1
|
Ma Y, Tang Q, Cheng X, Athertya JS, Coughlin D, Chang EY, Johnson CE, Cui J, Gu Z, Du J. UTE MRI for assessing demyelination in an mTBI mouse model: An open-field low-intensity blast study. Neuroimage 2025; 310:121103. [PMID: 40024556 DOI: 10.1016/j.neuroimage.2025.121103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/28/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025] Open
Abstract
Mild traumatic brain injury (mTBI) is a leading cause of long-term disability. Following mTBI, secondary chemical cascades and neuroinflammation can result in myelin damage, significantly impairing cognitive function. This study aims to assess demyelination in mice with mTBI induced by open-field low-intensity blast (LIB) using a novel three-dimensional short repetition time adiabatic inversion recovery UTE (3D STAIR-UTE) magnetic resonance imaging (MRI) sequence. Thirty male C57BL/6 mice, with 15 experiencing mTBI and 15 serving as sham controls, were included in this study. Behavioral tests were performed starting at 5 days post-injury to assess motor activity and anxiety-like responses followed by STAIR-UTE imaging using a pre-clinical 3T MRI scanner. Additionally, a proton density-weighted UTE sequence was scanned alongside the STAIR-UTE for quantification of myelin proton fraction (MPF). Luxol fast blue (LFB) staining was performed to evaluate myelin changes between the mTBI group and the control group. The behavioral tests indicated decreased motor activity in the center zone and increased anxiety-like response in the mTBI mice compared to sham controls. The STAIR-UTE sequence revealed significantly lower MPFs in the corpus callosum of mTBI mice (8.4 ± 0.4 % vs. 8.7 ± 0.4 %; P = 0.003), consistent with the myelin reduction observed in the LFB staining (0.77 ± 0.22 vs. 1.09 ± 0.15; P = 0.004). Our findings demonstrate that the STAIR-UTE sequence facilitates quantitative myelin imaging at 3T MRI, enabling the detection of demyelination in the white matter of the mouse brain associated with alterations in motor and anxiety domains post-LIB exposure.
Collapse
Affiliation(s)
- Yajun Ma
- Department of Radiology, University of California San Diego, CA, USA.
| | - Qingbo Tang
- Department of Radiology, University of California San Diego, CA, USA; Radiology Service, VA San Diego Healthcare System, CA, USA
| | - Xin Cheng
- Department of Radiology, University of California San Diego, CA, USA; Radiology Service, VA San Diego Healthcare System, CA, USA
| | - Jiyo S Athertya
- Department of Radiology, University of California San Diego, CA, USA
| | - David Coughlin
- Department of Neurosciences, University of California San Diego, CA, USA
| | - Eric Y Chang
- Department of Radiology, University of California San Diego, CA, USA; Radiology Service, VA San Diego Healthcare System, CA, USA
| | - Catherine E Johnson
- Department of Explosive Engineering, Missouri University of Science and Technology, MO, USA
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri, MO, USA; Research Division, Harry S Truman Memorial Hospital, Columbia, MO, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri, MO, USA; Research Division, Harry S Truman Memorial Hospital, Columbia, MO, USA
| | - Jiang Du
- Department of Radiology, University of California San Diego, CA, USA; Radiology Service, VA San Diego Healthcare System, CA, USA; Department of Bioengineering, University of California San Diego, CA, USA.
| |
Collapse
|
2
|
Athertya JS, Cheng X, Tang Q, Moazamian D, Chang EY, Johnson CE, Cui J, Gu Z, Ma Y, Du J. Myelin Quantification Using Ultrashort Echo Time Magnetization Transfer Ratio in a Mouse Model of Traumatic Brain Injury. J Neuroimaging 2025; 35:e70029. [PMID: 39994838 DOI: 10.1111/jon.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 02/04/2025] [Accepted: 02/15/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND AND PURPOSE This study aims to assess the potential of ultrashort echo time imaging-based magnetization transfer ratio (UTE-MTR) in detecting demyelination in mice with mild traumatic brain injury (mTBI) caused by an open-field low-intensity blast (LIB) injury model. METHODS This study included 30 male C57BL/6 mice, approximately 8 weeks old, sourced from Jackson Laboratories in Bar Harbor, ME, and conducted under institutional guidelines. The mice were divided into the mTBI group (n = 15) and the sham control group (n = 15). All animal experiments followed the approved protocols for the Care and Use of Laboratory Animals and Animal Research. The mTBI group underwent the open-field LIB injury. Behavioral tests were conducted to assess motor activity and anxiety-like responses. UTE-MT imaging was performed using a 3 Tesla Bruker system to measure UTE-MTR from two UTE-MT datasets with saturation powers of 1500° and 500°, and two frequency offsets of 2 and 50 kHz, respectively. Luxol fast blue (LFB) staining was performed to evaluate myelin content. The mean UTE-MTR values for regions of interest centered at the medial section of the corpus callosum were computed. The behavioral tests, LFB myelin staining, and UTE-MTR values were compared between the two groups using the independent t-test. p values <0.05 were considered significant. RESULTS The mTBI mice demonstrated decreased motor activity and increased anxiety-like response over sham controls. The mTBI mice also showed significantly lower UTE-MTR values (0.399±0.007 vs. 0.393±0.005; p<0.05) and reduced LFB myelin staining (0.848±0.324 vs. 1.145±0.260; p = 0.048) over sham controls. CONCLUSION The significantly lower UTE-MTR values in the corpus callosum of mTBI mice are consistent with reduced LFB myelin staining, indicating that UTE-MTR can detect myelin loss and associated alterations in motor and anxiety domains post-LIB exposure.
Collapse
Affiliation(s)
- Jiyo S Athertya
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Xin Cheng
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Qingbo Tang
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Dina Moazamian
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Eric Y Chang
- Department of Radiology, University of California San Diego, La Jolla, California, USA
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
| | - Catherine E Johnson
- Department of Explosive Engineering, Missouri University of Science and Technology, Rolla, Missouri, USA
| | - Jiankun Cui
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, USA
- Research Division, Harry S Truman Memorial Hospital, Columbia, Missouri, USA
| | - Zezong Gu
- Department of Pathology and Anatomical Sciences, University of Missouri, Columbia, Missouri, USA
- Research Division, Harry S Truman Memorial Hospital, Columbia, Missouri, USA
| | - Yajun Ma
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Jiang Du
- Department of Radiology, University of California San Diego, La Jolla, California, USA
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
3
|
Mhaske A, Shukla S, Ahirwar K, Singh KK, Shukla R. Receptor-Assisted Nanotherapeutics for Overcoming the Blood-Brain Barrier. Mol Neurobiol 2024; 61:8702-8738. [PMID: 38558360 PMCID: PMC11496374 DOI: 10.1007/s12035-024-04015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/03/2024] [Indexed: 04/04/2024]
Abstract
Blood-brain barrier (BBB) is a distinguishing checkpoint that segregates peripheral organs from neural compartment. It protects the central nervous system from harmful ambush of antigens and pathogens. Owing to such explicit selectivity, the BBB hinders passage of various neuroprotective drug molecules that escalates into poor attainability of neuroprotective agents towards the brain. However, few molecules can surpass the BBB and gain access in the brain parenchyma by exploiting surface transporters and receptors. For successful development of brain-targeted therapy, understanding of BBB transporters and receptors is crucial. This review focuses on the transporter and receptor-based mechanistic pathway that can be manoeuvred for better comprehension of reciprocity of receptors and nanotechnological vehicle delivery. Nanotechnology has emerged as one of the expedient noninvasive approaches for brain targeting via manipulating the hurdle of the BBB. Various nanovehicles are being reported for brain-targeted delivery such as nanoparticles, nanocrystals, nanoemulsion, nanolipid carriers, liposomes and other nanovesicles. Nanotechnology-aided brain targeting can be a strategic approach to circumvent the BBB without altering the inherent nature of the BBB.
Collapse
Affiliation(s)
- Akshada Mhaske
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Shalini Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Kailash Ahirwar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India
| | - Kamalinder K Singh
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, PR1 2HE, UK.
- Biomedical Evidence-based Transdisciplinary Health Research Institute, University of Central Lancashire, Preston, PR1 2HE, UK.
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Bijnor-Sisendi Road, Sarojini Nagar, Lucknow, Uttar Pradesh, 226002, India.
| |
Collapse
|
4
|
Tachino J, Togami Y, Matsumoto H, Matsubara T, Seno S, Ogura H, Oda J. Plasma proteomics profile-based comparison of torso versus brain injury: A prospective cohort study. J Trauma Acute Care Surg 2024; 97:557-565. [PMID: 38595266 PMCID: PMC11446512 DOI: 10.1097/ta.0000000000004356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/11/2024]
Abstract
BACKGROUND Trauma-related deaths and posttraumatic sequelae are a global health concern, necessitating a deeper understanding of the pathophysiology to advance trauma therapy. Proteomics offers insights into identifying and analyzing plasma proteins associated with trauma and inflammatory conditions; however, current proteomic methods have limitations in accurately measuring low-abundance plasma proteins. This study compared plasma proteomics profiles of patients from different acute trauma subgroups to identify new therapeutic targets and devise better strategies for personalized medicine. METHODS This prospective observational single-center cohort study was conducted between August 2020 and September 2021 in the intensive care unit of Osaka University Hospital in Japan. Enrolling 59 consecutive patients with blunt trauma, we meticulously analyzed plasma proteomics profiles in participants with torso or head trauma, comparing them with those of controls (mild trauma). Using the Olink Explore 3072 instrument (Olink Proteomics AB, Uppsala, Sweden), we identified five endotypes (α-ε) via unsupervised hierarchical clustering. RESULTS The median time from injury to blood collection was 47 minutes [interquartile range, 36-64 minutes]. The torso trauma subgroup exhibited 26 unique proteins with significantly altered expression, while the head trauma subgroup showed 68 unique proteins with no overlap between the two. The identified endotypes included α (torso trauma, n = 8), β (young patients with brain injury, n = 5), γ (severe brain injury postsurgery, n = 8), δ (torso or brain trauma with mild hyperfibrinolysis, n = 18), and ε (minor trauma, n = 20). Patients with torso trauma showed changes in blood pressure, smooth muscle adaptation, hypermetabolism, and hypoxemia. Patients with traumatic brain injury had dysregulated blood coagulation and altered nerves regeneration and differentiation. CONCLUSION This study identified unique plasma protein expression patterns in patients with torso trauma and traumatic brain injury, helping categorize five distinct endotypes. Our findings may offer new insights for clinicians, highlighting potential strategies for personalized medicine and improved trauma-related care. LEVEL OF EVIDENCE Prognostic and Epidemiological; Level III.
Collapse
|
5
|
Clay AM, Carr RL, DuBien JL, To F. Short-term behavioral and histological findings following a single concussive and repeated subconcussive brain injury in a rodent model. Brain Inj 2024; 38:827-834. [PMID: 38704844 DOI: 10.1080/02699052.2024.2349144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 04/23/2024] [Indexed: 05/07/2024]
Abstract
PRIMARY OBJECTIVE It is unclear of the correlation between a mild traumatic brain injury (mTBI) and repeated subconcussive (RSC) impacts with respect to injury biomechanics. Thus, the present study was designed to determine the behavioral and histological differences between a single mTBI impact and RSC impacts with subdivided cumulative kinetic energies of the single mTBI impact. RESEARCH DESIGN Adult male Sprague-Dawley rats were randomly assigned to a single mTBI impact, RSC impact, sham, or repeated sham groups. METHODS AND PROCEDURES Following a weight drop injury, anxiety-like behavior and general locomotive activity and were assessed using the open field test, while motor coordination was evaluated using a rotarod unit. Neuronal loss, astrogliosis, and microgliosis were assessed using NeuN, GFAP and Iba-1 immunohistochemistry. All assessments were undertaken at 3- and 7-days post impact. MAIN OUTCOMES AND RESULTS No behavioral disturbances were observed in injury groups, however, both injury groups did lead to microgliosis following 3-days post-impact. CONCLUSIONS No pathophysiological differences were observed between a single mTBI impact and RSC impacts of the same energy input. Even though a cumulative injury threshold for RSC impacts was not determined, a threshold still may exist where no pathodynamic shift occurs.
Collapse
Affiliation(s)
- Anna Marie Clay
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi, USA
| | - Russell L Carr
- Center for Environmental Health Sciences, College of Veterinary Medicine, Mississippi University, Mississippi, USA
| | - Janice L DuBien
- Department of Statistics, Mississippi University, Mississippi, USA
| | - Filip To
- Department of Agricultural and Biological Engineering, Mississippi State University, Mississippi, USA
| |
Collapse
|
6
|
Poddar J, Rangasamy SB, Pahan K. Therapeutic efficacy of cinnamein, a component of balsam of Tolu/Peru, in controlled cortical impact mouse model of TBI. Neurochem Int 2024; 176:105742. [PMID: 38641028 DOI: 10.1016/j.neuint.2024.105742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 04/21/2024]
Abstract
Traumatic brain injury (TBI) remains a major health concern which causes long-term neurological disability particularly in war veterans, athletes and young adults. In spite of intense clinical and research investigations, there is no effective therapy to cease the pathogenesis of the disease. It is believed that axonal injury during TBI is potentiated by neuroinflammation and demyelination and/or failure to remyelination. This study highlights the use of naturally available cinnamein, also chemically known as benzyl cinnamate, in inhibiting neuroinflammation, promoting remyelination and combating the disease process of controlled cortical impact (CCI)-induced TBI in mice. Oral delivery of cinnamein through gavage brought down the activation of microglia and astrocytes to decrease the expression of inducible nitric oxide synthase (iNOS), glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1 (Iba1) in hippocampus and cortex of TBI mice. Cinnamein treatment also stimulated remyelination in TBI mice as revealed by PLP and A2B5 double-labeling, luxol fast blue (LFB) staining and axonal double-labeling for neurofilament and MBP. Furthermore, oral cinnamein reduced the size of lesion cavity in the brain, improved locomotor functions and restored memory and learning in TBI mice. These results suggest a new neuroprotective property of cinnamein that may be valuable in the treatment of TBI.
Collapse
Affiliation(s)
- Jit Poddar
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Suresh B Rangasamy
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Kalipada Pahan
- Division of Research and Development, Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60612, USA; Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, 60612, USA.
| |
Collapse
|
7
|
Amlerova Z, Chmelova M, Anderova M, Vargova L. Reactive gliosis in traumatic brain injury: a comprehensive review. Front Cell Neurosci 2024; 18:1335849. [PMID: 38481632 PMCID: PMC10933082 DOI: 10.3389/fncel.2024.1335849] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/01/2024] [Indexed: 01/03/2025] Open
Abstract
Traumatic brain injury (TBI) is one of the most common pathological conditions impacting the central nervous system (CNS). A neurological deficit associated with TBI results from a complex of pathogenetic mechanisms including glutamate excitotoxicity, inflammation, demyelination, programmed cell death, or the development of edema. The critical components contributing to CNS response, damage control, and regeneration after TBI are glial cells-in reaction to tissue damage, their activation, hypertrophy, and proliferation occur, followed by the formation of a glial scar. The glial scar creates a barrier in damaged tissue and helps protect the CNS in the acute phase post-injury. However, this process prevents complete tissue recovery in the late/chronic phase by producing permanent scarring, which significantly impacts brain function. Various glial cell types participate in the scar formation, but this process is mostly attributed to reactive astrocytes and microglia, which play important roles in several brain pathologies. Novel technologies including whole-genome transcriptomic and epigenomic analyses, and unbiased proteomics, show that both astrocytes and microglia represent groups of heterogenic cell subpopulations with different genomic and functional characteristics, that are responsible for their role in neurodegeneration, neuroprotection and regeneration. Depending on the representation of distinct glia subpopulations, the tissue damage as well as the regenerative processes or delayed neurodegeneration after TBI may thus differ in nearby or remote areas or in different brain structures. This review summarizes TBI as a complex process, where the resultant effect is severity-, region- and time-dependent and determined by the model of the CNS injury and the distance of the explored area from the lesion site. Here, we also discuss findings concerning intercellular signaling, long-term impacts of TBI and the possibilities of novel therapeutical approaches. We believe that a comprehensive study with an emphasis on glial cells, involved in tissue post-injury processes, may be helpful for further research of TBI and be the decisive factor when choosing a TBI model.
Collapse
Affiliation(s)
- Zuzana Amlerova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Chmelova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Miroslava Anderova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Lydia Vargova
- Department of Neuroscience, Second Faculty of Medicine, Charles University, Prague, Czechia
- Department of Cellular Neurophysiology, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
8
|
Obenaus A, Noarbe BP, Lee JB, Panchenko PE, Noarbe SD, Lee YC, Badaut J. Progressive lifespan modifications in the corpus callosum following a single juvenile concussion in male mice monitored by diffusion MRI. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572925. [PMID: 38187748 PMCID: PMC10769374 DOI: 10.1101/2023.12.21.572925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Introduction The sensitivity of white matter (WM) in acute and chronic moderate-severe traumatic brain injury (TBI) has been established. In concussion syndromes, particularly in preclinical rodent models, there is lacking a comprehensive longitudinal study spanning the lifespan of the mouse. We previously reported early modifications to WM using clinically relevant neuroimaging and histological measures in a model of juvenile concussion at one month post injury (mpi) who then exhibited cognitive deficits at 12mpi. For the first time, we assess corpus callosum (CC) integrity across the lifespan after a single juvenile concussion utilizing diffusion MRI (dMRI). Methods C57Bl/6 mice were exposed to sham or two severities of closed-head concussion (Grade 1, G1, speed 2 m/sec, depth 1mm; Grade 2, G2, 3m/sec, 3mm) using an electromagnetic impactor at postnatal day 17. In vivo diffusion tensor imaging was conducted at 1, 3, 6, 12 and 18 mpi (21 directions, b=2000 mm2/sec) and processed for dMRI parametric maps: fractional anisotropy (FA), axial (AxD), radial (RD) and mean diffusivity (MD). Whole CC and regional CC data were extracted. To identify the biological basis of altered dMRI metrics, astrocyte and microglia in the CC were characterized at 1 and 12 mpi by immunohistochemistry. Results Whole CC analysis revealed altered FA and RD trajectories following juvenile concussion. Shams exhibited a temporally linear increase in FA with age while G1/G2 mice had plateaued FA values. G2 concussed mice exhibited high variance of dMRI metrics at 12mpi, which was attributed to the heterogeneity of TBI on the anterior CC. Regional analysis of dMRI metrics at the impact site unveiled significant differences between G2 and sham mice. The dMRI findings appear to be driven, in part, by loss of astrocyte process lengths and increased circularity and decreased cell span ratios in microglia. Conclusion For the first time, we demonstrate progressive perturbations to WM of male mice after a single juvenile concussion across the mouse lifespan. The CC alterations were dependent on concussion severity with elevated sensitivity in the anterior CC that was related to astrocyte and microglial morphology. Our findings suggest that long-term monitoring of children with juvenile concussive episodes using dMRI is warranted, focusing on vulnerable WM tracts.
Collapse
Affiliation(s)
- Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Brenda P. Noarbe
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Jeong Bin Lee
- Basic Science Department, Loma Linda University School of Medicine, Loma Linda, CA, US
| | | | - Sean D. Noarbe
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Yu Chiao Lee
- Department of Pediatrics, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Jerome Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France
| |
Collapse
|
9
|
Machado CA, Oliveira BDS, Dias TL, Barros JLVMD, Ferreira GMF, Cordeiro TM, Feracin V, Alexandre CH, Abreu LKS, Silva WND, Carvalho BC, Fernandes HDB, Vieira ÉLM, Castro PR, Ferreira RN, Kangussu LM, Franco GR, Guatimosim C, Barcelos LDS, Simões E Silva AC, Toscano ECDB, Rachid MA, Teixeira AL, Miranda ASD. Weight-drop model as a valuable tool to study potential neurobiological processes underlying behavioral and cognitive changes secondary to mild traumatic brain injury. J Neuroimmunol 2023; 385:578242. [PMID: 37951202 DOI: 10.1016/j.jneuroim.2023.578242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 10/27/2023] [Accepted: 11/05/2023] [Indexed: 11/13/2023]
Abstract
The pathophysiology of post-traumatic brain injury (TBI) behavioral and cognitive changes is not fully understood, especially in its mild presentation. We designed a weight drop TBI model in mice to investigate the role of neuroinflammation in behavioral and cognitive sequelae following mild TBI. C57BL/6 mice displayed depressive-like behavior at 72 h after mild TBI compared with controls, as indicated by a decrease in the latency to first immobility and climbing time in the forced swim test. Additionally, anxiety-like behavior and hippocampal-associated spatial learning and memory impairment were found in the elevated plus maze and in the Barnes maze, respectively. Levels of a set of inflammatory mediators and neurotrophic factors were analyzed at 6 h, 24 h, 72 h, and 30 days after injury in ipsilateral and contralateral hemispheres of the prefrontal cortex and hippocampus. Principal components analysis revealed two principal components (PC), which represented 59.1% of data variability. PC1 (cytokines and chemokines) expression varied between both hemispheres, while PC2 (neurotrophic factors) expression varied only across the investigated brain areas. Our model reproduces mild TBI-associated clinical signs and pathological features and might be a valuable tool to broaden the knowledge regarding mild TBI pathophysiology as well as to test potential therapeutic targets.
Collapse
Affiliation(s)
- Caroline Amaral Machado
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Bruna da Silva Oliveira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Thomaz Lüscher Dias
- Department of Biochemistry and Immunology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Thiago Macedo Cordeiro
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Victor Feracin
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cristian Henrique Alexandre
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Larissa Katharina Sabino Abreu
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Walison Nunes da Silva
- Department of Physiology and Biophysics, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Brener Cunha Carvalho
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Heliana de Barros Fernandes
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Érica Leandro Marciano Vieira
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Pollyana Ribeiro Castro
- Department of Physiology and Biophysics, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rodrigo Novaes Ferreira
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lucas Miranda Kangussu
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gloria Regina Franco
- Department of Biochemistry and Immunology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Cristina Guatimosim
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lucíola da Silva Barcelos
- Department of Physiology and Biophysics, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana Cristina Simões E Silva
- Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Milene Alvarenga Rachid
- Department of Pathology, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX.
| | - Aline Silva de Miranda
- Department of Morphology, Institute of Biological Science, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil; Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
10
|
Hoogenboom WS, Rubin TG, Ambadipudi K, Cui MH, Ye K, Foster H, Elkouby E, Liu J, Branch CA, Lipton ML. Evolving brain and behaviour changes in rats following repetitive subconcussive head impacts. Brain Commun 2023; 5:fcad316. [PMID: 38046094 PMCID: PMC10691880 DOI: 10.1093/braincomms/fcad316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/26/2023] [Accepted: 11/19/2023] [Indexed: 12/05/2023] Open
Abstract
There is growing concern that repetitive subconcussive head impacts, independent of concussion, alter brain structure and function, and may disproportionately affect the developing brain. Animal studies of repetitive subconcussive head impacts are needed to begin to characterize the pathological basis and mechanisms underlying imaging and functional effects of repetitive subconcussive head impacts seen in humans. Since repetitive subconcussive head impacts have been largely unexplored in animals, we aimed to characterize the evolution of imaging, behavioural and pathological effects of repetitive subconcussive head impacts in awake adolescent rodents. Awake male and female Sprague Dawley rats (postnatal Day 35) received 140 closed-head impacts over the course of a week. Impacted and sham-impacted animals were restrained in a plastic cone, and unrestrained control animals were included to account for effects of restraint and normal development. Animals (n = 43) underwent repeated diffusion tensor imaging prior to and over 1 month following the final impact. A separate cohort (n = 53) was assessed behaviourally for fine motor control, emotional-affective behaviour and memory at acute and chronic time points. Histological and immunohistochemical analyses, which were exploratory in nature due to smaller sample sizes, were completed at 1 month following the final impact. All animals tolerated the protocol with no overt changes in behaviour or stigmata of traumatic brain injury, such as alteration of consciousness, intracranial haemorrhage or skull fracture. We detected longitudinal, sex-dependent diffusion tensor imaging changes (fractional anisotropy and axial diffusivity decline) in corpus callosum and external capsule of repetitive subconcussive head impact animals, which diverged from both sham and control. Compared to sham animals, repetitive subconcussive head impact animals exhibited acute but transient mild motor deficits. Repetitive subconcussive head impact animals also exhibited chronic anxiety and spatial memory impairment that differed from the control animals, but these effects were not different from those seen in the sham condition. We observed trends in the data for thinning of the corpus callosum as well as regions with elevated Iba-1 in the corpus callosum and cerebral white matter among repetitive subconcussive head impact animals. While replication with larger study samples is needed, our findings suggest that subconcussive head impacts cause microstructural tissue changes in the developing rat brain, which are detectable with diffusion tensor imaging, with suggestion of correlates in tissue pathology and behaviour. The results point to potential mechanisms underpinning consequences of subconcussive head impacts that have been described in humans. The congruence of our imaging findings with human subconcussive head impacts suggests that neuroimaging could serve as a translational bridge to advance study of injury mechanisms and development of interventions.
Collapse
Affiliation(s)
- Wouter S Hoogenboom
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Clinical Investigation, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Todd G Rubin
- Department of Neurology, Icahn School of Medicine at Mount Sinai, NewYork, NY 10029, USA
| | - Kamalakar Ambadipudi
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Min-Hui Cui
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Kenny Ye
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Henry Foster
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Esther Elkouby
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Jinyuan Liu
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
| | - Craig A Branch
- The Gruss Magnetic Resonance Research Center, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10641, USA
- Department of Radiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
- Department of Physiology and Biophysics, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY 10461, USA
| | - Michael L Lipton
- Department of Radiology, Columbia University Irving Medical Center, NewYork, NY 10032, USA
- Department of Biomedical Engineering, Columbia University, NewYork, NY 10032, USA
| |
Collapse
|
11
|
Lee MY, Son M, Lee HH, Kang MG, Yun SJ, Seo HG, Kim Y, Oh BM. Proteomic discovery of prognostic protein biomarkers for persisting problems after mild traumatic brain injury. Sci Rep 2023; 13:19786. [PMID: 37957236 PMCID: PMC10643618 DOI: 10.1038/s41598-023-45965-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Some individuals with mild traumatic brain injury (mTBI), also known as concussion, have neuropsychiatric and physical problems that last longer than a few months. Symptoms following mTBI are not only impacted by the kind and severity of the injury but also by the post-injury experience and the individual's responses to it, making the persistence of mTBI particularly difficult to predict. We aimed to identify prognostic blood-based protein biomarkers predicting 6-month outcomes, in light of the clinical course after the injury, in a longitudinal mTBI cohort (N = 42). Among 420 target proteins quantified by multiple-reaction monitoring-mass spectrometry assays of blood samples, 31, 43, and 15 proteins were significantly associated with the poor recovery of neuropsychological symptoms at < 72 h, 1 week, and 1 month after the injury, respectively. Sequential associations among clinical assessments (depressive symptoms and cognitive function) affecting the 6-month outcomes were evaluated. Then, candidate biomarker proteins indirectly affecting the outcome via neuropsychological symptoms were identified. Using the identified proteins, prognostic models that can predict the 6-month outcome of mTBI were developed. These protein biomarkers established in the context of the clinical course of mTBI may have potential for clinical application.
Collapse
Affiliation(s)
- Min-Yong Lee
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Rehabilitation Medicine, National Traffic Injury Rehabilitation Hospital, Yangpyeong, Korea
| | - Minsoo Son
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Korea
- Mass Spectrometry Technology Access Center, McDonnell Genome Institute, Washington University School of Medicine in Saint Louis, St. Louis, MO, USA
| | - Hyun Haeng Lee
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Rehabilitation Medicine, Konkuk University School of Medicine and Konkuk University Medical Center, Seoul, Korea
| | - Min-Gu Kang
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Korea
| | - Seo Jung Yun
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Korea
| | - Han Gil Seo
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Youngsoo Kim
- Interdisciplinary Program of Bioengineering, Seoul National University College of Engineering, Seoul, Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea.
- Department of Biomedical Science, School of Medicine, CHA University, Seongnam-si, Kyeonggi-do, Korea.
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University Hospital, Seoul, Korea.
- Department of Rehabilitation Medicine, National Traffic Injury Rehabilitation Hospital, Yangpyeong, Korea.
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul, Korea.
- Institute on Aging, Seoul National University, Seoul, Korea.
| |
Collapse
|
12
|
Maliha F, Adnan A. Mechanical Responses of a Single Myelin Layer: A Molecular Simulation Study. Biomolecules 2023; 13:1525. [PMID: 37892207 PMCID: PMC10605433 DOI: 10.3390/biom13101525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/06/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The myelin sheath provides insulation to the brain's neuron cells, which aids in signal transmission and communication with the body. Degenerated myelin hampers the connection between the glial cells, which are the front row responders during traumatic brain injury mitigation. Thus, the structural integrity of the myelin layer is critical for protecting the brain tissue from traumatic injury. At the molecular level, myelin consists of a lipid bilayer, myelin basic proteins (MBP), proteolipid proteins (PLP), water and ions. Structurally, the myelin sheath is formed by repeatedly wrapping forty or more myelin layers around an axon. Here, we have used molecular dynamic simulations to model and capture the tensile response of a single myelin layer. An openly available molecular dynamic solver, LAMMPS, was used to conduct the simulations. The interatomic potentials for the interacting atoms and molecules were defined using CHARMM force fields. Following a standard equilibration process, the molecular model was stretched uniaxially at a deformation rate of 5 Å/ps. We observed that, at around 10% applied strain, the myelin started to cohesively fail via flaw formation inside the bilayers. Further stretching led to a continued expansion of the defect inside the bilayer, both radially and transversely. This study provides the cellular-level mechanisms of myelin damage due to mechanical load.
Collapse
Affiliation(s)
| | - Ashfaq Adnan
- Department of Mechanical and Aerospace Engineering, The University of Texas at Arlington, Arlington, TX 76019, USA;
| |
Collapse
|
13
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. Biomaterials 2023; 301:122210. [PMID: 37413842 PMCID: PMC10528716 DOI: 10.1016/j.biomaterials.2023.122210] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over the chronic implantation period. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
14
|
Kobayashi M, Moro N, Yoshino A, Kumagawa T, Shijo K, Maeda T, Oshima H. Inhibition of P2X4 and P2X7 receptors improves histological and behavioral outcomes after experimental traumatic brain injury in rats. Exp Ther Med 2023; 26:378. [PMID: 37456165 PMCID: PMC10347371 DOI: 10.3892/etm.2023.12077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Release of large amounts of adenosine triphosphate (ATP), a gliotransmitter, into the extracellular space by traumatic brain injury (TBI) is considered to activate the microglia followed by release of inflammatory cytokines resulting in excessive inflammatory response that induces secondary brain injury. The present study investigated whether antagonists of ATP receptors (P2X4 and/or P2X7) on microglia are beneficial for reducing the post-injury inflammatory response that leads to secondary injury, a prognostic aggravation factor of TBI. Adult male Sprague-Dawley rats were subjected to cortical contusion injury (CCI) and randomly assigned to injury and drug treatment conditions, as follows: i) No surgical intervention (naïve group); ii) dimethyl sulfoxide treatment after CCI (CCI-control group); iii) 5-BDBD (antagonist of P2X4 receptor) treatment after CCI (CCI-5-BDBD group); iv) CCI-AZ11645373 (antagonist of P2X7 receptor) treatment after CCI (CCI-AZ11645373 group); v) or 5-BDBD and AZ11645373 treatment after CCI (CCI-5-BDBD + AZ11645373 group). In the CCI-5-BDBD, CCI-AZ11645373, and CCI-5-BDBD + AZ11645373 groups, expression of activated microglia was suppressed in the ipsilateral cortex and hippocampus 3 days after the CCI. Western blotting with ionized calcium-binding adaptor molecule 1 antibody revealed that administration of CCI-5-BDBD and/or CCI-AZ11645373 suppressed expression of microglia and reduced expression of inflammatory cytokine mRNA 3 days after the CCI. Furthermore, the plus maze test, which reflects the spatial memory function and involves the hippocampal function, showed improvement 28 days after secondary injury to the hippocampus. These findings confirmed that blocking the P2X4 and P2X7 receptors, which are ATP receptors central in gliotransmission, suppresses microglial activation and subsequent cytokine expression after brain injury, and demonstrates the potential as an effective treatment for reducing secondary brain injury.
Collapse
Affiliation(s)
- Masato Kobayashi
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Nobuhiro Moro
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
- Department of Neurological Surgery, Honjo-General Hospital, Saitama 367-0031, Japan
| | - Atsuo Yoshino
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takahiro Kumagawa
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Katsunori Shijo
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takeshi Maeda
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hideki Oshima
- Division of Neurosurgery, Department of Neurological Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
15
|
Okeke O, Elbasheir A, Carter SE, Powers A, Mekawi Y, Gillespie CF, Schwartz AC, Bradley B, Fani N. Indirect Effects of Racial Discrimination on Health Outcomes Through Prefrontal Cortical White Matter Integrity. BIOLOGICAL PSYCHIATRY. COGNITIVE NEUROSCIENCE AND NEUROIMAGING 2023; 8:741-749. [PMID: 35597432 DOI: 10.1016/j.bpsc.2022.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/18/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Racial discrimination is consistently associated with adverse health outcomes and has been linked to structural decrements in brain white matter. However, it is unclear whether discrimination-related neuroplastic changes could indirectly affect health outcomes. Our goal was to evaluate indirect associations of racial discrimination on health outcomes through white matter microstructure in a sample of trauma-exposed Black women. METHODS A trauma study in an urban hospital setting recruited 79 Black women who received a history and physical examination to assess medical disorders (compiled into a summed total of disorder types). Participants reported on experiences of racial discrimination and underwent diffusion tensor imaging; fractional anisotropy values were extracted from white matter pathways previously linked to racial discrimination (corpus callosum, including the body and genu; anterior cingulum bundle; and superior longitudinal fasciculus) and entered into mediational models. RESULTS Indirect effects of racial discrimination on medical disorders through left anterior cingulum bundle fractional anisotropy were significant (β = 0.07, SE = 0.04, 95% CI [0.003, 0.14]) after accounting for trauma and economic disadvantage. Indirect effects of racial discrimination on medical disorders through corpus callosum genu fractional anisotropy were also significant (β = 0.08, SE = 0.04, 95% CI [0.01, 0.16]). CONCLUSIONS Racial discrimination may increase risk for medical disorders via neuroplastic effects on microstructural integrity of stress-sensitive prefrontal white matter tracts. Racial discrimination-related changes in these tracts may affect health behaviors, which, in turn, influence vulnerability for medical disorders. These data highlight the connections between racial discrimination, prefrontal white matter connections, and incidence of medical disorders in Black Americans.
Collapse
Affiliation(s)
- Onyebuchi Okeke
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Aziz Elbasheir
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Sierra E Carter
- Department of Psychology, Georgia State University, Atlanta, Georgia
| | - Abigail Powers
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Yara Mekawi
- Department of Psychological and Brain Sciences, University of Louisville, Louisville, Kentucky
| | - Charles F Gillespie
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Ann C Schwartz
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia
| | - Bekh Bradley
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia; Atlanta VA Medical Center, Decatur, Georgia
| | - Negar Fani
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, Georgia.
| |
Collapse
|
16
|
Adams AA, Wood TL, Kim HA. Mature and Myelinating Oligodendrocytes Are Specifically Vulnerable to Mild Fluid Percussion Injury in Mice. Neurotrauma Rep 2023; 4:433-446. [PMID: 37435356 PMCID: PMC10331160 DOI: 10.1089/neur.2023.0037] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023] Open
Abstract
Myelin loss and oligodendrocyte death are well documented in patients with traumatic brain injury (TBI), as well as in experimental animal models after moderate-to-severe TBI. In comparison, mild TBI (mTBI) does not necessarily result in myelin loss or oligodendrocyte death, but causes structural alterations in the myelin. To gain more insight into the impact of mTBI on oligodendrocyte lineage in the adult brain, we subjected mice to mild lateral fluid percussion injury (mFPI) and characterized the early impact (1 and 3 days post-injury) on oligodendrocytes in the corpus callosum using multiple oligodendrocyte lineage markers (platelet-derived growth factor receptor [PDGFR]-α, glutathione S-transferase [GST]-π, CC1, breast carcinoma-amplified sequence 1 [BCAS1], myelin basic protein [MBP], myelin-associated glycoprotein [MAG], proteolipid protein [PLP], and FluoroMyelin™). Two regions of the corpus callosum in relation to the impact site were analyzed: areas near (focal) and anterior (distal) to the impact site. mFPI did not result in oligodendrocyte death in either the focal or distal corpus callosum, nor impact on oligodendrocyte precursors (PDGFR-α+) and GST-π+ oligodendrocyte numbers. In the focal but not distal corpus callosum, mFPI caused a decrease in CC1+ as well as BCAS1+ actively myelinating oligodendrocytes and reduced FluoroMyelin intensity without altering myelin protein expression (MBP, PLP, and MAG). Disruption in node-paranode organization and loss of Nav1.6+ nodes were observed in both the focal and distal regions, even in areas without obvious axonal damage. Altogether, our study shows regional differences in mature and myelinating oligodendrocyte in response to mFPI. Further, mFPI elicits a widespread impact on node-paranode organization that affects regions both close to and remotely located from the site of injury.
Collapse
Affiliation(s)
- Alexandra A. Adams
- Department of Biological Sciences, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Teresa L. Wood
- Department of Pharmacology, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| | - Haesun A. Kim
- Department of Biological Sciences, Physiology, and Neuroscience, New Jersey Medical School, Rutgers University, Newark, New Jersey, USA
| |
Collapse
|
17
|
Glänzel NM, Parmeggiani B, Grings M, Seminotti B, Brondani M, Bobermin LD, Ribeiro CAJ, Quincozes-Santos A, Vockley J, Leipnitz G. Myelin Disruption, Neuroinflammation, and Oxidative Stress Induced by Sulfite in the Striatum of Rats Are Mitigated by the pan-PPAR agonist Bezafibrate. Cells 2023; 12:1557. [PMID: 37371027 DOI: 10.3390/cells12121557] [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: 04/01/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Sulfite predominantly accumulates in the brain of patients with isolated sulfite oxidase (ISOD) and molybdenum cofactor (MoCD) deficiencies. Patients present with severe neurological symptoms and basal ganglia alterations, the pathophysiology of which is not fully established. Therapies are ineffective. To elucidate the pathomechanisms of ISOD and MoCD, we investigated the effects of intrastriatal administration of sulfite on myelin structure, neuroinflammation, and oxidative stress in rat striatum. Sulfite administration decreased FluoromyelinTM and myelin basic protein staining, suggesting myelin abnormalities. Sulfite also increased the staining of NG2, a protein marker of oligodendrocyte progenitor cells. In line with this, sulfite also reduced the viability of MO3.13 cells, which express oligodendroglial markers. Furthermore, sulfite altered the expression of interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-10 (IL-10), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS) and heme oxygenase-1 (HO-1), indicating neuroinflammation and redox homeostasis disturbances. Iba1 staining, another marker of neuroinflammation, was also increased by sulfite. These data suggest that myelin changes and neuroinflammation induced by sulfite contribute to the pathophysiology of ISOD and MoCD. Notably, post-treatment with bezafibrate (BEZ), a pan-PPAR agonist, mitigated alterations in myelin markers and Iba1 staining, and IL-1β, IL-6, iNOS and HO-1 expression in the striatum. MO3.13 cell viability decrease was further prevented. Moreover, pre-treatment with BEZ also attenuated some effects. These findings show the modulation of PPAR as a potential opportunity for therapeutic intervention in these disorders.
Collapse
Affiliation(s)
- Nícolas Manzke Glänzel
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Belisa Parmeggiani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Mateus Grings
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Bianca Seminotti
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Morgana Brondani
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Larissa D Bobermin
- Programa de Pós-Graduação Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - César A J Ribeiro
- Natural and Humanities Sciences Center, Universidade Federal do ABC, São Bernardo do Campo 09606-070, SP, Brazil
| | - André Quincozes-Santos
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Programa de Pós-Graduação Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| | - Jerry Vockley
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Programa de Pós-Graduação Neurociências, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, Porto Alegre 90035-003, RS, Brazil
| |
Collapse
|
18
|
Doust YV, Bindoff A, Holloway OG, Wilson R, King AE, Ziebell JM. Temporal changes in the microglial proteome of male and female mice after a diffuse brain injury using label-free quantitative proteomics. Glia 2023; 71:880-903. [PMID: 36468604 PMCID: PMC10952308 DOI: 10.1002/glia.24313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022]
Abstract
Traumatic brain injury (TBI) triggers neuroinflammatory cascades mediated by microglia, which promotes tissue repair in the short-term. These cascades may exacerbate TBI-induced tissue damage and symptoms in the months to years post-injury. However, the progression of the microglial function across time post-injury and whether this differs between biological sexes is not well understood. In this study, we examined the microglial proteome at 3-, 7-, or 28-days after a midline fluid percussion injury (mFPI) in male and female mice using label-free quantitative proteomics. Data are available via ProteomeXchange with identifier PXD033628. We identified a reduction in microglial proteins involved with clearance of neuronal debris via phagocytosis at 3- and 7-days post-injury. At 28 days post-injury, pro-inflammatory proteins were decreased and anti-inflammatory proteins were increased in microglia. These results indicate a reduction in microglial clearance of neuronal debris in the days post-injury with a shift to anti-inflammatory function by 28 days following TBI. The changes in the microglial proteome that occurred across time post-injury did not differ between biological sexes. However, we did identify an increase in microglial proteins related to pro-inflammation and phagocytosis as well as insulin and estrogen signaling in males compared with female mice that occurred with or without a brain injury. Although the microglial response was similar between males and females up to 28 days following TBI, biological sex differences in the microglial proteome, regardless of TBI, has implications for the efficacy of treatment strategies targeting the microglial response post-injury.
Collapse
Affiliation(s)
- Yasmine V. Doust
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Olivia G. Holloway
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Richard Wilson
- Central Science Laboratory (CSL)University of TasmaniaHobartTasmaniaAustralia
| | - Anna E. King
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| | - Jenna M. Ziebell
- Wicking Dementia Research and Education Centre, College of Health and MedicineUniversity of TasmaniaHobartTasmaniaAustralia
| |
Collapse
|
19
|
Chen K, Cambi F, Kozai TDY. Pro-myelinating Clemastine administration improves recording performance of chronically implanted microelectrodes and nearby neuronal health. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.31.526463. [PMID: 36778360 PMCID: PMC9915570 DOI: 10.1101/2023.01.31.526463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Intracortical microelectrodes have become a useful tool in neuroprosthetic applications in the clinic and to understand neurological disorders in basic neurosciences. Many of these brain-machine interface technology applications require successful long-term implantation with high stability and sensitivity. However, the intrinsic tissue reaction caused by implantation remains a major failure mechanism causing loss of recorded signal quality over time. Oligodendrocytes remain an underappreciated intervention target to improve chronic recording performance. These cells can accelerate action potential propagation and provides direct metabolic support for neuronal health and functionality. However, implantation injury causes oligodendrocyte degeneration and leads to progressive demyelination in surrounding brain tissue. Previous work highlighted that healthy oligodendrocytes are necessary for greater electrophysiological recording performance and the prevention of neuronal silencing around implanted microelectrodes over chronic implantation. Thus, we hypothesize that enhancing oligodendrocyte activity with a pharmaceutical drug, Clemastine, will prevent the chronic decline of microelectrode recording performance. Electrophysiological evaluation showed that the promyelination Clemastine treatment significantly elevated the signal detectability and quality, rescued the loss of multi-unit activity, and increased functional interlaminar connectivity over 16-weeks of implantation. Additionally, post-mortem immunohistochemistry showed that increased oligodendrocyte density and myelination coincided with increased survival of both excitatory and inhibitory neurons near the implant. Overall, we showed a positive relationship between enhanced oligodendrocyte activity and neuronal health and functionality near the chronically implanted microelectrode. This study shows that therapeutic strategy that enhance oligodendrocyte activity is effective for integrating the functional device interface with brain tissue over chronic implantation period. Abstract Figure
Collapse
|
20
|
Cramer SW, Haley SP, Popa LS, Carter RE, Scott E, Flaherty EB, Dominguez J, Aronson JD, Sabal L, Surinach D, Chen CC, Kodandaramaiah SB, Ebner TJ. Wide-field calcium imaging reveals widespread changes in cortical functional connectivity following mild traumatic brain injury in the mouse. Neurobiol Dis 2023; 176:105943. [PMID: 36476979 PMCID: PMC9972226 DOI: 10.1016/j.nbd.2022.105943] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/29/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
>2.5 million individuals in the United States suffer mild traumatic brain injuries (mTBI) annually. Mild TBI is characterized by a brief period of altered consciousness, without objective findings of anatomic injury on clinical imaging or physical deficit on examination. Nevertheless, a subset of mTBI patients experience persistent subjective symptoms and repeated mTBI can lead to quantifiable neurological deficits, suggesting that each mTBI alters neurophysiology in a deleterious manner not detected using current clinical methods. To better understand these effects, we performed mesoscopic Ca2+ imaging in mice to evaluate how mTBI alters patterns of neuronal interactions across the dorsal cerebral cortex. Spatial Independent Component Analysis (sICA) and Localized semi-Nonnegative Matrix Factorization (LocaNMF) were used to quantify changes in cerebral functional connectivity (FC). Repetitive, mild, controlled cortical impacts induce temporary neuroinflammatory responses, characterized by increased density of microglia exhibiting de-ramified morphology. These temporary neuro-inflammatory changes were not associated with compromised cognitive performance in the Barnes maze or motor function as assessed by rotarod. However, long-term alterations in functional connectivity (FC) were observed. Widespread, bilateral changes in FC occurred immediately following impact and persisted for up to 7 weeks, the duration of the experiment. Network alterations include decreases in global efficiency, clustering coefficient, and nodal strength, thereby disrupting functional interactions and information flow throughout the dorsal cerebral cortex. A subnetwork analysis shows the largest disruptions in FC were concentrated near the impact site. Therefore, mTBI induces a transient neuroinflammation, without alterations in cognitive or motor behavior, and a reorganized cortical network evidenced by the widespread, chronic alterations in cortical FC.
Collapse
Affiliation(s)
- Samuel W Cramer
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Samuel P Haley
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Laurentiu S Popa
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Russell E Carter
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Earl Scott
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Evelyn B Flaherty
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Judith Dominguez
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Justin D Aronson
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Luke Sabal
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel Surinach
- Department of Mechanical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Timothy J Ebner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
21
|
Ross DE, Seabaugh JD, Seabaugh JM, Alvarez C, Ellis LP, Powell C, Reese C, Cooper L, Shepherd K, Alzheimer's Disease Neuroimaging Initiative FT. Journey to the other side of the brain: asymmetry in patients with chronic mild or moderate traumatic brain injury. Concussion 2022; 8:CNC101. [PMID: 36874877 PMCID: PMC9979152 DOI: 10.2217/cnc-2022-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 12/01/2022] [Indexed: 02/01/2023] Open
Abstract
Aim Patients with chronic mild or moderate traumatic brain injury have some regions of brain atrophy (including cerebral white matter) but even more regions of abnormal brain enlargement (including other cerebral regions). Hypothesis Ipsilateral injury and atrophy cause the eventual development of contralateral compensatory hypertrophy. Materials & methods 50 patients with mild or moderate traumatic brain injury were compared to 80 normal controls (n = 80) with respect to MRI brain volume asymmetry. Asymmetry-based correlations were used to test the primary hypothesis. Results The group of patients had multiple regions of abnormal asymmetry. Conclusion The correlational analyses supported the conclusion that acute injury to ipsilateral cerebral white matter regions caused atrophy, leading eventually to abnormal enlargement of contralateral regions due to compensatory hypertrophy.
Collapse
Affiliation(s)
- David E Ross
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA
| | - John D Seabaugh
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA
| | - Jan M Seabaugh
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA
| | - Claudia Alvarez
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA.,Neuroscience Department, Randolph Macon College, Ashland, VA 23005, USA
| | - Laura Peyton Ellis
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA.,Neuroscience Department, Randolph Macon College, Ashland, VA 23005, USA
| | - Christopher Powell
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA.,Virginia Commonwealth University, Medical College of Virginia, Richmond, VA 23219, USA
| | - Christopher Reese
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA.,Neuroscience Department, University of North Carolina at Wilmington, Wilmington, NC 28403, USA
| | - Leah Cooper
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA.,Neuroscience Department, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA
| | - Katherine Shepherd
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA.,Neuroscience Department, James Madison University, Harrisonburg, VA 22807, USA
| | - For The Alzheimer's Disease Neuroimaging Initiative
- Virginia Institute of Neuropsychiatry, Midlothian, VA 23114, USA.,Neuroscience Department, Randolph Macon College, Ashland, VA 23005, USA.,Virginia Commonwealth University, Medical College of Virginia, Richmond, VA 23219, USA.,Neuroscience Department, University of North Carolina at Wilmington, Wilmington, NC 28403, USA.,Neuroscience Department, Virginia Polytechnic Institute & State University, Blacksburg, VA 24061, USA.,Neuroscience Department, James Madison University, Harrisonburg, VA 22807, USA
| |
Collapse
|
22
|
Clay AM, Carr R, Dubien J, To F. Short-term behavioral and histological changes in a rodent model of mild traumatic brain injury. BIOMEDICAL ENGINEERING ADVANCES 2022. [DOI: 10.1016/j.bea.2022.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
23
|
Juan SMA, Daglas M, Adlard PA. Altered amyloid precursor protein, tau-regulatory proteins, neuronal numbers and behaviour, but no tau pathology, synaptic and inflammatory changes or memory deficits, at 1 month following repetitive mild traumatic brain injury. Eur J Neurosci 2022; 56:5342-5367. [PMID: 35768153 DOI: 10.1111/ejn.15752] [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: 09/14/2021] [Revised: 05/31/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022]
Abstract
Repetitive mild traumatic brain injury, commonly experienced following sports injuries, results in various secondary injury processes and is increasingly recognised as a risk factor for the development of neurodegenerative conditions such as chronic traumatic encephalopathy, which is characterised by tau pathology. We aimed to characterise the underlying pathological mechanisms that might contribute to the onset of neurodegeneration and behavioural changes in the less-explored subacute (1-month) period following single or repetitive controlled cortical impact injury (five impacts, 48 h apart) in 12-week-old male and female C57Bl6 mice. We conducted motor and cognitive testing, extensively characterised the status of tau and its regulatory proteins via western blot and quantified neuronal populations using stereology. We report that r-mTBI resulted in neurobehavioural deficits, gait impairments and anxiety-like behaviour at 1 month post-injury, effects not seen following a single injury. R-mTBI caused a significant increase in amyloid precursor protein, an increased trend towards tau phosphorylation and significant changes in kinase/phosphatase proteins that may promote a downstream increase in tau phosphorylation, but no changes in synaptic or neuroinflammatory markers. Lastly, we report neuronal loss in various brain regions following both single and repeat injuries. We demonstrate herein that repeated impacts are required to promote the initiation of a cascade of biochemical events that are consistent with the onset of neurodegeneration subacutely post-injury. Identifying the timeframe in which these changes occur and the pathological mechanisms involved will be crucial for the development of future therapeutics to prevent the onset or mitigate the progression of neurodegeneration following r-mTBI.
Collapse
Affiliation(s)
- Sydney M A Juan
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The Melbourne Dementia Research Centre and The University of Melbourne, Melbourne, Australia
| | - Maria Daglas
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The Melbourne Dementia Research Centre and The University of Melbourne, Melbourne, Australia
| | - Paul A Adlard
- Synaptic Neurobiology Laboratory, The Florey Institute of Neuroscience and Mental Health, The Melbourne Dementia Research Centre and The University of Melbourne, Melbourne, Australia
| |
Collapse
|
24
|
Song S, Hasan MN, Yu L, Paruchuri SS, Bielanin JP, Metwally S, Oft HCM, Fischer SG, Fiesler VM, Sen T, Gupta RK, Foley LM, Hitchens TK, Dixon CE, Cambi F, Sen N, Sun D. Microglial-oligodendrocyte interactions in myelination and neurological function recovery after traumatic brain injury. J Neuroinflammation 2022; 19:246. [PMID: 36199097 PMCID: PMC9533529 DOI: 10.1186/s12974-022-02608-6] [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: 06/24/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
Differential microglial inflammatory responses play a role in regulation of differentiation and maturation of oligodendrocytes (OLs) in brain white matter. How microglia-OL crosstalk is altered by traumatic brain injury (TBI) and its impact on axonal myelination and neurological function impairment remain poorly understood. In this study, we investigated roles of a Na+/H+ exchanger (NHE1), an essential microglial pH regulatory protein, in microglial proinflammatory activation and OL survival and differentiation in a murine TBI model induced by controlled cortical impact. Similar TBI-induced contusion volumes were detected in the Cx3cr1-CreERT2 control (Ctrl) mice and selective microglial Nhe1 knockout (Cx3cr1-CreERT2;Nhe1flox/flox, Nhe1 cKO) mice. Compared to the Ctrl mice, the Nhe1 cKO mice displayed increased resistance to initial TBI-induced white matter damage and accelerated chronic phase of OL regeneration at 30 days post-TBI. The cKO brains presented increased anti-inflammatory phenotypes of microglia and infiltrated myeloid cells, with reduced proinflammatory transcriptome profiles. Moreover, the cKO mice exhibited accelerated post-TBI sensorimotor and cognitive functional recovery than the Ctrl mice. These phenotypic outcomes in cKO mice were recapitulated in C57BL6J wild-type TBI mice receiving treatment of a potent NHE1 inhibitor HOE642 for 1-7 days post-TBI. Taken together, these findings collectively demonstrated that blocking NHE1 protein stimulates restorative microglial activation in oligodendrogenesis and neuroprotection, which contributes to accelerated brain repair and neurological function recovery after TBI.
Collapse
Affiliation(s)
- Shanshan Song
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA
| | - Md Nabiul Hasan
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA
| | - Lauren Yu
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Satya S Paruchuri
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - John P Bielanin
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Shamseldin Metwally
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Helena C M Oft
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Sydney G Fischer
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Victoria M Fiesler
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA
| | - Tanusree Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Rajaneesh K Gupta
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Lesley M Foley
- Animal Imaging Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - T Kevin Hitchens
- Animal Imaging Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - C Edward Dixon
- Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA.,Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Franca Cambi
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA.,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA.,Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA
| | - Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Dandan Sun
- Department of Neurology, University of Pittsburgh, 3501 Fifth Avenue, Pittsburgh, PA, 15213, USA. .,Pittsburgh Institute for Neurodegenerative Disorders, University of Pittsburgh, Pittsburgh, PA, 15213, USA. .,Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, 15213, USA.
| |
Collapse
|
25
|
Arneson D, Zhang G, Ahn IS, Ying Z, Diamante G, Cely I, Palafox-Sanchez V, Gomez-Pinilla F, Yang X. Systems spatiotemporal dynamics of traumatic brain injury at single-cell resolution reveals humanin as a therapeutic target. Cell Mol Life Sci 2022; 79:480. [PMID: 35951114 PMCID: PMC9372016 DOI: 10.1007/s00018-022-04495-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/10/2022] [Accepted: 07/17/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND The etiology of mild traumatic brain injury (mTBI) remains elusive due to the tissue and cellular heterogeneity of the affected brain regions that underlie cognitive impairments and subsequent neurological disorders. This complexity is further exacerbated by disrupted circuits within and between cell populations across brain regions and the periphery, which occur at different timescales and in spatial domains. METHODS We profiled three tissues (hippocampus, frontal cortex, and blood leukocytes) at the acute (24-h) and subacute (7-day) phases of mTBI at single-cell resolution. RESULTS We demonstrated that the coordinated gene expression patterns across cell types were disrupted and re-organized by TBI at different timescales with distinct regional and cellular patterns. Gene expression-based network modeling implied astrocytes as a key regulator of the cell-cell coordination following mTBI in both hippocampus and frontal cortex across timepoints, and mt-Rnr2, which encodes the mitochondrial peptide humanin, as a potential target for intervention based on its broad regional and dynamic dysregulation following mTBI. Treatment of a murine mTBI model with humanin reversed cognitive impairment caused by mTBI through the restoration of metabolic pathways within astrocytes. CONCLUSIONS Our results offer a systems-level understanding of the dynamic and spatial regulation of gene programs by mTBI and pinpoint key target genes, pathways, and cell circuits that are amenable to therapeutics.
Collapse
Affiliation(s)
- Douglas Arneson
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Zhe Ying
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Ingrid Cely
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Victoria Palafox-Sanchez
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Fernando Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Department of Neurosurgery, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
26
|
Cruz-Méndez JS, Herrera-Sánchez MP, Céspedes-Rubio ÁE, Rondón-Barragán IS. Molecular characterization of myelin basic protein a (mbpa) gene from red-bellied pacu (Piaractus brachypomus). JOURNAL OF GENETIC ENGINEERING AND BIOTECHNOLOGY 2022; 20:8. [PMID: 35024999 PMCID: PMC8758815 DOI: 10.1186/s43141-022-00296-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/04/2022] [Indexed: 12/18/2022]
Abstract
Background Myelin basic protein (MBP) is one of the most important structural components of the myelin sheaths in both central and peripheral nervous systems. MBP has several functions including organization of the myelin membranes, reorganization of the cytoskeleton during the myelination process, and interaction with the SH3 domain in signaling pathways. Likewise, MBP has been proposed as a marker of demyelination in traumatic brain injury and chemical exposure. Methods The aim of this study was to molecularly characterize the myelin basic protein a (mbpa) gene from the Colombian native fish, red-bellied pacu, Piaractus brachypomus. Bioinformatic tools were used to identify the phylogenetic relationships, physicochemical characteristics, exons, intrinsically disordered regions, and conserved domains of the protein. Gene expression was assessed by qPCR in three models corresponding to sublethal chlorpyrifos exposure, acute brain injury, and anesthesia experiments. Results mbpa complete open reading frame was identified with 414 nucleotides distributed in 7 exons that encode 137 amino acids. MBPa was recognized as belonging to the myelin basic protein family, closely related with orthologous proteins, and two intrinsically disordered regions were established within the sequence. Gene expression of mbpa was upregulated in the optic chiasm of the chlorpyrifos exposed fish in contrast to the control group. Conclusions The physicochemical computed features agree with the biological functions of MBP, and basal gene expression was according to the anatomical distribution in the tissues analyzed. This study is the first molecular characterization of mbpa from the native species Piaractus brachypomus.
Collapse
Affiliation(s)
- Juan Sebastian Cruz-Méndez
- Research Group in Immunobiology and Pathogenesis, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Santa Helena Highs, 730006299, Ibague, Tolima, Colombia
| | - María Paula Herrera-Sánchez
- Research Group in Immunobiology and Pathogenesis, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Santa Helena Highs, 730006299, Ibague, Tolima, Colombia
| | - Ángel Enrique Céspedes-Rubio
- Research Group in Neurodegenerative Diseases, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Santa Helena Highs, 730006299, Ibague, Tolima, Colombia
| | - Iang Schroniltgen Rondón-Barragán
- Research Group in Immunobiology and Pathogenesis, Laboratory of Immunology and Molecular Biology, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Santa Helena Highs, 730006299, Ibague, Tolima, Colombia. .,Research Group in Neurodegenerative Diseases, Faculty of Veterinary Medicine and Zootechnics, Universidad del Tolima, Santa Helena Highs, 730006299, Ibague, Tolima, Colombia. .,Laboratory of Immunology and Molecular Biology, Department of Animal Health, Faculty of Veterinary Medicine and Zootechnics, University of Tolima, Ibague, 730006299, Colombia.
| |
Collapse
|
27
|
Kumagawa T, Moro N, Maeda T, Kobayashi M, Furukawa Y, Shijo K, Yoshino A. Anti-inflammatory effect of P2Y1 receptor blocker MRS2179 in a rat model of traumatic brain injury. Brain Res Bull 2022; 181:46-54. [DOI: 10.1016/j.brainresbull.2022.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 01/16/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023]
|
28
|
Delage C, Vignal N, Guerin C, Taib T, Barboteau C, Mamma C, Khacef K, Margaill I, Sarda-Mantel L, Rizzo-Padoin N, Hontonnou F, Marchand-Leroux C, Lerouet D, Hosten B, Besson V. From positron emission tomography to cell analysis of the 18-kDa Translocator Protein in mild traumatic brain injury. Sci Rep 2021; 11:24009. [PMID: 34907268 PMCID: PMC8671393 DOI: 10.1038/s41598-021-03416-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 11/17/2021] [Indexed: 11/24/2022] Open
Abstract
Traumatic brain injury (TBI) leads to a deleterious neuroinflammation, originating from microglial activation. Monitoring microglial activation is an indispensable step to develop therapeutic strategies for TBI. In this study, we evaluated the use of the 18-kDa translocator protein (TSPO) in positron emission tomography (PET) and cellular analysis to monitor microglial activation in a mild TBI mouse model. TBI was induced on male Swiss mice. PET imaging analysis with [18F]FEPPA, a TSPO radiotracer, was performed at 1, 3 and 7 days post-TBI and flow cytometry analysis on brain at 1 and 3 days post-TBI. PET analysis showed no difference in TSPO expression between non-operated, sham-operated and TBI mice. Flow cytometry analysis demonstrated an increase in TSPO expression in ipsilateral brain 3 days post-TBI, especially in microglia, macrophages, lymphocytes and neutrophils. Moreover, microglia represent only 58.3% of TSPO+ cells in the brain. Our results raise the question of the use of TSPO radiotracer to monitor microglial activation after TBI. More broadly, flow cytometry results point the lack of specificity of TSPO for microglia and imply that microglia contribute to the overall increase in TSPO in the brain after TBI, but is not its only contributor.
Collapse
Affiliation(s)
- Clément Delage
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France.
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France.
| | - Nicolas Vignal
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
| | - Coralie Guerin
- Université de Paris, Innovative Therapies in Haemostasis, Inserm, 75006, Paris, France
- Institut Curie, Cytometry Core, 75005, Paris, France
- Université de Paris, Inserm UMS 3612 CNRS - US25 Inserm -Faculté de Pharmacie de Paris, Paris, France
| | - Toufik Taib
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Clément Barboteau
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Célia Mamma
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Kahina Khacef
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
| | - Isabelle Margaill
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1140, Paris, France
| | - Laure Sarda-Mantel
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service de Médecine Nucléaire, Hôpital Lariboisière, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
| | - Nathalie Rizzo-Padoin
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- CHU de Martinique, Service Pharmacie, Hôpital Pierre Zobda-Quitman, Fort-de-France, France
| | - Fortune Hontonnou
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- Université de Paris, Inserm UMR-S 942, Hôpital Lariboisière, Paris, France
| | - Catherine Marchand-Leroux
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Dominique Lerouet
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Benoit Hosten
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
- Université de Paris, Institut de Recherche Saint-Louis, Unité Claude Kellershohn, Paris, France
- Assistance Publique - Hôpitaux de Paris (AP-HP), Service Pharmacie, Hôpital Saint-Louis, Paris, France
| | - Valérie Besson
- Faculté de Pharmacie de Paris, Université Paris Descartes, EA4475 - Pharmacologie de la circulation cérébrale, Paris, France
- Faculté de Pharmacie de Paris, Université de Paris, Inserm UMR-S 1144 - Optimisation Thérapeutique en Neuropsychopharmacologie, 4 avenue de l'Observatoire, 75006, Paris, France
| |
Collapse
|
29
|
Regulation of the Fructose Transporter Gene Slc2a5 Expression by Glucose in Cultured Microglial Cells. Int J Mol Sci 2021; 22:ijms222312668. [PMID: 34884473 PMCID: PMC8657830 DOI: 10.3390/ijms222312668] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 01/16/2023] Open
Abstract
Microglia play a role in the regulation of metabolism and pathogenesis of obesity. Microglial activity is altered in response to changes in diet and the body’s metabolic state. Solute carrier family 2 member 5 (Slc2a5) that encodes glucose transporter 5 (GLUT5) is a fructose transporter primarily expressed in microglia within the central nervous system. However, little is known about the nutritional regulation of Slc2a5 expression in microglia and its role in the regulation of metabolism. The present study aimed to address the hypothesis that nutrients affect microglial activity by altering the expression of glucose transporter genes. Murine microglial cell line SIM-A9 cells and primary microglia from mouse brain were exposed to different concentrations of glucose and levels of microglial activation markers and glucose transporter genes were measured. High concentration of glucose increased levels of the immediate-early gene product c-Fos, a marker of cell activation, Slc2a5 mRNA, and pro-inflammatory cytokine genes in microglial cells in a time-dependent manner, while fructose failed to cause these changes. Glucose-induced changes in pro-inflammatory gene expression were partially attenuated in SIM-A9 cells treated with the GLUT5 inhibitor. These findings suggest that an increase in local glucose availability leads to the activation of microglia by controlling their carbohydrate sensing mechanism through both GLUT5-dependent and –independent mechanisms.
Collapse
|
30
|
Mira RG, Lira M, Cerpa W. Traumatic Brain Injury: Mechanisms of Glial Response. Front Physiol 2021; 12:740939. [PMID: 34744783 PMCID: PMC8569708 DOI: 10.3389/fphys.2021.740939] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/20/2021] [Indexed: 11/17/2022] Open
Abstract
Traumatic brain injury (TBI) is a heterogeneous disorder that involves brain damage due to external forces. TBI is the main factor of death and morbidity in young males with a high incidence worldwide. TBI causes central nervous system (CNS) damage under a variety of mechanisms, including synaptic dysfunction, protein aggregation, mitochondrial dysfunction, oxidative stress, and neuroinflammation. Glial cells comprise most cells in CNS, which are mediators in the brain’s response to TBI. In the CNS are present astrocytes, microglia, oligodendrocytes, and polydendrocytes (NG2 cells). Astrocytes play critical roles in brain’s ion and water homeostasis, energy metabolism, blood-brain barrier, and immune response. In response to TBI, astrocytes change their morphology and protein expression. Microglia are the primary immune cells in the CNS with phagocytic activity. After TBI, microglia also change their morphology and release both pro and anti-inflammatory mediators. Oligodendrocytes are the myelin producers of the CNS, promoting axonal support. TBI causes oligodendrocyte apoptosis, demyelination, and axonal transport disruption. There are also various interactions between these glial cells and neurons in response to TBI that contribute to the pathophysiology of TBI. In this review, we summarize several glial hallmarks relevant for understanding the brain injury and neuronal damage under TBI conditions.
Collapse
Affiliation(s)
- Rodrigo G Mira
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Matías Lira
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Waldo Cerpa
- Laboratorio de Función y Patología Neuronal, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| |
Collapse
|
31
|
Lucas-Ruiz F, Galindo-Romero C, Albaladejo-García V, Vidal-Sanz M, Agudo-Barriuso M. Mechanisms implicated in the contralateral effect in the central nervous system after unilateral injury: focus on the visual system. Neural Regen Res 2021; 16:2125-2131. [PMID: 33818483 PMCID: PMC8354113 DOI: 10.4103/1673-5374.310670] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 11/21/2020] [Accepted: 01/11/2021] [Indexed: 12/21/2022] Open
Abstract
The retina, as part of the central nervous system is an ideal model to study the response of neurons to injury and disease and to test new treatments. During the last decade is becoming clear that unilateral lesions in bilateral areas of the central nervous system trigger an inflammatory response in the contralateral uninjured site. This effect has been better studied in the visual system where, as a rule, one retina is used as experimental and the other as control. Contralateral retinas in unilateral models of retinal injury show neuronal degeneration and glial activation. The mechanisms by which this adverse response in the central nervous system occurs are discussed in this review, focusing primarily on the visual system.
Collapse
Affiliation(s)
- Fernando Lucas-Ruiz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Caridad Galindo-Romero
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Virginia Albaladejo-García
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Manuel Vidal-Sanz
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| | - Marta Agudo-Barriuso
- Departamento de Oftalmología, Facultad de Medicina, Universidad de Murcia and Instituto Murciano de Investigación Biosanitaria-Virgen de la Arrixaca (IMIBArrixaca) Murcia, Spain
| |
Collapse
|
32
|
Neuroprotective and anti-neuropathic actions of pulsed magnetic fields with low frequencies in rats with chronic peripheral neuropathic pain. Brain Res Bull 2021; 177:273-281. [PMID: 34688831 DOI: 10.1016/j.brainresbull.2021.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/15/2021] [Accepted: 10/19/2021] [Indexed: 12/14/2022]
Abstract
The management of chronic peripheral neuropathic pain conditions with conventional treatments is still limited. In this present study, we aimed to determine the anti-neuropathic actions of pulsed magnetic field (PMF) treatments as a therapeutic. Effects of daily PMF treatments for 4 weeks were investigated by examining pain behaviors, hyperalgesia and allodynia, electrophysiological parameters, amplitude of compound action potential (CAP) and sciatic nerve conduction velocity (SNCV) and histopathological changes in rats with chronic constriction injury (CCI). Peripheral and central pro-inflammatory cytokines (TNF α, IL-1β and IL-17), chemokines (CCL3 and CXCL1) and angiogenic factors (VEGF and bFGF) in sciatic nerves and spinal cord tissues were also measured for determining the possible molecular action mechanisms of PMF treatment. Hyperalgesia and allodynia were observed at the first week and lasted for 4 weeks after CCI. PMF treatments caused time-dependent anti-hyperalgesic and anti-allodynic effects. PMF treatment alleviated the histopathological consequences of CCI on sciatic nerve and significantly improved the amplitude of the CAP and SNCV. PMF treatment inhibited the pro-inflammatory molecules and promoted the anti-inflammatory cytokines in neural tissues. PMF treatment also suppressed the VEGF levels and enhanced the bFGF levels in both neural tissues. The results of the present study suggested that daily PMF treatment may have neuroprotective and anti-neuropathic pain actions in rats with CCI-induced neuropathy due to its modulating effects on neuro-inflammatory and neuro-angiogenic mediators in central and peripheral neural tissues.
Collapse
|
33
|
Traumatic Brain Injury: An Age-Dependent View of Post-Traumatic Neuroinflammation and Its Treatment. Pharmaceutics 2021; 13:pharmaceutics13101624. [PMID: 34683918 PMCID: PMC8537402 DOI: 10.3390/pharmaceutics13101624] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability all over the world. TBI leads to (1) an inflammatory response, (2) white matter injuries and (3) neurodegenerative pathologies in the long term. In humans, TBI occurs most often in children and adolescents or in the elderly, and it is well known that immune responses and the neuroregenerative capacities of the brain, among other factors, vary over a lifetime. Thus, age-at-injury can influence the consequences of TBI. Furthermore, age-at-injury also influences the pharmacological effects of drugs. However, the post-TBI inflammatory, neuronal and functional consequences have been mostly studied in experimental young adult animal models. The specificity and the mechanisms underlying the consequences of TBI and pharmacological responses are poorly understood in extreme ages. In this review, we detail the variations of these age-dependent inflammatory responses and consequences after TBI, from an experimental point of view. We investigate the evolution of microglial, astrocyte and other immune cells responses, and the consequences in terms of neuronal death and functional deficits in neonates, juvenile, adolescent and aged male animals, following a single TBI. We also describe the pharmacological responses to anti-inflammatory or neuroprotective agents, highlighting the need for an age-specific approach to the development of therapies of TBI.
Collapse
|
34
|
McCunn P, Xu X, Moszczynski A, Li A, Brown A, Bartha R. Neurite orientation dispersion and density imaging in a rodent model of acute mild traumatic brain injury. J Neuroimaging 2021; 31:879-892. [PMID: 34473386 DOI: 10.1111/jon.12917] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 07/24/2021] [Accepted: 07/27/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Identification of changesin brain microstructure following mild traumatic brain injury (mTBI) could be instrumental in understanding the underlying pathophysiology. The purpose of this study was to apply neurite orientation dispersion and density imaging (NODDI) to a rodent model of mTBI to determine whether microstructural changes could be detected immediately following injury. METHODS Fifteen adult male Wistar rats were scanned on a Bruker 9.4 Tesla small animal MRI using a multi-shell acquisition (30 b = 1000 s/mm2 and 60 b = 2000 s/mm2 ). Nine animals experienced a single closed head controlled cortical impact followed by NODDI from 1 to 4 h post injury. Region of interest analysis focused on the corpus callosum and hippocampus. A mixed analysis of variance (ANOVA) was used to determine statistically significant interactions in neurite density index (NDI), orientation dispersion index (ODI), fractional anisotropy (FA), mean diffusivity (MD), axial diffusivity (AD), and radial diffusivity. Follow up repeated-measures ANOVAs were used to determine individual changes over time. RESULTS NDI showed a significant increase in the hippocampus and corpus callosum following injury, while ODI showed increases in the corpus callosum. No significant changes were observed in the sham control animals. No changes were found in FA, MD, AD, or RD. Histological analysis revealed increased glial fibrillary acidic protein staining relative to controls in both the hippocampus and corpus callosum, with evidence of activated astrocytes in these regions. CONCLUSIONS Changes in NODDI metrics were detected as early as 1 h following mTBI. No changes were detected with conventional diffusion tensor imaging (DTI) metrics, suggesting that NODDI provides greater sensitivity to microstructural changes than conventional DTI.
Collapse
Affiliation(s)
- Patrick McCunn
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada
| | - Xiaoyun Xu
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada
| | | | - Alex Li
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada.,Departments of Psychiatry and Medical Imaging, University of Western Ontario, London, Ontario, Canada
| | - Arthur Brown
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Neuroscience, University of Western Ontario, London, Ontario, Canada
| | - Robert Bartha
- Robarts Research Institute, University of Western Ontario, London, Ontario, Canada.,Department of Medical Biophysics, University of Western Ontario, London, Ontario, Canada.,Department of Anatomy and Cell Biology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
35
|
Lyu Z, Park J, Kim KM, Jin HJ, Wu H, Rajadas J, Kim DH, Steinberg GK, Lee W. A neurovascular-unit-on-a-chip for the evaluation of the restorative potential of stem cell therapies for ischaemic stroke. Nat Biomed Eng 2021; 5:847-863. [PMID: 34385693 PMCID: PMC8524779 DOI: 10.1038/s41551-021-00744-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 04/30/2021] [Indexed: 02/07/2023]
Abstract
The therapeutic efficacy of stem cells transplanted into an ischaemic brain depends primarily on the responses of the neurovascular unit. Here, we report the development and applicability of a functional neurovascular unit on a microfluidic chip as a microphysiological model of ischaemic stroke that recapitulates the function of the blood-brain barrier as well as interactions between therapeutic stem cells and host cells (human brain microvascular endothelial cells, pericytes, astrocytes, microglia and neurons). We used the model to track the infiltration of a number of candidate stem cells and to characterize the expression levels of genes associated with post-stroke pathologies. We observed that each type of stem cell showed unique neurorestorative effects, primarily by supporting endogenous recovery rather than through direct cell replacement, and that the recovery of synaptic activities is correlated with the recovery of the structural and functional integrity of the neurovascular unit rather than with the regeneration of neurons.
Collapse
Affiliation(s)
- Zhonglin Lyu
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jon Park
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kwang-Min Kim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hye-Jin Jin
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Haodi Wu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jayakumar Rajadas
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deok-Ho Kim
- Departments of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, U.S.A.,Departments of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, U.S.A
| | - Gary K. Steinberg
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Wonjae Lee
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.,Stanford Stroke Center, Stanford University School of Medicine, Stanford, CA 94305, USA.,Correspondence and requests for materials should be addressed to: Corresponding author, Wonjae Lee, or
| |
Collapse
|
36
|
Serpa RO, Ferguson L, Larson C, Bailard J, Cooke S, Greco T, Prins ML. Pathophysiology of Pediatric Traumatic Brain Injury. Front Neurol 2021; 12:696510. [PMID: 34335452 PMCID: PMC8319243 DOI: 10.3389/fneur.2021.696510] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/21/2021] [Indexed: 11/23/2022] Open
Abstract
The national incidence of traumatic brain injury (TBI) exceeds that of any other disease in the pediatric population. In the United States the Centers for Disease Control and Prevention (CDC) reports 697,347 annual TBIs in children ages 0–19 that result in emergency room visits, hospitalization or deaths. There is a bimodal distribution within the pediatric TBI population, with peaks in both toddlers and adolescents. Preclinical TBI research provides evidence for age differences in acute pathophysiology that likely contribute to long-term outcome differences between age groups. This review will examine the timecourse of acute pathophysiological processes during cerebral maturation, including calcium accumulation, glucose metabolism and cerebral blood flow. Consequences of pediatric TBI are complicated by the ongoing maturational changes allowing for substantial plasticity and windows of vulnerabilities. This review will also examine the timecourse of later outcomes after mild, repeat mild and more severe TBI to establish developmental windows of susceptibility and altered maturational trajectories. Research progress for pediatric TBI is critically important to reveal age-associated mechanisms and to determine knowledge gaps for future studies.
Collapse
Affiliation(s)
- Rebecka O Serpa
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lindsay Ferguson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Cooper Larson
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Julie Bailard
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha Cooke
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tiffany Greco
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mayumi L Prins
- Department of Neurosurgery, Brain Injury Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States.,Steve Tisch BrainSPORT Program, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
37
|
Modeling links softening of myelin and spectrin scaffolds of axons after a concussion to increased vulnerability to repeated injuries. Proc Natl Acad Sci U S A 2021; 118:2024961118. [PMID: 34234016 DOI: 10.1073/pnas.2024961118] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Damage to the microtubule lattice, which serves as a rigid cytoskeletal backbone for the axon, is a hallmark mechanical initiator of pathophysiology after concussion. Understanding the mechanical stress transfer from the brain tissue to the axonal cytoskeleton is essential to determine the microtubule lattice's vulnerability to mechanical injury. Here, we develop an ultrastructural model of the axon's cytoskeletal architecture to identify the components involved in the dynamic load transfer during injury. Corroborative in vivo studies were performed using a gyrencephalic swine model of concussion via single and repetitive head rotational acceleration. Computational analysis of the load transfer mechanism demonstrates that the myelin sheath and the actin/spectrin cortex play a significant role in effectively shielding the microtubules from tissue stress. We derive failure maps in the space spanned by tissue stress and stress rate to identify physiological conditions in which the microtubule lattice can rupture. We establish that a softer axonal cortex leads to a higher susceptibility of the microtubules to failure. Immunohistochemical examination of tissue from the swine model of single and repetitive concussion confirms the presence of postinjury spectrin degradation, with more extensive pathology observed following repetitive injury. Because the degradation of myelin and spectrin occurs over weeks following the first injury, we show that softening of the myelin layer and axonal cortex exposes the microtubules to higher stress during repeated incidences of traumatic brain injuries. Our predictions explain how mechanical injury predisposes axons to exacerbated responses to repeated injuries, as observed in vitro and in vivo.
Collapse
|
38
|
Ross DE, Seabaugh JD, Seabaugh JM, Plumley J, Ha J, Burton JA, Vandervaart A, Mischel R, Blount A, Seabaugh D, Shepherd K, Barcelona J, Ochs AL. Patients with chronic mild or moderate traumatic brain injury have abnormal longitudinal brain volume enlargement more than atrophy. JOURNAL OF CONCUSSION 2021. [DOI: 10.1177/20597002211018049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Introduction Many studies have found brain atrophy in patients with traumatic brain injury (TBI), but most of those studies examined patients with moderate or severe TBI. A few recent studies in patients with chronic mild or moderate TBI found abnormally large brain volume. Some of these studies used NeuroQuant®, FDA-cleared software for measuring MRI brain volume. It is not known if the abnormal enlargement occurs before or after injury. The purpose of the current study was to test the hypothesis that it occurs after injury. Methods 55 patients with chronic mild or moderate TBI were compared to NeuroQuant® normal controls ( n > 4000) with respect to MRI brain volume change from before injury (time 0 [t0], estimated volume) to after injury (t1, measured volume). A subset of 36 patients were compared to the normal controls with respect to longitudinal change of brain volume after injury from t1 to t2. Results The patients had abnormally fast increase of brain volume for multiple brain regions, including whole brain, cerebral cortical gray matter, and subcortical regions. Discussion This is the first report of extensive abnormal longitudinal brain volume enlargement in patients with TBI. In particular, the findings suggested that the previously reported findings of cross-sectional brain volume abnormal enlargement were due to longitudinal enlargement after, not before, injury. Abnormal longitudinal enlargement of the posterior cingulate cortex correlated with neuropathic headaches, partially replicating a previously reported finding that was associated with neuroinflammation.
Collapse
Affiliation(s)
- David E Ross
- Virginia Institute of Neuropsychiatry, Midlothian, USA
| | | | | | | | - Junghoon Ha
- Virginia Commonwealth University, School of Medicine, Richmond, USA
| | - Jason A Burton
- Virginia Commonwealth University, School of Medicine, Richmond, USA
| | | | - Ryan Mischel
- Virginia Commonwealth University, School of Medicine, Richmond, USA
| | - Alyson Blount
- Randolph Macon College, Undergraduate Program, Ashland, USA
| | | | - Katherine Shepherd
- Virginia Institute of Neuropsychiatry, Midlothian, USA
- James Madison University, Undergraduate Program, Harrisonburg, USA
| | | | - Alfred L Ochs
- Virginia Institute of Neuropsychiatry, Midlothian, USA
- Virginia Commonwealth University, School of Medicine, Richmond, USA
| | | |
Collapse
|
39
|
Bodnar CN, Watson JB, Higgins EK, Quan N, Bachstetter AD. Inflammatory Regulation of CNS Barriers After Traumatic Brain Injury: A Tale Directed by Interleukin-1. Front Immunol 2021; 12:688254. [PMID: 34093593 PMCID: PMC8176952 DOI: 10.3389/fimmu.2021.688254] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/05/2021] [Indexed: 01/13/2023] Open
Abstract
Several barriers separate the central nervous system (CNS) from the rest of the body. These barriers are essential for regulating the movement of fluid, ions, molecules, and immune cells into and out of the brain parenchyma. Each CNS barrier is unique and highly dynamic. Endothelial cells, epithelial cells, pericytes, astrocytes, and other cellular constituents each have intricate functions that are essential to sustain the brain's health. Along with damaging neurons, a traumatic brain injury (TBI) also directly insults the CNS barrier-forming cells. Disruption to the barriers first occurs by physical damage to the cells, called the primary injury. Subsequently, during the secondary injury cascade, a further array of molecular and biochemical changes occurs at the barriers. These changes are focused on rebuilding and remodeling, as well as movement of immune cells and waste into and out of the brain. Secondary injury cascades further damage the CNS barriers. Inflammation is central to healthy remodeling of CNS barriers. However, inflammation, as a secondary pathology, also plays a role in the chronic disruption of the barriers' functions after TBI. The goal of this paper is to review the different barriers of the brain, including (1) the blood-brain barrier, (2) the blood-cerebrospinal fluid barrier, (3) the meningeal barrier, (4) the blood-retina barrier, and (5) the brain-lesion border. We then detail the changes at these barriers due to both primary and secondary injury following TBI and indicate areas open for future research and discoveries. Finally, we describe the unique function of the pro-inflammatory cytokine interleukin-1 as a central actor in the inflammatory regulation of CNS barrier function and dysfunction after a TBI.
Collapse
Affiliation(s)
- Colleen N. Bodnar
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - James B. Watson
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Emma K. Higgins
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Ning Quan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, FL, United States
| | - Adam D. Bachstetter
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
40
|
Wang R, Chu C, Wei Z, Chen L, Xu J, Liang Y, Janowski M, Stevens RD, Walczak P. Traumatic brain injury does not disrupt costimulatory blockade-induced immunological tolerance to glial-restricted progenitor allografts. J Neuroinflammation 2021; 18:104. [PMID: 33931070 PMCID: PMC8088005 DOI: 10.1186/s12974-021-02152-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 04/09/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cell transplantation-based treatments for neurological disease are promising, yet graft rejection remains a major barrier to successful regenerative therapies. Our group and others have shown that long-lasting tolerance of transplanted stem cells can be achieved in the brain with systemic application of monoclonal antibodies blocking co-stimulation signaling. However, it is unknown if subsequent injury and the blood-brain barrier breach could expose the transplanted cells to systemic immune system spurring fulminant rejection and fatal encephalitis. Therefore, we investigated whether delayed traumatic brain injury (TBI) could trigger graft rejection. METHODS Glial-restricted precursor cells (GRPs) were intracerebroventricularly transplanted in immunocompetent neonatal mice and co-stimulation blockade (CoB) was applied 0, 2, 4, and 6 days post-grafting. Bioluminescence imaging (BLI) was performed to monitor the grafted cell survival. Mice were subjected to TBI 12 weeks post-transplantation. MRI and open-field test were performed to assess the brain damage and behavioral change, respectively. The animals were decapitated at week 16 post-transplantation, and the brains were harvested. The survival and distribution of grafted cells were verified from brain sections. Hematoxylin and eosin staining (HE) was performed to observe TBI-induced brain legion, and neuroinflammation was evaluated immunohistochemically. RESULTS BLI showed that grafted GRPs were rejected within 4 weeks after transplantation without CoB, while CoB administration resulted in long-term survival of allografts. BLI signal had a steep rise following TBI and subsequently declined but remained higher than the preinjury level. Open-field test showed TBI-induced anxiety for all animals but neither CoB nor GRP transplantation intensified the symptom. HE and MRI demonstrated a reduction in TBI-induced lesion volume in GRP-transplanted mice compared with non-transplanted mice. Brain sections further validated the survival of grafted GRPs and showed more GRPs surrounding the injured tissue. Furthermore, the brains of post-TBI shiverer mice had increased activation of microglia and astrocytes compared to post-TBI wildtype mice, but infiltration of CD45+ leukocytes remained low. CONCLUSIONS CoB induces sustained immunological tolerance towards allografted cerebral GRPs which is not disrupted following TBI, and unexpectedly TBI may enhance GRPs engraftment and contribute to post-injury brain tissue repair.
Collapse
Affiliation(s)
- Rui Wang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,Departments of Anesthesiology and Critical Care Medicine, Neurology, Neurosurgery, Johns Hopkins University, Baltimore, MD, 21287, USA.,Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, 110006, Liaoning, China
| | - Chengyan Chu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA
| | - Zhiliang Wei
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Lin Chen
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Jiadi Xu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Yajie Liang
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA
| | - Miroslaw Janowski
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA
| | - Robert D Stevens
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, 21205, USA.,Departments of Anesthesiology and Critical Care Medicine, Neurology, Neurosurgery, Johns Hopkins University, Baltimore, MD, 21287, USA.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institution, Baltimore, MD, 21205, USA
| | - Piotr Walczak
- Diagnostic Radiology and Nuclear Medicine, University of Maryland Baltimore, 670 W. Baltimore St., HSF III rm 1176, Baltimore, MD, 21201, USA.
| |
Collapse
|
41
|
Corne R, Besson V, Ait Si Slimane S, Coutan M, Palhas MLC, Shen FX, Marchand-Leroux C, Ogier M, Mongeau R. Insulin-like Growth Factors may be Markers of both Traumatic Brain Injury and Fear-Related Stress. Neuroscience 2021; 466:205-221. [PMID: 33895341 DOI: 10.1016/j.neuroscience.2021.04.013] [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: 06/18/2020] [Revised: 04/08/2021] [Accepted: 04/15/2021] [Indexed: 10/21/2022]
Abstract
Insulin-like growth factors (IGF) are potent neurotrophic and neurorepair factors that were recently proposed as biomarkers of traumatic brain injury (TBI) and associated psychiatric comorbidities, in particular post-traumatic stress disorder (PSTD). We tested the hypothesis that the IGF system is differentially deregulated in the acute and early chronic stages of TBI, and under acute stress. Plasma and brain IGF1 and IGF2 levels were evaluated in mice 3 weeks and 3 days after a controlled cortical impact (CCI)-induced mild-to-moderate TBI. The effects of conditioned fear on IGF levels and its interaction with TBI (TBI followed, 3 weeks later, by fear-inducing procedures) were also evaluated. In the plasma, IGF1 decreased 3 weeks post-TBI only (-9%), whereas IGF2 remained unaffected. In the brain, IGF1 increased only in the cortex and hippocampus at 3 weeks post-TBI (up to +650%). At 3 days, surpringly, this increase was more diffuse and more important in sham (craniotomized) animals. Additionally, IGF2 immunostaining in brain ventricles was reorganized in TBI animals at both post-TBI stages. Conditioned fear exposure did not influence the effects of early chronic TBI on plasma IGF1 levels, but reduced plasma IGF2 (-6%) levels. It also dampened the effects of TBI on brain IGF systems, but brain IGF1 level and IGF2 tissue distribution remained statistically different from controls under these conditions. In co-exposed animals, DNA methylation increased at the hippocampal Igf1 gene promoter. These results show that blood IGF1 and IGF2 are most reduced in the early chronic phase of TBI and after exposure to a stressful event, and that the brain IGF system is up-regulated after TBI, and more so in the acute phase.
Collapse
Affiliation(s)
- Rémi Corne
- EA4475 Pharmacologie de la Circulation Cérébrale, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Valérie Besson
- EA4475 Pharmacologie de la Circulation Cérébrale, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France; UMR_S1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Sofiane Ait Si Slimane
- EA4475 Pharmacologie de la Circulation Cérébrale, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Mathieu Coutan
- Institut de Recherche Biomédicale de Armées, 1 place du Général Valérie André, 91223 Brétigny sur Orge Cedex, France
| | - Marta L C Palhas
- EA4475 Pharmacologie de la Circulation Cérébrale, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Fang Xue Shen
- EA4475 Pharmacologie de la Circulation Cérébrale, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Catherine Marchand-Leroux
- EA4475 Pharmacologie de la Circulation Cérébrale, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France; UMR_S1144 Optimisation Thérapeutique en Neuropsychopharmacologie, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France
| | - Michaël Ogier
- Institut de Recherche Biomédicale de Armées, 1 place du Général Valérie André, 91223 Brétigny sur Orge Cedex, France
| | - Raymond Mongeau
- EA4475 Pharmacologie de la Circulation Cérébrale, Université Paris Descartes, 4 avenue de l'Observatoire, 75006 Paris, France; CNRS ERL 3649 T3S-1124 - UMR-S 1124 - Addictions, Pharmacology and Therapy, Université Paris Descartes, 45, rue des Saint-Pères, 75006 Paris, France.
| |
Collapse
|
42
|
Sharma R, Zamani A, Dill LK, Sun M, Chu E, Robinson MJ, O'Brien TJ, Shultz SR, Semple BD. A systemic immune challenge to model hospital-acquired infections independently regulates immune responses after pediatric traumatic brain injury. J Neuroinflammation 2021; 18:72. [PMID: 33731173 PMCID: PMC7968166 DOI: 10.1186/s12974-021-02114-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 02/16/2021] [Indexed: 11/10/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a major cause of disability in young children, yet the factors contributing to poor outcomes in this population are not well understood. TBI patients are highly susceptible to nosocomial infections, which are mostly acquired within the first week of hospitalization, and such infections may modify TBI pathobiology and recovery. In this study, we hypothesized that a peripheral immune challenge such as lipopolysaccharide (LPS)—mimicking a hospital-acquired infection—would worsen outcomes after experimental pediatric TBI, by perpetuating the inflammatory immune response. Methods Three-week-old male mice received either a moderate controlled cortical impact or sham surgery, followed by a single LPS dose (1 mg/kg i.p.) or vehicle (0.9% saline) at 4 days post-surgery, then analysis at 5 or 8 days post-injury (i.e., 1 or 4 days post-LPS). Results LPS-treated mice exhibited a time-dependent reduction in general activity and social investigation, and increased anxiety, alongside substantial body weight loss, indicating transient sickness behaviors. Spleen-to-body weight ratios were also increased in LPS-treated mice, indicative of persistent activation of adaptive immunity at 4 days post-LPS. TBI + LPS mice showed an impaired trajectory of weight gain post-LPS, reflecting a synergistic effect of TBI and the LPS-induced immune challenge. Flow cytometry analysis demonstrated innate immune cell activation in blood, brain, and spleen post-LPS; however, this was not potentiated by TBI. Cytokine protein levels in serum, and gene expression levels in the brain, were altered in response to LPS but not TBI across the time course. Immunofluorescence analysis of brain sections revealed increased glia reactivity due to injury, but no additive effect of LPS was observed. Conclusions Together, we found that a transient, infection-like systemic challenge had widespread effects on the brain and immune system, but these were not synergistic with prior TBI in pediatric mice. These findings provide novel insight into the potential influence of a secondary immune challenge to the injured pediatric brain, with future studies needed to elucidate the chronic effects of this two-hit insult. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02114-1.
Collapse
Affiliation(s)
- Rishabh Sharma
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Larissa K Dill
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Erskine Chu
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Marcus J Robinson
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia.,Department of Neurology, Alfred Health, Prahran, VIC, Australia.,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne, VIC, 3004, Australia. .,Department of Neurology, Alfred Health, Prahran, VIC, Australia. .,Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
43
|
Ved R, Sharouf F, Harari B, Muzaffar M, Manivannan S, Ormonde C, Gray WP, Zaben M. Disulfide HMGB1 acts via TLR2/4 receptors to reduce the numbers of oligodendrocyte progenitor cells after traumatic injury in vitro. Sci Rep 2021; 11:6181. [PMID: 33731757 PMCID: PMC7971069 DOI: 10.1038/s41598-021-84932-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 02/05/2021] [Indexed: 01/31/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with poor clinical outcomes; autopsy studies of TBI victims demonstrate significant oligodendrocyte progenitor cell (OPC) death post TBI; an observation, which may explain the lack of meaningful repair of injured axons. Whilst high-mobility group box-1 (HMGB1) and its key receptors TLR2/4 are identified as key initiators of neuroinflammation post-TBI, they have been identified as attractive targets for development of novel therapeutic approaches to improve post-TBI clinical outcomes. In this report we establish unequivocal evidence that HMGB1 released in vitro impairs OPC response to mechanical injury; an effect that is pharmacologically reversible. We show that needle scratch injury hyper-acutely induced microglial HMGB1 nucleus-to-cytoplasm translocation and subsequent release into culture medium. Application of injury-conditioned media resulted in significant decreases in OPC number through anti-proliferative effects. This effect was reversed by co-treatment with the TLR2/4 receptor antagonist BoxA. Furthermore, whilst injury conditioned medium drove OPCs towards an activated reactive morphology, this was also abolished after BoxA co-treatment. We conclude that HMGB1, through TLR2/4 dependant mechanisms, may be detrimental to OPC proliferation following injury in vitro, negatively affecting the potential for restoring a mature oligodendrocyte population, and subsequent axonal remyelination. Further study is required to assess how HMGB1-TLR signalling influences OPC maturation and myelination capacity.
Collapse
Affiliation(s)
- R Ved
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK
| | - F Sharouf
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK
| | - B Harari
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK
| | - M Muzaffar
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK
| | - S Manivannan
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK
| | - C Ormonde
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK
| | - W P Gray
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK
- Division of Psychological Medicine and Clinical Neurosciences (DPMCN), School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
| | - M Zaben
- Neuroscience and Mental Health Research Institute, Haydn Ellis Building, Cathays, Cardiff, CF24 4HQ, UK.
- Division of Psychological Medicine and Clinical Neurosciences (DPMCN), School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
44
|
Microglia and Neuroinflammation: What Place for P2RY12? Int J Mol Sci 2021; 22:ijms22041636. [PMID: 33561958 PMCID: PMC7915979 DOI: 10.3390/ijms22041636] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/18/2022] Open
Abstract
Microglia are immune brain cells involved in neuroinflammation. They express a lot of proteins on their surface such as receptors that can be activated by mediators released in the microglial environment. Among these receptors, purinergic receptor expression could be modified depending on the activation status of microglia. In this review, we focus on P2Y receptors and more specifically on P2RY12 that is involved in microglial motility and migration, the first step of neuroinflammation process. We describe the purinergic receptor families, P2RY12 structure, expression and physiological functions. The pharmacological and genetic tools for studying this receptor are detailed thereafter. Last but not least, we report the contribution of microglial P2RY12 to neuroinflammation in acute and chronic brain pathologies in order to better understand P2RY12 microglial role.
Collapse
|
45
|
Jia Y, Wang G, Ye Y, Kang E, Chen H, Guo Z, He X. Niche Cells Crosstalk In Neuroinflammation After Traumatic Brain Injury. Int J Biol Sci 2021; 17:368-378. [PMID: 33390856 PMCID: PMC7757042 DOI: 10.7150/ijbs.52169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Traumatic brain injury (TBI) is recognized as the disease with high morbidity and disability around world in spite of the work ongoing in neural protection. Due to heterogeneity among the patients, it's still hard to acquire satisfying achievements in clinic. Neuroinflammation, which exists since primary injury occurs, with elusive duality, appear to be of significance from recovery of injury to neurogenesis. In recent years, studied have revealed that communication in neurogenic niche is more than “cell to cell” communication, and study on NSCs represent it as central role in the progress of neural regeneration. Hence, the neuroinflammation-affecting crosstalk after TBI, and clarifying definitive role of NSCs in the course of regeneration is a promising subject for researchers, for its great potential in overcoming the frustrating status quo in clinic, promoting welfare of TBI patient.
Collapse
Affiliation(s)
- Yibin Jia
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Guanyi Wang
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Yuqing Ye
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China.,Department of Neurosurgery, PLA 163rd Hospital (Second Affiliated Hospital of Hunan Normal University), Changsha 410000, China
| | - Enming Kang
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Huijun Chen
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| | - Zishuo Guo
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, China
| | - Xiaosheng He
- Department of Neurosurgery, Xijing Hospital, Airforce Military Medical University (Fourth Military Medical University), Xi'an 710032, China
| |
Collapse
|
46
|
Gagne C, Piot A, Brake WG. Depression, Estrogens, and Neuroinflammation: A Preclinical Review of Ketamine Treatment for Mood Disorders in Women. Front Psychiatry 2021; 12:797577. [PMID: 35115970 PMCID: PMC8804176 DOI: 10.3389/fpsyt.2021.797577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/24/2021] [Indexed: 12/11/2022] Open
Abstract
Ketamine has been shown to acutely and rapidly ameliorate depression symptoms and suicidality. Given that women suffer from major depression at twice the rate of men, it is important to understand how ketamine works in the female brain. This review explores three themes. First, it examines our current understanding of the etiology of depression in women. Second, it examines preclinical research on ketamine's antidepressant effects at a neurobiological level as well as how ovarian hormones present a unique challenge in interpreting these findings. Lastly, the neuroinflammatory hypothesis of depression is highlighted to help better understand how ovarian hormones might interact with ketamine in the female brain.
Collapse
Affiliation(s)
- Collin Gagne
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| | - Alexandre Piot
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| | - Wayne G Brake
- Department of Psychology, Centre for Studies in Behavioural Neurobiology Concordia University, Montreal, QC, Canada
| |
Collapse
|
47
|
Montivero AJ, Ghersi MS, Catalán-Figueroa J, Formica ML, Camacho N, Culasso AF, Hereñú CB, Palma SD, Pérez MF. Beyond Acute Traumatic Brain Injury: Molecular Implications of Associated Neuroinflammation in Higher-Order Cognitive Processes. PSYCHIATRY AND NEUROSCIENCE UPDATE 2021:237-259. [DOI: 10.1007/978-3-030-61721-9_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
48
|
Martin B, Seguin J, Annereau M, Fleury T, Lai-Kuen R, Neri G, Lam A, Bally M, Mignet N, Corvis Y. Preparation of parenteral nanocrystal suspensions of etoposide from the excipient free dry state of the drug to enhance in vivo antitumoral properties. Sci Rep 2020; 10:18059. [PMID: 33093456 PMCID: PMC7581827 DOI: 10.1038/s41598-020-74809-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Nanoparticle technology in cancer chemotherapy is a promising approach to enhance active ingredient pharmacology and pharmacodynamics. Indeed, drug nanoparticles display various assets such as extended blood lifespan, high drug loading and reduced cytotoxicity leading to better drug compliance. In this context, organic nanocrystal suspensions for pharmaceutical use have been developed in the past ten years. Nanocrystals offer new possibilities by combining the nanoformulation features with the properties of solid dispersed therapeutic ingredients including (i) high loading of the active ingredient, (ii) its bioavailability improvement, and (iii) reduced drug systemic cytotoxicity. However, surprisingly, no antitumoral drug has been marketed as a nanocrystal suspension until now. Etoposide, which is largely used as an anti-cancerous agent against testicular, ovarian, small cell lung, colon and breast cancer in its liquid dosage form, has been selected to develop injectable nanocrystal suspensions designed to be transferred to the clinic. The aim of the present work is to provide optimized formulations for nanostructured etoposide solutions and validate by means of in vitro and in vivo evaluations the efficiency of this multiphase system. Indeed, the etoposide formulated as a nanosuspension by a bottom-up approach showed higher blood life span, reduced tumor growth and higher tolerance in a murine carcinoma cancer model. The results obtained are promising for future clinical evaluation of these etoposide nanosuspensions.
Collapse
Affiliation(s)
- Brice Martin
- Université de Paris, CNRS, Inserm, UTCBS, Chemical and Biological Technologies for Health Group (utcbs.cnrs.fr), Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006, Paris, France.,Department of Neurological Surgery, Weill Medical College of Cornell University, New York, NY, USA
| | - Johanne Seguin
- Université de Paris, CNRS, Inserm, UTCBS, Chemical and Biological Technologies for Health Group (utcbs.cnrs.fr), Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Maxime Annereau
- Gustave Roussy, 114 rue Edouard Vaillant, 94800, PharmacyVillejuif, France
| | - Thomas Fleury
- Gustave Roussy, 114 rue Edouard Vaillant, 94800, PharmacyVillejuif, France
| | - René Lai-Kuen
- Université de Paris, CNRS, Inserm, Cellular and Molecular Imaging Technology Platform, Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Giovanni Neri
- Université de Paris, CNRS, Inserm, UTCBS, Chemical and Biological Technologies for Health Group (utcbs.cnrs.fr), Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Anita Lam
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC, V6T 1Z1, Canada
| | - Marcel Bally
- Department of Experimental Therapeutics, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Nathalie Mignet
- Université de Paris, CNRS, Inserm, UTCBS, Chemical and Biological Technologies for Health Group (utcbs.cnrs.fr), Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Yohann Corvis
- Université de Paris, CNRS, Inserm, UTCBS, Chemical and Biological Technologies for Health Group (utcbs.cnrs.fr), Faculté de Pharmacie, 4 Avenue de l'Observatoire, 75006, Paris, France.
| |
Collapse
|
49
|
Wang H, Chen J, Gao C, Chen W, Chen G, Zhang M, Luo C, Wang T, Chen X, Tao L. TMT-based proteomics analysis to screen potential biomarkers of acute-phase TBI in rats. Life Sci 2020; 264:118631. [PMID: 33131748 DOI: 10.1016/j.lfs.2020.118631] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 10/09/2020] [Accepted: 10/17/2020] [Indexed: 01/19/2023]
Abstract
AIMS Traumatic brain injury (TBI) is a common nervous system injury. However, the detailed mechanisms about functional dysregulation and dignostic biomarkers post-TBI are still unclear. So we aimed to identify potential differentially expressed proteins and genes in TBI for clinical diagnosis and therapeutic purposes. MAIN METHODS Rat TBI model was established by the weight-drop method. First, through TMT-proteomics, we screened for the change in the proteins expression profile acute phase post-TBI. The DAVID and Reactome databases were used to analyze and visualize the dysregulation proteins. Then, using publicly available microarray datasets GSE45997, differentially expressed genes (DGEs) were identified for the 24 h post-TBI stage. Also, the proteomic data were compared with microarray data to analyze the similarity. KEY FINDINGS We found significant proteomics and transcriptomic changes in post-TBI samples. 989, 881, 832, 1057 proteins were quantitated at 1 h, 6 h, 24 h, and 3 d post-injury correspondingly. Concerning proteomics findings, oxygen transport, acute-phase response, and negative regulation of endopeptidase activity were influenced throughout the acute phrase of TBI. Also, pathways related to scavenging of heme from plasma, binding, and uptake of ligands by scavenger receptors were highly enriched in all time-points of TBI samples. SIGNIFICANCE We noticed that the interaction-networks trend to get complicated with more node connections following the progression of TBI. We inferred that Hk-1, PRKAR2A, and MBP could be novel candidate biomarkers related to time-injury in acute-phase TBI. Also, Ceruloplasmin and Complement C3 were found to be important proteins and genes are involved in the TBI.
Collapse
Affiliation(s)
- Haochen Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China.
| | - Jie Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Cheng Gao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Wei Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Guang Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Mingyang Zhang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Chengliang Luo
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Tao Wang
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China
| | - Xiping Chen
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China.
| | - Luyang Tao
- Department of Forensic Medicine, Medical School of Soochow University, Suzhou 215123, China.
| |
Collapse
|
50
|
Beaver JT, Mills LK, Swieboda D, Lelutiu N, Esser ES, Antao OQ, Scountzou E, Williams DT, Papaioannou N, Littauer EQ, Romanyuk A, Compans RW, Prausnitz MR, Skountzou I. Cutaneous vaccination ameliorates Zika virus-induced neuro-ocular pathology via reduction of anti-ganglioside antibodies. Hum Vaccin Immunother 2020; 16:2072-2091. [PMID: 32758106 PMCID: PMC7553697 DOI: 10.1080/21645515.2020.1775460] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Zika virus (ZIKV) causes moderate to severe neuro-ocular sequelae, with symptoms ranging from conjunctivitis to Guillain-Barré Syndrome (GBS). Despite the international threat ZIKV poses, no licensed vaccine exists. As ZIKV and DENV are closely related, antibodies against one virus have demonstrated the ability to enhance the other. To examine if vaccination can confer robust, long-term protection against ZIKV, preventing neuro-ocular pathology and long-term inflammation in immune-privileged compartments, BALB/c mice received two doses of unadjuvanted inactivated whole ZIKV vaccine (ZVIP) intramuscularly (IM) or cutaneously with dissolving microneedle patches (MNP). MNP immunization induced significantly higher B and T cell responses compared to IM vaccination, resulting in increased antibody titers with greater avidity for ZPIV as well as increased numbers of IFN-γ, TNF-α, IL- and IL-4 secreting T cells. When compared to IM vaccination, antibodies generated by cutaneous vaccination demonstrated greater neutralization activity, increased cross-reactivity with Asian and African lineage ZIKV strains (PRVABC59, FLR, and MR766) and Dengue virus (DENV) serotypes, limited ADE, and lower reactivity to GBS-associated gangliosides. MNP vaccination effectively controlled viremia and inflammation, preventing neuro-ocular pathology. Conversely, IM vaccination exacerbated ocular pathology, resulting in uncontrolled, long-term inflammation. Importantly, neuro-ocular pathology correlated with anti-ganglioside antibodies implicated in demyelination and GBS. This study highlights the importance of longevity studies in ZIKV immunization, and the need of exploring alternative vaccination platforms to improve the quality of vaccine-induced immune responses.
Collapse
Affiliation(s)
- Jacob T Beaver
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Lisa K Mills
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Dominika Swieboda
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Nadia Lelutiu
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Edward S Esser
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Olivia Q Antao
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | | | - Dahnide T Williams
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Nikolaos Papaioannou
- Faculty of Veterinary Medicine, Laboratory of Pathologic Anatomy, Aristotle University of Thessaloniki , Greece
| | - Elizabeth Q Littauer
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Andrey Romanyuk
- Department of Biomedical Engineering, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology , Atlanta, GA, USA
| | - Richard W Compans
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| | - Mark R Prausnitz
- Department of Biomedical Engineering, School of Chemical and Biomolecular Engineering, Georgia Institute of Technology , Atlanta, GA, USA
| | - Ioanna Skountzou
- Department of Microbiology & Immunology, Emory University School of Medicine , Atlanta, GA, USA
| |
Collapse
|