1
|
Zheng W, Chotipanvithayakul R, Ingviya T, Guo F. Effects of home-based integrated sensory stimulation program to preterm infants on parents' depression and anxiety: a randomized controlled trial. Glob Health Action 2025; 18:2491848. [PMID: 40314668 PMCID: PMC12051557 DOI: 10.1080/16549716.2025.2491848] [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: 12/14/2024] [Accepted: 04/07/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Preterm parents face higher risks of postpartum depression and anxiety, affecting bonding and infant development. Sensory stimulation shows promise, but its long-term impact on parental mental health needs further study. OBJECTIVES This study aimed to evaluate whether a home-based integrated sensory stimulation program, administered to preterm infants by their parents, could alleviate parental mental health issues and enhance maternal bonding and parenting competence. METHODS The program, including tactile, auditory, visual, gustatory, and olfactory stimuli, was assessed in a block-randomized controlled trial from November 2018 to January 2020. A total of 200 parents of preterm infants were recruited, and the intervention continued at home until the infants reached six months corrected age. Parents' depression and anxiety were assessed using validated scales at baseline, and at first, third, and sixth month follow-ups. RESULTS The intervention group included 98 parents, and the control group comprised 102 parents. At the six-month follow-up, the intervention group demonstrated significant improvements in maternal depression, state anxiety, and trait anxiety compared to the control group. In the mixed linear model, the intervention was associated with reductions in maternal trait anxiety (d =-2.18; 95% CI: -4.30, -0.06), paternal trait anxiety (d =-3.37; 95% CI: -5.62, -1.11) and state anxiety (d =-4.63; 95% CI: -7.00, -2.26). CONCLUSION The home-based integrated sensory stimulation program, when provided by parents to preterm infants, was effective in improving parents' mental health and can serve as an alternative treatment for postpartum depression and anxiety in parents of preterm infants at home.
Collapse
Affiliation(s)
- Wenjing Zheng
- Department of Pediatrics, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Rassamee Chotipanvithayakul
- Department of Epidemiology, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Research Center for Kids and Youth Development, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Thammasin Ingviya
- Department of Family and Preventive Medicine, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
- Department of Clinical Research and Medical Data Science, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Fang Guo
- Department of Neonatology, Affiliated Hospital of Kunming University of Science and Technology Clinical College, Kunming, Yunnan, China
| |
Collapse
|
2
|
Piña Novo D, Gao M, Fischer R, Richevaux L, Yu J, Barrett JM, Shepherd GMG. Cortical dynamics in hand/forelimb S1 and M1 evoked by brief photostimulation of the mouse's hand. eLife 2025; 14:RP105112. [PMID: 40387088 PMCID: PMC12088680 DOI: 10.7554/elife.105112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025] Open
Abstract
Spiking activity along synaptic circuits linking primary somatosensory (S1) and motor (M1) areas is fundamental for sensorimotor integration in cortex. Circuits along the ascending somatosensory pathway through mouse hand/forelimb S1 and M1 were recently described in detail (Yamawaki et al., 2021). Here, we characterize the peripherally evoked spiking dynamics in these two cortical areas. Brief (5 ms) optogenetic photostimulation of the hand generated short (~25 ms) barrages of activity first in S1 (onset latency 15 ms) then M1 (10 ms later). The estimated propagation speed was 20-fold faster from hand to S1 than from S1 to M1. Amplitudes in M1 were strongly attenuated. Responses were typically triphasic, with suppression and rebound following the initial peak. Evoked activity in S1 was biased to middle layers, consistent with thalamocortical connectivity, while that in M1 was biased to upper layers, consistent with corticocortical connectivity. Parvalbumin (PV) inhibitory interneurons were involved in each phase, accounting for three quarters of the initial spikes generated in S1, and their selective photostimulation sufficed to evoke suppression and rebound in both S1 and M1. Partial silencing of S1 by PV activation during hand stimulation reduced the M1 sensory responses. Overall, these results characterize how evoked spiking activity propagates along the hand/forelimb transcortical loop, and illuminate how in vivo cortical dynamics relate to the underlying synaptic circuit organization in this system.
Collapse
Affiliation(s)
- Daniela Piña Novo
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Mang Gao
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Rita Fischer
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Louis Richevaux
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Jianing Yu
- School of Life Sciences, Peking UniversityBeijingChina
| | - John M Barrett
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| | - Gordon MG Shepherd
- Department of Neuroscience, Feinberg School of Medicine, Northwestern UniversityChicagoUnited States
| |
Collapse
|
3
|
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
|
4
|
Carranza E, Franovic S, Boos A, Pirondini E. Assessing age-related proprioceptive changes through active and passive tasks: implications for stroke assessment. J Neural Eng 2025; 22:026047. [PMID: 40153871 DOI: 10.1088/1741-2552/adc6bc] [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: 12/19/2024] [Accepted: 03/28/2025] [Indexed: 04/01/2025]
Abstract
Objective.Voluntary control of motor actions requires precise regulation of proprioceptive and somatosensory functions. While aging is known to impair sensory processing, its effect on proprioception remains unclear. Previous studies report conflicting findings on whether passive proprioception (i.e. during externally driven movements) declines with age, and research on age-related changes in active proprioception (i.e. during voluntary movements) remains limited, particularly in the upper limb. Understanding these changes is critical for identifying and preventing impairments that may affect movement performance and mobility, particularly in neurological conditions such as stroke or Parkinson's disease.Approach.We refined a robotic protocol to assess upper-limb active proprioception and validated its robustness and reliability over multiple sessions. Using this protocol, we compared the performance between young and elderly neurologically healthy adults during both active and passive proprioceptive tasks.Main results.Elderly participants exhibited a significant decline in accuracy when sensing limb position in both active and passive proprioceptive tasks, whereas their precision remained unchanged. These findings indicate that aging primarily affects proprioceptive accuracy rather than variability in position sense.Significance.Our findings contribute to the ongoing debate on age-related proprioceptive decline and highlight the importance of distinguishing between active and passive proprioception. Furthermore, our validated robotic protocol provides a reliable tool for assessing proprioception, with potential applications in studying neurological conditions in clinical settings.
Collapse
Affiliation(s)
- Erick Carranza
- Rehab and Neural Engineering Laboratory, University of Pittsburgh, 3520 Fifth Avenue, Suite 300, Pittsburgh, PA 15213, United States of America
- Department of Bioengineering, University of Pittsburgh, 151 Benedum Hall, Pittsburgh, PA 15261, United States of America
- Center for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, PA 15213, United States of America
| | - Sreten Franovic
- Rehab and Neural Engineering Laboratory, University of Pittsburgh, 3520 Fifth Avenue, Suite 300, Pittsburgh, PA 15213, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA 15213, United States of America
| | - Amy Boos
- Rehab and Neural Engineering Laboratory, University of Pittsburgh, 3520 Fifth Avenue, Suite 300, Pittsburgh, PA 15213, United States of America
- Department of Neurology, University of Pittsburgh, 3471, Fifth Avenue, Suite 910, Pittsburgh, PA 15213, United States of America
- VA Pittsburgh Healthcare System, VA, Pittsburgh, PA 15240, United States of America
| | - Elvira Pirondini
- Rehab and Neural Engineering Laboratory, University of Pittsburgh, 3520 Fifth Avenue, Suite 300, Pittsburgh, PA 15213, United States of America
- Department of Bioengineering, University of Pittsburgh, 151 Benedum Hall, Pittsburgh, PA 15261, United States of America
- Center for the Neural Basis of Cognition, 4400 Fifth Avenue, Suite 115, Pittsburgh, PA 15213, United States of America
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, 3471 Fifth Avenue, Suite 910, Pittsburgh, PA 15213, United States of America
- University of Pittsburgh Clinical and Translational Science Institute (CTSI), Pittsburgh, PA 15213, United States of America
| |
Collapse
|
5
|
Kaiser J, Patel P, Fedde S, Lammers A, Kenwood MR, Iqbal A, Goldberg M, Sahni V. Developmental molecular signatures define de novo cortico-brainstem circuit for skilled forelimb movement. RESEARCH SQUARE 2025:rs.3.rs-6150344. [PMID: 40196004 PMCID: PMC11975033 DOI: 10.21203/rs.3.rs-6150344/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Skilled movement relies on descending cortical projections to the brainstem and spinal cord. While corticospinal neurons (CSN) have long been recognized for their role in fine motor control, the contribution of cortical projections to the brainstem remains poorly understood. Here, we identify a previously unrecognized direct cortico-brainstem circuit that emerges early in development and persists into adulthood. A subset of subcerebral projection neurons (SCPN) limit their projections to the brainstem from the earliest stages of axon extension without ever extending to the spinal cord. Using FACS purification and single-cell RNA sequencing, we show that these cortico-brainstem neurons (CBN) can be prospectively identified by the expression of Neuropeptide Y (Npy) in development. Functional silencing of Npy+ CBN in adulthood leads to impaired skilled forelimb reaching, demonstrating their essential role in adult motor control. Npy+ CBN project preferentially to rostral brainstem regions, including the midbrain reticular formation. These findings reveal developmental molecular signatures that define cortico-brainstem pathways for adult skilled movement. Our work provides new insights into the developmental logic that establishes descending cortical circuits and opens avenues for targeted investigation of their roles in motor function and recovery after injury.
Collapse
Affiliation(s)
- Julia Kaiser
- Burke Neurological Institute, White Plains, NY, 10605
| | - Payal Patel
- Burke Neurological Institute, White Plains, NY, 10605
| | - Sam Fedde
- Burke Neurological Institute, White Plains, NY, 10605
| | | | | | - Asim Iqbal
- Burke Neurological Institute, White Plains, NY, 10605
- Tibbling Technologies, Redmond, WA, 98052
| | - Mark Goldberg
- Department of Neurology, UT Health Sciences Center San Antonio, San Antonio, TX, USA
| | - Vibhu Sahni
- Burke Neurological Institute, White Plains, NY, 10605
- Department of Neurology, UT Health Sciences Center San Antonio, San Antonio, TX, USA
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York City, NY, 10065
| |
Collapse
|
6
|
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
|
7
|
Piña Novo D, Gao M, Fischer R, Richevaux L, Yu J, Barrett JM, Shepherd GMG. Cortical dynamics in hand/forelimb S1 and M1 evoked by brief photostimulation of the mouse's hand. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.02.626335. [PMID: 39677687 PMCID: PMC11642753 DOI: 10.1101/2024.12.02.626335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Spiking activity along synaptic circuits linking primary somatosensory (S1) and motor (M1) areas is fundamental for sensorimotor integration in cortex. Circuits along the ascending somatosensory pathway through mouse hand/forelimb S1 and M1 were recently described in detail (Yamawaki et al., 2021). Here, we characterize the peripherally evoked spiking dynamics in these two cortical areas. Brief (5 ms) optogenetic photostimulation of the hand generated short (~25 ms) barrages of activity first in S1 (onset latency 15 ms) then M1 (10 ms later). The estimated propagation speed was 20-fold faster from hand to S1 than from S1 to M1. Amplitudes in M1 were strongly attenuated. Responses were typically triphasic, with suppression and rebound following the initial peak. Evoked activity in S1 was biased to middle layers, consistent with thalamocortical connectivity, while that in M1 was biased to upper layers, consistent with corticocortical connectivity. Parvalbumin (PV) inhibitory interneurons were involved in each phase, accounting for three quarters of the initial spikes generated in S1, and their selective photostimulation sufficed to evoke suppression and rebound in both S1 and M1. Partial silencing of S1 by PV activation during hand stimulation reduced the M1 sensory responses. Overall, these results characterize how evoked spiking activity propagates along the hand/forelimb transcortical loop, and illuminate how in vivo cortical dynamics relate to the underlying synaptic circuit organization in this system.
Collapse
Affiliation(s)
- Daniela Piña Novo
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Mang Gao
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Rita Fischer
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Louis Richevaux
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Jianing Yu
- School of Life Sciences, Peking University, Beijing 100871, China
| | - John M. Barrett
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Gordon M. G. Shepherd
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
8
|
Chapman PD, Kulkarni AS, Trevisan AJ, Han K, Hinton JM, Deltuvaite P, Fenno LE, Ramakrishnan C, Patton MH, Schwarz LA, Zakharenko SS, Deisseroth K, Bikoff JB. A brain-wide map of descending inputs onto spinal V1 interneurons. Neuron 2025; 113:524-538.e6. [PMID: 39719703 PMCID: PMC11842218 DOI: 10.1016/j.neuron.2024.11.019] [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: 09/13/2023] [Revised: 10/11/2024] [Accepted: 11/26/2024] [Indexed: 12/26/2024]
Abstract
Motor output results from the coordinated activity of neural circuits distributed across multiple brain regions that convey information to the spinal cord via descending motor pathways. Yet the organizational logic through which supraspinal systems target discrete components of spinal motor circuits remains unclear. Here, using viral transsynaptic tracing along with serial two-photon tomography, we have generated a whole-brain map of monosynaptic inputs to spinal V1 interneurons, a major inhibitory population involved in motor control. We identified 26 distinct brain structures that directly innervate V1 interneurons, spanning medullary and pontine regions in the hindbrain as well as cortical, midbrain, cerebellar, and neuromodulatory systems. Moreover, we identified broad but biased input from supraspinal systems onto V1Foxp2 and V1Pou6f2 neuronal subsets. Collectively, these studies reveal elements of biased connectivity and convergence in descending inputs to molecularly distinct interneuron subsets and provide an anatomical foundation for understanding how supraspinal systems influence spinal motor circuits.
Collapse
Affiliation(s)
- Phillip D Chapman
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anand S Kulkarni
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alexandra J Trevisan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Katie Han
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer M Hinton
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Paulina Deltuvaite
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lief E Fenno
- Department of Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Psychiatry & Behavioral Sciences, University of Texas Dell Medical School, Austin, TX 78712, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Mary H Patton
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Lindsay A Schwarz
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
| | - Jay B Bikoff
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
9
|
Chaterji S, Belliappa PH, Sathyamurthy A. The superior colliculus directs goal-oriented forelimb movements. Cell Rep 2025; 44:115097. [PMID: 39723891 DOI: 10.1016/j.celrep.2024.115097] [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: 07/11/2024] [Revised: 10/23/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024] Open
Abstract
Skilled forelimb control is essential for daily living, yet our understanding of its neural mechanisms, although extensive, remains incomplete. Here, we present evidence that the superior colliculus (SC), a major midbrain structure, is necessary for accurate forelimb reaching in mice. We found that neurons in the lateral SC are active during goal-directed reaching, and by employing chemogenetic and phase-specific optogenetic silencing of these neurons, we show that the SC causally facilitates reach accuracy. Anatomical studies identified the deep cerebellar nuclei and substantia nigra pars reticulata as sources of inputs to the SC, while functional studies revealed a role for nigrotectal, but not cerebellotectal, neurons in controlling reach endpoints. Silencing the nigrotectal pathway caused paw deviations opposite to those seen with SC silencing, emphasizing the coordinated role of the substantia nigra and SC in regulating optimal reaching. Together, these findings establish the SC as a crucial regulator of skilled forelimb control.
Collapse
Affiliation(s)
- Shrivas Chaterji
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Punarva H Belliappa
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Anupama Sathyamurthy
- Centre for Neuroscience, Indian Institute of Science, Bengaluru, Karnataka 560012, India.
| |
Collapse
|
10
|
Lin M, Calabrese GB, Incognito AV, Moore MT, Agarwal A, Wilson RJ, Zagoraiou L, Sharples SA, Miles GB, Philippidou P. A cholinergic spinal pathway for the adaptive control of breathing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.20.633641. [PMID: 39896653 PMCID: PMC11785070 DOI: 10.1101/2025.01.20.633641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The ability to amplify motor neuron (MN) output is essential for generating high intensity motor actions. This is critical for breathing that must be rapidly adjusted to accommodate changing metabolic demands. While brainstem circuits generate the breathing rhythm, the pathways that directly augment respiratory MN output are not well understood. Here, we mapped first-order inputs to phrenic motor neurons (PMNs), a key respiratory MN population that initiates diaphragm contraction to drive breathing. We identified a predominant spinal input from a distinct subset of genetically-defined V0C cholinergic interneurons. We found that these interneurons receive phasic excitation from brainstem respiratory centers, augment phrenic output through M2 muscarinic receptors, and are highly activated under a hypercapnia challenge. Specifically silencing cholinergic interneuron neurotransmission impairs the breathing response to hypercapnia. Collectively, our findings identify a novel spinal pathway that amplifies breathing, presenting a potential target for promoting recovery of breathing following spinal cord injury.
Collapse
Affiliation(s)
- Minshan Lin
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | | | - Anthony V. Incognito
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Canada
| | - Matthew T. Moore
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Aambar Agarwal
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| | - Richard J.A. Wilson
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Canada
| | - Laskaro Zagoraiou
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Simon A. Sharples
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, United Kingdom
| | - Gareth B. Miles
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, United Kingdom
| | - Polyxeni Philippidou
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
11
|
Osborne KJ, Walther S, Mittal VA. Motor actions across psychiatric disorders: A research domain criteria (RDoC) perspective. Clin Psychol Rev 2024; 114:102511. [PMID: 39510028 DOI: 10.1016/j.cpr.2024.102511] [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: 03/14/2024] [Revised: 08/19/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024]
Abstract
The motor system is critical for understanding the pathophysiology and treatment of mental illness. Abnormalities in the processes that allow us to plan and execute movement in a goal-directed, context-appropriate manner (i.e., motor actions) are especially central to clinical motor research. Within this context, the NIMH Research Domain Criteria (RDoC) framework now includes a Motor Actions construct within the recently incorporated Sensorimotor Systems Domain, providing a useful framework for conducting research on motor action processes. However, there is limited available resources for understanding or implementing this framework. We address this gap by providing a comprehensive critical review and conceptual integration of the current clinical literature on the subconstructs comprising the Motor Actions construct. This includes a detailed discussion of each Motor Action subconstruct (e.g., action planning/execution) and its measurement across different units of analysis (e.g., molecules to behavior), the temporal and conceptual relationships among the Motor Action subconstructs (and other relevant RDoC domain constructs), and how abnormalities in these Motor Action subconstructs manifest in mental illness. Together, the review illustrates how motor system dysfunction is implicated in the pathophysiology of many psychiatric conditions and demonstrates shared and distinct mechanisms that may account for similar manifestations of motor abnormalities across disorders.
Collapse
Affiliation(s)
- K Juston Osborne
- Washington University in St. Louis, Department of Psychiatry, 4444 Forest Park Ave., St. Louis, MO, USA; Northwestern University, Department of Psychology, 633 Clark St. Evanston, IL, USA.
| | - Sebastian Walther
- University Hospital Würzburg, Department of Psychiatry, Psychosomatics, and Psychotherapy, Center of Mental Health, Margarete-Höppel-Platz 1, 97080 Würzburg, Germany
| | - Vijay A Mittal
- Northwestern University, Department of Psychology, 633 Clark St. Evanston, IL, USA; Northwestern University, Department of Psychiatry, 676 N. St. Claire, Chicago, IL, USA; Northwestern University, Department of Psychiatry, Institute for Policy Research, Department of Medical Social Sciences, Institute for Innovations in Developmental Sciences (DevSci), 633 Clark St., Evanston, Chicago, IL, USA
| |
Collapse
|
12
|
Han J, Wang R, Wang M, Yu Z, Zhu L, Zhang J, Zhu J, Zhang S, Xi W, Wu H. Dynamic lateralization in contralateral-projecting corticospinal neurons during motor learning. iScience 2024; 27:111078. [PMID: 39493873 PMCID: PMC11530912 DOI: 10.1016/j.isci.2024.111078] [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: 06/15/2024] [Accepted: 09/27/2024] [Indexed: 11/05/2024] Open
Abstract
Understanding the adaptability of the motor cortex in response to bilateral motor tasks is crucial for advancing our knowledge of neural plasticity and motor learning. Here we aim to investigate the dynamic lateralization of contralateral-projecting corticospinal neurons (cpCSNs) during such tasks. Utilizing in vivo two-photon calcium imaging, we observe cpCSNs in mice performing a "left-right" lever-press task. Our findings reveal heterogeneous populational dynamics in cpCSNs: a marked decrease in activity during ipsilateral motor learning, in contrast to maintained activity during contralateral motor learning. Notably, individual cpCSNs show dynamic shifts in engagement with ipsilateral and contralateral movements, displaying an evolving pattern of activation over successive days. It suggests that cpCSNs exhibit adaptive changes in activation patterns in response to ipsilateral and contralateral movements, highlighting a flexible reorganization during motor learning This reconfiguration underscores the dynamic nature of cortical lateralization in motor learning and offers insights for neuromotor rehabilitation.
Collapse
Affiliation(s)
- Jiawei Han
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Disease of Zhejiang Province, Hangzhou 310058, China
| | - Ruixue Wang
- Department of Neurosurgery, Third Affiliated Hospital, Naval Medical University, Shanghai 200438, China
- Qiushi Academy for Advanced Studies, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Minmin Wang
- Qiushi Academy for Advanced Studies, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Zhihua Yu
- Department of Critical Care Medicine, Hangzhou Third People’s Hospital, Hangzhou 310058, China
| | - Liang Zhu
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Disease of Zhejiang Province, Hangzhou 310058, China
| | - Junming Zhu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Disease of Zhejiang Province, Hangzhou 310058, China
| | - Shaomin Zhang
- Qiushi Academy for Advanced Studies, Key Laboratory of Biomedical Engineering of Ministry of Education, Zhejiang Provincial Key Laboratory of Cardio-Cerebral Vascular Detection Technology and Medicinal Effectiveness Appraisal, Zhejiang University, Hangzhou 310027, China
| | - Wang Xi
- Interdisciplinary Institute of Neuroscience and Technology (ZIINT), the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310020, China
- MOE Frontier Science Center for Brain Research and Brain Machine Integration, Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou 310027, China
| | - Hemmings Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
- Clinical Research Center for Neurological Disease of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Tsuji S, Kuramoto Y, Rajbhandari S, Takeda Y, Yamahara K, Yoshimura S. Intravenous administration of human amnion-derived mesenchymal stem cells improves gait and sensory function in mouse models of spinal cord injury. Front Cell Dev Biol 2024; 12:1464727. [PMID: 39324071 PMCID: PMC11422150 DOI: 10.3389/fcell.2024.1464727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Spinal cord injury (SCI) leads to severe disabilities and remains a significant social and economic challenge. Despite advances in medical research, there are still no effective treatments for SCI. Human amnion-derived mesenchymal stem cells (hAMSCs) have shown potential due to their anti-inflammatory and neuroprotective effects. This study evaluates the therapeutic potential of intravenously administered hAMSCs in SCI models. Methods Three days after induction of SCI with forceps calibrated with a 0.2 mm gap, hAMSCs or vehicle were administered intravenously. Up to 4 weeks of SCI induction, motor function was assessed by scores on the Basso Mouse Locomotor Scale (BMS) and the Basso-Beattie-Bresnahan Scale (BBB), and sensory function by hindlimb withdrawal reflex using von Frey filaments. Six weeks after SCI induction, gait function was assessed using three-dimensional motion analysis. Immunohistochemistry, polymerase chain reaction (PCR), flow cytometry, and ELISA assay were performed to clarify the mechanisms of functional improvement. Results The hAMSC treatment significantly improved sensory response and gait function. In the SCI site, immunohistochemistry showed a reduction in Iba1-positive cells and PCR revealed decreased TNFα and increased BDNF levels in the hAMSC-treated group. In assessing the systemic inflammatory response, hAMSC treatment reduced monocytic bone marrow-derived suppressor cells (M-MDSCs) and Ly6C-positive inflammatory macrophages in the bone marrow by flow cytometry and serum NO levels by ELISA assay. Discussion This study demonstrates the therapeutic potential of the hAMSC in SCI, with improvements in gait and sensory functions and reduced inflammation both locally and systemically. The findings support further investigation of the hAMSC as a potential treatment for SCI, focusing on their ability to modulate inflammation and promote neuroprotection.
Collapse
Affiliation(s)
- Shoichiro Tsuji
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | - Yoji Kuramoto
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | | | - Yuki Takeda
- Department of Neurosurgery, Hyogo Medical University, Hyogo, Japan
| | - Kenichi Yamahara
- Laboratory of Molecular and Cellular Therapy, Institute for Advanced Medical Sciences, Hyogo Medical University, Hyogo, Japan
| | | |
Collapse
|
16
|
Isumi S, Futamura D, Hanasaki T, Sako Y, Miyata S, Kan H, Suzuki Y, Hasegawa N, Mushiake H, Kametaka S, Uchiyama Y, Osanai M, Lee-Hotta S. Association of medullary reticular formation ventral part with spasticity in mice suffering from photothrombotic stroke. Neuroimage 2024; 298:120791. [PMID: 39147291 DOI: 10.1016/j.neuroimage.2024.120791] [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: 03/22/2024] [Revised: 07/15/2024] [Accepted: 08/13/2024] [Indexed: 08/17/2024] Open
Abstract
Strokes cause spasticity via stretch reflex hyperexcitability in the spinal cord, and spastic paralysis due to involuntary muscle contraction in the hands and fingers can severely restrict skilled hand movements. However, the underlying neurological mechanisms remain unknown. Using a mouse model of spasticity after stroke, we demonstrate changes in neuronal activity with and without electrostimulation of the afferent nerve to induce the stretch reflex, measured using quantitative activation-induced manganese-enhanced magnetic resonance imaging. Neuronal activity increased within the ventral medullary reticular formation (MdV) in the contralesional brainstem during the acute post-stroke phase, and this increase was characterised by activation of circuits involved in spasticity. Interestingly, ascending electrostimulation inhibited the MdV activity on the stimulation side in normal conditions. Moreover, immunohistochemical staining showed that, in the acute phase, the density of GluA1, one of the α-amino-3 hydroxy‑5 methyl -4 isoxazolepropionic acid receptor (AMPAR) subunits, at the synapses of MdV neurons was significantly increased. In addition, the GluA1/GluA2 ratio in these receptors was altered at 2 weeks post-stroke, confirming homeostatic plasticity as the underlying mechanisms of spasticity. These results provide new insights into the relationship between impaired skilled movements and spasticity at the acute post-stroke phase.
Collapse
Affiliation(s)
- Shogo Isumi
- Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Daiki Futamura
- Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Takuto Hanasaki
- Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yukito Sako
- Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Shotaro Miyata
- Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hirohito Kan
- Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yumika Suzuki
- Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Naoki Hasegawa
- Department of Radiological Imaging and Informatics, School of Medicine, Tohoku University, Sendai, Japan
| | - Hajime Mushiake
- Department of Physiology, School of Medicine, Tohoku University, Sendai, Japan
| | - Satoshi Kametaka
- Division of Biofunctional Sciences, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yasushi Uchiyama
- Division of Creative Physical Therapy, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, Aichi 461-8673, Japan
| | - Makoto Osanai
- Department of Physiology, School of Medicine, Tohoku University, Sendai, Japan; Laboratory for Physiological Functional Imaging, Department of Medical Physics and Engineering, Division of Health Sciences, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Sachiko Lee-Hotta
- Division of Creative Physical Therapy, Department of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, 1-1-20 Daiko-Minami, Higashi-ku, Nagoya, Aichi 461-8673, Japan.
| |
Collapse
|
17
|
Imai F, Matsuura K, Yang E, Klinefelter K, Alexandrou G, Letelier A, Takatani H, Osakada F, Yoshida Y. Layer Va neurons, as major presynaptic partners of corticospinal neurons, play critical roles in skilled movements. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.28.601172. [PMID: 38979259 PMCID: PMC11230360 DOI: 10.1101/2024.06.28.601172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Corticospinal neurons (CSNs) are located in the cortex and projecting into the spinal cord. The activation of CSNs, which is associated with skilled motor behaviors, induces the activation of interneurons in the spinal cord. Eventually, motor neuron activation is induced by corticospinal circuits to coordinate muscle activation. Therefore, elucidating how the activation of CSNs in the brain is regulated is necessary for understanding the roles of CSNs in skilled motor behaviors. However, the presynaptic partners of CSNs in the brain remain to be identified. Here, we performed transsynaptic rabies virus-mediated brain-wide mapping to identify presynaptic partners of CSNs (pre-CSNs). We found that pre-CSNs are located in all cortical layers, but major pre-CSNs are located in layer Va. A small population of pre-CSNs are also located outside the cortex, such as in the thalamus. Inactivation of layer Va neurons in Tlx3-Cre mice results in deficits in skilled reaching and grasping behaviors, suggesting that, similar to CSNs, layer Va neurons are critical for skilled movements. Finally, we examined whether the connectivity of CSNs is altered after spinal cord injury (SCI). We found that unlike connections between CNSs and postsynaptic neurons, connections between pre-CSNs and CSNs do not change after SCI.
Collapse
|
18
|
Fan F, Yin T, Wu B, Zheng J, Deng J, Wu G, Hu S. The role of spinal neurons targeted by corticospinal neurons in central poststroke neuropathic pain. CNS Neurosci Ther 2024; 30:e14813. [PMID: 38887838 PMCID: PMC11183184 DOI: 10.1111/cns.14813] [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: 01/14/2024] [Revised: 05/15/2024] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Central poststroke pain (CPSP) is one of the primary sequelae following stroke, yet its underlying mechanisms are poorly understood. METHODS By lesioning the lateral thalamic nuclei, we first established a CPSP model that exhibits mechanical and thermal hypersensitivity. Innocuous mechanical stimuli following the thalamic lesion evoked robust neural activation in somatosensory corticospinal neurons (CSNs), as well as in the deep dorsal horn, where low threshold mechanosensory afferents terminate. In this study, we used viral-based mapping and intersectional functional manipulations to decipher the role of somatosensory CSNs and their spinal targets in the CPSP pathophysiology. RESULTS We first mapped the post-synaptic spinal targets of lumbar innervating CSNs using an anterograde trans-synaptic AAV1-based strategy and showed these spinal interneurons were activated by innocuous tactile stimuli post-thalamic lesion. Functionally, tetanus toxin-based chronic inactivation of spinal neurons targeted by CSNs prevented the development of CPSP. Consistently, transient chemogenetic silencing of these neurons alleviated established mechanical pain hypersensitivity and innocuous tactile stimuli evoked aversion linked to the CPSP. In contrast, chemogenetic activation of these neurons was insufficient to induce robust mechanical allodynia typically observed in the CPSP. CONCLUSION The CSNs and their spinal targets are required but insufficient for the establishment of CPSP hypersensitivity. Our study provided novel insights into the neural mechanisms underlying CPSP and potential therapeutic interventions to treat refractory central neuropathic pain conditions.
Collapse
Affiliation(s)
- Fenqqi Fan
- Department of Pain, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Tianze Yin
- Department of Pain, Yueyang Hospital of Integrated Traditional Chinese and Western MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Biwu Wu
- Department of Neurosurgery and Neurocritical Care, Huashan HospitalFudan UniversityShanghaiChina
| | - Jiajun Zheng
- Department of Neurosurgery and Neurocritical Care, Huashan HospitalFudan UniversityShanghaiChina
| | - Jiaojiao Deng
- Department of Neurosurgery and Neurocritical Care, Huashan HospitalFudan UniversityShanghaiChina
| | - Gang Wu
- Department of Neurosurgery and Neurocritical Care, Huashan HospitalFudan UniversityShanghaiChina
| | - Shukun Hu
- Department of Neurosurgery and Neurocritical Care, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
19
|
Weman HM, Ceder MM, Ahemaiti A, Magnusson KA, Henriksson K, Andréasson L, Lagerström MC. Spinal Glycine Receptor Alpha 3 Cells Communicate Sensations of Chemical Itch in Hairy Skin. J Neurosci 2024; 44:e1585232024. [PMID: 38553047 PMCID: PMC11079978 DOI: 10.1523/jneurosci.1585-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 01/31/2024] [Accepted: 02/15/2024] [Indexed: 05/12/2024] Open
Abstract
Glycinergic neurons regulate nociceptive and pruriceptive signaling in the spinal cord, but the identity and role of the glycine-regulated neurons are not fully known. Herein, we have characterized spinal glycine receptor alpha 3 (Glra3) subunit-expressing neurons in Glra3-Cre female and male mice. Glra3-Cre(+) neurons express Glra3, are located mainly in laminae III-VI, and respond to glycine. Chemogenetic activation of spinal Glra3-Cre(+) neurons induced biting/licking, stomping, and guarding behaviors, indicative of both a nociceptive and pruriceptive role for this population. Chemogenetic inhibition did not affect mechanical or thermal responses but reduced behaviors evoked by compound 48/80 and chloroquine, revealing a pruriceptive role for these neurons. Spinal cells activated by compound 48/80 or chloroquine express Glra3, further supporting the phenotype. Retrograde tracing revealed that spinal Glra3-Cre(+) neurons receive input from afferents associated with pain and itch, and dorsal root stimulation validated the monosynaptic input. In conclusion, these results show that spinal Glra3(+) neurons contribute to acute communication of compound 48/80- and chloroquine-induced itch in hairy skin.
Collapse
Affiliation(s)
- Hannah M Weman
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Mikaela M Ceder
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Aikeremu Ahemaiti
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Kajsa A Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Katharina Henriksson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Linn Andréasson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| | - Malin C Lagerström
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala 75108, Sweden
| |
Collapse
|
20
|
Wang Y, Chen Y, Chen L, Herron BJ, Chen XY, Wolpaw JR. Motor learning changes the axon initial segment of the spinal motoneuron. J Physiol 2024; 602:2107-2126. [PMID: 38568869 PMCID: PMC11196014 DOI: 10.1113/jp283875] [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: 02/26/2023] [Accepted: 03/12/2024] [Indexed: 04/05/2024] Open
Abstract
We are studying the mechanisms of H-reflex operant conditioning, a simple form of learning. Modelling studies in the literature and our previous data suggested that changes in the axon initial segment (AIS) might contribute. To explore this, we used blinded quantitative histological and immunohistochemical methods to study in adult rats the impact of H-reflex conditioning on the AIS of the spinal motoneuron that produces the reflex. Successful, but not unsuccessful, H-reflex up-conditioning was associated with greater AIS length and distance from soma; greater length correlated with greater H-reflex increase. Modelling studies in the literature suggest that these increases may increase motoneuron excitability, supporting the hypothesis that they may contribute to H-reflex increase. Up-conditioning did not affect AIS ankyrin G (AnkG) immunoreactivity (IR), p-p38 protein kinase IR, or GABAergic terminals. Successful, but not unsuccessful, H-reflex down-conditioning was associated with more GABAergic terminals on the AIS, weaker AnkG-IR, and stronger p-p38-IR. More GABAergic terminals and weaker AnkG-IR correlated with greater H-reflex decrease. These changes might potentially contribute to the positive shift in motoneuron firing threshold underlying H-reflex decrease; they are consistent with modelling suggesting that sodium channel change may be responsible. H-reflex down-conditioning did not affect AIS dimensions. This evidence that AIS plasticity is associated with and might contribute to H-reflex conditioning adds to evidence that motor learning involves both spinal and brain plasticity, and both neuronal and synaptic plasticity. AIS properties of spinal motoneurons are likely to reflect the combined influence of all the motor skills that share these motoneurons. KEY POINTS: Neuronal action potentials normally begin in the axon initial segment (AIS). AIS plasticity affects neuronal excitability in development and disease. Whether it does so in learning is unknown. Operant conditioning of a spinal reflex, a simple learning model, changes the rat spinal motoneuron AIS. Successful, but not unsuccessful, H-reflex up-conditioning is associated with greater AIS length and distance from soma. Successful, but not unsuccessful, down-conditioning is associated with more AIS GABAergic terminals, less ankyrin G, and more p-p38 protein kinase. The associations between AIS plasticity and successful H-reflex conditioning are consistent with those between AIS plasticity and functional changes in development and disease, and with those predicted by modelling studies in the literature. Motor learning changes neurons and synapses in spinal cord and brain. Because spinal motoneurons are the final common pathway for behaviour, their AIS properties probably reflect the combined impact of all the behaviours that use these motoneurons.
Collapse
Affiliation(s)
- Yu Wang
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
| | - Yi Chen
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
| | - Lu Chen
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
| | - Bruce J. Herron
- Wadsworth Center, New York State Department of Health, 150 New Scotland Ave, Albany, NY 12208
- Department of Biomedical Sciences, School of Public Health, State University of New York, Albany, New York
| | - Xiang Yang Chen
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
- Department of Biomedical Sciences, School of Public Health, State University of New York, Albany, New York
| | - Jonathan R. Wolpaw
- National Center for Adaptive Neurotechnologies, Albany Stratton VA Medical Center, 113 Holland Ave, Albany, NY 12208
- Department of Biomedical Sciences, School of Public Health, State University of New York, Albany, New York
| |
Collapse
|
21
|
Carmona LM, Thomas ED, Smith K, Tasic B, Costa RM, Nelson A. Topographical and cell type-specific connectivity of rostral and caudal forelimb corticospinal neuron populations. Cell Rep 2024; 43:113993. [PMID: 38551963 PMCID: PMC11100358 DOI: 10.1016/j.celrep.2024.113993] [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: 12/01/2023] [Revised: 02/07/2024] [Accepted: 03/07/2024] [Indexed: 04/09/2024] Open
Abstract
Corticospinal neurons (CSNs) synapse directly on spinal neurons, a diverse assortment of cells with unique structural and functional properties necessary for body movements. CSNs modulating forelimb behavior fractionate into caudal forelimb area (CFA) and rostral forelimb area (RFA) motor cortical populations. Despite their prominence, the full diversity of spinal neurons targeted by CFA and RFA CSNs is uncharted. Here, we use anatomical and RNA sequencing methods to show that CSNs synapse onto a remarkably selective group of spinal cell types, favoring inhibitory populations that regulate motoneuron activity and gate sensory feedback. CFA and RFA CSNs target similar spinal neuron types, with notable exceptions that suggest that these populations differ in how they influence behavior. Finally, axon collaterals of CFA and RFA CSNs target similar brain regions yet receive highly divergent inputs. These results detail the rules of CSN connectivity throughout the brain and spinal cord for two regions critical for forelimb behavior.
Collapse
Affiliation(s)
- Lina Marcela Carmona
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA
| | - Eric D Thomas
- Allen Institute for Brain Science, Allen Institute, Seattle, WA, USA
| | - Kimberly Smith
- Allen Institute for Brain Science, Allen Institute, Seattle, WA, USA
| | - Bosiljka Tasic
- Allen Institute for Brain Science, Allen Institute, Seattle, WA, USA
| | - Rui M Costa
- Department of Neuroscience, Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY, USA; Allen Institute for Brain Science, Allen Institute, Seattle, WA, USA
| | - Anders Nelson
- Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
22
|
Gradwell MA, Ozeri-Engelhard N, Eisdorfer JT, Laflamme OD, Gonzalez M, Upadhyay A, Medlock L, Shrier T, Patel KR, Aoki A, Gandhi M, Abbas-Zadeh G, Oputa O, Thackray JK, Ricci M, George A, Yusuf N, Keating J, Imtiaz Z, Alomary SA, Bohic M, Haas M, Hernandez Y, Prescott SA, Akay T, Abraira VE. Multimodal sensory control of motor performance by glycinergic interneurons of the mouse spinal cord deep dorsal horn. Neuron 2024; 112:1302-1327.e13. [PMID: 38452762 DOI: 10.1016/j.neuron.2024.01.027] [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: 06/13/2023] [Revised: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 03/09/2024]
Abstract
Sensory feedback is integral for contextually appropriate motor output, yet the neural circuits responsible remain elusive. Here, we pinpoint the medial deep dorsal horn of the mouse spinal cord as a convergence point for proprioceptive and cutaneous input. Within this region, we identify a population of tonically active glycinergic inhibitory neurons expressing parvalbumin. Using anatomy and electrophysiology, we demonstrate that deep dorsal horn parvalbumin-expressing interneuron (dPV) activity is shaped by convergent proprioceptive, cutaneous, and descending input. Selectively targeting spinal dPVs, we reveal their widespread ipsilateral inhibition onto pre-motor and motor networks and demonstrate their role in gating sensory-evoked muscle activity using electromyography (EMG) recordings. dPV ablation altered limb kinematics and step-cycle timing during treadmill locomotion and reduced the transitions between sub-movements during spontaneous behavior. These findings reveal a circuit basis by which sensory convergence onto dorsal horn inhibitory neurons modulates motor output to facilitate smooth movement and context-appropriate transitions.
Collapse
Affiliation(s)
- Mark A Gradwell
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Nofar Ozeri-Engelhard
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Neuroscience PhD program, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Jaclyn T Eisdorfer
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Olivier D Laflamme
- Dalhousie PhD program, Dalhousie University, Halifax, NS, Canada; Department of Medical Neuroscience, Atlantic Mobility Action Project, Brain Repair Center, Dalhousie University, Halifax, NS, Canada
| | - Melissa Gonzalez
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Department of Biomedical Engineering, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Aman Upadhyay
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Neuroscience PhD program, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Laura Medlock
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Tara Shrier
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Komal R Patel
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Adin Aoki
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Melissa Gandhi
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Gloria Abbas-Zadeh
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Olisemaka Oputa
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Joshua K Thackray
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Human Genetics Institute of New Jersey, Rutgers University, The State University of New Jersey, Piscataway, NJ, USA; Tourette International Collaborative Genetics Study (TIC Genetics)
| | - Matthew Ricci
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Arlene George
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Nusrath Yusuf
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; Neuroscience PhD program, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Jessica Keating
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Zarghona Imtiaz
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Simona A Alomary
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Manon Bohic
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Michael Haas
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Yurdiana Hernandez
- W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA
| | - Steven A Prescott
- Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Turgay Akay
- Department of Medical Neuroscience, Atlantic Mobility Action Project, Brain Repair Center, Dalhousie University, Halifax, NS, Canada
| | - Victoria E Abraira
- Cell Biology and Neuroscience Department, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA; W.M. Keck Center for Collaborative Neuroscience, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
23
|
Nekanti U, Sakthivel PS, Zahedi A, Creasman DA, Nishi RA, Dumont CM, Piltti KM, Guardamondo GL, Hernandez N, Chen X, Song H, Lin X, Martinez J, On L, Lakatos A, Pawar K, David BT, Guo Z, Seidlits SK, Xu X, Shea LD, Cummings BJ, Anderson AJ. Multichannel bridges and NSC synergize to enhance axon regeneration, myelination, synaptic reconnection, and recovery after SCI. NPJ Regen Med 2024; 9:12. [PMID: 38499577 PMCID: PMC10948859 DOI: 10.1038/s41536-024-00356-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 02/15/2024] [Indexed: 03/20/2024] Open
Abstract
Regeneration in the injured spinal cord is limited by physical and chemical barriers. Acute implantation of a multichannel poly(lactide-co-glycolide) (PLG) bridge mechanically stabilizes the injury, modulates inflammation, and provides a permissive environment for rapid cellularization and robust axonal regrowth through this otherwise inhibitory milieu. However, without additional intervention, regenerated axons remain largely unmyelinated (<10%), limiting functional repair. While transplanted human neural stem cells (hNSC) myelinate axons after spinal cord injury (SCI), hNSC fate is highly influenced by the SCI inflammatory microenvironment, also limiting functional repair. Accordingly, we investigated the combination of PLG scaffold bridges with hNSC to improve histological and functional outcome after SCI. In vitro, hNSC culture on a PLG scaffold increased oligodendroglial lineage selection after inflammatory challenge. In vivo, acute PLG bridge implantation followed by chronic hNSC transplantation demonstrated a robust capacity of donor human cells to migrate into PLG bridge channels along regenerating axons and integrate into the host spinal cord as myelinating oligodendrocytes and synaptically integrated neurons. Axons that regenerated through the PLG bridge formed synaptic circuits that connected the ipsilateral forelimb muscle to contralateral motor cortex. hNSC transplantation significantly enhanced the total number of regenerating and myelinated axons identified within the PLG bridge. Finally, the combination of acute bridge implantation and hNSC transplantation exhibited robust improvement in locomotor recovery. These data identify a successful strategy to enhance neurorepair through a temporally layered approach using acute bridge implantation and chronic cell transplantation to spare tissue, promote regeneration, and maximize the function of new axonal connections.
Collapse
Affiliation(s)
- Usha Nekanti
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA.
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
| | - Pooja S Sakthivel
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Atena Zahedi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Dana A Creasman
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Rebecca A Nishi
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Courtney M Dumont
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Katja M Piltti
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Glenn L Guardamondo
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Norbert Hernandez
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Xingyuan Chen
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Hui Song
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Xiaoxiao Lin
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
| | - Joshua Martinez
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Lillian On
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Anita Lakatos
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
| | - Kiran Pawar
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Brian T David
- Department of Neurosurgery, Rush University Medical Center, Chicago, IL, USA
| | - Zhiling Guo
- Department of Medicine & Susan Samueli Integrative Health Institute, University of California, Irvine, CA, USA
| | - Stephanie K Seidlits
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Center for Neural Circuit Mapping, University of California Irvine, Irvine, CA, USA
| | - Lonnie D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Brian J Cummings
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA
- Institute for Memory Impairments & Neurological Disorder, University of California Irvine, Irvine, CA, USA
| | - Aileen J Anderson
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, USA.
- Department of Anatomy and Neurobiology, University of California, Irvine, CA, USA.
- Department of Physical Medicine and Rehabilitation, University of California, Irvine, CA, USA.
- Institute for Memory Impairments & Neurological Disorder, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
24
|
Golan N, Ehrlich D, Bonanno J, O'Brien RF, Murillo M, Kauer SD, Ravindra N, Van Dijk D, Cafferty WB. Anatomical Diversity of the Adult Corticospinal Tract Revealed by Single-Cell Transcriptional Profiling. J Neurosci 2023; 43:7929-7945. [PMID: 37748862 PMCID: PMC10669816 DOI: 10.1523/jneurosci.0811-22.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 09/27/2023] Open
Abstract
The corticospinal tract (CST) forms a central part of the voluntary motor apparatus in all mammals. Thus, injury, disease, and subsequent degeneration within this pathway result in chronic irreversible functional deficits. Current strategies to repair the damaged CST are suboptimal in part because of underexplored molecular heterogeneity within the adult tract. Here, we combine spinal retrograde CST tracing with single-cell RNA sequencing (scRNAseq) in adult male and female mice to index corticospinal neuron (CSN) subtypes that differentially innervate the forelimb and hindlimb. We exploit publicly available datasets to confer anatomic specialization among CSNs and show that CSNs segregate not only along the forelimb and hindlimb axis but also by supraspinal axon collateralization. These anatomically defined transcriptional data allow us to use machine learning tools to build classifiers that discriminate between CSNs and cortical layer 2/3 and nonspinally terminating layer 5 neurons in M1 and separately identify limb-specific CSNs. Using these tools, CSN subtypes can be differentially identified to study postnatal patterning of the CST in vivo, leveraged to screen for novel limb-specific axon growth survival and growth activators in vitro, and ultimately exploited to repair the damaged CST after injury and disease.SIGNIFICANCE STATEMENT Therapeutic interventions designed to repair the damaged CST after spinal cord injury have remained functionally suboptimal in part because of an incomplete understanding of the molecular heterogeneity among subclasses of CSNs. Here, we combine spinal retrograde labeling with scRNAseq and annotate a CSN index by the termination pattern of their primary axon in the cervical or lumbar spinal cord and supraspinal collateral terminal fields. Using machine learning we have confirmed the veracity of our CSN gene lists to train classifiers to identify CSNs among all classes of neurons in primary motor cortex to study the development, patterning, homeostasis, and response to injury and disease, and ultimately target streamlined repair strategies to this critical motor pathway.
Collapse
Affiliation(s)
- Noa Golan
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Daniel Ehrlich
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Psychiatry, Yale University School, New Haven, Connecticut 06511
| | - James Bonanno
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Rory F O'Brien
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Matias Murillo
- Interdepartmental Neuroscience Program, Yale University School, New Haven, Connecticut 06511
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Sierra D Kauer
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
| | - Neal Ravindra
- Department of Internal Medicine, Yale University School, New Haven, Connecticut 06511
- Department of Computer Science, Yale University School, New Haven, Connecticut 06511
| | - David Van Dijk
- Department of Internal Medicine, Yale University School, New Haven, Connecticut 06511
- Department of Computer Science, Yale University School, New Haven, Connecticut 06511
| | - William B Cafferty
- Department of Neurology, Yale University School, New Haven, Connecticut 06511
- Department of Neuroscience, Yale University School, New Haven, Connecticut 06511
| |
Collapse
|
25
|
Carmona LM, Thomas ET, Smith K, Tasic B, Costa RM, Nelson A. Topographical and cell type-specific connectivity of rostral and caudal forelimb corticospinal neuron populations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.17.567623. [PMID: 38014164 PMCID: PMC10680840 DOI: 10.1101/2023.11.17.567623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Corticospinal neurons (CSNs) synapse directly on spinal neurons, a diverse group of neurons with unique structural and functional properties necessary for body movements. CSNs modulating forelimb behavior fractionate into caudal forelimb area (CFA) and rostral forelimb area (RFA) motor cortical populations. Despite their prominence, no studies have mapped the diversity of spinal cell types targeted by CSNs, let alone compare CFA and RFA populations. Here we use anatomical and RNA-sequencing methods to show that CSNs synapse onto a remarkably selective group of spinal cell types, favoring inhibitory populations that regulate motoneuron activity and gate sensory feedback. CFA and RFA CSNs target similar spinal cell types, with notable exceptions that suggest these populations differ in how they influence behavior. Finally, axon collaterals of CFA and RFA CSNs target similar brain regions yet receive surprisingly divergent inputs. These results detail the rules of CSN connectivity throughout the brain and spinal cord for two regions critical for forelimb behavior.
Collapse
|
26
|
Shinotsuka T, Tanaka YR, Terada SI, Hatano N, Matsuzaki M. Layer 5 Intratelencephalic Neurons in the Motor Cortex Stably Encode Skilled Movement. J Neurosci 2023; 43:7130-7148. [PMID: 37699714 PMCID: PMC10601372 DOI: 10.1523/jneurosci.0428-23.2023] [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: 03/08/2023] [Revised: 07/29/2023] [Accepted: 08/26/2023] [Indexed: 09/14/2023] Open
Abstract
The primary motor cortex (M1) and the dorsal striatum play a critical role in motor learning and the retention of learned behaviors. Motor representations of corticostriatal ensembles emerge during motor learning. In the coordinated reorganization of M1 and the dorsal striatum for motor learning, layer 5a (L5a) which connects M1 to the ipsilateral and contralateral dorsal striatum, should be a key layer. Although M1 L5a neurons represent movement-related activity in the late stage of learning, it is unclear whether the activity is retained as a memory engram. Here, using Tlx3-Cre male transgenic mice, we conducted two-photon calcium imaging of striatum-projecting L5a intratelencephalic (IT) neurons in forelimb M1 during late sessions of a self-initiated lever-pull task and in sessions after 6 d of nontraining following the late sessions. We found that trained male animals exhibited stable motor performance before and after the nontraining days. At the same time, we found that M1 L5a IT neurons strongly represented the well-learned forelimb movement but not uninstructed orofacial movements. A subset of M1 L5a IT neurons consistently coded the well-learned forelimb movement before and after the nontraining days. Inactivation of M1 IT neurons after learning impaired task performance when the lever was made heavier or when the target range of the pull distance was narrowed. These results suggest that a subset of M1 L5a IT neurons continuously represent skilled movement after learning and serve to fine-tune the kinematics of well-learned movement.SIGNIFICANCE STATEMENT Motor memory persists even when it is not used for a while. IT neurons in L5a of the M1 gradually come to represent skilled forelimb movements during motor learning. However, it remains to be determined whether these changes persist over a long period and how these neurons contribute to skilled movements. Here, we show that a subset of M1 L5a IT neurons retain information for skilled forelimb movements even after nontraining days. Furthermore, suppressing the activity of these neurons during skilled forelimb movements impaired behavioral stability and adaptability. Our results suggest the importance of M1 L5a IT neurons for tuning skilled forelimb movements over a long period.
Collapse
Affiliation(s)
- Takanori Shinotsuka
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Yasuhiro R Tanaka
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
- Brain Science Institute, Tamagawa University, Machida, Tokyo 194-8610, Japan
| | - Shin-Ichiro Terada
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Natsuki Hatano
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Masanori Matsuzaki
- Department of Physiology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence, University of Tokyo Institutes for Advanced Study, Tokyo 113-0033, Japan
- Brain Functional Dynamics Collaboration Laboratory, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| |
Collapse
|
27
|
Montoro-Gámez C, Nolte H, Molinié T, Evangelista G, Tröder SE, Barth E, Popovic M, Trifunovic A, Zevnik B, Langer T, Rugarli EI. SARM1 deletion delays cerebellar but not spinal cord degeneration in an enhanced mouse model of SPG7 deficiency. Brain 2023; 146:4117-4131. [PMID: 37086482 DOI: 10.1093/brain/awad136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/16/2023] [Accepted: 04/10/2023] [Indexed: 04/24/2023] Open
Abstract
Hereditary spastic paraplegia is a neurological condition characterized by predominant axonal degeneration in long spinal tracts, leading to weakness and spasticity in the lower limbs. The nicotinamide adenine dinucleotide (NAD+)-consuming enzyme SARM1 has emerged as a key executioner of axonal degeneration upon nerve transection and in some neuropathies. An increase in the nicotinamide mononucleotide/NAD+ ratio activates SARM1, causing catastrophic NAD+ depletion and axonal degeneration. However, the role of SARM1 in the pathogenesis of hereditary spastic paraplegia has not been investigated. Here, we report an enhanced mouse model for hereditary spastic paraplegia caused by mutations in SPG7. The eSpg7 knockout mouse carries a deletion in both Spg7 and Afg3l1, a redundant homologue expressed in mice but not in humans. The eSpg7 knockout mice recapitulate the phenotypic features of human patients, showing progressive symptoms of spastic-ataxia and degeneration of axons in the spinal cord as well as the cerebellum. We show that the lack of SPG7 rewires the mitochondrial proteome in both tissues, leading to an early onset decrease in mito-ribosomal subunits and a remodelling of mitochondrial solute carriers and transporters. To interrogate mechanisms leading to axonal degeneration in this mouse model, we explored the involvement of SARM1. Deletion of SARM1 delays the appearance of ataxic signs, rescues mitochondrial swelling and axonal degeneration of cerebellar granule cells and dampens neuroinflammation in the cerebellum. The loss of SARM1 also prevents endoplasmic reticulum abnormalities in long spinal cord axons, but does not halt the degeneration of these axons. Our data thus reveal a neuron-specific interplay between SARM1 and mitochondrial dysfunction caused by lack of SPG7 in hereditary spastic paraplegia.
Collapse
Affiliation(s)
- Carolina Montoro-Gámez
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Hendrik Nolte
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Thibaut Molinié
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Giovanna Evangelista
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Simon E Tröder
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- in vivo Research Facility, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne 50931, Germany
| | - Esther Barth
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
| | - Milica Popovic
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne 50931, Germany
| | - Aleksandra Trifunovic
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| | - Branko Zevnik
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- in vivo Research Facility, Medical Faculty and University Hospital Cologne, University of Cologne, Cologne 50931, Germany
| | - Thomas Langer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- Max Planck Institute for Biology of Ageing, Cologne 50931, Germany
| | - Elena I Rugarli
- Institute for Genetics, University of Cologne, Cologne 50931, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne 50931, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| |
Collapse
|
28
|
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
|
29
|
Lin S, Hari K, Black S, Khatmi A, Fouad K, Gorassini MA, Li Y, Lucas-Osma AM, Fenrich KK, Bennett DJ. Locomotor-related propriospinal V3 neurons produce primary afferent depolarization and modulate sensory transmission to motoneurons. J Neurophysiol 2023; 130:799-823. [PMID: 37609680 PMCID: PMC10650670 DOI: 10.1152/jn.00482.2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023] Open
Abstract
When a muscle is stretched, sensory feedback not only causes reflexes but also leads to a depolarization of sensory afferents throughout the spinal cord (primary afferent depolarization, PAD), readying the whole limb for further disturbances. This sensory-evoked PAD is thought to be mediated by a trisynaptic circuit, where sensory input activates first-order excitatory neurons that activate GABAergic neurons that in turn activate GABAA receptors on afferents to cause PAD, though the identity of these first-order neurons is unclear. Here, we show that these first-order neurons include propriospinal V3 neurons, as they receive extensive sensory input and in turn innervate GABAergic neurons that cause PAD, because optogenetic activation or inhibition of V3 neurons in mice mimics or inhibits sensory-evoked PAD, respectively. Furthermore, persistent inward sodium currents intrinsic to V3 neurons prolong their activity, explaining the prolonged duration of PAD. Also, local optogenetic activation of V3 neurons at one segment causes PAD in other segments, due to the long propriospinal tracts of these neurons, helping to explain the radiating nature of PAD. This in turn facilitates monosynaptic reflex transmission to motoneurons across the spinal cord. In addition, V3 neurons directly innervate proprioceptive afferents (including Ia), causing a glutamate receptor-mediated PAD (glutamate PAD). Finally, increasing the spinal cord excitability with either GABAA receptor blockers or chronic spinal cord injury causes an increase in the glutamate PAD. Overall, we show the V3 neuron has a prominent role in modulating sensory transmission, in addition to its previously described role in locomotion.NEW & NOTEWORTHY Locomotor-related propriospinal neurons depolarize sensory axons throughout the spinal cord by either direct glutamatergic axoaxonic contacts or indirect innervation of GABAergic neurons that themselves form axoaxonic contacts on sensory axons. This depolarization (PAD) increases sensory transmission to motoneurons throughout the spinal cord, readying the sensorimotor system for external disturbances. The glutamate-mediated PAD is particularly adaptable, increasing with either an acute block of GABA receptors or chronic spinal cord injury, suggesting a role in motor recovery.
Collapse
Affiliation(s)
- Shihao Lin
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Krishnapriya Hari
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sophie Black
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Aysan Khatmi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Monica A Gorassini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yaqing Li
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Ana M Lucas-Osma
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - David J Bennett
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
30
|
Alwood JS, Mulavara AP, Iyer J, Mhatre SD, Rosi S, Shelhamer M, Davis C, Jones CW, Mao XW, Desai RI, Whitmire AM, Williams TJ. Circuits and Biomarkers of the Central Nervous System Relating to Astronaut Performance: Summary Report for a NASA-Sponsored Technical Interchange Meeting. Life (Basel) 2023; 13:1852. [PMID: 37763256 PMCID: PMC10532466 DOI: 10.3390/life13091852] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Biomarkers, ranging from molecules to behavior, can be used to identify thresholds beyond which performance of mission tasks may be compromised and could potentially trigger the activation of countermeasures. Identification of homologous brain regions and/or neural circuits related to operational performance may allow for translational studies between species. Three discussion groups were directed to use operationally relevant performance tasks as a driver when identifying biomarkers and brain regions or circuits for selected constructs. Here we summarize small-group discussions in tables of circuits and biomarkers categorized by (a) sensorimotor, (b) behavioral medicine and (c) integrated approaches (e.g., physiological responses). In total, hundreds of biomarkers have been identified and are summarized herein by the respective group leads. We hope the meeting proceedings become a rich resource for NASA's Human Research Program (HRP) and the community of researchers.
Collapse
Affiliation(s)
| | | | - Janani Iyer
- Universities Space Research Association (USRA), Moffett Field, CA 94035, USA
| | | | - Susanna Rosi
- Department of Physical Therapy & Rehabilitation Science, University of California, San Francisco, CA 94110, USA
- Department of Neurological Surgery, University of California, San Francisco, CA 94110, USA
| | - Mark Shelhamer
- Department of Otolaryngology–Head and Neck Surgery, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Catherine Davis
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences (USUHS), Bethesda, MD 20814, USA
| | - Christopher W. Jones
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Biomedical Engineering Sciences (BMES), Loma Linda University Health, Loma Linda, CA 92354, USA
| | - Rajeev I. Desai
- Integrative Neurochemistry Laboratory, Behavioral Biology Program, McLean Hospital-Harvard Medical School, Belmont, MA 02478, USA
| | | | | |
Collapse
|
31
|
Swanson OK, Yevoo PE, Richard D, Maffei A. Altered Thalamocortical Signaling in a Mouse Model of Parkinson's Disease. J Neurosci 2023; 43:6021-6034. [PMID: 37527923 PMCID: PMC10451150 DOI: 10.1523/jneurosci.2871-20.2023] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 06/05/2023] [Accepted: 06/10/2023] [Indexed: 08/03/2023] Open
Abstract
Activation of the primary motor cortex (M1) is important for the execution of skilled movements and motor learning, and its dysfunction contributes to the pathophysiology of Parkinson's disease (PD). A well-accepted idea in PD research, albeit not tested experimentally, is that the loss of midbrain dopamine leads to decreased activation of M1 by the motor thalamus. Here, we report that midbrain dopamine loss altered motor thalamus input in a laminar- and cell type-specific fashion and induced laminar-specific changes in intracortical synaptic transmission. Frequency-dependent changes in synaptic dynamics were also observed. Our results demonstrate that loss of midbrain dopaminergic neurons alters thalamocortical activation of M1 in both male and female mice, and provide novel insights into circuit mechanisms for motor cortex dysfunction in a mouse model of PD.SIGNIFICANCE STATEMENT Loss of midbrain dopamine neurons increases inhibition from the basal ganglia to the motor thalamus, suggesting that it may ultimately lead to reduced activation of primary motor cortex (M1). In contrast with this line of thinking, analysis of M1 activity in patients and animal models of Parkinson's disease report hyperactivation of this region. Our results are the first report that midbrain dopamine loss alters the input-output function of M1 through laminar and cell type specific effects. These findings support and expand on the idea that loss of midbrain dopamine reduces motor cortex activation and provide experimental evidence that reconciles reduced thalamocortical input with reports of altered activation of motor cortex in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Olivia K Swanson
- Department of Neurobiology and Behavior, State University of New York-Stony Brook, Stony Brook, New York 11794
- Graduate Program in Neuroscience, State University of New York-Stony Brook, Stony Brook, New York 11794
| | - Priscilla E Yevoo
- Department of Neurobiology and Behavior, State University of New York-Stony Brook, Stony Brook, New York 11794
- Graduate Program in Neuroscience, State University of New York-Stony Brook, Stony Brook, New York 11794
| | - Dave Richard
- Department of Neurobiology and Behavior, State University of New York-Stony Brook, Stony Brook, New York 11794
| | - Arianna Maffei
- Department of Neurobiology and Behavior, State University of New York-Stony Brook, Stony Brook, New York 11794
- Graduate Program in Neuroscience, State University of New York-Stony Brook, Stony Brook, New York 11794
| |
Collapse
|
32
|
Mykins M, Layo-Carris D, Dunn LR, Skinner DW, McBryar AH, Perez S, Shultz TR, Willems A, Lau BYB, Hong T, Krishnan K. Wild-type MECP2 expression coincides with age-dependent sensory phenotypes in a female mouse model for Rett syndrome. J Neurosci Res 2023; 101:1236-1258. [PMID: 37026482 PMCID: PMC10332853 DOI: 10.1002/jnr.25190] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/07/2023] [Accepted: 03/12/2023] [Indexed: 04/08/2023]
Abstract
Rett syndrome is characterized by an early period of typical development and then, regression of learned motor and speech skills in girls. Loss of MECP2 protein is thought to cause Rett syndrome phenotypes. The specific underlying mechanisms from typical developmental trajectory to regression features throughout life are unclear. Lack of established timelines to study the molecular, cellular, and behavioral features of regression in female mouse models is a major contributing factor. Due to random X-chromosome inactivation, female patients with Rett syndrome and female mouse models for Rett syndrome (Mecp2Heterozygous , Het) express a functional copy of wild-type MECP2 protein in approximately half of all cells. As MECP2 expression is regulated during early postnatal development and experience, we characterized the expression of wild-type MECP2 in the primary somatosensory cortex of female Het mice. Here, we report increased MECP2 levels in non-parvalbumin-positive neurons of 6-week-old adolescent Het relative to age-matched wild-type controls, while also displaying typical levels of perineuronal net expression in the barrel field subregion of the primary somatosensory cortex, mild tactile sensory perception deficits, and efficient pup retrieval behavior. In contrast, 12-week-old adult Het express MECP2 at levels similar to age-matched wild-type mice, show increased perineuronal net expression in the cortex, and display significant tactile sensory perception deficits. Thus, we have identified a set of behavioral metrics and the cellular substrates to study regression during a specific time in the female Het mouse model, which coincides with changes in wild-type MECP2 expression. We speculate that the precocious increase in MECP2 expression within specific cell types of adolescent Het may provide compensatory benefits at the behavioral level, while the inability to further increase MECP2 levels leads to regressive behavioral phenotypes over time.
Collapse
Affiliation(s)
- Michael Mykins
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Dana Layo-Carris
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Logan Reid Dunn
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - David Wilson Skinner
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Alexandra Hart McBryar
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Sarah Perez
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Trinity Rose Shultz
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Andrew Willems
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Billy You Bun Lau
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Tian Hong
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| | - Keerthi Krishnan
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, Tennessee, USA
| |
Collapse
|
33
|
Xiong W, Jin L, Zhao Y, Wu Y, Dong J, Guo Z, Zhu M, Dai Y, Pan Y, Zhu X. Deletion of Transferrin Receptor 1 in Parvalbumin Interneurons Induces a Hereditary Spastic Paraplegia-Like Phenotype. J Neurosci 2023; 43:5092-5113. [PMID: 37308296 PMCID: PMC10325000 DOI: 10.1523/jneurosci.2277-22.2023] [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: 12/12/2022] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/14/2023] Open
Abstract
Hereditary spastic paraplegia (HSP) is a severe neurodegenerative movement disorder, the underlying pathophysiology of which remains poorly understood. Mounting evidence has suggested that iron homeostasis dysregulation can lead to motor function impairment. However, whether deficits in iron homeostasis are involved in the pathophysiology of HSP remains unknown. To address this knowledge gap, we focused on parvalbumin-positive (PV+) interneurons, a large category of inhibitory neurons in the central nervous system, which play a critical role in motor regulation. The PV+ interneuron-specific deletion of the gene encoding transferrin receptor 1 (TFR1), a key component of the neuronal iron uptake machinery, induced severe progressive motor deficits in both male and female mice. In addition, we observed skeletal muscle atrophy, axon degeneration in the spinal cord dorsal column, and alterations in the expression of HSP-related proteins in male mice with Tfr1 deletion in the PV+ interneurons. These phenotypes were highly consistent with the core clinical features of HSP cases. Furthermore, the effects on motor function induced by Tfr1 ablation in PV+ interneurons were mostly concentrated in the dorsal spinal cord; however, iron repletion partly rescued the motor defects and axon loss seen in both sexes of conditional Tfr1 mutant mice. Our study describes a new mouse model for mechanistic and therapeutic studies relating to HSP and provides novel insights into iron metabolism in spinal cord PV+ interneurons and its role in the regulation of motor functions.SIGNIFICANCE STATEMENT Iron is crucial for neuronal functioning. Mounting evidence suggests that iron homeostasis dysregulation can induce motor function deficits. Transferrin receptor 1 (TFR1) is thought to be the key component in neuronal iron uptake. We found that deletion of Tfr1 in parvalbumin-positive (PV+) interneurons in mice induced severe progressive motor deficits, skeletal muscle atrophy, axon degeneration in the spinal cord dorsal column, and alterations in the expression of hereditary spastic paraplegia (HSP)-related proteins. These phenotypes were highly consistent with the core clinical features of HSP cases and partly rescued by iron repletion. This study describes a new mouse model for the study of HSP and provides novel insights into iron metabolism in spinal cord PV+ interneurons.
Collapse
Affiliation(s)
- Wenchao Xiong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liqiang Jin
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yulu Zhao
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yu Wu
- School of Psychology, Shenzhen University, Shenzhen 518060, China
| | - Jinghua Dong
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhixin Guo
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minzhen Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongfeng Dai
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yida Pan
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xinhong Zhu
- School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
- School of Psychology, Shenzhen University, Shenzhen 518060, China
- Research Center for Brain Health, Pazhou Lab, Guangzhou 510330, China
| |
Collapse
|
34
|
Maurer L, Brown M, Saggi T, Cardiges A, Kolarcik CL. Hindlimb muscle representations in mouse motor cortex defined by viral tracing. Front Neuroanat 2023; 17:965318. [PMID: 37303816 PMCID: PMC10248224 DOI: 10.3389/fnana.2023.965318] [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: 06/09/2022] [Accepted: 05/02/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Descending pathways from the cortex to the spinal cord are involved in the control of natural movement. Although mice are widely used to study the neurobiology of movement and as models of neurodegenerative disease, an understanding of motor cortical organization is lacking, particularly for hindlimb muscles. Methods In this study, we used the retrograde transneuronal transport of rabies virus to compare the organization of descending cortical projections to fast- and slow-twitch hindlimb muscles surrounding the ankle joint in mice. Results Although the initial stage of virus transport from the soleus muscle (predominantly slow-twitch) appeared to be more rapid than that associated with the tibialis anterior muscle (predominantly fast-twitch), the rate of further transport of virus to cortical projection neurons in layer V was equivalent for the two injected muscles. After appropriate survival times, dense concentrations of layer V projection neurons were identified in three cortical areas: the primary motor cortex (M1), secondary motor cortex (M2), and primary somatosensory cortex (S1). Discussion The origin of the cortical projections to each of the two injected muscles overlapped almost entirely within these cortical areas. This organization suggests that cortical projection neurons maintain a high degree of specificity; that is, even when cortical projection neurons are closely located, each neuron could have a distinct functional role (controlling fast- versus slow-twitch and/or extensor versus flexor muscles). Our results represent an important addition to the understanding of the mouse motor system and lay the foundation for future studies investigating the mechanisms underlying motor system dysfunction and degeneration in diseases such as amyotrophic lateral sclerosis and spinal muscular atrophy.
Collapse
Affiliation(s)
- Lauren Maurer
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Maia Brown
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tamandeep Saggi
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Alexia Cardiges
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Christi L. Kolarcik
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
- LiveLikeLou Center for ALS Research, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
- Center for the Neural Basis of Cognition, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
35
|
Metz K, Matos IC, Hari K, Bseis O, Afsharipour B, Lin S, Singla R, Fenrich KK, Li Y, Bennett DJ, Gorassini MA. Post-activation depression from primary afferent depolarization (PAD) produces extensor H-reflex suppression following flexor afferent conditioning. J Physiol 2023; 601:1925-1956. [PMID: 36928599 PMCID: PMC11064783 DOI: 10.1113/jp283706] [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: 08/08/2022] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Suppression of the extensor H-reflex by flexor afferent conditioning is thought to be produced by a long-lasting inhibition of extensor Ia afferent terminals via GABAA receptor-activated primary afferent depolarization (PAD). Given the recent finding that PAD does not produce presynaptic inhibition of Ia afferent terminals, we examined in 28 participants if H-reflex suppression is instead mediated by post-activation depression of the extensor Ia afferents triggered by PAD-evoked spikes and/or by a long-lasting inhibition of the extensor motoneurons. A brief conditioning vibration of the flexor tendon suppressed both the extensor soleus H-reflex and the tonic discharge of soleus motor units out to 150 ms following the vibration, suggesting that part of the H-reflex suppression during this period was mediated by postsynaptic inhibition of the extensor motoneurons. When activating the flexor afferents electrically to produce conditioning, the soleus H-reflex was also suppressed but only when a short-latency reflex was evoked in the soleus muscle by the conditioning input itself. In mice, a similar short-latency reflex was evoked when optogenetic or afferent activation of GABAergic (GAD2+ ) neurons produced a large enough PAD to evoke orthodromic spikes in the test Ia afferents, causing post-activation depression of subsequent monosynaptic EPSPs. The long duration of this post-activation depression and related H-reflex suppression (seconds) was similar to rate-dependent depression that is also due to post-activation depression. We conclude that extensor H-reflex inhibition by brief flexor afferent conditioning is produced by both post-activation depression of extensor Ia afferents and long-lasting inhibition of extensor motoneurons, rather than from PAD inhibiting Ia afferent terminals. KEY POINTS: Suppression of extensor H-reflexes by flexor afferent conditioning was thought to be mediated by GABAA receptor-mediated primary afferent depolarization (PAD) shunting action potentials in the Ia afferent terminal. In line with recent findings that PAD has a facilitatory role in Ia afferent conduction, we show here that when large enough, PAD can evoke orthodromic spikes that travel to the Ia afferent terminal to evoke EPSPs in the motoneuron. These PAD-evoked spikes also produce post-activation depression of Ia afferent terminals and may mediate the short- and long-lasting suppression of extensor H-reflexes in response to flexor afferent conditioning. Our findings highlight that we must re-examine how changes in the activation of GABAergic interneurons and PAD following nervous system injury or disease affects the regulation of Ia afferent transmission to spinal neurons and ultimately motor dysfunction in these disorders.
Collapse
Affiliation(s)
- Krista Metz
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Isabel Concha Matos
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Krishnapriya Hari
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Omayma Bseis
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Babak Afsharipour
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Shihao Lin
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Rahul Singla
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Yaqing Li
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - David J Bennett
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
| | - Monica A Gorassini
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
36
|
Yang L, Martin JH. Effects of motor cortex neuromodulation on the specificity of corticospinal tract spinal axon outgrowth and targeting in rats. Brain Stimul 2023; 16:759-771. [PMID: 37094762 PMCID: PMC10501380 DOI: 10.1016/j.brs.2023.04.014] [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: 12/18/2022] [Revised: 04/01/2023] [Accepted: 04/19/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND Neural activity helps construct neural circuits during development and this function is leveraged by neuromodulation protocols to promote connectivity and repair in maturity. Neuromodulation targeting the motor cortex (MCX) strengthens connections for evoking muscle contraction (MEPs). Mechanisms include promoting local MCX and corticospinal tract (CST) synaptic efficacy and also axon terminal structural changes. OBJECTIVE In this study, we address the question of potential causality between neuronal activation and the neuronal structural response. METHODS We used patterned optogenetic activation (ChR2-EYFP), daily for 10-days, to deliver intermittent theta burst stimulation (iTBS) to activate MCX neurons within the forelimb representation in healthy rats, while differentiating them from neurons in the same population that were not activated. We used chemogenetic DREADD activation to produce a daily period of non-patterned neuronal activation. RESULTS We found a significant increase in CST axon length, axon branching, contacts targeted to a class of premotor interneuron (Chx10), as well as projections into the motor pools in the ventral horn in optically activated but not neighboring non-activated neurons. A period of 2-h of continuous activation daily for 10 days using DREADD chemogenetic activation with systemic clozapine N-oxide (CNO) administration also increased CST axon length and branching, but not the ventral horn and Chx10 targeting effects. Both patterned optical and chemogenetic activation reduced MCX MEP thresholds. CONCLUSION Our findings show that targeting of CST axon sprouting is dependent on patterned activation, but that CST spinal axon outgrowth and branching are not. Our optogenetic findings, by distinguishing optically activated and non-activated CST axons, suggests that the switch for activity-dependent axonal outgrowth is neuron-intrinsic.
Collapse
Affiliation(s)
- Lillian Yang
- Department of Molecular, Cellular, and Biomedical Sciences, Center for Discovery and Innovation, City University of New York School of Medicine, New York, NY, USA
| | - John H Martin
- Department of Molecular, Cellular, and Biomedical Sciences, Center for Discovery and Innovation, City University of New York School of Medicine, New York, NY, USA; Neuroscience Program, Graduate Center of the City University of New York, New York, NY, USA.
| |
Collapse
|
37
|
Metz K, Matos IC, Li Y, Afsharipour B, Thompson CK, Negro F, Quinlan KA, Bennett DJ, Gorassini MA. Facilitation of sensory transmission to motoneurons during cortical or sensory-evoked primary afferent depolarization (PAD) in humans. J Physiol 2023; 601:1897-1924. [PMID: 36916205 PMCID: PMC11037101 DOI: 10.1113/jp284275] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Sensory and corticospinal tract (CST) pathways activate spinal GABAergic interneurons that have axoaxonic connections onto proprioceptive (Ia) afferents that cause long-lasting depolarizations (termed primary afferent depolarization, PAD). In rodents, sensory-evoked PAD is produced by GABAA receptors at nodes of Ranvier in Ia afferents, rather than at presynaptic terminals, and facilitates spike propagation to motoneurons by preventing branch-point failures, rather than causing presynaptic inhibition. We examined in 40 human participants whether putative activation of Ia-PAD by sensory or CST pathways can also facilitate Ia afferent activation of motoneurons via the H-reflex. H-reflexes in several leg muscles were facilitated by prior conditioning from low-threshold proprioceptive, cutaneous or CST pathways, with a similar long-lasting time course (∼200 ms) to phasic PAD measured in rodent Ia afferents. Long trains of cutaneous or proprioceptive afferent conditioning produced longer-lasting facilitation of the H-reflex for up to 2 min, consistent with tonic PAD in rodent Ia afferents mediated by nodal α5-GABAA receptors for similar stimulation trains. Facilitation of H-reflexes by this conditioning was likely not mediated by direct facilitation of the motoneurons because isolated stimulation of sensory or CST pathways did not alone facilitate the tonic firing rate of motor units. Furthermore, cutaneous conditioning increased the firing probability of single motor units (motoneurons) during the H-reflex without increasing their firing rate at this time, indicating that the underlying excitatory postsynaptic potential was more probable, but not larger. These results are consistent with sensory and CST pathways activating nodal GABAA receptors that reduce intermittent failure of action potentials propagating into Ia afferent branches. KEY POINTS: Controlled execution of posture and movement requires continually adjusted feedback from peripheral sensory pathways, especially those that carry proprioceptive information about body position, movement and effort. It was previously thought that the flow of proprioceptive feedback from Ia afferents was only reduced by GABAergic neurons in the spinal cord that sent axoaxonic projections to the terminal endings of sensory axons (termed GABAaxo neurons). Based on new findings in rodents, we provide complementary evidence in humans to suggest that sensory and corticospinal pathways known to activate GABAaxo neurons that project to dorsal parts of the Ia afferent also increase the flow of proprioceptive feedback to motoneurons in the spinal cord. These findings support a new role for spinal GABAaxo neurons in facilitating afferent feedback to the spinal cord during voluntary or reflexive movements.
Collapse
Affiliation(s)
- Krista Metz
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Isabel Concha Matos
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Yaqing Li
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Babak Afsharipour
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | | | - Francesco Negro
- Clinical and Experimental Sciences, Universita degli Studi di Brescia, Brescia, Italy
| | - Katharina A Quinlan
- George and Anne Ryan Institute for Neuroscience, Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, USA
| | - David J Bennett
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Monica A Gorassini
- Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| |
Collapse
|
38
|
Frezel N, Ranucci M, Foster E, Wende H, Pelczar P, Mendes R, Ganley RP, Werynska K, d'Aquin S, Beccarini C, Birchmeier C, Zeilhofer HU, Wildner H. c-Maf-positive spinal cord neurons are critical elements of a dorsal horn circuit for mechanical hypersensitivity in neuropathy. Cell Rep 2023; 42:112295. [PMID: 36947543 PMCID: PMC10157139 DOI: 10.1016/j.celrep.2023.112295] [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: 01/13/2022] [Revised: 02/02/2023] [Accepted: 03/06/2023] [Indexed: 03/23/2023] Open
Abstract
Corticospinal tract (CST) neurons innervate the deep spinal dorsal horn to sustain chronic neuropathic pain. The majority of neurons targeted by the CST are interneurons expressing the transcription factor c-Maf. Here, we used intersectional genetics to decipher the function of these neurons in dorsal horn sensory circuits. We find that excitatory c-Maf (c-MafEX) neurons receive sensory input mainly from myelinated fibers and target deep dorsal horn parabrachial projection neurons and superficial dorsal horn neurons, thereby connecting non-nociceptive input to nociceptive output structures. Silencing c-MafEX neurons has little effect in healthy mice but alleviates mechanical hypersensitivity in neuropathic mice. c-MafEX neurons also receive input from inhibitory c-Maf and parvalbumin neurons, and compromising inhibition by these neurons caused mechanical hypersensitivity and spontaneous aversive behaviors reminiscent of c-MafEX neuron activation. Our study identifies c-MafEX neurons as normally silent second-order nociceptors that become engaged in pathological pain signaling upon loss of inhibitory control.
Collapse
Affiliation(s)
- Noémie Frezel
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | - Matteo Ranucci
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | - Edmund Foster
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | | | - Pawel Pelczar
- Center for Transgenic Models (CTM), University of Basel, 4001 Basel, Switzerland
| | - Raquel Mendes
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | - Robert P Ganley
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | - Karolina Werynska
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | - Simon d'Aquin
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | - Camilla Beccarini
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland
| | | | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland; Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zürich, 8092 Zürich, Switzerland.
| | - Hendrik Wildner
- Institute of Pharmacology and Toxicology, University of Zürich, 8057 Zürich, Switzerland.
| |
Collapse
|
39
|
Itoh Y, Sahni V, Shnider SJ, McKee H, Macklis JD. Inter-axonal molecular crosstalk via Lumican proteoglycan sculpts murine cervical corticospinal innervation by distinct subpopulations. Cell Rep 2023; 42:112182. [PMID: 36934325 PMCID: PMC10167627 DOI: 10.1016/j.celrep.2023.112182] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/07/2022] [Accepted: 02/14/2023] [Indexed: 03/19/2023] Open
Abstract
How CNS circuits sculpt their axonal arbors into spatially and functionally organized domains is not well understood. Segmental specificity of corticospinal connectivity is an exemplar for such regional specificity of many axon projections. Corticospinal neurons (CSN) innervate spinal and brainstem targets with segmental precision, controlling voluntary movement. Multiple molecularly distinct CSN subpopulations innervate the cervical cord for evolutionarily enhanced precision of forelimb movement. Evolutionarily newer CSNBC-lat exclusively innervate bulbar-cervical targets, while CSNmedial are heterogeneous; distinct subpopulations extend axons to either bulbar-cervical or thoraco-lumbar segments. We identify that Lumican controls balance of cervical innervation between CSNBC-lat and CSNmedial axons during development, which is maintained into maturity. Lumican, an extracellular proteoglycan expressed by CSNBC-lat, non-cell-autonomously suppresses cervical collateralization by multiple CSNmedial subpopulations. This inter-axonal molecular crosstalk between CSN subpopulations controls murine corticospinal circuitry refinement and forelimb dexterity. Such crosstalk is generalizable beyond the corticospinal system for evolutionary incorporation of new neuron populations into preexisting circuitry.
Collapse
Affiliation(s)
- Yasuhiro Itoh
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Vibhu Sahni
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Sara J Shnider
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Holly McKee
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA
| | - Jeffrey D Macklis
- Department of Stem Cell and Regenerative Biology and Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
40
|
Wang Q, Zhao S, He Z, Zhang S, Jiang X, Zhang T, Liu T, Liu C, Han J. Modeling functional difference between gyri and sulci within intrinsic connectivity networks. Cereb Cortex 2023; 33:933-947. [PMID: 35332916 DOI: 10.1093/cercor/bhac111] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/17/2022] [Accepted: 02/18/2022] [Indexed: 11/12/2022] Open
Abstract
Recently, the functional roles of the human cortical folding patterns have attracted increasing interest in the neuroimaging community. However, most existing studies have focused on the gyro-sulcal functional relationship on a whole-brain scale but possibly overlooked the localized and subtle functional differences of brain networks. Actually, accumulating evidences suggest that functional brain networks are the basic unit to realize the brain function; thus, the functional relationships between gyri and sulci still need to be further explored within different functional brain networks. Inspired by these evidences, we proposed a novel intrinsic connectivity network (ICN)-guided pooling-trimmed convolutional neural network (I-ptFCN) to revisit the functional difference between gyri and sulci. By testing the proposed model on the task functional magnetic resonance imaging (fMRI) datasets of the Human Connectome Project, we found that the classification accuracy of gyral and sulcal fMRI signals varied significantly for different ICNs, indicating functional heterogeneity of cortical folding patterns in different brain networks. The heterogeneity may be contributed by sulci, as only sulcal signals show heterogeneous frequency features across different ICNs, whereas the frequency features of gyri are homogeneous. These results offer novel insights into the functional difference between gyri and sulci and enlighten the functional roles of cortical folding patterns.
Collapse
Affiliation(s)
- Qiyu Wang
- School of Automation, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Shijie Zhao
- School of Automation, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Zhibin He
- School of Automation, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Shu Zhang
- School of Computer Science, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Xi Jiang
- School of Life Science and Technology, MOE Key Lab for Neuroinformation, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Tuo Zhang
- School of Automation, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Tianming Liu
- Cortical Architecture Imaging and Discovery Lab, Department of Computer Science and Bioimaging Research Center, The University of Georgia, Athens, GA 30605, United States
| | - Cirong Liu
- CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai 200031, China
| | - Junwei Han
- School of Automation, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| |
Collapse
|
41
|
Berzanskyte I, Riccio F, Machado CB, Bradbury EJ, Lieberam I. Enrichment of human embryonic stem cell-derived V3 interneurons using an Nkx2-2 gene-specific reporter. Sci Rep 2023; 13:2008. [PMID: 36737643 PMCID: PMC9898512 DOI: 10.1038/s41598-023-29165-z] [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: 08/19/2022] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
V3 spinal interneurons are a key element of the spinal circuits, which control motor function. However, to date, there are no effective ways of deriving a pure V3 population from human pluripotent stem cells. Here, we report a method for differentiation and isolation of spinal V3 interneurons, combining extrinsic factor-mediated differentiation and magnetic activated cell sorting. We found that differentiation of V3 progenitors can be enhanced with a higher concentration of Sonic Hedgehog agonist, as well as culturing cells in 3D format. To enable V3 progenitor purification from mixed differentiation cultures, we developed a transgene reporter, with a part of the regulatory region of V3-specific gene Nkx2-2 driving the expression of a membrane marker CD14. We found that in human cells, NKX2-2 initially exhibited co-labelling with motor neuron progenitor marker, but V3 specificity emerged as the differentiation culture progressed. At these later differentiation timepoints, we were able to enrich V3 progenitors labelled with CD14 to ~ 95% purity, and mature them to postmitotic V3 interneurons. This purification tool for V3 interneurons will be useful for in vitro disease modeling, studies of normal human neural development and potential cell therapies for disorders of the spinal cord.
Collapse
Affiliation(s)
- Ieva Berzanskyte
- Centre for Gene Therapy and Regenerative Medicine, Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, 28th Floor Tower Wing, Guy's Campus, Great Maze Pond, London, SE1 9RT, UK.
- The Wolfson Centre for Age-Related Diseases, King's College London, London, UK.
| | - Federica Riccio
- Centre for Gene Therapy and Regenerative Medicine, Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, 28th Floor Tower Wing, Guy's Campus, Great Maze Pond, London, SE1 9RT, UK
| | - Carolina Barcellos Machado
- Centre for Gene Therapy and Regenerative Medicine, Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, 28th Floor Tower Wing, Guy's Campus, Great Maze Pond, London, SE1 9RT, UK
| | | | - Ivo Lieberam
- Centre for Gene Therapy and Regenerative Medicine, Centre for Developmental Neurobiology, MRC Centre for Neurodevelopmental Disorders, King's College London, 28th Floor Tower Wing, Guy's Campus, Great Maze Pond, London, SE1 9RT, UK.
| |
Collapse
|
42
|
Yang W, Kanodia H, Arber S. Structural and functional map for forelimb movement phases between cortex and medulla. Cell 2023; 186:162-177.e18. [PMID: 36608651 PMCID: PMC9842395 DOI: 10.1016/j.cell.2022.12.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/10/2022] [Accepted: 12/05/2022] [Indexed: 01/07/2023]
Abstract
The cortex influences movement by widespread top-down projections to many nervous system regions. Skilled forelimb movements require brainstem circuitry in the medulla; however, the logic of cortical interactions with these neurons remains unexplored. Here, we reveal a fine-grained anatomical and functional map between anterior cortex (AC) and medulla in mice. Distinct cortical regions generate three-dimensional synaptic columns tiling the lateral medulla, topographically matching the dorso-ventral positions of postsynaptic neurons tuned to distinct forelimb action phases. Although medial AC (MAC) terminates ventrally and connects to forelimb-reaching-tuned neurons and its silencing impairs reaching, lateral AC (LAC) influences dorsally positioned neurons tuned to food handling, and its silencing impairs handling. Cortico-medullary neurons also extend collaterals to other subcortical structures through a segregated channel interaction logic. Our findings reveal a precise alignment between cortical location, its function, and specific forelimb-action-tuned medulla neurons, thereby clarifying interaction principles between these two key structures and beyond.
Collapse
Affiliation(s)
- Wuzhou Yang
- Biozentrum, Department of Cell Biology, University of Basel, 4056 Basel, Switzerland,Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Harsh Kanodia
- Biozentrum, Department of Cell Biology, University of Basel, 4056 Basel, Switzerland,Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Silvia Arber
- Biozentrum, Department of Cell Biology, University of Basel, 4056 Basel, Switzerland,Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland,Corresponding author
| |
Collapse
|
43
|
Vieillard J, Franck MCM, Hartung S, Jakobsson JET, Ceder MM, Welsh RE, Lagerström MC, Kullander K. Adult spinal Dmrt3 neurons receive direct somatosensory inputs from ipsi- and contralateral primary afferents and from brainstem motor nuclei. J Comp Neurol 2023; 531:5-24. [PMID: 36214727 PMCID: PMC9828095 DOI: 10.1002/cne.25405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 01/12/2023]
Abstract
In the spinal cord, sensory-motor circuits controlling motor activity are situated in the dorso-ventral interface. The neurons identified by the expression of the transcription factor Doublesex and mab-3 related transcription factor 3 (Dmrt3) have previously been associated with the coordination of locomotion in horses (Equus caballus, Linnaeus, 1758), mice (Mus musculus, Linnaeus, 1758), and zebrafish (Danio rerio, F. Hamilton, 1822). Based on earlier studies, we hypothesized that, in mice, these neurons may be positioned to receive sensory and central inputs to relay processed commands to motor neurons. Thus, we investigated the presynaptic inputs to spinal Dmrt3 neurons using monosynaptic retrograde replication-deficient rabies tracing. The analysis showed that lumbar Dmrt3 neurons receive inputs from intrasegmental neurons, and intersegmental neurons from the cervical, thoracic, and sacral segments. Some of these neurons belong to the excitatory V2a interneurons and to plausible Renshaw cells, defined by the expression of Chx10 and calbindin, respectively. We also found that proprioceptive primary sensory neurons of type Ia2, Ia3, and Ib, defined by the expression of calbindin, calretinin, and Brn3c, respectively, provide presynaptic inputs to spinal Dmrt3 neurons. In addition, we demonstrated that Dmrt3 neurons receive inputs from brain areas involved in motor regulation, including the red nucleus, primary sensory-motor cortex, and pontine nuclei. In conclusion, adult spinal Dmrt3 neurons receive inputs from motor-related brain areas as well as proprioceptive primary sensory neurons and have been shown to connect directly to motor neurons. Dmrt3 neurons are thus positioned to provide sensory-motor control and their connectivity is suggestive of the classical reflex pathways present in the spinal cord.
Collapse
Affiliation(s)
- Jennifer Vieillard
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Marina C. M. Franck
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden,Present address: Department of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Sunniva Hartung
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Jon E. T. Jakobsson
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Mikaela M. Ceder
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Robert E. Welsh
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Malin C. Lagerström
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| | - Klas Kullander
- Department of Immunology, Genetics and PathologyUppsala UniversityUppsalaSweden
| |
Collapse
|
44
|
Nakamura Y, Kurabe M, Matsumoto M, Sato T, Miytashita S, Hoshina K, Kamiya Y, Tainaka K, Matsuzawa H, Ohno N, Ueno M. Cerebrospinal fluid-contacting neuron tracing reveals structural and functional connectivity for locomotion in the mouse spinal cord. eLife 2023; 12:83108. [PMID: 36805807 PMCID: PMC9943067 DOI: 10.7554/elife.83108] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 02/03/2023] [Indexed: 02/22/2023] Open
Abstract
Cerebrospinal fluid-contacting neurons (CSF-cNs) are enigmatic mechano- or chemosensory cells lying along the central canal of the spinal cord. Recent studies in zebrafish larvae and lampreys have shown that CSF-cNs control postures and movements via spinal connections. However, the structures, connectivity, and functions in mammals remain largely unknown. Here we developed a method to genetically target mouse CSF-cNs that highlighted structural connections and functions. We first found that intracerebroventricular injection of adeno-associated virus with a neuron-specific promoter and Pkd2l1-Cre mice specifically labeled CSF-cNs. Single-cell labeling of 71 CSF-cNs revealed rostral axon extensions of over 1800 μm in unmyelinated bundles in the ventral funiculus and terminated on CSF-cNs to form a recurrent circuitry, which was further determined by serial electron microscopy and electrophysiology. CSF-cNs were also found to connect with axial motor neurons and premotor interneurons around the central canal and within the axon bundles. Chemogenetic CSF-cNs inactivation reduced speed and step frequency during treadmill locomotion. Our data revealed the basic structures and connections of mouse CSF-cNs to control spinal motor circuits for proper locomotion. The versatile methods developed in this study will contribute to further understanding of CSF-cN functions in mammals.
Collapse
Affiliation(s)
- Yuka Nakamura
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Miyuki Kurabe
- Division of Anesthesiology, Niigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Mami Matsumoto
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological SciencesOkazakiJapan,Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical SciencesNagoyaJapan
| | - Tokiharu Sato
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Satoshi Miytashita
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Kana Hoshina
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Yoshinori Kamiya
- Division of Anesthesiology, Niigata University Graduate School of Medical and Dental SciencesNiigataJapan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| | - Hitoshi Matsuzawa
- Center for Advanced Medicine and Clinical Research, Kashiwaba Neurosurgical HospitalSapporoJapan,Center for Integrated Human Brain Science, Niigata UniversityNiigataJapan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, School of MedicineShimotsukeJapan,Division of Ultrastructural Research, National Institute for Physiological SciencesOkazakiJapan
| | - Masaki Ueno
- Department of System Pathology for Neurological Disorders, Brain Research Institute, Niigata UniversityNiigataJapan
| |
Collapse
|
45
|
Wang Z, Duan H, Hao F, Hao P, Zhao W, Gao Y, Gu Y, Song J, Li X, Yang Z. Circuit reconstruction of newborn neurons after spinal cord injury in adult rats via an NT3-chitosan scaffold. Prog Neurobiol 2023; 220:102375. [PMID: 36410665 DOI: 10.1016/j.pneurobio.2022.102375] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 11/07/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
An implanted neurotrophin-3 (NT3)-chitosan scaffold can recruit endogenous neural stem cells to migrate to a lesion region and differentiate into mature neurons after adult spinal cord injury (SCI). However, the identities of these newborn neurons and whether they can form functional synapses and circuits to promote recovery after paraplegia remain unknown. By using combined advanced technologies, we revealed here that the newborn neurons of several subtypes received synaptic input from the corticospinal tract (CST), rubrospinal tract (RST), and supraspinal tracts. They formed a functional neural circuit at the injured spinal region, further driving the local circuits beneath the lesion. Our results showed that the NT3-chitosan scaffold facilitated the maturation of spinal neurons and the reestablishment of the spinal neural circuit in the lesion region 12 weeks after SCI. Transsynaptic virus experiments revealed that these newborn spinal neurons received synaptic connections from the CST and RST and drove the neural circuit beneath the lesion via newly formed synapses. These re-established circuits successfully recovered the formation and function of the neuromuscular junction (NMJ) beneath the lesion spinal segments. These findings suggest that the NT3-chitosan scaffold promotes the formation of relay neural circuits to accommodate various types of brain descending inputs and facilitate functional recovery after paraplegia.
Collapse
Affiliation(s)
- Zijue Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Fei Hao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing 100191, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Yiming Gu
- Physical Education Department, Capital University of Economics and Business, Beijing 100070, China
| | - Jianren Song
- Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai 200434, China; Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China.
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
46
|
Huang Z, Sun L, Zheng X, Zhang Y, Zhu Y, Chen T, Chen Z, Ja L, OuYang L, Zhu Y, Chen S, Lei W. A neural tract tracing study on synaptic connections for cortical glutamatergic terminals and cervical spinal calretinin neurons in rats. Front Neural Circuits 2023; 17:1086873. [PMID: 37187913 PMCID: PMC10175624 DOI: 10.3389/fncir.2023.1086873] [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: 11/01/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
The cerebral cortex innervates motor neurons in the anterior horn of the spinal cord by regulating of interneurons. At present, nerve tracing, immunohistochemistry, and immunoelectron microscopy are used to explore and confirm the characteristics of synaptic connections between the corticospinal tract (CST) and cervical spinal calretinin (Cr) interneurons. Our morphological results revealed that (1) biotinylated dextran amine labeled (BDA+) fibers from the cerebral cortex primarily presented a contralateral spinal distribution, with a denser distribution in the ventral horn (VH) than in the dorsal horn (DH). An electron microscope (EM) showed that BDA+ terminals formed asymmetric synapses with spinal neurons, and their mean labeling rate was not different between the DH and VH. (2) Cr-immunoreactive (Cr+) neurons were unevenly distributed throughout the spinal gray matter, and were denser and larger in the VH than in the DH. At the single labeling electron microscope (EM) level, the labeling rate of Cr+ dendrites was higher in the VH than in the DH, in which Cr+ dendrites mainly received asymmetric synaptic inputs, and between the VH and DH. (3) Immunofluorescence triple labeling showed obvious apposition points among BDA+ terminals, synaptophysin and Cr+ dendrites, with a higher density in the VH than in the DH. (4) Double labeling in EM, BDA+ terminals and Cr+ dendrites presented the same pattern, BDA+ terminals formed asymmetric synapses either with Cr+ dendrites or Cr negative (Cr-) dendrites, and Cr+ dendrites received either BDA+ terminals or BDA- synaptic inputs. The average percentage of BDA+ terminals targeting Cr+ dendrites was higher in the VH than in the DH, but the percentage of BDA+ terminals targeting Cr- dendrites was prominently higher than that targeting Cr+ dendrites. There was no difference in BDA+ terminal size. The percentage rate for Cr+ dendrites receiving BDA+ terminal inputs was lower than that receiving BDA- terminal inputs, and the BDA+ terminal size was larger than the BDA- terminal size received by Cr+ dendrites. The present morphological results suggested that spinal Cr+ interneurons are involved in the regulatory process of the cortico-spinal pathway.
Collapse
Affiliation(s)
- Ziyun Huang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Liping Sun
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xuefeng Zheng
- Neuroscience Laboratory for Cognitive and Developmental Disorders, Department of Anatomy, Medical College of Jinan University, Guangzhou, China
| | - Ye Zhang
- Neuroscience Laboratory for Cognitive and Developmental Disorders, Department of Anatomy, Medical College of Jinan University, Guangzhou, China
| | - Yaxi Zhu
- Department of Pathology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhi Chen
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Linju Ja
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Lisi OuYang
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yaofeng Zhu
- College of Medicine, Institute of Medical Sciences, Jishou University, Jishou, China
- Yaofeng Zhu, ,
| | - Si Chen
- Department of Human Anatomy, Histology and Embryology, Zunyi Medical University, Zhuhai, China
- Si Chen, ,
| | - Wanlong Lei
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Wanlong Lei, ,
| |
Collapse
|
47
|
Younger DS. Spinal cord motor disorders. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:3-42. [PMID: 37620076 DOI: 10.1016/b978-0-323-98817-9.00007-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
Spinal cord diseases are frequently devastating due to the precipitous and often permanently debilitating nature of the deficits. Spastic or flaccid paraparesis accompanied by dermatomal and myotomal signatures complementary to the incurred deficits facilitates localization of the insult within the cord. However, laboratory studies often employing disease-specific serology, neuroradiology, neurophysiology, and cerebrospinal fluid analysis aid in the etiologic diagnosis. While many spinal cord diseases are reversible and treatable, especially when recognized early, more than ever, neuroscientists are being called to investigate endogenous mechanisms of neural plasticity. This chapter is a review of the embryology, neuroanatomy, clinical localization, evaluation, and management of adult and childhood spinal cord motor disorders.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
48
|
Hirota R, Sasaki M, Kataoka-Sasaki Y, Oshigiri T, Kurihara K, Fukushi R, Oka S, Ukai R, Yoshimoto M, Kocsis JD, Yamashita T, Honmou O. Enhanced Network in Corticospinal Tracts after Infused Mesenchymal Stem Cells in Spinal Cord Injury. J Neurotrauma 2022; 39:1665-1677. [PMID: 35611987 PMCID: PMC9734021 DOI: 10.1089/neu.2022.0106] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Although limited spontaneous recovery occurs after spinal cord injury (SCI), current knowledge reveals that multiple forms of axon growth in spared axons can lead to circuit reorganization and a detour or relay pathways. This hypothesis has been derived mainly from studies of the corticospinal tract (CST), which is the primary descending motor pathway in mammals. The major CST is the dorsal CST (dCST), being the major projection from cortex to spinal cord. Two other components often called "minor" pathways are the ventral and the dorsal lateral CSTs, which may play an important role in spontaneous recovery. Intravenous infusion of mesenchymal stem cells (MSCs) provides functional improvement after SCI with an enhancement of axonal sprouting of CSTs. Detailed morphological changes of CST pathways, however, have not been fully elucidated. The primary objective was to evaluate detailed changes in descending CST projections in SCI after MSC infusion. The MSCs were infused intravenously one day after SCI. A combination of adeno-associated viral vector (AAV), which is an anterograde and non-transsynaptic axonal tracer, was injected 14 days after SCI induction. The AAV with advanced tissue clearing techniques were used to visualize the distribution pattern and high-resolution features of the individual axons coursing from above to below the lesion. The results demonstrated increased observable axonal connections between the dCST and axons in the lateral funiculus, both rostral and caudal to the lesion core, and an increase in observable axons in the dCST below the lesion. This increased axonal network could contribute to functional recovery by providing greater input to the spinal cord below the lesion.
Collapse
Affiliation(s)
- Ryosuke Hirota
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masanori Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Neurology, and Yale University School of Medicine, New Haven, Connecticut, USA.,Address correspondence to: Masanori Sasaki, MD, PhD, Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, S1W17, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan
| | - Yuko Kataoka-Sasaki
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsutomu Oshigiri
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kota Kurihara
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ryunosuke Fukushi
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shinichi Oka
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ryo Ukai
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsunori Yoshimoto
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Jeffery D. Kocsis
- Department of Neurology, and Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA.,Center for Neuroscience and Regeneration Research, VA Connecticut Healthcare System, West Haven, Connecticut, USA
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Osamu Honmou
- Department of Neural Regenerative Medicine, Research Institute for Frontier Medicine, and Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Neurology, and Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
49
|
Macías M, Lopez-Virgen V, Olivares-Moreno R, Rojas-Piloni G. Corticospinal neurons from motor and somatosensory cortices exhibit different temporal activity dynamics during motor learning. Front Hum Neurosci 2022; 16:1043501. [PMID: 36504625 PMCID: PMC9732016 DOI: 10.3389/fnhum.2022.1043501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022] Open
Abstract
The ability to learn motor skills implicates an improvement in accuracy, speed and consistency of movements. Motor control is related to movement execution and involves corticospinal neurons (CSp), which are broadly distributed in layer 5B of the motor and somatosensory cortices. CSp neurons innervate the spinal cord and are functionally diverse. However, whether CSp activity differs between different cortical areas throughout motor learning has been poorly explored. Given the importance and interaction between primary motor (M1) and somatosensory (S1) cortices related to movement, we examined the functional roles of CSp neurons in both areas. We induced the expression of GCaMP7s calcium indicator to perform photometric calcium recordings from layer 5B CSp neurons simultaneously in M1 and S1 cortices and track their activity while adult mice learned and performed a cued lever-press task. We found that during early learning sessions, the population calcium activity of CSp neurons in both cortices during movement did not change significantly. In late learning sessions the peak amplitude and duration of calcium activity CSp neurons increased in both, M1 and S1 cortices. However, S1 and M1 CSp neurons display a different temporal dynamic during movements that occurred when animals learned the task; both M1 and S1 CSp neurons activate before movement initiation, however, M1 CSp neurons continue active during movement performance, reinforcing the idea of the diversity of the CSp system and suggesting that CSp neuron activity in M1 and S1 cortices throughout motor learning have different functional roles for sensorimotor integration.
Collapse
|
50
|
Hausmann FS, Barrett JM, Martin ME, Zhan H, Shepherd GMG. Axonal Barcode Analysis of Pyramidal Tract Projections from Mouse Forelimb M1 and M2. J Neurosci 2022; 42:7733-7743. [PMID: 36414009 PMCID: PMC9581560 DOI: 10.1523/jneurosci.1062-22.2022] [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: 06/01/2022] [Revised: 08/22/2022] [Accepted: 08/25/2022] [Indexed: 12/14/2022] Open
Abstract
Forelimb-related areas of the motor cortex communicate directly to downstream areas in the brainstem and spinal cord via axons that project to and through the pyramidal tract (PT). To better understand the diversity of the brainstem branching patterns of these pyramidal tract projections, we used MAPseq, a molecular barcode technique for population-scale sampling with single-axon resolution. In experiments using mice of both sexes, we first confirmed prior results demonstrating the basic efficacy of axonal barcode identification of primary motor cortex (M1) PT-type axons, including corticobulbar (CBULB) and corticospinal (CSPI) subclasses. We then used multiplexed MAPseq to analyze projections from M1 and M2 (caudal and rostral forelimb areas). The four basic axon subclasses comprising these projections (M1-CSPI, M1-CBULB, M2-CSPI, M2-CBULB) showed a complex mix of differences and similarities in their brainstem projection profiles. This included relatively abundant branching by all classes in the dorsal midbrain, by M2 subclasses in the pons, and by CSPI subclasses in the dorsal medulla. Cluster analysis showed graded distributions of the basic subclasses within the PT class. Clusters were of diversely mixed subclass composition and showed distinct rostrocaudal and/or dorsomedial projection biases. Exemplifying these patterns was a subcluster likely enriched in corticocuneate branches. Overall, the results indicate high yet systematic PT axon diversity at the level of brainstem branching patterns; projections of M1 and M2 appear qualitatively similar, yet with quantitative differences in subclasses and clusters.SIGNIFICANCE STATEMENT Axons of the PT class of cortical projection neurons, which includes corticospinal and corticobulbar neurons, anatomically link motor cortex to brainstem and spinal cord circuits. Both of these subclasses can form branches to brainstem destinations along the way, but the extent and diversity of these branching patterns is incompletely understood. Here, we used MAPseq to tag PT axons with individual molecular barcodes for high-throughput quantification of branching patterns across the brainstem. The results reveal diverse, complex, yet systematic branching patterns of corticospinal and corticobulbar neurons arising from two motor cortex areas, M1 and M2.
Collapse
Affiliation(s)
- Frances S Hausmann
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - John M Barrett
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Megan E Martin
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Huiqing Zhan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, New York 11724
| | - Gordon M G Shepherd
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|