1
|
Othman AMM, Abdallah OY, Elnaggar YSR. Topical hyalubilosomes of dantrolene sodium as muscle targeted nanocarrier for muscle spasms: fabrication, ex-vivo permeation and behavioral animal model. Pharm Dev Technol 2025:1-14. [PMID: 40371687 DOI: 10.1080/10837450.2025.2504999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/16/2025]
Abstract
Topical muscle relaxants are gaining interest in pharmaceuticals. Dantrolene sodium (DS), an FDA-approved relaxant targeting ryanodine receptors, is limited in topical use by poor physicochemical properties, delayed onset, and hepatotoxicity. This study introduces the first optimized hyalubilosome-based nanocarrier for non-invasive DS delivery. Two anionic surfactants were used as edge activators to improve drug encapsulation and permeation. The optimized nanocarrier had a spherical shape, 165 nm particle size, -31.2 mV zeta potential, and 97.47% entrapment efficiency. Ex vivo studies showed superior permeation compared to DS suspension (10% in water, pH 6.8), with 30% of the dose permeating within 15 min. In vivo, efficacy was tested in Wistar mice using the Straub tail test with a single 30 mg/kg topical dose. Behavioral analysis showed a fivefold increase in muscle relaxation vs. untreated controls (p < 0.0001). The formulation had an onset within one minute and complete relief within two minutes, unlike the conventional topical DS, which showed no effect for 90 min. This highlights hyaluronic acid-based transbilosomes as a promising nanoplatform for fast, effective topical DS delivery and potential muscle spasm treatment.
Collapse
Affiliation(s)
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Head of Research & International Publishing Administration, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
2
|
Roze E, Dubacq C, Welniarz Q. Corticospinal Tract Development, Evolution, and Skilled Movements. Mov Disord 2025. [PMID: 40277091 DOI: 10.1002/mds.30199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
The evolution of the corticospinal tract (CST) is closely linked to the development of skilled voluntary movements in mammals. The main evolutionary divergence concerns the position of the CST within the spinal cord white matter and its postsynaptic targets in the grey matter. Here, we examine the developmental steps contributing to the CST projection pattern from an evolutionary point of view. Recent studies have highlighted the molecular mechanisms involved in these processes and how they relate to the acquisition of skilled movements. Comparison of the evolution of the CST in different species offers a new perspective on manual dexterity. In particular, it adds a new level of complexity to the classic view linking the evolution of the CST and the sequential improvement of skilled hand movements from rodents to primates. © 2025 The Author(s). Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Emmanuel Roze
- Sorbonne Université, INSERM, CNRS, Paris Brain Institute Institut du Cerveau, Paris, France
- Département de Neurologie, Assistance Publique - Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| | - Caroline Dubacq
- Sorbonne Université, INSERM, CNRS, Paris Brain Institute Institut du Cerveau, Paris, France
| | - Quentin Welniarz
- Sorbonne Université, INSERM, CNRS, Paris Brain Institute Institut du Cerveau, Paris, France
- Département de Neurologie, Assistance Publique - Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
3
|
Eisen A, Vucic S, Kiernan MC. Amyotrophic lateral sclerosis represents corticomotoneuronal system failure. Muscle Nerve 2025; 71:499-511. [PMID: 39511939 PMCID: PMC11887532 DOI: 10.1002/mus.28290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
Several decades have passed since the anterograde corticomotoneuronal hypothesis for amyotrophic lateral sclerosis (ALS) was proposed. The intervening years have witnessed its emergent support based on anatomical, pathological, physiological, neuroimaging, and molecular biological studies. The evolution of an extensive corticomotoneuronal system appears restricted to the human species, with ALS representing a uniquely human disease. While some, very select non-human primates have limited corticomotoneuronal projections, these tend to be absent in all other animals. From a general perspective, the early clinical features of ALS may be considered to reflect failure of the corticomotoneuronal system. The characteristic loss of skilled motor dexterity involving the limbs, and speech impairment through progressive bulbar dysfunction specifically involve those motor units having the strongest corticomotoneuronal projections. A similar explanation likely underlies the unique "split phenotypes" that have now been well characterized in ALS. Large Betz cells and other pyramidal corticomotoneuronal projecting neurons, with their extensive dendritic arborization, are particularly vulnerable to the elements of the ALS exposome such as aging, environmental stress and lifestyle changes. Progressive failure of the proteosome impairs nucleocytoplasmic shuffling and induces toxic but soluble TDP-43 to aggregate in corticomotoneurons. Betz cell failure is further accentuated through dysfunction of its profuse dendritic arborizations. Clarification of system specific genomes and neural networks will likely promote the initiation of precision medicine approaches directed to support the key structure that underlies the neurological manifestations of ALS, the corticomotoneuronal system.
Collapse
Affiliation(s)
- Andrew Eisen
- Division of Neurology, Department of MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Steve Vucic
- Brain and Nerve Research CenterConcord Clinical School, University of SydneySydneyNew South WalesAustralia
| | - Matthew C. Kiernan
- Neuroscience Research AustraliaSydneyNew South WalesAustralia
- NeuroscienceUniversity of New South WalesSydneyNew South WalesAustralia
| |
Collapse
|
4
|
Inoue T, Ueno M. The diversity and plasticity of descending motor pathways rewired after stroke and trauma in rodents. Front Neural Circuits 2025; 19:1566562. [PMID: 40191711 PMCID: PMC11968733 DOI: 10.3389/fncir.2025.1566562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Descending neural pathways to the spinal cord plays vital roles in motor control. They are often damaged by brain injuries such as stroke and trauma, which lead to severe motor impairments. Due to the limited capacity for regeneration of neural circuits in the adult central nervous system, currently no essential treatments are available for complete recovery. Notably, accumulating evidence shows that residual circuits of the descending pathways are dynamically reorganized after injury and contribute to motor recovery. Furthermore, recent technological advances in cell-type classification and manipulation have highlighted the structural and functional diversity of these pathways. Here, we focus on three major descending pathways, namely, the corticospinal tract from the cerebral cortex, the rubrospinal tract from the red nucleus, and the reticulospinal tract from the reticular formation, and summarize the current knowledge of their structures and functions, especially in rodent models (mice and rats). We then review and discuss the process and patterns of reorganization induced in these pathways following injury, which compensate for lost connections for recovery. Understanding the basic structural and functional properties of each descending pathway and the principles of the induction and outcome of the rewired circuits will provide therapeutic insights to enhance interactive rewiring of the multiple descending pathways for motor recovery.
Collapse
Affiliation(s)
- Takahiro Inoue
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| |
Collapse
|
5
|
Tian Y, Wu X, Luo S, Xiong D, Liu R, Hu L, Yuan Y, Shi G, Yao J, Huang Z, Fu F, Yang X, Tang Z, Zhang J, Hu K. A multi-omic single-cell landscape of cellular diversification in the developing human cerebral cortex. Comput Struct Biotechnol J 2024; 23:2173-2189. [PMID: 38827229 PMCID: PMC11141146 DOI: 10.1016/j.csbj.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 06/04/2024] Open
Abstract
The vast neuronal diversity in the human neocortex is vital for high-order brain functions, necessitating elucidation of the regulatory mechanisms underlying such unparalleled diversity. However, recent studies have yet to comprehensively reveal the diversity of neurons and the molecular logic of neocortical origin in humans at single-cell resolution through profiling transcriptomic or epigenomic landscapes, owing to the application of unimodal data alone to depict exceedingly heterogeneous populations of neurons. In this study, we generated a comprehensive compendium of the developing human neocortex by simultaneously profiling gene expression and open chromatin from the same cell. We computationally reconstructed the differentiation trajectories of excitatory projection neurons of cortical origin and inferred the regulatory logic governing lineage bifurcation decisions for neuronal diversification. We demonstrated that neuronal diversity arises from progenitor cell lineage specificity and postmitotic differentiation at distinct stages. Our data paves the way for understanding the primarily coordinated regulatory logic for neuronal diversification in the neocortex.
Collapse
Affiliation(s)
- Yuhan Tian
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Xia Wu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Songhao Luo
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China
| | - Dan Xiong
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Rong Liu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Lanqi Hu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Yuchen Yuan
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Guowei Shi
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Junjie Yao
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Zhiwei Huang
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China
| | - Fang Fu
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 511436, China
| | - Xin Yang
- Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhonghui Tang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
| | - Jiajun Zhang
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China
| | - Kunhua Hu
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Public Platform Laboratory, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510630, China
| |
Collapse
|
6
|
Isko EC, Harpole CE, Zheng XM, Zhan H, Davis MB, Zador AM, Banerjee A. Selective expansion of motor cortical projections in the evolution of vocal novelty. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612752. [PMID: 39484467 PMCID: PMC11526862 DOI: 10.1101/2024.09.13.612752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Deciphering how cortical architecture evolves to drive behavioral innovations is a long-standing challenge in neuroscience and evolutionary biology. Here, we leverage a striking behavioral novelty in the Alston's singing mouse (Scotinomys teguina), compared to the laboratory mouse (Mus musculus), to quantitatively test models of motor cortical evolution. We used bulk tracing, serial two-photon tomography, and high-throughput DNA sequencing of over 76,000 barcoded neurons to discover a specific and substantial expansion (200%) of orofacial motor cortical (OMC) projections to the auditory cortical region (AudR) and the midbrain periaqueductal gray (PAG), both implicated in vocal behaviors. Moreover, analysis of individual OMC neurons' projection motifs revealed preferential expansion of exclusive projections to AudR. Our results imply that selective expansion of ancestral motor cortical projections can underlie behavioral divergence over short evolutionary timescales, suggesting potential mechanisms for the evolution of enhanced cortical control over vocalizations-a crucial preadaptation for human language.
Collapse
Affiliation(s)
- Emily C Isko
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
- Cold Spring Harbor Laboratory School for Biological Sciences, Cold Spring Harbor, NY
| | | | - Xiaoyue Mike Zheng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
- Cold Spring Harbor Laboratory School for Biological Sciences, Cold Spring Harbor, NY
| | - Huiqing Zhan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | | | - Anthony M Zador
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
- Cold Spring Harbor Laboratory School for Biological Sciences, Cold Spring Harbor, NY
| | - Arkarup Banerjee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
- Cold Spring Harbor Laboratory School for Biological Sciences, Cold Spring Harbor, NY
| |
Collapse
|
7
|
Poinsatte K, Kenwood M, Betz D, Nawaby A, Ajay AD, Xu W, Plautz EJ, Kong X, Ramirez DMO, Goldberg MP. SpinalTRAQ: A novel volumetric cervical spinal cord atlas identifies the corticospinal tract synaptic projectome in healthy and post-stroke mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609434. [PMID: 39416130 PMCID: PMC11482800 DOI: 10.1101/2024.08.23.609434] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Descending corticospinal tract (CST) connections to the neurons of the cervical spinal cord are vital for performance of forelimb-specific fine motor skills. In rodents, CST axons are almost entirely crossed at the level of the medullary decussation. While specific contralateral axon projections have been well-characterized using anatomic and molecular approaches, the field currently lacks a cohesive imaging modality allowing rapid quantitative assessment of the entire, bilateral cervical cord projectome at the level of individual laminae and cervical levels. This is potentially important as the CST is known to undergo marked structural remodeling in development, injury, and disease. We developed SpinalTRAQ (Spinal cord Tomographic Registration and Automated Quantification), a novel volumetric cervical spinal cord atlas and machine learning-driven microscopy acquisition and analysis pipeline that uses serial two-photon tomography- images to generate unbiased, region-specific quantification of the fluorescent pixels of anterograde AAV-labeled CST pre-synaptic terminals. In adult mice, the CST synaptic projectome densely innervates the contralateral hemicord, particularly in laminae 5 and 7, with sparse, monosynaptic input to motoneurons in lamina 9. Motor pools supplying axial musculature in the upper cervical cord are bilaterally innervated. The remainder of the ipsilateral cord has sparse labeling in a distinct distribution compared to the contralateral side. Following a focal stroke of the motor cortex, there is a complete loss of descending corticospinal axons from the injured side. Consistent with prior reports of axon collateralization, the CST spinal projectome increases at four weeks post-stroke and continues to elevate by six weeks post stroke. At six weeks post-stroke, we observed striking synapse formation in the denervated hemicord from the uninjured CST in a homotopic distribution. Additionally, CST synaptic reinnervation increases in the denervated lamina 9 in nearly all motoneuron pools, exhibiting novel patterns of connectivity. Detailed level- and lamina-specific quantification of the bilateral cervical spinal cord synaptic projectome reveals previously undescribed patterns of CST connectivity in health and injury-related plasticity.
Collapse
Affiliation(s)
- Katherine Poinsatte
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Matthew Kenwood
- Department of Neurology, Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Graduate School of Biomedical Science (Neuroscience), University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Dene Betz
- Department of Neurology, Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Graduate School of Biomedical Science (Neuroscience), University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
| | - Ariana Nawaby
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Apoorva D Ajay
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Wei Xu
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Erik J Plautz
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xiangmei Kong
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Denise M O Ramirez
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mark P Goldberg
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, Long School of Medicine, University of Texas Health Science Center San Antonio, San Antonio, Texas, USA
- Peter O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Lead Contact
| |
Collapse
|
8
|
Abe P, Lavalley A, Morassut I, Santinha AJ, Roig-Puiggros S, Javed A, Klingler E, Baumann N, Prados J, Platt RJ, Jabaudon D. Molecular programs guiding arealization of descending cortical pathways. Nature 2024; 634:644-651. [PMID: 39261725 DOI: 10.1038/s41586-024-07895-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
Layer 5 extratelencephalic (ET) neurons are present across neocortical areas and send axons to multiple subcortical targets1-6. Two cardinal subtypes exist7,8: (1) Slco2a1-expressing neurons (ETdist), which predominate in the motor cortex and project distally to the pons, medulla and spinal cord; and (2) Nprs1- or Hpgd-expressing neurons (ETprox), which predominate in the visual cortex and project more proximally to the pons and thalamus. An understanding of how area-specific ETdist and ETprox emerge during development is important because they are critical for fine motor skills and are susceptible to spinal cord injury and amyotrophic lateral sclerosis9-12. Here, using cross-areal mapping of axonal projections in the mouse neocortex, we identify the subtype-specific developmental dynamics of ET neurons. Whereas subsets of ETprox emerge by pruning of ETdist axons, others emerge de novo. We outline corresponding subtype-specific developmental transcriptional programs using single-nucleus sequencing. Leveraging these findings, we use postnatal in vivo knockdown of subtype-specific transcription factors to reprogram ET neuron connectivity towards more proximal targets. Together, these results show the functional transcriptional programs driving ET neuron diversity and uncover cell subtype-specific gene regulatory networks that can be manipulated to direct target specificity in motor corticofugal pathways.
Collapse
Affiliation(s)
- Philipp Abe
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Dresden, Germany
| | - Adrien Lavalley
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland
| | - Ilaria Morassut
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Antonio J Santinha
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Sergi Roig-Puiggros
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Awais Javed
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Esther Klingler
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Natalia Baumann
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Julien Prados
- Bioinformatic Support Platform, University of Geneva, Geneva, Switzerland
| | - Randall J Platt
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Basel Research Center for Child Health, Basel, Switzerland
- Department of Chemistry, University of Basel, Basel, Switzerland
- NCCR Molecular Systems Engineering, Basel, Switzerland
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland.
- Department of Clinical Neurosciences, Geneva University Hospital, Geneva, Switzerland.
- Université Paris Cité, Imagine Institute, Paris, France.
| |
Collapse
|
9
|
Zhao HT, Schmidt ER. Human-specific genetic modifiers of cortical architecture and function. Curr Opin Genet Dev 2024; 88:102241. [PMID: 39111228 PMCID: PMC11547859 DOI: 10.1016/j.gde.2024.102241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/30/2024] [Accepted: 07/23/2024] [Indexed: 09/11/2024]
Abstract
Evolution of the cerebral cortex is thought to have been critical for the emergence of our cognitive abilities. Major features of cortical evolution include increased neuron number and connectivity and altered morpho-electric properties of cortical neurons. Significant progress has been made in identifying human-specific genetic modifiers (HSGMs), some of which are involved in shaping these features of cortical architecture. But how did these evolutionary changes support the emergence of our cognitive abilities? Here, we highlight recent studies aimed at examining the impact of HSGMs on cortical circuit function and behavior. We also discuss the need for greater insight into the link between evolution of cortical architecture and the functional and computational properties of neuronal circuits, as we seek to provide a neurobiological foundation for human cognition.
Collapse
Affiliation(s)
- Hanzhi T Zhao
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA
| | - Ewoud Re Schmidt
- Department of Neuroscience, Medical University of South Carolina, Suite 403 BSB, MSC510, 173 Ashley Ave, Charleston, SC 29425, USA.
| |
Collapse
|
10
|
Griffith EC, West AE, Greenberg ME. Neuronal enhancers fine-tune adaptive circuit plasticity. Neuron 2024; 112:3043-3057. [PMID: 39208805 PMCID: PMC11550865 DOI: 10.1016/j.neuron.2024.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
Neuronal activity-regulated gene expression plays a crucial role in sculpting neural circuits that underpin adaptive brain function. Transcriptional enhancers are now recognized as key components of gene regulation that orchestrate spatiotemporally precise patterns of gene transcription. We propose that the dynamics of enhancer activation uniquely position these genomic elements to finely tune activity-dependent cellular plasticity. Enhancer specificity and modularity can be exploited to gain selective genetic access to specific cell states, and the precise modulation of target gene expression within restricted cellular contexts enabled by targeted enhancer manipulation allows for fine-grained evaluation of gene function. Mounting evidence also suggests that enduring stimulus-induced changes in enhancer states can modify target gene activation upon restimulation, thereby contributing to a form of cell-wide metaplasticity. We advocate for focused exploration of activity-dependent enhancer function to gain new insight into the mechanisms underlying brain plasticity and cognitive dysfunction.
Collapse
Affiliation(s)
- Eric C Griffith
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Anne E West
- Department of Neurobiology, Duke University Medical Center, Durham, NC, USA.
| | | |
Collapse
|
11
|
Fait BW, Cotto B, Murakami TC, Hagemann-Jensen M, Zhan H, Freivald C, Turbek I, Gao Y, Yao Z, Way SW, Zeng H, Tasic B, Steward O, Heintz N, Schmidt EF. Spontaneously regenerative corticospinal neurons in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.09.612115. [PMID: 39314356 PMCID: PMC11419066 DOI: 10.1101/2024.09.09.612115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
The spinal cord receives inputs from the cortex via corticospinal neurons (CSNs). While predominantly a contralateral projection, a less-investigated minority of its axons terminate in the ipsilateral spinal cord. We analyzed the spatial and molecular properties of these ipsilateral axons and their post-synaptic targets in mice and found they project primarily to the ventral horn, including directly to motor neurons. Barcode-based reconstruction of the ipsilateral axons revealed a class of primarily bilaterally-projecting CSNs with a distinct cortical distribution. The molecular properties of these ipsilaterally-projecting CSNs (IP-CSNs) are strikingly similar to the previously described molecular signature of embryonic-like regenerating CSNs. Finally, we show that IP-CSNs are spontaneously regenerative after spinal cord injury. The discovery of a class of spontaneously regenerative CSNs may prove valuable to the study of spinal cord injury. Additionally, this work suggests that the retention of juvenile-like characteristics may be a widespread phenomenon in adult nervous systems.
Collapse
|
12
|
Nakanishi Y, Izumi M, Matsushita H, Koyama Y, Diez D, Takamatsu H, Koyama S, Nishide M, Naito M, Mizuno Y, Yamaguchi Y, Mae T, Noda Y, Nakaya K, Nojima S, Sugihara F, Okuzaki D, Ikawa M, Shimada S, Kang S, Kumanogoh A. Semaphorin 6D tunes amygdalar circuits for emotional, metabolic, and inflammatory outputs. Neuron 2024; 112:2955-2972.e9. [PMID: 39002542 DOI: 10.1016/j.neuron.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/20/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024]
Abstract
Regulated neural-metabolic-inflammatory responses are essential for maintaining physiological homeostasis. However, the molecular machinery that coordinates neural, metabolic, and inflammatory responses is largely unknown. Here, we show that semaphorin 6D (SEMA6D) coordinates anxiogenic, metabolic, and inflammatory outputs from the amygdala by maintaining synaptic homeostasis. Using genome-wide approaches, we identify SEMA6D as a pleiotropic gene for both psychiatric and metabolic traits in human. Sema6d deficiency increases anxiety in mice. When fed a high-fat diet, Sema6d-/- mice display attenuated obesity and enhanced myelopoiesis compared with control mice due to higher sympathetic activity via the β3-adrenergic receptor. Genetic manipulation and spatial and single-nucleus transcriptomics reveal that SEMA6D in amygdalar interneurons is responsible for regulating anxiogenic and autonomic responses. Mechanistically, SEMA6D is required for synaptic maturation and γ-aminobutyric acid transmission. These results demonstrate that SEMA6D is important for the normal functioning of the neural circuits in the amygdala, coupling emotional, metabolic, and inflammatory responses.
Collapse
Affiliation(s)
- Yoshimitsu Nakanishi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan; Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan
| | - Mayuko Izumi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan; Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan
| | - Hiroaki Matsushita
- Department of Advanced Clinical and Translational Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Discovery Pharmacology Department, Research Division, Chugai Pharmaceutical Co. Ltd., Kanagawa 247-8530, Japan
| | - Yoshihisa Koyama
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan; Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Diego Diez
- Quantitative Immunology Research Unit, WPI-IFReC, Osaka University, Osaka 565-0871, Japan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan
| | - Maiko Naito
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan
| | - Yumiko Mizuno
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan
| | - Yuta Yamaguchi
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan
| | - Tomoki Mae
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yu Noda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Kamon Nakaya
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Satoshi Nojima
- Department of Pathology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Fuminori Sugihara
- Laboratory of Biofunctional Imaging, WPI-IFReC, Osaka University, Osaka 565-0871, Japan
| | - Daisuke Okuzaki
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan; Laboratory of Human Immunology (Single Cell Genomics), WPI-IFReC, Osaka University, Osaka 565-0871, Japan; Genome Information Research Center, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0871, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan; Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka 565-0871, Japan
| | - Masahito Ikawa
- Department of Experimental Genome Research, RIMD, Osaka University, Osaka 565-0871, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka 565-0871, Japan
| | - Shoichi Shimada
- Department of Neuroscience and Cell Biology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Addiction Research Unit, Osaka Psychiatric Research Center, Osaka Psychiatric Medical Center, Osaka 541-8567, Japan
| | - Sujin Kang
- Laboratory of Immune Regulation, WPI-IFReC, Osaka University, Osaka 565-0871, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan.
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan; Department of Immunopathology, World Premier International Research Center Initiative Immunology Frontier Research Center (WPI-IFReC), Osaka University, Osaka 565-0871, Japan; Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Osaka 565-0871, Japan; Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Osaka 565-0871, Japan; Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Osaka 565-0871, Japan; Center for Advanced Modalities and DDS (CAMaD), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
13
|
Dong J, Zhu XN, Zeng PM, Cao DD, Yang Y, Hu J, Luo ZG. A hominoid-specific signaling axis regulating the tempo of synaptic maturation. Cell Rep 2024; 43:114548. [PMID: 39052482 DOI: 10.1016/j.celrep.2024.114548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 04/15/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
Human cortical neurons (hCNs) exhibit high dendritic complexity and synaptic density, and the maturation process is greatly protracted. However, the molecular mechanism governing these specific features remains unclear. Here, we report that the hominoid-specific gene TBC1D3 promotes dendritic arborization and protracts the pace of synaptogenesis. Ablation of TBC1D3 in induced hCNs causes reduction of dendritic growth and precocious synaptic maturation. Forced expression of TBC1D3 in the mouse cortex protracts synaptic maturation while increasing dendritic growth. Mechanistically, TBC1D3 functions via interaction with MICAL1, a monooxygenase that mediates oxidation of actin filament. At the early stage of differentiation, the TBC1D3/MICAL1 interaction in the cytosol promotes dendritic growth via F-actin oxidation and enhanced actin dynamics. At late stages, TBC1D3 escorts MICAL1 into the nucleus and downregulates the expression of genes related with synaptic maturation through interaction with the chromatin remodeling factor ATRX. Thus, this study delineates the molecular mechanisms underlying human neuron development.
Collapse
Affiliation(s)
- Jian Dong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Xiao-Na Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Peng-Ming Zeng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Dong-Dong Cao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yang Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Zhen-Ge Luo
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
14
|
Pritz MB. Interconnections between the dorsal thalamus and the basal nuclei in a reptile. Neurosci Lett 2024; 836:137894. [PMID: 38997083 DOI: 10.1016/j.neulet.2024.137894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 07/14/2024]
Abstract
Reciprocal connections between the thalamus and the cortex are one of the most characteristic features of forebrain organization in mammals. To date, this circuit has been documented only in turtles. However, reptiles, including turtles, have an additional path from the dorsal thalamus to the telencephalon. This terminates in a pallial structure known as the dorsal ventricular ridge. Yet, no reciprocal connection from the dorsal ventricular ridge to thalamic nuclei has been uncovered. Since axons from the thalamus pass through the basal nuclei on route to the dorsal ventricular ridge, the basal nuclei might be a source of reciprocal connections. Accordingly, the location and distribution of neurons after retrograde tracer placement into the dorsal thalamus were examined. Retrogradely labeled neurons in the basal nuclei were indeed found. One possibility to explain this observation is that connections with the dorsal ventricular ridge are present during development but later pruned during embryogenesis.
Collapse
Affiliation(s)
- Michael B Pritz
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; DENLABS, Draper, UT, USA.
| |
Collapse
|
15
|
Woo MS, Mayer C, Binkle-Ladisch L, Sonner JK, Rosenkranz SC, Shaposhnykov A, Rothammer N, Tsvilovskyy V, Lorenz SM, Raich L, Bal LC, Vieira V, Wagner I, Bauer S, Glatzel M, Conrad M, Merkler D, Freichel M, Friese MA. STING orchestrates the neuronal inflammatory stress response in multiple sclerosis. Cell 2024; 187:4043-4060.e30. [PMID: 38878778 DOI: 10.1016/j.cell.2024.05.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/08/2024] [Accepted: 05/16/2024] [Indexed: 07/28/2024]
Abstract
Inflammation-induced neurodegeneration is a defining feature of multiple sclerosis (MS), yet the underlying mechanisms remain unclear. By dissecting the neuronal inflammatory stress response, we discovered that neurons in MS and its mouse model induce the stimulator of interferon genes (STING). However, activation of neuronal STING requires its detachment from the stromal interaction molecule 1 (STIM1), a process triggered by glutamate excitotoxicity. This detachment initiates non-canonical STING signaling, which leads to autophagic degradation of glutathione peroxidase 4 (GPX4), essential for neuronal redox homeostasis and thereby inducing ferroptosis. Both genetic and pharmacological interventions that target STING in neurons protect against inflammation-induced neurodegeneration. Our findings position STING as a central regulator of the detrimental neuronal inflammatory stress response, integrating inflammation with glutamate signaling to cause neuronal cell death, and present it as a tractable target for treating neurodegeneration in MS.
Collapse
Affiliation(s)
- Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christina Mayer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Binkle-Ladisch
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sina C Rosenkranz
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Artem Shaposhnykov
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nicola Rothammer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Volodymyr Tsvilovskyy
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Svenja M Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas C Bal
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Wagner
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Doron Merkler
- Department of Pathology and Immunology, Division of Clinical Pathology, Faculty of Medicine, University and University Hospital of Geneva, Geneva, Switzerland
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
16
|
Nishiyama M, Kalambogias J, Imai F, Yang E, Lang S, de Nooij JC, Yoshida Y. Anatomical and functional analysis of the corticospinal tract in an FRDA mouse model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601178. [PMID: 39005321 PMCID: PMC11244874 DOI: 10.1101/2024.06.28.601178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Friedreich's ataxia (FRDA) is one of the most common hereditary ataxias. It is caused by a GAA repeat in the first intron of the FXN gene, which encodes an essential mitochondrial protein. Patients suffer from progressive motor dysfunction due to the degeneration of mechanoreceptive and proprioceptive neurons in dorsal root ganglia (DRG) and cerebellar dentate nucleus neurons, especially at early disease stages. Postmortem analyses of FRDA patients also indicate pathological changes in motor cortex including in the projection neurons that give rise to the cortical spinal tract (CST). Yet, it remains poorly understood how early in the disease cortical spinal neurons (CSNs) show these alterations, or whether CSN/CST pathology resembles the abnormalities observed in other tissues affected by FXN loss. To address these questions, we examined CSN driven motor behaviors and pathology in the YG8JR FRDA mouse model. We find that FRDA mice show impaired motor skills, exhibit significant reductions in CSN functional output, and, among other pathological changes, show abnormal mitochondrial distributions in CSN neurons and CST axonal tracts. Moreover, some of these alterations were observed as early as two months of age, suggesting that CSN/CST pathology may be an earlier event in FRDA disease than previously appreciated. These studies warrant a detailed mechanistic understanding of how FXN loss impacts CSN health and functionality.
Collapse
Affiliation(s)
- Misa Nishiyama
- Burke Neurological Institute, White Plains, New York, United States
| | - John Kalambogias
- Burke Neurological Institute, White Plains, New York, United States
- Department of Neurology, Columbia University, New York, NY, USA
| | - Fumiyasu Imai
- Burke Neurological Institute, White Plains, New York, United States
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
| | - Emily Yang
- Burke Neurological Institute, White Plains, New York, United States
| | - Sonia Lang
- Burke Neurological Institute, White Plains, New York, United States
| | | | - Yutaka Yoshida
- Burke Neurological Institute, White Plains, New York, United States
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, United States
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, Japan
| |
Collapse
|
17
|
Simon CM, Delestree N, Montes J, Gerstner F, Carranza E, Sowoidnich L, Buettner JM, Pagiazitis JG, Prat-Ortega G, Ensel S, Donadio S, Garcia JL, Kratimenos P, Chung WK, Sumner CJ, Weimer LH, Pirondini E, Capogrosso M, Pellizzoni L, De Vivo DC, Mentis GZ. Dysfunction of proprioceptive sensory synapses is a pathogenic event and therapeutic target in mice and humans with spinal muscular atrophy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.03.24308132. [PMID: 38883729 PMCID: PMC11177917 DOI: 10.1101/2024.06.03.24308132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by a varying degree of severity that correlates with the reduction of SMN protein levels. Motor neuron degeneration and skeletal muscle atrophy are hallmarks of SMA, but it is unknown whether other mechanisms contribute to the spectrum of clinical phenotypes. Here, through a combination of physiological and morphological studies in mouse models and SMA patients, we identify dysfunction and loss of proprioceptive sensory synapses as key signatures of SMA pathology. We demonstrate that SMA patients exhibit impaired proprioception, and their proprioceptive sensory synapses are dysfunctional as measured by the neurophysiological test of the Hoffmann reflex (H-reflex). We further show that loss of excitatory afferent synapses and altered potassium channel expression in SMA motor neurons are conserved pathogenic events found in both severely affected patients and mouse models. Lastly, we report that improved motor function and fatigability in ambulatory SMA patients and mouse models treated with SMN-inducing drugs correlate with increased function of sensory-motor circuits that can be accurately captured by the H-reflex assay. Thus, sensory synaptic dysfunction is a clinically relevant event in SMA, and the H-reflex is a suitable assay to monitor disease progression and treatment efficacy of motor circuit pathology.
Collapse
Affiliation(s)
- CM Simon
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - N Delestree
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
| | - J Montes
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Rehabilitation and Regenerative Medicine, Columbia University, NY, USA
| | - F Gerstner
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - E Carranza
- Depts. Physical Medicine & Rehabilitation & Bioengineering, University of Pittsburgh, PA, USA
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
| | - L Sowoidnich
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - JM Buettner
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig, Germany
| | - JG Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
| | - G Prat-Ortega
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - S Ensel
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - S Donadio
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - JL Garcia
- Dept. of Neurology, Columbia University, NY, USA
| | - P Kratimenos
- Center for Neuroscience Research, Children’s National Res. Institute, Washington, DC, USA
- Dept. of Pediatrics, G Washington Univ. Sch. of Medicine, Washington, DC, USA
| | - WK Chung
- Dept. of Pediatrics, Boston Children’s Hospital and Harvard Medical School, Boston, MA USA
| | - CJ Sumner
- Depts. of Neurology, Neuroscience and Genetic Medicine, Johns Hopkins University School of Medicine, MD, USA
| | - LH Weimer
- Dept. of Neurology, Columbia University, NY, USA
| | - E Pirondini
- Depts. Physical Medicine & Rehabilitation & Bioengineering, University of Pittsburgh, PA, USA
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
| | - M Capogrosso
- Rehab and Neural Engineering Labs, University of Pittsburgh, PA, USA
- Depts. of Neurological Surgery & Bioengineering, University of Pittsburgh, PA, USA
| | - L Pellizzoni
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
- Dept. of Pathology and Cell Biology, Columbia University, NY, USA
| | - DC De Vivo
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
| | - GZ Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, NY, USA
- Dept. of Neurology, Columbia University, NY, USA
- Dept. of Pathology and Cell Biology, Columbia University, NY, USA
| |
Collapse
|
18
|
Lemon R. The Corticospinal System and Amyotrophic Lateral Sclerosis: IFCN handbook chapter. Clin Neurophysiol 2024; 160:56-67. [PMID: 38401191 DOI: 10.1016/j.clinph.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/23/2023] [Accepted: 02/03/2024] [Indexed: 02/26/2024]
Abstract
Corticospinal neurons located in motor areas of the cerebral neocortex project corticospinal axons which synapse with the spinal network; a parallel corticobulbar system projects to the cranial motor network and to brainstem motor pathways. The primate corticospinal system has a widespread cortical origin and an extensive range of different fibre diameters, including thick, fast-conducting axons. Direct cortico-motoneuronal (CM) projections from the motor cortex to arm and hand alpha motoneurons are a recent evolutionary feature, that is well developed in dexterous primates and particularly in humans. Many of these projections originate from the caudal subdivision of area 4 ('new' M1: primary motor cortex). They arise from corticospinal neurons of varied soma size, including those with fast- and relatively slow-conducting axons. This CM system has been shown to be involved in the control of skilled movements, carried out with fractionation of the distal extremities and at low force levels. During movement, corticospinal neurons are activated quite differently from 'lower' motoneurons, and there is no simple or fixed functional relationship between a so-called 'upper' motoneuron and its target lower motoneuron. There are key differences in the organisation and function of the corticospinal and CM system in primates versus non-primates, such as rodents. These differences need to be recognized when making the choice of animal model for understanding disorders such as amyotrophic lateral sclerosis (ALS). In this neurodegenerative brain disease there is a selective loss of fast-conducting corticospinal axons, and their synaptic connections, and this is reflected in responses to non-invasive cortical stimuli and measures of cortico-muscular coherence. The loss of CM connections influencing distal limb muscles results in a differential loss of muscle strength or 'split-hand' phenotype. Importantly, there is also a unique impairment in the coordination of skilled hand tasks that require fractionation of digit movement. Scores on validated tests of skilled hand function could be used to assess disease progression.
Collapse
Affiliation(s)
- Roger Lemon
- Department of Clinical and Movement Sciences, Queen Square Institute of Neurology, UCL, London WC1N 3BG, UK.
| |
Collapse
|
19
|
Abstract
Brain development in humans is achieved through precise spatiotemporal genetic control, the mechanisms of which remain largely elusive. Recently, integration of technological advances in human stem cell-based modelling with genome editing has emerged as a powerful platform to establish causative links between genotypes and phenotypes directly in the human system. Here, we review our current knowledge of complex genetic regulation of each key step of human brain development through the lens of evolutionary specialization and neurodevelopmental disorders and highlight the use of human stem cell-derived 2D cultures and 3D brain organoids to investigate human-enriched features and disease mechanisms. We also discuss opportunities and challenges of integrating new technologies to reveal the genetic architecture of human brain development and disorders.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Yang F, Wang F, Ma X, Zhou M, Jiang S, Xu W. Longitudinal optogenetic mapping reveals enhanced motor control by the contralesional cortex after traumatic brain injury in mice. Exp Neurol 2023; 369:114546. [PMID: 37751813 DOI: 10.1016/j.expneurol.2023.114546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/13/2023] [Accepted: 09/21/2023] [Indexed: 09/28/2023]
Abstract
Traumatic brain injury (TBI) is a significant cause of human disability, and understanding its spontaneous recovery pattern after injury is critical for potential treatments. However, studies on the function of the contralesional cortex after TBI have mostly focused on acute-phase changes, and long-term dynamic changes in the control of the affected limb by the contralesional cortex are less understood. To unravel long-term adaptations in the contralesional cortex, we developed a mouse model of TBI and used longitudinal optogenetic motor mapping to observe the function of contralesional corticospinal neurons (CSNs) projecting to the unilateral seventh cervical (C7) segment of the spinal cord. We injected a retrograde adeno-associated virus (AAV) expressing channelrhodopsin-2 to optogenetically stimulate and map the functional connections of the motor-sensory cortex. We validated the effectiveness of transcranial optogenetic stimulation for functional mapping and observed a general increase in the control of the affected limb by the contralesional cortex over time. Using retrograde labeling techniques, we showed that TBI does not affect the distribution of C7-CSNs but alters their function, and the labeled CSNs are concentrated in the caudal and rostral forelimb areas. Our findings provide new insights into harnessing contralesional cortical plasticity to improve treatment for affected limbs. This study sheds light on the long-term adaptations in the contralesional cortex after TBI, paving the way for potential clinical applications of optogenetic stimulation to improve motor control and rehabilitation outcomes.
Collapse
Affiliation(s)
- Fangjing Yang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Fei Wang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China; The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China; Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital, Fudan University, Shanghai, China
| | - Xingyi Ma
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Mingjie Zhou
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Su Jiang
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wendong Xu
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, China; The National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China; Department of Hand and Upper Extremity Surgery, Jing'an District Central Hospital, Fudan University, Shanghai, China; Institutes of Brain Science, Fudan University, Shanghai, China; State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center of Brain Science, Fudan University, Shanghai, China; Co-innovation Center of Neuroregeneration, Nantong University,226000 Nantong, China; Research Unit of Synergistic Reconstruction of Upper and Lower Limbs After Brain Injury, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
21
|
Kogan E, Lu J, Zuo Y. Cortical circuit dynamics underlying motor skill learning: from rodents to humans. Front Mol Neurosci 2023; 16:1292685. [PMID: 37965043 PMCID: PMC10641381 DOI: 10.3389/fnmol.2023.1292685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/16/2023] Open
Abstract
Motor learning is crucial for the survival of many animals. Acquiring a new motor skill involves complex alterations in both local neural circuits in many brain regions and long-range connections between them. Such changes can be observed anatomically and functionally. The primary motor cortex (M1) integrates information from diverse brain regions and plays a pivotal role in the acquisition and refinement of new motor skills. In this review, we discuss how motor learning affects the M1 at synaptic, cellular, and circuit levels. Wherever applicable, we attempt to relate and compare findings in humans, non-human primates, and rodents. Understanding the underlying principles shared by different species will deepen our understanding of the neurobiological and computational basis of motor learning.
Collapse
Affiliation(s)
| | | | - Yi Zuo
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
22
|
Tsuboguchi S, Nakamura Y, Ishihara T, Kato T, Sato T, Koyama A, Mori H, Koike Y, Onodera O, Ueno M. TDP-43 differentially propagates to induce antero- and retrograde degeneration in the corticospinal circuits in mouse focal ALS models. Acta Neuropathol 2023; 146:611-629. [PMID: 37555859 DOI: 10.1007/s00401-023-02615-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/22/2023] [Accepted: 07/15/2023] [Indexed: 08/10/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by TDP-43 inclusions in the cortical and spinal motor neurons. It remains unknown whether and how pathogenic TDP-43 spreads across neural connections to progress degenerative processes in the cortico-spinal motor circuitry. Here we established novel mouse ALS models that initially induced mutant TDP-43 inclusions in specific neuronal or cell types in the motor circuits, and investigated whether TDP-43 and relevant pathological processes spread across neuronal or cellular connections. We first developed ALS models that primarily induced TDP-43 inclusions in the corticospinal neurons, spinal motor neurons, or forelimb skeletal muscle, by using adeno-associated virus (AAV) expressing mutant TDP-43. We found that TDP-43 induced in the corticospinal neurons was transported along the axons anterogradely and transferred to the oligodendrocytes along the corticospinal tract (CST), coinciding with mild axon degeneration. In contrast, TDP-43 introduced in the spinal motor neurons did not spread retrogradely to the cortical or spinal neurons; however, it induced an extreme loss of spinal motor neurons and subsequent degeneration of neighboring spinal neurons, suggesting a degenerative propagation in a retrograde manner in the spinal cord. The intraspinal degeneration further led to severe muscle atrophy. Finally, TDP-43 induced in the skeletal muscle did not propagate pathological events to spinal neurons retrogradely. Our data revealed that mutant TDP-43 spread across neuro-glial connections anterogradely in the corticospinal pathway, whereas it exhibited different retrograde degenerative properties in the spinal circuits. This suggests that pathogenic TDP-43 may induce distinct antero- and retrograde mechanisms of degeneration in the motor system in ALS.
Collapse
Affiliation(s)
- Shintaro Tsuboguchi
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tomohiko Ishihara
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Taisuke Kato
- Department of Molecular Neuroscience, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tokiharu Sato
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan
| | - Akihide Koyama
- Division of Legal Medicine, Graduate School of Medicine and Dental Sciences, Niigata University, Niigata, Japan
| | - Hideki Mori
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Yuka Koike
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, 951-8585, Japan.
- Department of Molecular Neuroscience, Brain Research Institute, Niigata University, Niigata, Japan.
| | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata University, Niigata, Japan.
| |
Collapse
|
23
|
Gu Z, Matsuura K, Letelier A, Basista M, Craig C, Imai F, Yoshida Y. Axon Fasciculation, Mediated by Transmembrane Semaphorins, Is Critical for the Establishment of Segmental Specificity of Corticospinal Circuits. J Neurosci 2023; 43:5753-5768. [PMID: 37344234 PMCID: PMC10423052 DOI: 10.1523/jneurosci.0073-22.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 06/23/2023] Open
Abstract
Axon fasciculation is thought to be a critical step in neural circuit formation and function. Recent studies have revealed various molecular mechanisms that underlie axon fasciculation; however, the impacts of axon fasciculation, and its corollary, defasciculation, on neural circuit wiring remain unclear. Corticospinal (CS) neurons in the sensorimotor cortex project axons to the spinal cord to control skilled movements. In rodents, the axons remain tightly fasciculated in the brain and traverse the dorsal funiculus of the spinal cord. Here we show that plexinA1 (PlexA1) and plexinA3 (PlexA3) receptors are expressed by CS neurons, whereas their ligands, semaphorin-5A (Sema5A) and semaphorin-5B (Sema5B) are expressed in the medulla at the decussation site of CS axons to inhibit premature defasciculation of these axons. In the absence of Sema5A/5B-PlexA1/A3 signaling, some CS axons are prematurely defasciculated in the medulla of the brainstem, and those defasciculated CS axons aberrantly transverse in the spinal gray matter instead of the spinal dorsal funiculus. In the absence of Sema5A/Sema5B-PlexA1/A3 signaling, CS axons, which would normally innervate the lumbar spinal cord, are unbundled in the spinal gray matter, and prematurely innervate the cervical gray matter with reduced innervation of the lumbar gray matter. In both Sema5A/5B and PlexA1/A3 mutant mice (both sexes), stimulation of the hindlimb motor cortex aberrantly evokes robust forelimb muscle activation. Finally, Sema5A/5B and PlexA1/A3 mutant mice show deficits in skilled movements. These results suggest that proper fasciculation of CS axons is required for appropriate neural circuit wiring and ultimately affect the ability to perform skilled movements.SIGNIFICANCE STATEMENT Axon fasciculation is believed to be essential for neural circuit formation and function. However, whether and how defects in axon fasciculation affect the formation and function of neural circuits remain unclear. Here we examine whether the transmembrane proteins semaphorin-5A (Sema5A) and semaphorin-5B (Sema5B), and their receptors, plexinA1 (PlexA1) and plexinA3 (PlexA3) play roles in the development of corticospinal circuits. We find that Sema5A/Sema5B and PlexA1/A3 are required for proper axon fasciculation of corticospinal neurons. Furthermore, Sema5A/5B and PlexA1/A3 mutant mice show marked deficits in skilled motor behaviors. Therefore, these results strongly suggest that proper corticospinal axon fasciculation is required for the appropriate formation and functioning of corticospinal circuits in mice.
Collapse
Affiliation(s)
- Zirong Gu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Ken Matsuura
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| | | | - Mark Basista
- Burke Neurological Institute, White Plains, New York 10605
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Corey Craig
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
| | - Fumiyasu Imai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Burke Neurological Institute, White Plains, New York 10605
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229
- Burke Neurological Institute, White Plains, New York 10605
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, New York 10065
- Neural Circuit Unit, Okinawa Institute of Science and Technology Graduate University, Okinawa, 904-0495, Japan
| |
Collapse
|
24
|
Prieur DS, Francius C, Gaspar P, Mason CA, Rebsam A. Semaphorin-6D and Plexin-A1 Act in a Non-Cell-Autonomous Manner to Position and Target Retinal Ganglion Cell Axons. J Neurosci 2023; 43:5769-5778. [PMID: 37344233 PMCID: PMC10423046 DOI: 10.1523/jneurosci.0072-22.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 04/04/2023] [Accepted: 05/01/2023] [Indexed: 06/23/2023] Open
Abstract
Semaphorins and Plexins form ligand/receptor pairs that are crucial for a wide range of developmental processes from cell proliferation to axon guidance. The ability of semaphorins to act both as signaling receptors and ligands yields a multitude of responses. Here, we describe a novel role for Semaphorin-6D (Sema6D) and Plexin-A1 in the positioning and targeting of retinogeniculate axons. In Plexin-A1 or Sema6D mutant mice of either sex, the optic tract courses through, rather than along, the border of the dorsal lateral geniculate nucleus (dLGN), and some retinal axons ectopically arborize adjacent and lateral to the optic tract rather than defasciculating and entering the target region. We find that Sema6D and Plexin-A1 act together in a dose-dependent manner, as the number of the ectopic retinal projections is altered in proportion to the level of Sema6D or Plexin-A1 expression. Moreover, using retinal in utero electroporation of Sema6D or Plexin-A1 shRNA, we show that Sema6D and Plexin-A1 are both required in retinal ganglion cells for axon positioning and targeting. Strikingly, nonelectroporated retinal ganglion cell axons also mistarget in the tract region, indicating that Sema6D and Plexin-A1 can act non-cell-autonomously, potentially through axon-axon interactions. These data provide novel evidence for a dose-dependent and non-cell-autonomous role for Sema6D and Plexin-A1 in retinal axon organization in the optic tract and dLGN.SIGNIFICANCE STATEMENT Before innervating their central brain targets, retinal ganglion cell axons fasciculate in the optic tract and then branch and arborize in their target areas. Upon deletion of the guidance molecules Plexin-A1 or Semaphorin-6D, the optic tract becomes disorganized near and extends within the dorsal lateral geniculate nucleus. In addition, some retinal axons form ectopic aggregates within the defasciculated tract. Sema6D and Plexin-A1 act together as a receptor-ligand pair in a dose-dependent manner, and non-cell-autonomously, to produce this developmental aberration. Such a phenotype highlights an underappreciated role for axon guidance molecules in tract cohesion and appropriate defasciculation near, and arborization within, targets.
Collapse
Affiliation(s)
- Delphine S Prieur
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
| | - Cédric Francius
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
| | - Patricia Gaspar
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
| | - Carol A Mason
- Departments of Pathology and Cell Biology, Neuroscience, and Ophthalmology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027
| | - Alexandra Rebsam
- Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche-S 839, Paris, 75005, France
- Sorbonne Université, Paris, 75005, France
- Institut du Fer à Moulin, Paris, 75005, France
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, F-75012, France
| |
Collapse
|
25
|
Andersen J, Thom N, Shadrach JL, Chen X, Onesto MM, Amin ND, Yoon SJ, Li L, Greenleaf WJ, Müller F, Pașca AM, Kaltschmidt JA, Pașca SP. Single-cell transcriptomic landscape of the developing human spinal cord. Nat Neurosci 2023; 26:902-914. [PMID: 37095394 DOI: 10.1038/s41593-023-01311-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
Understanding spinal cord assembly is essential to elucidate how motor behavior is controlled and how disorders arise. The human spinal cord is exquisitely organized, and this complex organization contributes to the diversity and intricacy of motor behavior and sensory processing. But how this complexity arises at the cellular level in the human spinal cord remains unknown. Here we transcriptomically profiled the midgestation human spinal cord with single-cell resolution and discovered remarkable heterogeneity across and within cell types. Glia displayed diversity related to positional identity along the dorso-ventral and rostro-caudal axes, while astrocytes with specialized transcriptional programs mapped into white and gray matter subtypes. Motor neurons clustered at this stage into groups suggestive of alpha and gamma neurons. We also integrated our data with multiple existing datasets of the developing human spinal cord spanning 22 weeks of gestation to investigate the cell diversity over time. Together with mapping of disease-related genes, this transcriptomic mapping of the developing human spinal cord opens new avenues for interrogating the cellular basis of motor control in humans and guides human stem cell-based models of disease.
Collapse
Affiliation(s)
- Jimena Andersen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - Nicholas Thom
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | | | - Xiaoyu Chen
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Massimo Mario Onesto
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA
| | - Neal D Amin
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Se-Jin Yoon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA
| | - Li Li
- Department of Human Genetics, Emory University, Atlanta, GA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| | - Fabian Müller
- Department of Genetics, Stanford University, Stanford, CA, USA
- Center for Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Anca M Pașca
- Department of Pediatrics, Division of Neonatology, Stanford University, Stanford, CA, USA
| | | | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.
- Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford, CA, USA.
| |
Collapse
|
26
|
Vanderhaeghen P, Polleux F. Developmental mechanisms underlying the evolution of human cortical circuits. Nat Rev Neurosci 2023; 24:213-232. [PMID: 36792753 PMCID: PMC10064077 DOI: 10.1038/s41583-023-00675-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2023] [Indexed: 02/17/2023]
Abstract
The brain of modern humans has evolved remarkable computational abilities that enable higher cognitive functions. These capacities are tightly linked to an increase in the size and connectivity of the cerebral cortex, which is thought to have resulted from evolutionary changes in the mechanisms of cortical development. Convergent progress in evolutionary genomics, developmental biology and neuroscience has recently enabled the identification of genomic changes that act as human-specific modifiers of cortical development. These modifiers influence most aspects of corticogenesis, from the timing and complexity of cortical neurogenesis to synaptogenesis and the assembly of cortical circuits. Mutations of human-specific genetic modifiers of corticogenesis have started to be linked to neurodevelopmental disorders, providing evidence for their physiological relevance and suggesting potential relationships between the evolution of the human brain and its sensitivity to specific diseases.
Collapse
Affiliation(s)
- Pierre Vanderhaeghen
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.
- Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
| | - Franck Polleux
- Department of Neuroscience, Columbia University Medical Center, New York, NY, USA.
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA.
| |
Collapse
|
27
|
Arbat-Plana A, Bolívar S, Navarro X, Udina E, Alvarez FJ. Massive Loss of Proprioceptive Ia Synapses in Rat Spinal Motoneurons after Nerve Crush Injuries in the Postnatal Period. eNeuro 2023; 10:ENEURO.0436-22.2023. [PMID: 36759186 PMCID: PMC9948128 DOI: 10.1523/eneuro.0436-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/15/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
Peripheral nerve injuries (PNIs) induce the retraction from the ventral horn of the synaptic collaterals of Ia afferents injured in the nerve, effectively removing Ia synapses from α-motoneurons. The loss of Ia input impairs functional recovery and could explain, in part, better recovery after PNIs with better Ia synaptic preservation. Synaptic losses correlate with injury severity, speed, and efficiency of muscle reinnervation and requires ventral microglia activation. It is unknown whether this plasticity is age dependent. In neonates, axotomized motoneurons and sensory neurons undergo apoptosis, but after postnatal day 10 most survive. The goal of this study was to analyze vesicular glutamate transporter 1 (VGluT1)-labeled Ia synapses (which also include II afferents) after nerve crush in 10 day old rats, a PNI causing little Ia/II synapse loss in adult rats. We confirmed fast and efficient reinnervation of leg muscles; however, a massive number of VGluT1/Ia/II synapses were permanently lost. This synapse loss was similar to that after more severe nerve injuries involving full transection in adults. In adults, disappearance of ventrally directed Ia/II collaterals targeting α-motoneurons was associated with a prolonged microglia reaction and a CCR2 mechanism that included infiltration of CCR2 blood immune cells. By contrast, microgliosis after P10 injuries was fast, resolved in about a week, and there was no evidence of peripheral immune cell infiltration. We conclude that VGluT1/Ia/II synapse loss in young animals differs in mechanism, perhaps associated with higher microglia synaptic pruning activity at this age and results in larger losses after milder nerve injuries.
Collapse
Affiliation(s)
- Ariadna Arbat-Plana
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193 Bellaterra, Spain
- Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Sara Bolívar
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193 Bellaterra, Spain
- Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193 Bellaterra, Spain
| | - Esther Udina
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08193 Bellaterra, Spain
| | | |
Collapse
|
28
|
Gellért L, Luhmann HJ, Kilb W. Axonal connections between S1 barrel, M1, and S2 cortex in the newborn mouse. Front Neuroanat 2023; 17:1105998. [PMID: 36760662 PMCID: PMC9905141 DOI: 10.3389/fnana.2023.1105998] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
The development of functionally interconnected networks between primary (S1), secondary somatosensory (S2), and motor (M1) cortical areas requires coherent neuronal activity via corticocortical projections. However, the anatomical substrate of functional connections between S1 and M1 or S2 during early development remains elusive. In the present study, we used ex vivo carbocyanine dye (DiI) tracing in paraformaldehyde-fixed newborn mouse brain to investigate axonal projections of neurons in different layers of S1 barrel field (S1Bf), M1, and S2 toward the subplate (SP), a hub layer for sensory information transfer in the immature cortex. In addition, we performed extracellular recordings in neocortical slices to unravel the functional connectivity between these areas. Our experiments demonstrate that already at P0 neurons from the cortical plate (CP), layer 5/6 (L5/6), and the SP of both M1 and S2 send projections through the SP of S1Bf. Reciprocally, neurons from CP to SP of S1Bf send projections through the SP of M1 and S2. Electrophysiological recordings with multi-electrode arrays in cortical slices revealed weak, but functional synaptic connections between SP and L5/6 within and between S1 and M1. An even lower functional connectivity was observed between S1 and S2. In summary, our findings demonstrate that functional connections between SP and upper cortical layers are not confined to the same cortical area, but corticocortical connection between adjacent cortical areas exist already at the day of birth. Hereby, SP can integrate early cortical activity of M1, S1, and S2 and shape the development of sensorimotor integration at an early stage.
Collapse
|
29
|
Restoring After Central Nervous System Injuries: Neural Mechanisms and Translational Applications of Motor Recovery. Neurosci Bull 2022; 38:1569-1587. [DOI: 10.1007/s12264-022-00959-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/29/2022] [Indexed: 11/06/2022] Open
Abstract
AbstractCentral nervous system (CNS) injuries, including stroke, traumatic brain injury, and spinal cord injury, are leading causes of long-term disability. It is estimated that more than half of the survivors of severe unilateral injury are unable to use the denervated limb. Previous studies have focused on neuroprotective interventions in the affected hemisphere to limit brain lesions and neurorepair measures to promote recovery. However, the ability to increase plasticity in the injured brain is restricted and difficult to improve. Therefore, over several decades, researchers have been prompted to enhance the compensation by the unaffected hemisphere. Animal experiments have revealed that regrowth of ipsilateral descending fibers from the unaffected hemisphere to denervated motor neurons plays a significant role in the restoration of motor function. In addition, several clinical treatments have been designed to restore ipsilateral motor control, including brain stimulation, nerve transfer surgery, and brain–computer interface systems. Here, we comprehensively review the neural mechanisms as well as translational applications of ipsilateral motor control upon rehabilitation after CNS injuries.
Collapse
|
30
|
Sanders ZB, Fleming MK, Smejka T, Marzolla MC, Zich C, Rieger SW, Lührs M, Goebel R, Sampaio-Baptista C, Johansen-Berg H. Self-modulation of motor cortex activity after stroke: a randomized controlled trial. Brain 2022; 145:3391-3404. [PMID: 35960166 PMCID: PMC9586541 DOI: 10.1093/brain/awac239] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 06/01/2022] [Accepted: 06/20/2022] [Indexed: 11/14/2022] Open
Abstract
Real-time functional MRI neurofeedback allows individuals to self-modulate their ongoing brain activity. This may be a useful tool in clinical disorders that are associated with altered brain activity patterns. Motor impairment after stroke has previously been associated with decreased laterality of motor cortex activity. Here we examined whether chronic stroke survivors were able to use real-time fMRI neurofeedback to increase laterality of motor cortex activity and assessed effects on motor performance and on brain structure and function. We carried out a randomized, double-blind, sham-controlled trial (ClinicalTrials.gov: NCT03775915) in which 24 chronic stroke survivors with mild to moderate upper limb impairment experienced three training days of either Real (n = 12) or Sham (n = 12) neurofeedback. Assessments of brain structure, brain function and measures of upper-limb function were carried out before and 1 week after neurofeedback training. Additionally, measures of upper-limb function were repeated 1 month after neurofeedback training. Primary outcome measures were (i) changes in lateralization of motor cortex activity during movements of the stroke-affected hand throughout neurofeedback training days; and (ii) changes in motor performance of the affected limb on the Jebsen Taylor Test (JTT). Stroke survivors were able to use Real neurofeedback to increase laterality of motor cortex activity within (P = 0.019), but not across, training days. There was no group effect on the primary behavioural outcome measure, which was average JTT performance across all subtasks (P = 0.116). Secondary analysis found improvements in the performance of the gross motor subtasks of the JTT in the Real neurofeedback group compared to Sham (P = 0.010). However, there were no improvements on the Action Research Arm Test or the Upper Extremity Fugl-Meyer score (both P > 0.5). Additionally, decreased white-matter asymmetry of the corticospinal tracts was detected 1 week after neurofeedback training (P = 0.008), indicating that the tracts become more similar with Real neurofeedback. Changes in the affected corticospinal tract were positively correlated with participants neurofeedback performance (P = 0.002). Therefore, here we demonstrate that chronic stroke survivors are able to use functional MRI neurofeedback to self-modulate motor cortex activity in comparison to a Sham control, and that training is associated with improvements in gross hand motor performance and with white matter structural changes.
Collapse
Affiliation(s)
- Zeena-Britt Sanders
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| | - Melanie K Fleming
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| | - Tom Smejka
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| | - Marilien C Marzolla
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| | - Catharina Zich
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| | - Sebastian W Rieger
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| | - Michael Lührs
- Department of Cognitive Neuroscience, Maastricht University, 6229 EV Maastricht, The Netherlands
- Research Department, Brain Innovation B.V., 6229 EV Maastricht, The Netherlands
| | - Rainer Goebel
- Department of Cognitive Neuroscience, Maastricht University, 6229 EV Maastricht, The Netherlands
- Research Department, Brain Innovation B.V., 6229 EV Maastricht, The Netherlands
| | - Cassandra Sampaio-Baptista
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
- Institute of Neuroscience and Psychology, University of Glasgow, Glasgow G61 1QH, UK
| | - Heidi Johansen-Berg
- Wellcome Centre for Integrative Neuroimaging, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
31
|
Sinopoulou E, Rosenzweig ES, Conner JM, Gibbs D, Weinholtz CA, Weber JL, Brock JH, Nout-Lomas YS, Royz E, Takashima Y, Biane JS, Kumamaru H, Havton LA, Beattie MS, Bresnahan JC, Tuszynski MH. Rhesus macaque versus rat divergence in the corticospinal projectome. Neuron 2022; 110:2970-2983.e4. [PMID: 35917818 PMCID: PMC9509478 DOI: 10.1016/j.neuron.2022.07.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 04/14/2022] [Accepted: 07/06/2022] [Indexed: 01/14/2023]
Abstract
We used viral intersectional tools to map the entire projectome of corticospinal neurons associated with fine distal forelimb control in Fischer 344 rats and rhesus macaques. In rats, we found an extraordinarily diverse set of collateral projections from corticospinal neurons to 23 different brain and spinal regions. Remarkably, the vast weighting of this "motor" projection was to sensory systems in both the brain and spinal cord, confirmed by optogenetic and transsynaptic viral intersectional tools. In contrast, rhesus macaques exhibited far heavier and narrower weighting of corticospinal outputs toward spinal and brainstem motor systems. Thus, corticospinal systems in macaques primarily constitute a final output system for fine motor control, whereas this projection in rats exerts a multi-modal integrative role that accesses far broader CNS regions. Unique structural-functional correlations can be achieved by mapping and quantifying a single neuronal system's total axonal output and its relative weighting across CNS targets.
Collapse
Affiliation(s)
- Eleni Sinopoulou
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Ephron S Rosenzweig
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - James M Conner
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Daniel Gibbs
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Chase A Weinholtz
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Janet L Weber
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - John H Brock
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Veterans Administration Medical Center, La Jolla, CA, USA
| | - Yvette S Nout-Lomas
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Eric Royz
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Yoshio Takashima
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Jeremy S Biane
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Hiromi Kumamaru
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Leif A Havton
- Departments of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Veterans Administration Medical Center, Bronx, NY, USA
| | - Michael S Beattie
- Department of Neurosurgery, University of California, San Francisco, CA, USA
| | | | - Mark H Tuszynski
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA; Veterans Administration Medical Center, La Jolla, CA, USA.
| |
Collapse
|
32
|
Chen B, Li F, Jia B, So KF, Wei JA, Liu Y, Qu Y, Zhou L. Celsr3 Inactivation in the Brainstem Impairs Rubrospinal Tract Development and Mouse Behaviors in Motor Coordination and Mechanic-Induced Response. Mol Neurobiol 2022; 59:5179-5192. [PMID: 35678978 PMCID: PMC9363480 DOI: 10.1007/s12035-022-02910-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022]
Abstract
Inactivation of Celsr3 in the forebrain results in defects of longitudinal axonal tracts such as the corticospinal tract. In this study, we inactivated Celsr3 in the brainstem using En1-Cre mice (Celsr3 cKO) and analyzed axonal and behavioral phenotypes. Celsr3 cKO animals showed an 83% reduction of rubrospinal axons and 30% decrease of corticospinal axons in spinal segments, associated with increased branching of dopaminergic fibers in the ventral horn. Decreases of spinal motoneurons, neuromuscular junctions, and electromyographic signal amplitude of the biceps were also found in mutant animals. Mutant mice had impaired motor coordination and defective response to heavy mechanical stimulation, but no disability in walking and food pellet handling. Transsynaptic tracing demonstrated that rubrospinal axons synapse on spinal neurons in the deep layer of the dorsal horn, and mechanical stimulation of hindpaws induced strong calcium signal of red nuclei in control mice, which was less prominent in mutant mice. In conclusion, Celsr3 regulates development of spinal descending axons and the motor network in cell and non-cell autonomous manners, and the maturation of the rubrospinal system is required for motor coordination and response to mechanical stimulation.
Collapse
Affiliation(s)
- Boli Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Jinan University, Huangpu Avenue West 601, Guangzhou, 510632, People's Republic of China
| | - Fuxiang Li
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Jinan University, Huangpu Avenue West 601, Guangzhou, 510632, People's Republic of China
| | - Bin Jia
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Jinan University, Huangpu Avenue West 601, Guangzhou, 510632, People's Republic of China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Jinan University, Huangpu Avenue West 601, Guangzhou, 510632, People's Republic of China
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266071, Shandong, People's Republic of China
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical, Neuroscience Institute of Jinan University, Guangzhou, 510632, People's Republic of China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, People's Republic of China
- Co-Innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, People's Republic of China
| | - Ji-An Wei
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yuchu Liu
- School of Biological Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Yibo Qu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Jinan University, Huangpu Avenue West 601, Guangzhou, 510632, People's Republic of China
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, People's Republic of China
| | - Libing Zhou
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Jinan University, Huangpu Avenue West 601, Guangzhou, 510632, People's Republic of China.
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266071, Shandong, People's Republic of China.
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical, Neuroscience Institute of Jinan University, Guangzhou, 510632, People's Republic of China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, People's Republic of China.
- Co-Innovation Center of Neuroregeneration, Nantong University, Jiangsu, People's Republic of China.
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
33
|
Zschüntzsch J, Meyer S, Shahriyari M, Kummer K, Schmidt M, Kummer S, Tiburcy M. The Evolution of Complex Muscle Cell In Vitro Models to Study Pathomechanisms and Drug Development of Neuromuscular Disease. Cells 2022; 11:1233. [PMID: 35406795 PMCID: PMC8997482 DOI: 10.3390/cells11071233] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/25/2022] [Accepted: 03/31/2022] [Indexed: 12/04/2022] Open
Abstract
Many neuromuscular disease entities possess a significant disease burden and therapeutic options remain limited. Innovative human preclinical models may help to uncover relevant disease mechanisms and enhance the translation of therapeutic findings to strengthen neuromuscular disease precision medicine. By concentrating on idiopathic inflammatory muscle disorders, we summarize the recent evolution of the novel in vitro models to study disease mechanisms and therapeutic strategies. A particular focus is laid on the integration and simulation of multicellular interactions of muscle tissue in disease phenotypes in vitro. Finally, the requirements of a neuromuscular disease drug development workflow are discussed with a particular emphasis on cell sources, co-culture systems (including organoids), functionality, and throughput.
Collapse
Affiliation(s)
- Jana Zschüntzsch
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Stefanie Meyer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Mina Shahriyari
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Karsten Kummer
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
| | - Matthias Schmidt
- Department of Neurology, University Medical Center Goettingen, 37075 Goettingen, Germany; (S.M.); (K.K.); (M.S.)
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| | - Susann Kummer
- Risk Group 4 Pathogens–Stability and Persistence, Biosafety Level-4 Laboratory, Center for Biological Threats and Special Pathogens, Robert Koch Institute, 13353 Berlin, Germany;
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, 37075 Goettingen, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Goettingen, Germany
| |
Collapse
|
34
|
Moya MV, Kim RD, Rao MN, Cotto BA, Pickett SB, Sferrazza CE, Heintz N, Schmidt EF. Unique molecular features and cellular responses differentiate two populations of motor cortical layer 5b neurons in a preclinical model of ALS. Cell Rep 2022; 38:110556. [PMID: 35320722 PMCID: PMC9059890 DOI: 10.1016/j.celrep.2022.110556] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 01/31/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Many neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS), lead to the selective degeneration of discrete cell types in the CNS despite the ubiquitous expression of many genes linked to disease. Therapeutic advancement depends on understanding the unique cellular adaptations that underlie pathology of vulnerable cells in the context of disease-causing mutations. Here, we employ bacTRAP molecular profiling to elucidate cell type-specific molecular responses of cortical upper motor neurons in a preclinical ALS model. Using two bacTRAP mouse lines that label distinct vulnerable or resilient projection neuron populations in motor cortex, we show that the regulation of oxidative phosphorylation (Oxphos) pathways is a common response in both cell types. However, differences in the baseline expression of genes involved in Oxphos and the handling of reactive oxygen species likely lead to the selective degeneration of the vulnerable cells. These results provide a framework to identify cell-type-specific processes in neurodegenerative disease. Moya et al. use bacTRAP mouse lines to characterize two highly related subpopulations of layer 5b projection neurons in motor cortex that are differentially susceptible to neurodegeneration in the SOD1-G93A mouse model of ALS. They identify the regulation of genes involved in bioenergetics as a key factor regulating susceptibility.
Collapse
Affiliation(s)
- Maria V Moya
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA
| | - Rachel D Kim
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA
| | - Meghana N Rao
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA
| | - Bianca A Cotto
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA
| | - Sarah B Pickett
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA
| | - Caroline E Sferrazza
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA
| | - Nathaniel Heintz
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Eric F Schmidt
- Laboratory of Molecular Biology, The Rockefeller University, 1230 York Avenue, Box 260, New York, NY 10065, USA.
| |
Collapse
|
35
|
Tocco C, Øvsthus M, Bjaalie JG, Leergaard TB, Studer M. The topography of corticopontine projections is controlled by postmitotic expression of the area-mapping gene Nr2f1. Development 2022; 149:274658. [PMID: 35262177 PMCID: PMC8959144 DOI: 10.1242/dev.200026] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 12/20/2021] [Indexed: 12/27/2022]
Abstract
Axonal projections from layer V neurons of distinct neocortical areas are topographically organized into discrete clusters within the pontine nuclei during the establishment of voluntary movements. However, the molecular determinants controlling corticopontine connectivity are insufficiently understood. Here, we show that an intrinsic cortical genetic program driven by Nr2f1 graded expression is directly implicated in the organization of corticopontine topographic mapping. Transgenic mice lacking cortical expression of Nr2f1 and exhibiting areal organization defects were used as model systems to investigate the arrangement of corticopontine projections. By combining three-dimensional digital brain atlas tools, Cre-dependent mouse lines and axonal tracing, we show that Nr2f1 expression in postmitotic neurons spatially and temporally controls somatosensory topographic projections, whereas expression in progenitor cells influences the ratio between corticopontine and corticospinal fibres passing the pontine nuclei. We conclude that cortical gradients of area-patterning genes are directly implicated in the establishment of a topographic somatotopic mapping from the cortex onto pontine nuclei. Summary: Cortical gradient expression of the area patterning gene Nr2f1 spatially and temporally controls corticopontine topographic connectivity in layer V projection neurons.
Collapse
Affiliation(s)
- Chiara Tocco
- University Côte d'Azur, CNRS, Inserm, iBV, Nice 06108, France
| | - Martin Øvsthus
- Institute of Basic Medical Sciences, University of Oslo, Oslo N-0317, Norway
| | - Jan G Bjaalie
- Institute of Basic Medical Sciences, University of Oslo, Oslo N-0317, Norway
| | - Trygve B Leergaard
- Institute of Basic Medical Sciences, University of Oslo, Oslo N-0317, Norway
| | - Michèle Studer
- University Côte d'Azur, CNRS, Inserm, iBV, Nice 06108, France
| |
Collapse
|
36
|
cis-Regulatory changes in locomotor genes are associated with the evolution of burrowing behavior. Cell Rep 2022; 38:110360. [PMID: 35172153 DOI: 10.1016/j.celrep.2022.110360] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/24/2021] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
How evolution modifies complex, innate behaviors is largely unknown. Divergence in many morphological traits, and some behaviors, is linked to cis-regulatory changes in gene expression. Given this, we compare brain gene expression of two interfertile sister species of Peromyscus mice that show large and heritable differences in burrowing behavior. Species-level differential expression and allele-specific expression in F1 hybrids indicate a preponderance of cis-regulatory divergence, including many genes whose cis-regulation is affected by burrowing behavior. Genes related to locomotor coordination show the strongest signals of lineage-specific selection on burrowing-induced cis-regulatory changes. Furthermore, genetic markers closest to these candidate genes associate with variation in burrow shape in a genetic cross, suggesting an enrichment for loci affecting burrowing behavior near these candidate locomotor genes. Our results provide insight into how cis-regulated gene expression can depend on behavioral context and how this dynamic regulatory divergence between species may contribute to behavioral evolution.
Collapse
|
37
|
Laliberte AM, Farah C, Steiner KR, Tariq O, Bui TV. Changes in Sensorimotor Connectivity to dI3 Interneurons in Relation to the Postnatal Maturation of Grasping. Front Neural Circuits 2022; 15:768235. [PMID: 35153680 PMCID: PMC8828486 DOI: 10.3389/fncir.2021.768235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/31/2021] [Indexed: 11/23/2022] Open
Abstract
Primitive reflexes are evident shortly after birth. Many of these reflexes disappear during postnatal development as part of the maturation of motor control. This study investigates the changes of connectivity related to sensory integration by spinal dI3 interneurons during the time in which the palmar grasp reflex gradually disappears in postnatal mice pups. Our results reveal an increase in GAD65/67-labeled terminals to perisomatic Vglut1-labeled sensory inputs contacting cervical and lumbar dI3 interneurons between postnatal day 3 and day 25. In contrast, there were no changes in the number of perisomatic Vglut1-labeled sensory inputs to lumbar and cervical dI3 interneurons other than a decrease between postnatal day 15 and day 25. Changes in postsynaptic GAD65/67-labeled inputs to dI3 interneurons were inconsistent with a role in the sustained loss of the grasp reflex. These results suggest a possible link between the maturation of hand grasp during postnatal development and increased presynaptic inhibition of sensory inputs to dI3 interneurons.
Collapse
Affiliation(s)
- Alex M. Laliberte
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Carl Farah
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Kyra R. Steiner
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Omar Tariq
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Tuan V. Bui
- Brain and Mind Research Institute, Department of Biology, University of Ottawa, Ottawa, ON, Canada
- *Correspondence: Tuan V. Bui
| |
Collapse
|
38
|
Schmidt ERE, Polleux F. Genetic Mechanisms Underlying the Evolution of Connectivity in the Human Cortex. Front Neural Circuits 2022; 15:787164. [PMID: 35069126 PMCID: PMC8777274 DOI: 10.3389/fncir.2021.787164] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/09/2021] [Indexed: 12/22/2022] Open
Abstract
One of the most salient features defining modern humans is our remarkable cognitive capacity, which is unrivaled by any other species. Although we still lack a complete understanding of how the human brain gives rise to these unique abilities, the past several decades have witnessed significant progress in uncovering some of the genetic, cellular, and molecular mechanisms shaping the development and function of the human brain. These features include an expansion of brain size and in particular cortical expansion, distinct physiological properties of human neurons, and modified synaptic development. Together they specify the human brain as a large primate brain with a unique underlying neuronal circuit architecture. Here, we review some of the known human-specific features of neuronal connectivity, and we outline how novel insights into the human genome led to the identification of human-specific genetic modifiers that played a role in the evolution of human brain development and function. Novel experimental paradigms are starting to provide a framework for understanding how the emergence of these human-specific genomic innovations shaped the structure and function of neuronal circuits in the human brain.
Collapse
Affiliation(s)
- Ewoud R. E. Schmidt
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, United States
- *Correspondence: Ewoud R. E. Schmidt
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY, United States
- Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, United States
- Kavli Institute for Brain Science, Columbia University, New York, NY, United States
- Franck Polleux
| |
Collapse
|
39
|
Spinal cord representation of motor cortex plasticity reflects corticospinal tract LTP. Proc Natl Acad Sci U S A 2021; 118:2113192118. [PMID: 34934000 PMCID: PMC8719859 DOI: 10.1073/pnas.2113192118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2021] [Indexed: 11/18/2022] Open
Abstract
Although it is well known that activity-dependent motor cortex (MCX) plasticity produces long-term potentiation (LTP) of local cortical circuits, leading to enhanced muscle function, the effects on the corticospinal projection to spinal neurons has not yet been thoroughly studied. Here, we investigate a spinal locus for corticospinal tract (CST) plasticity in anesthetized rats using multichannel recording of motor-evoked, intraspinal local field potentials (LFPs) at the sixth cervical spinal cord segment. We produced LTP by intermittent theta burst electrical stimulation (iTBS) of the wrist area of MCX. Approximately 3 min of MCX iTBS potentiated the monosynaptic excitatory LFP recorded within the CST termination field in the dorsal horn and intermediate zone for at least 15 min after stimulation. Ventrolaterally, in the spinal cord gray matter, which is outside the CST termination field in rats, iTBS potentiated an oligosynaptic negative LFP that was localized to the wrist muscle motor pool. Spinal LTP remained robust, despite pharmacological blockade of iTBS-induced LTP within MCX using MK801, showing that activity-dependent spinal plasticity can be induced without concurrent MCX LTP. Pyramidal tract iTBS, which preferentially activates the CST, also produced significant spinal LTP, indicating the capacity for plasticity at the CST-spinal interneuron synapse. Our findings show CST monosynaptic LTP in spinal interneurons and demonstrate that spinal premotor circuits are capable of further modifying descending MCX control signals in an activity-dependent manner.
Collapse
|
40
|
Belyk M, Eichert N, McGettigan C. A dual larynx motor networks hypothesis. Philos Trans R Soc Lond B Biol Sci 2021; 376:20200392. [PMID: 34719252 PMCID: PMC8558777 DOI: 10.1098/rstb.2020.0392] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 01/14/2023] Open
Abstract
Humans are vocal modulators par excellence. This ability is supported in part by the dual representation of the laryngeal muscles in the motor cortex. Movement, however, is not the product of motor cortex alone but of a broader motor network. This network consists of brain regions that contain somatotopic maps that parallel the organization in motor cortex. We therefore present a novel hypothesis that the dual laryngeal representation is repeated throughout the broader motor network. In support of the hypothesis, we review existing literature that demonstrates the existence of network-wide somatotopy and present initial evidence for the hypothesis' plausibility. Understanding how this uniquely human phenotype in motor cortex interacts with broader brain networks is an important step toward understanding how humans evolved the ability to speak. We further suggest that this system may provide a means to study how individual components of the nervous system evolved within the context of neuronal networks. This article is part of the theme issue 'Voice modulation: from origin and mechanism to social impact (Part I)'.
Collapse
Affiliation(s)
- Michel Belyk
- Department of Speech Hearing and Phonetic Sciences, University College London, London WC1N 1PJ, UK
- Department of Psychology, Edge Hill University, Ormskirk, L39 4QP, UK
| | - Nicole Eichert
- Wellcome Centre for Integrative Neuroimaging, Centre for Functional MRI of the Brain (FMRIB), Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Carolyn McGettigan
- Department of Speech Hearing and Phonetic Sciences, University College London, London WC1N 1PJ, UK
| |
Collapse
|
41
|
Ohno T, Fukuda S, Murabe N, Niido M, Sakurai M. Temporal Course of Transient Direct Corticomotoneuronal Connections during Development in Rodents. Neuroscience 2021; 478:89-99. [PMID: 34534634 DOI: 10.1016/j.neuroscience.2021.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 11/28/2022]
Abstract
We previously observed in rodents that during the 2nd postnatal week corticospinal axons make monosynaptic connections with motoneurons. Prior to that finding, it had been believed that such contacts only occur in higher primates. Although an in vitro electrophysiological study is prerequisite for studying the developmental time course of synaptic connections, the technical difficulty of reliably recording synaptic responses from spinal motoneurons in animals over 2 weeks old hampered the study. Instead, we used retrograde transsynaptic labeling with a genetically modified rabies virus to confirm the presence of direct corticomotoneuronal connections at an early developmental stage and to show that these connections were subsequently eliminated. However, determination of an accurate elimination time course and quantitative evaluation of synaptic connectivity cannot be achieved through viral-tracing experiments. For the present study, we improved the slice preparation procedure and maintenance of slice viability, which enabled us to record postsynaptic responses using the whole cell patch-clamp technique from retrogradely labeled forearm motoneurons up until postnatal week 7. We examined the extent of corticomotoneuronal monosynaptic connections and studied the time course of their accumulation and loss. Positive ratios of monosynaptic corticomotoneuronal EPSCs increased from P6 to P8 and then plateaued (P8-P13: 65%). Thereafter, the monosynaptic connections declined until P21, at which time they were no longer detected. The time course of the falling phase and elimination was confirmed by experiments using optogenetic stimulation. The timing of the elimination fell within the same range (P18-22) estimated in our earlier study using retrograde transsynaptic labeling.
Collapse
Affiliation(s)
- Takae Ohno
- Department of Physiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Satoshi Fukuda
- Department of Physiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Naoyuki Murabe
- Department of Physiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Mizuho Niido
- Department of Physiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan.
| | - Masaki Sakurai
- Department of Physiology, Teikyo University School of Medicine, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan.
| |
Collapse
|
42
|
Baxter DE, Allinson LM, Al Amri WS, Poulter JA, Pramanik A, Thorne JL, Verghese ET, Hughes TA. MiR-195 and Its Target SEMA6D Regulate Chemoresponse in Breast Cancer. Cancers (Basel) 2021; 13:cancers13235979. [PMID: 34885090 PMCID: PMC8656586 DOI: 10.3390/cancers13235979] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/23/2021] [Accepted: 11/26/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND poor prognosis primary breast cancers are typically treated with cytotoxic chemotherapy. However, recurrences remain relatively common even after this aggressive therapy. Comparison of matched tumours pre- and post-chemotherapy can allow identification of molecular characteristics of therapy resistance and thereby potentially aid discovery of novel predictive markers or targets for chemosensitisation. Through this comparison, we aimed to identify microRNAs associated with chemoresistance, define microRNA target genes, and assess targets as predictors of chemotherapy response. METHODS cancer cells were laser microdissected from matched breast cancer tissues pre- and post-chemotherapy from estrogen receptor positive/HER2 negative breast cancers showing partial responses to epirubicin/cyclophosphamide chemotherapy (n = 5). MicroRNA expression was profiled using qPCR arrays. MicroRNA/mRNA expression was manipulated in estrogen receptor positive/HER2 negative breast cancer cell lines (MCF7 and MDA-MB-175 cells) with mimics, inhibitors or siRNAs, and chemoresponse was assessed using MTT and colony forming survival assays. MicroRNA targets were identified by RNA-sequencing of microRNA mimic pull-downs, and comparison of these with mRNAs containing predicted microRNA binding sites. Survival correlations were tested using the METABRIC expression dataset (n = 1979). RESULTS miR-195 and miR-26b were consistently up-regulated after therapy, and changes in their expression in cell lines caused significant differences in chemotherapy sensitivity, in accordance with up-regulation driving resistance. SEMA6D was defined and confirmed as a target of the microRNAs. Reduced SEMA6D expression was significantly associated with chemoresistance, in accordance with SEMA6D being a down-stream effector of the microRNAs. Finally, low SEMA6D expression in breast cancers was significantly associated with poor survival after chemotherapy, but not after other therapies. CONCLUSIONS microRNAs and their targets influence chemoresponse, allowing the identification of SEMA6D as a predictive marker for chemotherapy response that could be used to direct therapy or as a target in chemosensitisation strategies.
Collapse
Affiliation(s)
- Diana E. Baxter
- School of Medicine, University of Leeds, Leeds LS9 7TF, UK; (D.E.B.); (J.A.P.); (A.P.)
- Cancer Research UK Manchester Institute, University of Manchester, Manchester SK10 4TG, UK
| | - Lisa M. Allinson
- Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4AD, UK;
| | - Waleed S. Al Amri
- Department of Histopathology and Cytopathology, The Royal Hospital, Muscat, Oman;
| | - James A. Poulter
- School of Medicine, University of Leeds, Leeds LS9 7TF, UK; (D.E.B.); (J.A.P.); (A.P.)
| | - Arindam Pramanik
- School of Medicine, University of Leeds, Leeds LS9 7TF, UK; (D.E.B.); (J.A.P.); (A.P.)
| | - James L. Thorne
- School of Food Science and Nutrition, University of Leeds, Leeds LS2 9JT, UK;
| | - Eldo T. Verghese
- Department of Histopathology, St. James’s University Hospital, Leeds LS9 7JX, UK;
| | - Thomas A. Hughes
- School of Medicine, University of Leeds, Leeds LS9 7TF, UK; (D.E.B.); (J.A.P.); (A.P.)
- Correspondence:
| |
Collapse
|
43
|
Del Cerro P, Rodríguez-De-Lope Á, Collazos-Castro JE. The Cortical Motor System in the Domestic Pig: Origin and Termination of the Corticospinal Tract and Cortico-Brainstem Projections. Front Neuroanat 2021; 15:748050. [PMID: 34790101 PMCID: PMC8591036 DOI: 10.3389/fnana.2021.748050] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
The anatomy of the cortical motor system and its relationship to motor repertoire in artiodactyls is for the most part unknown. We studied the origin and termination of the corticospinal tract (CST) and cortico-brainstem projections in domestic pigs. Pyramidal neurons were retrogradely labeled by injecting aminostilbamidine in the spinal segment C1. After identifying the dual origin of the porcine CST in the primary motor cortex (M1) and premotor cortex (PM), the axons descending from those regions to the spinal cord and brainstem were anterogradely labeled by unilateral injections of dextran alexa-594 in M1 and dextran alexa-488 in PM. Numerous corticospinal projections from M1 and PM were detected up to T6 spinal segment and showed a similar pattern of decussation and distribution in the white matter funiculi and the gray matter laminae. They terminated mostly on dendrites of the lateral intermediate laminae and the internal basilar nucleus, and some innervated the ventromedial laminae, but were essentially absent in lateral laminae IX. Corticofugal axons terminated predominantly ipsilaterally in the midbrain and bilaterally in the medulla oblongata. Most corticorubral projections arose from M1, whereas the mesencephalic reticular formation, superior colliculus, lateral reticular nucleus, gigantocellular reticular nucleus, and raphe received abundant axonal contacts from both M1 and PM. Our data suggest that the porcine cortical motor system has some common features with that of primates and humans and may control posture and movement through parallel motor descending pathways. However, less cortical regions project to the spinal cord in pigs, and the CST neither seems to reach the lumbar enlargement nor to have a significant direct innervation of cervical, foreleg motoneurons.
Collapse
Affiliation(s)
- Patricia Del Cerro
- Neural Repair and Biomaterials Laboratory, Hospital Nacional de Parapléjicos, Toledo, Spain.,Ph.D. Program in Neuroscience, Autonoma de Madrid University, Madrid, Spain
| | | | | |
Collapse
|
44
|
Sobinov AR, Bensmaia SJ. The neural mechanisms of manual dexterity. Nat Rev Neurosci 2021; 22:741-757. [PMID: 34711956 DOI: 10.1038/s41583-021-00528-7] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2021] [Indexed: 01/22/2023]
Abstract
The hand endows us with unparalleled precision and versatility in our interactions with objects, from mundane activities such as grasping to extraordinary ones such as virtuoso pianism. The complex anatomy of the human hand combined with expansive and specialized neuronal control circuits allows a wide range of precise manual behaviours. To support these behaviours, an exquisite sensory apparatus, spanning the modalities of touch and proprioception, conveys detailed and timely information about our interactions with objects and about the objects themselves. The study of manual dexterity provides a unique lens into the sensorimotor mechanisms that endow the nervous system with the ability to flexibly generate complex behaviour.
Collapse
Affiliation(s)
- Anton R Sobinov
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL, USA.,Neuroscience Institute, University of Chicago, Chicago, IL, USA
| | - Sliman J Bensmaia
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL, USA. .,Neuroscience Institute, University of Chicago, Chicago, IL, USA. .,Committee on Computational Neuroscience, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
45
|
Moreno-Lopez Y, Bichara C, Delbecq G, Isope P, Cordero-Erausquin M. The corticospinal tract primarily modulates sensory inputs in the mouse lumbar cord. eLife 2021; 10:65304. [PMID: 34497004 PMCID: PMC8439650 DOI: 10.7554/elife.65304] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 07/27/2021] [Indexed: 01/01/2023] Open
Abstract
It is generally assumed that the main function of the corticospinal tract (CST) is to convey motor commands to bulbar or spinal motoneurons. Yet the CST has also been shown to modulate sensory signals at their entry point in the spinal cord through primary afferent depolarization (PAD). By sequentially investigating different routes of corticofugal pathways through electrophysiological recordings and an intersectional viral strategy, we here demonstrate that motor and sensory modulation commands in mice belong to segregated paths within the CST. Sensory modulation is executed exclusively by the CST via a population of lumbar interneurons located in the deep dorsal horn. In contrast, the cortex conveys the motor command via a relay in the upper spinal cord or supraspinal motor centers. At lumbar level, the main role of the CST is thus the modulation of sensory inputs, which is an essential component of the selective tuning of sensory feedback used to ensure well-coordinated and skilled movement.
Collapse
Affiliation(s)
- Yunuen Moreno-Lopez
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Charlotte Bichara
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Gilles Delbecq
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Philippe Isope
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| | - Matilde Cordero-Erausquin
- Institut des Neurosciences Cellulaires et Intégrées, CNRS - Université de Strasbourg, Strasbourg, France
| |
Collapse
|
46
|
Bennett MS. Five Breakthroughs: A First Approximation of Brain Evolution From Early Bilaterians to Humans. Front Neuroanat 2021; 15:693346. [PMID: 34489649 PMCID: PMC8418099 DOI: 10.3389/fnana.2021.693346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/13/2021] [Indexed: 11/13/2022] Open
Abstract
Retracing the evolutionary steps by which human brains evolved can offer insights into the underlying mechanisms of human brain function as well as the phylogenetic origin of various features of human behavior. To this end, this article presents a model for interpreting the physical and behavioral modifications throughout major milestones in human brain evolution. This model introduces the concept of a "breakthrough" as a useful tool for interpreting suites of brain modifications and the various adaptive behaviors these modifications enabled. This offers a unique view into the ordered steps by which human brains evolved and suggests several unique hypotheses on the mechanisms of human brain function.
Collapse
|
47
|
Unraveling Axon Guidance during Axotomy and Regeneration. Int J Mol Sci 2021; 22:ijms22158344. [PMID: 34361110 PMCID: PMC8347220 DOI: 10.3390/ijms22158344] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/06/2023] Open
Abstract
During neuronal development and regeneration axons extend a cytoskeletal-rich structure known as the growth cone, which detects and integrates signals to reach its final destination. The guidance cues “signals” bind their receptors, activating signaling cascades that result in the regulation of the growth cone cytoskeleton, defining growth cone advance, pausing, turning, or collapse. Even though much is known about guidance cues and their isolated mechanisms during nervous system development, there is still a gap in the understanding of the crosstalk between them, and about what happens after nervous system injuries. After neuronal injuries in mammals, only axons in the peripheral nervous system are able to regenerate, while the ones from the central nervous system fail to do so. Therefore, untangling the guidance cues mechanisms, as well as their behavior and characterization after axotomy and regeneration, are of special interest for understanding and treating neuronal injuries. In this review, we present findings on growth cone guidance and canonical guidance cues mechanisms, followed by a description and comparison of growth cone pathfinding mechanisms after axotomy, in regenerative and non-regenerative animal models.
Collapse
|
48
|
Ribeiro Gomes AR, Olivier E, Killackey HP, Giroud P, Berland M, Knoblauch K, Dehay C, Kennedy H. Refinement of the Primate Corticospinal Pathway During Prenatal Development. Cereb Cortex 2021; 30:656-671. [PMID: 31343065 DOI: 10.1093/cercor/bhz116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 11/14/2022] Open
Abstract
Perturbation of the developmental refinement of the corticospinal (CS) pathway leads to motor disorders. While non-primate developmental refinement is well documented, in primates invasive investigations of the developing CS pathway have been confined to neonatal and postnatal stages when refinement is relatively modest. Here, we investigated the developmental changes in the distribution of CS projection neurons in cynomolgus monkey (Macaca fascicularis). Injections of retrograde tracer at cervical levels of the spinal cord at embryonic day (E) 95 and E105 show that: (i) areal distribution of back-labeled neurons is more extensive than in the neonate and dense labeling is found in prefrontal, limbic, temporal, and occipital cortex; (ii) distributions of contralateral and ipsilateral projecting CS neurons are comparable in terms of location and numbers of labeled neurons, in contrast to the adult where the contralateral projection is an order of magnitude higher than the ipsilateral projection. Findings from one largely restricted injection suggest a hitherto unsuspected early innervation of the gray matter. In the fetus there was in addition dense labeling in the central nucleus of the amygdala, the hypothalamus, the subthalamic nucleus, and the adjacent region of the zona incerta, subcortical structures with only minor projections in the adult control.
Collapse
Affiliation(s)
- Ana Rita Ribeiro Gomes
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Etienne Olivier
- Institute of Neuroscience, Université Catholique de Louvain, Belgium
| | - Herbert P Killackey
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Pascale Giroud
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Michel Berland
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Kenneth Knoblauch
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Colette Dehay
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France
| | - Henry Kennedy
- Univ Lyon, Université Claude Bernard Lyon 1, Inserm, Stem Cell and Brain Research Institute, Bron, France.,Institute of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
49
|
Modelling and Refining Neuronal Circuits with Guidance Cues: Involvement of Semaphorins. Int J Mol Sci 2021; 22:ijms22116111. [PMID: 34204060 PMCID: PMC8201269 DOI: 10.3390/ijms22116111] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022] Open
Abstract
The establishment of neuronal circuits requires neurons to develop and maintain appropriate connections with cellular partners in and out the central nervous system. These phenomena include elaboration of dendritic arborization and formation of synaptic contacts, initially made in excess. Subsequently, refinement occurs, and pruning takes places both at axonal and synaptic level, defining a homeostatic balance maintained throughout the lifespan. All these events require genetic regulations which happens cell-autonomously and are strongly influenced by environmental factors. This review aims to discuss the involvement of guidance cues from the Semaphorin family.
Collapse
|
50
|
Strick PL, Dum RP, Rathelot JA. The Cortical Motor Areas and the Emergence of Motor Skills: A Neuroanatomical Perspective. Annu Rev Neurosci 2021; 44:425-447. [PMID: 33863253 DOI: 10.1146/annurev-neuro-070918-050216] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
What changes in neural architecture account for the emergence and expansion of dexterity in primates? Dexterity, or skill in performing motor tasks, depends on the ability to generate highly fractionated patterns of muscle activity. It also involves the spatiotemporal coordination of activity in proximal and distal muscles across multiple joints. Many motor skills require the generation of complex movement sequences that are only acquired and refined through extensive practice. Improvements in dexterity have enabled primates to manufacture and use tools and humans to engage in skilled motor behaviors such as typing, dance, musical performance, and sports. Our analysis leads to the following synthesis: The neural substrate that endows primates with their enhanced motor capabilities is due, in part, to (a) major organizational changes in the primary motor cortex and (b) the proliferation of output pathways from other areas of the cerebral cortex, especially from the motor areas on the medial wall of the hemisphere.
Collapse
Affiliation(s)
- Peter L Strick
- Department of Neurobiology, Systems Neuroscience Center, and Brain Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA;
| | - Richard P Dum
- Department of Neurobiology, Systems Neuroscience Center, and Brain Institute, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, USA;
| | - Jean-Alban Rathelot
- Institut des Neurosciences de la Timone, CNRS, and Aix-Marseille Université, 13005 Marseille, France
| |
Collapse
|