves the formation of the neural tube and the generation of multiple distinct cell types. The process starts during gastrulation, combining axial elongation with specification of neural cells and the formation of the neuroepithelium. Tissue movements produce the neural tube which is then exposed to signals that provide patterning information to neural progenitors. The intracellular response to these signals, via a gene regulatory network, governs the spatial and temporal differentiation of progenitors into specific cell types, facilitating the assembly of functional neuronal circuits. The interplay between the gene regulatory network, cell movement, and tissue mechanics generates the conserved neural tube pattern observed across species. In this review we offer an overview of the molecular and cellular processes governing the formation and patterning of the neural tube, highlighting how the remarkable complexity and precision of vertebrate nervous system arises. We argue that a multidisciplinary and multiscale understanding of the neural tube development, paired with the study of species-specific strategies, will be crucial to tackle the open questions.


Sujet(s)
Plan d'organisation du corps , Régulation de l'expression des gènes au cours du développement , Tube neural , Transduction du signal , Tube neural/embryologie , Tube neural/métabolisme , Tube neural/cytologie , Animaux , Plan d'organisation du corps/génétique , Humains , Réseaux de régulation génique , Moelle spinale/embryologie , Moelle spinale/cytologie , Moelle spinale/métabolisme , Différenciation cellulaire , Mouvement cellulaire
7.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article de Anglais | MEDLINE | ID: mdl-38731901

RÉSUMÉ

Growing demand for therapeutic tissue repair recurrently focusses scientists' attention on critical assessment of postmortal collection of live cells, especially stem cells. Our study aimed to assess the survival of neuronal progenitors in postmortal spinal cord and their differentiation potential. Postmortal samples of spinal cords were obtained from human-sized animals (goats) at 6, 12, 24, 36, and 54 h after slaughter. Samples were studied by immunohistology, differentiation assay, Western blot and flow cytometry for the presence and location of GD2-positive neural progenitors and their susceptibility to cell death. TUNEL staining of the goat spinal cord samples over 6-54 h postmortem revealed no difference in the number of positive cells per cross-section. Many TUNEL-positive cells were located in the gray commissure around the central canal of the spinal cord; no increase in TUNEL-positive cells was recorded in either posterior or anterior horns of the gray matter where many GD2-positive neural progenitors can be found. The active caspase 3 amount as measured by Western blot at the same intervals was moderately increasing over time. Neuronal cells were enriched by magnetic separation with antibodies against CD24; among them, the GD2-positive neural progenitor subpopulation did not overlap with apoptotic cells having high pan-caspase activity. Apoptotic cell death events are relatively rare in postmortal spinal cords and are not increased in areas of the neural progenitor cell's location, within measured postmortal intervals, or among the CD24/GD2-positive cells. Data from our study suggest postmortal spinal cords as a valuable source for harvesting highly viable allogenic neural progenitor cells.


Sujet(s)
Apoptose , Capra , Cellules souches neurales , Moelle spinale , Animaux , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Moelle spinale/métabolisme , Moelle spinale/cytologie , Différenciation cellulaire , Survie cellulaire , Caspase-3/métabolisme
8.
Mol Brain ; 17(1): 25, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38773624

RÉSUMÉ

A growing body of evidence indicates intra- and inter-regional heterogeneity of astrocytes in the brain. However, because of a lack of an efficient method for isolating astrocytes from the spinal cord, little is known about how much spinal cord astrocytes are heterogeneous in adult mice. In this study, we developed a new method for isolating spinal astrocytes from adult mice using a cold-active protease from Bacillus licheniformis with an astrocyte cell surface antigen-2 (ACSA-2) antibody. Using fluorescence-activated cell sorting, isolated spinal ACSA-2+ cells were divided into two distinct populations, ACSA-2high and ACSA-2low. By analyzing the expression of cell-type marker genes, the ACSA-2high and ACSA-2low populations were identified as astrocytes and ependymal cells, respectively. Furthermore, ACSA-2high cells had mRNAs encoding genes that were abundantly expressed in the gray matter (GM) but not white matter astrocytes. By optimizing enzymatic isolation procedures, the yield of GM astrocytes also increased. Therefore, our newly established method enabled the selective and efficient isolation of GM astrocytes from the spinal cord of adult mice and may be useful for bulk- or single-cell RNA-sequencing under physiological and pathological conditions.


Sujet(s)
Astrocytes , Séparation cellulaire , Substance grise , Moelle spinale , Animaux , Astrocytes/métabolisme , Astrocytes/cytologie , Moelle spinale/cytologie , Séparation cellulaire/méthodes , Souris de lignée C57BL , Souris , Mâle , ARN messager/métabolisme , ARN messager/génétique , Vieillissement
9.
J Physiol ; 602(9): 2107-2126, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38568869

RÉSUMÉ

We are studying the mechanisms of H-reflex operant conditioning, a simple form of learning. Modelling studies in the literature and our previous data suggested that changes in the axon initial segment (AIS) might contribute. To explore this, we used blinded quantitative histological and immunohistochemical methods to study in adult rats the impact of H-reflex conditioning on the AIS of the spinal motoneuron that produces the reflex. Successful, but not unsuccessful, H-reflex up-conditioning was associated with greater AIS length and distance from soma; greater length correlated with greater H-reflex increase. Modelling studies in the literature suggest that these increases may increase motoneuron excitability, supporting the hypothesis that they may contribute to H-reflex increase. Up-conditioning did not affect AIS ankyrin G (AnkG) immunoreactivity (IR), p-p38 protein kinase IR, or GABAergic terminals. Successful, but not unsuccessful, H-reflex down-conditioning was associated with more GABAergic terminals on the AIS, weaker AnkG-IR, and stronger p-p38-IR. More GABAergic terminals and weaker AnkG-IR correlated with greater H-reflex decrease. These changes might potentially contribute to the positive shift in motoneuron firing threshold underlying H-reflex decrease; they are consistent with modelling suggesting that sodium channel change may be responsible. H-reflex down-conditioning did not affect AIS dimensions. This evidence that AIS plasticity is associated with and might contribute to H-reflex conditioning adds to evidence that motor learning involves both spinal and brain plasticity, and both neuronal and synaptic plasticity. AIS properties of spinal motoneurons are likely to reflect the combined influence of all the motor skills that share these motoneurons. KEY POINTS: Neuronal action potentials normally begin in the axon initial segment (AIS). AIS plasticity affects neuronal excitability in development and disease. Whether it does so in learning is unknown. Operant conditioning of a spinal reflex, a simple learning model, changes the rat spinal motoneuron AIS. Successful, but not unsuccessful, H-reflex up-conditioning is associated with greater AIS length and distance from soma. Successful, but not unsuccessful, down-conditioning is associated with more AIS GABAergic terminals, less ankyrin G, and more p-p38 protein kinase. The associations between AIS plasticity and successful H-reflex conditioning are consistent with those between AIS plasticity and functional changes in development and disease, and with those predicted by modelling studies in the literature. Motor learning changes neurons and synapses in spinal cord and brain. Because spinal motoneurons are the final common pathway for behaviour, their AIS properties probably reflect the combined impact of all the behaviours that use these motoneurons.


Sujet(s)
Segment initial de l'axone , Réflexe H , Motoneurones , Rat Sprague-Dawley , Animaux , Motoneurones/physiologie , Rats , Mâle , Réflexe H/physiologie , Segment initial de l'axone/physiologie , Apprentissage/physiologie , Moelle spinale/physiologie , Moelle spinale/cytologie , Axones/physiologie , Plasticité neuronale/physiologie , Conditionnement opérant/physiologie , Ankyrines/métabolisme
10.
ACS Biomater Sci Eng ; 10(5): 3203-3217, 2024 05 13.
Article de Anglais | MEDLINE | ID: mdl-38557027

RÉSUMÉ

The intricate electrophysiological functions and anatomical structures of spinal cord tissue render the establishment of in vitro models for spinal cord-related diseases highly challenging. Currently, both in vivo and in vitro models for spinal cord-related diseases are still underdeveloped, complicating the exploration and development of effective therapeutic drugs or strategies. Organoids cultured from human induced pluripotent stem cells (hiPSCs) hold promise as suitable in vitro models for spinal cord-related diseases. However, the cultivation of spinal cord organoids predominantly relies on Matrigel, a matrix derived from murine sarcoma tissue. Tissue-specific extracellular matrices are key drivers of complex organ development, thus underscoring the urgent need to research safer and more physiologically relevant organoid culture materials. Herein, we have prepared a rat decellularized brain extracellular matrix hydrogel (DBECMH), which supports the formation of hiPSC-derived spinal cord organoids. Compared with Matrigel, organoids cultured in DBECMH exhibited higher expression levels of markers from multiple compartments of the natural spinal cord, facilitating the development and maturation of spinal cord organoid tissues. Our study suggests that DBECMH holds potential to replace Matrigel as the standard culture medium for human spinal cord organoids, thereby advancing the development of spinal cord organoid culture protocols and their application in in vitro modeling of spinal cord-related diseases.


Sujet(s)
Encéphale , Hydrogels , Cellules souches pluripotentes induites , Organoïdes , Moelle spinale , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/cytologie , Organoïdes/métabolisme , Humains , Animaux , Moelle spinale/cytologie , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Hydrogels/composition chimique , Hydrogels/pharmacologie , Encéphale/métabolisme , Rats , Matrice extracellulaire décellularisée/composition chimique , Matrice extracellulaire décellularisée/pharmacologie , Matrice extracellulaire/métabolisme , Matrice extracellulaire/composition chimique , Laminine/pharmacologie , Laminine/composition chimique , Protéoglycanes/composition chimique , Rat Sprague-Dawley , Association médicamenteuse , Collagène
11.
ACS Biomater Sci Eng ; 10(5): 3218-3231, 2024 05 13.
Article de Anglais | MEDLINE | ID: mdl-38593429

RÉSUMÉ

Spinal cord organoids are of significant value in the research of spinal cord-related diseases by simulating disease states, thereby facilitating the development of novel therapies. However, the complexity of spinal cord structure and physiological functions, along with the lack of human-derived inducing components, presents challenges in the in vitro construction of human spinal cord organoids. Here, we introduce a novel human decellularized placenta-derived extracellular matrix hydrogel (DPECMH) and, combined with a new induction protocol, successfully construct human spinal cord organoids. The human placenta-sourced decellularized extracellular matrix (dECM), verified through hematoxylin and eosin staining, DNA quantification, and immunofluorescence staining, retained essential ECM components such as elastin, fibronectin, type I collagen, laminin, and so forth. The temperature-sensitive hydrogel made from human placenta dECM demonstrated good biocompatibility and promoted the differentiation of human induced pluripotent stem cell (hiPSCs)-derived spinal cord organoids into neurons. It displayed enhanced expression of laminar markers in comparison to Matrigel and showed higher expression of laminar markers compared to Matrigel, accelerating the maturation process of spinal cord organoids and demonstrating its potential as an organoid culture substrate. DPECMH has the potential to replace Matrigel as the standard additive for human spinal cord organoids, thus advancing the development of spinal cord organoid culture protocols and their application in the in vitro modeling of spinal cord-related diseases.


Sujet(s)
Différenciation cellulaire , Matrice extracellulaire décellularisée , Hydrogels , Cellules souches pluripotentes induites , Organoïdes , Placenta , Moelle spinale , Humains , Organoïdes/cytologie , Organoïdes/métabolisme , Organoïdes/effets des médicaments et des substances chimiques , Femelle , Placenta/cytologie , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Cellules souches pluripotentes induites/métabolisme , Grossesse , Hydrogels/composition chimique , Hydrogels/pharmacologie , Moelle spinale/cytologie , Moelle spinale/métabolisme , Différenciation cellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire décellularisée/pharmacologie , Matrice extracellulaire décellularisée/composition chimique , Matrice extracellulaire/métabolisme , Matrice extracellulaire/composition chimique , Laminine/pharmacologie , Laminine/composition chimique
12.
J Vis Exp ; (206)2024 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-38682940

RÉSUMÉ

Resolutive cures for spinal cord injuries (SCIs) are still lacking, due to the complex pathophysiology. One of the most promising regenerative approaches is based on stem cell transplantation to replace lost tissue and promote functional recovery. This approach should be further explored better in vitro and ex vivo for safety and efficacy before proceeding with more expensive and time-consuming animal testing. In this work, we show the establishment of a long-term platform based on mouse spinal cord (SC) organotypic slices transplanted with human neural stem cells to test cellular replacement therapies for SCIs. Standard SC organotypic cultures are maintained for around 2 or 3 weeks in vitro. Here, we describe an optimized protocol for long-term maintenance (≥30 days) for up to 90 days. The medium used for long-term culturing of SC slices was also optimized for transplanting neural stem cells into the organotypic model. Human SC-derived neuroepithelial stem (h-SC-NES) cells carrying a green fluorescent protein (GFP) reporter were transplanted into mouse SC slices. Thirty days after the transplant, cells still show GFP expression and a low apoptotic rate, suggesting that the optimized environment sustained their survival and integration inside the tissue. This protocol represents a robust reference for efficiently testing cell replacement therapies in the SC tissue. This platform will allow researchers to perform an ex vivo pre-screening of different cell transplantation therapies, helping them to choose the most appropriate strategy before proceeding with in vivo experiments.


Sujet(s)
Cellules souches neurales , Traumatismes de la moelle épinière , Moelle spinale , Animaux , Souris , Traumatismes de la moelle épinière/thérapie , Humains , Cellules souches neurales/cytologie , Cellules souches neurales/transplantation , Moelle spinale/cytologie , Techniques de culture d'organes/méthodes , Transplantation de cellules souches/méthodes
13.
Cells ; 13(8)2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38667267

RÉSUMÉ

The differential expression of transcription factors during embryonic development has been selected as the main feature to define the specific subclasses of spinal interneurons. However, recent studies based on single-cell RNA sequencing and transcriptomic experiments suggest that this approach might not be appropriate in the adult spinal cord, where interneurons show overlapping expression profiles, especially in the ventral region. This constitutes a major challenge for the identification and direct targeting of specific populations that could be involved in locomotor recovery after a traumatic spinal cord injury in adults. Current experimental therapies, including electrical stimulation, training, pharmacological treatments, or cell implantation, that have resulted in improvements in locomotor behavior rely on the modulation of the activity and connectivity of interneurons located in the surroundings of the lesion core for the formation of detour circuits. However, very few publications clarify the specific identity of these cells. In this work, we review the studies where premotor interneurons were able to create new intraspinal circuits after different kinds of traumatic spinal cord injury, highlighting the difficulties encountered by researchers, to classify these populations.


Sujet(s)
Interneurones , Récupération fonctionnelle , Traumatismes de la moelle épinière , Adulte , Animaux , Humains , Interneurones/métabolisme , Moelle spinale/cytologie , Moelle spinale/anatomopathologie , Traumatismes de la moelle épinière/thérapie , Traumatismes de la moelle épinière/physiopathologie
14.
Exp Neurol ; 376: 114779, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38621449

RÉSUMÉ

Neural stem cells have exhibited efficacy in pre-clinical models of spinal cord injury (SCI) and are on a translational path to human testing. We recently reported that neural stem cells must be driven to a spinal cord fate to optimize host axonal regeneration into sites of implantation in the injured spinal cord, where they subsequently form neural relays across the lesion that support significant functional improvement. We also reported methods of deriving and culturing human spinal cord neural stem cells derived from embryonic stem cells that can be sustained over serial high passage numbers in vitro, providing a potentially optimized cell source for human clinical trials. We now report further optimization of methods for deriving and sustaining cultures of human spinal cord neural stem cell lines that result in improved karyotypic stability while retaining anatomical efficacy in vivo. This development improves prospects for safe human translation.


Sujet(s)
Différenciation cellulaire , Cellules souches neurales , Traumatismes de la moelle épinière , Moelle spinale , Humains , Cellules souches neurales/cytologie , Moelle spinale/cytologie , Animaux , Traumatismes de la moelle épinière/thérapie , Différenciation cellulaire/physiologie , Techniques de culture cellulaire/méthodes , Cellules cultivées , Souris , Transplantation de cellules souches/méthodes
15.
eNeuro ; 11(4)2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38627062

RÉSUMÉ

Proprioception, the sense of limb and body position, is required to produce accurate and precise movements. Proprioceptive sensory neurons transmit muscle length and tension information to the spinal cord. The function of excitatory neurons in the intermediate spinal cord, which receive this proprioceptive information, remains poorly understood. Using genetic labeling strategies and patch-clamp techniques in acute spinal cord preparations in mice, we set out to uncover how two sets of spinal neurons, Clarke's column (CC) and Atoh1-lineage neurons, respond to electrical activity and how their inputs are organized. Both sets of neurons are located in close proximity in laminae V-VII of the thoracolumbar spinal cord and have been described to receive proprioceptive signals. We find that a majority of CC neurons have a tonic-firing type and express a distinctive hyperpolarization-activated current (Ih). Atoh1-lineage neurons, which cluster into two spatially distinct populations, are mostly a fading-firing type and display similar electrophysiological properties to each other, possibly due to their common developmental lineage. Finally, we find that CC neurons respond to stimulation of lumbar dorsal roots, consistent with prior knowledge that CC neurons receive hindlimb proprioceptive information. In contrast, using a combination of electrical stimulation, optogenetic stimulation, and transsynaptic rabies virus tracing, we find that Atoh1-lineage neurons receive heterogeneous, predominantly local thoracic inputs that include parvalbumin-lineage sensory afferents and local interneuron presynaptic inputs. Altogether, we find that CC and Atoh1-lineage neurons have distinct membrane properties and sensory input organization, representing different subcircuit modes of proprioceptive information processing.


Sujet(s)
Proprioception , Moelle spinale , Animaux , Proprioception/physiologie , Moelle spinale/physiologie , Moelle spinale/cytologie , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Souris transgéniques , Souris , Mâle , Femelle , Potentiels d'action/physiologie , Cellules réceptrices sensorielles/physiologie , Techniques de patch-clamp , Souris de lignée C57BL , Vertèbres thoraciques
16.
Cell Rep ; 43(4): 113993, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38551963

RÉSUMÉ

Corticospinal neurons (CSNs) synapse directly on spinal neurons, a diverse assortment of cells with unique structural and functional properties necessary for body movements. CSNs modulating forelimb behavior fractionate into caudal forelimb area (CFA) and rostral forelimb area (RFA) motor cortical populations. Despite their prominence, the full diversity of spinal neurons targeted by CFA and RFA CSNs is uncharted. Here, we use anatomical and RNA sequencing methods to show that CSNs synapse onto a remarkably selective group of spinal cell types, favoring inhibitory populations that regulate motoneuron activity and gate sensory feedback. CFA and RFA CSNs target similar spinal neuron types, with notable exceptions that suggest that these populations differ in how they influence behavior. Finally, axon collaterals of CFA and RFA CSNs target similar brain regions yet receive highly divergent inputs. These results detail the rules of CSN connectivity throughout the brain and spinal cord for two regions critical for forelimb behavior.


Sujet(s)
Membre thoracique , Tractus pyramidaux , Animaux , Membre thoracique/physiologie , Tractus pyramidaux/physiologie , Moelle spinale/physiologie , Moelle spinale/cytologie , Souris , Cortex moteur/physiologie , Neurones/physiologie , Motoneurones/physiologie , Femelle , Mâle , Axones/physiologie , Synapses/physiologie
17.
J Neurosci ; 44(18)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38438260

RÉSUMÉ

Locomotion allows us to move and interact with our surroundings. Spinal networks that control locomotion produce rhythm and left-right and flexor-extensor coordination. Several glutamatergic populations, Shox2 non-V2a, Hb9-derived interneurons, and, recently, spinocerebellar neurons have been proposed to be involved in the mouse rhythm generating networks. These cells make up only a smaller fraction of the excitatory cells in the ventral spinal cord. Here, we set out to identify additional populations of excitatory spinal neurons that may be involved in rhythm generation or other functions in the locomotor network. We use RNA sequencing from glutamatergic, non-glutamatergic, and Shox2 cells in the neonatal mice from both sexes followed by differential gene expression analyses. These analyses identified transcription factors that are highly expressed by glutamatergic spinal neurons and differentially expressed between Shox2 neurons and glutamatergic neurons. From this latter category, we identified the Lhx9-derived neurons as having a restricted spinal expression pattern with no Shox2 neuron overlap. They are purely glutamatergic and ipsilaterally projecting. Ablation of the glutamatergic transmission or acute inactivation of the neuronal activity of Lhx9-derived neurons leads to a decrease in the frequency of locomotor-like activity without change in coordination pattern. Optogenetic activation of Lhx9-derived neurons promotes locomotor-like activity and modulates the frequency of the locomotor activity. Calcium activities of Lhx9-derived neurons show strong left-right out-of-phase rhythmicity during locomotor-like activity. Our study identifies a distinct population of spinal excitatory neurons that regulates the frequency of locomotor output with a suggested role in rhythm-generation in the mouse alongside other spinal populations.


Sujet(s)
Interneurones , Protéines à homéodomaine LIM , Locomotion , Moelle spinale , Facteurs de transcription , Animaux , Interneurones/physiologie , Souris , Protéines à homéodomaine LIM/génétique , Protéines à homéodomaine LIM/métabolisme , Locomotion/physiologie , Moelle spinale/physiologie , Moelle spinale/cytologie , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Mâle , Femelle , Acide glutamique/métabolisme , Animaux nouveau-nés , Protéines à homéodomaine/génétique , Protéines à homéodomaine/métabolisme
18.
Nature ; 628(8007): 391-399, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38408487

RÉSUMÉ

The human nervous system is a highly complex but organized organ. The foundation of its complexity and organization is laid down during regional patterning of the neural tube, the embryonic precursor to the human nervous system. Historically, studies of neural tube patterning have relied on animal models to uncover underlying principles. Recently, models of neurodevelopment based on human pluripotent stem cells, including neural organoids1-5 and bioengineered neural tube development models6-10, have emerged. However, such models fail to recapitulate neural patterning along both rostral-caudal and dorsal-ventral axes in a three-dimensional tubular geometry, a hallmark of neural tube development. Here we report a human pluripotent stem cell-based, microfluidic neural tube-like structure, the development of which recapitulates several crucial aspects of neural patterning in brain and spinal cord regions and along rostral-caudal and dorsal-ventral axes. This structure was utilized for studying neuronal lineage development, which revealed pre-patterning of axial identities of neural crest progenitors and functional roles of neuromesodermal progenitors and the caudal gene CDX2 in spinal cord and trunk neural crest development. We further developed dorsal-ventral patterned microfluidic forebrain-like structures with spatially segregated dorsal and ventral regions and layered apicobasal cellular organizations that mimic development of the human forebrain pallium and subpallium, respectively. Together, these microfluidics-based neurodevelopment models provide three-dimensional lumenal tissue architectures with in vivo-like spatiotemporal cell differentiation and organization, which will facilitate the study of human neurodevelopment and disease.


Sujet(s)
Plan d'organisation du corps , Microfluidique , Tube neural , Humains , Techniques de cultures cellulaires tridimensionnelles , Différenciation cellulaire , Crête neurale/cytologie , Crête neurale/embryologie , Tube neural/cytologie , Tube neural/embryologie , Cellules souches pluripotentes/cytologie , Prosencéphale/cytologie , Prosencéphale/embryologie , Moelle spinale/cytologie , Moelle spinale/embryologie
19.
Cell Prolif ; 57(5): e13594, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38155412

RÉSUMÉ

The study of neurogenesis is essential to understanding fundamental developmental processes and for the development of cell replacement therapies for central nervous system disorders. Here, we designed an in vivo drug screening protocol in developing zebrafish to find new molecules and signalling pathways regulating neurogenesis in the ventral spinal cord. This unbiased drug screen revealed that 4 cyclooxygenase (COX) inhibitors reduced the generation of serotonergic interneurons in the developing spinal cord. These results fitted very nicely with available single-cell RNAseq data revealing that floor plate cells show differential expression of 1 of the 2 COX2 zebrafish genes (ptgs2a). Indeed, several selective COX2 inhibitors and two different morpholinos against ptgs2a reduced the number of serotonergic neurons in the ventral spinal cord and led to locomotor deficits. Single-cell RNAseq data and different pharmacological manipulations further revealed that COX2-floor plate-derived prostaglandin D2 promotes neurogenesis in the developing spinal cord by promoting mitotic activity in progenitor cells. Rescue experiments using a phosphodiesterase-4 inhibitor suggest that intracellular changes in cAMP levels underlie the effects of COX inhibitors on neurogenesis and locomotion. Our study provides compelling in vivo evidence showing that prostaglandin signalling promotes neurogenesis in the ventral spinal cord.


Sujet(s)
Cyclooxygenase 2 , Neurogenèse , Moelle spinale , Danio zébré , Animaux , Danio zébré/métabolisme , Neurogenèse/effets des médicaments et des substances chimiques , Moelle spinale/métabolisme , Moelle spinale/cytologie , Moelle spinale/effets des médicaments et des substances chimiques , Cyclooxygenase 2/métabolisme , Cyclooxygenase 2/génétique , Évaluation préclinique de médicament/méthodes , Inhibiteurs de la cyclooxygénase 2/pharmacologie , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Inhibiteurs des cyclooxygénases/pharmacologie
20.
Nature ; 624(7991): 403-414, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-38092914

RÉSUMÉ

The brain controls nearly all bodily functions via spinal projecting neurons (SPNs) that carry command signals from the brain to the spinal cord. However, a comprehensive molecular characterization of brain-wide SPNs is still lacking. Here we transcriptionally profiled a total of 65,002 SPNs, identified 76 region-specific SPN types, and mapped these types into a companion atlas of the whole mouse brain1. This taxonomy reveals a three-component organization of SPNs: (1) molecularly homogeneous excitatory SPNs from the cortex, red nucleus and cerebellum with somatotopic spinal terminations suitable for point-to-point communication; (2) heterogeneous populations in the reticular formation with broad spinal termination patterns, suitable for relaying commands related to the activities of the entire spinal cord; and (3) modulatory neurons expressing slow-acting neurotransmitters and/or neuropeptides in the hypothalamus, midbrain and reticular formation for 'gain setting' of brain-spinal signals. In addition, this atlas revealed a LIM homeobox transcription factor code that parcellates the reticulospinal neurons into five molecularly distinct and spatially segregated populations. Finally, we found transcriptional signatures of a subset of SPNs with large soma size and correlated these with fast-firing electrophysiological properties. Together, this study establishes a comprehensive taxonomy of brain-wide SPNs and provides insight into the functional organization of SPNs in mediating brain control of bodily functions.


Sujet(s)
Encéphale , Analyse de profil d'expression de gènes , Voies nerveuses , Neurones , Moelle spinale , Animaux , Souris , Hypothalamus , Neurones/métabolisme , Neuropeptides , Moelle spinale/cytologie , Moelle spinale/métabolisme , Encéphale/cytologie , Encéphale/métabolisme , Agents neuromédiateurs , Mésencéphale/cytologie , Formation réticulaire/cytologie , Électrophysiologie , Cervelet/cytologie , Cortex cérébral/cytologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...