1
|
Piasecki M. Motor unit adaptation to disuse: crossing the threshold from firing rate suppression to neuromuscular junction transmission. J Physiol 2024. [PMID: 39496497 DOI: 10.1113/jp284159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
Neural conditioning to scenarios of muscle disuse is undoubtedly a cause of functional decrements that typically exceed losses of muscle size. Yet establishing the relative contribution of neural adaptation and the specific location in the motor pathway remains technically challenging. Several studies of healthy humans have targeted this system and have established that motor unit firing rate is suppressed following disuse, with a number of critical caveats. It is suppressed in the immobilized limb only, at relative and absolute force levels, and preferentially targets lower-threshold motor units. Concomitantly, electrophysiological investigation of neuromuscular junction transmission (NMJ) stability of lower-threshold motor units reveals minimal change following disuse. These findings contrast with numerous other methods, which show clear involvement of the NMJ but are unable to characterize the motor unit to which they belong. It is physiologically plausible that decrements observed following disuse are a result of suppressed firing rate of lower-threshold motor units and impairment of transmission of the NMJ of higher-threshold motor units. As such, motor units within the pool should be viewed in light of their varying susceptibility to disuse.
Collapse
Affiliation(s)
- Mathew Piasecki
- Centre of Metabolism, Ageing & Physiology (CoMAP), Medical Research Council/Versus Arthritis UK Centre of Excellence for Musculoskeletal Ageing Research (CMAR), NIHR Nottingham Biomedical Research Centre, University of Nottingham, Derby, UK
| |
Collapse
|
2
|
Martynyuk T, Ricard J, Bracchi-Ricard V, Price S, McGrath JR, Dougherty KJ, Tom V, Bethea JR. Mitigating sTNF/TNFR1 activation on VGluT2 + spinal cord interneurons improves immune function after mid-thoracic spinal cord injury. Brain Behav Immun 2024; 123:633-643. [PMID: 39414179 DOI: 10.1016/j.bbi.2024.10.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with 250,000 to 500,000 new cases globally each year. Respiratory infections, e.g., pneumonia and influenza are the leading cause of death after SCI. Unfortunately, there is a poor understanding of how altered neuro-immune communication impacts an individual's outcome to infection. In humans and rodents, SCI leads to maladaptive changes in the spinal-sympathetic reflex (SSR) circuit which is crucial to sympathetic function. The cause of the impaired immune function may be related to harmful neuroinflammation which is detrimental to homeostatic neuronal function, aberrant plasticity, and hyperexcitable circuits. Soluble tumor necrosis factor (sTNF) is a pro-inflammatory cytokine that is elevated in the CNS after SCI and remains elevated for several months after injury. By pharmacologically attenuating sTNF in the CNS after SCI we were able to demonstrate improved immune function. Furthermore, when we investigated the specific cellular population which may be involved in altered neuro-immune communication we reported that excessive TNFR1 activity on excitatory INs promotes immune dysfunction. Furthermore, this observation is NF-kβ dependent in VGluT2 + INs. Our data is the first report of a target within the CNS, TNFR1, that contributes to SCI-induced immune dysfunction after T9-SCI and is a potential avenue for future therapeutics.
Collapse
Affiliation(s)
- Tetyana Martynyuk
- Drexel University, Department of Biology, Philadelphia, PA, United States.
| | - Jerome Ricard
- Drexel University, Department of Biology, Philadelphia, PA, United States
| | | | - Samuel Price
- Drexel University, Department of Biology, Philadelphia, PA, United States
| | - Jenna R McGrath
- Drexel University, Department of Neurobiology and Anatomy, Philadelphia, PA, United States
| | - Kimberly J Dougherty
- Drexel University, Department of Neurobiology and Anatomy, Philadelphia, PA, United States
| | - Veronica Tom
- Drexel University, Department of Neurobiology and Anatomy, Philadelphia, PA, United States
| | - John R Bethea
- Drexel University, Department of Biology, Philadelphia, PA, United States; George Washington University, Department of Anatomy and Cell Biology, Washington, D.C., United States.
| |
Collapse
|
3
|
Hung 洪瑋辰 WC, Chen 陳志成 CC, Yen 嚴震東 CT, Min 閔明源 MY. Presynaptic Enhancement of Transmission from Nociceptors Expressing Nav1.8 onto Lamina-I Spinothalamic Tract Neurons by Spared Nerve Injury in Mice. eNeuro 2024; 11:ENEURO.0087-24.2024. [PMID: 39256039 PMCID: PMC11391502 DOI: 10.1523/eneuro.0087-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 08/19/2024] [Accepted: 08/23/2024] [Indexed: 09/12/2024] Open
Abstract
Alteration of synaptic function in the dorsal horn (DH) has been implicated as a cellular substrate for the development of neuropathic pain, but certain details remain unclear. In particular, the lack of information on the types of synapses that undergo functional changes hinders the understanding of disease pathogenesis from a synaptic plasticity perspective. Here, we addressed this issue by using optogenetic and retrograde tracing ex vivo to selectively stimulate first-order nociceptors expressing Nav1.8 (NRsNav1.8) and record the responses of spinothalamic tract neurons in spinal lamina I (L1-STTNs). We found that spared nerve injury (SNI) increased excitatory postsynaptic currents (EPSCs) in L1-STTNs evoked by photostimulation of NRsNav1.8 (referred to as Nav1.8-STTN EPSCs). This effect was accompanied by a significant change in the failure rate and paired-pulse ratio of synaptic transmission from NRsNav1.8 to L1-STTN and in the frequency (not amplitude) of spontaneous EPSCs recorded in L1-STTNs. However, no change was observed in the ratio of AMPA to NMDA receptor-mediated components of Nav1.8-STTN EPSCs or in the amplitude of unitary EPSCs constituting Nav1.8-STTN EPSCs recorded with extracellular Ca2+ replaced by Sr2+ In addition, there was a small increase (approximately 10%) in the number of L1-STTNs showing immunoreactivity for phosphorylated extracellular signal-regulated kinases in mice after SNI compared with sham. Similarly, only a small percentage of L1-STTNs showed a lower action potential threshold after SNI. In conclusion, our results show that SNI induces presynaptic modulation at NRNav1.8 (consisting of both peptidergic and nonpeptidergic nociceptors) synapses on L1-STTNs forming the lateral spinothalamic tract.
Collapse
Affiliation(s)
- Wei-Chen Hung 洪瑋辰
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Neurobiology and Cognitive Science Centre, National Taiwan University, Taipei 10617, Taiwan
| | | | - Cheng-Tung Yen 嚴震東
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Neurobiology and Cognitive Science Centre, National Taiwan University, Taipei 10617, Taiwan
| | - Ming-Yuan Min 閔明源
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
- Neurobiology and Cognitive Science Centre, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
4
|
Martynyuk T, Ricard J, Bracchi-Ricard V, Price S, McGrath J, Dougherty K, Tom V, Bethea JR. Mitigating sTNF/TNFR1 activation on VGluT2+ spinal cord interneurons improves immune function after mid-thoracic spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602690. [PMID: 39026822 PMCID: PMC11257617 DOI: 10.1101/2024.07.09.602690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Spinal cord injury (SCI) is a devastating condition with 250,000 to 500,000 new cases globally each year. Respiratory infections, e.g., pneumonia and influenza are the leading cause of death after SCI. Unfortunately, there is a poor understanding of how altered neuro-immune communication impacts an individual's outcome to infection. In humans and rodents, SCI leads to maladaptive changes in the spinal-sympathetic reflex (SSR) circuit which is crucial to sympathetic function. The cause of the impaired immune function may be related to harmful neuroinflammation which is detrimental to homeostatic neuronal function, aberrant plasticity, and hyperexcitable circuits. Soluble tumor necrosis factor (sTNF) is a pro-inflammatory cytokine that is elevated in the CNS after SCI and remains elevated for several months after injury. By pharmacologically attenuating sTNF in the CNS after SCI we were able to demonstrate improved immune function. Furthermore, when we investigated the specific cellular population which may be involved in altered neuro-immune communication we reported that excessive TNFR1 activity on excitatory INs promotes immune dysfunction. Furthermore, this observation is NF-κB dependent in VGluT2+ INs. Our data is the first report of a target within the CNS, TNFR1, that contributes to SCI-induced immune dysfunction after T9-SCI and is a potential avenue for future therapeutics.
Collapse
Affiliation(s)
| | - Jerome Ricard
- Drexel University, Department of Biology, Philadelphia, PA
| | | | - Samuel Price
- Drexel University, Department of Biology, Philadelphia, PA
| | - Jenna McGrath
- Drexel University, Department of Neurobiology and Anatomy, Philadelphia, PA
| | - Kimberly Dougherty
- Drexel University, Department of Neurobiology and Anatomy, Philadelphia, PA
| | - Veronica Tom
- Drexel University, Department of Neurobiology and Anatomy, Philadelphia, PA
| | - John R. Bethea
- Drexel University, Department of Biology, Philadelphia, PA
- George Washington University, Department of Anatomy and Cell Biology, Washington D.C
| |
Collapse
|
5
|
Yoshinaga Y, Sato N. Reach-to-Grasp and tactile discrimination task: A new task for the study of sensory-motor learning. Behav Brain Res 2024; 466:115007. [PMID: 38648867 DOI: 10.1016/j.bbr.2024.115007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Although active touch in rodents arises from the forepaws as well as whiskers, most research on active touch only focuses on whiskers. This results in a paucity of tasks designed to assess the process of active touch with a forepaw. We develop a new experimental task, the Reach-to-Grasp and Tactile Discrimination task (RGTD task), to examine active touch with a forepaw in rodents, particularly changes in processes of active touch during motor skill learning. In the RGTD task, animals are required to (1) extend their forelimb to an object, (2) grasp the object, and (3) manipulate the grasped object with the forelimb. The animals must determine the direction of the manipulation based on active touch sensations arising during the period of the grasping. In experiment 1 of the present study, we showed that rats can learn the RGTD task. In experiment 2, we confirmed that the rats are capable of reversal learning of the RGTD task. The RGTD task shared most of the reaching movements involved with conventional forelimb reaching tasks. From the standpoint of a discrimination task, the RGTD task enables rigorous experimental control, for example by removing bias in the stimulus-response correspondence, and makes it possible to utilize diverse experimental procedures that have been difficult in prior tasks.
Collapse
Affiliation(s)
- Yudai Yoshinaga
- Department of Psychological Sciences, Kwansei Gakuin University, 1-1-155, Uegahara, Nishinomiya, Hyogo 662-8501, Japan; Research Fellow of Japan Society for the Promotion of Science, Japan
| | - Nobuya Sato
- Department of Psychological Sciences, Kwansei Gakuin University, 1-1-155, Uegahara, Nishinomiya, Hyogo 662-8501, Japan; Center for Applied Psychological Science (CAPS), Kwansei Gakuin University, 1-1-155, Uegahara, Nishinomiya, Hyogo, Japan.
| |
Collapse
|
6
|
Yoo J, Shin JC, Lim KB, Kim SH, Kim HS, Kim SH, Baek D, Jo S, Kim J, Baek A, Cho SR. Exposure to an enriched environment modulates the synaptic vesicle cycle in a mouse spinal cord injury model. Sci Rep 2024; 14:11946. [PMID: 38789574 PMCID: PMC11126684 DOI: 10.1038/s41598-024-62112-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Spinal cord injury (SCI) leads to motor and sensory impairment below the site of injury, thereby necessitating rehabilitation. An enriched environment (EE) increases social interaction and locomotor activity in a mouse model, similar to human rehabilitation. However, the impact of EE on presynaptic plasticity in gene expression levels remains unclear. Hence, this study aimed to investigate the therapeutic potential of EE in an SCI mouse model. Mice with spinal cord contusion were divided into two groups: those housed in standard cages (control) and those in EE conditions (EE). Each group was housed separately for either 2- or 8-weeks post-injury, after which RNA sequencing was performed and compared to a sham group (receiving only a dorsal laminectomy). The synaptic vesicle cycle (SVC) pathway and related genes showed significant downregulation after SCI at both time points. Subsequently, we investigated whether exposure to EE for 2- and 8-weeks post-SCI could modulate the SVC pathway and its related genes. Notably, exposure to EE for 8 weeks resulted in a marked reversal effect of SVC-related gene expression, along with stimulation of axon regeneration and mitigation of locomotor activity loss. Thus, prolonged exposure to EE increased presynaptic activity, fostering axon regeneration and functional improvement by modulating the SVC in the SCI mouse model. These findings suggest that EE exposure proves effective in inducing activity-dependent plasticity, offering a promising therapeutic approach akin to rehabilitation training in patients with SCI.
Collapse
Affiliation(s)
- Jeehyun Yoo
- Department of Rehabilitation Medicine, Ilsan Paik Hospital, Inje University, Gyeonggi-do, South Korea
- Department of Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Ji Cheol Shin
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Kil-Byung Lim
- Department of Rehabilitation Medicine, Ilsan Paik Hospital, Inje University, Gyeonggi-do, South Korea
| | - Se Hoon Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Seok Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, South Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Sung Hoon Kim
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Dawoon Baek
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea
| | - Seongmoon Jo
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Jinyoung Kim
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, South Korea
| | - Ahreum Baek
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.
- Department of Rehabilitation Medicine, Yonsei University Wonju College of Medicine, Wonju, South Korea.
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul, South Korea.
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea.
- Graduate Program of Biomedical Engineering, Yonsei University College of Medicine, Seoul, South Korea.
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
7
|
Karafoulidou E, Kesidou E, Theotokis P, Konstantinou C, Nella MK, Michailidou I, Touloumi O, Polyzoidou E, Salamotas I, Einstein O, Chatzisotiriou A, Boziki MK, Grigoriadis N. Systemic LPS Administration Stimulates the Activation of Non-Neuronal Cells in an Experimental Model of Spinal Muscular Atrophy. Cells 2024; 13:785. [PMID: 38727321 PMCID: PMC11083572 DOI: 10.3390/cells13090785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/27/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by deficiency of the survival motor neuron (SMN) protein. Although SMA is a genetic disease, environmental factors contribute to disease progression. Common pathogen components such as lipopolysaccharides (LPS) are considered significant contributors to inflammation and have been associated with muscle atrophy, which is considered a hallmark of SMA. In this study, we used the SMNΔ7 experimental mouse model of SMA to scrutinize the effect of systemic LPS administration, a strong pro-inflammatory stimulus, on disease outcome. Systemic LPS administration promoted a reduction in SMN expression levels in CNS, peripheral lymphoid organs, and skeletal muscles. Moreover, peripheral tissues were more vulnerable to LPS-induced damage compared to CNS tissues. Furthermore, systemic LPS administration resulted in a profound increase in microglia and astrocytes with reactive phenotypes in the CNS of SMNΔ7 mice. In conclusion, we hereby show for the first time that systemic LPS administration, although it may not precipitate alterations in terms of deficits of motor functions in a mouse model of SMA, it may, however, lead to a reduction in the SMN protein expression levels in the skeletal muscles and the CNS, thus promoting synapse damage and glial cells' reactive phenotype.
Collapse
Affiliation(s)
- Eleni Karafoulidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Chrystalla Konstantinou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Maria-Konstantina Nella
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Iliana Michailidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Olga Touloumi
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Eleni Polyzoidou
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Ilias Salamotas
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Ofira Einstein
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, Ariel 40700, Israel;
| | - Athanasios Chatzisotiriou
- Department of Physiology, Medical School, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - Marina-Kleopatra Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, 2nd Neurological University Department, AHEPA General Hospital of Thessaloniki, Faculty of Health Science, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece; (E.K.); (E.K.); (P.T.); (C.K.); (M.-K.N.); (I.M.); (O.T.); (E.P.); (I.S.)
| |
Collapse
|
8
|
Mangiamele LA, Dawn A, LeCure KM, Mantica GE, Racicot R, Fuxjager MJ, Preininger D. How new communication behaviors evolve: Androgens as modifiers of neuromotor structure and function in foot-flagging frogs. Horm Behav 2024; 161:105502. [PMID: 38382227 DOI: 10.1016/j.yhbeh.2024.105502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/08/2023] [Accepted: 02/05/2024] [Indexed: 02/23/2024]
Abstract
How diverse animal communication signals have arisen is a question that has fascinated many. Xenopus frogs have been a model system used for three decades to reveal insights into the neuroendocrine mechanisms and evolution of vocal diversity. Due to the ease of studying central nervous system control of the laryngeal muscles in vitro, Xenopus has helped us understand how variation in vocal communication signals between sexes and between species is produced at the molecular, cellular, and systems levels. Yet, it is becoming easier to make similar advances in non-model organisms. In this paper, we summarize our research on a group of frog species that have evolved a novel hind limb signal known as 'foot flagging.' We have previously shown that foot flagging is androgen dependent and that the evolution of foot flagging in multiple unrelated species is accompanied by the evolution of higher androgen hormone sensitivity in the leg muscles. Here, we present new preliminary data that compare patterns of androgen receptor expression and neuronal cell density in the lumbar spinal cord - the neuromotor system that controls the hind limb - between foot-flagging and non-foot-flagging frog species. We then relate our work to prior findings in Xenopus, highlighting which patterns of hormone sensitivity and neuroanatomical structure are shared between the neuromotor systems underlying Xenopus vocalizations and foot-flagging frogs' limb movement and which appear to be species-specific. Overall, we aim to illustrate the power of drawing inspiration from experiments in model organisms, in which the mechanistic details have been worked out, and then applying these ideas to a non-model species to reveal new details, further complexities, and fresh hypotheses.
Collapse
Affiliation(s)
- Lisa A Mangiamele
- Department of Biological Sciences, Smith College, Northampton, MA 01063, United States of America.
| | - AllexAndrya Dawn
- Department of Biological Sciences, Smith College, Northampton, MA 01063, United States of America
| | - Kerry M LeCure
- Department of Biological Sciences, Smith College, Northampton, MA 01063, United States of America
| | - Gina E Mantica
- Department of Biological Sciences, Smith College, Northampton, MA 01063, United States of America
| | - Riccardo Racicot
- Department of Biological Sciences, Smith College, Northampton, MA 01063, United States of America
| | - Matthew J Fuxjager
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI 02912, United States of America
| | - Doris Preininger
- Department of Evolutionary Biology, University of Vienna, Vienna, Austria; Vienna Zoo, Vienna, Austria
| |
Collapse
|
9
|
Taga M, Hong YNG, Charalambous CC, Raju S, Hayes L, Lin J, Zhang Y, Shao Y, Houston M, Zhang Y, Mazzoni P, Roh J, Schambra HM. Corticospinal and corticoreticulospinal projections benefit motor behaviors in chronic stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.04.588112. [PMID: 38645144 PMCID: PMC11030245 DOI: 10.1101/2024.04.04.588112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
After corticospinal tract (CST) stroke, several motor deficits in the upper extremity (UE) emerge, including diminished muscle strength, motor control, and muscle individuation. Both the ipsilesional CST and contralesional corticoreticulospinal tract (CReST) innervate the paretic UE and may have different innervation patterns for the proximal and distal UE segments. These patterns may underpin distinct pathway relationships to separable motor behaviors. In this cross-sectional study of 15 chronic stroke patients and 28 healthy subjects, we examined two key questions: (1) whether segmental motor behaviors differentially relate to ipsilesional CST and contralesional CReST projection strengths, and (2) whether motor behaviors segmentally differ in the paretic UE. We measured strength, motor control, and muscle individuation in a proximal (biceps, BIC) and distal muscle (first dorsal interosseous, FDI) of the paretic UE. We measured the projection strengths of the ipsilesional CST and contralesional CReST to these muscles using transcranial magnetic stimulation (TMS). Stroke subjects had abnormal motor control and muscle individuation despite strength comparable to healthy subjects. In stroke subjects, stronger ipsilesional CST projections were linked to superior motor control in both UE segments, whereas stronger contralesional CReST projections were linked to superior muscle strength and individuation in both UE segments. Notably, both pathways also shared associations with behaviors in the proximal segment. Motor control deficits were segmentally comparable, but muscle individuation was worse for distal motor performance. These results suggest that each pathway has specialized contributions to chronic motor behaviors but also work together, with varying levels of success in supporting chronic deficits. Key points summary Individuals with chronic stroke typically have deficits in strength, motor control, and muscle individuation in their paretic upper extremity (UE). It remains unclear how these altered behaviors relate to descending motor pathways and whether they differ by proximal and distal UE segment.In this study, we used transcranial magnetic stimulation (TMS) to examine projection strengths of the ipsilesional corticospinal tract (CST) and contralesional corticoreticulospinal tract (CReST) with respect to quantitated motor behaviors in chronic stroke.We found that stronger ipsilesional CST projections were associated with better motor control in both UE segments, whereas stronger contralesional CReST projections were associated with better strength and individuation in both UE segments. In addition, projections of both pathways shared associations with motor behaviors in the proximal UE segment.We also found that deficits in strength and motor control were comparable across UE segments, but muscle individuation was worse with controlled movement in the distal UE segment.These results suggest that the CST and CReST have specialized contributions to chronic motor behaviors and also work together, although with different degrees of efficacy.
Collapse
|
10
|
Dominguez-Bajo A, Clotman F. Potential Roles of Specific Subclasses of Premotor Interneurons in Spinal Cord Function Recovery after Traumatic Spinal Cord Injury in Adults. Cells 2024; 13:652. [PMID: 38667267 PMCID: PMC11048910 DOI: 10.3390/cells13080652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
The differential expression of transcription factors during embryonic development has been selected as the main feature to define the specific subclasses of spinal interneurons. However, recent studies based on single-cell RNA sequencing and transcriptomic experiments suggest that this approach might not be appropriate in the adult spinal cord, where interneurons show overlapping expression profiles, especially in the ventral region. This constitutes a major challenge for the identification and direct targeting of specific populations that could be involved in locomotor recovery after a traumatic spinal cord injury in adults. Current experimental therapies, including electrical stimulation, training, pharmacological treatments, or cell implantation, that have resulted in improvements in locomotor behavior rely on the modulation of the activity and connectivity of interneurons located in the surroundings of the lesion core for the formation of detour circuits. However, very few publications clarify the specific identity of these cells. In this work, we review the studies where premotor interneurons were able to create new intraspinal circuits after different kinds of traumatic spinal cord injury, highlighting the difficulties encountered by researchers, to classify these populations.
Collapse
Affiliation(s)
- Ana Dominguez-Bajo
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology (LIBST), Animal Molecular and Cellular Biology Group (AMCB), Place Croix du Sud 4–5, 1348 Louvain la Neuve, Belgium
| | - Frédéric Clotman
- Université catholique de Louvain, Louvain Institute of Biomolecular Science and Technology (LIBST), Animal Molecular and Cellular Biology Group (AMCB), Place Croix du Sud 4–5, 1348 Louvain la Neuve, Belgium
| |
Collapse
|
11
|
Lemieux M, Karimi N, Bretzner F. Functional plasticity of glutamatergic neurons of medullary reticular nuclei after spinal cord injury in mice. Nat Commun 2024; 15:1542. [PMID: 38378819 PMCID: PMC10879492 DOI: 10.1038/s41467-024-45300-4] [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: 07/26/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Spinal cord injury disrupts the descending command from the brain and causes a range of motor deficits. Here, we use optogenetic tools to investigate the functional plasticity of the glutamatergic reticulospinal drive of the medullary reticular formation after a lateral thoracic hemisection in female mice. Sites evoking stronger excitatory descending drive in intact conditions are the most impaired after injury, whereas those associated with a weaker drive are potentiated. After lesion, pro- and anti-locomotor activities (that is, initiation/acceleration versus stop/deceleration) are overall preserved. Activating the descending reticulospinal drive improves stepping ability on a flat surface of chronically impaired injured mice, and its priming enhances recovery of skilled locomotion on a horizontal ladder. This study highlights the resilience and capacity for reorganization of the glutamatergic reticulospinal command after injury, along with its suitability as a therapeutical target to promote functional recovery.
Collapse
Affiliation(s)
- Maxime Lemieux
- Centre de Recherche du CHU de Québec, CHUL-Neurosciences, 2705 Boul. Laurier, Québec, QC, G1V 4G2, Canada
| | - Narges Karimi
- Centre de Recherche du CHU de Québec, CHUL-Neurosciences, 2705 Boul. Laurier, Québec, QC, G1V 4G2, Canada
- Faculty of Medicine, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, G1V 4G2, Canada
| | - Frederic Bretzner
- Centre de Recherche du CHU de Québec, CHUL-Neurosciences, 2705 Boul. Laurier, Québec, QC, G1V 4G2, Canada.
- Faculty of Medicine, Department of Psychiatry and Neurosciences, Université Laval, Québec, QC, G1V 4G2, Canada.
| |
Collapse
|
12
|
Zholudeva LV, Fortino T, Agrawal A, Vila OF, Williams M, McDevitt T, Lane MA, Srivastava D. Human spinal interneurons repair the injured spinal cord through synaptic integration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.11.575264. [PMID: 38260390 PMCID: PMC10802598 DOI: 10.1101/2024.01.11.575264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Advances in cell therapy offer promise for some of the most devastating neural injuries, including spinal cord injury (SCI). Endogenous VSX2-expressing spinal V2a interneurons have been implicated as a key component in plasticity and therapeutically driven recovery post-SCI. While transplantation of generic V2a neurons may have therapeutic value, generation of human spinal V2a neurons with rostro-caudal specificity and assessment of their functional synaptic integration with the injured spinal cord has been elusive. Here, we efficiently differentiated optogenetically engineered cervical V2a spinal interneurons (SpINs) from human induced pluripotent stem cells and tested their capacity to form functional synapses with injured diaphragm motor networks in a clinically-relevant sub-acute model of cervical contusion injury. Neuroanatomical tracing and immunohistochemistry demonstrated transplant integration and synaptic connectivity with injured host tissue. Optogenetic activation of transplanted human V2a SpINs revealed functional synaptic connectivity to injured host circuits, culminating in improved diaphragm activity assessed by electromyography. Furthermore, optogenetic activation of host supraspinal pathways revealed functional innervation of transplanted cells by host neurons, which also led to enhanced diaphragm contraction indicative of a functional neuronal relay. Single cell analyses pre- and post-transplantation suggested the in vivo environment resulted in maturation of cervical SpINs that mediate the formation of neuronal relays, as well as differentiation of glial progenitors involved in repair of the damaged spinal cord. This study rigorously demonstrates feasibility of generating human cervical spinal V2a interneurons that develop functional host-transplant and transplant-host connectivity resulting in improved muscle activity post-SCI.
Collapse
|
13
|
Roman A, Huntemer-Silveira A, Waldron MA, Khalid Z, Blake J, Parr AM, Low WC. Cell Transplantation for Repair of the Spinal Cord and Prospects for Generating Region-Specific Exogenic Neuronal Cells. Cell Transplant 2024; 33:9636897241241998. [PMID: 38590295 PMCID: PMC11005494 DOI: 10.1177/09636897241241998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 03/05/2024] [Accepted: 03/11/2024] [Indexed: 04/10/2024] Open
Abstract
Spinal cord injury (SCI) is associated with currently irreversible consequences in several functional components of the central nervous system. Despite the severity of injury, there remains no approved treatment to restore function. However, with a growing number of preclinical studies and clinical trials, cell transplantation has gained significant potential as a treatment for SCI. Researchers have identified several cell types as potential candidates for transplantation. To optimize successful functional outcomes after transplantation, one key factor concerns generating neuronal cells with regional and subtype specificity, thus calling on the developmental transcriptome patterning of spinal cord cells. A potential source of spinal cord cells for transplantation is the generation of exogenic neuronal progenitor cells via the emerging technologies of gene editing and blastocyst complementation. This review highlights the use of cell transplantation to treat SCI in the context of relevant developmental gene expression patterns useful for producing regionally specific exogenic spinal cells via in vitro differentiation and blastocyst complementation.
Collapse
Affiliation(s)
- Alex Roman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Anne Huntemer-Silveira
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Madison A. Waldron
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Zainab Khalid
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey Blake
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Ann M. Parr
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| | - Walter C. Low
- Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Department of Neurosurgery, Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
14
|
Braaß H, Feldheim J, Chu Y, Tinnermann A, Finsterbusch J, Büchel C, Schulz R, Gerloff C. Association between activity in the ventral premotor cortex and spinal cord activation during force generation-A combined cortico-spinal fMRI study. Hum Brain Mapp 2023; 44:6471-6483. [PMID: 37873743 PMCID: PMC10681651 DOI: 10.1002/hbm.26523] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/04/2023] [Accepted: 10/08/2023] [Indexed: 10/25/2023] Open
Abstract
Force generation is a crucial element of dexterity and a highly relevant skill of the human motor system. How cerebral and spinal components interact and how spinal activation is associated with the activity in the cerebral primary motor and premotor areas is poorly understood. Here, we conducted combined cortico-spinal functional magnetic resonance imaging during a simple visually guided isometric force generation task in 20 healthy young subjects. Activation was localized in the right cervical spinal cord and left primary motor and premotor areas. The main finding is that spinal activation was negatively correlated with ventral premotor cortex activation. Spinal activation was furthermore significantly correlated with primary motor cortex activation, while increasing target forces led to an increase in the amount of activation. These data indicate that human premotor areas such as the ventral premotor cortex might be functionally connected to the lower cervical spinal cord contributing to distal upper limb functions, a finding that extends our understanding of human motor function beyond the animal literature.
Collapse
Affiliation(s)
- Hanna Braaß
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jan Feldheim
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Ying Chu
- Institute of Systems NeuroscienceUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Alexandra Tinnermann
- Institute of Systems NeuroscienceUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Jürgen Finsterbusch
- Institute of Systems NeuroscienceUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christian Büchel
- Institute of Systems NeuroscienceUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Robert Schulz
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Christian Gerloff
- Department of NeurologyUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
15
|
Kuehn N, Schwarz A, Beretta CA, Schwarte Y, Schmitt F, Motsch M, Weidner N, Puttagunta R. Intermediate gray matter interneurons in the lumbar spinal cord play a critical and necessary role in coordinated locomotion. PLoS One 2023; 18:e0291740. [PMID: 37906544 PMCID: PMC10617729 DOI: 10.1371/journal.pone.0291740] [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: 05/28/2023] [Accepted: 09/05/2023] [Indexed: 11/02/2023] Open
Abstract
Locomotion is a complex task involving excitatory and inhibitory circuitry in spinal gray matter. While genetic knockouts examine the function of individual spinal interneuron (SpIN) subtypes, the phenotype of combined SpIN loss remains to be explored. We modified a kainic acid lesion to damage intermediate gray matter (laminae V-VIII) in the lumbar spinal enlargement (spinal L2-L4) in female rats. A thorough, tailored behavioral evaluation revealed deficits in gross hindlimb function, skilled walking, coordination, balance and gait two weeks post-injury. Using a Random Forest algorithm, we combined these behavioral assessments into a highly predictive binary classification system that strongly correlated with structural deficits in the rostro-caudal axis. Machine-learning quantification confirmed interneuronal damage to laminae V-VIII in spinal L2-L4 correlates with hindlimb dysfunction. White matter alterations and lower motoneuron loss were not observed with this KA lesion. Animals did not regain lost sensorimotor function three months after injury, indicating that natural recovery mechanisms of the spinal cord cannot compensate for loss of laminae V-VIII neurons. As gray matter damage accounts for neurological/walking dysfunction in instances of spinal cord injury affecting the cervical or lumbar enlargement, this research lays the groundwork for new neuroregenerative therapies to replace these lost neuronal pools vital to sensorimotor function.
Collapse
Affiliation(s)
- Naëmi Kuehn
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Schwarz
- Laboratory for Experimental Neurorehabilitation, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Carlo Antonio Beretta
- Department of Functional Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Yvonne Schwarte
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Francesca Schmitt
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie Motsch
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Radhika Puttagunta
- Laboratory for Experimental Neuroregeneration, Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
16
|
Lin S, Hari K, Black S, Khatmi A, Fouad K, Gorassini MA, Li Y, Lucas-Osma AM, Fenrich KK, Bennett DJ. Locomotor-related propriospinal V3 neurons produce primary afferent depolarization and modulate sensory transmission to motoneurons. J Neurophysiol 2023; 130:799-823. [PMID: 37609680 PMCID: PMC10650670 DOI: 10.1152/jn.00482.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/24/2023] Open
Abstract
When a muscle is stretched, sensory feedback not only causes reflexes but also leads to a depolarization of sensory afferents throughout the spinal cord (primary afferent depolarization, PAD), readying the whole limb for further disturbances. This sensory-evoked PAD is thought to be mediated by a trisynaptic circuit, where sensory input activates first-order excitatory neurons that activate GABAergic neurons that in turn activate GABAA receptors on afferents to cause PAD, though the identity of these first-order neurons is unclear. Here, we show that these first-order neurons include propriospinal V3 neurons, as they receive extensive sensory input and in turn innervate GABAergic neurons that cause PAD, because optogenetic activation or inhibition of V3 neurons in mice mimics or inhibits sensory-evoked PAD, respectively. Furthermore, persistent inward sodium currents intrinsic to V3 neurons prolong their activity, explaining the prolonged duration of PAD. Also, local optogenetic activation of V3 neurons at one segment causes PAD in other segments, due to the long propriospinal tracts of these neurons, helping to explain the radiating nature of PAD. This in turn facilitates monosynaptic reflex transmission to motoneurons across the spinal cord. In addition, V3 neurons directly innervate proprioceptive afferents (including Ia), causing a glutamate receptor-mediated PAD (glutamate PAD). Finally, increasing the spinal cord excitability with either GABAA receptor blockers or chronic spinal cord injury causes an increase in the glutamate PAD. Overall, we show the V3 neuron has a prominent role in modulating sensory transmission, in addition to its previously described role in locomotion.NEW & NOTEWORTHY Locomotor-related propriospinal neurons depolarize sensory axons throughout the spinal cord by either direct glutamatergic axoaxonic contacts or indirect innervation of GABAergic neurons that themselves form axoaxonic contacts on sensory axons. This depolarization (PAD) increases sensory transmission to motoneurons throughout the spinal cord, readying the sensorimotor system for external disturbances. The glutamate-mediated PAD is particularly adaptable, increasing with either an acute block of GABA receptors or chronic spinal cord injury, suggesting a role in motor recovery.
Collapse
Affiliation(s)
- Shihao Lin
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Krishnapriya Hari
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sophie Black
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Aysan Khatmi
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Monica A Gorassini
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Yaqing Li
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Ana M Lucas-Osma
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Keith K Fenrich
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - David J Bennett
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
- Faculty of Rehabilitation Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
17
|
Huntemer-Silveira A, Malone D, Frie A, Walsh P, Parr AM. Accelerated differentiation of human induced pluripotent stem cells into regionally specific dorsal and ventral spinal neural progenitor cells for application in spinal cord therapeutics. Front Neurosci 2023; 17:1251906. [PMID: 37781243 PMCID: PMC10540309 DOI: 10.3389/fnins.2023.1251906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023] Open
Abstract
Spinal cord injury can attenuate both motor and sensory function with minimal potential for full recovery. Research utilizing human induced pluripotent stem cell (hiPSC) -derived spinal cell types for in vivo remodeling and neuromodulation after spinal cord injury has grown substantially in recent years. However, the majority of protocols for the differentiation of spinal neurons are lengthy, lack the appropriate dorsoventral or rostrocaudal specification, and are not typically replicated in more than one cell line. Furthermore, most researchers currently utilize hiPSC-derived motor neurons for cell transplantation after injury, with very little exploration of spinal sensory neuron transplantation. The lack of studies that utilize sensory populations may be due in part to the relative scarcity of dorsal horn differentiation protocols. Building upon our previously published work that demonstrated the rapid establishment of a primitive ectoderm population from hiPSCs, we describe here the production of a diverse population of both ventral spinal and dorsal horn progenitor cells. Our work creates a novel system allowing dorsal and ventral spinal neurons to be differentiated from the same intermediate ectoderm population, making it possible to construct the dorsal and ventral domains of the spinal cord while decreasing variability. This technology can be used in tandem with biomaterials and pharmacology to improve cell transplantation for spinal cord injury, increasing the potential for neuroregeneration.
Collapse
Affiliation(s)
| | - Dane Malone
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Anna Frie
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Patrick Walsh
- Anatomic Incorporated, Minneapolis, MN, United States
| | - Ann M. Parr
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
18
|
Cheng J, Guan NN. A fresh look at propriospinal interneurons plasticity and intraspinal circuits remodeling after spinal cord injury. IBRO Neurosci Rep 2023. [DOI: 10.1016/j.ibneur.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023] Open
|
19
|
Cao B, Scherrer G, Chen L. Spinal cord retinoic acid receptor signaling gates mechanical hypersensitivity in neuropathic pain. Neuron 2022; 110:4108-4124.e6. [PMID: 36223767 PMCID: PMC9789181 DOI: 10.1016/j.neuron.2022.09.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 08/27/2022] [Accepted: 09/22/2022] [Indexed: 02/08/2023]
Abstract
Central sensitization caused by spinal disinhibition is a key mechanism of mechanical allodynia in neuropathic pain. However, the molecular mechanisms underlying spinal disinhibition after nerve injury remain unclear. Here, we show in mice that spared nerve injury (SNI), which induces mechanical hypersensitivity and neuropathic pain, triggers homeostatic reduction of inhibitory outputs from dorsal horn parvalbumin-positive (PV+) interneurons onto both primary afferent terminals and excitatory interneurons. The reduction in inhibitory outputs drives hyperactivation of the spinal cord nociceptive pathway, causing mechanical hypersensitivity. We identified the retinoic acid receptor RARα, a central regulator of homeostatic plasticity, as the key molecular mediator for this synaptic disinhibition. Deletion of RARα in spinal PV+ neurons or application of an RARα antagonist in the spinal cord prevented the development of SNI-induced mechanical hypersensitivity. Our results identify RARα as a crucial molecular effector for neuropathic pain and a potential target for its treatment.
Collapse
Affiliation(s)
- Bing Cao
- Department of Neurosurgery, Wu Tsai Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Gregory Scherrer
- Department of Cell Biology and Physiology, UNC Neuroscience Center, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lu Chen
- Department of Neurosurgery, Wu Tsai Neuroscience Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
20
|
Anderson MA, Squair JW, Gautier M, Hutson TH, Kathe C, Barraud Q, Bloch J, Courtine G. Natural and targeted circuit reorganization after spinal cord injury. Nat Neurosci 2022; 25:1584-1596. [PMID: 36396975 DOI: 10.1038/s41593-022-01196-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/05/2022] [Indexed: 11/18/2022]
Abstract
A spinal cord injury disrupts communication between the brain and the circuits in the spinal cord that regulate neurological functions. The consequences are permanent paralysis, loss of sensation and debilitating dysautonomia. However, the majority of circuits located above and below the injury remain anatomically intact, and these circuits can reorganize naturally to improve function. In addition, various neuromodulation therapies have tapped into these processes to further augment recovery. Emerging research is illuminating the requirements to reconstitute damaged circuits. Here, we summarize these natural and targeted reorganizations of circuits after a spinal cord injury. We also advocate for new concepts of reorganizing circuits informed by multi-omic single-cell atlases of recovery from injury. These atlases will uncover the molecular logic that governs the selection of 'recovery-organizing' neuronal subpopulations, and are poised to herald a new era in spinal cord medicine.
Collapse
Affiliation(s)
- Mark A Anderson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Jordan W Squair
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Matthieu Gautier
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Thomas H Hutson
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.,Wyss Center for Bio and Neuroengineering, Geneva, Switzerland
| | - Claudia Kathe
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Quentin Barraud
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Jocelyne Bloch
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland
| | - Grégoire Courtine
- NeuroX Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland. .,Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland. .,Defitech Center for Interventional Neurotherapies (NeuroRestore), EPFL/CHUV/UNIL, Lausanne, Switzerland.
| |
Collapse
|
21
|
Kathe C, Skinnider MA, Hutson TH, Regazzi N, Gautier M, Demesmaeker R, Komi S, Ceto S, James ND, Cho N, Baud L, Galan K, Matson KJE, Rowald A, Kim K, Wang R, Minassian K, Prior JO, Asboth L, Barraud Q, Lacour SP, Levine AJ, Wagner F, Bloch J, Squair JW, Courtine G. The neurons that restore walking after paralysis. Nature 2022; 611:540-547. [DOI: 10.1038/s41586-022-05385-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 09/23/2022] [Indexed: 11/10/2022]
Abstract
AbstractA spinal cord injury interrupts pathways from the brain and brainstem that project to the lumbar spinal cord, leading to paralysis. Here we show that spatiotemporal epidural electrical stimulation (EES) of the lumbar spinal cord1–3 applied during neurorehabilitation4,5 (EESREHAB) restored walking in nine individuals with chronic spinal cord injury. This recovery involved a reduction in neuronal activity in the lumbar spinal cord of humans during walking. We hypothesized that this unexpected reduction reflects activity-dependent selection of specific neuronal subpopulations that become essential for a patient to walk after spinal cord injury. To identify these putative neurons, we modelled the technological and therapeutic features underlying EESREHAB in mice. We applied single-nucleus RNA sequencing6–9 and spatial transcriptomics10,11 to the spinal cords of these mice to chart a spatially resolved molecular atlas of recovery from paralysis. We then employed cell type12,13 and spatial prioritization to identify the neurons involved in the recovery of walking. A single population of excitatory interneurons nested within intermediate laminae emerged. Although these neurons are not required for walking before spinal cord injury, we demonstrate that they are essential for the recovery of walking with EES following spinal cord injury. Augmenting the activity of these neurons phenocopied the recovery of walking enabled by EESREHAB, whereas ablating them prevented the recovery of walking that occurs spontaneously after moderate spinal cord injury. We thus identified a recovery-organizing neuronal subpopulation that is necessary and sufficient to regain walking after paralysis. Moreover, our methodology establishes a framework for using molecular cartography to identify the neurons that produce complex behaviours.
Collapse
|
22
|
Hall A, Fortino T, Spruance V, Niceforo A, Harrop JS, Phelps PE, Priest CA, Zholudeva LV, Lane MA. Cell transplantation to repair the injured spinal cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 166:79-158. [PMID: 36424097 PMCID: PMC10008620 DOI: 10.1016/bs.irn.2022.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Adam Hall
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Tara Fortino
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - Victoria Spruance
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Division of Kidney, Urologic, & Hematologic Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Alessia Niceforo
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States
| | - James S Harrop
- Department of Neurological and Orthopedic Surgery, Thomas Jefferson University, Philadelphia, PA, United States
| | - Patricia E Phelps
- Department of Integrative Biology & Physiology, UCLA, Los Angeles, CA, United States
| | | | - Lyandysha V Zholudeva
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States; Gladstone Institutes, San Francisco, CA, United States
| | - Michael A Lane
- Drexel University, Philadelphia, PA, United States; Marion Murray Spinal Cord Research Center, Drexel University, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Locke KC, Randelman ML, Hoh DJ, Zholudeva LV, Lane MA. Respiratory plasticity following spinal cord injury: perspectives from mouse to man. Neural Regen Res 2022; 17:2141-2148. [PMID: 35259820 PMCID: PMC9083159 DOI: 10.4103/1673-5374.335839] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/18/2021] [Accepted: 10/20/2021] [Indexed: 12/03/2022] Open
Abstract
The study of respiratory plasticity in animal models spans decades. At the bench, researchers use an array of techniques aimed at harnessing the power of plasticity within the central nervous system to restore respiration following spinal cord injury. This field of research is highly clinically relevant. People living with cervical spinal cord injury at or above the level of the phrenic motoneuron pool at spinal levels C3-C5 typically have significant impairments in breathing which may require assisted ventilation. Those who are ventilator dependent are at an increased risk of ventilator-associated co-morbidities and have a drastically reduced life expectancy. Pre-clinical research examining respiratory plasticity in animal models has laid the groundwork for clinical trials. Despite how widely researched this injury is in animal models, relatively few treatments have broken through the preclinical barrier. The three goals of this present review are to define plasticity as it pertains to respiratory function post-spinal cord injury, discuss plasticity models of spinal cord injury used in research, and explore the shift from preclinical to clinical research. By investigating current targets of respiratory plasticity research, we hope to illuminate preclinical work that can influence future clinical investigations and the advancement of treatments for spinal cord injury.
Collapse
Affiliation(s)
- Katherine C. Locke
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA, USA
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| | - Margo L. Randelman
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA, USA
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| | - Daniel J. Hoh
- Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Lyandysha V. Zholudeva
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
- Cardiovascular Disease, Gladstone Institutes, San Francisco, CA, USA
| | - Michael A. Lane
- Department of Neurobiology & Anatomy, Drexel University, Philadelphia, PA, USA
- Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| |
Collapse
|
24
|
Eisdorfer JT, Sobotka-Briner H, Schramfield S, Moukarzel G, Chen J, Campion TJ, Smit R, Rauscher BC, Lemay MA, Smith GM, Spence AJ. Chemogenetic modulation of sensory afferents induces locomotor changes and plasticity after spinal cord injury. Front Mol Neurosci 2022; 15:872634. [PMID: 36090254 PMCID: PMC9461563 DOI: 10.3389/fnmol.2022.872634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 07/26/2022] [Indexed: 12/12/2022] Open
Abstract
Neuromodulatory therapies for spinal cord injury (SCI) such as electrical epidural stimulation (EES) are increasingly effective at improving patient outcomes. These improvements are thought to be due, at least in part, to plasticity in neuronal circuits. Precisely which circuits are influenced and which afferent classes are most effective in stimulating change remain important open questions. Genetic tools, such as Designer Receptors Exclusively Activated by Designer Drugs (DREADDs), support targeted and reversible neuromodulation as well as histological characterization of manipulated neurons. We therefore transduced and activated lumbar large diameter peripheral afferents with excitatory (hM3Dq) DREADDs, in a manner analogous to EES, in a rat hemisection model, to begin to trace plasticity and observe concomitant locomotor changes. Chronic DREADDs activation, coupled with thrice weekly treadmill training, was observed to increase afferent fluorescent labeling within motor pools and Clarke's column when compared to control animals. This plasticity may underlie kinematic differences that we observed across stages of recovery, including an increased and less variable hindquarters height in DREADDs animals, shorter step durations, a more flexed ankle joint early in recovery, a less variable ankle joint angle in swing phase, but a more variable hip joint angle. Withdrawal of DREADDs agonist, clozapine-N-oxide (CNO) left these kinematic differences largely unaffected; suggesting that DREADDs activation is not necessary for them later in recovery. However, we observed an intermittent “buckling” phenomenon in DREADDs animals without CNO activation, that did not occur with CNO re-administration. Future studies could use more refined genetic targeted of specific afferent classes, and utilize muscle recordings to find where afferent modulation is most influential in altering motor output.
Collapse
Affiliation(s)
- Jaclyn T. Eisdorfer
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, United States
| | - Hannah Sobotka-Briner
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - Susan Schramfield
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - George Moukarzel
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - Jie Chen
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Thomas J. Campion
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Rupert Smit
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Bradley C. Rauscher
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - Michel A. Lemay
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
| | - George M. Smith
- Department of Neuroscience, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Andrew J. Spence
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, United States
- *Correspondence: Andrew J. Spence
| |
Collapse
|
25
|
Chen M, Chen Z, Xiao X, Zhou L, Fu R, Jiang X, Pang M, Xia J. Corticospinal circuit neuroplasticity may involve silent synapses: Implications for functional recovery facilitated by neuromodulation after spinal cord injury. IBRO Neurosci Rep 2022; 14:185-194. [PMID: 36824667 PMCID: PMC9941655 DOI: 10.1016/j.ibneur.2022.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022] Open
Abstract
Spinal cord injury (SCI) leads to devastating physical consequences, such as severe sensorimotor dysfunction even lifetime disability, by damaging the corticospinal system. The conventional opinion that SCI is intractable due to the poor regeneration of neurons in the adult central nervous system (CNS) needs to be revisited as the CNS is capable of considerable plasticity, which underlie recovery from neural injury. Substantial spontaneous neuroplasticity has been demonstrated in the corticospinal motor circuitry following SCI. Some of these plastic changes appear to be beneficial while others are detrimental toward locomotor function recovery after SCI. The beneficial corticospinal plasticity in the spared corticospinal circuits can be harnessed therapeutically by multiple contemporary neuromodulatory approaches, especially the electrical stimulation-based modalities, in an activity-dependent manner to improve functional outcomes in post-SCI rehabilitation. Silent synapse generation and unsilencing contribute to profound neuroplasticity that is implicated in a variety of neurological disorders, thus they may be involved in the corticospinal motor circuit neuroplasticity following SCI. Exploring the underlying mechanisms of silent synapse-mediated neuroplasticity in the corticospinal motor circuitry that may be exploited by neuromodulation will inform a novel direction for optimizing therapeutic repair strategies and rehabilitative interventions in SCI patients.
Collapse
Key Words
- AMPARs, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors
- BDNF, brain-derived neurotrophic factor
- BMIs, brain-machine interfaces
- CPG, central pattern generator
- CST, corticospinal tract
- Corticospinal motor circuitry
- DBS, deep brain stimulation
- ESS, epidural spinal stimulation
- MEPs, motor-evoked potentials
- NHPs, non-human primates
- NMDARs, N-methyl-d-aspartate receptors
- Neuromodulation
- Neuroplasticity
- PSNs, propriospinal neurons
- Rehabilitation
- SCI, spinal cord injury
- STDP, spike timing-dependent plasticity
- Silent synapses
- Spinal cord injury
- TBS, theta burst stimulation
- TMS, transcranial magnetic stimulation
- TrkB, tropomyosin-related kinase B
- cTBS, continuous TBS
- iTBS, intermittent TBS
- mTOR, mammalian target of rapamycin
- rTMS, repetitive TMS
- tDCS, transcranial direct current stimulation
- tcSCS, transcutaneous spinal cord stimulation
Collapse
Affiliation(s)
- Mingcong Chen
- Department of Orthopedics and Traumatology, Shenzhen University General Hospital, Shenzhen, Guangdong 518055, China
| | - Zuxin Chen
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS); Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong 518055, China
| | - Xiao Xiao
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education; Behavioral and Cognitive Neuroscience Center, Institute of Science and Technology for Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200433, China
| | - Libing Zhou
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Jinan University)-Ministry of Education, Guangzhou, Guangdong 510632, China
| | - Rao Fu
- Department of Anatomy, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong 518100, China
| | - Xian Jiang
- Institute of Neurological and Psychiatric Disorder, Shenzhen Bay laboratory, Shenzhen, Guangdong 518000, China
| | - Mao Pang
- Department of Spine Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, Guangzhou, Guangdong 510630, China
| | - Jianxun Xia
- Department of Basic Medical Sciences, Yunkang School of Medicine and Health, Nanfang College, Guangzhou, Guangdong 510970, China,Corresponding author.
| |
Collapse
|
26
|
Wang Z, Romanski A, Mehra V, Wang Y, Brannigan M, Campbell BC, Petsko GA, Tsoulfas P, Blackmore MG. Brain-wide analysis of the supraspinal connectome reveals anatomical correlates to functional recovery after spinal injury. eLife 2022; 11:76254. [PMID: 35838234 PMCID: PMC9345604 DOI: 10.7554/elife.76254] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 07/12/2022] [Indexed: 11/15/2022] Open
Abstract
The supraspinal connectome is essential for normal behavior and homeostasis and consists of numerous sensory, motor, and autonomic projections from brain to spinal cord. Study of supraspinal control and its restoration after damage has focused mostly on a handful of major populations that carry motor commands, with only limited consideration of dozens more that provide autonomic or crucial motor modulation. Here, we assemble an experimental workflow to rapidly profile the entire supraspinal mesoconnectome in adult mice and disseminate the output in a web-based resource. Optimized viral labeling, 3D imaging, and registration to a mouse digital neuroanatomical atlas assigned tens of thousands of supraspinal neurons to 69 identified regions. We demonstrate the ability of this approach to clarify essential points of topographic mapping between spinal levels, measure population-specific sensitivity to spinal injury, and test the relationships between region-specific neuronal sparing and variability in functional recovery. This work will spur progress by broadening understanding of essential but understudied supraspinal populations.
Collapse
Affiliation(s)
- Zimei Wang
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Adam Romanski
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Vatsal Mehra
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Yunfang Wang
- Department of Neurological Surgery, University of Miami, Miami, United States
| | - Matthew Brannigan
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| | - Benjamin C Campbell
- Helen and Robert Appel Alzheimer's Disease Research Institute, Cornell University, New York, United States
| | - Gregory A Petsko
- Helen and Robert Appel Alzheimer's Disease Research Institute, Cornell University, New York, United States
| | - Pantelis Tsoulfas
- Department of Neurological Surgery, University of Miami, Miami, United States
| | - Murray G Blackmore
- Department of Biomedical Sciences, Marquette University, Milwaukee, United States
| |
Collapse
|
27
|
Noble BT, Brennan FH, Wang Y, Guan Z, Mo X, Schwab JM, Popovich PG. Thoracic VGluT2 + Spinal Interneurons Regulate Structural and Functional Plasticity of Sympathetic Networks after High-Level Spinal Cord Injury. J Neurosci 2022; 42:3659-3675. [PMID: 35304427 PMCID: PMC9053847 DOI: 10.1523/jneurosci.2134-21.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 11/21/2022] Open
Abstract
Traumatic spinal cord injury (SCI) above the major spinal sympathetic outflow (T6 level) disinhibits sympathetic neurons from supraspinal control, causing systems-wide "dysautonomia." We recently showed that remarkable structural remodeling and plasticity occurs within spinal sympathetic circuitry, creating abnormal sympathetic reflexes that exacerbate dysautonomia over time. As an example, thoracic VGluT2+ spinal interneurons (SpINs) become structurally and functionally integrated with neurons that comprise the spinal-splenic sympathetic network and immunological dysfunction becomes progressively worse after SCI. To test whether the onset and progression of SCI-induced sympathetic plasticity is neuron activity dependent, we selectively inhibited (or excited) thoracic VGluT2+ interneurons using chemogenetics. New data show that silencing VGluT2+ interneurons in female and male mice with a T3 SCI, using hM4Di designer receptors exclusively activated by designer drugs (Gi DREADDs), blocks structural plasticity and the development of dysautonomia. Specifically, silencing VGluT2+ interneurons prevents the structural remodeling of spinal sympathetic networks that project to lymphoid and endocrine organs, reduces the frequency of spontaneous autonomic dysreflexia (AD), and reduces the severity of experimentally induced AD. Features of SCI-induced structural plasticity can be recapitulated in the intact spinal cord by activating excitatory hM3Dq-DREADDs in VGluT2+ interneurons. Collectively, these data implicate VGluT2+ excitatory SpINs in the onset and propagation of SCI-induced structural plasticity and dysautonomia, and reveal the potential for neuromodulation to block or reduce dysautonomia after severe high-level SCI.SIGNIFICANCE STATEMENT In response to stress or dangerous stimuli, autonomic spinal neurons coordinate a "fight or flight" response marked by increased cardiac output and release of stress hormones. After a spinal cord injury (SCI), normally harmless stimuli like bladder filling can result in a "false" fight or flight response, causing pathological changes throughout the body. We show that progressive hypertension and immune suppression develop after SCI because thoracic excitatory VGluT2+ spinal interneurons (SpINs) provoke structural remodeling in autonomic networks within below-lesion spinal levels. These pathological changes can be prevented in SCI mice or phenocopied in uninjured mice using chemogenetics to selectively manipulate activity in VGluT2+ SpINs. Targeted neuromodulation of SpINs could prevent structural plasticity and subsequent autonomic dysfunction in people with SCI.
Collapse
Affiliation(s)
- Benjamin T Noble
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Faith H Brennan
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Yan Wang
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Zhen Guan
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Xiaokui Mo
- Center for Biostatistics, The Ohio State University, Columbus, Ohio 43210
| | - Jan M Schwab
- Department of Neurology, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| | - Phillip G Popovich
- Department of Neuroscience, Center for Brain and Spinal Cord Repair, Belford Center for Spinal Cord Injury, Wexner Medical Center, The Ohio State University, Columbus, Ohio 43210
| |
Collapse
|
28
|
Diversified physiological sensory input connectivity questions the existence of distinct classes of spinal interneurons. iScience 2022; 25:104083. [PMID: 35372805 PMCID: PMC8971951 DOI: 10.1016/j.isci.2022.104083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 12/21/2022] Open
Abstract
The spinal cord is engaged in all forms of motor performance but its functions are far from understood. Because network connectivity defines function, we explored the connectivity of muscular, tendon, and tactile sensory inputs among a wide population of spinal interneurons in the lower cervical segments. Using low noise intracellular whole cell recordings in the decerebrated, non-anesthetized cat in vivo, we could define mono-, di-, and trisynaptic inputs as well as the weights of each input. Whereas each neuron had a highly specific input, and each indirect input could moreover be explained by inputs in other recorded neurons, we unexpectedly also found the input connectivity of the spinal interneuron population to form a continuum. Our data hence contrasts with the currently widespread notion of distinct classes of interneurons. We argue that this suggested diversified physiological connectivity, which likely requires a major component of circuitry learning, implies a more flexible functionality.
Collapse
|
29
|
Xu J, Huang LJ, Fang Z, Luo HM, Chen YQ, Li YJ, Gong CZ, Chen H. Spinal dI4 Interneuron Differentiation From Human Pluripotent Stem Cells. Front Mol Neurosci 2022; 15:845875. [PMID: 35465095 PMCID: PMC9026311 DOI: 10.3389/fnmol.2022.845875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/10/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal interneurons (INs) form intricate local networks in the spinal cord and regulate not only the ascending and descending nerve transduction but also the central pattern generator function. They are therefore potential therapeutic targets in spinal cord injury and diseases. In this study, we devised a reproducible protocol to differentiate human pluripotent stem cells (hPSCs) from enriched spinal dI4 inhibitory GABAergic INs. The protocol is designed based on developmental principles and optimized by using small molecules to maximize its reproducibility. The protocol comprises induction of neuroepithelia, patterning of neuroepithelia to dorsal spinal progenitors, expansion of the progenitors in suspension, and finally differentiation into mature neurons. In particular, we employed both morphogen activators and inhibitors to restrict or “squeeze” the progenitor fate during the stage of neural patterning. We use retinoic acid (RA) which ventralizes cells up to the mid-dorsal region, with cyclopamine (CYC), an SHH inhibitor, to antagonize the ventralization effect of RA, yielding highly enriched dI4 progenitors (90% Ptf1a+, 90.7% Ascl1+). The ability to generate enriched spinal dI4 GABAergicINs will likely facilitate the study of human spinal IN development and regenerative therapies for traumatic injuries and diseases of the spinal cord.
Collapse
Affiliation(s)
- Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang-Jiang Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengyu Fang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Mei Luo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun-Qiang Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Jie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chen-Zi Gong
- Department of Neurological Rehabilitation, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Hong Chen
| |
Collapse
|
30
|
Selective Loss of MATR3 in Spinal Interneurons, Upper Motor Neurons and Hippocampal CA1 Neurons in a MATR3 S85C Knock-In Mouse Model of Amyotrophic Lateral Sclerosis. BIOLOGY 2022; 11:biology11020298. [PMID: 35205163 PMCID: PMC8869279 DOI: 10.3390/biology11020298] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/28/2022] [Accepted: 02/08/2022] [Indexed: 11/17/2022]
Abstract
Simple Summary Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting the motor neurons in the brain and spinal cord. Mutations in the gene Matr3 have been linked to ALS, including the autosomal dominant missense mutation S85C. We previously created a mouse model containing the S85C mutation in the Matr3 gene to understand how it causes ALS. The S85C mice exhibited MATR3 staining loss in selective populations of degenerating neurons, such as Purkinje cells in the cerebellum and α-motor neurons in the lumbar spinal cord. However, studies have shown that neurons other than motor neurons may be involved in contributing to ALS; therefore, we investigated additional neuronal cell types in the spinal cord and brain. Here, we found that MATR3 staining is selectively reduced in interneurons and α-motor neurons of the cervical and thoracic regions of the spinal cord, as well as in subsets of upper motor neurons and hippocampal neurons. These neurons did not exhibit cell body loss; however, how the MATR3 loss affects neuronal function remains to be determined. Overall, these findings demonstrate that the MATR3 S85C mutation affects other neuronal types of the brain and spinal cord in addition to motor neurons, suggesting that these additional neuronal types are involved in ALS pathogenesis. Abstract The neuropathological hallmark of amyotrophic lateral sclerosis (ALS) is motor neuron degeneration in the spinal cord and cortex. Accumulating studies report that other neurons in the central nervous system (CNS) are also affected in ALS. Mutations in Matr3, which encodes a nuclear matrix protein involved in RNA splicing, have been linked to ALS. Previously, we generated a MATR3 S85C knock-in (KI) mouse model that recapitulates early-stage features of ALS. We reported that MATR3 S85C KI mice exhibit defects in lumbar spinal cord motor neurons and in cerebellar Purkinje cells, which are associated with reduced MATR3 immunoreactivity. Here, we show that neurons in various other regions of the CNS are affected in MATR3 S85C KI mice. Using histological analyses, we found selective loss of MATR3 staining in α-motor neurons, but not γ-motor neurons in the cervical and thoracic spinal cord. Loss of MATR3 was also found in parvalbumin-positive interneurons in the cervical, thoracic and lumbar spinal cord. In addition, we found the loss of MATR3 in subsets of upper motor neurons and hippocampal CA1 neurons. Collectively, our findings suggest that these additional neuronal types may contribute to the disease process in MATR3 S85C KI mice.
Collapse
|
31
|
Fortino TA, Randelman ML, Hall AA, Singh J, Bloom DC, Engel E, Hoh DJ, Hou S, Zholudeva LV, Lane MA. Transneuronal tracing to map connectivity in injured and transplanted spinal networks. Exp Neurol 2022; 351:113990. [DOI: 10.1016/j.expneurol.2022.113990] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 11/24/2022]
|
32
|
Randelman M, Zholudeva LV, Vinit S, Lane MA. Respiratory Training and Plasticity After Cervical Spinal Cord Injury. Front Cell Neurosci 2021; 15:700821. [PMID: 34621156 PMCID: PMC8490715 DOI: 10.3389/fncel.2021.700821] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 08/11/2021] [Indexed: 12/30/2022] Open
Abstract
While spinal cord injuries (SCIs) result in a vast array of functional deficits, many of which are life threatening, the majority of SCIs are anatomically incomplete. Spared neural pathways contribute to functional and anatomical neuroplasticity that can occur spontaneously, or can be harnessed using rehabilitative, electrophysiological, or pharmacological strategies. With a focus on respiratory networks that are affected by cervical level SCI, the present review summarizes how non-invasive respiratory treatments can be used to harness this neuroplastic potential and enhance long-term recovery. Specific attention is given to "respiratory training" strategies currently used clinically (e.g., strength training) and those being developed through pre-clinical and early clinical testing [e.g., intermittent chemical stimulation via altering inhaled oxygen (hypoxia) or carbon dioxide stimulation]. Consideration is also given to the effect of training on non-respiratory (e.g., locomotor) networks. This review highlights advances in this area of pre-clinical and translational research, with insight into future directions for enhancing plasticity and improving functional outcomes after SCI.
Collapse
Affiliation(s)
- Margo Randelman
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States.,Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Lyandysha V Zholudeva
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States.,Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States.,Gladstone Institutes, San Francisco, CA, United States
| | - Stéphane Vinit
- INSERM, END-ICAP, Université Paris-Saclay, UVSQ, Versailles, France
| | - Michael A Lane
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States.,Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
33
|
Walker JR, Detloff MR. Plasticity in Cervical Motor Circuits following Spinal Cord Injury and Rehabilitation. BIOLOGY 2021; 10:biology10100976. [PMID: 34681075 PMCID: PMC8533179 DOI: 10.3390/biology10100976] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Spinal cord injury results in a decreased quality of life and impacts hundreds of thousands of people in the US alone. This review discusses the underlying cellular mechanisms of injury and the concurrent therapeutic hurdles that impede recovery. It then describes the phenomena of neural plasticity—the nervous system’s ability to change. The primary focus of the review is on the impact of cervical spinal cord injury on control of the upper limbs. The neural plasticity that occurs without intervention is discussed, which shows new connections growing around the injury site and the involvement of compensatory movements. Rehabilitation-driven neural plasticity is shown to have the ability to guide connections to create more normal functions. Various novel stimulation and recording technologies are outlined for their role in further improving rehabilitative outcomes and gains in independence. Finally, the importance of sensory input, an often-overlooked aspect of motor control, is shown in driving neural plasticity. Overall, this review seeks to delineate the historical and contemporary research into neural plasticity following injury and rehabilitation to guide future studies. Abstract Neuroplasticity is a robust mechanism by which the central nervous system attempts to adapt to a structural or chemical disruption of functional connections between neurons. Mechanical damage from spinal cord injury potentiates via neuroinflammation and can cause aberrant changes in neural circuitry known as maladaptive plasticity. Together, these alterations greatly diminish function and quality of life. This review discusses contemporary efforts to harness neuroplasticity through rehabilitation and neuromodulation to restore function with a focus on motor recovery following cervical spinal cord injury. Background information on the general mechanisms of plasticity and long-term potentiation of the nervous system, most well studied in the learning and memory fields, will be reviewed. Spontaneous plasticity of the nervous system, both maladaptive and during natural recovery following spinal cord injury is outlined to provide a baseline from which rehabilitation builds. Previous research has focused on the impact of descending motor commands in driving spinal plasticity. However, this review focuses on the influence of physical therapy and primary afferent input and interneuron modulation in driving plasticity within the spinal cord. Finally, future directions into previously untargeted primary afferent populations are presented.
Collapse
|
34
|
Karnup S. Spinal interneurons of the lower urinary tract circuits. Auton Neurosci 2021; 235:102861. [PMID: 34391124 DOI: 10.1016/j.autneu.2021.102861] [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: 04/01/2021] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
The storage and elimination of urine requires coordinated activity between muscles of the bladder and the urethra. This coordination is orchestrated by a complex system containing spinal, midbrain and forebrain networks. Normally there is a reciprocity between patterns of activity in urinary bladder sacral parasympathetic efferents and somatic motoneurons innervating the striatal external urethral sphincter muscle. At the spinal level this reciprocity is mediated by ensembles of excitatory and inhibitory interneurons located in the lumbar-sacral segments. In this review I will present an overview of currently identified spinal interneurons and circuits relevant to the lower urinary tract and will discuss their established or hypothetical roles in the cycle of micturition. In addition, a recently discovered auxiliary spinal neuronal ensemble named lumbar spinal coordinating center will be described. Sexual dimorphism and developmental features of the lower urinary tract which may play a significant role in designing treatments for patients with urine storage and voiding dysfunctions are also considered. Spinal cord injuries seriously damage or even eliminate the ability to urinate. Treatment of this abnormality requires detailed knowledge of supporting neural mechanisms, therefore various experiments in normal and spinalized animals will be discussed. Finally, a possible intraspinal mechanism will be proposed for organization of external urethral sphincter (EUS) bursting which represents a form of intermittent EUS relaxation in rats and mice.
Collapse
Affiliation(s)
- Sergei Karnup
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 200 Lothrop St. BST, R.1303, Pittsburgh, 15213, PA, United States.
| |
Collapse
|
35
|
Unusual Quadrupedal Locomotion in Rat during Recovery from Lumbar Spinal Blockade of 5-HT 7 Receptors. Int J Mol Sci 2021; 22:ijms22116007. [PMID: 34199392 PMCID: PMC8199611 DOI: 10.3390/ijms22116007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 01/18/2023] Open
Abstract
Coordination of four-limb movements during quadrupedal locomotion is controlled by supraspinal monoaminergic descending pathways, among which serotoninergic ones play a crucial role. Here we investigated the locomotor pattern during recovery from blockade of 5-HT7 or 5-HT2A receptors after intrathecal application of SB269970 or cyproheptadine in adult rats with chronic intrathecal cannula implanted in the lumbar spinal cord. The interlimb coordination was investigated based on electromyographic activity recorded from selected fore- and hindlimb muscles during rat locomotion on a treadmill. In the time of recovery after hindlimb transient paralysis, we noticed a presence of an unusual pattern of quadrupedal locomotion characterized by a doubling of forelimb stepping in relation to unaffected hindlimb stepping (2FL-1HL) after blockade of 5-HT7 receptors but not after blockade of 5-HT2A receptors. The 2FL-1HL pattern, although transient, was observed as a stable form of fore-hindlimb coupling during quadrupedal locomotion. We suggest that modulation of the 5-HT7 receptors on interneurons located in lamina VII with ascending projections to the forelimb spinal network can be responsible for the 2FL-1HL locomotor pattern. In support, our immunohistochemical analysis of the lumbar spinal cord demonstrated the presence of the 5-HT7 immunoreactive cells in the lamina VII, which were rarely 5-HT2A immunoreactive.
Collapse
|
36
|
Spinal Inhibitory Interneurons: Gatekeepers of Sensorimotor Pathways. Int J Mol Sci 2021; 22:ijms22052667. [PMID: 33800863 PMCID: PMC7961554 DOI: 10.3390/ijms22052667] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 03/04/2021] [Indexed: 12/20/2022] Open
Abstract
The ability to sense and move within an environment are complex functions necessary for the survival of nearly all species. The spinal cord is both the initial entry site for peripheral information and the final output site for motor response, placing spinal circuits as paramount in mediating sensory responses and coordinating movement. This is partly accomplished through the activation of complex spinal microcircuits that gate afferent signals to filter extraneous stimuli from various sensory modalities and determine which signals are transmitted to higher order structures in the CNS and to spinal motor pathways. A mechanistic understanding of how inhibitory interneurons are organized and employed within the spinal cord will provide potential access points for therapeutics targeting inhibitory deficits underlying various pathologies including sensory and movement disorders. Recent studies using transgenic manipulations, neurochemical profiling, and single-cell transcriptomics have identified distinct populations of inhibitory interneurons which express an array of genetic and/or neurochemical markers that constitute functional microcircuits. In this review, we provide an overview of identified neural components that make up inhibitory microcircuits within the dorsal and ventral spinal cord and highlight the importance of inhibitory control of sensorimotor pathways at the spinal level.
Collapse
|