1
|
Ruiz-Vitte A, Gutiérrez-Fernández M, Laso-García F, Piniella D, Gómez-de Frutos MC, Díez-Tejedor E, Gutiérrez Á, Alonso de Leciñana M. Ledged Beam Walking Test Automatic Tracker: Artificial intelligence-based functional evaluation in a stroke model. Comput Biol Med 2025; 186:109689. [PMID: 39862465 DOI: 10.1016/j.compbiomed.2025.109689] [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: 06/25/2024] [Revised: 01/05/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025]
Abstract
The quantitative evaluation of motor function in experimental stroke models is essential for the preclinical assessment of new therapeutic strategies that can be transferred to clinical research; however, conventional assessment tests are hampered by the evaluator's subjectivity. We present an artificial intelligence-based system for the automatic, accurate, and objective analysis of target parameters evaluated by the ledged beam walking test, which offers higher sensitivity than the current methodology based on manual and visual counting. This system employs a residual deep network model, trained with DeepLabCut (DLC) to extract target paretic hindlimb coordinates, which are categorized to provide a ratio measurement of the animal's neurological deficit. The results correlate with the measurements performed by a professional observer and have greater reproducibility, easing the analysis of motor deficits and providing a reliable and useful tool applicable to other diseases causing motor deficits.
Collapse
Affiliation(s)
- Ainhoa Ruiz-Vitte
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area La Paz Institute for Health Research (idiPAZ), (La Paz University Hospital- Universidad Autónoma de Madrid), Spain; ETSI Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain
| | - María Gutiérrez-Fernández
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area La Paz Institute for Health Research (idiPAZ), (La Paz University Hospital- Universidad Autónoma de Madrid), Spain
| | - Fernando Laso-García
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area La Paz Institute for Health Research (idiPAZ), (La Paz University Hospital- Universidad Autónoma de Madrid), Spain
| | - Dolores Piniella
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area La Paz Institute for Health Research (idiPAZ), (La Paz University Hospital- Universidad Autónoma de Madrid), Spain; Universidad Autónoma de Madrid and IdiPAZ Health Research Institute, La Paz University Hospital, Madrid, Spain
| | - Mari Carmen Gómez-de Frutos
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area La Paz Institute for Health Research (idiPAZ), (La Paz University Hospital- Universidad Autónoma de Madrid), Spain
| | - Exuperio Díez-Tejedor
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area La Paz Institute for Health Research (idiPAZ), (La Paz University Hospital- Universidad Autónoma de Madrid), Spain
| | - Álvaro Gutiérrez
- ETSI Telecomunicación, Universidad Politécnica de Madrid, Madrid, Spain
| | - María Alonso de Leciñana
- Neurological Sciences and Cerebrovascular Research Laboratory, Department of Neurology and Stroke Centre, Neurology and Cerebrovascular Disease Group, Neuroscience Area La Paz Institute for Health Research (idiPAZ), (La Paz University Hospital- Universidad Autónoma de Madrid), Spain.
| |
Collapse
|
2
|
Hubertus V, Meyer L, Waldmann L, Roolfs L, Taheri N, Kersting K, von Bronewski E, Nieminen-Kelhä M, Kremenetskaia I, Uhl C, Fiedler KC, Ode JE, Rex A, Prüß H, Rakhymzhan A, Hauser AE, Niesner R, Heppner FL, Fehlings MG, Vajkoczy P. Identification of a Therapeutic Window for Neurovascular Unit Repair after Experimental Spinal Cord Injury. J Neurotrauma 2024. [PMID: 39585767 DOI: 10.1089/neu.2024.0233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating condition for which effective neuroregenerative and neuroreparative strategies are lacking. The post-traumatic disruption of the blood-spinal cord barrier (BSCB) as part of the neurovascular unit (NVU) is one major factor in the complex pathophysiology of SCI, which is associated with edema, inflammation, and cell death in the penumbra regions of the spinal cord adjacent to the lesion epicenter. Thus, the preservation of an intact NVU and vascular integrity to facilitate the regenerative capacity following SCI is a desirable therapeutic target. This study aims to identify a therapeutic window of opportunity for NVU repair after SCI by characterizing the timeframe of its post-traumatic disintegration and reintegration with implications for functional spinal cord recovery. Following thoracic clip-compression SCI or sham injury, adult C57BL/6J mice were followed up from one to 28 days. At one, three, seven, 14, and 28 days after SCI/sham, seven-Tesla magnetic resonance imaging (MRI), neurobehavioral analysis (Basso mouse scale, Tally subscore, CatWalk® gait analysis), and following sacrifice immunohistochemistry were performed, assessing vessel permeability via Evans blue (EVB) extravasation, (functional) vessel density, and NVU integrity. Thy1-yellow fluorescent protein+ mice were additionally implanted with a customized spinal window chamber and received longitudinal in vivo two-photon excitation imaging (2PM) with the injection of rhodamine-B-isothiocyanate-dextran for the combined imaging of axons and vasculature up to 14 days after SCI/sham injury. Post-traumatic edema formation as assessed by MRI volumetry peaked at one to three days after injury, while EVB permeability quantification revealed a thoroughly injured BSCB up to 14 days after SCI. Partial regeneration of functional vasculature via endogenous revascularization was detected after one to four weeks, however, with only 50-54% of existing vessels regaining functional perfusion. Longitudinal in vivo 2PM visualized the progressive degeneration of initially preserved spinal cord axons in the peri-traumatic zone after SCI while displaying a rarefication of functionally perfused vessels up to two weeks after injury. Neurobehavioral recovery started after one week but remained impaired over the whole observation period of four weeks after SCI. With this study, a therapeutic window to address the impaired NVU starting from the first days to two weeks after SCI is identified. A number of lines of evidence including in vivo 2PM, assessment of NVU integrity, and neurobehavioral assessments point to the critical nature of targeting the NVU to enhance axonal preservation and regeneration after SCI. Continuous multifactorial therapy applications targeting the integrity of the NVU over the identified therapeutic window of opportunity appears promising to ameliorate functional vessel perseverance and the spinal cord's regenerative capacity.
Collapse
Affiliation(s)
- Vanessa Hubertus
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lea Meyer
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lilly Waldmann
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Laurens Roolfs
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nima Taheri
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Katharina Kersting
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Emily von Bronewski
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Christian Uhl
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kim C Fiedler
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan-Erik Ode
- Scientific Workshops, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andre Rex
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Harald Prüß
- Department of Experimental Neurology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Asylkhan Rakhymzhan
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Anja E Hauser
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raluca Niesner
- Deutsches Rheuma-Forschungszentrum, A Leibniz Institute, Berlin, Germany and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Frank L Heppner
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Michael G Fehlings
- Division of Neurosurgery and Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network and University of Toronto, Toronto, Canada
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
3
|
Yuan M, Tang Y, Huang T, Ke L, Huang E. In situ direct reprogramming of astrocytes to neurons via polypyrimidine tract-binding protein 1 knockdown in a mouse model of ischemic stroke. Neural Regen Res 2024; 19:2240-2248. [PMID: 38488558 PMCID: PMC11034579 DOI: 10.4103/1673-5374.390957] [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: 05/06/2023] [Revised: 08/09/2023] [Accepted: 10/16/2023] [Indexed: 04/24/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202410000-00025/figure1/v/2024-02-06T055622Z/r/image-tiff In situ direct reprogramming technology can directly convert endogenous glial cells into functional neurons in vivo for central nervous system repair. Polypyrimidine tract-binding protein 1 (PTB) knockdown has been shown to reprogram astrocytes to functional neurons in situ. In this study, we used AAV-PHP.eB-GFAP-shPTB to knockdown PTB in a mouse model of ischemic stroke induced by endothelin-1, and investigated the effects of GFAP-shPTB-mediated direct reprogramming to neurons. Our results showed that in the mouse model of ischemic stroke, PTB knockdown effectively reprogrammed GFAP-positive cells to neurons in ischemic foci, restored neural tissue structure, reduced inflammatory response, and improved behavioral function. These findings validate the effectiveness of in situ transdifferentiation of astrocytes, and suggest that the approach may be a promising strategy for stroke treatment.
Collapse
Affiliation(s)
- Meng Yuan
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Yao Tang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Scientific Research Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Tianwen Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, China
- Fujian Key Laboratory of Vascular Aging, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Lining Ke
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Department of Human Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| | - En Huang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases of Fujian Province, Fujian Medical University, Fuzhou, Fujian Province, China
- Scientific Research Center, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
4
|
Gargareta VI, Berghoff SA, Krauter D, Hümmert S, Marshall-Phelps KLH, Möbius W, Nave KA, Fledrich R, Werner HB, Eichel-Vogel MA. Myelinated peripheral axons are more vulnerable to mechanical trauma in a model of enlarged axonal diameters. Glia 2024; 72:1572-1589. [PMID: 38895764 DOI: 10.1002/glia.24568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 06/21/2024]
Abstract
The velocity of axonal impulse propagation is facilitated by myelination and axonal diameters. Both parameters are frequently impaired in peripheral nerve disorders, but it is not known if the diameters of myelinated axons affect the liability to injury or the efficiency of functional recovery. Mice lacking the adaxonal myelin protein chemokine-like factor-like MARVEL-transmembrane domain-containing family member-6 (CMTM6) specifically from Schwann cells (SCs) display appropriate myelination but increased diameters of peripheral axons. Here we subjected Cmtm6-cKo mice as a model of enlarged axonal diameters to a mild sciatic nerve compression injury that causes temporarily reduced axonal diameters but otherwise comparatively moderate pathology of the axon/myelin-unit. Notably, both of these pathological features were worsened in Cmtm6-cKo compared to genotype-control mice early post-injury. The increase of axonal diameters caused by CMTM6-deficiency thus does not override their injury-dependent decrease. Accordingly, we did not detect signs of improved regeneration or functional recovery after nerve compression in Cmtm6-cKo mice; depleting CMTM6 in SCs is thus not a promising strategy toward enhanced recovery after nerve injury. Conversely, the exacerbated axonal damage in Cmtm6-cKo nerves early post-injury coincided with both enhanced immune response including foamy macrophages and SCs and transiently reduced grip strength. Our observations support the concept that larger peripheral axons are particularly susceptible toward mechanical trauma.
Collapse
Affiliation(s)
- Vasiliki-Ilya Gargareta
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefan A Berghoff
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Doris Krauter
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sophie Hümmert
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | | | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Robert Fledrich
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Biology and Psychology, University of Göttingen, Göttingen, Germany
| | - Maria A Eichel-Vogel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Yang C, Xu T, Lu Y, Liu J, Chen C, Wang H, Chen X. Quercetin-loaded Human Umbilical cord Mesenchymal Stem Cell-derived sEVs for Spinal Cord Injury Recovery. Neuroscience 2024; 552:14-28. [PMID: 38806069 DOI: 10.1016/j.neuroscience.2024.05.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/03/2024] [Accepted: 05/22/2024] [Indexed: 05/30/2024]
Abstract
Following spinal cord injury, the inflammatory environment at the injury site causes local microglia and astrocytes to activate, which worsens the nerve damage in the affected area. Quercetin, an anti-inflammatory agent, has been limited in spinal cord injury due to its poor water solubility and easy degradation. Stem cell-derived extracellular vesicles can go through the blood-brain barrier and are an ideal drug delivery system. In this study, umbilical cord mesenchymal stem cell-derived extracellular vesicles were used to load quercetin to prevent its degradation and allow it to accumulate at the site of spinal cord injury. Our results showed that quercetin-loaded extracellular vesicles could inhibit the activation of microglia to M1 phenotype through the TLR4/NF-κB pathway, and the activation of astrocytes to A1 phenotype through the JAK2/STAT3 pathway. This reduced the production of inflammatory factors, mitigated neuronal damage, and inhibited the growth of astroglial scar, but promoted the recovery of motor function in rats with spinal cord injury.
Collapse
Affiliation(s)
- Changwei Yang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Tao Xu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Yang Lu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Jianhang Liu
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Cheng Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Heng Wang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Xiaoqing Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
6
|
Deppen JN, Ginn SC, Tang EO, Wang L, Brockman ML, Levit RD. Alginate-Encapsulated Mesenchymal Stromal Cells Improve Hind Limb Ischemia in a Translational Swine Model. J Am Heart Assoc 2024; 13:e029880. [PMID: 38639336 PMCID: PMC11179867 DOI: 10.1161/jaha.123.029880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 03/01/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Cellular therapies have been investigated to improve blood flow and prevent amputation in peripheral artery disease with limited efficacy in clinical trials. Alginate-encapsulated mesenchymal stromal cells (eMSCs) demonstrated improved retention and survival and promoted vascular generation in murine hind limb ischemia through their secretome, but large animal evaluation is necessary for human applicability. We sought to determine the efficacy of eMSCs for peripheral artery disease-induced limb ischemia through assessment in our durable swine hind limb ischemia model. METHODS AND RESULTS Autologous bone marrow eMSCs or empty alginate capsules were intramuscularly injected 2 weeks post-hind limb ischemia establishment (N=4/group). Improvements were quantified for 4 weeks through walkway gait analysis, contrast angiography, blood pressures, fluorescent microsphere perfusion, and muscle morphology and histology. Capsules remained intact with mesenchymal stromal cells retained for 4 weeks. Adenosine-induced perfusion deficits and muscle atrophy in ischemic limbs were significantly improved by eMSCs versus empty capsules (mean±SD, 1.07±0.19 versus 0.41±0.16, P=0.002 for perfusion ratios and 2.79±0.12 versus 1.90±0.62 g/kg, P=0.029 for ischemic muscle mass). Force- and temporal-associated walkway parameters normalized (ratio, 0.63±0.35 at week 3 versus 1.02±0.19 preligation; P=0.17), and compensatory footfall patterning was diminished in eMSC-administered swine (12.58±8.46% versus 34.85±15.26%; P=0.043). Delivery of eMSCs was associated with trending benefits in collateralization, local neovascularization, and muscle fibrosis. Hypoxia-cultured porcine mesenchymal stromal cells secreted vascular endothelial growth factor and tissue inhibitor of metalloproteinase 2. CONCLUSIONS This study demonstrates the promise of the mesenchymal stromal cell secretome at improving peripheral artery disease outcomes and the potential for this novel swine model to serve as a component of the preclinical pipeline for advanced therapies.
Collapse
Affiliation(s)
- Juline N. Deppen
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Sydney C. Ginn
- Wallace H. Coulter Department of Biomedical EngineeringGeorgia Institute of Technology and Emory UniversityAtlantaGA
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Erica O. Tang
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | - Lanfang Wang
- Division of CardiologyEmory University School of MedicineAtlantaGA
| | | | - Rebecca D. Levit
- Division of CardiologyEmory University School of MedicineAtlantaGA
| |
Collapse
|
7
|
Radlicka-Borysewska A, Jabłońska J, Lenarczyk M, Szumiec Ł, Harda Z, Bagińska M, Barut J, Pera J, Kreiner G, Wójcik DK, Rodriguez Parkitna J. Non-motor symptoms associated with progressive loss of dopaminergic neurons in a mouse model of Parkinson's disease. Front Neurosci 2024; 18:1375265. [PMID: 38745938 PMCID: PMC11091341 DOI: 10.3389/fnins.2024.1375265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/09/2024] [Indexed: 05/16/2024] Open
Abstract
Parkinson's disease (PD) is characterized by three main motor symptoms: bradykinesia, rigidity and tremor. PD is also associated with diverse non-motor symptoms that may develop in parallel or precede motor dysfunctions, ranging from autonomic system dysfunctions and impaired sensory perception to cognitive deficits and depression. Here, we examine the role of the progressive loss of dopaminergic transmission in behaviors related to the non-motor symptoms of PD in a mouse model of the disease (the TIF-IADATCreERT2 strain). We found that in the period from 5 to 12 weeks after the induction of a gradual loss of dopaminergic neurons, mild motor symptoms became detectable, including changes in the distance between paws while standing as well as the swing speed and step sequence. Male mutant mice showed no apparent changes in olfactory acuity, no anhedonia-like behaviors, and normal learning in an instrumental task; however, a pronounced increase in the number of operant responses performed was noted. Similarly, female mice with progressive dopaminergic neuron degeneration showed normal learning in the probabilistic reversal learning task and no loss of sweet-taste preference, but again, a robustly higher number of choices were performed in the task. In both males and females, the higher number of instrumental responses did not affect the accuracy or the fraction of rewarded responses. Taken together, these data reveal discrete, dopamine-dependent non-motor symptoms that emerge in the early stages of dopaminergic neuron degeneration.
Collapse
Affiliation(s)
- Anna Radlicka-Borysewska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Judyta Jabłońska
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Michał Lenarczyk
- Faculty of Management and Social Communication, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
| | - Łukasz Szumiec
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Zofia Harda
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Monika Bagińska
- Department of Brain Biochemistry, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Justyna Barut
- Department of Brain Biochemistry, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Joanna Pera
- Department of Neurology, Jagiellonian University Medical College, Kraków, Poland
| | - Grzegorz Kreiner
- Department of Brain Biochemistry, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| | - Daniel K. Wójcik
- Faculty of Management and Social Communication, Institute of Applied Psychology, Jagiellonian University, Kraków, Poland
- Laboratory of Neuroinformatics, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Jan Rodriguez Parkitna
- Department of Molecular Neuropharmacology, Maj Institute of Pharmacology of the Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
8
|
Fogarty NL, Johnson T, Kwok B, Lin A, Tsinman TK, Jiang X, Koyama E, Han L, Baxter JR, Mauck RL, Dyment NA. Reduction in postnatal weight-bearing does not alter the trajectory of murine meniscus growth and maturation. J Orthop Res 2024; 42:894-904. [PMID: 37804210 PMCID: PMC10978302 DOI: 10.1002/jor.25711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/27/2023] [Accepted: 10/05/2023] [Indexed: 10/09/2023]
Abstract
The early postnatal period represents a critical window for the maturation and development of orthopedic tissues, including those within the knee joint. To understand how mechanical loading impacts the maturational trajectory of the meniscus and other tissues of the hindlimb, perturbation of postnatal weight bearing was achieved through surgical resection of the sciatic nerve in neonatal mice at 1 or 14 days old. Sciatic nerve resection (SNR) produced significant and persistent disruptions in gait, leading to reduced tibial length and reductions in Achilles tendon mechanical properties. However, SNR resulted in minimal disruptions in morphometric parameters of the menisci and other structures in the knee joint, with no detectable differences in Col1a1-YFP or Col2a1-CFP expressing cells within the menisci. Furthermore, micromechanical properties of the meniscus and cartilage (as assessed by atomic force microscopy-based nanoindentation testing) were not different between experimental groups. In contrast to our initial hypothesis, reduced hindlimb weight bearing via neonatal SNR did not significantly impact the growth and development of the knee meniscus. This unexpected finding demonstrates that the input mechanical threshold required to sustain meniscus development may be lower than previously hypothesized, though future studies incorporating skeletal kinematic models coupled with force plate measurements will be required to calculate the loads passing through the affected hindlimb and precisely define these thresholds. Collectively, these results provide insight into the mechanobiological responses of the meniscus to alterations in load, and contribute to our understanding of the factors that influence normal postnatal development.
Collapse
Affiliation(s)
- Natalie L Fogarty
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Talayah Johnson
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Alisia Lin
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tonia K Tsinman
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xi Jiang
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eiki Koyama
- Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, Pennsylvania, USA
| | - Josh R Baxter
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Laboratory, CMC VA Medical Center, Philadelphia, Pennsylvania, USA
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
李 文, 宋 娟, 张 含, 杨 禄, 岳 宇, 张 新, 王 永. [Effects of α1-antitrypsin on motor function in mice with immature brain white matter injury]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:181-187. [PMID: 38436317 PMCID: PMC10921877 DOI: 10.7499/j.issn.1008-8830.2309003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/11/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVES To investigate the effects of α1-antitrypsin (AAT) on motor function in adult mice with immature brain white matter injury. METHODS Five-day-old C57BL/6J mice were randomly assigned to the sham surgery group (n=27), hypoxia-ischemia (HI) + saline group (n=27), and HI+AAT group (n=27). The HI white matter injury mouse model was established using HI methods. The HI+AAT group received intraperitoneal injections of AAT (50 mg/kg) 24 hours before HI, immediately after HI, and 72 hours after HI; the HI+saline group received intraperitoneal injections of the same volume of saline at the corresponding time points. Brain T2-weighted magnetic resonance imaging scans were performed at 7 and 55 days after modeling. At 2 months of age, adult mice were evaluated for static, dynamic, and coordination parameters using the Catwalk gait analysis system. RESULTS Compared to the sham surgery group, mice with HI injury showed high signal intensity on brain T2-weighted magnetic resonance imaging at 7 days after modeling, indicating significant white matter injury. The white matter injury persisted at 55 days after modeling. In comparison to the sham surgery group, the HI+saline group exhibited decreased paw print area, maximum contact area, average pressure, maximum pressure, paw print width, average velocity, body velocity, stride length, swing speed, percentage of gait pattern AA, and percentage of inter-limb coordination (left hind paw → left front paw) (P<0.05). The HI+saline group showed increased inter-paw distance, percentage of gait pattern AB, and percentage of phase lag (left front paw → left hind paw) compared to the sham surgery group (P<0.05). In comparison to the HI+saline group, the HI+AAT group showed increased average velocity, body velocity, stride length, and swing speed (right front paw) (P<0.05). CONCLUSIONS The mice with immature brain white matter injury may exhibit significant motor dysfunction in adulthood, while the use of AAT can improve some aspects of their motor function.
Collapse
|
10
|
Islam R, Ahlfors JE, Siu R, Noman H, Akbary R, Morshead CM. Inhibition of Apoptosis in a Model of Ischemic Stroke Leads to Enhanced Cell Survival, Endogenous Neural Precursor Cell Activation and Improved Functional Outcomes. Int J Mol Sci 2024; 25:1786. [PMID: 38339065 PMCID: PMC10855341 DOI: 10.3390/ijms25031786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/24/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Stroke results in neuronal cell death, which causes long-term disabilities in adults. Treatment options are limited and rely on a narrow window of opportunity. Apoptosis inhibitors demonstrate efficacy in improving neuronal cell survival in animal models of stroke. However, many inhibitors non-specifically target apoptosis pathways and high doses are needed for treatment. We explored the use of a novel caspase-3/7 inhibitor, New World Laboratories (NWL) 283, with a lower IC50 than current caspase-3/7 inhibitors. We performed in vitro and in vivo assays to determine the efficacy of NWL283 in modulating cell death in a preclinical model of stroke. In vitro and in vivo assays show that NWL283 enhances cell survival of neural precursor cells. Delivery of NWL283 following stroke enhances endogenous NPC migration and leads to increased neurogenesis in the stroke-injured cortex. Furthermore, acute NWL283 administration is neuroprotective at the stroke injury site, decreasing neuronal cell death and reducing microglia activation. Coincident with NWL283 delivery for 8 days, stroke-injured mice exhibited improved functional outcomes that persisted following cessation of the drug. Therefore, we propose that NWL283 is a promising therapeutic warranting further investigation to enhance stroke recovery.
Collapse
Affiliation(s)
- Rehnuma Islam
- Institute of Medical Science, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
| | - Jan-Eric Ahlfors
- New World Laboratories, 275 Boul. Armand-Frappier, Laval, QC H7V 4A7, Canada
| | - Ricky Siu
- Department of Surgery, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
| | - Humna Noman
- Institute of Medical Science, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
| | - Roya Akbary
- Department of Surgery, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
| | - Cindi M. Morshead
- Institute of Medical Science, University of Toronto, 1 King’s College Circle, Toronto, ON M5S 3E1, Canada
- Department of Surgery, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, ON M5S 3G9, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON M5S 3E1, Canada
| |
Collapse
|
11
|
Zhong Y, Li S, Chen Y, Tang Y, Xiao X, Nie T. Combining PLGA microspheres loaded with Liver X receptor agonist GW3965 with a chitosan nerve conduit can promote the healing and regeneration of the wounded sciatic nerve. J Biomed Mater Res B Appl Biomater 2024; 112:e35378. [PMID: 38356051 DOI: 10.1002/jbm.b.35378] [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: 11/30/2023] [Accepted: 01/06/2024] [Indexed: 02/16/2024]
Abstract
Globally, peripheral nerve injury (PNI) is a common clinical issue. Successfully repairing severe PNIs has posed a major challenge for clinicians. GW3965 is a highly selective LXR agonist, and previous studies have demonstrated its positive protective effects in both central and peripheral nerve diseases. In this work, we examined the potential reparative effects of GW3965-loaded polylactic acid co-glycolic acid microspheres in conjunction with a chitosan nerve conduit for peripheral nerve damage. The experiment revealed that GW3965 promoted Schwann cell proliferation and neurotrophic factor release in vitro. In vivo experiments conducted on rats showed that GW3965 facilitated the restoration of motor function, promoted axon and myelin regeneration in the sciatic nerve, and enhanced the microenvironment of nerve regeneration. These results offer a novel therapeutic approach for the healing of nerve damage. Overall, this work provides valuable insights and presents a promising therapeutic strategy for addressing PNI.
Collapse
Affiliation(s)
- Yuanwu Zhong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Shiqi Li
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yanzhen Chen
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yuan Tang
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xinmao Xiao
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tao Nie
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
12
|
Pradier B, Segelcke D, Reichl S, Zahn PK, Pogatzki-Zahn EM. Spinal GABA transporter 1 contributes to evoked-pain related behavior but not resting pain after incision injury. Front Mol Neurosci 2023; 16:1282151. [PMID: 38130683 PMCID: PMC10734427 DOI: 10.3389/fnmol.2023.1282151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023] Open
Abstract
The inhibitory function of GABA at the spinal level and its central modulation in the brain are essential for pain perception. However, in post-surgical pain, the exact mechanism and modes of action of GABAergic transmission have been poorly studied. This work aimed to investigate GABA synthesis and uptake in the incisional pain model in a time-dependent manner. Here, we combined assays for mechanical and heat stimuli-induced withdrawal reflexes with video-based assessments and assays for non-evoked (NEP, guarding of affected hind paw) and movement-evoked (MEP, gait pattern) pain-related behaviors in a plantar incision model in male rats to phenotype the effects of the inhibition of the GABA transporter (GAT-1), using a specific antagonist (NO711). Further, we determined the expression profile of spinal dorsal horn GAT-1 and glutamate decarboxylase 65/67 (GAD65/67) by protein expression analyses at four time points post-incision. Four hours after incision, we detected an evoked pain phenotype (mechanical, heat and movement), which transiently ameliorated dose-dependently following spinal inhibition of GAT-1. However, the NEP-phenotype was not affected. Four hours after incision, GAT-1 expression was significantly increased, whereas GAD67 expression was significantly reduced. Our data suggest that GAT-1 plays a role in balancing spinal GABAergic signaling in the spinal dorsal horn shortly after incision, resulting in the evoked pain phenotype. Increased GAT-1 expression leads to increased GABA uptake from the synaptic cleft and reduces tonic GABAergic inhibition at the post-synapse. Inhibition of GAT-1 transiently reversed this imbalance and ameliorated the evoked pain phenotype.
Collapse
Affiliation(s)
- Bruno Pradier
- Department of Anesthesiology, Operative Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Daniel Segelcke
- Department of Anesthesiology, Operative Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - Sylvia Reichl
- Department of Anesthesiology, Operative Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| | - P. K. Zahn
- Department of Anesthesiology, Intensive Care and Pain Medicine, BG University Hospital Bergmannsheil, Ruhr-Universität Bochum, Bochum, Germany
| | - E. M. Pogatzki-Zahn
- Department of Anesthesiology, Operative Intensive Care and Pain Medicine, University Hospital Muenster, Muenster, Germany
| |
Collapse
|
13
|
Ma S, Zhang J, Hua X, Wu J, Zheng M, Xu J. Tuina therapy promotes behavioral improvement and brain plasticity in rats with peripheral nerve injury and repair. Brain Behav 2023; 13:e3174. [PMID: 37522806 PMCID: PMC10498059 DOI: 10.1002/brb3.3174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 06/16/2023] [Accepted: 07/09/2023] [Indexed: 08/01/2023] Open
Abstract
INTRODUCTION Tuina is currently one of the popular complementary and alternative methods of rehabilitation therapy. Tuina can improve patients' pain and mobility function. However, the underlying physiological mechanism remains largely unknown, which might limit its further popularization in clinical practice. The aim of this study is to explore the short-term and long-term changes in brain functional activity following Tuina intervention for peripheral nerve injury repair. METHODS A total of 16 rats were equally divided into the intervention group and the control group. Rats in the intervention group received Tuina therapy applying on the gastrocnemius muscle of the right side for 4 months following sciatic nerve transection and immediate repair, while the control group received nerve transection and repair only. The block-design functional magnetic resonance imaging scan was applied in both groups at 1 and 4 months after the surgery. During the scan, both the injured and intact hindpaw was electrically stimulated according to a "boxcar" paradigm. RESULTS When stimulating the intact hindpaw, the intervention group exhibited significantly lower activation in the somatosensory area, limbic/paralimbic areas, pain-regulation areas, and basal ganglia compared to the control group, with only the prefrontal area showing higher activation. After 4 months of sciatic nerve injury, the control group exhibited decreased motor cortex activity compared to the activity observed at 1 month, and the intervention group demonstrated stronger bilateral motor cortex activity compared to the control group. CONCLUSION Tuina therapy on the gastrocnemius muscle of rats with sciatic nerve injury can effectively alleviate pain and maintain the motor function of the affected limb. In addition, Tuina therapy reduced the activation level of pain-related brain regions and inhibited the decreased activity of the motor cortex caused by nerve injury, reflecting the impact of peripheral stimulation on brain plasticity.
Collapse
Affiliation(s)
- Shu‐Jie Ma
- Department of Traditional Chinese Rehabilitation MedicineThe Second Rehabilitation Hospital of ShanghaiShanghaiChina
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
| | - Jun‐Peng Zhang
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Xu‐Yun Hua
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
- Department of Traumatology and Orthopedics, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jia‐Jia Wu
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
- Department of Rehabilitation Medicine, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Mou‐Xiong Zheng
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
- Department of Traumatology and Orthopedics, Yueyang HospitalShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Jian‐Guang Xu
- Engineering Research Center of Traditional Chinese Medicine Intelligent RehabilitationMinistry of EducationShanghaiChina
- School of Rehabilitation ScienceShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
14
|
Zhao Y, Liu J, Liu S, Yang P, Liang Y, Ma J, Mao S, Sun C, Yang Y. Fibroblast exosomal TFAP2C induced by chitosan oligosaccharides promotes peripheral axon regeneration via the miR-132-5p/CAMKK1 axis. Bioact Mater 2023; 26:249-263. [PMID: 36936807 PMCID: PMC10020534 DOI: 10.1016/j.bioactmat.2023.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Chitosan and its degradation product, oligosaccharides, have been shown to facilitate peripheral nerve regeneration. However, the underlying mechanisms are not well understood. In this study, we analyzed the protein expression profiles in sciatic nerves after injury using proteomics. A group of proteins related to exosome packaging and transport is up-regulated by chitosan oligosaccharides (COS), implying that exosomes are involved in COS-induced peripheral nerve regeneration. In fact, exosomes derived from fibroblasts (f-EXOs) treated with COS significantly promoted axon extension and regeneration. Exosomal protein identification and functional studies, revealed that TFAP2C is a key factor in neurite outgrowth induced by COS-f-EXOs. Furthermore, we showed that TFAP2C targets the pri-miRNA-132 gene and represses miR-132-5p expression in dorsal root ganglion neurons. Camkk1 is a downstream substrate of miR-132-5p that positively affects axon extension. In rats, miR-132-5p antagomir stimulates CAMKK1 expression and improves axon regeneration and functional recovery in sciatic nerves after injury. Our data reveal the mechanism for COS in axon regeneration, that is COS induce fibroblasts to produce TFAP2C-enriched EXOs, which are then transferred into axons to promote axon regeneration via miR-132-5p/CAMKK1. Moreover, these results show a new facet of fibroblasts in axon regeneration in peripheral nerves.
Collapse
Affiliation(s)
- Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Jina Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Sha Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Panpan Yang
- School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Yunyun Liang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Jinyu Ma
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Susu Mao
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-Innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, China
| |
Collapse
|
15
|
Timotius IK, Roelofs RF, Richmond-Hacham B, Noldus LPJJ, von Hörsten S, Bikovski L. CatWalk XT gait parameters: a review of reported parameters in pre-clinical studies of multiple central nervous system and peripheral nervous system disease models. Front Behav Neurosci 2023; 17:1147784. [PMID: 37351154 PMCID: PMC10284348 DOI: 10.3389/fnbeh.2023.1147784] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/16/2023] [Indexed: 06/24/2023] Open
Abstract
Automated gait assessment tests are used in studies of disorders characterized by gait impairment. CatWalk XT is one of the first commercially available automated systems for analyzing the gait of rodents and is currently the most used system in peer-reviewed publications. This automated gait analysis system can generate a large number of gait parameters. However, this creates a new challenge in selecting relevant parameters that describe the changes within a particular disease model. Here, for the first time, we performed a multi-disorder review on published CatWalk XT data. We identify commonly reported CatWalk XT gait parameters derived from 91 peer-reviewed experimental studies in mice, covering six disorders of the central nervous system (CNS) and peripheral nervous system (PNS). The disorders modeled in mice were traumatic brain injury (TBI), stroke, sciatic nerve injury (SNI), spinal cord injury (SCI), Parkinson's disease (PD), and ataxia. Our review consisted of parameter selection, clustering, categorization, statistical evaluation, and data visualization. It suggests that certain gait parameters serve as potential indicators of gait dysfunction across multiple disease models, while others are specific to particular models. The findings also suggest that the more site-specific the injury is, the fewer parameters are reported to characterize its gait abnormalities. This study strives to present a clearly organized picture of gait parameters used in each one of the different mouse models, potentially helping novel CatWalk XT users to apply this information to similar or related mouse models they are working on.
Collapse
Affiliation(s)
- Ivanna K. Timotius
- Department of Electronics Engineering, Satya Wacana Christian University, Salatiga, Indonesia
- Department of Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Bar Richmond-Hacham
- Myers Neuro-Behavioral Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Lucas P. J. J. Noldus
- Noldus Information Technology BV, Wageningen, Netherlands
- Donders Center for Neuroscience, Radboud University, Nijmegen, Netherlands
| | - Stephan von Hörsten
- Department of Experimental Therapy, University Hospital Erlangen and Preclinical Experimental Animal Center, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Lior Bikovski
- Myers Neuro-Behavioral Core Facility, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
- School of Behavioral Sciences, Netanya Academic College, Netanya, Israel
| |
Collapse
|
16
|
Liang M, Chen L, He Q, Mi X, Qu L, Xie J, Song N. Intraperitoneal injection of iron dextran induces peripheral iron overload and mild neurodegeneration in the nigrostriatal system in C57BL/6 mice. Life Sci 2023; 320:121508. [PMID: 36858315 DOI: 10.1016/j.lfs.2023.121508] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 03/03/2023]
Abstract
AIMS Elevated iron levels in the affected areas of brain are linked to several neurodegenerative diseases including Parkinson's disease (PD). This study investigated the influence of peripheral iron overload in peripheral tissues, as well as its entry into the brain regions on lysosomal functions. The survival of dopaminergic neurons in the nigrostriatal system and motor coordination were also investigated. MAIN METHODS An intraperitoneal injection of iron dextran (FeDx) mouse model was established. Western blot was used to detect iron deposition and lysosomal functions in the liver, spleen, hippocampal (HC), striatum (STR), substantia nigra (SN) and olfactory bulb (OB). Iron in serum and cerebrospinal fluid (CSF) was determined by an iron assay kit. Immunofluorescence and immunohistochemical staining were applied to detect dopaminergic neurons and fibers. Motor behavior was evaluated by gait analysis. KEY FINDINGS Iron was deposited consistently in the liver and spleen, and serum iron was elevated. While iron deposition occurred late in the HC, STR and SN, without apparently affecting CSF iron levels. Although cathepsin B (CTSB), cathepsin D (CTSD), glucocerebrosidase (GCase) and lysosome integrated membrane protein 2 (LIMP-2) protein levels were dramatically up-regulated in the liver and spleen, they were almost unchanged in the brain regions. However, CTSB was up-regulated in acute iron-overloaded OB and primary cultured astrocytes. The number of dopaminergic neurons in the SN remained unchanged, and mice did not exhibit significant motor incoordination. SIGNIFICANCE Intraperitoneal injection of FeDx in mice induces largely peripheral iron overload while not necessarily sufficient to cause severe disruption of the nigrostriatal system.
Collapse
Affiliation(s)
- Meiyu Liang
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Lei Chen
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Qing He
- Department of Neurology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200125, China
| | - Xiaoqing Mi
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Le Qu
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China
| | - Junxia Xie
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China.
| | - Ning Song
- School of Basic Medicine, Institute of Brain Science and Disease, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
17
|
Visco DB, Manhães de Castro R, da Silva MM, Costa de Santana BJR, Bezerra Gouveia HJC, de Moura Ferraz MLR, de Albuquerque GL, Lacerda DC, de Vasconcelos DAA, Guzman Quevedo O, Toscano AE. Neonatal kaempferol exposure attenuates gait and strength deficits and prevents altered muscle phenotype in a rat model of cerebral palsy. Int J Dev Neurosci 2023; 83:80-97. [PMID: 36342836 DOI: 10.1002/jdn.10239] [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: 06/11/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 11/09/2022] Open
Abstract
Cerebral palsy (CP) is characterized by brain damage at a critical period of development of the central nervous system, and, as a result, motor, behavioural and learning deficits are observed in those affected. Flavonoids such as kaempferol have demonstrated potential anti-inflammatory and neuroprotective properties for neurological disorders. This study aimed to assess the effects of neonatal treatment with kaempferol on the body development, grip strength, gait performance and morphological and biochemical phenotype of skeletal muscle in rats subjected to a model of CP. The groups were formed by randomly allocating male Wistar rats after birth to four groups as follows: C = control treated with vehicle, K = control treated with kaempferol, CP = CP treated with vehicle and CPK = CP treated with kaempferol. The model of CP involved perinatal anoxia and sensorimotor restriction of the hind paws during infancy, from the second to the 28th day of postnatal life. Treatment with kaempferol (1 mg/kg) was performed intraperitoneally during the neonatal period. Body weight and length, muscle strength, gait kinetics and temporal and spatial parameters were evaluated in the offspring. On the 36th day of postnatal life, the animals were euthanized for soleus muscle dissection. The muscle fibre phenotype was assessed using the myofibrillar ATPase technique, and the muscle protein expression was measured using the Western blot technique. A reduction in the impact of CP on body phenotype was observed, and this also attenuated deficits in muscle strength and gait. Treatment also mitigated the impact on muscle phenotype by preventing a reduction in the proportion of oxidative fibres and in the histomorphometric parameters in the soleus muscle of rats in the CP group. The results demonstrate that neonatal treatment with kaempferol attenuated gait deficits and impaired muscle strength and muscle maturation in rats subjected to a model of CP.
Collapse
Affiliation(s)
- Diego Bulcão Visco
- Laboratory of Neurofunctional, Department of Biological Science and Health, Federal University of Amapá, Macapá, Brazil.,Postgraduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil.,Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Raul Manhães de Castro
- Postgraduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil.,Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Márcia Maria da Silva
- Postgraduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil.,Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Bárbara Juacy Rodrigues Costa de Santana
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil.,Postgraduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Henrique José Cavalcanti Bezerra Gouveia
- Postgraduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil.,Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | | | - Glayciele Leandro de Albuquerque
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil.,Postgraduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Diego Cabral Lacerda
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Diogo Antonio Alves de Vasconcelos
- Postgraduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil.,Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Omar Guzman Quevedo
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil.,Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Mexico.,Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico.,Postgraduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Ana Elisa Toscano
- Postgraduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil.,Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil.,Nursing Unit, Vitória Academic Center, Federal University of Pernambuco, Vitória de Santo Antão, Brazil.,Postgraduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
18
|
Lopas LA, Shen H, Zhang N, Jang Y, Tawfik VL, Goodman SB, Natoli RM. Clinical Assessments of Fracture Healing and Basic Science Correlates: Is There Room for Convergence? Curr Osteoporos Rep 2022; 21:216-227. [PMID: 36534307 DOI: 10.1007/s11914-022-00770-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the clinical and basic science methods used to assess fracture healing and propose a framework to improve the translational possibilities. RECENT FINDINGS Mainstays of fracture healing assessment include clinical examination, various imaging modalities, and assessment of function. Pre-clinical studies have yielded insight into biomechanical progression as well as the genetic, molecular, and cellular processes of fracture healing. Efforts are emerging to identify early markers to predict impaired healing and possibly early intervention to alter these processes. Despite of the differences in clinical and preclinical research, opportunities exist to unify and improve the translational efforts between these arenas to develop and optimize our ability to assess and predict fracture healing, thereby improving the clinical care of these patients.
Collapse
Affiliation(s)
- Luke A Lopas
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1801 N. Senate Blvd Suite 535, Indianapolis, IN, USA.
| | - Huaishuang Shen
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Orthopaedic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ning Zhang
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Yohan Jang
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1801 N. Senate Blvd Suite 535, Indianapolis, IN, USA
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Roman M Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1801 N. Senate Blvd Suite 535, Indianapolis, IN, USA
| |
Collapse
|
19
|
Doyle AM, Bauer D, Hendrix C, Yu Y, Nebeck SD, Fergus S, Krieg J, Wilmerding LK, Blumenfeld M, Lecy E, Spencer C, Luo Z, Sullivan D, Brackman K, Ross D, Best S, Verma A, Havel T, Wang J, Johnson L, Vitek JL, Johnson MD. Spatiotemporal scaling changes in gait in a progressive model of Parkinson's disease. Front Neurol 2022; 13:1041934. [PMID: 36582611 PMCID: PMC9792983 DOI: 10.3389/fneur.2022.1041934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/30/2022] [Indexed: 12/14/2022] Open
Abstract
Objective Gait dysfunction is one of the most difficult motor signs to treat in patients with Parkinson's disease (PD). Understanding its pathophysiology and developing more effective therapies for parkinsonian gait dysfunction will require preclinical studies that can quantitatively and objectively assess the spatial and temporal features of gait. Design We developed a novel system for measuring volitional, naturalistic gait patterns in non-human primates, and then applied the approach to characterize the progression of parkinsonian gait dysfunction across a sequence of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatments that allowed for intrasubject comparisons across mild, moderate, and severe stages. Results Parkinsonian gait dysfunction was characterized across treatment levels by a slower stride speed, increased time in both the stance and swing phase of the stride cycle, and decreased cadence that progressively worsened with overall parkinsonian severity. In contrast, decreased stride length occurred most notably in the moderate to severe parkinsonian state. Conclusion The results suggest that mild parkinsonism in the primate model of PD starts with temporal gait deficits, whereas spatial gait deficits manifest after reaching a more severe parkinsonian state overall. This study provides important context for preclinical studies in non-human primates studying the neurophysiology of and treatments for parkinsonian gait.
Collapse
Affiliation(s)
- Alex M. Doyle
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Devyn Bauer
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Claudia Hendrix
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Ying Yu
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Shane D. Nebeck
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Sinta Fergus
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Jordan Krieg
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Lucius K. Wilmerding
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Madeline Blumenfeld
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Emily Lecy
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Chelsea Spencer
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Ziling Luo
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Disa Sullivan
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Krista Brackman
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Dylan Ross
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Sendréa Best
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| | - Ajay Verma
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Tyler Havel
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Jing Wang
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Luke Johnson
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Jerrold L. Vitek
- Department of Neurology, University of Minnesota, Minneapolis, MN, United States
| | - Matthew D. Johnson
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
20
|
Kong Y, Kuss M, Shi Y, Fang F, Xue W, Shi W, Liu Y, Zhang C, Zhong P, Duan B. Exercise facilitates regeneration after severe nerve transection and further modulates neural plasticity. Brain Behav Immun Health 2022; 26:100556. [PMID: 36405423 PMCID: PMC9673108 DOI: 10.1016/j.bbih.2022.100556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/03/2022] [Accepted: 11/11/2022] [Indexed: 11/13/2022] Open
Abstract
Patients with severe traumatic peripheral nerve injury (PNI) always suffer from incomplete recovery and poor functional outcome. Physical exercise-based rehabilitation, as a non-invasive interventional strategy, has been widely acknowledged to improve PNI recovery by promoting nerve regeneration and relieving pain. However, effects of exercise on chronic plastic changes following severe traumatic PNIs have been limitedly discussed. In this study, we created a long-gap sciatic nerve transection followed by autograft bridging in rats and tested the therapeutic functions of treadmill running with low intensity and late initiation. We demonstrated that treadmill running effectively facilitated nerve regeneration and prevented muscle atrophy and thus improved sensorimotor functions and walking performance. Furthermore, exercise could reduce inflammation at the injured nerve as well as prevent the overexpression of TRPV1, a pain sensor, in primary afferent sensory neurons. In the central nervous system, we found that PNI induced transcriptive changes at the ipsilateral lumber spinal dorsal horn, and exercise could reverse the differential expression for genes involved in the Notch signaling pathway. In addition, through neural imaging techniques, we found volumetric, microstructural, metabolite, and neuronal activity changes in supraspinal regions of interest (i.e., somatosensory cortex, motor cortex, hippocampus, etc.) after the PNI, some of which could be reversed through treadmill running. In summary, treadmill running with late initiation could promote recovery from long-gap nerve transection, and while it could reverse maladaptive plasticity after the PNI, exercise may also ameliorate comorbidities, such as chronic pain, mental depression, and anxiety in the long term.
Collapse
Affiliation(s)
- Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yu Shi
- School of Biological Sciences, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
| | - Fang Fang
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wen Xue
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Yutong Liu
- Department of Radiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Chi Zhang
- School of Biological Sciences, University of Nebraska Lincoln, Lincoln, NE, 68588, USA
- Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Peng Zhong
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Surgery, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| |
Collapse
|
21
|
Sun X, Li X, Zhang L, Zhang Y, Qi X, Wang S, Qin C. Longitudinal assessment of motor function following the unilateral intrastriatal 6-hydroxydopamine lesion model in mice. Front Behav Neurosci 2022; 16:982218. [PMID: 36505729 PMCID: PMC9730519 DOI: 10.3389/fnbeh.2022.982218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/04/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction Despite the widespread use of the unilateral striatal 6-hydroxydopamine (6-OHDA) lesion model in mice in recent years, the stability of behavioral deficits in the 6-OHDA striatal mouse model over time is not yet clear, raising concerns about using this model to evaluate a compound's long-term therapeutic effects. Materials and methods In the current study, mice were tested at regular intervals in the cylinder test and gait analysis beginning 3 days after 6-OHDA injection of 4 and 8 μg and lasting until 56 days post-lesion. Apomorphine-induced rotational test and rotarod test were also performed on Day 23 and 43 post-lesion, respectively. Immunohistochemistry for dopaminergic neurons stained by tyrosine hydroxylase (TH) was also performed. Results Our results showed that both the 4 and 8 μg 6-OHDA lesion groups exhibited forelimb use asymmetry with a preference for the ipsilateral (injection) side on Day 3 and until Day 21 post-lesion, but did not show forelimb asymmetry on Day 28 to 56 post-lesion. The 8 μg 6-OHDA lesion group still exhibited forelimb asymmetry on Day 28 and 42 post-lesion, but not on Day 56. The gait analysis showed that the contralateral front and hind step cycles increased from Day 3 to 42 post-lesion and recovered on Day 56 post-lesion. In addition, our results displayed a dose-dependent reduction in TH+ cells and TH+ fibers, as well as dose-dependent apomorphine-induced rotations. In the rotarod test, the 8 μg 6-OHDA lesion group, but not the 4 μg group, decreased the latency to fall on the rotarod on Day 43 post-lesion. Conclusion In summary, unilateral striatal 6-OHDA injections of 4 and 8 μg induced spontaneous motor impairment in mice, which partially recovered starting on Day 28 post-lesion. Forced motor deficits were observed in the 8 g 6-OHDA lesion group, which remained stable on Day 43 post-lesion. In addition, the rotarod test and apomorphine-induced rotational test can distinguish between lesions of different extents and are useful tools for the assessment of functional recovery in studies screening novel potential therapies.
Collapse
Affiliation(s)
- Xiuping Sun
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Xianglei Li
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Ling Zhang
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Yu Zhang
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Xiaolong Qi
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Siyuan Wang
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China
| | - Chuan Qin
- National Health Commission Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, National Center of Technology Innovation for Animal Model, Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences (CAMS) & Peking Union Medical College (PUMC), Beijing, China,Changping National Laboratory (CPNL), Beijing, China,*Correspondence: Chuan Qin,
| |
Collapse
|
22
|
Vinestock RC, Felsenthal N, Assaraf E, Katz E, Rubin S, Heinemann-Yerushalmi L, Krief S, Dezorella N, Levin-Zaidman S, Tsoory M, Thomopoulos S, Zelzer E. Neonatal Enthesis Healing Involves Noninflammatory Acellular Scar Formation through Extracellular Matrix Secretion by Resident Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1122-1135. [PMID: 35659946 PMCID: PMC9379688 DOI: 10.1016/j.ajpath.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/19/2022] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Wound healing typically recruits the immune and vascular systems to restore tissue structure and function. However, injuries to the enthesis, a hypocellular and avascular tissue, often result in fibrotic scar formation and loss of mechanical properties, severely affecting musculoskeletal function and life quality. This raises questions about the healing capabilities of the enthesis. Herein, this study established an injury model to the Achilles entheses of neonatal mice to study the effectiveness of early-age enthesis healing. Histology and immunohistochemistry analyses revealed an atypical process that did not involve inflammation or angiogenesis. Instead, healing was mediated by secretion of collagen types I and II by resident cells, which formed a permanent hypocellular and avascular scar. Transmission electron microscopy showed that the cellular response to injury, including endoplasmic reticulum stress, autophagy, and cell death, varied between the tendon and cartilage ends of the enthesis. Single-molecule in situ hybridization, immunostaining, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling assays verified these differences. Finally, gait analysis showed that these processes effectively restored function of the injured leg. These findings reveal a novel healing mechanism in neonatal entheses, whereby local extracellular matrix secretion by resident cells forms an acellular extracellular matrix deposit without inflammation, allowing gait restoration. These insights into the healing mechanism of a complex transitional tissue may lead to new therapeutic strategies for adult enthesis injuries.
Collapse
Affiliation(s)
- Ron C Vinestock
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Assaraf
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Eldad Katz
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Rubin
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sharon Krief
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nili Dezorella
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Smadar Levin-Zaidman
- Department of Electron Microscopy Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Michael Tsoory
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, New York; Department of Biomedical Engineering, Columbia University, New York, New York
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
23
|
Targosinski S, Henzi A, Engmann AK, Rushing EJ, Barth AA, Klein HJ, Kim BS, Giovanoli P, Schwab ME, Plock JA, Schweizer R. A swim test for functional assessment of rodent peripheral nerve regeneration. J Neurosci Methods 2022; 379:109663. [PMID: 35809863 DOI: 10.1016/j.jneumeth.2022.109663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 06/08/2022] [Accepted: 07/04/2022] [Indexed: 10/17/2022]
Affiliation(s)
- Stefan Targosinski
- Department of Plastic Surgery and Hand Surgery, UniversitätsSpital Zürich, Zurich, Switzerland
| | - Anna Henzi
- Institute of Neuropathology, UniversitätsSpital Zürich, Zurich, Switzerland
| | - Anne K Engmann
- Department of Health Sciences and Technology, ETH Zurich, Switzerland; Brain Research Institute, University of Zurich, Zurich, Switzerland
| | | | - André A Barth
- Department of Plastic Surgery and Hand Surgery, UniversitätsSpital Zürich, Zurich, Switzerland
| | - Holger J Klein
- Department of Plastic Surgery and Hand Surgery, UniversitätsSpital Zürich, Zurich, Switzerland; Department of Plastic Surgery and Hand Surgery, Kantonsspital Aarau, Aarau, Switzerland
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, UniversitätsSpital Zürich, Zurich, Switzerland
| | - Pietro Giovanoli
- Department of Plastic Surgery and Hand Surgery, UniversitätsSpital Zürich, Zurich, Switzerland
| | - Martin E Schwab
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Jan A Plock
- Department of Plastic Surgery and Hand Surgery, UniversitätsSpital Zürich, Zurich, Switzerland; Department of Plastic Surgery and Hand Surgery, Kantonsspital Aarau, Aarau, Switzerland
| | - Riccardo Schweizer
- Department of Plastic Surgery and Hand Surgery, UniversitätsSpital Zürich, Zurich, Switzerland.
| |
Collapse
|
24
|
Dallazen JL, da Luz BB, Maria-Ferreira D, Nascimento AM, Cipriani TR, de Souza LM, Geppetti P, de Paula Werner MF. Local effects of natural alkylamides from Acmella oleracea and synthetic isobutylalkyl amide on neuropathic and postoperative pain models in mice. Fitoterapia 2022; 160:105224. [PMID: 35659524 DOI: 10.1016/j.fitote.2022.105224] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/25/2022]
Abstract
Neuropathic and postoperative pain are clinical conditions that impair the patient's quality of life. The current pharmacotherapy of both painful states is ineffective and accompanied by several side effects. In order to develop new therapeutics targets, the secondary metabolites of plants have been extensively studied. Acmella oleracea ("jambu") is a native plant from the Amazon region and rich in alkylamides, bioactive compounds responsible for inducing anesthetic and chemesthetic sensations. We previously demonstrated that the intraplantar administration of an hexanic fraction (HF) rich in alkylamides from jambu and the synthetic isobutylalkyl amide (IBA) at 0.1 μg/20 μL can promote antinociceptive and anti-inflammatory effects. Thus, this study aimed to evaluate the local effect of HF and IBA (0.1 μg/20 μL) on neuropathic (partial sciatic nerve ligation, PSNL) and postoperative pain (plantar incision surgery, PIS) models in mice. Seven days after the PSNL, the mechanical (von Frey test) and cold (acetone-evoked evaporative cooling) allodynia, and digital gait parameters were analyzed. The intraplantar HF and IBA treatments attenuated the mechanical and cold allodynia as well as the static (max. Contact and print area) and dynamic (stand duration) parameters of digital gait analyses. On the day after PIS, the mechanical allodynia, heat hyperalgesia (hot plate, 52 ± 0.1°C), and spontaneous nociception scores were evaluated. Topical treatment with HF reduced the mechanical allodynia, heat hyperalgesia, and spontaneous nociception scores. In contrast, IBA treatment only partially reduced the mechanical allodynia. In summary, the local treatment with HF was effective on both neuropathic and postoperative pain, as opposed to IBA, which only had an effect on neuropathic pain.
Collapse
Affiliation(s)
| | | | - Daniele Maria-Ferreira
- Department of Pharmacology, Federal University of Parana, Curitiba, Brazil; Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Adamara Machado Nascimento
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, Brazil; Multidisciplinary Center, Federal University of Acre, Cruzeiro do Sul, Brazil
| | - Thales Ricardo Cipriani
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, Brazil
| | - Lauro Mera de Souza
- Instituto de Pesquisa Pelé Pequeno Príncipe, Faculdades Pequeno Príncipe, Curitiba, Brazil
| | - Pierangelo Geppetti
- Department of Health Sciences, Section of Clinical Pharmacology and Oncology, University of Florence, Florence, Italy
| | | |
Collapse
|
25
|
Chou MY, Hu MC, Chen PY, Hsu CL, Lin TY, Tan MJ, Lee CY, Kuo MF, Huang PH, Wu VC, Yang SH, Fan PC, Huang HY, Akbarian S, Loo TH, Stewart CL, Huang HP, Gau SSF, Huang HS. RTL1/PEG11 imprinted in human and mouse brain mediates anxiety-like and social behaviors and regulates neuronal excitability in the locus coeruleus. Hum Mol Genet 2022; 31:3161-3180. [PMID: 35567414 PMCID: PMC9476620 DOI: 10.1093/hmg/ddac110] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/01/2022] [Accepted: 05/09/2022] [Indexed: 11/24/2022] Open
Abstract
RTL1/PEG11, which has been associated with anxiety disorders, is a retrotransposon-derived imprinted gene in the placenta. However, imprinting patterns and functions of RTL1 in the brain have not been well-investigated. We found Rtl1 was paternally, but not maternally, expressed in brain stem, thalamus, and hypothalamus of mice, and imprinting status of RTL1 was maintained in human brain. Paternal Rtl1 knockout (Rtl1m+/p-) mice had higher neonatal death rates due to impaired suckling, and low body weights beginning on embryonic day 16.5. High paternal expression of Rtl1 was detected in the locus coeruleus (LC) and Rtl1m+/p- mice showed an increased delay in time of onset for action potentials and inward currents with decreased neuronal excitability of LC neurons. Importantly, Rtl1m+/p- mice exhibited behaviors associated with anxiety, depression, fear-related learning and memory, social dominance, and low locomotor activity. Taken together, our findings demonstrate RTL1 is imprinted in brain, mediates emotional and social behaviors, and regulates excitability in LC neurons.
Collapse
Affiliation(s)
- Ming-Yi Chou
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Meng-Chuen Hu
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Pin-Yu Chen
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chi-Lin Hsu
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Ting-Yu Lin
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Mao-Jia Tan
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Chih-Yu Lee
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Meng-Fai Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 10051, Taiwan
| | - Pei-Hsin Huang
- Department of Pathology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Vin-Cent Wu
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Shih-Hung Yang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 10051, Taiwan
| | - Pi-Chuan Fan
- Department of Pediatrics, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Hsin-Yi Huang
- Department of Pathology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, NY, 10029, USA
| | - Tsui-Han Loo
- A*STAR Skin Research Labs, Agency for Science, Technology and Research, 138632, Singapore
| | - Colin L Stewart
- A*STAR Skin Research Labs, Agency for Science, Technology and Research, 138632, Singapore
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Susan Shur-Fen Gau
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.,Department of Psychiatry, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| | - Hsien-Sung Huang
- Graduate Institute of Brain and Mind Sciences, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
| |
Collapse
|
26
|
Licero J, Illan MS, Descorbeth M, Cordero K, Figueroa JD, De Leon M. Fatty acid-binding protein 4 (FABP4) inhibition promotes locomotor and autonomic recovery in rats following spinal cord injury. J Neurotrauma 2022; 39:1099-1112. [PMID: 35297679 PMCID: PMC9347423 DOI: 10.1089/neu.2021.0346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The inflammatory response associated with traumatic spinal cord injury (SCI) contributes to locomotor and sensory impairments. Pro-inflammatory (M1) macrophages/microglia (MφMG) are the major cellular players in this response as they promote chronic inflammation resulting in injury expansion and tissue damage. Fatty Acid-Binding Protein 4 (FABP4) promotes M1 MφMG differentiation; however, it is unknown if FABP4 also plays a role in the etiology of SCI. The present study investigates whether FABP4's gene expression influences functional recovery following SCI. Analysis of qPCR data shows a robust induction of FABP4 mRNA (>100 fold) in rats subjected to a T9-T10 contusion injury compared to control. Western blot experiments reveal significant upregulation of FABP4 protein at the injury epicenter, and immunofluorescence analysis identifies this upregulation occurs in CD11b+ MφMG. Furthermore, upregulation of FABP4 gene expression correlates with PPARγ downregulation, inactivation of Iκβα, and the activation of the NF-κB pathway. Analysis of locomotor recovery using the Basso-Beattie-Bresnahan's (BBB) locomotor scale and the CatWalk gait analysis system shows that injured rats treated with FABP4 inhibitor BMS309403 have significant improvements in locomotion compared to vehicle controls. Additionally, inhibitor-treated rats exhibit enhanced autonomic bladder reflex recovery. Immunofluorescence experiments also show the administration of the FABP4 inhibitor increases the number of CD163+ and Liver Arginase+ M2 MφMG within the epicenter and penumbra of the injured spinal cord 28 dpi. These findings show that FABP4 may significantly exacerbate locomotor and sensory impairments during SCI by modulating macrophage/microglial activity.
Collapse
Affiliation(s)
- Jenniffer Licero
- Loma Linda University, Center for Health Disparities and Molecular Medicine, 142 Mortensen Hall, 11085 Campus St, Loma Linda, California, United States, 92354;
| | - Miguel S Illan
- Loma Linda University, Center for Health Disparities and Molecular Medicine, 142 Mortensen Hall, 11085 Campus St, Loma Linda, California, United States, 92354;
| | - Magda Descorbeth
- Loma Linda University, Center for Health Disparities and Molecular Medicine, Loma Linda, California, United States;
| | - Kathia Cordero
- Loma Linda University, Center for Health Disparities and Molecular Medicine, Loma Linda, California, United States;
| | - Johnny D Figueroa
- Loma Linda University, Center for Health Disparities and Molecular Medicine, Loma Linda, California, United States;
| | - Marino De Leon
- Loma Linda University, Center for Health Disparities and Molecular Medicine, 142 Mortensen Hall, 11085 Campus St, Loma Linda, California, United States, 92354;
| |
Collapse
|
27
|
Samejima S, Ievins AM, Boissenin A, Tolley NM, Khorasani A, Mondello SE, Moritz CT. Automated lever task with minimum antigravity movement for rats with cervical spinal cord injury. J Neurosci Methods 2022; 366:109433. [PMID: 34863839 DOI: 10.1016/j.jneumeth.2021.109433] [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: 07/15/2021] [Revised: 10/31/2021] [Accepted: 11/28/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Although there is currently no cure for paralysis due to spinal cord injury (SCI), the highest treatment priority is restoring arm and hand function for people with cervical SCI. Preclinical animal models provide an opportunity to test innovative treatments, but severe cervical injury models require significant time and effort to assess responses to novel interventions. Moreover, there is no behavioral task that can assess forelimb movement in rats with severe cervical SCI unable to perform antigravity movements. NEW METHOD We developed a novel lever pressing task for rats with severe cervical SCI. We employed an automated adaptive algorithm to train animals using open-source software and commercially available hardware. We found that using the adaptive training required only 13.3 ± 2.5 training days to achieve behavioral proficiency. The lever press task could quantify immediate and long-term improvements in severely impaired forelimb function effectively. This behavior platform has potential to facilitate rehabilitative training and assess effects of therapeutic modalities following SCI. COMPARISON WITH EXISTING METHODS There is no existing assessment aiming to quantify forelimb extension movement in rodents without function against gravity. We found that the new lever press task in the antigravity position could assess the severity of cervical SCI as well as the compensatory movement in the proximal forelimb less affected by the injury. CONCLUSIONS This study demonstrates that the new behavioral task is capable of tracking the functional changes with various therapies in rats with severe forelimb impairments in a cost- and time-efficient manner.
Collapse
Affiliation(s)
- Soshi Samejima
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States; Department of Electrical & Computer Engineering, University of Washington, Seattle, WA, United States; UW Institute for Neural Engineering, University of Washington, Seattle, WA, United States; The Center for Neurotechnology, University of Washington, Seattle, WA, United States
| | - Aiva M Ievins
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States; Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
| | - Adrien Boissenin
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States
| | - Nicholas M Tolley
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States
| | - Abed Khorasani
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States
| | - Sarah E Mondello
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States
| | - Chet T Moritz
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, United States; Department of Electrical & Computer Engineering, University of Washington, Seattle, WA, United States; Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States; UW Institute for Neural Engineering, University of Washington, Seattle, WA, United States; The Center for Neurotechnology, University of Washington, Seattle, WA, United States; Department of Physiology & Biophysics, University of Washington, Seattle, WA, United States.
| |
Collapse
|
28
|
McDonald BZ, Gee CC, Kievit FM. The Nanotheranostic Researcher’s Guide for Use of Animal Models of Traumatic Brain Injury. JOURNAL OF NANOTHERANOSTICS 2021; 2:224-268. [PMID: 35655793 PMCID: PMC9159501 DOI: 10.3390/jnt2040014] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Traumatic brain injury (TBI) is currently the leading cause of injury-related morbidity and mortality worldwide, with an estimated global cost of USD 400 billion annually. Both clinical and preclinical behavioral outcomes associated with TBI are heterogeneous in nature and influenced by the mechanism and frequency of injury. Previous literature has investigated this relationship through the development of animal models and behavioral tasks. However, recent advancements in these methods may provide insight into the translation of therapeutics into a clinical setting. In this review, we characterize various animal models and behavioral tasks to provide guidelines for evaluating the therapeutic efficacy of treatment options in TBI. We provide a brief review into the systems utilized in TBI classification and provide comparisons to the animal models that have been developed. In addition, we discuss the role of behavioral tasks in evaluating outcomes associated with TBI. Our goal is to provide those in the nanotheranostic field a guide for selecting an adequate TBI animal model and behavioral task for assessment of outcomes to increase research in this field.
Collapse
|
29
|
Wang X, Zang J, Yang Y, Lu S, Guan Q, Ye D, Wang Z, Zhou H, Li K, Wang Q, Wu Y, Luan Z. Transplanted Human Oligodendrocyte Progenitor Cells Restore Neurobehavioral Deficits in a Rat Model of Preterm White Matter Injury. Front Neurol 2021; 12:749244. [PMID: 34858313 PMCID: PMC8631304 DOI: 10.3389/fneur.2021.749244] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/18/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Preterm white matter injury (PWMI) is a common brain injury and a leading cause of life-long neurological deficits in premature infants; however, no effective treatment is available yet. This study aimed to investigate the fate and effectiveness of transplanted human oligodendrocyte progenitor cells (hOPCs) in a rat model of PWMI. Methods: Hypoxia-ischemia was induced in rats at postnatal day 3, and hOPCs (6 × 105 cells/5 μL) were intracerebroventricularly transplanted at postnatal day 7. Neurobehavior was assessed 12 weeks post-transplant using the CatWalk test and Morris water maze test. Histological analyses, as well as immunohistochemical and transmission electron microscopy, were performed after transcardial perfusion. Results: Transplanted hOPCs survived for 13 weeks in PWMI brains. They were widely distributed in the injured white matter, and migrated along the corpus callosum to the contralateral hemisphere. Notably, 82.77 ± 3.27% of transplanted cells differentiated into mature oligodendrocytes, which produced myelin around the axons. Transplantation of hOPCs increased the fluorescence intensity of myelin basic protein and the thickness of myelin sheaths as observed in immunostaining and transmission electron microscopy, while it reduced white matter atrophy at the level of gross morphology. With regard to neurobehavior, the CatWalk test revealed improved locomotor function and inter-paw coordination after transplantation, and the cognitive functions of hOPC-transplanted rats were restored as revealed by the Morris water maze test. Conclusions: Myelin restoration through the transplantation of hOPCs led to neurobehavioral improvements in PWMI rats, suggesting that transplanting hOPCs may provide an effective and promising therapeutic strategy in children with PWMI.
Collapse
Affiliation(s)
- Xiaohua Wang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China.,Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Jing Zang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Yinxiang Yang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Siliang Lu
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Qian Guan
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Dou Ye
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Zhaoyan Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Haipeng Zhou
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Ke Li
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Qian Wang
- Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| | - Youjia Wu
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, China
| | - Zuo Luan
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Pediatrics, The Sixth Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
30
|
Liu YJ, Chen XF, Zhou LP, Rao F, Zhang DY, Wang YH. A nerve conduit filled with Wnt5a-loaded fibrin hydrogels promotes peripheral nerve regeneration. CNS Neurosci Ther 2021; 28:145-157. [PMID: 34729936 PMCID: PMC8673702 DOI: 10.1111/cns.13752] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 10/09/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Aims Peripheral nerve injury is a significant clinical problem with a substantial impact on quality of life, for which no optimal treatment has been found. This study aimed to analyze the effect and mechanism of Wnt5a‐loaded fibrin hydrogel on a 10‐mm rat sciatic nerve defect. Methods The Wnt5a‐loaded fibrin hydrogel was synthesized by mixing a Wnt5a solution with thrombin and fibrinogen solutions. The loading capacity and release profile of Wnt5a‐loaded fibrin hydrogel and the effect of Wnt5a on Schwann cells were evaluated in vitro. We also assessed the in vivo repair status via histological analysis of the regenerative nerve and gastrocnemius muscle, electrophysiological examination, gait analysis, and muscle wet weight. Results We developed a nerve conduit filled with Wnt5a‐loaded fibrin hydrogel (Fn) as a sustained‐release system to repair a 10‐mm rat sciatic nerve defect. In vitro, Wnt5a could promote SC proliferation and the gene expression of vascular endothelial growth factor (VEGF), nerve growth factor (NGF), and cholinergic neurotrophic factor (CNTF), as well as the protein secretion of VEGF and NGF. In vivo, the Wnt5a/Fn group was superior to the hollow, fibrin hydrogel, and Wnt5a groups in terms of axonal growth, myelination, electrophysiological recovery, target organ innervation, and motor function recovery 12 weeks after the operation. Conclusion The Wnt5a/Fn nerve conduit can promote peripheral nerve defect regeneration, with potential clinical applications. The mechanism for this may be the facilitation of multiple neurotrophin secretion, combining vascularization and neurotrophic growth cues.
Collapse
Affiliation(s)
- Yi-Jun Liu
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, China.,Department of Foot and Ankle Surgery, Center for Orthopaedic Surgery, the Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xiao-Feng Chen
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, China.,Department of Orthopedics and Trauma, Key Laboratory of Trauma and Neural Regeneration (Ministry of Education/ Peking University), Beijing, China
| | - Li-Ping Zhou
- Beijing Key Laboratory for Bioengineering and Sensing Technology, Daxing Research Institute, School of Chemistry & Biological Engineering, University of Science & Technology Beijing, Beijing, China
| | - Feng Rao
- Trauma Medicine Center, Peking University People's Hospital, Beijing, China
| | - Dian-Ying Zhang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, China.,Department of Orthopedics and Trauma, Key Laboratory of Trauma and Neural Regeneration (Ministry of Education/ Peking University), Beijing, China
| | - Yan-Hua Wang
- Department of Orthopedics and Trauma, Peking University People's Hospital, Beijing, China.,Department of Orthopedics and Trauma, Key Laboratory of Trauma and Neural Regeneration (Ministry of Education/ Peking University), Beijing, China
| |
Collapse
|
31
|
Pitzer C, Kurpiers B, Eltokhi A. Gait performance of adolescent mice assessed by the CatWalk XT depends on age, strain and sex and correlates with speed and body weight. Sci Rep 2021; 11:21372. [PMID: 34725364 PMCID: PMC8560926 DOI: 10.1038/s41598-021-00625-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/06/2021] [Indexed: 11/17/2022] Open
Abstract
The automatization of behavioral tests assessing motor activity in rodent models is important for providing robust and reproducible results and evaluating new therapeutics. The CatWalk system is an observer-independent, automated and computerized technique for the assessment of gait performance in rodents. This method has previously been used in adult rodent models of CNS-based movement disorders such as Parkinson's and Huntington's diseases. As motor and gait abnormalities in neuropsychiatric disorders are observed during infancy and adolescence, it became important to validate the CatWalk XT in the gait analysis of adolescent mice and unravel factors that may cause variations in gait performance. Three adolescent wild-type inbred mouse strains, C57BL/6N, DBA/2 and FVB/N, were tested using the CatWalk XT (Version 10.6) for suitable detection settings to characterize several gait parameters at P32 and P42. The same detection settings being suitable for C57BL/6N and DBA/2 mice allowed a direct comparison between the two strains. On the other hand, due to their increased body weight and size, FVB/N mice required different detection settings. The CatWalk XT reliably measured the temporal, spatial, and interlimb coordination parameters in the investigated strains during adolescence. Additionally, significant effects of sex, development, speed and body weight within each strain confirmed the sensitivity of motor and gait functions to these factors. The CatWalk gait analysis of rodents during adolescence, taking the effect of age, strain, sex, speed and body weight into consideration, will decrease intra-laboratory discrepancies and increase the face validity of rodent models of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany.
| | - Barbara Kurpiers
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, USA.
| |
Collapse
|
32
|
Distribution, fine structure, and three-dimensional innervation of lamellar corpuscles in rat plantar skin. Cell Tissue Res 2021; 386:477-490. [PMID: 34562148 DOI: 10.1007/s00441-021-03525-5] [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: 12/09/2020] [Accepted: 09/09/2021] [Indexed: 10/20/2022]
Abstract
Lamellar corpuscles function as mechanoreceptors in the skin, composed of axon terminals and lamellae constructed by terminal Schwann cells. They are classified into Pacinian, Meissner, and simple corpuscles based on histological criteria. Lamellar corpuscles in rat dermal papilla cells have been reported; however, the morphological aspects have yet to be thoroughly investigated. In the present study, we analyzed the enzyme activity, distribution, fine structure, and three-dimensional innervation of lamellar corpuscles in rat plantar skin. The lamellar corpuscles exhibiting non-specific cholinesterase were densely distributed in rat footpads, evident as notable skin elevations, especially at the apex, the highest portion of the ridges in each footpad. In contrast, only a few lamellar corpuscles were found in other plantar skin areas. Lamellar corpuscle was considered composed of a flat axon terminal Schwann cell lamellae, which were roughly concentrically arranged in the dermal papilla. These histological characteristics correspond to those of the simple corpuscle. Moreover, the axon tracing method revealed that one trunk axon innervated several simple corpuscles. The territory of the trunk axons overlapped with each other. Finally, the animals' footprints were analyzed. During the pausing and walking phases, footpads are often in contact with the floor. These results demonstrate that the type of lamellar corpuscles in the dermal papillae of rat plantar skin is a simple corpuscle and implies that their distribution pattern in the plantar skin is convenient for efficient sensing and transmission of mechanical stimuli from the ground.
Collapse
|
33
|
Isvoranu G, Manole E, Neagu M. Gait Analysis Using Animal Models of Peripheral Nerve and Spinal Cord Injuries. Biomedicines 2021; 9:1050. [PMID: 34440252 PMCID: PMC8392642 DOI: 10.3390/biomedicines9081050] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 11/17/2022] Open
Abstract
The present review discusses recent data regarding rodent models of spinal cord and peripheral nerve injuries in terms of gait analysis using the CatWalk system (CW), an automated and exceptionally reliable system for assessing gait abnormalities and motor coordination. CW is a good tool for both studying improvements in the walking of animals after suffering a peripheral nerve and spinal cord lesion and to select the best therapies and procedures after tissue destruction, given that it provides objective and quantifiable data. Most studies using CW for gait analysis that were published in recent years focus on injuries inflicted in the peripheral nerve, spinal cord, and brain. CW has been used in the assessment of rodent motor function through high-resolution videos, whereby specialized software was used to measure several aspects of the animal's gait, and the main characteristics of the automated system are presented here. CW was developed to assess footfall and gait changes, and it can calculate many parameters based on footprints and time. However, given the multitude of parameters, it is necessary to evaluate which are the most important under the employed experimental circumstances. By selecting appropriate animal models and evaluating peripheral nerve and spinal cord lesion regeneration using standardized methods, suggestions for new therapies can be provided, which represents the translation of this methodology into clinical application.
Collapse
Affiliation(s)
- Gheorghita Isvoranu
- Husbandry Unit, Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania;
| | - Emilia Manole
- Laboratory of Cellular Biology, Neuroscience and Experimental Myology, Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Pathology Department, Colentina University Hospital, 19-21 Sos. Stefan cel Mare, 020125 Bucharest, Romania;
| | - Monica Neagu
- Pathology Department, Colentina University Hospital, 19-21 Sos. Stefan cel Mare, 020125 Bucharest, Romania;
- Immunology Laboratory, Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania
- Doctoral School of Biology, Faculty of Biology, University of Bucharest, 91-93 Splaiul Independentei, 050095 Bucharest, Romania
| |
Collapse
|
34
|
Garrick JM, Costa LG, Cole TB, Marsillach J. Evaluating Gait and Locomotion in Rodents with the CatWalk. Curr Protoc 2021; 1:e220. [PMID: 34370398 PMCID: PMC8363132 DOI: 10.1002/cpz1.220] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Motor deficits can significantly affect the completion of daily life activities and have a negative impact on quality of life. Consequently, motor function is an important behavioral endpoint to measure for in vivo pathophysiologic studies in a variety of research areas, such as toxicant exposure, drug development, disease characterization, and transgenic phenotyping. Evaluation of motor function is also critical to the interpretation of cognitive behavioral assays, as many rely on intact motor abilities to derive meaningful data. As such, gait analysis is an important component of behavioral research and can be achieved by manual or video-assisted methods. Manual gait analysis methods, however, are prone to observer bias and are unable to capture many critical parameters. In contrast, automated video-assisted gait analysis can quickly and reliably assess gait and locomotor abnormalities that were previously difficult to collect manually. Here, we describe the evaluation of gait and locomotion in rodents using the automated Noldus CatWalk XT system. We include a step-by-step guide for running an experiment using the CatWalk XT system and discuss theory and considerations when evaluating rodent gait. The protocol and discussion provided here act as a supplemental resource to the manual for this commercially available system and can assist CatWalk users in their experimental design and implementation. © 2021 Wiley Periodicals LLC.
Collapse
Affiliation(s)
- Jacqueline M. Garrick
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| | - Lucio G. Costa
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
- Dept. of Medicine and Surgery, University of Parma, Italy
| | - Toby B. Cole
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
- Center on Human Development and Disabilities, University of Washington, United States
| | - Judit Marsillach
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, United States
| |
Collapse
|
35
|
Jia D, Sun Y, Liu G, Xue M, Huang Z, Huang C. Trifluoro-icaritin alleviates mechanical hypersensitivity and improves motor coordination and balance in rats with spared nerve injury-induced neuropathic pain. Neurosci Lett 2021; 761:136125. [PMID: 34302890 DOI: 10.1016/j.neulet.2021.136125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/21/2021] [Accepted: 07/09/2021] [Indexed: 12/25/2022]
Abstract
Neuropathic pain is still one of the unsolved public health problems worldwide. Although the current reagents can attenuate neuropathic pain to a certain extent, their clinical application is very limited owing to larger toxicity and serious side effects. Trifluoro-icaritin (ICTF) has been documented to possess profound anti-inflammatory and neuroprotective activities, but whether ICTF exerts an anti-nociceptive effect on neuropathic pain remains unknown. Here, a rat model of spared nerve injury (SNI)-induced neuropathic pain was used. SNI rats were administrated with ICTF (i.p.) once daily lasting for 21 days, and subsequently the pain-related behaviors were evaluated by applying mechanical or thermal pain threshold, CatWalk gait parameter, and rotarod test on day 1 before and day 1, 3, 7, 10, 14, and 21 after SNI surgery, respectively. The results showed that ICTF (0.5 mg/kg, 1.5 mg/kg, and 5.0 mg/kg, i.p.) treatment alleviated SNI-induced mechanical allodynia but not thermal hyperalgesia in a dose-dependent manner. After administration of ICTF at the most effective dose of 5.0 mg/kg to SNI rats, CatWalk gait analysis revealed that ICTF not only significantly enhanced gait parameters including max contact max intensity, max intensity, print area, and stand time but also decreased the swing time; Rotarod test further exhibited that ICTF could effectively prolong the time on rod and increase the rotating speed in SNI rats. Additionally, following ICTF (5.0 mg/kg) treatment of SNI rats for 21 consecutive days, the max contact max intensity was found to be positively correlated with the rotating speed. Taken together, ICTF successfully ameliorates mechanical hypersensitivity and improves the motor coordination and balance in SNI rats, suggesting that ICTF may be exploited as a potential candidate in the management of neuropathic pain.
Collapse
Affiliation(s)
- Dandan Jia
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, PR China
| | - Yalan Sun
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, PR China
| | - Guangsen Liu
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, PR China
| | - Meng Xue
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, PR China
| | - Zhihua Huang
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, PR China; Pain Medicine Research Institute, Gannan Medical University, Ganzhou 341000, PR China
| | - Cheng Huang
- Department of Physiology, School of Basic Medicine Sciences, Gannan Medical University, Ganzhou 341000, PR China; Pain Medicine Research Institute, Gannan Medical University, Ganzhou 341000, PR China.
| |
Collapse
|
36
|
Mah KM, Torres-Espín A, Hallworth BW, Bixby JL, Lemmon VP, Fouad K, Fenrich KK. Automation of training and testing motor and related tasks in pre-clinical behavioural and rehabilitative neuroscience. Exp Neurol 2021; 340:113647. [PMID: 33600814 PMCID: PMC10443427 DOI: 10.1016/j.expneurol.2021.113647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/25/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Testing and training animals in motor and related tasks is a cornerstone of pre-clinical behavioural and rehabilitative neuroscience. Yet manually testing and training animals in these tasks is time consuming and analyses are often subjective. Consequently, there have been many recent advances in automating both the administration and analyses of animal behavioural training and testing. This review is an in-depth appraisal of the history of, and recent developments in, the automation of animal behavioural assays used in neuroscience. We describe the use of common locomotor and non-locomotor tasks used for motor training and testing before and after nervous system injury. This includes a discussion of how these tasks help us to understand the underlying mechanisms of neurological repair and the utility of some tasks for the delivery of rehabilitative training to enhance recovery. We propose two general approaches to automation: automating the physical administration of behavioural tasks (i.e., devices used to facilitate task training, rehabilitative training, and motor testing) and leveraging the use of machine learning in behaviour analysis to generate large volumes of unbiased and comprehensive data. The advantages and disadvantages of automating various motor tasks as well as the limitations of machine learning analyses are examined. In closing, we provide a critical appraisal of the current state of automation in animal behavioural neuroscience and a prospective on some of the advances in machine learning we believe will dramatically enhance the usefulness of these approaches for behavioural neuroscientists.
Collapse
Affiliation(s)
- Kar Men Mah
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Abel Torres-Espín
- Brain and Spinal Injury Center, Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Ben W Hallworth
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Mechanical Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - John L Bixby
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA; Department of Molecular & Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL 33136, USA
| | - Vance P Lemmon
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami, Miami, FL 33136, USA
| | - Karim Fouad
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Physical Therapy, 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.
| |
Collapse
|
37
|
Plesia M, Stevens OA, Lloyd GR, Kendall CA, Coldicott I, Kennerley AJ, Miller G, Shaw PJ, Mead RJ, Day JCC, Alix JJP. In Vivo Fiber Optic Raman Spectroscopy of Muscle in Preclinical Models of Amyotrophic Lateral Sclerosis and Duchenne Muscular Dystrophy. ACS Chem Neurosci 2021; 12:1768-1776. [PMID: 33950665 PMCID: PMC8154326 DOI: 10.1021/acschemneuro.0c00794] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/30/2021] [Indexed: 12/13/2022] Open
Abstract
Neuromuscular diseases result in muscle weakness, disability, and, in many instances, death. Preclinical models form the bedrock of research into these disorders, and the development of in vivo and potentially translational biomarkers for the accurate identification of disease is crucial. Spontaneous Raman spectroscopy can provide a rapid, label-free, and highly specific molecular fingerprint of tissue, making it an attractive potential biomarker. In this study, we have developed and tested an in vivo intramuscular fiber optic Raman technique in two mouse models of devastating human neuromuscular diseases, amyotrophic lateral sclerosis, and Duchenne muscular dystrophy (SOD1G93A and mdx, respectively). The method identified diseased and healthy muscle with high classification accuracies (area under the receiver operating characteristic curves (AUROC): 0.76-0.92). In addition, changes in diseased muscle over time were also identified (AUROCs 0.89-0.97). Key spectral changes related to proteins and the loss of α-helix protein structure. Importantly, in vivo recording did not cause functional motor impairment and only a limited, resolving tissue injury was seen on high-resolution magnetic resonance imaging. Lastly, we demonstrate that ex vivo muscle from human patients with these conditions produced similar spectra to those observed in mice. We conclude that spontaneous Raman spectroscopy of muscle shows promise as a translational research tool.
Collapse
Affiliation(s)
- Maria Plesia
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | - Oliver A. Stevens
- Interface
Analysis Centre, School of Physics, University
of Bristol, Bristol BS8 1TL, UK
| | - Gavin R. Lloyd
- Phenome
Centre Birmingham, University of Birmingham, Birmingham B15 2TT, UK
- Biophotonics
Research Unit, Gloucestershire Hospitals
NHS Foundation Trust, Gloucester GL1 3NN, UK
| | - Catherine A. Kendall
- Biophotonics
Research Unit, Gloucestershire Hospitals
NHS Foundation Trust, Gloucester GL1 3NN, UK
| | - Ian Coldicott
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
| | | | - Gaynor Miller
- Department
of Oncology and Metabolism, University of
Sheffield, Sheffield S10 2RX, UK
| | - Pamela J. Shaw
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Cross-Faculty
Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| | - Richard J. Mead
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Cross-Faculty
Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| | - John C. C. Day
- Interface
Analysis Centre, School of Physics, University
of Bristol, Bristol BS8 1TL, UK
| | - James J. P. Alix
- Sheffield
Institute for Translational Neuroscience, University of Sheffield, Sheffield S10 2HQ, UK
- Cross-Faculty
Neuroscience Institute, University of Sheffield, Sheffield S10 2HQ, UK
| |
Collapse
|
38
|
Islam R, Drecun S, Varga BV, Vonderwalde I, Siu R, Nagy A, Morshead CM. Transplantation of Human Cortically-Specified Neuroepithelial Progenitor Cells Leads to Improved Functional Outcomes in a Mouse Model of Stroke. Front Cell Neurosci 2021; 15:654290. [PMID: 33994947 PMCID: PMC8116536 DOI: 10.3389/fncel.2021.654290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/08/2021] [Indexed: 12/02/2022] Open
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Current therapeutic options are limited in terms of their time for implementation and efficacy in promoting recovery. Cell transplantation has been shown to have promise in several animal models however significant challenges remain, including the optimal source of cells to promote neural repair. Here, we report on the use of a population of human ESC derived, cortically specified, neuroepithelial precursor cells (cNEPs) that are neurally restricted in their lineage potential. CNEPs have the potential to give rise to mature neural cell types following transplantation, including neurons, astrocytes and oligodendrocytes. With a view towards translation, we sought to determine whether this human cell source was effective in promoting improved functional outcomes following stroke. Undifferentiated cNEPs were transplanted in a pre-clinical endothelin-1 (ET-1) model of ischemic motor cortical stroke in immunocompromised SCID-beige mice and cellular and functional outcomes were assessed. We demonstrate that cNEP transplantation in the acute phase (4 days post-stroke) improves motor function as early as 20 days post-stroke, compared to stroke-injured, non-transplanted mice. At the time of recovery, a small fraction (<6%) of the transplanted cNEPs are observed within the stroke injury site. The surviving cells expressed the immature neuronal marker, doublecortin, with no differentiation into mature neural phenotypes. At longer survival times (40 days), the majority of recovered, transplanted mice had a complete absence of surviving cNEPS. Hence, human cNEPs grafted at early times post-stroke support the observed functional recovery following ET-1 stroke but their persistence is not required, thereby supporting a by-stander effect rather than cell replacement.
Collapse
Affiliation(s)
- Rehnuma Islam
- Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Stasja Drecun
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Balazs V. Varga
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Ilan Vonderwalde
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Ricky Siu
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Andras Nagy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Cindi M. Morshead
- Faculty of Medicine, Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
39
|
Rui F, Jiawei K, Yuntao H, Xinran L, Jiani H, Ruixue M, Rui L, Na Z, Meihong X, Yong L. Undenatured type II collagen prevents and treats osteoarthritis and motor function degradation in T2DM patients and db/db mice. Food Funct 2021; 12:4373-4391. [PMID: 33890588 DOI: 10.1039/d0fo03011b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Osteoarthritis (OA) has been scarcely researched among patients with diabetes mellitus. This study aims to confirm the preventive and therapeutic effects of undenatured type II collagen (UC II) on OA in aging db/db mice and in patients with T2DM. Firstly, aging db/db mice were randomly assigned to three groups: the UC II intervention (UC II) group, old model (OM) group and positive control group. Meanwhile db/m mice and young db/db mice were used as the normal control and young control groups, respectively. Secondly, fifty-five T2DM patients diagnosed with knee OA were randomly assigned to two groups: UC-II and placebo control groups. After a three-month intervention in both mice and T2DM patients, the subjects' gait and physical activities were assessed and the serum biomarkers including inflammatory cytokines, oxidative stress factors and matrix metalloproteinases (MMPs) were measured. Compared with the OM group mice, those in the UC II group showed a significantly greater superiority in terms of motor functions including the movement trajectories area (163.25 ± 20.3 vs. 78.52 ± 20.14 cm2), the tremor index (0.42 vs. 1.23), standing time (left hind: 0.089 ± 0.03 vs. 0.136 ± 0.04 s), swing (right front: 0.12 ± 0.02 vs. 0.216 ± 0.02 s), stride length (right hind: 7.2 ± 0.9 vs. 5.7 ± 1.1 cm), step cycle (right hind: 0.252 ± 0.05 vs. 0.478 ± 0.11 s) and cadence (14.12 ± 2.7 vs. 7.35 ± 4.4 steps per s). In addition, the levels of IL-4, IL-10, CTX- II and TGF-β in the UC II group were 1.74, 2.23, 1.67 and 1.84 times higher than those in the OM group, respectively, while the levels of MMP-3 and MMP-13 in the UC II group were half those in the OM group. Correspondingly, UC II intervention significantly decreased the scores of pain, stiffness and physical function (p < 0.05), whereas the 6 MWT and total MET distances in the UC II group increased remarkably (p < 0.05). After a three-month period of intervention, the varus angle significantly decreased from 4.6 ± 2.0° to 3.0 ± 1.4° and the knee flexion range obviously increased from 57.9 ± 14.0° to 66.9 ± 10.4°. Importantly, the declining trend in the levels of hs-CRP and MDA and the incremental trend in the SOD level were consistent in the db/db mice and OA patients following UC II administration.
Collapse
Affiliation(s)
- Fan Rui
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Combination of Defined CatWalk Gait Parameters for Predictive Locomotion Recovery in Experimental Spinal Cord Injury Rat Models. eNeuro 2021; 8:ENEURO.0497-20.2021. [PMID: 33593735 PMCID: PMC7986542 DOI: 10.1523/eneuro.0497-20.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 12/27/2022] Open
Abstract
In many preclinical spinal cord injury (SCI) studies, assessment of locomotion recovery is key to understanding the effectiveness of the experimental intervention. In such rat SCI studies, the most basic locomotor recovery scoring system is a subjective observation of animals freely roaming in an open field, the Basso Beattie Bresnahan (BBB) score. In comparison, CatWalk is an automated gait analysis system, providing further parameter specifications. Although together the CatWalk parameters encompass gait, studies consistently report single parameters, which differ in significance from other behavioral assessments. Therefore, we believe no single parameter produced by the CatWalk can represent the fully-coordinated motion of gait. Typically, other locomotor assessments, such as the BBB score, combine several locomotor characteristics into a representative score. For this reason, we ranked the most distinctive CatWalk parameters between uninjured and SC injured rats. Subsequently, we combined nine of the topmost parameters into an SCI gait index score based on linear discriminant analysis (LDA). The resulting combination was applied to assess gait recovery in SCI experiments comprising of three thoracic contusions, a thoracic dorsal hemisection, and a cervical dorsal column lesion model. For thoracic lesions, our unbiased machine learning model revealed gait differences in lesion type and severity. In some instances, our LDA was found to be more sensitive in differentiating recovery than the BBB score alone. We believe the newly developed gait parameter combination presented here should be used in CatWalk gait recovery work with preclinical thoracic rat SCI models.
Collapse
|
41
|
Defining the relative impact of muscle versus Schwann cell denervation on functional recovery after delayed nerve repair. Exp Neurol 2021; 339:113650. [PMID: 33607079 DOI: 10.1016/j.expneurol.2021.113650] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 12/16/2022]
Abstract
Functional recovery following peripheral nerve injury worsens with increasing durations of delay prior to repair. From the time of injury until re-innervation occurs, denervated muscle undergoes progressive atrophy that limits the extent to which motor function can be restored. Similarly, Schwann cells (SC) in the distal nerve lacking axonal interaction progressively lose their capacity to proliferate and support regenerating axons. The relative contributions of these processes to diminished functional recovery is unclear. We developed a novel rat model to isolate the effects of SC vs. muscle denervation on functional recovery. Four different groups underwent the following interventions for 12 weeks prior to nerve transfer: 1) muscle denervation; 2) SC denervation; 3) muscle + SC denervation (negative control); 4) no denervation (positive control). Functional recovery was measured weekly using the stimulated grip strength testing (SGST). Animals were sacrificed 13 weeks post nerve transfer. Retrograde labeling was used to assess the number of motor neurons that regenerated their axons. Immunofluorescence was performed to evaluate target muscle re-innervation and atrophy, and to assess the phenotype of the SC within the distal nerve segment. Functional recovery in the muscle denervation and SC denervation groups mirrored that of the negative and positive control groups, respectively. The SC denervation group achieved better functional recovery, with a greater number of reinnervated motor endplates and less muscle atrophy, than the muscle denervation group. Retrograde labeling suggested a higher number of neurons contributing to muscle reinnervation in the muscle denervation group as compared to SC denervation (p > 0.05). The distal nerve segment in the muscle denervation group had a greater proportion of SCs expressing the proliferation marker Ki67 as compared to the SC denervation group (p < 0.05). Conversely, the SC denervation group had a higher percentage of senescent SCs expressing p16 as compared to the muscle denervation group (p < 0.05). The deleterious effects of muscle denervation are more consequential than the effects of SC denervation on functional recovery. The effects of 12 weeks of SC denervation on functional outcome were negligible. Future studies are needed to determine whether longer periods of SC denervation negatively impact functional recovery.
Collapse
|
42
|
Shen H, Gardner AM, Vyas J, Ishida R, Tawfik VL. Modeling Complex Orthopedic Trauma in Rodents: Bone, Muscle and Nerve Injury and Healing. Front Pharmacol 2021; 11:620485. [PMID: 33597884 PMCID: PMC7882733 DOI: 10.3389/fphar.2020.620485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/21/2020] [Indexed: 12/24/2022] Open
Abstract
Orthopedic injury can occur from a variety of causes including motor vehicle collision, battlefield injuries or even falls from standing. Persistent limb pain is common after orthopedic injury or surgery and presents a unique challenge, as the initiating event may result in polytrauma to bone, muscle, and peripheral nerves. It is imperative that we understand the tissue-specific and multicellular response to this unique type of injury in order to best develop targeted treatments that improve healing and regeneration. In this Mini Review we will first discuss current rodent models of orthopedic trauma/complex orthotrauma. In the second section, we will focus on bone-specific outcomes including imaging modalities, biomechanical testing and immunostaining for markers of bone healing/turnover. In the third section, we will discuss muscle-related pathology including outcome measures of fibrosis, muscle regeneration and tensile strength measurements. In the fourth section, we will discuss nervous system-related pathology including outcome measures of pain-like responses, both reflexive and non-reflexive. In all sections we will consider parallels between preclinical outcome measures and the functional and mechanistic findings of the human condition.
Collapse
Affiliation(s)
- Huaishuang Shen
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States.,Department of Orthopaedic Surgery, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Aysha M Gardner
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Juhee Vyas
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States
| | - Ryosuke Ishida
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States.,Department of Anesthesiology, Shimane University, Shimane, Japan
| | - Vivianne L Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University, Stanford, CA, United States.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
| |
Collapse
|
43
|
Heinzel JC, Oberhauser V, Keibl C, Swiadek N, Längle G, Frick H, Kolbenschlag J, Prahm C, Grillari J, Hercher D. Evaluation of Functional Recovery in Rats After Median Nerve Resection and Autograft Repair Using Computerized Gait Analysis. Front Neurosci 2021; 14:593545. [PMID: 33551723 PMCID: PMC7859340 DOI: 10.3389/fnins.2020.593545] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Computerized gait analysis is a common evaluation method in rat models of hind limb nerve injuries, but its use remains unpublished in models of segmental nerve injury of the forelimb. It was the aim of this work to investigate if computerized gait analysis is a feasible evaluation method in a rat model of segmental median nerve injury and autograft repair. Ten male Lewis rats underwent 7-mm resection of the right median nerve with immediate autograft repair. The left median nerve was resected without repair and served as an internal control. Animals were assessed for 12 weeks after surgery via CatWalk (CW) gait analysis every 2 weeks. Evaluation of motor recovery by means of the grasping test was performed weekly while electrophysiological measurements were performed at the end of the observation period. CW data were correlated with grasping strength at each post-operative time point. CW data were also correlated with electrophysiology using linear regression analysis. Principal component analysis was performed to identify clusters of outcome metrics. Recovery of motor function was observable 4 weeks after surgery, but grasping strength was significantly reduced (p < 0.01) compared to baseline values until post-operative week 6. In terms of sensory recovery, the pain-related parameter Duty Cycle showed significant (p < 0.05) recovery starting from post-operative week 8. The Print Area of the right paw was significantly (p < 0.05) increased compared to the left side starting from post-operative week 10. Various parameters of gait correlated significantly (p < 0.05) with mean and maximum grasping strength. However, only Stand Index showed a significant correlation with compound muscle action potential (CMAP) amplitude (p < 0.05). With this work, we prove that computerized gait analysis is a valid and feasible method to evaluate functional recovery after autograft repair of the rat median nerve. We were able to identify parameters such as Print Area, Duty Cycle, and Stand Index, which allow assessment of nerve regeneration. The course of these parameters following nerve resection without repair was also assessed. Additionally, external paw rotation was identified as a valid parameter to evaluate motor reinnervation. In summary, computerized gait analysis is a valuable additional tool to study nerve regeneration in rats with median nerve injury.
Collapse
Affiliation(s)
- Johannes C Heinzel
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Viola Oberhauser
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Claudia Keibl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Nicole Swiadek
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Gregor Längle
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Helen Frick
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Jonas Kolbenschlag
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Cosima Prahm
- Department of Hand, Plastic, Reconstructive and Burn Surgery, BG Trauma Center Tuebingen, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Biotechnology, Institute of Molecular Biotechnology, BOKU-University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
44
|
Huang ZH, Feng AY, Liu J, Zhou L, Zhou B, Yu P. Inhibitor of DNA binding 2 accelerates nerve regeneration after sciatic nerve injury in mice. Neural Regen Res 2021; 16:2542-2548. [PMID: 33907046 PMCID: PMC8374553 DOI: 10.4103/1673-5374.313054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Inhibitor of DNA binding 2 (Id2) can promote axonal regeneration after injury of the central nervous system. However, whether Id2 can promote axonal regeneration and functional recovery after peripheral nerve injury is currently unknown. In this study, we established a mouse model of bilateral sciatic nerve crush injury. Two weeks before injury, AAV9-Id2-3×Flag-GFP was injected stereotaxically into the bilateral ventral horn of lumbar spinal cord. Our results showed that Id2 was successfully delivered into spinal cord motor neurons projecting to the sciatic nerve, and the number of regenerated motor axons in the sciatic nerve distal to the crush site was increased at 2 weeks after injury, arriving at the tibial nerve and reinnervating a few endplates in the gastrocnemius muscle. By 1 month after injury, extensive neuromuscular reinnervation occurred. In addition, the amplitude of compound muscle action potentials of the gastrocnemius muscle was markedly recovered, and their latency was shortened. These findings suggest that Id2 can accelerate axonal regeneration, promote neuromuscular reinnervation, and enhance functional improvement following sciatic nerve injury. Therefore, elevating the level of Id2 in adult neurons may present a promising strategy for peripheral nerve repair following injury. The study was approved by the Experimental Animal Ethics Committee of Jinan University (approval No. 20160302003) on March 2, 2016.
Collapse
Affiliation(s)
- Zhong-Hai Huang
- Guangdong-Hong Kong-Macau Institute of Central Nervous System Regeneration; Ministry of Education Joint International Research Laboratory of Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Ai-Ying Feng
- Guangdong-Hong Kong-Macau Institute of Central Nervous System Regeneration; Ministry of Education Joint International Research Laboratory of Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Jing Liu
- Guangdong-Hong Kong-Macau Institute of Central Nervous System Regeneration; Ministry of Education Joint International Research Laboratory of Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Libing Zhou
- Guangdong-Hong Kong-Macau Institute of Central Nervous System Regeneration; Ministry of Education Joint International Research Laboratory of Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| | - Bing Zhou
- Interdisciplinary Innovation Institute of Medicine and Engineering, Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| | - Panpan Yu
- Guangdong-Hong Kong-Macau Institute of Central Nervous System Regeneration; Ministry of Education Joint International Research Laboratory of Central Nervous System Regeneration, Jinan University, Guangzhou, Guangdong Province, China
| |
Collapse
|
45
|
Heinzel J, Längle G, Oberhauser V, Hausner T, Kolbenschlag J, Prahm C, Grillari J, Hercher D. Use of the CatWalk gait analysis system to assess functional recovery in rodent models of peripheral nerve injury – a systematic review. J Neurosci Methods 2020; 345:108889. [DOI: 10.1016/j.jneumeth.2020.108889] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
|
46
|
McVeigh LG, Perugini AJ, Fehrenbacher JC, White FA, Kacena MA. Assessment, Quantification, and Management of Fracture Pain: from Animals to the Clinic. Curr Osteoporos Rep 2020; 18:460-470. [PMID: 32827293 PMCID: PMC7541703 DOI: 10.1007/s11914-020-00617-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW Fractures are painful and disabling injuries that can occur due to trauma, especially when compounded with pathologic conditions, such as osteoporosis in older adults. It is well documented that acute pain management plays an integral role in the treatment of orthopedic patients. There is no current therapy available to completely control post-fracture pain that does not interfere with bone healing or have major adverse effects. In this review, we focus on recent advances in the understanding of pain behaviors post-fracture. RECENT FINDINGS We review animal models of bone fracture and the assays that have been developed to assess and quantify spontaneous and evoked pain behaviors, including the two most commonly used assays: dynamic weight bearing and von Frey testing to assess withdrawal from a cutaneous (hindpaw) stimulus. Additionally, we discuss the assessment and quantification of fracture pain in the clinical setting, including the use of numeric pain rating scales, satisfaction with pain relief, and other biopsychosocial factor measurements. We review how pain behaviors in animal models and clinical cases can change with the use of current pain management therapies. We conclude by discussing the use of pain behavioral analyses in assessing potential therapeutic treatment options for addressing acute and chronic fracture pain without compromising fracture healing. There currently is a lack of effective treatment options for fracture pain that reliably relieve pain without potentially interfering with bone healing. Continued development and verification of reliable measurements of fracture pain in both pre-clinical and clinical settings is an essential aspect of continued research into novel analgesic treatments for fracture pain.
Collapse
Affiliation(s)
- Luke G McVeigh
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, 46202, USA
| | - Anthony J Perugini
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, 46202, USA
| | - Jill C Fehrenbacher
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fletcher A White
- Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN, USA
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, 1130 W. Michigan St, FH 115, Indianapolis, IN, 46202, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| |
Collapse
|
47
|
An Analysis of Variability in "CatWalk" Locomotor Measurements to Aid Experimental Design and Interpretation. eNeuro 2020; 7:ENEURO.0092-20.2020. [PMID: 32647037 PMCID: PMC7458803 DOI: 10.1523/eneuro.0092-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 06/17/2020] [Accepted: 06/22/2020] [Indexed: 12/02/2022] Open
Abstract
Preclinical studies in models of neurologic injury and disease rely on behavioral outcomes to measure intervention efficacy. For spinal cord injury, the CatWalk system provides unbiased quantitative assessment of subtle aspects of locomotor function in rodents and so can powerfully detect significant differences between experimental and control groups. Although clearly of key importance, summary group-level data can obscure the variability within and between individual subjects and therefore make it difficult to understand the magnitude of effect in individual animals and the proportion of a group that may show benefit. Here, we calculate reference change intervals (RCIs) that define boundaries of normal variability for measures of rat locomotion on the CatWalk. Our results indicate that many commonly-used outcome measures are highly variable, such that differences of up to 70% from baseline value must be considered normal variation. Many CatWalk outcome variables are also highly correlated and dependent on run speed. Application of calculated RCIs to open access data (https://scicrunch.org/odc-sci) on hindlimb stride length in spinal cord-injured rats illustrates the complementarity between group-level (16 mm change; p = 0.0009) and individual-level (5/32 animals show change outside RCI boundaries) analysis between week 3 and week 6 after injury. We also conclude that interdependence among CatWalk variables implies that test “batteries” require careful composition to ensure that different aspects of defective gait are analyzed. Calculation of RCIs aids in experimental design by quantifying variability and enriches overall data analysis by providing details of change at an individual level that complement group-level analysis.
Collapse
|
48
|
Effect of Systemic Adipose-derived Stem Cell Therapy on Functional Nerve Regeneration in a Rodent Model. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2020; 8:e2953. [PMID: 32802651 PMCID: PMC7413771 DOI: 10.1097/gox.0000000000002953] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 05/07/2020] [Indexed: 12/15/2022]
Abstract
Supplemental Digital Content is available in the text. Regardless of etiology, peripheral nerve injuries (PNI) result in disruption/loss of neuromuscular junctions, target muscle denervation, and poor sensorimotor outcomes with associated pain and disability. Adipose-derived stem cells (ASCs) have shown promise in neuroregeneration. However, there is a paucity of objective assessments reflective of functional neuroregeneration in experimental PNI. Here, we use a multimodal, static, and dynamic approach to evaluate functional outcomes after ASC therapy in a rodent PNI model.
Collapse
|
49
|
Yuan YS, Niu SP, Yu F, Zhang YJ, Han N, Lu H, Yin XF, Xu HL, Kou YH. Intraoperative single administration of neutrophil peptide 1 accelerates the early functional recovery of peripheral nerves after crush injury. Neural Regen Res 2020; 15:2108-2115. [PMID: 32394969 PMCID: PMC7716025 DOI: 10.4103/1673-5374.282270] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neutrophil peptide 1 belongs to a family of peptides involved in innate immunity. Continuous intramuscular injection of neutrophil peptide 1 can promote the regeneration of peripheral nerves, but clinical application in this manner is not convenient. To this end, the effects of a single intraoperative administration of neutrophil peptide 1 on peripheral nerve regeneration were experimentally observed. A rat model of sciatic nerve crush injury was established using the clamp method. After model establishment, a normal saline group and a neutrophil peptide 1 group were injected with a single dose of normal saline or 10 μg/mL neutrophil peptide 1, respectively. A sham group, without sciatic nerve crush was also prepared as a control. Sciatic nerve function tests, neuroelectrophysiological tests, and hematoxylin-eosin staining showed that the nerve conduction velocity, sciatic functional index, and tibialis anterior muscle fiber cross-sectional area were better in the neutrophil peptide 1 group than in the normal saline group at 4 weeks after surgery. At 4 and 8 weeks after surgery, there were no differences in the wet weight of the tibialis anterior muscle between the neutrophil peptide 1 and saline groups. Histological staining of the sciatic nerve showed no significant differences in the number of myelinated nerve fibers or the axon cross-sectional area between the neutrophil peptide 1 and normal saline groups. The above data confirmed that a single dose of neutrophil peptide 1 during surgery can promote the recovery of neurological function 4 weeks after sciatic nerve injury. All the experiments were approved by the Medical Ethics Committee of Peking University People’s Hospital, China (approval No. 2015-50) on December 9, 2015.
Collapse
Affiliation(s)
- Yu-Song Yuan
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| | - Su-Ping Niu
- Office of Academic Research, Peking University People's Hospital, Beijing, China
| | - Fei Yu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University, Beijing; National and Local Joint Engineering Research Center of Orthopaedic Biomaterials, Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, China
| | - Ya-Jun Zhang
- National Center for Trauma Medicine, Beijing, China
| | - Na Han
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education; Office of Academic Research, Peking University People's Hospital, Beijing, China
| | - Hao Lu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Diabetic Foot Treatment Center, Peking University People's Hospital, Peking University, Beijing, China
| | - Xiao-Feng Yin
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| | - Hai-Lin Xu
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Diabetic Foot Treatment Center, Peking University People's Hospital, Peking University, Beijing, China
| | - Yu-Hui Kou
- Department of Trauma and Orthopedics, Peking University People's Hospital, Peking University; Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
50
|
Heinzel JC, Hercher D, Redl H. The course of recovery of locomotor function over a 10-week observation period in a rat model of femoral nerve resection and autograft repair. Brain Behav 2020; 10:e01580. [PMID: 32097542 PMCID: PMC7177579 DOI: 10.1002/brb3.1580] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/26/2019] [Accepted: 01/30/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND A great extent of knowledge on peripheral nerve regeneration has been gathered using the rat sciatic nerve model. The femoral nerve model of the rat offers an interesting alternative, as it lacks disadvantageous features such as automutilation. For the analysis of locomotor behavior in rats after sciatic nerve injury, the CatWalk™ XT Gait Analysis System is often used. However, lesions of the femoral nerve in the rat have yet remained unstudied with this method. MATERIAL AND METHODS Ten male Sprague Dawley rats were evaluated with the CatWalk XT to study their gait after a 6-mm resection of the right femoral nerve and reconstruction with an autologous nerve graft. Animals were observed for 10 weeks after surgery. RESULTS Print Area, Print Length, Swing Speed, and Duty Cycle decreased to a minimum of 40% of baseline 2 weeks after surgery. Swing Time was elevated more than twofold at this time point. However, all these parameters recovered back to >90% of baseline values at 10 weeks after surgery. This degree of functional recovery has not been reported after sciatic nerve resection and autograft repair. Base of support varied minimally postoperatively in contrast to a strong decrement after sciatic nerve resection and repair. CONCLUSION We hereby provide a comprehensive in-depth analysis of how to study functional recovery after injury of the femoral nerve in the rat via the CatWalk XT. We place special emphasis on highlighting the differences between the femoral nerve and sciatic nerve injury model in this context.
Collapse
Affiliation(s)
- Johannes Christoph Heinzel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in the AUVA Trauma Research Center, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|