1
|
Wireman OH, Sams EL, Richey LE, Hammers GV, Stewart AN, Bailey WM, Patel SP, Gensel JC. Complete High Thoracic Spinal Cord Injury Causes Bowel Dysfunction in Mice. J Neurotrauma 2025. [PMID: 40179002 DOI: 10.1089/neu.2024.0277] [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: 04/05/2025] Open
Abstract
Bowel dysfunction, is a prevalent and life-impacting comorbidity of spinal cord injury (SCI) with no long-term treatment available. SCI-induced colon changes including motility and fibrosis are understudied as are strategies to address SCI bowel dysfunction. This need remains partly due to the lack of a mouse model that recapitulates the human condition. We hypothesized that a high thoracic spinal transection in mice would trigger bowel dysfunction with coincident colon pathology similar to humans and rats after SCI. We observed bowel dysfunction as increased fecal pellet numbers within the colon, smaller pellet size, and decreased motility. Fecal pellets numbers in the colon increased significantly in SCI animals versus sham (laminectomy only) injuries by 4 days postinjury (dpi) and persisted to 7 and 21 dpi. The number of pellets expelled (fecal output) significantly decreased in SCI versus sham animals at both 7 and 20 dpi. Pellet size was significantly decreased in SCI animals at 7 and 14 dpi, collectively indicative of decreased motility with SCI. SCI caused non-significant reductions in colonic motility (bead expulsion assay) at all three timepoints. Through ex vivo myograph analyses of live colon sections, we detected significant increase in the maximal contractility of the circular musculature from both the proximal and distal colon after SCI at 21 dpi. At the same time point, distal colons displayed significant collagen deposition in the musculature after SCI. Collectively, these findings demonstrate bowel dysfunction immediately after injury that continues in the distal colon over time. Establishing this mouse model enables further interrogation using transgenic models.
Collapse
Affiliation(s)
- Olivia H Wireman
- Department of Physiology, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Ellie L Sams
- Department of Physiology, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Lynnet E Richey
- Department of Physiology, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Gabrielle V Hammers
- Department of Physiology, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Andrew N Stewart
- Department of Neuroscience, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - William M Bailey
- Department of Physiology, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Samir P Patel
- Department of Physiology, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - John C Gensel
- Department of Physiology, Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
2
|
Li Y, Wu J. Sex-dependent alterations in extracellular vesicles linking chronic spinal cord injury to brain neuroinflammation and neurodegeneration. Neural Regen Res 2025; 20:483-484. [PMID: 38819058 PMCID: PMC11317934 DOI: 10.4103/nrr.nrr-d-24-00189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/18/2024] [Accepted: 03/30/2024] [Indexed: 06/01/2024] Open
Affiliation(s)
- Yun Li
- Department of Anesthesiology & Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Junfang Wu
- Department of Anesthesiology & Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
3
|
Kumari R, Hammers GV, Hammons RH, Stewart AN, MacLean SM, Niedzielko T, Schneider LE, Floyd CL, Gensel JC. Cross-species comparisons between pigs and mice reveal conserved sex-specific intraspinal inflammatory responses after spinal cord injury. J Neuroinflammation 2025; 22:16. [PMID: 39849507 PMCID: PMC11759441 DOI: 10.1186/s12974-025-03338-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 01/07/2025] [Indexed: 01/25/2025] Open
Abstract
OBJECTIVE Therapeutic translation is challenging in spinal cord injury (SCI) and large animal models with high clinical relevance may accelerate therapeutic development. Pigs have important anatomical and physiological similarities to humans. Intraspinal inflammation mediates SCI pathophysiology. The purpose of this study was to evaluate the effect of sex on inflammation and outcomes in a pig thoracic contusion/compression SCI model. METHODS Adult (gonad-intact) male and female Yucatan miniature swine were subjected to either SCI or sham (laminectomy-only) injury. RESULTS SCI caused locomotor dysfunction (measured with the Porcine Thoracic Injury Behavior Score) with some recovery over 6 weeks and limited tissue sparing at 6 weeks with no difference between sexes. Immunohistological evaluations of spinal cord tissue at 2 days and 6 weeks post-injury revealed intraspinal microglia/macrophage (IBA-1, CD68) and lymphocyte responses (T-cells (CD3) and B-cells (CD79a)) consistent with observations in rodents and humans. Astrocyte (GFAP) immunoreactivity was observed within the lesion core at 6 weeks in contrast to observations in rodents. No differences were seen for astrocytes, microglia, macrophages, B-cells, and neutrophil infiltration between males and females. Intraspinal CD3 + T-cell counts and T-cell microclusters were significantly higher in females compared to males 6 weeks post-injury. Interestingly, we observed a similar significant increase in intraspinal CD3 + T-cell accumulation in female vs. male mice at 6 weeks post-thoracic contusion SCI. INTERPRETATION Our observations indicate that sex is a potential biological variable for T-cell infiltration and may contribute to sex-based differences in SCI pathophysiology and recovery outcomes.
Collapse
Affiliation(s)
- Reena Kumari
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington Kentucky, USA
| | - Gabrielle V Hammers
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington Kentucky, USA
| | - Robert H Hammons
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington Kentucky, USA
| | - Andrew N Stewart
- Spinal Cord and Brain Injury Research Center, Department of Neuroscience, College of Medicine, University of Kentucky, Lexington Kentucky, USA
| | - Steven M MacLean
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington Kentucky, USA
| | - Tracy Niedzielko
- Department of Emergency Medicine, School of Medicine, Emory University, Atlanta Georgia, USA
| | - Lonnie E Schneider
- Department of Emergency Medicine, School of Medicine, Emory University, Atlanta Georgia, USA
| | - Candace L Floyd
- Department of Emergency Medicine, School of Medicine, Emory University, Atlanta Georgia, USA.
- , Whitehead Biomedical Research Building, Room 605L 615 Michael Street, Atlanta, GA, 30322, USA.
| | - John C Gensel
- Spinal Cord and Brain Injury Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington Kentucky, USA.
- University of Kentucky College of Medicine, B483 Biomedical & Biological Sciences Research Building (BBSRB), 741 S. Limestone Street, Lexington, KY, 40536-0509, USA.
| |
Collapse
|
4
|
Entenmann CJ, von Bronewski EJ, Waldmann L, Meyer L, Kersting K, Roolfs LT, Schleker LM, Nieminen-Kelhä M, Kremenetskaia I, Heppner FL, Fehlings MG, Vajkoczy P, Hubertus V. Analysis of the spatiotemporal dynamics of vascular injury and regeneration following experimental Spinal Cord Injury. BRAIN & SPINE 2025; 5:104191. [PMID: 39935529 PMCID: PMC11810697 DOI: 10.1016/j.bas.2025.104191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Introduction The loss of vasculature in Spinal Cord Injury (SCI) contributes to secondary injury, expanding the injury to unharmed spinal cord (SC) regions. Understanding these mechanisms is crucial for developing therapeutic interventions. Research question Comprehensive analysis of the temporospatial dynamics of vascular injury and regeneration following SCI. Materials and methods Adult C57BL/6J mice were subjected to clip-compression SCI (Th 6/7, 5g, 60s, n = 20) or sham injury (laminectomy, n = 4), and sacrificed at 1, 3, 7, 14, and 28 days (d) post-injury following intracardial fluorescein isothiocyanate (FITC)-Lectin perfusion. Histological analysis (CD31, FITC-Lectin, Ki-67, IgG, TER-119) assessed vascular changes, permeability, and proliferation within the injury epicenter (region 0 (R0), ± 0,5 mm) and two adjacent SC regions (R1: ± 1 mm, R2: ± 2.5 mm). Results Perfusion loss (FITC-Lectin+/CD31+), was most severe in R0 and R1 at d3 (p < 0.01). Significant vascular loss in R2 started at d3 (p = 0.043). Perfusion was restored at d28 in R0 and R1, and at d7 in R2. Vessel density (CD31+) returned to baseline quicker (R0: d3, R1 and R2: d14). Vascular proliferation (CD31+/Ki-67+) manifested across all regions at d3 (p < 0.01), and most notably in R2 (p < 0.01). Vascular permeability for IgG remained disrupted until d3 in R0 and R1 and until d14 in R2. Discussion and conclusion Vascular injury is most severe initially and spreads to the surrounding SC regions. Gradual vascular regeneration occurs early and up to a considerable distance from the injury epicenter, highlighting the potential of early therapeutic interventions targeted at vascular repair and regeneration.
Collapse
Affiliation(s)
- Christian J. Entenmann
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Emily J. von Bronewski
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Lilly Waldmann
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Lea Meyer
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Katharina Kersting
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Laurens T. Roolfs
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Lasse M. Schleker
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, 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, Berlin Institute of Health, Berlin, Germany
| | - Irina Kremenetskaia
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Frank L. Heppner
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Cluster of Excellence, NeuroCure, Berlin, Germany
| | - Michael G. Fehlings
- Division of Neurosurgery and Krembil Neuroscience Center, 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, Berlin Institute of Health, Berlin, Germany
| | - Vanessa Hubertus
- Department of Neurosurgery, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health (BIH) – Charité Clinician Scientist Program, Berlin, Germany
| |
Collapse
|
5
|
Stefanov A, Brakel K, Rau J, Joseph RM, Guice C, Araguz K, Hemphill A, Madry J, Irion A, Dash S, Souza KA, Hook MA. Depression-like behavior is associated with deficits in cognition and hippocampal neurogenesis in a subset of spinally contused male, but not female, rats. Brain Behav Immun 2025; 123:270-287. [PMID: 39288895 DOI: 10.1016/j.bbi.2024.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/25/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024] Open
Abstract
Depression and cognitive deficits present at higher rates among people with spinal cord injury (SCI) compared to the general population, yet these SCI comorbidities are poorly addressed. Sex and age appear to play roles in depression incidence, but consensus on the direction of their effects is limited. Systemic and cortical inflammation and disruptions in hippocampal neurogenesis have been identified as potential treatment targets, but a comprehensive understanding of these mechanisms remains elusive. We used a rodent SCI model to interrogate these gaps in knowledge. We examined post-injury depression-like behavior and cognitive deficits, as well as the association between affect, cognition, chronic hippocampal inflammation and hippocampal neurogenesis, in young and middle-aged male and female Sprague-Dawley rats. Depression-like behavior manifested in male and female subsets of SCI rats irrespective of age, at rates commensurate with the incidence of clinical depression. Changes in components of behavior were driven by sex and age, and affective outcomes were independent of common post-injury pathophysiological outcomes including locomotor functional deficits and spinal lesion severity. Interestingly, however, only male depression-like SCI rats exhibited deficits in hippocampal-associated spatial cognition. Neurogenesis was also disrupted in only SCI males in regions of the hippocampus responsible for affective outcomes. Decreased neurogenesis among middle-aged male subjects coincided with increases in numbers of the pro-inflammatory markers CD86 and iNOS, while middle-aged females had increased numbers of cells expressing Iba-1 and anti-inflammatory marker CD206. Overall, the present data suggest that post-SCI depression and cognition may be affected, in part, by sex- and age-dependent changes in hippocampal neurogenesis and inflammation. Hippocampal neurogenesis is a potential target to address psychological wellbeing after SCI, but therapeutic strategies must carefully consider sex and age as biological variables.
Collapse
Affiliation(s)
- Alex Stefanov
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843.
| | - Kiralyn Brakel
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843
| | - Josephina Rau
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843
| | - Rose M Joseph
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Corey Guice
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Kendall Araguz
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Annebel Hemphill
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Jessica Madry
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Andrew Irion
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Swapnil Dash
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Karienn A Souza
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807
| | - Michelle A Hook
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, College of Medicine, Bryan, TX 77807; Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX 77843
| |
Collapse
|
6
|
Rigo YR, Benvenutti R, Portela LV, Strogulski NR. Neurogenic potential of NG2 in neurotrauma: a systematic review. Neural Regen Res 2024; 19:2673-2683. [PMID: 38595286 PMCID: PMC11168526 DOI: 10.4103/nrr.nrr-d-23-01031] [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: 06/21/2023] [Revised: 12/20/2023] [Accepted: 02/07/2024] [Indexed: 04/11/2024] Open
Abstract
Regenerative approaches towards neuronal loss following traumatic brain or spinal cord injury have long been considered a dogma in neuroscience and remain a cutting-edge area of research. This is reflected in a large disparity between the number of studies investigating primary and secondary injury as therapeutic targets in spinal cord and traumatic brain injuries. Significant advances in biotechnology may have the potential to reshape the current state-of-the-art and bring focus to primary injury neurotrauma research. Recent studies using neural-glial factor/antigen 2 (NG2) cells indicate that they may differentiate into neurons even in the developed brain. As these cells show great potential to play a regenerative role, studies have been conducted to test various manipulations in neurotrauma models aimed at eliciting a neurogenic response from them. In the present study, we systematically reviewed the experimental protocols and findings described in the scientific literature, which were peer-reviewed original research articles (1) describing preclinical experimental studies, (2) investigating NG2 cells, (3) associated with neurogenesis and neurotrauma, and (4) in vitro and/or in vivo, available in PubMed/MEDLINE, Web of Science or SCOPUS, from 1998 to 2022. Here, we have reviewed a total of 1504 papers, and summarized findings that ultimately suggest that NG2 cells possess an inducible neurogenic potential in animal models and in vitro. We also discriminate findings of NG2 neurogenesis promoted by different pharmacological and genetic approaches over functional and biochemical outcomes of traumatic brain injury and spinal cord injury models, and provide mounting evidence for the potential benefits of manipulated NG2 cell ex vivo transplantation in primary injury treatment. These findings indicate the feasibility of NG2 cell neurogenesis strategies and add new players in the development of therapeutic alternatives for neurotrauma.
Collapse
Affiliation(s)
- Yuri R. Rigo
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Radharani Benvenutti
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luis V. Portela
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
7
|
Funnell JL, Fougere J, Zahn D, Dutz S, Gilbert RJ. Delivery of TGFβ3 from Magnetically Responsive Coaxial Fibers Reduces Spinal Cord Astrocyte Reactivity In Vitro. Adv Biol (Weinh) 2024; 8:e2300531. [PMID: 38935534 PMCID: PMC11473240 DOI: 10.1002/adbi.202300531] [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: 10/02/2023] [Revised: 03/29/2024] [Indexed: 06/29/2024]
Abstract
A spinal cord injury (SCI) compresses the spinal cord, killing neurons and glia at the injury site and resulting in prolonged inflammation and scarring that prevents regeneration. Astrocytes, the main glia in the spinal cord, become reactive following SCI and contribute to adverse outcomes. The anti-inflammatory cytokine transforming growth factor beta 3 (TGFβ3) has been shown to mitigate astrocyte reactivity; however, the effects of prolonged TGFβ3 exposure on reactive astrocyte phenotype have not yet been explored. This study investigates whether magnetic core-shell electrospun fibers can be used to alter the release rate of TGFβ3 using externally applied magnetic fields, with the eventual application of tailored drug delivery based on SCI severity. Magnetic core-shell fibers are fabricated by incorporating superparamagnetic iron oxide nanoparticles (SPIONs) into the shell and TGFβ3 into the core solution for coaxial electrospinning. Magnetic field stimulation increased the release rate of TGFβ3 from the fibers by 25% over 7 days and released TGFβ3 reduced gene expression of key astrocyte reactivity markers by at least twofold. This is the first study to magnetically deliver bioactive proteins from magnetic fibers and to assess the effect of sustained release of TGFβ3 on reactive astrocyte phenotype.
Collapse
Affiliation(s)
- Jessica L Funnell
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St., Troy, NY, 12180, USA
| | - Jasper Fougere
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St., Troy, NY, 12180, USA
| | - Diana Zahn
- Institut für Biomedizinische Technik und Informatik, Technische Universität Ilmenau, Gustav-Kirchhoff-Str. 2, 98693, Ilmenau, Germany
| | - Silvio Dutz
- Institut für Biomedizinische Technik und Informatik, Technische Universität Ilmenau, Gustav-Kirchhoff-Str. 2, 98693, Ilmenau, Germany
- Westsächsische Hochschule Zwickau, Kornmarkt 1, 08056, Zwickau, Germany
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th St, Troy, NY, 12180, USA
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th St., Troy, NY, 12180, USA
- Albany Stratton Veteran Affairs Medical Center, 113 Holland Ave., Albany, NY, 12208, USA
| |
Collapse
|
8
|
Li Y, Lei Z, Ritzel RM, He J, Liu S, Zhang L, Wu J. Ablation of the Integrin CD11b Mac-1 Limits Deleterious Responses to Traumatic Spinal Cord Injury and Improves Functional Recovery in Mice. Cells 2024; 13:1584. [PMID: 39329765 PMCID: PMC11430243 DOI: 10.3390/cells13181584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 09/28/2024] Open
Abstract
Spinal cord injury (SCI) triggers microglial/monocytes activation with distinct pro-inflammatory or inflammation-resolving phenotypes, which potentiate tissue damage or facilitate functional repair, respectively. The major integrin Mac-1 (CD11b/CD18), a heterodimer consisting of CD11b and CD18 chains, is expressed in multiple immune cells of the myeloid lineage. Here, we examined the effects of CD11b gene ablation in neuroinflammation and functional outcomes after SCI. qPCR analysis of C57BL/6 female mice showed upregulation of CD11b mRNA starting from 1 d after injury, which persisted up to 28 d. CD11b knockout (KO) mice and their wildtype littermates were subjected to moderate SCI. At 1 d post-injury, qPCR showed increased expression of genes involved with inflammation-resolving processes in CD11b KO mice. Flow cytometry analysis of CD45intLy6C-CX3CR1+ microglia, CD45hiLy6C+Ly6G- monocytes, and CD45hiLy6C+Ly6G+ neutrophils revealed significantly reduced cell counts as well as reactive oxygen species (ROS) production in CD11b KO mice at d3 post-injury. Further examination with NanoString and RNA-seq showed upregulation of pro-inflammatory genes, but downregulation of the ROS pathway. Importantly, CD11b KO mice exhibited significantly improved locomotor function, reduced cutaneous mechanical/thermal hypersensitivity, and limited tissue damage at 8 weeks post-injury. Collectively, our data suggest an important role for CD11b in regulating tissue inflammation and functional outcome following SCI.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD 21201, USA; (Y.L.); (Z.L.); (R.M.R.); (J.H.); (S.L.)
| | - Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD 21201, USA; (Y.L.); (Z.L.); (R.M.R.); (J.H.); (S.L.)
| | - Rodney M. Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD 21201, USA; (Y.L.); (Z.L.); (R.M.R.); (J.H.); (S.L.)
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD 21201, USA; (Y.L.); (Z.L.); (R.M.R.); (J.H.); (S.L.)
| | - Simon Liu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD 21201, USA; (Y.L.); (Z.L.); (R.M.R.); (J.H.); (S.L.)
| | - Li Zhang
- Department of Physiology, Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD 21201, USA; (Y.L.); (Z.L.); (R.M.R.); (J.H.); (S.L.)
| |
Collapse
|
9
|
Lei Z, Ritzel RM, Li Y, Li H, Faden AI, Wu J. Old age alters inflammation and autophagy signaling in the brain, leading to exacerbated neurological outcomes after spinal cord injury in male mice. Brain Behav Immun 2024; 120:439-451. [PMID: 38925420 PMCID: PMC11269014 DOI: 10.1016/j.bbi.2024.06.023] [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: 01/27/2024] [Revised: 05/20/2024] [Accepted: 06/23/2024] [Indexed: 06/28/2024] Open
Abstract
Older patients with spinal cord injury (SCI) have different features with regard to neurological characteristics after injury. Recent large-scale longitudinal population-based studies showed that individuals with SCI are at a higher risk of developing dementia than non-SCI patients, indicating that SCI is a potential risk factor for dementia. Aging is known to potentiate inflammation and neurodegeneration at the injured site leading to impaired recovery from SCI. However, no research has been aimed at studying the mechanisms of SCI-mediated cognitive impairment in the elderly. The present study examined neurobehavioral and molecular changes in the brain and the underlying mechanisms associated with brain dysfunction in aged C57BL/6 male mice using a contusion SCI model. At 2 months post-injury, aged mice displayed worse performance in locomotor, cognitive and depressive-like behavioral tests compared to young adult animals. Histopathology in injured spinal cord tissue was exacerbated in aged SCI mice. In the brain, transcriptomic analysis with NanoString neuropathology panel identified activated microglia and dysregulated autophagy as the most significantly altered pathways by both age and injury. These findings were further validated by flow cytometry, which demonstrated increased myeloid and lymphocytes infiltration at both the injured site and brain of aged mice. Moreover, SCI in aged mice altered microglial function and dysregulated autophagy in microglia, resulting in worsened neurodegeneration. Taken together, our data indicate that old age exacerbates neuropathological changes in both the injured spinal cord and remote brain regions leading to poorer functional outcomes, at least in part, through altered inflammation and autophagy function.
Collapse
Affiliation(s)
- Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hui Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
10
|
Li Y, Ritzel RM, He J, Liu S, Zhang L, Wu J. Ablation of the integrin CD11b mac-1 limits deleterious responses to traumatic spinal cord injury and improves functional recovery in mice. RESEARCH SQUARE 2024:rs.3.rs-4196316. [PMID: 38645238 PMCID: PMC11030505 DOI: 10.21203/rs.3.rs-4196316/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background Spinal cord injury (SCI) causes long-term sensorimotor deficits and posttraumatic neuropathic pain, with no effective treatment. In part, this reflects an incomplete understanding of the complex secondary pathobiological mechanisms involved. SCI triggers microglial/macrophage activation with distinct pro-inflammatory or inflammation-resolving phenotypes, which potentiate tissue damage or facilitate functional repair, respectively. The major integrin Mac-1 (CD11b/CD18, αMβ2 or CR3), a heterodimer consisting of αM (CD11b) and β2 (CD18) chains, is generally regarded as a pro-inflammatory receptor in neurotrauma. Multiple immune cells of the myeloid lineage express CD11b, including microglia, macrophages, and neutrophils. In the present study, we examined the effects of CD11b gene ablation on posttraumatic neuroinflammation and functional outcomes after SCI. Methods Young adult age-matched female CD11b knockout (KO) mice and their wildtype (WT) littermates were subjected to moderate thoracic spinal cord contusion. Neuroinflammation in the injured spinal cord was assessed with qPCR, flow cytometry, NanoString, and RNAseq. Neurological function was evaluated with the Basso Mouse Scale (BMS), gait analysis, thermal hyperesthesia, and mechanical allodynia. Lesion volume was evaluated by GFAP-DAB immunohistochemistry, followed by analysis with unbiased stereology. Results qPCR analysis showed a rapid and persistent upregulation of CD11b mRNA starting from 1d after injury, which persisted up to 28 days. At 1d post-injury, increased expression levels of genes that regulate inflammation-resolving processes were observed in CD11b KO mice. Flow cytometry analysis of CD45intLy6C-CX3CR1+ microglia, CD45hiLy6C+Ly6G- monocytes, and CD45hiLy6C+Ly6G+ neutrophils revealed significantly reduced cell counts as well as reactive oxygen production in CD11b KO mice at d3 post-injury. Further examination of the injured spinal cord with NanoString Mouse Neuroinflammation Panel and RNAseq showed upregulated expression of pro-inflammatory genes, but downregulated expression of the reactive oxygen species pathway. Importantly, CD11b KO mice exhibited significantly improved locomotor function, reduced cutaneous mechanical/thermal hypersensitivity, and limited tissue damage at 8 weeks post-injury. Conclusion Collectively, our data suggest an important role for CD11b in regulating tissue inflammation and functional outcome following SCI. Thus, the integrin CD11b represents a potential target that may lead to novel therapeutic strategies for SCI.
Collapse
Affiliation(s)
- Yun Li
- University of Maryland School of Medicine
| | | | - Junyun He
- University of Maryland School of Medicine
| | - Simon Liu
- University of Maryland School of Medicine
| | - Li Zhang
- University of Maryland School of Medicine
| | - Junfang Wu
- University of Maryland School of Medicine
| |
Collapse
|
11
|
Cherian CM, Reeves HR, De Silva D, Tsao S, Marshall KE, Rideout EJ. Consideration of sex as a biological variable in diabetes research across twenty years. Biol Sex Differ 2024; 15:19. [PMID: 38409052 PMCID: PMC10895746 DOI: 10.1186/s13293-024-00595-2] [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: 11/14/2023] [Accepted: 02/16/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Sex differences exist in the risk of developing type 1 and type 2 diabetes, and in the risk of developing diabetes-associated complications. Sex differences in glucose homeostasis, islet and β cell biology, and peripheral insulin sensitivity have also been reported. Yet, we lack detailed information on the mechanisms underlying these differences, preventing the development of sex-informed therapeutic strategies for persons living with diabetes. To chart a path toward greater inclusion of biological sex as a variable in diabetes research, we first need a detailed assessment of common practices in the field. METHODS We developed a scoring system to evaluate the inclusion of biological sex in manuscripts published in Diabetes, a journal published by the American Diabetes Association. We chose Diabetes as this journal focuses solely on diabetes and diabetes-related research, and includes manuscripts that use both clinical and biomedical approaches. We scored papers published across 3 years within a 20-year period (1999, 2009, 2019), a timeframe that spans the introduction of funding agency and journal policies designed to improve the consideration of biological sex as a variable. RESULTS Our analysis showed fewer than 15% of papers used sex-based analysis in even one figure across all study years, a trend that was reproduced across journal-defined categories of diabetes research (e.g., islet studies, signal transduction). Single-sex studies accounted for approximately 40% of all manuscripts, of which > 87% used male subjects only. While we observed a modest increase in the overall inclusion of sex as a biological variable during our study period, our data highlight significant opportunities for improvement in diabetes research practices. We also present data supporting a positive role for journal policies in promoting better consideration of biological sex in diabetes research. CONCLUSIONS Our analysis provides significant insight into common practices in diabetes research related to the consideration of biological sex as a variable. Based on our analysis we recommend ways that diabetes researchers can improve inclusion of biological sex as a variable. In the long term, improved practices will reveal sex-specific mechanisms underlying diabetes risk and complications, generating knowledge to enable the development of sex-informed prevention and treatment strategies.
Collapse
Affiliation(s)
- Celena M Cherian
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, Canada
| | - Hayley R Reeves
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, Canada
- School of Molecular Biosciences, University of Glasgow, Glasgow, UK
| | - Duneesha De Silva
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, Canada
- Department of Orthopaedics, The University of British Columbia, Vancouver, Canada
| | - Serena Tsao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, Canada
- Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, McGill University, Montréal, Canada
| | - Katie E Marshall
- Department of Zoology, The University of British Columbia, Vancouver, Canada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, Canada.
- Life Sciences Center, 2350 Health Sciences Mall (RM3308), Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
12
|
Collins NJ, Campbell TS, Bozeman AL, Martes AC, Ross SE, Doherty TS, Brumley MR, Roth TL. Epigenetic processes associated with neonatal spinal transection. Dev Psychobiol 2024; 66:e22466. [PMID: 38388192 DOI: 10.1002/dev.22466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/29/2023] [Accepted: 01/18/2024] [Indexed: 02/24/2024]
Abstract
In early development, the spinal cord in healthy or disease states displays remarkable activity-dependent changes in plasticity, which may be in part due to the increased activity of brain derived neurotrophic factor (BDNF). Indeed, BDNF delivery has been efficacious in partially ameliorating many of the neurobiological and behavioral consequences of spinal cord injury (SCI), making elucidating the role of BDNF in the normative developing and injured spinal cord a critical research focus. Recent work in our laboratory provided evidence for aberrant global and locus-specific epigenetic changes in methylation of the Bdnf gene as a consequence of SCI. In the present study, animals underwent thoracic lesions on P1, with cervical and lumbar tissue being later collected on P7, P14, and P21. Levels of Bdnf expression and methylation (exon IX and exon IV), in addition to global methylation levels were quantified at each timepoint. Results indicated locus-specific reductions of Bdnf expression that was accompanied by a parallel increase in methylation caudal to the injury site, with animals displaying increased Bdnf expression at the P14 timepoint. Together, these findings suggest that epigenetic activity of the Bdnf gene may act as biomarker in the etiology and intervention effort efficacy following SCI.
Collapse
Affiliation(s)
- Nicholas J Collins
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Taylor S Campbell
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Aimee L Bozeman
- Department of Psychology, Idaho State University, Pocatello, Idaho, USA
| | - Alleyna C Martes
- Department of Psychology, Idaho State University, Pocatello, Idaho, USA
| | - Sydney E Ross
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Tiffany S Doherty
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| | - Michele R Brumley
- Department of Psychology, Idaho State University, Pocatello, Idaho, USA
| | - Tania L Roth
- Department of Psychological and Brain Sciences, University of Delaware, Newark, Delaware, USA
| |
Collapse
|
13
|
Vijayakumar Sreelatha H, Palekkodan H, Fasaludeen A, K. Krishnan L, Abelson KSP. Refinement of the motorised laminectomy-assisted rat spinal cord injury model by analgesic treatment. PLoS One 2024; 19:e0294720. [PMID: 38227583 PMCID: PMC10790998 DOI: 10.1371/journal.pone.0294720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/06/2023] [Indexed: 01/18/2024] Open
Abstract
Usage and reporting of analgesia in animal models of spinal cord injury (SCI) have been sparse and requires proper attention. The majority of experimental SCI research uses rats as an animal model. This study aimed to probe into the effects of some commonly used regimens with NSAIDs and opioids on well-being of the rats as well as on the functional outcome of the model. This eight-week study used forty-two female Wistar rats (Crl: WI), randomly and equally divided into 6 treatment groups, viz. I) tramadol (5mg/kg) and buprenorphine (0.05mg/kg); II) carprofen (5mg/kg) and buprenorphine (0.05mg/kg); III) carprofen (5mg/kg); IV) meloxicam (1mg/kg) and buprenorphine (0.05mg/kg); V) meloxicam (1mg/kg); and VI) no analgesia (0.5 ml sterile saline). Buprenorphine was administered twice daily whereas other treatments were given once daily for five days post-operatively. Injections were given subcutaneously. All animals underwent dental burr-assisted laminectomy at the T10-T11 vertebra level. A custom-built calibrated spring-loaded 200 kilodynes force deliverer was used to induce severe SCI. Weekly body weight scores, Rat Grimace Scale (RGS), and dark-phase home cage activity were used as markers for well-being. Weekly Basso Beattie and Bresnahan (BBB) scores served as markers for functionality together with Novel Object Recognition test (NOR) at week 8 and terminal histopathology using area of vacuolisation and live neuronal count from the ventral horns of spinal cord. It was concluded that the usage of analgesia improved animal wellbeing while having no effects on the functional aspects of the animal model in comparison to the animals that received no analgesics.
Collapse
Affiliation(s)
- Harikrishnan Vijayakumar Sreelatha
- Department of Applied Biology, Division of Laboratory Animal Science, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Thiruvananthapuram, Kerala, India
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hamza Palekkodan
- Department of Veterinary Pathology, College of Veterinary and Animal Sciences, Pookot, Wayanad, Kerala, India
| | - Ansar Fasaludeen
- Department of Veterinary Pathology, College of Veterinary and Animal Sciences, Pookot, Wayanad, Kerala, India
| | - Lissy K. Krishnan
- Biological Research and Innovation Wing, Dr. Moopen’s Medical College, Wayanad, Kerala, India
| | - Klas S. P. Abelson
- Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
14
|
Stewart AN, Gensel JC, Jones L, Fouad K. Challenges in Translating Regenerative Therapies for Spinal Cord Injury. Top Spinal Cord Inj Rehabil 2023; 29:23-43. [PMID: 38174141 PMCID: PMC10759906 DOI: 10.46292/sci23-00044s] [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] [Indexed: 01/05/2024]
Abstract
Regenerating the injured spinal cord is a substantial challenge with many obstacles that need to be overcome to achieve robust functional benefits. This abundance of hurdles can partly explain the limited success when applying regenerative intervention treatments in animal models and/or people. In this article, we elaborate on a few of these obstacles, starting with the applicability of animal models and how they compare to the clinical setting. We then discuss the requirement for combinatorial interventions and the associated problems in experimental design, including the addition of rehabilitative training. The article expands on differences in lesion sizes and locations between humans and common animal models, and how this difference can determine the success or failure of an intervention. An additional and frequently overlooked problem in the translation of interventions that applies beyond the field of neuroregeneration is the reporting bias and the lack of transparency in reporting findings. New data mandates are tackling this problem and will eventually result in a more balanced view of the field. Finally, we will discuss strategies to negotiate the challenging course of successful translation to facilitate successful translation of regeneration promoting interventions.
Collapse
Affiliation(s)
- Andrew N. Stewart
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - John C. Gensel
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky, USA
| | - Linda Jones
- Department of Occupational Therapy, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Karim Fouad
- Department of Physical Therapy, University of Alberta, Edmonton, Canada
| |
Collapse
|
15
|
Dehdashtian A, Timek JH, Svientek SR, Risch MJ, Bratley JV, Riegger AE, Kung TA, Cederna PS, Kemp SWP. Sexually Dimorphic Pattern of Pain Mitigation Following Prophylactic Regenerative Peripheral Nerve Interface (RPNI) in a Rat Neuroma Model. Neurosurgery 2023; 93:1192-1201. [PMID: 37227138 DOI: 10.1227/neu.0000000000002548] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 04/06/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Treating neuroma pain is a clinical challenge. Identification of sex-specific nociceptive pathways allows a more individualized pain management. The Regenerative Peripheral Nerve Interface (RPNI) consists of a neurotized autologous free muscle using a severed peripheral nerve to provide physiological targets for the regenerating axons. OBJECTIVE To evaluate prophylactic RPNI to prevent neuroma pain in male and female rats. METHODS F344 rats of each sex were assigned to neuroma, prophylactic RPNI, or sham groups. Neuromas and RPNIs were created in both male and female rats. Weekly pain assessments including neuroma site pain and mechanical, cold, and thermal allodynia were performed for 8 weeks. Immunohistochemistry was used to evaluate macrophage infiltration and microglial expansion in the corresponding dorsal root ganglia and spinal cord segments. RESULTS Prophylactic RPNI prevented neuroma pain in both sexes; however, female rats displayed delayed pain attenuation when compared with males. Cold allodynia and thermal allodynia were attenuated exclusively in males. Macrophage infiltration was mitigated in males, whereas females showed a reduced number of spinal cord microglia. CONCLUSION Prophylactic RPNI can prevent neuroma site pain in both sexes. However, attenuation of both cold allodynia and thermal allodynia occurred in males exclusively, potentially because of their sexually dimorphic effect on pathological changes of the central nervous system.
Collapse
Affiliation(s)
- Amir Dehdashtian
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
| | - Jagienka H Timek
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
| | - Shelby R Svientek
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
| | - Mary Jane Risch
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
| | - Jared V Bratley
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
| | - Anna E Riegger
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
| | - Theodore A Kung
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
| | - Paul S Cederna
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
- Department of Biomedical Engineering, The University of Michigan, Ann Arbor , Michigan , USA
| | - Stephen W P Kemp
- Department of Surgery, Section of Plastic Surgery, The University of Michigan Health System, Ann Arbor , Michigan , USA
- Department of Biomedical Engineering, The University of Michigan, Ann Arbor , Michigan , USA
| |
Collapse
|
16
|
Stewart AN, Kumari R, Bailey WM, Glaser EP, Bosse-Joseph CC, Park KA, Hammers GV, Wireman OH, Gensel JC. PTEN knockout using retrogradely transported AAVs transiently restores locomotor abilities in both acute and chronic spinal cord injury. Exp Neurol 2023; 368:114502. [PMID: 37558155 PMCID: PMC10498341 DOI: 10.1016/j.expneurol.2023.114502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/07/2023] [Accepted: 08/07/2023] [Indexed: 08/11/2023]
Abstract
Restoring function in chronic stages of spinal cord injury (SCI) has often been met with failure or reduced efficacy when regenerative strategies are delayed past the acute or sub-acute stages of injury. Restoring function in the chronically injured spinal cord remains a critical challenge. We found that a single injection of retrogradely transported adeno-associated viruses (AAVrg) to knockout the phosphatase and tensin homolog protein (PTEN) in chronic SCI can effectively target both damaged and spared axons and transiently restore locomotor functions in near-complete injury models. AAVrg's were injected to deliver cre recombinase and/or a red fluorescent protein (RFP) under the human Synapsin 1 promoter (hSyn1) into the spinal cords of C57BL/6 PTENFloxΔ/Δ mice to knockout PTEN (PTEN-KO) in a severe thoracic SCI crush model at both acute and chronic time points. PTEN-KO improved locomotor abilities in both acute and chronic SCI conditions over a 9-week period. Regardless of whether treatment was initiated at the time of injury (acute), or three months after SCI (chronic), mice with limited hindlimb joint movement gained hindlimb weight support after treatment. Interestingly, functional improvements were not sustained beyond 9 weeks coincident with a loss of RFP reporter-gene expression and a near-complete loss of treatment-associated functional recovery by 6 months post-treatment. Treatment effects were also specific to severely injured mice; animals with weight support at the time of treatment lost function over a 6-month period. Retrograde tracing with Fluorogold revealed viable neurons throughout the motor cortex despite a loss of RFP expression at 9 weeks post-PTEN-KO. However, few Fluorogold labeled neurons were detected within the motor cortex at 6 months post-treatment. BDA labeling from the motor cortex revealed a dense corticospinal tract (CST) bundle in all groups except chronically treated PTEN-KO mice, indicating a potential long-term toxic effect of PTEN-KO to neurons in the motor cortex which was corroborated by a loss of β-tubulin III labeling above the lesion within spinal cords after PTEN-KO. PTEN-KO mice had significantly more β-tubulin III labeled axons within the lesion when treatment was delivered acutely, but not chronically post-SCI. In conclusion, we have found that using AAVrg's to knockout PTEN is an effective manipulation capable of restoring motor functions in chronic SCI and can enhance axon growth of currently unidentified axon populations when delivered acutely after injury. However, the long-term consequences of PTEN-KO on neuronal health and viability should be further explored.
Collapse
Affiliation(s)
- Andrew N Stewart
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA.
| | - Reena Kumari
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - William M Bailey
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Ethan P Glaser
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Christopher C Bosse-Joseph
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Kennedy A Park
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Gabrielle V Hammers
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Olivia H Wireman
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - John C Gensel
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA; Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA; College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
17
|
Cabrera-Aldana EE, Balderas-Martínez YI, Velázquez-Cruz R, Tovar-y-Romo LB, Sevilla-Montoya R, Martínez-Cruz A, Martinez-Cordero C, Valdés-Flores M, Santamaria-Olmedo M, Hidalgo-Bravo A, Guízar-Sahagún G. Administration of Tamoxifen Can Regulate Changes in Gene Expression during the Acute Phase of Traumatic Spinal Cord Injury. Curr Issues Mol Biol 2023; 45:7476-7491. [PMID: 37754256 PMCID: PMC10529143 DOI: 10.3390/cimb45090472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Traumatic spinal cord injury (SCI) causes irreversible damage leading to incapacity. Molecular mechanisms underlying SCI damage are not fully understood, preventing the development of novel therapies. Tamoxifen (TMX) has emerged as a promising therapy. Our aim was to identify transcriptome changes in the acute phase of SCI and the effect of Tamoxifen on those changes in a rat model of SCI. Four groups were considered: (1) Non-injured without TMX (Sham/TMX-), (2) Non-injured with TMX (Sham/TMX+), (3) injured without TMX (SCI/TMX-), and (4) injured with TMX (SCI/TMX+). Tamoxifen was administered intraperitoneally 30 min after injury, and spinal cord tissues were collected 24 h after injury. Clariom S Assays Array was used for transcriptome analysis. After comparing Sham/TMX- versus SCI/TMX-, 708 genes showed differential expression. The enriched pathways were the SCI pathway and pathways related to the inflammatory response. When comparing SCI/TMX- versus SCI/TMX+, only 30 genes showed differential expression, with no pathways enriched. Our results showed differential expression of genes related to the inflammatory response after SCI, and Tamoxifen seems to regulate gene expression changes in Ccr2 and Mmp12. Our study contributes data regarding the potential value of tamoxifen as a therapeutic resource for traumatic SCI during the acute phase.
Collapse
Affiliation(s)
- Eibar E. Cabrera-Aldana
- Department of Genomics Medicine, National Institute of Rehabilitation (INR), Calzada Mexico-Xochimilco 289, Arenal de Guadalupe, Mexico City 14389, Mexico; (E.E.C.-A.); (M.V.-F.); (M.S.-O.)
| | - Yalbi I. Balderas-Martínez
- Laboratorio de Biología Computacional, Instituto Nacional de Enfermedades Respiratorias, Ismael Cosío Villegas, Calz. de Tlalpan 4502, Belisario Domínguez Secc 16, Tlalpan, Mexico City 14080, Mexico;
| | - Rafael Velázquez-Cruz
- Genomics of Bone Metabolism Laboratory, National Institute of Genomic Medicine (INMEGEN), Periférico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico;
| | - Luis B. Tovar-y-Romo
- Department of Molecular Neuropathology, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Circuito Exterior s/n, Mexico City 04510, Mexico;
| | - Rosalba Sevilla-Montoya
- Reproductive Research and Perinatal Health Department, National Institute of Perinatology, Montes Urales 800, Lomas de Virreyes, Mexico City 11000, Mexico;
| | - Angelina Martínez-Cruz
- Department of Experimental Surgery, Proyecto Camina, A.C. 4430 Calz. Tlalpan, Mexico City 14050, Mexico;
| | - Claudia Martinez-Cordero
- Regional Hospital of High Specialty of the Bajio, Blvd. Milenio 130, Col. San Carlos la Roncha, León 37660, Guanajuato, Mexico;
| | - Margarita Valdés-Flores
- Department of Genomics Medicine, National Institute of Rehabilitation (INR), Calzada Mexico-Xochimilco 289, Arenal de Guadalupe, Mexico City 14389, Mexico; (E.E.C.-A.); (M.V.-F.); (M.S.-O.)
| | - Monica Santamaria-Olmedo
- Department of Genomics Medicine, National Institute of Rehabilitation (INR), Calzada Mexico-Xochimilco 289, Arenal de Guadalupe, Mexico City 14389, Mexico; (E.E.C.-A.); (M.V.-F.); (M.S.-O.)
| | - Alberto Hidalgo-Bravo
- Department of Genomics Medicine, National Institute of Rehabilitation (INR), Calzada Mexico-Xochimilco 289, Arenal de Guadalupe, Mexico City 14389, Mexico; (E.E.C.-A.); (M.V.-F.); (M.S.-O.)
| | - Gabriel Guízar-Sahagún
- Research Unit for Neurological Diseases, Instituto Mexicano del Seguro Social, 330 Avenida Cuauhtémoc, Mexico City 06720, Mexico
| |
Collapse
|
18
|
Jovanovic LI, Jervis Rademeyer H, Pakosh M, Musselman KE, Popovic MR, Marquez-Chin C. Scoping Review on Brain-Computer Interface-Controlled Electrical Stimulation Interventions for Upper Limb Rehabilitation in Adults: A Look at Participants, Interventions, and Technology. Physiother Can 2023; 75:276-290. [PMID: 37736411 PMCID: PMC10510539 DOI: 10.3138/ptc-2021-0074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/07/2021] [Accepted: 12/07/2021] [Indexed: 09/23/2023]
Abstract
Purpose While current rehabilitation practice for improving arm and hand function relies on physical/occupational therapy, a growing body of research evaluates the effects of technology-enhanced rehabilitation. We review interventions that combine a brain-computer interface (BCI) with electrical stimulation (ES) for upper limb movement rehabilitation to summarize the evidence on (1) populations of study participants, (2) BCI-ES interventions, and (3) the BCI-ES systems. Method After searching seven databases, two reviewers identified 23 eligible studies. We consolidated information on the study participants, interventions, and approaches used to develop integrated BCI-ES systems. The included studies investigated the use of BCI-ES interventions with stroke and spinal cord injury (SCI) populations. All studies used electroencephalography to collect brain signals for the BCI, and functional electrical stimulation was the most common type of ES. The BCI-ES interventions were typically conducted without a therapist, with sessions varying in both frequency and duration. Results Of the 23 eligible studies, only 3 studies involved the SCI population, compared to 20 involving individuals with stroke. Conclusions Future BCI-ES interventional studies could address this gap. Additionally, standardization of device and rehabilitation modalities, and study-appropriate involvement with therapists, can be considered to advance this intervention towards clinical implementation.
Collapse
Affiliation(s)
- Lazar I. Jovanovic
- From the:
Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- KITE Research Institute, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
- The Center for Advancing Neurotechnological Innovation to Application (CRANIA), University Health Network, Toronto, Canada
| | - Hope Jervis Rademeyer
- KITE Research Institute, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
- Rehabilitation Sciences Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Maureen Pakosh
- Library & Information Services, University Health Network, Toronto Rehabilitation Institute, Toronto, Canada
| | - Kristin E. Musselman
- KITE Research Institute, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
- Rehabilitation Sciences Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
- Department of Physical Therapy, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Milos R. Popovic
- From the:
Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- KITE Research Institute, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
- The Center for Advancing Neurotechnological Innovation to Application (CRANIA), University Health Network, Toronto, Canada
- Rehabilitation Sciences Institute, Temerty Faculty of Medicine, University of Toronto, Toronto, Canada
| | - Cesar Marquez-Chin
- From the:
Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- KITE Research Institute, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
- The Center for Advancing Neurotechnological Innovation to Application (CRANIA), University Health Network, Toronto, Canada
| |
Collapse
|
19
|
Li Y, Khan N, Ritzel RM, Lei Z, Allen S, Faden AI, Wu J. Sexually dimorphic extracellular vesicle responses after chronic spinal cord injury are associated with neuroinflammation and neurodegeneration in the aged brain. J Neuroinflammation 2023; 20:197. [PMID: 37653491 PMCID: PMC10469550 DOI: 10.1186/s12974-023-02881-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/27/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND Medical advances have made it increasingly possible for spinal cord injury (SCI) survivors to survive decades after the insult. But how SCI affects aging changes and aging impacts the injury process have received limited attention. Extracellular vesicles (EVs) are recognized as critical mediators of neuroinflammation after CNS injury, including at a distance from the lesion site. We have previously shown that SCI in young male mice leads to robust changes in plasma EV count and microRNA (miR) content. Here, our goal was to investigate the impact of biological sex and aging on EVs and brain after SCI. METHODS Young adult age-matched male and female C57BL/6 mice were subjected to SCI. At 19 months post-injury, total plasma EVs were isolated by ultracentrifugation and characterized by nanoparticle tracking analysis (NTA). EVs miR cargo was examined using the Fireplex® assay. The transcriptional changes in the brain were assessed by a NanoString nCounter Neuropathology panel and validated by Western blot (WB) and flow cytometry (FC). A battery of behavioral tests was performed for assessment of neurological function. RESULTS Transcriptomic changes showed a high number of changes between sham and those with SCI. Sex-specific changes were found in transcription networks related to disease association, activated microglia, and vesicle trafficking. FC showed higher microglia and myeloid counts in the injured tissue of SCI/Female compared to their male counterparts, along with higher microglial production of ROS in both injured site and the brain. In the latter, increased levels of TNF and mitochondrial membrane potential were seen in microglia from SCI/Female. WB and NTA revealed that EV markers are elevated in the plasma of SCI/Male. Particle concentration in the cortex increased after injury, with SCI/Female showing higher counts than SCI/Male. EVs cargo analysis revealed changes in miR content related to injury and sex. Behavioral testing confirmed impairment of cognition and depression at chronic time points after SCI in both sexes, without significant differences between males and females. CONCLUSIONS Our study is the first to show sexually dimorphic changes in brain after very long-term SCI and supports a potential sex-dependent EV-mediated mechanism that contributes to SCI-induced brain changes.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Niaz Khan
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Rodney M Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Samantha Allen
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, 685 W. Baltimore Street, MSTF, Room 6-034D, Baltimore, MD, 21201, USA.
| |
Collapse
|
20
|
Fouad K, Wireman OH, Gensel JC. Data and subject heterogeneity and data sharing: keys to translational success in spinal cord injury research? Neural Regen Res 2023; 18:1730-1731. [PMID: 36751797 PMCID: PMC10154489 DOI: 10.4103/1673-5374.363191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/20/2022] [Accepted: 11/12/2022] [Indexed: 12/14/2022] Open
Affiliation(s)
- Karim Fouad
- Faculty of Rehabilitation Medicine, Department of Physical Therapy, Neuroscience and Mental Health Institute, University of Alberta, Alberta, Canada
| | - Olivia H. Wireman
- Spinal Cord and Brain Injury Research Center (SCoBIRC), Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - John C. Gensel
- Spinal Cord and Brain Injury Research Center (SCoBIRC), Department of Physiology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
21
|
Phillips AK, Keller MF, McClung JP, Steele N, Witkop CT, Wu TJ. Physical Health and Well-being: Updates and the Way Ahead. Mil Med 2023; 188:9-18. [PMID: 37490559 DOI: 10.1093/milmed/usac370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 02/24/2022] [Accepted: 11/14/2022] [Indexed: 07/27/2023] Open
Abstract
INTRODUCTION The Women in Combat Summit 2021 "Forging the Future: How Women Enhance the Fighting Force" took place during February 9-11, 2021, via a virtual conference platform. The third and final day of the Summit regarded the physical health and well-being of military women and included the topics of urogenital health, nutrition and iron-deficiency anemia, unintended pregnancy and contraception, and traumatic brain injury. MATERIALS AND METHODS After presentations on the topics earlier, interested conference attendees were invited to participate in focus groups to discuss and review policy recommendations for physical health and well-being in military women. Discussions centered around the topics discussed during the presentations, and suggestions for future Women in Combat Summits were noted. Specifics of the methods of the Summit are presented elsewhere in this supplement. RESULTS We formulated research and policy recommendations for urogenital health, nutrition and iron-deficiency anemia, contraception and unintended pregnancy, and traumatic brain injury. CONCLUSIONS In order to continue to develop the future health of military women, health care providers, researchers, and policymakers should consider the recommendations made in this supplement as they continue to build on the state of the science and forge the future.
Collapse
Affiliation(s)
- Angela K Phillips
- Malcolm Grow Medical Clinics and Surgery Center, Joint Base Andrews, MD 20762, USA
| | - Margaux F Keller
- Henry Jackson Foundation at the Daniel K. Inouye Graduate School of Nursing, Uniformed Services University, Bethesda, MD 20814, USA
| | - James P McClung
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA 01760, USA
| | - Nancy Steele
- School of Nursing, University of North Florida, Jacksonville, FL 32224, USA
| | - Catherine T Witkop
- Department of Preventive Medicine and Biostatistics, Uniformed Services University, Bethesda, MD 20814, USA
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University, Bethesda, MD 20814, USA
| | - T John Wu
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
22
|
Askarifirouzjaei H, Khajoueinejad L, Wei E, Cheruvu S, Ayala C, Chiang N, Theis T, Sun D, Fazeli M, Young W. Sex Differences in Immune Cell Infiltration and Hematuria in SCI-Induced Hemorrhagic Cystitis. PATHOPHYSIOLOGY 2023; 30:275-295. [PMID: 37489403 PMCID: PMC10366728 DOI: 10.3390/pathophysiology30030023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/26/2023] Open
Abstract
Rats manifest a condition called hemorrhagic cystitis after spinal cord injury (SCI). The mechanism of this condition is unknown, but it is more severe in male rats than in female rats. We assessed the role of sex regarding hemorrhagic cystitis and pathological chronic changes in the bladder. We analyzed the urine of male and female Sprague-Dawley and Fischer 344 rats after experimental spinal cord contusion, including unstained microscopic inspections of the urine, differential white blood cell counts colored by the Wright stain, and total leukocyte counts using fluorescent nuclear stains. We examined bladder histological changes in acute and chronic phases of SCI, using principal component analysis (PCA) and clustered heatmaps of Pearson correlation coefficients to interpret how measured variables correlated with each other. Male rats showed a distinct pattern of macroscopic hematuria after spinal cord injury. They had higher numbers of red blood cells with significantly more leukocytes and neutrophils than female rats, particularly hypersegmented neutrophils. The histological examination of the bladders revealed a distinct line of apoptotic umbrella cells and disrupted bladder vessels early after SCI and progressive pathological changes in multiple bladder layers in the chronic phase. Multivariate analyses indicated immune cell infiltration in the bladder, especially hypersegmented neutrophils, that correlated with red blood cell counts in male rats. Our study highlights a hitherto unreported sex difference of hematuria and pathological changes in males and females' bladders after SCI, suggesting an important role of immune cell infiltration, especially neutrophils, in SCI-induced hemorrhagic cystitis.
Collapse
Affiliation(s)
- Hadi Askarifirouzjaei
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Leila Khajoueinejad
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
- Department of Pharmacology, School of Veterinary Medicine, Shiraz University, Shiraz 71345, Iran
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Elena Wei
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Sruti Cheruvu
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Carlos Ayala
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Ning Chiang
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Dongming Sun
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Mehdi Fazeli
- Department of Pharmacology, School of Veterinary Medicine, Shiraz University, Shiraz 71345, Iran
| | - Wise Young
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| |
Collapse
|
23
|
Gilbert EAB, Livingston J, Garcia-Flores E, Kehtari T, Morshead CM. Metformin Improves Functional Outcomes, Activates Neural Precursor Cells, and Modulates Microglia in a Sex-Dependent Manner After Spinal Cord Injury. Stem Cells Transl Med 2023:7174953. [PMID: 37209417 DOI: 10.1093/stcltm/szad030] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 04/20/2023] [Indexed: 05/22/2023] Open
Abstract
Spinal cord injury (SCI) results in devastating patient outcomes with few treatment options. A promising approach to improve outcomes following SCI involves the activation of endogenous precursor populations including neural stem and progenitor cells (NSPCs) which are located in the periventricular zone (PVZ), and oligodendrocyte precursor cells (OPCs) found throughout the parenchyma. In the adult spinal cord, resident NSPCs are primarily mitotically quiescent and aneurogenic, while OPCs contribute to ongoing oligodendrogenesis into adulthood. Each of these populations is responsive to SCI, increasing their proliferation and migration to the site of injury; however, their activation is not sufficient to support functional recovery. Previous work has shown that administration of the FDA-approved drug metformin is effective at promoting endogenous brain repair following injury, and this is correlated with enhanced NSPC activation. Here, we ask whether metformin can promote functional recovery and neural repair following SCI in both males and females. Our results reveal that acute, but not delayed metformin administration improves functional outcomes following SCI in both sexes. The functional improvement is concomitant with OPC activation and oligodendrogenesis. Our data also reveal sex-dependent effects of metformin following SCI with increased activation of NSPCs in females and reduced microglia activation in males. Taken together, these findings support metformin as a viable therapeutic strategy following SCI and highlight its pleiotropic effects in the spinal cord.
Collapse
Affiliation(s)
- Emily A B Gilbert
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Jessica Livingston
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Emilio Garcia-Flores
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Tarlan Kehtari
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
| | - Cindi M Morshead
- Division of Anatomy, Department of Surgery, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
| |
Collapse
|
24
|
Stewart AN, Kumari R, Bailey WM, Glaser EP, Hammers GV, Wireman OH, Gensel JC. PTEN knockout using retrogradely transported AAVs restores locomotor abilities in both acute and chronic spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537179. [PMID: 37131840 PMCID: PMC10153160 DOI: 10.1101/2023.04.17.537179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Restoring function in chronic stages of spinal cord injury (SCI) has often been met with failure or reduced efficacy when regenerative strategies are delayed past the acute or sub-acute stages of injury. Restoring function in the chronically injured spinal cord remains a critical challenge. We found that a single injection of retrogradely transported adeno-associated viruses (AAVrg) to knockout the phosphatase and tensin homolog protein (PTEN) in chronic SCI can effectively target both damaged and spared axons and restore locomotor functions in near-complete injury models. AAVrg's were injected to deliver cre recombinase and/or a red fluorescent protein (RFP) under the human Synapsin 1 promoter (hSyn1) into the spinal cords of C57BL/6 PTEN FloxΔ / Δ mice to knockout PTEN (PTEN-KO) in a severe thoracic SCI crush model at both acute and chronic time points. PTEN-KO improved locomotor abilities in both acute and chronic SCI conditions over a 9-week period. Regardless of whether treatment was initiated at the time of injury (acute), or three months after SCI (chronic), mice with limited hindlimb joint movement gained hindlimb weight support after treatment. Interestingly, functional improvements were not sustained beyond 9 weeks coincident with a loss of RFP reporter-gene expression and a near-complete loss of treatment-associated functional recovery by 6 months post-treatment. Treatment effects were also specific to severely injured mice; animals with weight support at the time of treatment lost function over a 6-month period. Retrograde tracing with Fluorogold revealed viable neurons throughout the motor cortex despite a loss of RFP expression at 9 weeks post-PTEN-KO. However, few Fluorogold labeled neurons were detected within the motor cortex at 6 months post-treatment. BDA labeling from the motor cortex revealed a dense corticospinal tract (CST) bundle in all groups except chronically treated PTEN-KO mice indicating a potential long-term toxic effect of PTEN-KO to neurons in the motor cortex. PTEN-KO mice had significantly more β - tubulin III labeled axons within the lesion when treatment was delivered acutely, but not chronically post-SCI. In conclusion, we have found that using AAVrg's to knockout PTEN is an effective manipulation capable of restoring motor functions in chronic SCI and can enhance axon growth of currently unidentified axon populations when delivered acutely after injury. However, the long-term consequences of PTEN-KO may exert neurotoxic effects.
Collapse
Affiliation(s)
- Andrew N. Stewart
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Reena Kumari
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - William M. Bailey
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Ethan P. Glaser
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Gabrielle V. Hammers
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - Olivia H. Wireman
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
| | - John C. Gensel
- Department of Physiology, University of Kentucky, Lexington, Kentucky 40536, USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, Kentucky 40536, USA
- College of Medicine, University of Kentucky, Lexington, Kentucky 40536, USA
| |
Collapse
|
25
|
Liu R, Li Y, Wang Z, Chen P, Xie Y, Qu W, Wang M, Yu Z, Luo X. Regulatory T cells promote functional recovery after spinal cord injury by alleviating microglia inflammation via STAT3 inhibition. CNS Neurosci Ther 2023. [PMID: 36914969 DOI: 10.1111/cns.14161] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 02/08/2023] [Accepted: 02/24/2023] [Indexed: 03/15/2023] Open
Abstract
BACKGROUND Immediately after spinal trauma, immune cells, and proinflammatory cytokines infiltrate the spinal cord and disrupt the focal microenvironment, which impedes axon regeneration and functional recovery. Previous studies have reported that regulatory T cells (Tregs) enter the central nervous system and exert immunosuppressive effects on microglia during multiple sclerosis and stroke. However, whether and how Tregs interact with microglia and modulate injured microenvironments after spinal cord injury (SCI) remains unknown. METHOD Regulatory T cells spatiotemporal characteristics were analyzed in a mouse contusion SCI model. Microglia activation status was evaluated by immunostaining and RNA sequencing. Cytokine production in injured spinal cord was examined using Luminex. The role of STAT3 in Treg-microglia crosstalk was investigated in a transwell system with isolated Tregs and primary microglia. RESULTS Regulatory T cells infiltration of the spinal cord peaked on day 7 after SCI. Treg depletion promoted microglia switch to a proinflammatory phenotype. Inflammation-related genes, such as ApoD, as well as downstream cytokines IL-6 and TNF-α were upregulated in microglia in Treg-depleted mice. STAT3 inhibition was involved in Treg-microglia crosstalk, and STAT3 chemical blockade improved function recovery in Treg-depleted mice. CONCLUSION Our results suggest that Tregs promote functional recovery after SCI by alleviating microglia inflammatory reaction via STAT3.
Collapse
Affiliation(s)
- Rui Liu
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Li
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ziyue Wang
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Chen
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Xie
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wensheng Qu
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Minghuan Wang
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyuan Yu
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Luo
- Department of Neurology, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.,Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Cheng L, Cai B, Lu D, Zeng H. The role of mitochondrial energy metabolism in neuroprotection and axonal regeneration after spinal cord injury. Mitochondrion 2023; 69:57-63. [PMID: 36740158 DOI: 10.1016/j.mito.2023.01.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/12/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
Mitochondrial dysfunction occurs in the early stage of axonal degeneration after spinal cord injury and involves oxidative stress, energy deficiency, imbalance of mitochondrial dynamics, etc., which play a key role in axonal degeneration and regeneration under physiological and pathological conditions. Failure of axonal regeneration can lead to long-term structural and functional damage. Several recent studies have shown that improved mitochondrial energy metabolism provides conditions for axonal regeneration and central nervous system repair. Here, we describe the role of mitochondrial energy metabolism in neuroprotection and axonal regeneration after spinal cord injury and review recent advances in targeted mitochondrial therapy.
Collapse
Affiliation(s)
- Li Cheng
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Bin Cai
- Department of Rehabilitation Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dezhi Lu
- School of Medicine, Shanghai University, Shanghai, China
| | - Hong Zeng
- Department of Rehabilitation Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Furlan JC, Shen T, Kurban D. Sex-Related Discrepancies in the Access to Optimal Care and Outcomes After Traumatic Spinal Cord Injury: A Retrospective Cohort Study Using Data From a Canadian Registry. Arch Phys Med Rehabil 2023; 104:1-10. [PMID: 36170894 DOI: 10.1016/j.apmr.2022.09.006] [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: 07/08/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To compare males and females who were stratified into subgroups corresponding to premenopausal, perimenopausal, and postmenopausal ages, regarding access to optimal care and their outcomes after traumatic spinal cord injury (tSCI). STUDY DESIGN Retrospective cohort study. SETTING Eighteen acute care centers and 13 rehabilitation facilities across Canada. PARTICIPANTS This study included 5571 individuals with tSCI at C1-L2 who were enrolled in the Rick Hansen Spinal Cord Injury Registry from July 2004 to September 2019 (N=5571). Females were compared with males in the younger (aged ≤40 years), middle-aged (ages 41-50), and older (aged >50 years) subgroups. INTERVENTION Not applicable. MAIN OUTCOME MEASURES Females were compared with males in each subgroup with regard to their demographic data, pre-existing comorbidities, injury characteristics, management choices, access to optimal care, and clinical, neurologic, and functional outcomes after tSCI. RESULTS In the younger subgroups, females (n=408) were significantly younger, had a greater proportion of aboriginals and transportation-related tSCIs, underwent surgical treatment more often, and had a greater sensory score change than males (n=1613). In the middle-aged subgroups, females (n=174) had a greater proportion of high-thoracic tSCIs than males (n=666). In the older subgroups, females (n=660) were significantly older, had more fall-related and less severe tSCIs, had a shorter stay at the rehabilitation center, had less spasticity, and were discharged home less often than males (n=2050). CONCLUSIONS The results of this study suggest some sex-related differences in individuals' demographics and injury characteristics, but fewer discrepancies between females and males regarding their access to optimal care and outcomes after tSCI. Overall, future clinical trials could consider inclusion of males and females of all age groups to enhance recruitment and augment generalizability.
Collapse
Affiliation(s)
- Julio C Furlan
- Lyndhurst Centre, Toronto Rehabilitation Institute, University Health Network, Toronto, Canada; KITE Research Institute, University Health Network, Toronto, Canada; Department of Medicine, Division of Physical Medicine and Rehabilitation, University of Toronto, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Rehabilitation Sciences Institute, University of Toronto, Toronto, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Canada.
| | - Tian Shen
- Praxis Spinal Cord Institute, Vancouver, Canada
| | | |
Collapse
|
28
|
Stewart AN, Jones LAT, Gensel JC. Improving translatability of spinal cord injury research by including age as a demographic variable. Front Cell Neurosci 2022; 16:1017153. [PMID: 36467608 PMCID: PMC9714671 DOI: 10.3389/fncel.2022.1017153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Pre-clinical and clinical spinal cord injury (SCI) studies differ in study design, particularly in the demographic characteristics of the chosen population. In clinical study design, criteria such as such as motor scores, neurological level, and severity of injury are often key determinants for participant inclusion. Further, demographic variables in clinical trials often include individuals from a wide age range and typically include both sexes, albeit historically most cases of SCI occur in males. In contrast, pre-clinical SCI models predominately utilize young adult rodents and typically use only females. While it is often not feasible to power SCI clinical trials to test multi-variable designs such as contrasting different ages, recent pre-clinical findings in SCI animal models have emphasized the importance of considering age as a biological variable prior to human experiments. Emerging pre-clinical data have identified case examples of treatments that diverge in efficacy across different demographic variables and have elucidated several age-dependent effects in SCI. The extent to which these differing or diverging treatment responses manifest clinically can not only complicate statistical findings and trial interpretations but also may be predictive of worse outcomes in select clinical populations. This review highlights recent literature including age as a biological variable in pre-clinical studies and articulates the results with respect to implications for clinical trials. Based on emerging unpredictable treatment outcomes in older rodents, we argue for the importance of including age as a biological variable in pre-clinical animal models prior to clinical testing. We believe that careful analyses of how age interacts with SCI treatments and pathophysiology will help guide clinical trial design and may improve both the safety and outcomes of such important efforts.
Collapse
Affiliation(s)
- Andrew N. Stewart
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States,Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Linda A. T. Jones
- Center for Outcomes and Measurement, Jefferson College of Rehabilitation Sciences, Thomas Jefferson University, Philadelphia, PA, United States
| | - John C. Gensel
- Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY, United States,Spinal Cord and Brain Injury Research Center, College of Medicine, University of Kentucky, Lexington, KY, United States,*Correspondence: John C. Gensel,
| |
Collapse
|
29
|
Early Changes in Androgen Levels in Individuals with Spinal Cord Injury: A Longitudinal SwiSCI Study. J Clin Med 2022; 11:jcm11216559. [DOI: 10.3390/jcm11216559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/18/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
We aimed to explore longitudinal changes in androgen levels in individuals with spinal cord injury (SCI) within initial inpatient rehabilitation stay and identify clinical/injury characteristics associated with hormone levels. Linear regression analysis was applied to explore the association between personal/injury characteristics and androgen hormones (total testosterone, free testosterone, sex hormone-binding globulin (SHBG), dehydroepiandrosterone (DHEA), and dehydroepiandrosterone sulfate (DHEA-S)) at admission to rehabilitation. Longitudinal changes in androgen levels were studied using linear mixed models. Analyses were stratified by sex and by injury type. We included 70 men and 16 women with SCI. We observed a non-linear association between age, time since injury, and androgens at baseline. At admission to initial rehabilitation, mature serum SHBG (full-length, protein form which lacks the N-terminal signaling peptide) was higher, while DHEA and DHEA-S were lower among opioid users vs. non-users. Serum levels of total testosterone and DHEA-S increased over rehabilitation period [β 3.96 (95%CI 1.37, 6.56), p = 0.003] and [β 1.77 (95%CI 0.73, 2.81), p = 0.01], respectively. We observed no significant changes in other androgens. Restricting our analysis to men with traumatic injury did not materially change our findings. During first inpatient rehabilitation over a median follow up of 5.6 months, we observed an increase in total testosterone and DHEA-S in men with SCI. Future studies need to explore whether these hormonal changes influence neurological and functional recovery as well as metabolic parameters during initial rehabilitation stay.
Collapse
|
30
|
Pitonak M, Aceves M, Kumar PA, Dampf G, Green P, Tucker A, Dietz V, Miranda D, Letchuman S, Jonika MM, Bautista D, Blackmon H, Dulin JN. Effects of biological sex mismatch on neural progenitor cell transplantation for spinal cord injury in mice. Nat Commun 2022; 13:5380. [PMID: 36104357 PMCID: PMC9474813 DOI: 10.1038/s41467-022-33134-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 09/02/2022] [Indexed: 12/03/2022] Open
Abstract
Despite advancement of neural progenitor cell transplantation to spinal cord injury clinical trials, there remains a lack of understanding of how biological sex of transplanted cells influences outcomes after transplantation. To address this, we transplanted GFP-expressing sex-matched, sex-mismatched, or mixed donor cells into sites of spinal cord injury in adult male and female mice. Biological sex of the donor cells does not influence graft neuron density, glial differentiation, formation of the reactive glial cell border, or graft axon outgrowth. However, male grafts in female hosts feature extensive hypervascularization accompanied by increased vascular diameter and perivascular cell density. We show greater T-cell infiltration within male-to-female grafts than other graft types. Together, these findings indicate a biological sex-specific immune response of female mice to male donor cells. Our work suggests that biological sex should be considered in the design of future clinical trials for cell transplantation in human injury. In this study, Pitonak et al. report that transplantation of neural progenitor cells derived from male donors trigger an immune rejection response following transplantation into sites of spinal cord injury in female mice.
Collapse
|
31
|
Osimanjiang W, Allgood JE, Van Sandt RL, Burns DT, Bushman JS. Sexual Dimorphism in Lesion Size and Sensorimotor Responses Following Spinal Cord Injury. Front Neurol 2022; 13:925797. [PMID: 36994113 PMCID: PMC10041393 DOI: 10.3389/fneur.2022.925797] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 05/31/2022] [Indexed: 03/14/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating disorder, which impacts the lives of millions of people worldwide with no clinically standardized treatment. Both pro-recovery and anti-recovery factors contribute to the overall outcome after the initial SCI. Sex is emerging as an important variable, which can affect recovery post-SCI. Contusion SCI at T10 was generated in male and female rats. Open-field Basso, Beattie, Bresnahan (BBB) behavioral test, Von Frey test, and CatWalk gate analysis were performed. Histological analysis was performed at the 45-day post-SCI end point. Male/female differences in sensorimotor function recovery, lesion size, and the recruitment of immune cells to the lesion area were measured. A group of males with less severe injuries was included to compare the outcomes for severity. Our results show that both sexes with the same injury level plateaued at a similar final score for locomotor function. Males in the less severe injury group recovered faster and plateaued at a higher BBB score compared to the more severe injury group. Von Frey tests show faster recovery of sensory function in females compared to both male groups. All three groups exhibited reduced mechanical response thresholds after SCI. The lesion area was significantly larger in the male group with severe injury than in females, as well as in males of less severe injury. No significant differences in immune cell recruitment were identified when comparing the three groups. The faster sensorimotor recovery and significantly smaller lesion area in females potentially indicate that neuroprotection against the secondary injury is a likely reason for sex-dependent differences in functional outcomes after SCI.
Collapse
Affiliation(s)
- Wupu Osimanjiang
- Division of Pharmaceutical Sciences, University of Wyoming, Laramie, WY, United States
| | - JuliAnne E. Allgood
- Division of Pharmaceutical Sciences, University of Wyoming, Laramie, WY, United States
| | - Rae L. Van Sandt
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY, United States
| | - Daniel T. Burns
- Division of Pharmaceutical Sciences, University of Wyoming, Laramie, WY, United States
| | - Jared S. Bushman
- Division of Pharmaceutical Sciences, University of Wyoming, Laramie, WY, United States
- *Correspondence: Jared S. Bushman
| |
Collapse
|
32
|
Mun S, Han K, Hyun JK. The Time Sequence of Gene Expression Changes after Spinal Cord Injury. Cells 2022; 11:cells11142236. [PMID: 35883679 PMCID: PMC9324287 DOI: 10.3390/cells11142236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/15/2022] [Accepted: 07/17/2022] [Indexed: 02/01/2023] Open
Abstract
Gene expression changes following spinal cord injury (SCI) are time-dependent, and an accurate understanding of these changes can be crucial in determining time-based treatment options in a clinical setting. We performed RNA sequencing of the contused spinal cord of rats at five different time points from the very acute to chronic stages (1 hour, 1 day, 1 week, 1 month, and 3 months) following SCI. We identified differentially expressed genes (DEGs) and Gene Ontology (GO) terms at each time point, and 14,257 genes were commonly expressed at all time points. The biological process of the inflammatory response was increased at 1 hour and 1 day, and the cellular component of the integral component of the synaptic membrane was increased at 1 day. DEGs associated with cell activation and the innate immune response were highly enriched at 1 week and 1 month, respectively. A total of 2841 DEGs were differentially expressed at any of the five time points, and 18 genes (17 upregulated and 1 downregulated) showed common expression differences at all time points. We found that interleukin signaling, neutrophil degranulation, eukaryotic translation, collagen degradation, LGI–ADAM interactions, GABA receptor, and L1CAM-ankyrin interactions were prominent after SCI depending on the time post injury. We also performed gene–drug network analysis and found several potential antagonists and agonists which can be used to treat SCI. We expect to discover effective treatments in the clinical field through further studies revealing the efficacy and safety of potential drugs.
Collapse
Affiliation(s)
- Seyoung Mun
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea;
- Center for Bio Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea;
| | - Kyudong Han
- Center for Bio Medical Engineering Core Facility, Dankook University, Cheonan 31116, Korea;
- Department of Microbiology, College of Science & Technology, Dankook University, Cheonan 31116, Korea
| | - Jung Keun Hyun
- Department of Nanobiomedical Science & BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Korea;
- Department of Rehabilitation Medicine, College of Medicine, Dankook University, Cheonan 31116, Korea
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Correspondence: ; Tel.: +82-10-2293-3415
| |
Collapse
|
33
|
Zhao C, Zhou T, Zhao X, Pang Y, Li W, Fan B, Li M, Liu X, Ma L, Zhang J, Sun C, Shen W, Kong X, Yao X, Feng S. Delayed administration of nafamostat mesylate inhibits thrombin-mediated blood-spinal cord barrier breakdown during acute spinal cord injury in rats. J Neuroinflammation 2022; 19:189. [PMID: 35842640 PMCID: PMC9287720 DOI: 10.1186/s12974-022-02531-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 06/15/2022] [Indexed: 01/10/2023] Open
Abstract
Background Nafamostat mesylate (nafamostat, NM) is an FDA-approved serine protease inhibitor that exerts anti-neuroinflammation and neuroprotective effects following rat spinal cord injury (SCI). However, clinical translation of nafamostat has been limited by an unclear administration time window and mechanism of action. Methods Time to first dose of nafamostat administration was tested on rats after contusive SCI. The optimal time window of nafamostat was screened by evaluating hindlimb locomotion and electrophysiology. As nafamostat is a serine protease inhibitor known to target thrombin, we used argatroban (Arg), a thrombin-specific inhibitor, as a positive control in the time window experiments. Western blot and immunofluorescence of thrombin expression level and its enzymatic activity were assayed at different time points, as well its receptor, the protease activated receptor 1 (PAR1) and downstream protein matrix metalloproteinase-9 (MMP9). Blood–spinal cord barrier (BSCB) permeability leakage indicator Evans Blue and fibrinogen were analyzed along these time points. The infiltration of peripheral inflammatory cell was observed by immunofluorescence. Results The optimal administration time window of nafamostat was 2–12 h post-injury. Argatroban, the thrombin-specific inhibitor, had a similar pattern. Thrombin expression peaked at 12 h and returned to normal level at 7 days post-SCI. PAR1, the thrombin receptor, and MMP9 were significantly upregulated after SCI. The most significant increase of thrombin expression was detected in vascular endothelial cells (ECs). Nafamostat and argatroban significantly downregulated thrombin and MMP9 expression as well as thrombin activity in the spinal cord. Nafamostat inhibited thrombin enrichment in endothelial cells. Nafamostat administration at 2–12 h after SCI inhibited the leakage of Evans Blue in the epicenter and upregulated tight junction proteins (TJPs) expression. Nafamostat administration 8 h post-SCI effectively inhibited the infiltration of peripheral macrophages and neutrophils to the injury site. Conclusions Our study provides preclinical information of nafamostat about the administration time window of 2–12 h post-injury in contusive SCI. We revealed that nafamostat functions through inhibiting the thrombin-mediated BSCB breakdown and subsequent peripheral immune cells infiltration. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02531-w.
Collapse
Affiliation(s)
- Chenxi Zhao
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China.,Department of Orthopedics, Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Tiangang Zhou
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Xiaoqing Zhao
- Department of Orthopedics, Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yilin Pang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Wenxiang Li
- Department of Orthopedics, Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Baoyou Fan
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Ming Li
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Xinjie Liu
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Lei Ma
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Jiawei Zhang
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Chao Sun
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Wenyuan Shen
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China
| | - Xiaohong Kong
- Department of Orthopedics, Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xue Yao
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China. .,Department of Orthopedics, Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Shiqing Feng
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, China. .,Department of Orthopedics, Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
34
|
Brennan FH, Li Y, Wang C, Ma A, Guo Q, Li Y, Pukos N, Campbell WA, Witcher KG, Guan Z, Kigerl KA, Hall JCE, Godbout JP, Fischer AJ, McTigue DM, He Z, Ma Q, Popovich PG. Microglia coordinate cellular interactions during spinal cord repair in mice. Nat Commun 2022; 13:4096. [PMID: 35835751 PMCID: PMC9283484 DOI: 10.1038/s41467-022-31797-0] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/01/2022] [Indexed: 12/27/2022] Open
Abstract
Traumatic spinal cord injury (SCI) triggers a neuro-inflammatory response dominated by tissue-resident microglia and monocyte derived macrophages (MDMs). Since activated microglia and MDMs are morphologically identical and express similar phenotypic markers in vivo, identifying injury responses specifically coordinated by microglia has historically been challenging. Here, we pharmacologically depleted microglia and use anatomical, histopathological, tract tracing, bulk and single cell RNA sequencing to reveal the cellular and molecular responses to SCI controlled by microglia. We show that microglia are vital for SCI recovery and coordinate injury responses in CNS-resident glia and infiltrating leukocytes. Depleting microglia exacerbates tissue damage and worsens functional recovery. Conversely, restoring select microglia-dependent signaling axes, identified through sequencing data, in microglia depleted mice prevents secondary damage and promotes recovery. Additional bioinformatics analyses reveal that optimal repair after SCI might be achieved by co-opting key ligand-receptor interactions between microglia, astrocytes and MDMs.
Collapse
Affiliation(s)
- Faith H Brennan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Yang Li
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Cankun Wang
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Anjun Ma
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Qi Guo
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Yi Li
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Nicole Pukos
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Warren A Campbell
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristina G Witcher
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Zhen Guan
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Kristina A Kigerl
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Jodie C E Hall
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Jonathan P Godbout
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Andy J Fischer
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Dana M McTigue
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Qin Ma
- Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
- Belford Center for Spinal Cord Injury, Center for Brain and Spinal Cord Repair, The Ohio State University Wexner Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
35
|
The Inflammatory Response after Moderate Contusion Spinal Cord Injury: A Time Study. BIOLOGY 2022; 11:biology11060939. [PMID: 35741460 PMCID: PMC9220050 DOI: 10.3390/biology11060939] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/30/2022] [Accepted: 06/17/2022] [Indexed: 11/25/2022]
Abstract
Simple Summary The neuroinflammatory response is a rather complex event in spinal cord injury (SCI) and has the capacity to exacerbate cell damage but also to contribute to the repair of the injury. This complexity is thought to depend on a variety of inflammatory mediators, of which tumor necrosis factor (TNF) plays a key role. Evidence indicates that TNF can be both protective and detrimental in SCI. In the present study, we studied the temporal and cellular expression of TNF and its receptors after SCI in mice. We found TNF to be significantly increased in both the acute and the delayed phases after SCI, alongside a robust neuroinflammatory response. As we could verify some of our results in human postmortem tissue, our results imply that diminishing the detrimental immune signaling after SCI could also enhance recovery in humans. Abstract Spinal cord injury (SCI) initiates detrimental cellular and molecular events that lead to acute and delayed neuroinflammation. Understanding the role of the inflammatory response in SCI requires insight into the temporal and cellular synthesis of inflammatory mediators. We subjected C57BL/6J mice to SCI and investigated inflammatory reactions. We examined activation, recruitment, and polarization of microglia and infiltrating immune cells, focusing specifically on tumor necrosis factor (TNF) and its receptors TNFR1 and TNFR2. In the acute phase, TNF expression increased in glial cells and neuron-like cells, followed by infiltrating immune cells. TNFR1 and TNFR2 levels increased in the delayed phase and were found preferentially on neurons and glial cells, respectively. The acute phase was dominated by the infiltration of granulocytes and macrophages. Microglial/macrophage expression of Arg1 increased from 1–7 days after SCI, followed by an increase in Itgam, Cx3cr1, and P2ry12, which remained elevated throughout the study. By 21 and 28 days after SCI, the lesion core was populated by galectin-3+, CD68+, and CD11b+ microglia/macrophages, surrounded by a glial scar consisting of GFAP+ astrocytes. Findings were verified in postmortem tissue from individuals with SCI. Our findings support the consensus that future neuroprotective immunotherapies should aim to selectively neutralize detrimental immune signaling while sustaining pro-regenerative processes.
Collapse
|
36
|
Sartoretti T, Ganley RP, Ni R, Freund P, Zeilhofer HU, Klohs J. Structural MRI Reveals Cervical Spinal Cord Atrophy in the P301L Mouse Model of Tauopathy: Gender and Transgene-Dosing Effects. Front Aging Neurosci 2022; 14:825996. [PMID: 35585865 PMCID: PMC9108240 DOI: 10.3389/fnagi.2022.825996] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
In primary tauopathies, the deposition of tau neurofibrillary tangles and threads as well as neurodegenerative changes have been found within the brain and spinal cord. While degenerative changes have been intensively studied in the brain using structural magnetic resonance imaging (MRI), MRI studies investigating the spinal cord are still scarce. In the present study, we acquired ex vivo high resolution structural MRI of the cervical spinal cord of 8.5–9 month old hemizygous and homozygous P301L mice and non-transgenic littermates of both genders. We assessed the total cross-sectional area, and the gray and white matter anterior-posterior width and left-right width that are established imaging marker of spinal cord degeneration. We observed significant tissue-specific reductions in these parameters in female P301L mice that were stronger in homozygous than in hemizygous P301L mice, indicating both an effect of gender and transgene expression on cervical spinal cord atrophy. Moreover, atrophy was stronger in the gray matter than in the white matter. Immunohistochemical analysis revealed neurodegenerative and neuroinflammatory changes in the cervical spinal cord in both the gray and white matter of P301L mice. Collectively, our results provide evidence for cervical spinal cord atrophy that may directly contribute to the motor signs associated with tauopathy.
Collapse
Affiliation(s)
- Thomas Sartoretti
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
| | - Robert P. Ganley
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| | - Patrick Freund
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
- Balgrist University Hospital, University of Zurich, Zurich, Switzerland
| | - Hanns Ulrich Zeilhofer
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
- Institute for Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, ETH & University of Zurich, Zurich, Switzerland
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
- *Correspondence: Jan Klohs
| |
Collapse
|
37
|
Van Broeckhoven J, Erens C, Sommer D, Scheijen E, Sanchez S, Vidal PM, Dooley D, Van Breedam E, Quarta A, Ponsaerts P, Hendrix S, Lemmens S. Macrophage-based delivery of interleukin-13 improves functional and histopathological outcomes following spinal cord injury. J Neuroinflammation 2022; 19:102. [PMID: 35488301 PMCID: PMC9052547 DOI: 10.1186/s12974-022-02458-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 04/07/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) elicits a robust neuroinflammatory reaction which, in turn, exacerbates the initial mechanical damage. Pivotal players orchestrating this response are macrophages (Mφs) and microglia. After SCI, the inflammatory environment is dominated by pro-inflammatory Mφs/microglia, which contribute to secondary cell death and prevent regeneration. Therefore, reprogramming Mφ/microglia towards a more anti-inflammatory and potentially neuroprotective phenotype has gained substantial therapeutic interest in recent years. Interleukin-13 (IL-13) is a potent inducer of such an anti-inflammatory phenotype. In this study, we used genetically modified Mφs as carriers to continuously secrete IL-13 (IL-13 Mφs) at the lesion site. METHODS Mφs were genetically modified to secrete IL-13 (IL-13 Mφs) and were phenotypically characterized using qPCR, western blot, and ELISA. To analyze the therapeutic potential, the IL-13 Mφs were intraspinally injected at the perilesional area after hemisection SCI in female mice. Functional recovery and histopathological improvements were evaluated using the Basso Mouse Scale score and immunohistochemistry. Neuroprotective effects of IL-13 were investigated using different cell viability assays in murine and human neuroblastoma cell lines, human neurospheroids, as well as murine organotypic brain slice cultures. RESULTS In contrast to Mφs prestimulated with recombinant IL-13, perilesional transplantation of IL-13 Mφs promoted functional recovery following SCI in mice. This improvement was accompanied by reduced lesion size and demyelinated area. The local anti-inflammatory shift induced by IL-13 Mφs resulted in reduced neuronal death and fewer contacts between dystrophic axons and Mφs/microglia, suggesting suppression of axonal dieback. Using IL-4Rα-deficient mice, we show that IL-13 signaling is required for these beneficial effects. Whereas direct neuroprotective effects of IL-13 on murine and human neuroblastoma cell lines or human neurospheroid cultures were absent, IL-13 rescued murine organotypic brain slices from cell death, probably by indirectly modulating the Mφ/microglia responses. CONCLUSIONS Collectively, our data suggest that the IL-13-induced anti-inflammatory Mφ/microglia phenotype can preserve neuronal tissue and ameliorate axonal dieback, thereby promoting recovery after SCI.
Collapse
Affiliation(s)
- Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Céline Erens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Daniela Sommer
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Elle Scheijen
- Department of Neurosciences, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Selien Sanchez
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Pia M Vidal
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, 4090541, Concepción, Chile
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular & Biomedical Research University College Dublin, Belfield, Dublin 4, Ireland
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Sven Hendrix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium. .,Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany.
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| |
Collapse
|
38
|
Stewart A, Glaser E, Bailey WM, Gensel J. Immunoglobulin G is Increased in the Injured Spinal Cord in a Sex and Age Dependent Manner. J Neurotrauma 2022; 39:1090-1098. [PMID: 35373588 PMCID: PMC9347383 DOI: 10.1089/neu.2022.0011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
There are limited studies examining age and sex as biological variables in the pathophysiology of spinal cord injury (SCI). The use of older animals and sex-balanced groups in SCI models is increasingly prioritized to better match clinical demographics. Including older animals in SCI studies is technically challenging, and outcomes are unpredictable with respect to biological and treatment responses. Incidental discoveries that are unrelated to the question under investigation often emerge while including age and sex as biological variables. When probing tissue homogenates on Western blots of 4- and 14-month-old (MO) mice, we identified a sex- and age-dependent increase in immunoglobulin G (IgG) within the spinal cords of older, 14-MO mice acutely after SCI, with females having more IgG compared with males. We further probed to determine whether differences in hemorrhage exist between sexes or ages by evaluating hemoglobin within spinal homogenates. Differences in hemoglobin between sexes and ages were not consistently observed. Because IgG was elevated in an age- and sex-dependent manner without of evidence of differences in hemorrhage, our findings point to potential pre-existing differences in IgG within mouse plasma in an age- and sex-dependent manner. This report has identified age- and sex-dependent differences in infiltrating IgG into the injured spinal cord environment that may affect injury and recovery processes. Our findings highlight that systemic contributions to SCI can be sex- and age-dependent and illustrate the value of reporting incidental discoveries.
Collapse
Affiliation(s)
- Andrew Stewart
- University of Kentucky, Physiology, 741 S. Limestone Street, BBSRB B483, Lexington, Kentucky, United States, 40536-0509,
| | - Ethan Glaser
- University of Kentucky, Physiology, Lexington, Kentucky, United States,
| | - William M Bailey
- University of Kentucky, Spinal Cord and Brain Injury Research Center, Physiology, Lexington, Kentucky, United States
| | - John Gensel
- University of Kentucky, Physiology, 741 S. Limestone Street, B436 BBSRB, Lexington, Kentucky, United States, 40536-0509
| |
Collapse
|
39
|
Li Y, Ritzel RM, Lei Z, Cao T, He J, Faden AI, Wu J. Sexual dimorphism in neurological function after SCI is associated with disrupted neuroinflammation in both injured spinal cord and brain. Brain Behav Immun 2022; 101:1-22. [PMID: 34954073 PMCID: PMC8885910 DOI: 10.1016/j.bbi.2021.12.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/29/2021] [Accepted: 12/18/2021] [Indexed: 10/19/2022] Open
Abstract
Whereas human spinal cord injury (SCI) is more common in men, the prevalence is growing in women. However, little is known about the effect of biological sex on brain dysfunction and injury mechanisms. To model the highest per capita rate of injury (ages between 16 and 30 years old) in humans, in the present study, young adult or a young/middle-aged male and female C57BL/6 mice were subjected to moderate contusion SCI. When mice were injured at 10-12-week-old, transcriptomic analysis of inflammation-related genes and flow cytometry revealed a more aggressive neuroinflammatory profile in male than females following 3 d SCI, ostensibly driven by sex-specific changes myeloid cell function rather than cell number. Female mice were generally more active at baseline, as evidenced by greater distance traveled in the open field. After SCI, female mice had more favorable locomotor function than male animals. At 13 weeks post-injury, male mice showed poor performance in cognitive and depressive-like behavioral tests, while injured female mice showed fewer deficits in these tasks. However, when injured at 6 months old followed by 8 months post-injury, male mice had considerably less inflammatory activation compared with female animals despite having similar or worse outcomes in affective, cognitive, and motor tasks. Collectively, these findings indicate that sex differences in functional outcome after SCI are associated with the age at onset of injury, as well as disrupted neuroinflammation not only at the site of injury but also in remote brain regions. Thus, biological sex should be considered when designing new therapeutic agents.
Collapse
Affiliation(s)
- Yun Li
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Rodney M. Ritzel
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Zhuofan Lei
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Tuoxin Cao
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Junyun He
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA
| | - Alan I Faden
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD, 21201 USA,University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD, 21201 USA
| | - Junfang Wu
- Department of Anesthesiology and Center for Shock, Trauma and Anesthesiology Research (STAR), University of Maryland School of Medicine, Baltimore, MD 21201, USA; University of Maryland Center to Advance Chronic Pain Research, University of Maryland, Baltimore, MD 21201, USA.
| |
Collapse
|
40
|
Myelin and non-myelin debris contribute to foamy macrophage formation after spinal cord injury. Neurobiol Dis 2022; 163:105608. [PMID: 34979258 PMCID: PMC8783370 DOI: 10.1016/j.nbd.2021.105608] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 02/03/2023] Open
Abstract
Tissue damage after spinal cord injury (SCI) elicits a robust inflammatory cascade that fails to resolve in a timely manner, resulting in impaired wound healing and cellular regeneration. This inflammatory response is partly mediated by infiltrating immune cells, including macrophages. As professional phagocytes, macrophages initially play an important role in debris clearance at the injury site, which would be necessary for proper tissue regeneration. After SCI, most macrophages become filled with lipid droplets due to excessive uptake of lipid debris, assuming a "foamy" phenotype that is associated with a proinflammatory state. Myelin has been assumed to be the main source of lipid that induces foamy macrophage formation after injury given its abundance in the spinal cord. This assumption has led to the widespread use of purified myelin treatment to model foamy macrophage formation in vitro. However, the assumption that myelin is necessary for foamy macrophage formation remains untested. To this end, we developed a novel foamy macrophage assay utilizing total spinal cord homogenate to include all sources of lipid present at the injury site. Using the myelin basic protein knockout (MBP KO, i.e., Shiverer) mice that lack myelin, we investigated lipid accumulation in foamy macrophages. Primary macrophages treated with myelin-deficient spinal cord homogenate still formed large lipid droplets typically observed in foamy macrophages, although to a lesser degree than cells treated with normal homogenate. Similarly, MBP KO mice subjected to contusive spinal cord injury also formed foamy macrophages that exhibited reduced lipid content and associated with improved histological outcomes and reduced immune cell infiltration. Therefore, the absence of myelin does not preclude foamy macrophage formation, indicating that myelin is not the only major source of lipid that contributes this pathology, even though myelin may alter certain aspects of its inflammatory profile.
Collapse
|
41
|
Hook MA, Falck A, Dundumulla R, Terminel M, Cunningham R, Sefiani A, Callaway K, Gaddy D, Geoffroy CG. Osteopenia in a Mouse Model of Spinal Cord Injury: Effects of Age, Sex and Motor Function. BIOLOGY 2022; 11:biology11020189. [PMID: 35205056 PMCID: PMC8869334 DOI: 10.3390/biology11020189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/20/2022] [Accepted: 01/22/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary In the first two years following spinal cord injury, people lose up to 50% of bone below the injury. This injury-induced bone loss significantly affects rehabilitation and leaves people vulnerable to fractures and post-fracture complications, including lung and urinary tract infections, blood clots in the veins, and depression. Unfortunately, little is known about the factors driving this bone loss. In fact, even though we know that injury, age, and sex independently increase bone loss, there have been no studies looking at the cumulative effects of these variables. People with spinal injury are aging, and the age at which injuries occur is increasing. It is essential to know whether these factors together will further compromise bone. To examine this, we assessed bone loss in young and old, male and female mice after spinal injury. As expected, we found that aging alone decreased motor activity and bone volume. Spinal injury also reduced bone volume, but it did not worsen the effects of age. Instead, injury effects appeared related to reduced rearing activity. The data suggest that although partial weight-bearing does not reduce bone loss after spinal cord injury, therapies that put full weight on the legs may be clinically effective. Abstract After spinal cord injury (SCI), 80% of individuals are diagnosed with osteopenia or osteoporosis. The dramatic loss of bone after SCI increases the potential for fractures 100-fold, with post-fracture complications occurring in 54% of cases. With the age of new SCI injuries increasing, we hypothesized that a SCI-induced reduction in weight bearing could further exacerbate age-induced bone loss. To test this, young (2–3 months) and old (20–30 months) male and female mice were given a moderate spinal contusion injury (T9–T10), and recovery was assessed for 28 days (BMS, rearing counts, distance traveled). Tibial trabecular bone volume was measured after 28 days with ex vivo microCT. While BMS scores did not differ across groups, older subjects travelled less in the open field and there was a decrease in rearing with age and SCI. As expected, aging decreased trabecular bone volume and cortical thickness in both old male and female mice. SCI alone also reduced trabecular bone volume in young mice, but did not have an additional effect beyond the age-dependent decrease in trabecular and cortical bone volume seen in both sexes. Interestingly, both rearing and total activity correlated with decreased bone volume. These data underscore the importance of load and use on bone mass. While partial weight-bearing does not stabilize/reverse bone loss in humans, our data suggest that therapies that simulate complete loading may be effective after SCI.
Collapse
Affiliation(s)
- Michelle A. Hook
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
- Correspondence: ; Tel.: +1-979-436-0568
| | - Alyssa Falck
- Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; (A.F.); (D.G.)
| | - Ravali Dundumulla
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Mabel Terminel
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Rachel Cunningham
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Arthur Sefiani
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Kayla Callaway
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| | - Dana Gaddy
- Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, College Station, TX 77843, USA; (A.F.); (D.G.)
| | - Cédric G. Geoffroy
- Department of Neuroscience and Experimental Therapeutics, College of Medicine, Texas A&M Health Science Center, Bryan, TX 77807, USA; (R.D.); (M.T.); (R.C.); (A.S.); (K.C.); (C.G.G.)
| |
Collapse
|
42
|
Baine RE, Johnston DT, Strain MM, Henwood MK, Davis JA, Reynolds JA, Giles ED, Grau JW. Noxious Stimulation Induces Acute Hemorrhage and Impairs Long-Term Recovery after Spinal Cord Injury (SCI) in Female Rats: Evidence Estrous Cycle May Have a Modulatory Effect. Neurotrauma Rep 2022; 3:70-86. [PMID: 35112109 PMCID: PMC8804264 DOI: 10.1089/neur.2021.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Spinal cord injuries (SCIs) are often the result of traumatic accidents, which also produce multiple other injuries (polytrauma). Nociceptive input from associated injuries has been shown to significantly impair recovery post-SCI. Historically, work in our laboratory has focused exclusively on male animals; however, increasing incidence of SCI in females requires research to determine whether pain (nociceptive) input poses the same risk to their recovery. Some animal studies have shown that females demonstrate greater tissue preservation and better locomotor recovery post-SCI. Given this, we examined the effect of sex on SCI recovery in two pain models—intermittent electrical stimulation (shock) to the tail or capsaicin injection to the hindpaw. Female rats received a lower thoracic contusion injury and were exposed to noxious stimulation the next day. The acute effect of noxious input on cardiovascular function, locomotor performance, and hemorrhage were assessed. Treatment with capsaicin or noxious electrical stimulation disrupted locomotor performance, increased blood pressure, and disrupted stepping. Additional experiments examined the long-term consequences of noxious input, demonstrating that both noxious electrical stimulation and capsaicin impair long-term recovery in female rats. Interestingly, injury had a greater effect on behavioral performance when progesterone and estrogen were low (metestrus). Conversely, nociceptive input led to a greater disruption in locomotor performance and produced a greater rise in blood pressure in animals injured during estrus.
Collapse
Affiliation(s)
- Rachel E. Baine
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - David T. Johnston
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - Misty M. Strain
- Department of Cellular and Integrative Physiology, University of Texas Health Science, San Antonio, Texas, USA
| | - Melissa K. Henwood
- Department of Neuroscience, Cell Biology, Anatomy, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jacob A. Davis
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - Joshua A. Reynolds
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| | - Erin D. Giles
- Department of Nutrition, Texas A&M University, College Station, Texas, USA
| | - James W. Grau
- Department of Psychological and Brain Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
43
|
Liu W, Yu J, Wang YF, Shan QQ, Wang YX. Selection of suitable internal controls for gene expression normalization in rats with spinal cord injury. Neural Regen Res 2021; 17:1387-1392. [PMID: 34782586 PMCID: PMC8643046 DOI: 10.4103/1673-5374.327350] [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: 11/04/2022] Open
Abstract
There is a lack of systematic research on the expression of internal control genes used for gene expression normalization in real-time reverse transcription polymerase chain reaction in spinal cord injury research. In this study, we used rat models of spinal cord hemisection to analyze the expression stability of 13 commonly applied reference genes: Actb, Ankrd27, CypA, Gapdh, Hprt1, Mrpl10, Pgk1, Rictor, Rn18s, Tbp, Ubc, Ubxn11, and Ywhaz. Our results show that the expression of Ankrd27, Ubc, and Tbp were stable after spinal cord injury, while Actb was the most unstable internal control gene. Ankrd27, Ubc, Tbp, and Actb were consequently used to investigate the effects of internal control genes with differing stabilities on the normalization of target gene expression. Target gene expression levels and changes over time were similar when Ankrd27, Ubc, and Tbp were used as internal controls but different when Actb was used as an internal control. We recommend that Ankrd27, Ubc, and Tbp are used as internal control genes for real-time reverse transcription polymerase chain reaction in spinal cord injury research. This study was approved by the Administration Committee of Experimental Animals, Jiangsu Province, China (approval No. 20180304-008) on March 4, 2018.
Collapse
Affiliation(s)
- Wei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| | - Jie Yu
- Department of Nursing, The Affiliated Hospital of Nantong University; Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Yi-Fan Wang
- Department of Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu Province, China
| | - Qian-Qian Shan
- Department of Radiotherapy and Oncology, The Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Ya-Xian Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, National Medical Products Administration (NMPA) Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
44
|
Wang X, Li X, Zuo X, Liang Z, Ding T, Li K, Ma Y, Li P, Zhu Z, Ju C, Zhang Z, Song Z, Quan H, Zhang J, Hu X, Wang Z. Photobiomodulation inhibits the activation of neurotoxic microglia and astrocytes by inhibiting Lcn2/JAK2-STAT3 crosstalk after spinal cord injury in male rats. J Neuroinflammation 2021; 18:256. [PMID: 34740378 PMCID: PMC8571847 DOI: 10.1186/s12974-021-02312-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/29/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neurotoxic microglia and astrocytes begin to activate and participate in pathological processes after spinal cord injury (SCI), subsequently causing severe secondary damage and affecting tissue repair. We have previously reported that photobiomodulation (PBM) can promote functional recovery by reducing neuroinflammation after SCI, but little is known about the underlying mechanism. Therefore, we aimed to investigate whether PBM ameliorates neuroinflammation by modulating the activation of microglia and astrocytes after SCI. METHODS Male Sprague-Dawley rats were randomly divided into three groups: a sham control group, an SCI + vehicle group and an SCI + PBM group. PBM was performed for two consecutive weeks after clip-compression SCI models were established. The activation of neurotoxic microglia and astrocytes, the level of tissue apoptosis, the number of motor neurons and the recovery of motor function were evaluated at different days post-injury (1, 3, 7, 14, and 28 days post-injury, dpi). Lipocalin 2 (Lcn2) and Janus kinase-2 (JAK2)-signal transducer and activator of transcription-3 (STAT3) signaling were regarded as potential targets by which PBM affected neurotoxic microglia and astrocytes. In in vitro experiments, primary microglia and astrocytes were irradiated with PBM and cotreated with cucurbitacin I (a JAK2-STAT3 pathway inhibitor), an adenovirus (shRNA-Lcn2) and recombinant Lcn2 protein. RESULTS PBM promoted the recovery of motor function, inhibited the activation of neurotoxic microglia and astrocytes, alleviated neuroinflammation and tissue apoptosis, and increased the number of neurons retained after SCI. The upregulation of Lcn2 and the activation of the JAK2-STAT3 pathway after SCI were suppressed by PBM. In vitro experiments also showed that Lcn2 and JAK2-STAT3 were mutually promoted and that PBM interfered with this interaction, inhibiting the activation of microglia and astrocytes. CONCLUSION Lcn2/JAK2-STAT3 crosstalk is involved in the activation of neurotoxic microglia and astrocytes after SCI, and this process can be suppressed by PBM.
Collapse
Affiliation(s)
- Xuankang Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xin Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.,967 Hospital of People's Liberation Army Joint Logistic Support Force, Dalian, 116044, Liaoning, China
| | - Xiaoshuang Zuo
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhuowen Liang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Tan Ding
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Kun Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Yangguang Ma
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Penghui Li
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhijie Zhu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Cheng Ju
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Zhihao Zhang
- 967 Hospital of People's Liberation Army Joint Logistic Support Force, Dalian, 116044, Liaoning, China
| | - Zhiwen Song
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Huilin Quan
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jiawei Zhang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Xueyu Hu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Zhe Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
45
|
Oughourlian TC, Wang C, Salamon N, Holly LT, Ellingson BM. Sex-Dependent Cortical Volume Changes in Patients with Degenerative Cervical Myelopathy. J Clin Med 2021; 10:jcm10173965. [PMID: 34501413 PMCID: PMC8432178 DOI: 10.3390/jcm10173965] [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: 07/25/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 11/24/2022] Open
Abstract
Degenerative cervical myelopathy (DCM) is a progressive condition characterized by degeneration of osseocartilaginous structures within the cervical spine resulting in compression of the spinal cord and presentation of clinical symptoms. Compared to healthy controls (HCs), studies have shown DCM patients experience structural and functional reorganization in the brain; however, sex-dependent cortical differences in DCM patients remains largely unexplored. In the present study, we investigate the role of sex differences on the structure of the cerebral cortex in DCM and determine how structural differences may relate to clinical measures of neurological function. T1-weighted structural MRI scans were acquired in 85 symptomatic and asymptomatic patients with DCM and 90 age-matched HCs. Modified Japanese Orthopedic Association (mJOA) scores were obtained for patients. A general linear model was used to determine vertex-level significant differences in gray matter volume (GMV) between the following groups (1) male HCs and female HCs, (2) male patients and female patients, (3) male patients and male HCs, and (4) female patients and female HCs. Within patients, males exhibited larger GMV in motor, language, and vision related brain regions compared to female DCM patients. Males demonstrated a significant positive correlation between GMV and mJOA score, in which patients with worsening neurological symptoms exhibited decreasing GMV primarily across somatosensory and motor related cortical regions. Females exhibited a similar association, albeit across a broader range of cortical areas including those involved in pain processing. In sensorimotor regions, female patients consistently showed smaller GMV compared with male patients, independent of mJOA score. Results from the current study suggest strong sex-related differences in cortical volume in patients with DCM, which may reflect hormonal influence or differing compensation mechanisms.
Collapse
Affiliation(s)
- Talia C. Oughourlian
- UCLA Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (T.C.O.); (C.W.); (B.M.E.)
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Neuroscience Interdepartmental Graduate Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Chencai Wang
- UCLA Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (T.C.O.); (C.W.); (B.M.E.)
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Noriko Salamon
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Langston T. Holly
- Department of Neurosurgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +1-(310)-319-3475
| | - Benjamin M. Ellingson
- UCLA Center for Computer Vision and Imaging Biomarkers, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90024, USA; (T.C.O.); (C.W.); (B.M.E.)
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Neuroscience Interdepartmental Graduate Program, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
46
|
Moulson AJ, Squair JW, Franklin RJM, Tetzlaff W, Assinck P. Diversity of Reactive Astrogliosis in CNS Pathology: Heterogeneity or Plasticity? Front Cell Neurosci 2021; 15:703810. [PMID: 34381334 PMCID: PMC8349991 DOI: 10.3389/fncel.2021.703810] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
Astrocytes are essential for the development and homeostatic maintenance of the central nervous system (CNS). They are also critical players in the CNS injury response during which they undergo a process referred to as "reactive astrogliosis." Diversity in astrocyte morphology and gene expression, as revealed by transcriptional analysis, is well-recognized and has been reported in several CNS pathologies, including ischemic stroke, CNS demyelination, and traumatic injury. This diversity appears unique to the specific pathology, with significant variance across temporal, topographical, age, and sex-specific variables. Despite this, there is limited functional data corroborating this diversity. Furthermore, as reactive astrocytes display significant environmental-dependent plasticity and fate-mapping data on astrocyte subsets in the adult CNS is limited, it remains unclear whether this diversity represents heterogeneity or plasticity. As astrocytes are important for neuronal survival and CNS function post-injury, establishing to what extent this diversity reflects distinct established heterogeneous astrocyte subpopulations vs. environmentally dependent plasticity within established astrocyte subsets will be critical for guiding therapeutic development. To that end, we review the current state of knowledge on astrocyte diversity in the context of three representative CNS pathologies: ischemic stroke, demyelination, and traumatic injury, with the goal of identifying key limitations in our current knowledge and suggesting future areas of research needed to address them. We suggest that the majority of identified astrocyte diversity in CNS pathologies to date represents plasticity in response to dynamically changing post-injury environments as opposed to heterogeneity, an important consideration for the understanding of disease pathogenesis and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Aaron J. Moulson
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
| | - Jordan W. Squair
- Department of Clinical Neuroscience, Faculty of Life Sciences, Center for Neuroprosthetics and Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), NeuroRestore, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Robin J. M. Franklin
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Peggy Assinck
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
47
|
Zinger A, Soriano S, Baudo G, De Rosa E, Taraballi F, Villapol S. Biomimetic Nanoparticles as a Theranostic Tool for Traumatic Brain Injury. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2100722. [PMID: 34413716 PMCID: PMC8356641 DOI: 10.1002/adfm.202100722] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 03/08/2021] [Indexed: 05/04/2023]
Abstract
Traumatic brain injury (TBI) triggers both central and peripheral inflammatory responses. Existing pharmacological drugs are unable to effectively and quickly target the brain inflamed regions, setting up a major roadblock towards effective brain trauma treatments. Nanoparticles (NPs) have been used in multiple diseases as drug delivery tools with remarkable success due to their rapid diffusion and specificity in the target organ. Here, leukocyte-based biomimetic NPs are fabricated as a theranostic tool to directly access inflamed regions in a TBI mouse model. This NP systemic delivery is visualized using advanced in vivo imaging techniques, including intravital microscopy and in vivo imaging system. The results demonstrate selective targeting of NPs to the injured brain and increased NPs accumulation among the peripheral organs 24 h after TBI. Interestingly, increased microglial proliferation, decreased macrophage infiltration, and reduced brain lesion following the NPs treatments compared to sham vehicle-treated mice are also found. In summary, the results suggest that NPs represent a promising future theranostic tool for TBI treatment.
Collapse
Affiliation(s)
- Assaf Zinger
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Sirena Soriano
- Center for Neuroregeneration and Department of NeurosurgeryHouston Methodist Research InstituteHoustonTX77030USA
| | - Gherardo Baudo
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Enrica De Rosa
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Francesca Taraballi
- Center for Musculoskeletal RegenerationHouston Methodist Academic InstituteDepartment of Orthopedics and Sports MedicineHouston Methodist HospitalHoustonTX77030USA
| | - Sonia Villapol
- Center for Neuroregeneration and Department of NeurosurgeryHouston Methodist Research InstituteHoustonTX77030USA
- Weill Cornell Medical CollegeNew YorkNY10065USA
| |
Collapse
|
48
|
Stewart AN, Lowe JL, Glaser EP, Mott CA, Shahidehpour RK, McFarlane KE, Bailey WM, Zhang B, Gensel JC. Acute inflammatory profiles differ with sex and age after spinal cord injury. J Neuroinflammation 2021; 18:113. [PMID: 33985529 PMCID: PMC8120918 DOI: 10.1186/s12974-021-02161-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/29/2021] [Indexed: 01/05/2023] Open
Abstract
Background Sex and age are emerging as influential variables that affect spinal cord injury (SCI) recovery. Despite a changing demographic towards older age at the time of SCI, the effects of sex or age on inflammation remain to be elucidated. This study determined the sex- and age-dependency of the innate immune response acutely after SCI. Methods Male and female mice of ages 4- and 14-month-old received T9 contusion SCI and the proportion of microglia, monocyte-derived macrophages (MDM), and neutrophils surrounding the lesion were determined at 3- and 7-day post-injury (DPI) using flow cytometry. Cell counts of microglia and MDMs were obtained using immunohistochemistry to verify flow cytometry results at 3-DPI. Microglia and MDMs were separately isolated using fluorescence-activated cell sorting (FACS) at 3-day post-injury (DPI) to assess RNA expression of 27 genes associated with activation, redox, and debris metabolism/clearance. Results Flow cytometry revealed that being female and older at the time of injury significantly increased MDMs relative to other phagocytes, specifically increasing the ratio of MDMs to microglia at 3-DPI. Cell counts using immunohistochemistry revealed that male mice have more total microglia within SCI lesions that can account for a lower MDM/microglia ratio. With NanoString analyses of 27 genes, only 1 was differentially expressed between sexes in MDMs; specifically, complement protein C1qa was increased in males. No genes were affected by age in MDMs. Only 2 genes were differentially regulated in microglia between sexes after controlling for false discovery rate, specifically CYBB (NOX2) as a reactive oxygen species (ROS)-associated marker as well as MRC1 (CD206), a gene associated with reparative phenotypes. Both genes were increased in female microglia. No microglial genes were differentially regulated between ages. Differences between microglia and MDMs were found in 26 of 27 genes analyzed, all expressed higher in MDMs with three exceptions. Specifically, C1qa, cPLA2, and CD86 were expressed higher in microglia. Conclusions These findings indicate that inflammatory responses to SCI are sex-dependent at both the level of cellular recruitment and gene expression. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02161-8.
Collapse
Affiliation(s)
- Andrew N Stewart
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - John L Lowe
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.,Science Honors Program of Georgetown College, Georgetown, KY, 40324, USA
| | - Ethan P Glaser
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Caitlin A Mott
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Ryan K Shahidehpour
- Department of Neuroscience, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Katelyn E McFarlane
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - William M Bailey
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA
| | - Bei Zhang
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.,Present address: Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA
| | - John C Gensel
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky College of Medicine, Lexington, KY, 40536, USA.
| |
Collapse
|
49
|
Jacobson PB, Goody R, Lawrence M, Mueller BK, Zhang X, Hooker BA, Pfleeger K, Ziemann A, Locke C, Barraud Q, Droescher M, Bernhard J, Popp A, Boeser P, Huang L, Mollon J, Mordashova Y, Cui YF, Savaryn JP, Grinnell C, Dreher I, Gold M, Courtine G, Mothe A, Tator CH, Guest JD. Elezanumab, a human anti-RGMa monoclonal antibody, promotes neuroprotection, neuroplasticity, and neurorecovery following a thoracic hemicompression spinal cord injury in non-human primates. Neurobiol Dis 2021; 155:105385. [PMID: 33991647 DOI: 10.1016/j.nbd.2021.105385] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/10/2021] [Accepted: 04/30/2021] [Indexed: 01/21/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition characterized by loss of function, secondary to damaged spinal neurons, disrupted axonal connections, and myelin loss. Spontaneous recovery is limited, and there are no approved pharmaceutical treatments to reduce ongoing damage or promote repair. Repulsive guidance molecule A (RGMa) is upregulated following injury to the central nervous system (CNS), where it is believed to induce neuronal apoptosis and inhibit axonal growth and remyelination. We evaluated elezanumab, a human anti-RGMa monoclonal antibody, in a novel, newly characterized non-human primate (NHP) hemicompression model of thoracic SCI. Systemic intravenous (IV) administration of elezanumab over 6 months was well tolerated and associated with significant improvements in locomotor function. Treatment of animals for 16 weeks with a continuous intrathecal infusion of elezanumab below the lesion was not efficacious. IV elezanumab improved microstructural integrity of extralesional tissue as reflected by higher fractional anisotropy and magnetization transfer ratios in treated vs. untreated animals. IV elezanumab also reduced SCI-induced increases in soluble RGMa in cerebrospinal fluid, and membrane bound RGMa rostral and caudal to the lesion. Anterograde tracing of the corticospinal tract (CST) from the contralesional motor cortex following 20 weeks of IV elezanumab revealed a significant increase in the density of CST fibers emerging from the ipsilesional CST into the medial/ventral gray matter. There was a significant sprouting of serotonergic (5-HT) fibers rostral to the injury and in the ventral horn of lower thoracic regions. These data demonstrate that 6 months of intermittent IV administration of elezanumab, beginning within 24 h after a thoracic SCI, promotes neuroprotection and neuroplasticity of key descending pathways involved in locomotion. These findings emphasize the mechanisms leading to improved recovery of neuromotor functions with elezanumab in acute SCI in NHPs.
Collapse
Affiliation(s)
- Peer B Jacobson
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America.
| | - Robin Goody
- Virscio, New Haven, CT, United States of America
| | | | - Bernhard K Mueller
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Xiaomeng Zhang
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Bradley A Hooker
- Department of Translational Sciences, Imaging Research, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Kimberly Pfleeger
- Department of Neuroscience Development, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Adam Ziemann
- Department of Neuroscience Development, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Charles Locke
- Department of Biometrics, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Quentin Barraud
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland; Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland; Defitech Center for Interventional Neurotherapies, (NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Mathias Droescher
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Joerg Bernhard
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Andreas Popp
- Department of Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Preethne Boeser
- Department of Preclinical Safety, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Lili Huang
- AbbVie Biologics, AbbVie Bioresearch Center, 381 Plantation St., Worcester, MA 01605, United States of America
| | - Jennifer Mollon
- Data and Statistical Sciences, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Yulia Mordashova
- Data and Statistical Sciences, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Yi-Fang Cui
- Discovery Biology, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - John P Savaryn
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Christine Grinnell
- Department of Drug Metabolism and Pharmacokinetics, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Ingeborg Dreher
- Department of Bioanalytics, AbbVie Deutschland GmbH & Co. KG, Neuroscience Research, Knollstrasse, 67061 Ludwigshafen, Germany
| | - Michael Gold
- Department of Neuroscience Development, AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064, United States of America
| | - Grégoire Courtine
- Center for Neuroprosthetics and Brain Mind Institute, School of Life Sciences, Swiss Federal Institute of Technology (EPFL), Geneva, Switzerland; Department of Clinical Neuroscience, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland; Defitech Center for Interventional Neurotherapies, (NeuroRestore), CHUV/UNIL/EPFL, Lausanne, Switzerland
| | - Andrea Mothe
- Krembil Brain Institute, Toronto Western Hospital, University Health Network, Toronto, Canada
| | - Charles H Tator
- Division of Neurosurgery, Toronto Western Hospital, and University of Toronto, Toronto, Canada
| | - James D Guest
- Department of Neurosurgery and The Miami Project to Cure Paralysis, The Miller School of Medicine, University of Miami, Miami, FL, United States of America
| |
Collapse
|
50
|
Wei GZ, Saraswat Ohri S, Khattar NK, Listerman AW, Doyle CH, Andres KR, Karuppagounder SS, Ratan RR, Whittemore SR, Hetman M. Hypoxia-inducible factor prolyl hydroxylase domain (PHD) inhibition after contusive spinal cord injury does not improve locomotor recovery. PLoS One 2021; 16:e0249591. [PMID: 33819286 PMCID: PMC8021188 DOI: 10.1371/journal.pone.0249591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating neurological condition that involves both primary and secondary tissue loss. Various cytotoxic events including hypoxia, hemorrhage and blood lysis, bioenergetic failure, oxidative stress, endoplasmic reticulum (ER) stress, and neuroinflammation contribute to secondary injury. The HIF prolyl hydroxylase domain (PHD/EGLN) family of proteins are iron-dependent, oxygen-sensing enzymes that regulate the stability of hypoxia inducible factor-1α (HIF-1α) and also mediate oxidative stress caused by free iron liberated from the lysis of blood. PHD inhibition improves outcome after experimental intracerebral hemorrhage (ICH) by reducing activating transcription factor 4 (ATF4)-driven neuronal death. As the ATF4-CHOP (CCAAT-enhancer-binding protein homologous protein) pathway plays a role in the pathogenesis of contusive SCI, we examined the effects of PHD inhibition in a mouse model of moderate T9 contusive SCI in which white matter damage is the primary driver of locomotor dysfunction. Pharmacological inhibition of PHDs using adaptaquin (AQ) moderately lowers acute induction of Atf4 and Chop mRNAs and prevents the acute decline of oligodendrocyte (OL) lineage mRNAs, but does not improve long-term recovery of hindlimb locomotion or increase chronic white matter sparing. Conditional genetic ablation of all three PHD isoenzymes in OLs did not affect Atf4, Chop or OL mRNAs expression levels, locomotor recovery, and white matter sparing after SCI. Hence, PHDs may not be suitable targets to improve outcomes in traumatic CNS pathologies that involve acute white matter injury.
Collapse
Affiliation(s)
- George Z Wei
- University of Louisville School of Medicine, Louisville, Kentucky, United States of America.,Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Nicolas K Khattar
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Adam W Listerman
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Catherine H Doyle
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Saravanan S Karuppagounder
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Neurological Institute, White Plains, NY, United States of America.,Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY, United States of America
| | - Rajiv R Ratan
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Neurological Institute, White Plains, NY, United States of America.,Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY, United States of America
| | - Scott R Whittemore
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Michal Hetman
- University of Louisville School of Medicine, Louisville, Kentucky, United States of America.,Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States of America
| |
Collapse
|