1
|
Orlovsky K, Appel E, Hantisteanu S, Olender T, Lotem J, Levanon D, Groner Y. Runx3, Brn3a and Isl1 interplay orchestrates the transcriptional program in the early stages of proprioceptive neuron development. PLoS Genet 2024; 20:e1011401. [PMID: 39715266 PMCID: PMC11729954 DOI: 10.1371/journal.pgen.1011401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 01/13/2025] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND The development and diversification of sensory proprioceptive neurons, which reside in the dorsal root ganglia (DRG) and express the tropomyosin receptor kinase C (TrkC), depend on the transcription factor (TF) Runx3. Runx3-deficient mice develop severe limb ataxia due to TrkC neuron cell death. Two additional TFs Pou4f1 (also called Brn3a) and Isl1 also play an important role in sensory neuron development. Thus, we aimed to unravel the chromatin state of early-developing TrkC neurons and decipher the Runx3 high-confidence target genes (HCT) and the possible cooperation between Runx3, Brn3a and Isl1 in the regulation of these genes. METHODS Runx3 expression is driven by the gene proximal P2 promoter. Transcriptome analysis was conducted by RNA-seq on RNA isolated from heterozygous (P2+/-) vs. homozygous (P2-/-) TrkC neurons and differentially expressed genes (DEGs) were determined. Genome-wide occupancy of Runx3, Brn3a, Isl1 and histone H3 acetylated on lysine 27 (H3K27Ac) was determined using CUT&RUN. The landscape of Transposase-accessible chromatin was analyzed via ATAC-seq. FINDINGS The intersection of Runx3 genomic occupancy-associated genes and DEG data discovered 244 Runx3 HCT. Brn3a and Isl1 were found to bind to numerous genomic loci, some of which overlapped with Runx3. Most genomic regions bound by each of these three TFs or co-bound by them resided in distantly located enhancer regions rather than in gene promoters. In activated and suppressed neuronal Runx3 HCT, Runx3 cooperated mainly with Brn3a to regulate expression through distantly located enhancers. Interestingly, suppression of non-neuronal immune genes was mainly managed via Runx3 without Brn3a. The distribution of ATAC and H3K27Ac marked regions in Runx3 peaks containing at least one RUNX binding site (Runx3_RBS) revealed that while most promoter regions were marked by ATAC, a prominent fraction of intron/intergenic regions occupied by Runx3, Brn3a or Isl1 were unmarked by ATAC and/or H3K27Ac. CONCLUSIONS These analyses shed new light on the interplay of Runx3, Brn3a, Isl1, and open chromatin regions in regulating the Runx3 HCT in the early developmental stages of TrkC neurons.
Collapse
Affiliation(s)
- Kira Orlovsky
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Elena Appel
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shay Hantisteanu
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Joseph Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ditsa Levanon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Yoram Groner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
2
|
Qi L, Iskols M, Shi D, Reddy P, Walker C, Lezgiyeva K, Voisin T, Pawlak M, Kuchroo VK, Chiu IM, Ginty DD, Sharma N. A mouse DRG genetic toolkit reveals morphological and physiological diversity of somatosensory neuron subtypes. Cell 2024; 187:1508-1526.e16. [PMID: 38442711 PMCID: PMC10947841 DOI: 10.1016/j.cell.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 11/12/2023] [Accepted: 02/05/2024] [Indexed: 03/07/2024]
Abstract
Dorsal root ganglia (DRG) somatosensory neurons detect mechanical, thermal, and chemical stimuli acting on the body. Achieving a holistic view of how different DRG neuron subtypes relay neural signals from the periphery to the CNS has been challenging with existing tools. Here, we develop and curate a mouse genetic toolkit that allows for interrogating the properties and functions of distinct cutaneous targeting DRG neuron subtypes. These tools have enabled a broad morphological analysis, which revealed distinct cutaneous axon arborization areas and branching patterns of the transcriptionally distinct DRG neuron subtypes. Moreover, in vivo physiological analysis revealed that each subtype has a distinct threshold and range of responses to mechanical and/or thermal stimuli. These findings support a model in which morphologically and physiologically distinct cutaneous DRG sensory neuron subtypes tile mechanical and thermal stimulus space to collectively encode a wide range of natural stimuli.
Collapse
Affiliation(s)
- Lijun Qi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - David Shi
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Pranav Reddy
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Christopher Walker
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Karina Lezgiyeva
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Tiphaine Voisin
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Mathias Pawlak
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Vijay K Kuchroo
- Gene Lay Institute of Immunology and Inflammation, Brigham and Women's Hospital, Mass General Hospital, and Harvard Medical School, Boston, MA 02115, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| | - Nikhil Sharma
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
3
|
Shi Y, Huang L, Dong H, Yang M, Ding W, Zhou X, Lu T, Liu Z, Zhou X, Wang M, Zeng B, Sun Y, Zhong S, Wang B, Wang W, Yin C, Wang X, Wu Q. Decoding the spatiotemporal regulation of transcription factors during human spinal cord development. Cell Res 2024; 34:193-213. [PMID: 38177242 PMCID: PMC10907391 DOI: 10.1038/s41422-023-00897-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/02/2023] [Indexed: 01/06/2024] Open
Abstract
The spinal cord is a crucial component of the central nervous system that facilitates sensory processing and motor performance. Despite its importance, the spatiotemporal codes underlying human spinal cord development have remained elusive. In this study, we have introduced an image-based single-cell transcription factor (TF) expression decoding spatial transcriptome method (TF-seqFISH) to investigate the spatial expression and regulation of TFs during human spinal cord development. By combining spatial transcriptomic data from TF-seqFISH and single-cell RNA-sequencing data, we uncovered the spatial distribution of neural progenitor cells characterized by combinatorial TFs along the dorsoventral axis, as well as the molecular and spatial features governing neuronal generation, migration, and differentiation along the mediolateral axis. Notably, we observed a sandwich-like organization of excitatory and inhibitory interneurons transiently appearing in the dorsal horns of the developing human spinal cord. In addition, we integrated data from 10× Visium to identify early and late waves of neurogenesis in the dorsal horn, revealing the formation of laminas in the dorsal horns. Our study also illuminated the spatial differences and molecular cues underlying motor neuron (MN) diversification, and the enrichment of Amyotrophic Lateral Sclerosis (ALS) risk genes in MNs and microglia. Interestingly, we detected disease-associated microglia (DAM)-like microglia groups in the developing human spinal cord, which are predicted to be vulnerable to ALS and engaged in the TYROBP causal network and response to unfolded proteins. These findings provide spatiotemporal transcriptomic resources on the developing human spinal cord and potential strategies for spinal cord injury repair and ALS treatment.
Collapse
Affiliation(s)
- Yingchao Shi
- Guangdong Institute of Intelligence Science and Technology, Guangdong, China.
| | - Luwei Huang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Dong
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Meng Yang
- Changping Laboratory, Beijing, China
| | - Wenyu Ding
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China
| | - Xiang Zhou
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tian Lu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Xin Zhou
- Changping Laboratory, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China
| | - Mengdi Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Bo Zeng
- Changping Laboratory, Beijing, China
| | - Yinuo Sun
- Changping Laboratory, Beijing, China
| | - Suijuan Zhong
- Changping Laboratory, Beijing, China
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China
| | - Bosong Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China
| | - Wei Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | | | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- Changping Laboratory, Beijing, China.
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China.
| | - Qian Wu
- Changping Laboratory, Beijing, China.
- State Key Laboratory of Cognitive Neuroscience and Learning, IDG/McGovern Institute for Brain Research, New Cornerstone Science Laboratory, Beijing Normal University, Beijing, China.
| |
Collapse
|
4
|
Bornstein B, Watkins B, Passini FS, Blecher R, Assaraf E, Sui XM, Brumfeld V, Tsoory M, Kröger S, Zelzer E. The mechanosensitive ion channel ASIC2 mediates both proprioceptive sensing and spinal alignment. Exp Physiol 2024; 109:135-147. [PMID: 36951012 PMCID: PMC10988735 DOI: 10.1113/ep090776] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/20/2023] [Indexed: 03/24/2023]
Abstract
By translating mechanical forces into molecular signals, proprioceptive neurons provide the CNS with information on muscle length and tension, which is necessary to control posture and movement. However, the identities of the molecular players that mediate proprioceptive sensing are largely unknown. Here, we confirm the expression of the mechanosensitive ion channel ASIC2 in proprioceptive sensory neurons. By combining in vivo proprioception-related functional tests with ex vivo electrophysiological analyses of muscle spindles, we showed that mice lacking Asic2 display impairments in muscle spindle responses to stretch and motor coordination tasks. Finally, analysis of skeletons of Asic2 loss-of-function mice revealed a specific effect on spinal alignment. Overall, we identify ASIC2 as a key component in proprioceptive sensing and a regulator of spine alignment.
Collapse
Affiliation(s)
- Bavat Bornstein
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - Bridgette Watkins
- Department of Physiological Genomics, Biomedical CenterLudwig‐Maximilians‐UniversityPlanegg‐MartinsriedGermany
| | - Fabian S. Passini
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| | - Ronen Blecher
- Orthopedic DepartmentAssuta Ashdod University Hospital, Ashdod, Israel, affiliated to Ben Gurion University of the NegevBeer ShebaIsrael
| | - Eran Assaraf
- Department of Orthopedic SurgeryShamir Medical Center, Assaf HaRofeh Campus, Zeffifin, Israel, affiliated to Sackler Faculty of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Xiao Meng Sui
- Department of Chemical Research SupportWeizmann Institute of ScienceRehovotIsrael
| | - Vlad Brumfeld
- Department of Chemical Research SupportWeizmann Institute of ScienceRehovotIsrael
| | - Michael Tsoory
- Department of Veterinary ResourcesWeizmann Institute of ScienceRehovotIsrael
| | - Stephan Kröger
- Department of Physiological Genomics, Biomedical CenterLudwig‐Maximilians‐UniversityPlanegg‐MartinsriedGermany
| | - Elazar Zelzer
- Department of Molecular GeneticsWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
5
|
Di Russo A, Stanev D, Sabnis A, Danner SM, Ausborn J, Armand S, Ijspeert A. Investigating the roles of reflexes and central pattern generators in the control and modulation of human locomotion using a physiologically plausible neuromechanical model. J Neural Eng 2023; 20:066006. [PMID: 37757805 DOI: 10.1088/1741-2552/acfdcc] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 09/27/2023] [Indexed: 09/29/2023]
Abstract
Objective.Studying the neural components regulating movement in human locomotion is obstructed by the inability to perform invasive experimental recording in the human neural circuits. Neuromechanical simulations can provide insights by modeling the locomotor circuits. Past neuromechanical models proposed control of locomotion either driven by central pattern generators (CPGs) with simple sensory commands or by a purely reflex-based network regulated by state-machine mechanisms, which activate and deactivate reflexes depending on the detected gait cycle phases. However, the physiological interpretation of these state machines remains unclear. Here, we present a physiologically plausible model to investigate spinal control and modulation of human locomotion.Approach.We propose a bio-inspired controller composed of two coupled CPGs that produce the rhythm and pattern, and a reflex-based network simulating low-level reflex pathways and Renshaw cells. This reflex network is based on leaky-integration neurons, and the whole system does not rely on changing reflex gains according to the gait cycle state. The musculoskeletal model is composed of a skeletal structure and nine muscles per leg generating movement in sagittal plane.Main results.Optimizing the open parameters for effort minimization and stability, human kinematics and muscle activation naturally emerged. Furthermore, when CPGs were not activated, periodic motion could not be achieved through optimization, suggesting the necessity of this component to generate rhythmic behavior without a state machine mechanism regulating reflex activation. The controller could reproduce ranges of speeds from 0.3 to 1.9 m s-1. The results showed that the net influence of feedback on motoneurons (MNs) during perturbed locomotion is predominantly inhibitory and that the CPGs provide the timing of MNs' activation by exciting or inhibiting muscles in specific gait phases.Significance.The proposed bio-inspired controller could contribute to our understanding of locomotor circuits of the intact spinal cord and could be used to study neuromotor disorders.
Collapse
Affiliation(s)
| | | | | | - Simon M Danner
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States of America
| | - Jessica Ausborn
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States of America
| | - Stéphane Armand
- Kinesiology Laboratory, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | | |
Collapse
|
6
|
Qi L, Iskols M, Shi D, Reddy P, Walker C, Lezgiyeva K, Voisin T, Pawlak M, Kuchroo VK, Chiu I, Ginty DD, Sharma N. A DRG genetic toolkit reveals molecular, morphological, and functional diversity of somatosensory neuron subtypes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.22.537932. [PMID: 37131664 PMCID: PMC10153270 DOI: 10.1101/2023.04.22.537932] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Mechanical and thermal stimuli acting on the skin are detected by morphologically and physiologically distinct sensory neurons of the dorsal root ganglia (DRG). Achieving a holistic view of how this diverse neuronal population relays sensory information from the skin to the central nervous system (CNS) has been challenging with existing tools. Here, we used transcriptomic datasets of the mouse DRG to guide development and curation of a genetic toolkit to interrogate transcriptionally defined DRG neuron subtypes. Morphological analysis revealed unique cutaneous axon arborization areas and branching patterns of each subtype. Physiological analysis showed that subtypes exhibit distinct thresholds and ranges of responses to mechanical and/or thermal stimuli. The somatosensory neuron toolbox thus enables comprehensive phenotyping of most principal sensory neuron subtypes. Moreover, our findings support a population coding scheme in which the activation thresholds of morphologically and physiologically distinct cutaneous DRG neuron subtypes tile multiple dimensions of stimulus space.
Collapse
Affiliation(s)
- Lijun Qi
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Michael Iskols
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - David Shi
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| | - Pranav Reddy
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| | - Christopher Walker
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| | - Karina Lezgiyeva
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Tiphaine Voisin
- Department of Immunology, Harvard Medical School, Boston, MA 02115
| | - Mathias Pawlak
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Isaac Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02115
| | - David D. Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
| | - Nikhil Sharma
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115
- Department of Molecular Pharmacology and Therapeutics, Department of Systems Biology, Columbia University, New York, NY
| |
Collapse
|
7
|
Cao R, Chen P, Wang H, Jing H, Zhang H, Xing G, Luo B, Pan J, Yu Z, Xiong WC, Mei L. Intrafusal-fiber LRP4 for muscle spindle formation and maintenance in adult and aged animals. Nat Commun 2023; 14:744. [PMID: 36765071 PMCID: PMC9918736 DOI: 10.1038/s41467-023-36454-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 01/30/2023] [Indexed: 02/12/2023] Open
Abstract
Proprioception is sensed by muscle spindles for precise locomotion and body posture. Unlike the neuromuscular junction (NMJ) for muscle contraction which has been well studied, mechanisms of spindle formation are not well understood. Here we show that sensory nerve terminals are disrupted by the mutation of Lrp4, a gene required for NMJ formation; inducible knockout of Lrp4 in adult mice impairs sensory synapses and movement coordination, suggesting that LRP4 is required for spindle formation and maintenance. LRP4 is critical to the expression of Egr3 during development; in adult mice, it interacts in trans with APP and APLP2 on sensory terminals. Finally, spindle sensory endings and function are impaired in aged mice, deficits that could be diminished by LRP4 expression. These observations uncovered LRP4 as an unexpected regulator of muscle spindle formation and maintenance in adult and aged animals and shed light on potential pathological mechanisms of abnormal muscle proprioception.
Collapse
Affiliation(s)
- Rangjuan Cao
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.,Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Peng Chen
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongsheng Wang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongyang Jing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Hongsheng Zhang
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Guanglin Xing
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Bin Luo
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jinxiu Pan
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Zheng Yu
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Wen-Cheng Xiong
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| | - Lin Mei
- Department of Neurosciences, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA. .,Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, OH, 44106, USA.
| |
Collapse
|
8
|
Ladle DR, Hippenmeyer S. Loss of ETV1/ER81 in motor neurons leads to reduced monosynaptic inputs from proprioceptive sensory neurons. J Neurophysiol 2023; 129:501-512. [PMID: 36695533 DOI: 10.1152/jn.00172.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
Presynaptic inputs determine the pattern of activation of postsynaptic neurons in a neural circuit. Molecular and genetic pathways that regulate the selective formation of subsets of presynaptic inputs are largely unknown, despite significant understanding of the general process of synaptogenesis. In this study, we have begun to identify such factors using the spinal monosynaptic stretch reflex circuit as a model system. In this neuronal circuit, Ia proprioceptive afferents establish monosynaptic connections with spinal motor neurons that project to the same muscle (termed homonymous connections) or muscles with related or synergistic function. However, monosynaptic connections are not formed with motor neurons innervating muscles with antagonistic functions. The ETS transcription factor ER81 (also known as ETV1) is expressed by all proprioceptive afferents, but only a small set of motor neuron pools in the lumbar spinal cord of the mouse. Here we use conditional mouse genetic techniques to eliminate Er81 expression selectively from motor neurons. We find that ablation of Er81 in motor neurons reduces synaptic inputs from proprioceptive afferents conveying information from homonymous and synergistic muscles, with no change observed in the connectivity pattern from antagonistic proprioceptive afferents. In summary, these findings suggest a role for ER81 in defined motor neuron pools to control the assembly of specific presynaptic inputs and thereby influence the profile of activation of these motor neurons.
Collapse
Affiliation(s)
- David R Ladle
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, United States
| | - Simon Hippenmeyer
- Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| |
Collapse
|
9
|
Ma S, Dubin AE, Romero LO, Loud M, Salazar A, Chu S, Klier N, Masri S, Zhang Y, Wang Y, Chesler AT, Wilkinson KA, Vásquez V, Marshall KL, Patapoutian A. Excessive mechanotransduction in sensory neurons causes joint contractures. Science 2023; 379:201-206. [PMID: 36634173 PMCID: PMC10163824 DOI: 10.1126/science.add3598] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 11/04/2022] [Indexed: 01/13/2023]
Abstract
Distal arthrogryposis (DA) is a collection of rare disorders that are characterized by congenital joint contractures. Most DA mutations are in muscle- and joint-related genes, and the anatomical defects originate cell-autonomously within the musculoskeletal system. However, gain-of-function mutations in PIEZO2, a principal mechanosensor in somatosensation, cause DA subtype 5 (DA5) through unknown mechanisms. We show that expression of a gain-of-function PIEZO2 mutation in proprioceptive sensory neurons that mainly innervate muscle spindles and tendons is sufficient to induce DA5-like phenotypes in mice. Overactive PIEZO2 causes anatomical defects through increased activity within the peripheral nervous system during postnatal development. Furthermore, botulinum toxin (Botox) and a dietary fatty acid that modulates PIEZO2 activity reduce DA5-like deficits. This reveals a role for somatosensory neurons: Excessive mechanosensation within these neurons disrupts musculoskeletal development.
Collapse
Affiliation(s)
- Shang Ma
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Adrienne E. Dubin
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Luis O. Romero
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meaghan Loud
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Alexandra Salazar
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Sarah Chu
- Department of Biological Sciences, San Jose State University, San Jose, CA, USA
| | - Nikola Klier
- Department of Biological Sciences, San Jose State University, San Jose, CA, USA
| | - Sameer Masri
- Department of Biological Sciences, San Jose State University, San Jose, CA, USA
| | - Yunxiao Zhang
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Yu Wang
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| | - Alex T. Chesler
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
- National Center for Complementary and Integrative Health, National Institutes of Health, Bethesda, MD, USA
| | | | - Valeria Vásquez
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Kara L. Marshall
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Ardem Patapoutian
- Howard Hughes Medical Institute, Department of Neuroscience, Dorris Neuroscience Center, Scripps Research, La Jolla, CA 92037, USA
| |
Collapse
|
10
|
Enander JMD, Jones AM, Kirkland M, Hurless J, Jörntell H, Loeb GE. A model for self-organization of sensorimotor function: the spinal monosynaptic loop. J Neurophysiol 2022; 127:1460-1477. [PMID: 35264006 PMCID: PMC9208450 DOI: 10.1152/jn.00242.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 02/11/2022] [Accepted: 03/02/2022] [Indexed: 01/05/2023] Open
Abstract
Recent spinal cord literature abounds with descriptions of genetic preprogramming and the molecular control of circuit formation. In this paper, we explore to what extent circuit formation based on learning rather than preprogramming could explain the selective formation of the monosynaptic projections between muscle spindle primary afferents and homonymous motoneurons. We adjusted the initially randomized gains in the neural network according to a Hebbian plasticity rule while exercising the model system with spontaneous muscle activity patterns similar to those observed during early fetal development. Normal connectivity patterns developed only when we modeled β motoneurons, which are known to innervate both intrafusal and extrafusal muscle fibers in vertebrate muscles but were not considered in previous literature regarding selective formation of these synapses in animals with paralyzed muscles. It was also helpful to correctly model the greatly reduced contractility of extrafusal muscle fibers during early development. Stronger and more coordinated muscle activity patterns such as observed later during neonatal locomotion impaired projection selectivity. These findings imply a generic functionality of a musculoskeletal system to imprint important aspects of its mechanical dynamics onto a neural network, without specific preprogramming other than setting a critical period for the formation and maturation of this general pattern of connectivity. Such functionality would facilitate the successful evolution of new species with altered musculoskeletal anatomy, and it may help to explain patterns of connectivity and associated reflexes that appear during abnormal development.NEW & NOTEWORTHY A novel model of self-organization of early spinal circuitry based on a biologically realistic plant, sensors, and neuronal plasticity in conjunction with empirical observations of fetal development. Without explicit need for guiding genetic rules, connection matrices emerge that support functional self-organization of the mature pattern of Ia to motoneuron connectivity in the spinal circuitry.
Collapse
Affiliation(s)
- Jonas M D Enander
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Adam M Jones
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California
| | - Matthieu Kirkland
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California
| | - Jordan Hurless
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California
| | - Henrik Jörntell
- Department of Experimental Medical Science, Faculty of Medicine, Lund University, Lund, Sweden
| | - Gerald E Loeb
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California
| |
Collapse
|
11
|
Chalif JI, Mentis GZ. Normal Development and Pathology of Motoneurons: Anatomy, Electrophysiological Properties, Firing Patterns and Circuit Connectivity. ADVANCES IN NEUROBIOLOGY 2022; 28:63-85. [PMID: 36066821 DOI: 10.1007/978-3-031-07167-6_3] [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: 06/15/2023]
Abstract
This chapter will provide an introduction into motoneuron anatomy, electrophysiological properties, firing patterns focusing on development and also describing several pathological conditions that affect mononeurons. It starts with a historical retrospective describing the early landmark work into motoneurons. The next section lays out the various types of motoneurons (alpha, beta, and gamma) and their subclasses (fast-twitch fatigable, fast-twitch fatigue-resistant, and slow-twitch fatigue resistant), highlighting the functional relevance of this classification scheme. The third section describes the development of motoneurons' passive and active electrophysiological properties. This section also defines the major terms one uses in describing how a neuron functions electrophysiologically. The electrophysiological aspects of a neuron is critical to understanding how it behaves within a circuit and contributes to behavior since the firing of an action potential is how neurons communicate with each other and with muscles. The electrophysiological changes of motoneurons over development underlies how their function changes over the lifetime of an organism. After describing the properties of individual motoneurons, the chapter then turns to revealing how motoneurons interact within complex neural circuits, with other motoneurons as well as sensory neurons, and how these circuits change over development. Finally, this chapter ends with highlighting some recent advances made in motoneuron pathology, focusing on spinal muscular atrophy, amyotrophic lateral sclerosis, and axotomy.
Collapse
Affiliation(s)
- Joshua I Chalif
- Departments of Neurology and Pathology & Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - George Z Mentis
- Departments of Neurology and Pathology & Cell Biology, Center for Motor Neuron Biology and Disease, Columbia University, New York, NY, USA.
| |
Collapse
|
12
|
Dasen JS. Establishing the Molecular and Functional Diversity of Spinal Motoneurons. ADVANCES IN NEUROBIOLOGY 2022; 28:3-44. [PMID: 36066819 DOI: 10.1007/978-3-031-07167-6_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Spinal motoneurons are a remarkably diverse class of neurons responsible for facilitating a broad range of motor behaviors and autonomic functions. Studies of motoneuron differentiation have provided fundamental insights into the developmental mechanisms of neuronal diversification, and have illuminated principles of neural fate specification that operate throughout the central nervous system. Because of their relative anatomical simplicity and accessibility, motoneurons have provided a tractable model system to address multiple facets of neural development, including early patterning, neuronal migration, axon guidance, and synaptic specificity. Beyond their roles in providing direct communication between central circuits and muscle, recent studies have revealed that motoneuron subtype-specific programs also play important roles in determining the central connectivity and function of motor circuits. Cross-species comparative analyses have provided novel insights into how evolutionary changes in subtype specification programs may have contributed to adaptive changes in locomotor behaviors. This chapter focusses on the gene regulatory networks governing spinal motoneuron specification, and how studies of spinal motoneurons have informed our understanding of the basic mechanisms of neuronal specification and spinal circuit assembly.
Collapse
Affiliation(s)
- Jeremy S Dasen
- NYU Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY, USA.
| |
Collapse
|
13
|
Abstract
When animals walk overground, mechanical stimuli activate various receptors located in muscles, joints, and skin. Afferents from these mechanoreceptors project to neuronal networks controlling locomotion in the spinal cord and brain. The dynamic interactions between the control systems at different levels of the neuraxis ensure that locomotion adjusts to its environment and meets task demands. In this article, we describe and discuss the essential contribution of somatosensory feedback to locomotion. We start with a discussion of how biomechanical properties of the body affect somatosensory feedback. We follow with the different types of mechanoreceptors and somatosensory afferents and their activity during locomotion. We then describe central projections to locomotor networks and the modulation of somatosensory feedback during locomotion and its mechanisms. We then discuss experimental approaches and animal models used to investigate the control of locomotion by somatosensory feedback before providing an overview of the different functional roles of somatosensory feedback for locomotion. Lastly, we briefly describe the role of somatosensory feedback in the recovery of locomotion after neurological injury. We highlight the fact that somatosensory feedback is an essential component of a highly integrated system for locomotor control. © 2021 American Physiological Society. Compr Physiol 11:1-71, 2021.
Collapse
Affiliation(s)
- Alain Frigon
- Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Quebec, Canada
| | - Turgay Akay
- Department of Medical Neuroscience, Atlantic Mobility Action Project, Brain Repair Center, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Boris I Prilutsky
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Gras S, Blasco A, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Rueda R, Pereira SL, Navarro X, Esquerda JE, Calderó J. Beneficial effects of dietary supplementation with green tea catechins and cocoa flavanols on aging-related regressive changes in the mouse neuromuscular system. Aging (Albany NY) 2021; 13:18051-18093. [PMID: 34319911 PMCID: PMC8351677 DOI: 10.18632/aging.203336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/19/2021] [Indexed: 12/17/2022]
Abstract
Besides skeletal muscle wasting, sarcopenia entails morphological and molecular changes in distinct components of the neuromuscular system, including spinal cord motoneurons (MNs) and neuromuscular junctions (NMJs); moreover, noticeable microgliosis has also been observed around aged MNs. Here we examined the impact of two flavonoid-enriched diets containing either green tea extract (GTE) catechins or cocoa flavanols on age-associated regressive changes in the neuromuscular system of C57BL/6J mice. Compared to control mice, GTE- and cocoa-supplementation significantly improved the survival rate of mice, reduced the proportion of fibers with lipofuscin aggregates and central nuclei, and increased the density of satellite cells in skeletal muscles. Additionally, both supplements significantly augmented the number of innervated NMJs and their degree of maturity compared to controls. GTE, but not cocoa, prominently increased the density of VAChT and VGluT2 afferent synapses on MNs, which were lost in control aged spinal cords; conversely, cocoa, but not GTE, significantly augmented the proportion of VGluT1 afferent synapses on aged MNs. Moreover, GTE, but not cocoa, reduced aging-associated microgliosis and increased the proportion of neuroprotective microglial phenotypes. Our data indicate that certain plant flavonoids may be beneficial in the nutritional management of age-related deterioration of the neuromuscular system.
Collapse
Affiliation(s)
- Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Alejandro Barranco
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, Granada, Spain
| | - Tapas Das
- Abbott Nutrition, Research and Development, Columbus, OH 43215, USA
| | - Ricardo Rueda
- Abbott Nutrition, Research and Development, Granada, Spain
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cellular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona and CIBERNED, Bellaterra, Spain
| | - Josep E. Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida and Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
15
|
Oliveira MP, Menzel HJK, Cochrane DJ, Drummond MDM, Demicheli C, Lage G, Couto BP. Individual Responses to Different Vibration Frequencies Identified by Electromyography and Dynamometry in Different Types of Vibration Application. J Strength Cond Res 2021; 35:1748-1759. [PMID: 30844986 DOI: 10.1519/jsc.0000000000002985] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
ABSTRACT Oliveira, MP, Menzel, H-JK, Cochrane, DJ, Drummond, MD, Demicheli, C, Lage, G, and Couto, BP. Individual responses to different vibration frequencies identified by electromyography and dynamometry in different types of vibration application. J Strength Cond Res 35(6): 1748-1759, 2021-The application of mechanical vibration is a common neuromuscular training technique used in sports training programs to generate acute increases in muscle strength. The principal aim of the study was to compare the individual optimal vibration frequency (IOVF) identified by electromyography (EMG) activity and force production in strength training. Twenty well-trained male volunteers (age: 23.8 ± 3.3 years) performed a familiarization and 2 interventions sessions, which included 5 maximal voluntary contractions (MVCs) of the elbow flexors with a duration of 10 seconds and 5-minute intervals between each MVC. The first MVC was performed without vibration followed by 4 randomized MVCs with application of vibration in the direction of the resultant muscle forces' vector (VDF) or whole-body vibration (WBV) at frequencies of 10, 20, 30, or 40 Hz. The mechanical vibration stimulus was superimposed during the MVC. Individual optimal vibration frequency, as identified by EMG, did not coincide with IOVF identified by force production; low agreement was observed between the vibration frequencies in generating the higher EMG activity, maximal force, and root mean square of force. These findings suggest that the magnitude of the vibratory stimulus response is individualized. Therefore, if the aim is to use acute vibration in conjunction with strength training, a preliminary vibration exposure should be conducted to determine the individualized vibratory stimulus of the subject, so that training effects can be optimized.
Collapse
Affiliation(s)
- Mariana P Oliveira
- Laboratory of Load Evaluation, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Hans-Joachim K Menzel
- Laboratory of Biomechanics, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Marcos D M Drummond
- Laboratory of Load Evaluation, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carlo Demicheli
- School of Mathematics, Federal University of Minas Gerais, Belo Horizonte, Brazil; and
| | - Guilherme Lage
- Laboratory of Movements Analyses, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bruno P Couto
- Laboratory of Load Evaluation, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
16
|
Bornstein B, Konstantin N, Alessandro C, Tresch MC, Zelzer E. More than movement: the proprioceptive system as a new regulator of musculoskeletal biology. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2021.01.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
17
|
Lopez AJ, Xu J, Hoque MM, McMullen C, Kesar TM, Borich MR. Integration of Convergent Sensorimotor Inputs Within Spinal Reflex Circuits in Healthy Adults. Front Hum Neurosci 2020; 14:592013. [PMID: 33324184 PMCID: PMC7725688 DOI: 10.3389/fnhum.2020.592013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/03/2020] [Indexed: 11/25/2022] Open
Abstract
The output from motor neuron pools is influenced by the integration of synaptic inputs originating from descending corticomotor and spinal reflex pathways. In this study, using paired non-invasive brain and peripheral nerve stimulation, we investigated how descending corticomotor pathways influence the physiologic recruitment order of the soleus Hoffmann (H-) reflex. Eleven neurologically unimpaired adults (9 females; mean age 25 ± 3 years) completed an assessment of transcranial magnetic stimulation (TMS)-conditioning of the soleus H-reflex over a range of peripheral nerve stimulation (PNS) intensities. Unconditioned H-reflex recruitment curves were obtained by delivering PNS pulses to the posterior tibial nerve. Subsequently, TMS-conditioned H-reflex recruitment curves were obtained by pairing PNS with subthreshold TMS at short (−1.5 ms) and long (+10 ms) intervals. We evaluated unconditioned and TMS-conditioned H-reflex amplitudes along the ascending limb, peak, and descending limb of the H-reflex recruitment curve. Our results revealed that, for long-interval facilitation, TMS-conditioned H-reflex amplitudes were significantly larger than unconditioned H-reflex amplitudes along the ascending limb and peak of the H-reflex recruitment curve. Additionally, significantly lower PNS intensities were needed to elicit peak H-reflex amplitude (Hmax) for long-interval facilitation compared to unconditioned. These findings suggest that the influence of descending corticomotor pathways, particularly those mediating long-interval facilitation, contribute to changing the recruitment gain of the motor neuron pool, and can inform future methodological protocols for TMS-conditioning of H-reflexes. By characterizing and inducing short-term plasticity in circuitry mediating short- and long-interval TMS-conditioning of H-reflex amplitudes, future studies can investigate supraspinal and spinal circuit contributions to abnormal motor control, as well as develop novel therapeutic targets for neuromodulation.
Collapse
Affiliation(s)
- Alejandro J Lopez
- Neural Plasticity Research Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States.,Motion Analysis Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States
| | - Jiang Xu
- Neural Plasticity Research Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States.,Motion Analysis Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States.,Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Maruf M Hoque
- Neural Plasticity Research Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States.,Motion Analysis Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States
| | - Carly McMullen
- Neural Plasticity Research Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States.,Motion Analysis Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States
| | - Trisha M Kesar
- Motion Analysis Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States
| | - Michael R Borich
- Neural Plasticity Research Laboratory, Division of Physical Therapy, Department of Rehabilitation Medicine, Emory University, Atlanta, GA, United States
| |
Collapse
|
18
|
Thanawalla AR, Chen AI, Azim E. The Cerebellar Nuclei and Dexterous Limb Movements. Neuroscience 2020; 450:168-183. [PMID: 32652173 PMCID: PMC7688491 DOI: 10.1016/j.neuroscience.2020.06.046] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/03/2020] [Accepted: 06/30/2020] [Indexed: 01/21/2023]
Abstract
Dexterous forelimb movements like reaching, grasping, and manipulating objects are fundamental building blocks of the mammalian motor repertoire. These behaviors are essential to everyday activities, and their elaboration underlies incredible accomplishments by human beings in art and sport. Moreover, the susceptibility of these behaviors to damage and disease of the nervous system can lead to debilitating deficits, highlighting a need for a better understanding of function and dysfunction in sensorimotor control. The cerebellum is central to coordinating limb movements, as defined in large part by Joseph Babinski and Gordon Holmes describing motor impairment in patients with cerebellar lesions over 100 years ago (Babinski, 1902; Holmes, 1917), and supported by many important human and animal studies that have been conducted since. Here, with a focus on output pathways of the cerebellar nuclei across mammalian species, we describe forelimb movement deficits observed when cerebellar circuits are perturbed, the mechanisms through which these circuits influence motor output, and key challenges in defining how the cerebellum refines limb movement.
Collapse
Affiliation(s)
- Ayesha R Thanawalla
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Albert I Chen
- Nanyang Technological University (NTU), School of Biological Sciences, 11 Mandalay Road, Singapore 308232, Singapore; A*STAR, Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 308232, Singapore.
| | - Eiman Azim
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
19
|
Blasco A, Gras S, Mòdol-Caballero G, Tarabal O, Casanovas A, Piedrafita L, Barranco A, Das T, Pereira SL, Navarro X, Rueda R, Esquerda JE, Calderó J. Motoneuron deafferentation and gliosis occur in association with neuromuscular regressive changes during ageing in mice. J Cachexia Sarcopenia Muscle 2020; 11:1628-1660. [PMID: 32691534 PMCID: PMC7749545 DOI: 10.1002/jcsm.12599] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The cellular mechanisms underlying the age-associated loss of muscle mass and function (sarcopenia) are poorly understood, hampering the development of effective treatment strategies. Here, we performed a detailed characterization of age-related pathophysiological changes in the mouse neuromuscular system. METHODS Young, adult, middle-aged, and old (1, 4, 14, and 24-30 months old, respectively) C57BL/6J mice were used. Motor behavioural and electrophysiological tests and histological and immunocytochemical procedures were carried out to simultaneously analyse structural, molecular, and functional age-related changes in distinct cellular components of the neuromuscular system. RESULTS Ageing was not accompanied by a significant loss of spinal motoneurons (MNs), although a proportion (~15%) of them in old mice exhibited an abnormally dark appearance. Dark MNs were also observed in adult (~9%) and young (~4%) animals, suggesting that during ageing, some MNs undergo early deleterious changes, which may not lead to MN death. Old MNs were depleted of cholinergic and glutamatergic inputs (~40% and ~45%, respectively, P < 0.01), suggestive of age-associated alterations in MN excitability. Prominent microgliosis and astrogliosis [~93% (P < 0.001) and ~100% (P < 0.0001) increase vs. adults, respectively] were found in old spinal cords, with increased density of pro-inflammatory M1 microglia and A1 astroglia (25-fold and 4-fold increase, respectively, P < 0.0001). Ageing resulted in significant reductions in the nerve conduction velocity and the compound muscle action potential amplitude (~30%, P < 0.05, vs. adults) in old distal plantar muscles. Compared with adult muscles, old muscles exhibited significantly higher numbers of both denervated and polyinnervated neuromuscular junctions, changes in fibre type composition, higher proportion of fibres showing central nuclei and lipofuscin aggregates, depletion of satellite cells, and augmented expression of different molecules related to development, plasticity, and maintenance of neuromuscular junctions, including calcitonin gene-related peptide, growth associated protein 43, agrin, fibroblast growth factor binding protein 1, and transforming growth factor-β1. Overall, these alterations occurred at varying degrees in all the muscles analysed, with no correlation between the age-related changes observed and myofiber type composition or muscle topography. CONCLUSIONS Our data provide a global view of age-associated neuromuscular changes in a mouse model of ageing and help to advance understanding of contributing pathways leading to development of sarcopenia.
Collapse
Affiliation(s)
- Alba Blasco
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Sílvia Gras
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Guillem Mòdol-Caballero
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Olga Tarabal
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Anna Casanovas
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lídia Piedrafita
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | | | - Tapas Das
- Abbott Nutrition Research and Development, Columbus, OH, USA
| | | | - Xavier Navarro
- Grup de Neuroplasticitat i Regeneració, Institut de Neurociències, Departament de Biologia Cel·lular, Fisiologia i Immunologia, Universitat Autònoma de Barcelona, CIBERNED, Bellaterra, Spain
| | - Ricardo Rueda
- Abbott Nutrition Research and Development, Granada, Spain
| | - Josep E Esquerda
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Jordi Calderó
- Unitat de Neurobiologia Cel·lular, Departament de Medicina Experimental, Facultat de Medicina, Universitat de Lleida, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| |
Collapse
|
20
|
Vukojicic A, Delestrée N, Fletcher EV, Pagiazitis JG, Sankaranarayanan S, Yednock TA, Barres BA, Mentis GZ. The Classical Complement Pathway Mediates Microglia-Dependent Remodeling of Spinal Motor Circuits during Development and in SMA. Cell Rep 2020; 29:3087-3100.e7. [PMID: 31801075 PMCID: PMC6937140 DOI: 10.1016/j.celrep.2019.11.013] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 09/20/2019] [Accepted: 11/04/2019] [Indexed: 12/19/2022] Open
Abstract
Movement is an essential behavior requiring the assembly and refinement of spinal motor circuits. However, the mechanisms responsible for circuit refinement and synapse maintenance are poorly understood. Similarly, the molecular mechanisms by which gene mutations cause dysfunction and elimination of synapses in neurodegenerative diseases that occur during development are unknown. Here, we demonstrate that the complement protein C1q is required for the refinement of sensory-motor circuits during normal development, as well as for synaptic dysfunction and elimination in spinal muscular atrophy (SMA). C1q tags vulnerable SMA synapses, which triggers activation of the classical complement pathway leading to microglia-mediated elimination. Pharmacological inhibition of C1q or depletion of microglia rescues the number and function of synapses, conferring significant behavioral benefit in SMA mice. Thus, the classical complement pathway plays critical roles in the refinement of developing motor circuits, while its aberrant activation contributes to motor neuron disease.
Collapse
Affiliation(s)
- Aleksandra Vukojicic
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Nicolas Delestrée
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Emily V Fletcher
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - John G Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | | | - Ted A Yednock
- Annexon Biosciences, 180 Kimball Way, South San Francisco, CA 94080, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University, Palo Alto, CA, USA
| | - George Z Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, NY 10032, USA; Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA; Department of Neurology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
21
|
Shin MM, Catela C, Dasen J. Intrinsic control of neuronal diversity and synaptic specificity in a proprioceptive circuit. eLife 2020; 9:56374. [PMID: 32808924 PMCID: PMC7467731 DOI: 10.7554/elife.56374] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
Relay of muscle-derived sensory information to the CNS is essential for the execution of motor behavior, but how proprioceptive sensory neurons (pSNs) establish functionally appropriate connections is poorly understood. A prevailing model of sensory-motor circuit assembly is that peripheral, target-derived, cues instruct pSN identities and patterns of intraspinal connectivity. To date no known intrinsic determinants of muscle-specific pSN fates have been described in vertebrates. We show that expression of Hox transcription factors defines pSN subtypes, and these profiles are established independently of limb muscle. The Hoxc8 gene is expressed by pSNs and motor neurons (MNs) targeting distal forelimb muscles, and sensory-specific depletion of Hoxc8 in mice disrupts sensory-motor synaptic matching, without affecting pSN survival or muscle targeting. These results indicate that the diversity and central specificity of pSNs and MNs are regulated by a common set of determinants, thus linking early rostrocaudal patterning to the assembly of limb control circuits.
Collapse
Affiliation(s)
- Maggie M Shin
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, United States
| | - Catarina Catela
- Department of Neurobiology, University of Chicago, Chicago, United States
| | - Jeremy Dasen
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, United States
| |
Collapse
|
22
|
Baek M, Menon V, Jessell TM, Hantman AW, Dasen JS. Molecular Logic of Spinocerebellar Tract Neuron Diversity and Connectivity. Cell Rep 2020; 27:2620-2635.e4. [PMID: 31141687 PMCID: PMC6555431 DOI: 10.1016/j.celrep.2019.04.113] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/01/2019] [Accepted: 04/26/2019] [Indexed: 01/07/2023] Open
Abstract
Coordinated motor behaviors depend on feedback communication between peripheral sensory systems and central circuits in the brain and spinal cord. Relay of muscle- and tendon-derived sensory information to the CNS is facilitated by functionally and anatomically diverse groups of spinocerebellar tract neurons (SCTNs), but the molecular logic by which SCTN diversity and connectivity is achieved is poorly understood. We used single-cell RNA sequencing and genetic manipulations to define the mechanisms governing the molecular profile and organization of SCTN subtypes. We found that SCTNs relaying proprioceptive sensory information from limb and axial muscles are generated through segmentally restricted actions of specific Hox genes. Loss of Hox function disrupts SCTN-subtype-specific transcriptional programs, leading to defects in the connections between proprioceptive sensory neurons, SCTNs, and the cerebellum. These results indicate that Hox-dependent genetic programs play essential roles in the assembly of neural circuits necessary for communication between the brain and spinal cord. Baek et al. show that Hox-transcription factor-dependent programs govern the specification and connectivity of spinal interneurons that relay muscle-derived sensory information to the cerebellum. These findings shed light on the development of neural circuits required for proprioception—the perception of body position.
Collapse
Affiliation(s)
- Myungin Baek
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA; Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Republic of Korea
| | - Vilas Menon
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA; Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Thomas M Jessell
- Departments of Neuroscience and Biochemistry and Molecular Biophysics, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Adam W Hantman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA.
| | - Jeremy S Dasen
- Neuroscience Institute, Department of Neuroscience and Physiology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
23
|
Assaraf E, Blecher R, Heinemann-Yerushalmi L, Krief S, Carmel Vinestock R, Biton IE, Brumfeld V, Rotkopf R, Avisar E, Agar G, Zelzer E. Piezo2 expressed in proprioceptive neurons is essential for skeletal integrity. Nat Commun 2020; 11:3168. [PMID: 32576830 PMCID: PMC7311488 DOI: 10.1038/s41467-020-16971-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 05/26/2020] [Indexed: 11/24/2022] Open
Abstract
In humans, mutations in the PIEZO2 gene, which encodes for a mechanosensitive ion channel, were found to result in skeletal abnormalities including scoliosis and hip dysplasia. Here, we show in mice that loss of Piezo2 expression in the proprioceptive system recapitulates several human skeletal abnormalities. While loss of Piezo2 in chondrogenic or osteogenic lineages does not lead to human-like skeletal abnormalities, its loss in proprioceptive neurons leads to spine malalignment and hip dysplasia. To validate the non-autonomous role of proprioception in hip joint morphogenesis, we studied this process in mice mutant for proprioceptive system regulators Runx3 or Egr3. Loss of Runx3 in the peripheral nervous system, but not in skeletal lineages, leads to similar joint abnormalities, as does Egr3 loss of function. These findings expand the range of known regulatory roles of the proprioception system on the skeleton and provide a central component of the underlying molecular mechanism, namely Piezo2.
Collapse
Affiliation(s)
- Eran Assaraf
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 76100, Israel
- Department of Orthopedic Surgery, Assaf HaRofeh Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Zerrifin, 70300, Israel
| | - Ronen Blecher
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 76100, Israel
- Department of Orthopedic Surgery, Assuta Ashdod University Hospital, Ashdod, 7747629, Israel
- Ben Gurion University of the Negev, Beer-Sheva, 8410501, Israel
| | | | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Ron Carmel Vinestock
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Inbal E Biton
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Vlad Brumfeld
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Ron Rotkopf
- Bioinformatics Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Erez Avisar
- Department of Orthopedic Surgery, Assaf HaRofeh Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Zerrifin, 70300, Israel
| | - Gabriel Agar
- Department of Orthopedic Surgery, Assaf HaRofeh Medical Center, Sackler Faculty of Medicine, Tel Aviv University, Zerrifin, 70300, Israel
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
24
|
Helmbacher F, Stricker S. Tissue cross talks governing limb muscle development and regeneration. Semin Cell Dev Biol 2020; 104:14-30. [PMID: 32517852 DOI: 10.1016/j.semcdb.2020.05.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 05/07/2020] [Accepted: 05/08/2020] [Indexed: 12/14/2022]
Abstract
For decades, limb development has been a paradigm of three-dimensional patterning. Moreover, as the limb muscles and the other tissues of the limb's musculoskeletal system arise from distinct developmental sources, it has been a prime example of integrative morphogenesis and cross-tissue communication. As the limbs grow, all components of the musculoskeletal system (muscles, tendons, connective tissue, nerves) coordinate their growth and differentiation, ultimately giving rise to a functional unit capable of executing elaborate movement. While the molecular mechanisms governing global three-dimensional patterning and formation of the skeletal structures of the limbs has been a matter of intense research, patterning of the soft tissues is less understood. Here, we review the development of limb muscles with an emphasis on their interaction with other tissue types and the instructive roles these tissues play. Furthermore, we discuss the role of adult correlates of these embryonic accessory tissues in muscle regeneration.
Collapse
Affiliation(s)
| | - Sigmar Stricker
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195, Berlin, Germany.
| |
Collapse
|
25
|
Gabrych DR, Lau VZ, Niwa S, Silverman MA. Going Too Far Is the Same as Falling Short †: Kinesin-3 Family Members in Hereditary Spastic Paraplegia. Front Cell Neurosci 2019; 13:419. [PMID: 31616253 PMCID: PMC6775250 DOI: 10.3389/fncel.2019.00419] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/02/2019] [Indexed: 01/18/2023] Open
Abstract
Proper intracellular trafficking is essential for neuronal development and function, and when any aspect of this process is dysregulated, the resulting "transportopathy" causes neurological disorders. Hereditary spastic paraplegias (HSPs) are a family of such diseases attributed to over 80 spastic gait genes (SPG), specifically characterized by lower extremity spasticity and weakness. Multiple genes in the trafficking pathway such as those relating to microtubule structure and function and organelle biogenesis are representative disease loci. Microtubule motor proteins, or kinesins, are also causal in HSP, specifically mutations in Kinesin-I/KIF5A (SPG10) and two kinesin-3 family members; KIF1A (SPG30) and KIF1C (SPG58). KIF1A is a motor enriched in neurons, and involved in the anterograde transport of a variety of vesicles that contribute to pre- and post-synaptic assembly, autophagic processes, and neuron survival. KIF1C is ubiquitously expressed and, in addition to anterograde cargo transport, also functions in retrograde transport between the Golgi and the endoplasmic reticulum. Only a handful of KIF1C cargos have been identified; however, many have crucial roles such as neuronal differentiation, outgrowth, plasticity and survival. HSP-related kinesin-3 mutants are characterized mainly as loss-of-function resulting in deficits in motility, regulation, and cargo binding. Gain-of-function mutants are also seen, and are characterized by increased microtubule-on rates and hypermotility. Both sets of mutations ultimately result in misdelivery of critical cargos within the neuron. This likely leads to deleterious cell biological cascades that likely underlie or contribute to HSP clinical pathology and ultimately, symptomology. Due to the paucity of histopathological or cell biological data assessing perturbations in cargo localization, it has been difficult to positively link these mutations to the outcomes seen in HSPs. Ultimately, the goal of this review is to encourage future academic and clinical efforts to focus on "transportopathies" through a cargo-centric lens.
Collapse
Affiliation(s)
- Dominik R Gabrych
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Victor Z Lau
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Shinsuke Niwa
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Michael A Silverman
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada.,Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
26
|
Valdez G. Effects of disease-afflicted and aging neurons on the musculoskeletal system. Bone 2019; 122:31-37. [PMID: 30695738 PMCID: PMC6444351 DOI: 10.1016/j.bone.2019.01.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 01/09/2023]
Abstract
The musculoskeletal system includes skeletal muscles, bones and innervating axons from neurons in the central and peripheral nervous systems. Together, they form the largest structure in the body. They also initiate and coordinate locomotion, provide structural stability, and contribute to metabolism and homeostasis. Because of these functions, much effort has been devoted to ascertaining the impact of acute and chronic stress, such as disease, injury and aging, on the musculoskeletal system. This review will examine the role of the nervous system in the deleterious changes that accrue in skeletal muscles and bones during the progression of neurologic diseases and with advancing age.
Collapse
Affiliation(s)
- Gregorio Valdez
- Fralin Biomedical Research Institute, Virginia Tech Carilion, Roanoke, VA, USA; Department of Biological Sciences, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
27
|
Blecher R, Heinemann-Yerushalmi L, Assaraf E, Konstantin N, Chapman JR, Cope TC, Bewick GS, Banks RW, Zelzer E. New functions for the proprioceptive system in skeletal biology. Philos Trans R Soc Lond B Biol Sci 2018; 373:20170327. [PMID: 30249776 PMCID: PMC6158198 DOI: 10.1098/rstb.2017.0327] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2018] [Indexed: 01/13/2023] Open
Abstract
Muscle spindles and Golgi tendon organs (GTOs) are two types of sensory receptors that respond to changes in length or tension of skeletal muscles. These mechanosensors have long been known to participate in both proprioception and stretch reflex. Here, we present recent findings implicating these organs in maintenance of spine alignment as well as in realignment of fractured bones. These discoveries have been made in several mouse lines lacking functional mechanosensors in part or completely. In both studies, the absence of functional spindles and GTOs produced a more severe phenotype than that of spindles alone. Interestingly, the spinal curve phenotype, which appeared during peripubertal development, bears resemblance to the human condition adolescent idiopathic scoliosis. This similarity may contribute to the study of the disease by offering both an animal model and a clue as to its aetiology. Moreover, it raises the possibility that impaired proprioceptive signalling may be involved in the aetiology of other conditions. Overall, these new findings expand considerably the scope of involvement of proprioception in musculoskeletal development and function.This article is part of the Theo Murphy meeting issue 'Mechanics of development'.
Collapse
Affiliation(s)
- Ronen Blecher
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
- Department of Orthopedic Surgery, Assaf HaRofeh Medical Center, Zerrifin 70300, Israel, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Swedish Neuroscience Institute, Seattle, WA 98122, USA
| | | | - Eran Assaraf
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
- Department of Orthopedic Surgery, Assaf HaRofeh Medical Center, Zerrifin 70300, Israel, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nitzan Konstantin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | - Timothy C Cope
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA 30332, USA
| | - Guy S Bewick
- Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Robert W Banks
- Department of Biosciences, Durham University, Durham DH1 3LE, UK
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
28
|
Grb2-associated binder-1 is required for extrafusal and intrafusal muscle fiber development. Neuroreport 2018; 28:604-609. [PMID: 28542067 DOI: 10.1097/wnr.0000000000000807] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The neuregulin-1 (NRG1) signaling pathway plays an important role in the development of the peripheral neuromuscular system, including in muscle spindle and postnatal myelination. We previously showed that NRG1 on the axonal membrane regulates peripheral nerve myelination through Grb2-associated binder 1 (Gab1), a scaffolding mediator of receptor tyrosine kinase signaling. Here, we determined the role of Gab1 in the development of muscles and the muscle spindle using muscle-specific conditional Gab1 knockout mice. The mutant mice showed general retardation in muscular growth and hypotrophy of extrafusal muscle fibers. In addition, the muscle-specific Gab1 knockout mutant exhibited significant underdevelopment of muscle spindles, which are normally regulated by NRG1, and abnormal proprioceptive behavior. Furthermore, the selective knockdown of Gab1 in C2C12 muscle cells reduced NRG1-induced expression of Egr3, a critical transcription factor for muscle spindle development. However, Gab2 knockout mice did not show any defects in the development of muscles or muscle spindles. Our findings suggest that Gab1 is an essential signaling molecule in mediating axonal NRG1 signaling for the development of both extrafusal and intrafusal muscle fibers.
Collapse
|
29
|
Abstract
During embryogenesis, the musculoskeletal system develops while containing within itself a force generator in the form of the musculature. This generator becomes functional relatively early in development, exerting an increasing mechanical load on neighboring tissues as development proceeds. A growing body of evidence indicates that such mechanical forces can be translated into signals that combine with the genetic program of organogenesis. This unique situation presents both a major challenge and an opportunity to the other tissues of the musculoskeletal system, namely bones, joints, tendons, ligaments and the tissues connecting them. Here, we summarize the involvement of muscle-induced mechanical forces in the development of various vertebrate musculoskeletal components and their integration into one functional unit.
Collapse
Affiliation(s)
- Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
30
|
Imai F, Chen X, Weirauch MT, Yoshida Y. Requirement for Dicer in Maintenance of Monosynaptic Sensory-Motor Circuits in the Spinal Cord. Cell Rep 2017; 17:2163-2172. [PMID: 27880894 DOI: 10.1016/j.celrep.2016.10.083] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 08/08/2016] [Accepted: 10/24/2016] [Indexed: 01/19/2023] Open
Abstract
In contrast to our knowledge of mechanisms governing circuit formation, our understanding of how neural circuits are maintained is limited. Here, we show that Dicer, an RNaseIII protein required for processing microRNAs (miRNAs), is essential for maintenance of the spinal monosynaptic stretch reflex circuit in which group Ia proprioceptive sensory neurons form direct connections with motor neurons. In postnatal mice lacking Dicer in proprioceptor sensory neurons, there are no obvious defects in specificity or formation of monosynaptic sensory-motor connections. However, these circuits degrade through synapse loss and retraction of proprioceptive axonal projections from the ventral spinal cord. Peripheral terminals are also impaired without retracting from muscle targets. Interestingly, despite these central and peripheral axonal defects, proprioceptive neurons survive in the absence of Dicer-processed miRNAs. These findings reveal that Dicer, through its production of mature miRNAs, plays a key role in the maintenance of monosynaptic sensory-motor circuits.
Collapse
Affiliation(s)
- Fumiyasu Imai
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Xiaoting Chen
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Matthew T Weirauch
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA; Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Yutaka Yoshida
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| |
Collapse
|
31
|
Blecher R, Krief S, Galili T, Assaraf E, Stern T, Anekstein Y, Agar G, Zelzer E. The Proprioceptive System Regulates Morphologic Restoration of Fractured Bones. Cell Rep 2017; 20:1775-1783. [PMID: 28834742 PMCID: PMC5575358 DOI: 10.1016/j.celrep.2017.07.073] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 07/12/2017] [Accepted: 07/20/2017] [Indexed: 12/29/2022] Open
Abstract
Successful fracture repair requires restoration of bone morphology and mechanical integrity. Recent evidence shows that fractured bones of neonatal mice undergo spontaneous realignment, dubbed "natural reduction." Here, we show that natural reduction is regulated by the proprioceptive system and improves with age. Comparison among mice of different ages revealed, surprisingly, that 3-month-old mice exhibited more rapid and effective natural reduction than newborns. Fractured bones of null mutants for transcription factor Runx3, lacking functional proprioceptors, failed to realign properly. Blocking Runx3 expression in the peripheral nervous system, but not in limb mesenchyme, recapitulated the null phenotype, as did inactivation of muscles flanking the fracture site. Egr3 knockout mice, which lack muscle spindles but not Golgi tendon organs, displayed a less severe phenotype, suggesting that both receptor types, as well as muscle contraction, are required for this regulatory mechanism. These findings uncover a physiological role for proprioception in non-autonomous regulation of skeletal integrity.
Collapse
Affiliation(s)
- Ronen Blecher
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Orthopedic Surgery, Assaf Harofeh Medical Center, Zerrifin 70300, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Tal Galili
- Department of Statistics and Operations Research, Tel Aviv University, Tel Aviv 69978, Israel
| | - Eran Assaraf
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel; Department of Orthopedic Surgery, Assaf Harofeh Medical Center, Zerrifin 70300, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tomer Stern
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yoram Anekstein
- Department of Orthopedic Surgery, Assaf Harofeh Medical Center, Zerrifin 70300, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gabriel Agar
- Department of Orthopedic Surgery, Assaf Harofeh Medical Center, Zerrifin 70300, Israel, affiliated with the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
32
|
Vaughan SK, Stanley OL, Valdez G. Impact of Aging on Proprioceptive Sensory Neurons and Intrafusal Muscle Fibers in Mice. J Gerontol A Biol Sci Med Sci 2017; 72:771-779. [PMID: 27688482 DOI: 10.1093/gerona/glw175] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 08/09/2016] [Indexed: 11/13/2022] Open
Abstract
The impact of aging on proprioceptive sensory neurons and intrafusal muscle fibers (IMFs) remains largely unexplored despite the central function these cells play in modulating voluntary movements. Here, we show that proprioceptive sensory neurons undergo deleterious morphological changes in middle age (11- to 13-month-old) and old (15- to 21-month-old) mice. In the extensor digitorum longus and soleus muscles of middle age and old mice, there is a significant increase in the number of Ia afferents with large swellings that fail to properly wrap around IMFs compared with young adult (2- to 4-month-old) mice. Fewer II afferents were also found in the same muscles of middle age and old mice. Although these age-related changes in peripheral nerve endings were accompanied by degeneration of proprioceptive sensory neuron cell bodies in dorsal root ganglia (DRG), the morphology and number of IMFs remained unchanged. Our analysis also revealed normal levels of neurotrophin 3 (NT3) but dysregulated expression of the tyrosine kinase receptor C (TrkC) in aged muscles and DRGs, respectively. These results show that proprioceptive sensory neurons degenerate prior to atrophy of IMFs during aging, and in the presence of the NT3/TrkC signaling axis.
Collapse
Affiliation(s)
- Sydney K Vaughan
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke.,Graduate Program in Translational Biology, Medicine, and Health and
| | - Olivia L Stanley
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke
| | - Gregorio Valdez
- Virginia Tech Carilion Research Institute, Virginia Tech, Roanoke.,Department of Biological Sciences, Virginia Tech, Blacksburg
| |
Collapse
|
33
|
Wang F, Wang Q, Li C, Yu P, Qu Y, Zhou L. The role of Celsr3 in the development of central somatosensory projections from dorsal root ganglia. Neuroscience 2017; 359:267-276. [PMID: 28754314 DOI: 10.1016/j.neuroscience.2017.07.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 06/29/2017] [Accepted: 07/17/2017] [Indexed: 01/10/2023]
Abstract
Dorsal root ganglion (DRG) neurons receive peripheral somatosensory information and send orderly projections to second-order relay nuclei in the spinal cord and in the brainstem. Atypical cadherin Celsr3 is known to play a critical role in wiring of several central and peripheral axons. Although Celsr3 mRNA is heavily expressed in DRG neurons, its role in the development of somatosensory projections remains unexplored. Here we assessed the role of Celsr3 in DRG using conditional gene inactivation in crosses with Wnt1-Cre mice. Using Celsr3-GFP transgenic mice, we found that Celsr3 was highly expressed in different DRG cells, such as Pavalbumin-, TrkB-, and calcitonin gene-related peptide (CGRP)-positive neurons. Wnt1-Cre;Celsr3f/- animals survived for a few weeks and looked smaller than littermate controls. DiI tracing showed that early DRG axons entered the spinal cord and reached spinal cord targets similarly in mutant and control mice. CGRP-positive fiber density was significantly decreased in lamina I in the mutant versus control spinal cord at postnatal day (P) 7 and P14. Furthermore, more Pavalbumin-positive fibers invaded the gray matter and made more contacts with spinal motor neurons in mutant than in control samples. Behavioral analysis showed that mutant animals were less sensitive to pain and more sensitive to mechanical stimulation than controls. In conclusion, Celsr3 is dispensable for the patterning of central DRG projections, but it regulates for the fine mapping of sensory fibers in the gray matter, which is important for somatosensory processing.
Collapse
Affiliation(s)
- Feifei Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Qianghua Wang
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Chen Li
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Panpan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Yibo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China
| | - Libing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, PR China; Co-innovation Center of Neuroregeneration, Jiangsu, PR China; Key Laboratory of Neuroscience, School of Basic Medical Sciences, Institute of Neuroscience, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
34
|
Descending Systems Direct Development of Key Spinal Motor Circuits. J Neurosci 2017; 37:6372-6387. [PMID: 28576940 DOI: 10.1523/jneurosci.0149-17.2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/10/2017] [Accepted: 05/24/2017] [Indexed: 12/17/2022] Open
Abstract
The formation of mature spinal motor circuits is dependent on both activity-dependent and independent mechanisms during postnatal development. During this time, reorganization and refinement of spinal sensorimotor circuits occurs as supraspinal projections are integrated. However, specific features of postnatal spinal circuit development remain poorly understood. This study provides the first detailed characterization of rat spinal sensorimotor circuit development in the presence and absence of descending systems. We show that the development of proprioceptive afferent input to motoneurons (MNs) and Renshaw cells (RCs) is disrupted by thoracic spinal cord transection at postnatal day 5 (P5TX). P5TX also led to malformation of GABApre neuron axo-axonic contacts on Ia afferents and of the recurrent inhibitory circuit between MNs and RCs. Using a novel in situ perfused preparation for studying motor control, we show that malformation of these spinal circuits leads to hyperexcitability of the monosynaptic reflex. Our results demonstrate that removing descending input severely disrupts the development of spinal circuits and identifies key mechanisms contributing to motor dysfunction in conditions such as cerebral palsy and spinal cord injury.SIGNIFICANCE STATEMENT Acquisition of mature behavior during postnatal development correlates with the arrival and maturation of supraspinal projections to the spinal cord. However, we know little about the role that descending systems play in the maturation of spinal circuits. Here, we characterize postnatal development of key spinal microcircuits in the presence and absence of descending systems. We show that formation of these circuits is abnormal after early (postnatal day 5) removal of descending systems, inducing hyperexcitability of the monosynaptic reflex. The study is a detailed characterization of spinal circuit development elucidating how these mechanisms contribute to motor dysfunction in conditions such as cerebral palsy and spinal cord injury. Understanding these circuits is crucial to developing new therapeutics and improving existing ones in such conditions.
Collapse
|
35
|
Geertsen SS, Willerslev-Olsen M, Lorentzen J, Nielsen JB. Development and aging of human spinal cord circuitries. J Neurophysiol 2017; 118:1133-1140. [PMID: 28566459 DOI: 10.1152/jn.00103.2017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/25/2017] [Accepted: 05/25/2017] [Indexed: 01/25/2023] Open
Abstract
The neural motor circuitries in the spinal cord receive information from our senses and the rest of the nervous system and translate it into purposeful movements, which allow us to interact with the rest of the world. In this review, we discuss how these circuitries are established during early development and the extent to which they are shaped according to the demands of the body that they control and the environment with which the body has to interact. We also discuss how aging processes and physiological changes in our body are reflected in adaptations of activity in the spinal cord motor circuitries. The complex, multifaceted connectivity of the spinal cord motor circuitries allows them to generate vastly different movements and to adapt their activity to meet new challenges imposed by bodily changes or a changing environment. There are thus plenty of possibilities for adaptive changes in the spinal motor circuitries both early and late in life.
Collapse
Affiliation(s)
- Svend Sparre Geertsen
- Neural Control of Movement Research Group, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark.,Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen N, Denmark; and
| | - Maria Willerslev-Olsen
- Neural Control of Movement Research Group, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark.,Elsass Institute, Charlottenlund, Denmark
| | - Jakob Lorentzen
- Neural Control of Movement Research Group, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark.,Elsass Institute, Charlottenlund, Denmark
| | - Jens Bo Nielsen
- Neural Control of Movement Research Group, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark; .,Elsass Institute, Charlottenlund, Denmark
| |
Collapse
|
36
|
Fletcher EV, Simon CM, Pagiazitis JG, Chalif JI, Vukojicic A, Drobac E, Wang X, Mentis GZ. Reduced sensory synaptic excitation impairs motor neuron function via Kv2.1 in spinal muscular atrophy. Nat Neurosci 2017; 20:905-916. [PMID: 28504671 PMCID: PMC5487291 DOI: 10.1038/nn.4561] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 04/04/2017] [Indexed: 12/30/2022]
Abstract
Behavioral deficits in neurodegenerative diseases are often attributed to the selective dysfunction of vulnerable neurons via cell-autonomous mechanisms. Although vulnerable neurons are embedded in neuronal circuits, the contribution of their synaptic partners to the disease process is largely unknown. Here, we show that in a mouse model of spinal muscular atrophy (SMA), a reduction in proprioceptive synaptic drive leads to motor neuron dysfunction and motor behavior impairments. In SMA mice or after the blockade of proprioceptive synaptic transmission we observed a decrease in the motor neuron firing which could be explained by the reduction in the expression of the potassium channel Kv2.1 at the surface of motor neurons. Increasing neuronal activity pharmacologically by chronic exposure in vivo led to a normalization of Kv2.1 expression and an improvement in motor function. Our results demonstrate a key role of excitatory synaptic drive in shaping the function of motor neurons during development and the contribution of its disruption to a neurodegenerative disease.
Collapse
Affiliation(s)
- Emily V Fletcher
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Christian M Simon
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - John G Pagiazitis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Joshua I Chalif
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Aleksandra Vukojicic
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Estelle Drobac
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Xiaojian Wang
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - George Z Mentis
- Center for Motor Neuron Biology and Disease, Columbia University, New York, New York, USA.,Department of Pathology and Cell Biology, Columbia University, New York, New York, USA.,Department of Neurology, Columbia University, New York, New York, USA
| |
Collapse
|
37
|
Guo X, Colon A, Akanda N, Spradling S, Stancescu M, Martin C, Hickman JJ. Tissue engineering the mechanosensory circuit of the stretch reflex arc with human stem cells: Sensory neuron innervation of intrafusal muscle fibers. Biomaterials 2017; 122:179-187. [PMID: 28129596 DOI: 10.1016/j.biomaterials.2017.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 12/28/2016] [Accepted: 01/03/2017] [Indexed: 12/28/2022]
Abstract
Muscle spindles are sensory organs embedded in the belly of skeletal muscles that serve as mechanoreceptors detecting static and dynamic information about muscle length and stretch. Through their connection with proprioceptive sensory neurons, sensation of axial body position and muscle movement are transmitted to the central nervous system. Impairment of this sensory circuit causes motor deficits and has been linked to a wide range of diseases. To date, no defined human-based in vitro model of the proprioceptive sensory circuit has been developed. The goal of this study was to develop a human-based in vitro muscle sensory circuit utilizing human stem cells. A serum-free medium was developed to drive the induction of intrafusal fibers from human satellite cells by actuation of a neuregulin signaling pathway. Both bag and chain intrafusal fibers were generated and subsequently validated by phase microscopy and immunocytochemistry. When co-cultured with proprioceptive sensory neurons derived from human neuroprogenitors, mechanosensory nerve terminal structural features with intrafusal fibers were demonstrated. Most importantly, patch-clamp electrophysiological analysis of the intrafusal fibers indicated repetitive firing of human intrafusal fibers, which has not been observed in human extrafusal fibers.
Collapse
Affiliation(s)
- Xiufang Guo
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Alisha Colon
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, 12722 Research Parkway, Orlando, FL 32826, USA
| | - Nesar Akanda
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - Severo Spradling
- Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, 12722 Research Parkway, Orlando, FL 32826, USA
| | - Maria Stancescu
- Department of Chemistry, 4000 Central Florida Blvd., Physical Sciences Building (PS) Room 255, University of Central Florida, Orlando, FL 32816-2366, USA
| | - Candace Martin
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA
| | - James J Hickman
- Hybrid Systems Lab, NanoScience Technology Center, University of Central Florida, 12424 Research Parkway, Suite 400, Orlando, FL 32826, USA; Biomolecular Science Center, Burnett School of Biomedical Sciences, University of Central Florida, 12722 Research Parkway, Orlando, FL 32826, USA; Department of Chemistry, 4000 Central Florida Blvd., Physical Sciences Building (PS) Room 255, University of Central Florida, Orlando, FL 32816-2366, USA.
| |
Collapse
|
38
|
Nikolaou S, Hu L, Cornwall R. Afferent Innervation, Muscle Spindles, and Contractures Following Neonatal Brachial Plexus Injury in a Mouse Model. J Hand Surg Am 2015; 40:2007-16. [PMID: 26319770 DOI: 10.1016/j.jhsa.2015.07.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 02/02/2023]
Abstract
PURPOSE We used an established mouse model of elbow flexion contracture after neonatal brachial plexus injury (NBPI) to test the hypothesis that preservation of afferent innervation protects against contractures and is associated with preservation of muscle spindles and ErbB signaling. METHODS A model of preganglionic C5 through C7 NBPI was first tested in mice with fluorescent axons using confocal imaging to confirm preserved afferent innervation of spindles despite motor end plate denervation. Preganglionic and postganglionic injuries were then created in wild-type mice. Four weeks later, we assessed total and afferent denervation of the elbow flexors by musculocutaneous nerve immunohistochemistry. Biceps muscle volume and cross-sectional area were measured by micro computed tomography. An observer who was blinded to the study protocol measured elbow flexion contractures. Biceps spindle and muscle fiber morphology and ErbB signaling pathway activity were assessed histologically and immunohistochemically. RESULTS Preganglionic and postganglionic injuries caused similar total denervation and biceps muscle atrophy. However, after preganglionic injuries, afferent innervation was partially preserved and elbow flexion contractures were significantly less severe. Spindles degenerated after postganglionic injury but were preserved after preganglionic injury. ErbB signaling was inactivated in denervated spindles after postganglionic injury but ErbB signaling activity was preserved in spindles after preganglionic injury with retained afferent innervation. Preganglionic and postganglionic injuries were associated with upregulation of ErbB signaling in extrafusal muscle fibers. CONCLUSIONS Contractures after NBPI are associated with muscle spindle degeneration and loss of spindle ErbB signaling activity. Preservation of afferent innervation maintained spindle development and ErbB signaling activity, and protected against contractures. CLINICAL RELEVANCE Pharmacologic modulation of ErbB signaling, which is being investigated as a therapy for congestive heart failure, may be able to recapitulate the protective effects of afferent innervation in spindle development and contracture prevention. Muscle spindle preservation may also have implications in proprioception and motor learning, both of which are impaired in NBPI.
Collapse
Affiliation(s)
- Sia Nikolaou
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Liangjun Hu
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Roger Cornwall
- Division of Orthopaedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| |
Collapse
|
39
|
Huang L, Xian Q, Shen N, Shi L, Qu Y, Zhou L. Congenital absence of corticospinal tract does not severely affect plastic changes of the developing postnatal spinal cord. Neuroscience 2015; 301:338-50. [DOI: 10.1016/j.neuroscience.2015.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2015] [Revised: 06/06/2015] [Accepted: 06/08/2015] [Indexed: 11/25/2022]
|
40
|
Development of twitching in sleeping infant mice depends on sensory experience. Curr Biol 2015; 25:656-62. [PMID: 25702578 DOI: 10.1016/j.cub.2015.01.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/23/2014] [Accepted: 01/07/2015] [Indexed: 01/15/2023]
Abstract
Myoclonic twitches are jerky movements that occur exclusively and abundantly during active (or REM) sleep in mammals, especially in early development [1-4]. In rat pups, limb twitches exhibit a complex spatiotemporal structure that changes across early development [5]. However, it is not known whether this developmental change is influenced by sensory experience, which is a prerequisite to the notion that sensory feedback from twitches not only activates sensorimotor circuits but modifies them [4]. Here, we investigated the contributions of proprioception to twitching in newborn ErbB2 conditional knockout mice that lack muscle spindles and grow up to exhibit dysfunctional proprioception [6-8]. High-speed videography of forelimb twitches unexpectedly revealed a category of reflex-like twitching-comprising an agonist twitch followed immediately by an antagonist twitch-that developed postnatally in wild-types/heterozygotes, but not in knockouts. Contrary to evidence from adults that spinal reflexes are inhibited during twitching [9-11], this finding suggests that twitches trigger the monosynaptic stretch reflex and, by doing so, contribute to its activity-dependent development [12-14]. Next, we assessed developmental changes in the frequency and organization (i.e., entropy) of more-complex, multi-joint patterns of twitching; again, wild-types/heterozygotes exhibited developmental changes in twitch patterning that were not seen in knockouts. Thus, targeted deletion of a peripheral sensor alters the normal development of local and global features of twitching, demonstrating that twitching is shaped by sensory experience. These results also highlight the potential use of twitching as a uniquely informative diagnostic tool for assessing the functional status of spinal and supraspinal circuits.
Collapse
|
41
|
Clowry GJ, Basuodan R, Chan F. What are the Best Animal Models for Testing Early Intervention in Cerebral Palsy? Front Neurol 2014; 5:258. [PMID: 25538677 PMCID: PMC4255621 DOI: 10.3389/fneur.2014.00258] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 11/21/2014] [Indexed: 11/13/2022] Open
Abstract
Interventions to treat cerebral palsy should be initiated as soon as possible in order to restore the nervous system to the correct developmental trajectory. One drawback to this approach is that interventions have to undergo exceptionally rigorous assessment for both safety and efficacy prior to use in infants. Part of this process should involve research using animals but how good are our animal models? Part of the problem is that cerebral palsy is an umbrella term that covers a number of conditions. There are also many causal pathways to cerebral palsy, such as periventricular white matter injury in premature babies, perinatal infarcts of the middle cerebral artery, or generalized anoxia at the time of birth, indeed multiple causes, including intra-uterine infection or a genetic predisposition to infarction, may need to interact to produce a clinically significant injury. In this review, we consider which animal models best reproduce certain aspects of the condition, and the extent to which the multifactorial nature of cerebral palsy has been modeled. The degree to which the corticospinal system of various animal models human corticospinal system function and development is also explored. Where attempts have already been made to test early intervention in animal models, the outcomes are evaluated in light of the suitability of the model.
Collapse
Affiliation(s)
- Gavin John Clowry
- Institute of Neuroscience, Newcastle University , Newcastle upon Tyne , UK
| | - Reem Basuodan
- Institute of Neuroscience, Newcastle University , Newcastle upon Tyne , UK
| | - Felix Chan
- Institute of Neuroscience, Newcastle University , Newcastle upon Tyne , UK
| |
Collapse
|
42
|
Zhang Y, Lin S, Karakatsani A, Rüegg MA, Kröger S. Differential regulation of AChR clustering in the polar and equatorial region of murine muscle spindles. Eur J Neurosci 2014; 41:69-78. [PMID: 25377642 DOI: 10.1111/ejn.12768] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 09/29/2014] [Accepted: 10/02/2014] [Indexed: 12/16/2022]
Abstract
Intrafusal fibers of muscle spindles are innervated in the central region by afferent sensory axons and at both polar regions by efferent γ-motoneurons. We previously demonstrated that both neuron-muscle contact sites contain cholinergic synapse-like specialisation, including aggregates of the nicotinic acetylcholine receptor (AChR). In this study we tested the hypothesis that agrin and its receptor complex (consisting of LRP4 and the tyrosine kinase MuSK) are involved in the aggregation of AChRs in muscle spindles, similar to their role at the neuromuscular junction. We show that agrin, MuSK and LRP4 are concentrated at the contact site between the intrafusal fibers and the sensory- and γ-motoneuron, respectively, and that they are expressed in the cell bodies of proprioceptive neurons in dorsal root ganglia. Moreover, agrin and LRP4, but not MuSK, are expressed in γ-motoneuron cell bodies in the ventral horn of the spinal cord. In agrin- and in MuSK-deficient mice, AChR aggregates are absent from the polar regions. In contrast, the subcellular concentration of AChRs in the central region where the sensory neuron contacts the intrafusal muscle fiber is apparently unaffected. Skeletal muscle-specific expression of miniagrin in agrin(-/-) mice in vivo is sufficient to restore the formation of γ-motoneuron endplates. These results show that agrin and MuSK are major determinants during the formation of γ-motoneuron endplates but appear dispensable for the aggregation of AChRs at the central region. Our results therefore suggest different molecular mechanisms for AChR clustering within two domains of intrafusal fibers.
Collapse
Affiliation(s)
- Yina Zhang
- Department of Physiological Genomics, Ludwig-Maximilians-University, Pettenkoferstrasse 12, D-80336, Munich, Germany; Helmholtz Center Munich, Neuherberg, Germany
| | | | | | | | | |
Collapse
|
43
|
Formation of cholinergic synapse-like specializations at developing murine muscle spindles. Dev Biol 2014; 393:227-235. [DOI: 10.1016/j.ydbio.2014.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/30/2022]
|
44
|
Marques HG, Bharadwaj A, Iida F. From spontaneous motor activity to coordinated behaviour: a developmental model. PLoS Comput Biol 2014; 10:e1003653. [PMID: 25057775 PMCID: PMC4109855 DOI: 10.1371/journal.pcbi.1003653] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 04/18/2014] [Indexed: 01/09/2023] Open
Abstract
In mammals, the developmental path that links the primary behaviours observed during foetal stages to the full fledged behaviours observed in adults is still beyond our understanding. Often theories of motor control try to deal with the process of incremental learning in an abstract and modular way without establishing any correspondence with the mammalian developmental stages. In this paper, we propose a computational model that links three distinct behaviours which appear at three different stages of development. In order of appearance, these behaviours are: spontaneous motor activity (SMA), reflexes, and coordinated behaviours, such as locomotion. The goal of our model is to address in silico four hypotheses that are currently hard to verify in vivo: First, the hypothesis that spinal reflex circuits can be self-organized from the sensor and motor activity induced by SMA. Second, the hypothesis that supraspinal systems can modulate reflex circuits to achieve coordinated behaviour. Third, the hypothesis that, since SMA is observed in an organism throughout its entire lifetime, it provides a mechanism suitable to maintain the reflex circuits aligned with the musculoskeletal system, and thus adapt to changes in body morphology. And fourth, the hypothesis that by changing the modulation of the reflex circuits over time, one can switch between different coordinated behaviours. Our model is tested in a simulated musculoskeletal leg actuated by six muscles arranged in a number of different ways. Hopping is used as a case study of coordinated behaviour. Our results show that reflex circuits can be self-organized from SMA, and that, once these circuits are in place, they can be modulated to achieve coordinated behaviour. In addition, our results show that our model can naturally adapt to different morphological changes and perform behavioural transitions.
Collapse
Affiliation(s)
| | - Arjun Bharadwaj
- Dept. of Mechanical and Process Engineering, ETH, Zurich, Switzerland
| | - Fumiya Iida
- Dept. of Mechanical and Process Engineering, ETH, Zurich, Switzerland
| |
Collapse
|
45
|
Boyce VS, Mendell LM. Neurotrophins and spinal circuit function. Front Neural Circuits 2014; 8:59. [PMID: 24926235 PMCID: PMC4046666 DOI: 10.3389/fncir.2014.00059] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2014] [Accepted: 05/19/2014] [Indexed: 01/19/2023] Open
Abstract
Work early in the last century emphasized the stereotyped activity of spinal circuits based on studies of reflexes. However, the last several decades have focused on the plasticity of these spinal circuits. These considerations began with studies of the effects of monoamines on descending and reflex circuits. In recent years new classes of compounds called growth factors that are found in peripheral nerves and the spinal cord have been shown to affect circuit behavior in the spinal cord. In this review we will focus on the effects of neurotrophins, particularly nerve growth factor (NGF), brain derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3), on spinal circuits. We also discuss evidence that these molecules can modify functions including nociceptive behavior, motor reflexes and stepping behavior. Since these substances and their receptors are normally present in the spinal cord, they could potentially be useful in improving function in disease states and after injury. Here we review recent findings relevant to these translational issues.
Collapse
Affiliation(s)
- Vanessa S Boyce
- Department of Neurobiology and Behavior, Stony Brook University Stony Brook, NY, USA
| | - Lorne M Mendell
- Department of Neurobiology and Behavior, Stony Brook University Stony Brook, NY, USA
| |
Collapse
|
46
|
Normal distribution of VGLUT1 synapses on spinal motoneuron dendrites and their reorganization after nerve injury. J Neurosci 2014; 34:3475-92. [PMID: 24599449 DOI: 10.1523/jneurosci.4768-13.2014] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Peripheral nerve injury induces permanent alterations in spinal cord circuitries that are not reversed by regeneration. Nerve injury provokes the loss of many proprioceptive IA afferent synapses (VGLUT1-IR boutons) from motoneurons, the reduction of IA EPSPs in motoneurons, and the disappearance of stretch reflexes. After motor and sensory axons successfully reinnervate muscle, lost IA VGLUT1 synapses are not re-established and the stretch reflex does not recover; however, electrically evoked EPSPs do recover. The reasons why remaining IA synapses can evoke EPSPs on motoneurons, but fail to transmit useful stretch signals are unknown. To better understand changes in the organization of VGLUT1 IA synapses that might influence their input strength, we analyzed their distribution over the entire dendritic arbor of motoneurons before and after nerve injury. Adult rats underwent complete tibial nerve transection followed by microsurgical reattachment and 1 year later motoneurons were intracellularly recorded and filled with neurobiotin to map the distribution of VGLUT1 synapses along their dendrites. We found in control motoneurons an average of 911 VGLUT1 synapses; ~62% of them were lost after injury. In controls, VGLUT1 synapses were focused to proximal dendrites where they were grouped in tight clusters. After injury, most synaptic loses occurred in the proximal dendrites and remaining synapses were declustered, smaller, and uniformly distributed throughout the dendritic arbor. We conclude that this loss and reorganization renders IA afferent synapses incompetent for efficient motoneuron synaptic depolarization in response to natural stretch, while still capable of eliciting EPSPs when synchronously fired by electrical volleys.
Collapse
|
47
|
Drummond MDM, Couto BP, Augusto IG, Rodrigues SA, Szmuchrowski LA. Effects of 12 weeks of dynamic strength training with local vibration. Eur J Sport Sci 2014; 14:695-702. [PMID: 24571184 DOI: 10.1080/17461391.2014.889757] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The purpose of this study was to investigate the chronic effects of dynamic strength training (ST) with local vibration on the maximum strength of elbow flexor muscles. Twenty healthy male untrained volunteers were divided randomly into the following two groups: the conventional training group (CTG) or the vibration training group (VTG). Both groups performed ST for 12 weeks, three times a week. The ST protocol included four sets of 8-10 repetition maximums (RMs) of unilateral elbow flexion exercise. The VTG performed this training protocol with local vibration at a frequency of 30 Hz and amplitude of 6 mm. The mean values of the one repetition maximum (1RM) tests for both groups increased significantly from the pretest week to the fourth week and from the fourth week to the eighth week (CTG: mean 19.02, s = 7.88%, p = 0.01; mean 10.50, s = 6.86%, p = 0.019, respectively; VTG: mean 16.02, s = 8.30%, p = 0.017; mean 12.55, s = 8.76%, p = 0.019, respectively). The increases in the maximal voluntary contraction (MVC) tests were also statistically significant from the pretest week to the fourth week and from the fourth week to the eighth week (CTG: mean 12.32, s = 8.33%, p = 0.004; mean 9.95, s = 5.32%, p = 0.006, respectively; VTG: mean 10.16, s = 11.71%, p = 0.003; mean 10.36, s = 2.96%, p = 0.01, respectively). There was no significant difference between the 1RM and MVC test results in the eighth and twelfth weeks in either group. No significant differences were observed between the groups (p < 0.05). In conclusion, the application of local vibration does not change the chronic effects of dynamic ST in untrained individuals.
Collapse
Affiliation(s)
- Marcos D M Drummond
- a Load Evaluation Laboratory , Federal University of Minas Gerais , Belo Horizonte , Brazil
| | | | | | | | | |
Collapse
|
48
|
Heath M, Sutherland C, Bartel K, Gradisar M, Williamson P, Lovato N, Micic G. Does one hour of bright or short-wavelength filtered tablet screenlight have a meaningful effect on adolescents’ pre-bedtime alertness, sleep, and daytime functioning? Chronobiol Int 2014; 31:496-505. [DOI: 10.3109/07420528.2013.872121] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Abstract
A major challenge in repairing the injured spinal cord is to assure survival of damaged cells and to encourage regrowth of severed axons. Because neurotrophins are known to affect these processes during development, many experimental approaches to improving function of the injured spinal cord have made use of these agents, particularly Brain derived neurotrophic factor (BDNF) and Neurotrophin-3 (NT-3). More recently, neurotrophins have also been shown to affect the physiology of cells and synapses in the spinal cord. The effect of neurotrophins on circuit performance adds an important dimension to their consideration as agents for repairing the injured spinal cord. In this chapter we discuss the role of neurotrophins in promoting recovery after spinal cord injury from both a structural and functional perspective.
Collapse
Affiliation(s)
- Vanessa S Boyce
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | | |
Collapse
|
50
|
Cheret C, Willem M, Fricker FR, Wende H, Wulf-Goldenberg A, Tahirovic S, Nave KA, Saftig P, Haass C, Garratt AN, Bennett DL, Birchmeier C. Bace1 and Neuregulin-1 cooperate to control formation and maintenance of muscle spindles. EMBO J 2013; 32:2015-28. [PMID: 23792428 PMCID: PMC3715864 DOI: 10.1038/emboj.2013.146] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 05/29/2013] [Indexed: 01/18/2023] Open
Abstract
The protease β-secretase 1 (Bace1) was identified through its critical role in production of amyloid-β peptides (Aβ), the major component of amyloid plaques in Alzheimer's disease. Bace1 is considered a promising target for the treatment of this pathology, but processes additional substrates, among them Neuregulin-1 (Nrg1). Our biochemical analysis indicates that Bace1 processes the Ig-containing β1 Nrg1 (IgNrg1β1) isoform. We find that a graded reduction in IgNrg1 signal strength in vivo results in increasingly severe deficits in formation and maturation of muscle spindles, a proprioceptive organ critical for muscle coordination. Further, we show that Bace1 is required for formation and maturation of the muscle spindle. Finally, pharmacological inhibition and conditional mutagenesis in adult animals demonstrate that Bace1 and Nrg1 are essential to sustain muscle spindles and to maintain motor coordination. Our results assign to Bace1 a role in the control of coordinated movement through its regulation of muscle spindle physiology, and implicate IgNrg1-dependent processing as a molecular mechanism. Bace1 is required for Nrg1 processing for muscle spindle development. Bace1 inhibition leads to loss of motor coordination even in adult mice, suggesting potentially serious side effects for drugs targeting Bace1 as a treatment for Alzheimer's disease.
Collapse
Affiliation(s)
- Cyril Cheret
- Entwicklungsbiologie/Signaltransduktion, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|