1
|
Alonaizan R, K Alotaibi W, Alsulami A, M Alkhulaifi F, Alomar S. Sex-Differences Influence Depressive-Like Behaviour via Alterations in Microglial Expression of GIF-1, TREM2, and IL-1β in an Acute Lipopolysaccharide-Induced Murine Neuroinflammation Model. Immunol Invest 2025; 54:317-333. [PMID: 39701694 DOI: 10.1080/08820139.2024.2440006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
BACKGROUND Neurodegenerative diseases (NDs) have caused serious health issues worldwide. A growing body of evidence suggests a correlation between neuroinflammation and abnormal microglial activity with ND symptoms. Microglia survey play crucial roles in CNS during health and the injury. It is proposed that sex affects microglial roles during inflammation, resulting in mouse behavioural changes and expression alterations in key markers related to microglia functions. METHODS Male and female C57BL/6 mice were injected with a single dose of LPS (5 mg/kg, i.p.) or saline. After 48 h, an open field test was conducted, followed by brain tissues collection for measuring the expression of IGF-1, IL-1β and TREM2 and Immunohistochemistry (IHC) analysis for NLRP3 level. RESULTS Males displayed greater depressive-like behaviour in the OFT, with lower levels of IGF-1, IL-1β, and NLRP3 and high TREM2 expression. Female mice did not exhibit this behaviour, in contrast to male mice, they exhibited increased IL-1β and NLRP3 expression. DISCUSSION This study revealed that LPS-induced sex-specific changes in genes involved in neuronal cell survival caused behavioural alterations in male mice. Moreover, females had observed inflammatory responses that had no impact on behavioural alterations. Overall, both sexes exhibited sex-specific microglial activation states.
Collapse
Affiliation(s)
- Rasha Alonaizan
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Wafa K Alotaibi
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Asma Alsulami
- Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Fadwa M Alkhulaifi
- Biology Department, College of Science, Imam Abdulrahman bin Faisal University, Dammam, Saudi Arabia
| | - Suliman Alomar
- Doping Research Chair, Zoology Department, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
2
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [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: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
3
|
Afridi R, Bhusal A, Lee SE, Hwang EM, Ryu H, Kim JH, Suk K. A microglial kinase ITK mediating neuroinflammation and behavioral deficits in traumatic brain injury. Mol Cell Neurosci 2025; 132:103994. [PMID: 39864680 DOI: 10.1016/j.mcn.2025.103994] [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: 10/22/2024] [Revised: 01/04/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025] Open
Abstract
Microglia-mediated neuroinflammation has been implicated in the neuropathology of traumatic brain injuries (TBI). Recently, the expression of interleukin-2-inducible T-cell kinase (ITK) has been detected in brain microglia, regulating their inflammatory activities. However, the role of microglial ITK in TBI has not been investigated. In this study, we demonstrate that ITK expression and activation are upregulated in microglia following an injury caused by controlled cortical impact (CCI) - a mouse model of TBI. Pharmacological inhibition of ITK protein or knockdown of microglial ITK gene expression using adeno-associated virus mitigates neuroinflammation and improves neurological outcomes in the CCI model. Additionally, ITK mRNA expression was found to be increased in the brains of patients with chronic traumatic encephalopathy. An ITK inhibitor reduced the activation of inflammatory responses in both human and mouse microglia in vitro. Collectively, these results suggest that microglial ITK plays a pivotal role in neuroinflammation and mediating behavioral deficits following TBI. Thus, targeting the signaling pathway of microglial ITK may exert protective effects by alleviating neuroinflammation associated with TBI.
Collapse
Affiliation(s)
- Ruqayya Afridi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Anup Bhusal
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Seung Eun Lee
- Virus Facility, Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Eun Mi Hwang
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Jong-Heon Kim
- Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea.
| | - Kyoungho Suk
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
4
|
Packer JM, Giammo SG, Wangler LM, Davis AC, Bray CE, Godbout JP. Diffuse Traumatic Brain Injury Induced Stimulator of Interferons (STING) Signaling in Microglia Drives Cortical Neuroinflammation, Neuronal Dysfunction, and Impaired Cognition. RESEARCH SQUARE 2025:rs.3.rs-5960640. [PMID: 40034431 PMCID: PMC11875282 DOI: 10.21203/rs.3.rs-5960640/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Neuropsychiatric complications including depression and cognitive impairment develop, persist, and worsen in the years after traumatic brain injury (TBI), negatively affecting life and lifespan. Inflammatory responses mediated by microglia are associated with the transition from acute to chronic neuroinflammation after TBI. Moreover, type I interferon (IFN-I) signaling is a key mediator of inflammation during this transition. Thus, the purpose of this study was to determine the degree to which a microglia-specific knockout of the stimulator of interferons (STING) influenced TBI-induced neuroinflammation, neuronal dysfunction, and cognitive impairment. Here, microglial inducible STING knockout (CX₃CR1Cre/ERT2 x STING fl/fl ) mice were created and validated (mSTING -/- ). Diffuse brain injury (midline fluid percussion) in male and female mice increased STING expression in microglia, promoted microglial morphological restructuring, and induced robust cortical inflammation and pathology 7 days post injury (dpi). These TBI-associated responses were attenuated in mSTING -/- mice. Increased cortical astrogliosis and rod-shaped microglia induced by TBI were independent of mSTING -/- . 7 dpi, TBI induced 237 differentially expressed genes (DEG) in the cortex of functionally wildtype (STING +/+ ) associated with STING, NF- κB, and Interferon Alpha signaling and 85% were attenuated by mSTING -/- . Components of neuronal injury including reduced NeuN expression, increased cortical lipofuscin, and increased neurofilament light chain in plasma were increased by TBI and dependent on mSTING. TBI-associated cognitive tasks (novel object recognition/location, NOR/NOL) at 7 dpi were dependent on mSTING. Notably, the TBI-induced cognitive deficits in NOR/NOL and increased cortical inflammation 7 dpi were unaffected in global interferon-α/β receptor 1 knockout (IFNAR1) mice. In the final study, the RNA profile of neurons after TBI in STING +/+ and mSTING -/- mice was assessed 7 dpi by single nucleus RNA-sequencing. There was a TBI-dependent suppression of cortical neuronal homeostasis with reductions in CREB signaling, synaptogenesis, and oxytocin signaling and increases in cilium assembly and PTEN signaling. Overall, mSTING -/- prevented 50% of TBI-induced DEGs in cortical neurons. Collectively, ablation of STING in microglia attenuates TBI-induced IFN-dependent responses, cortical inflammation, neuronal dysfunction, neuronal pathology, and cognitive impairment.
Collapse
|
5
|
Downing HC, Glover AB, Gebhardt JE, Thompson KL, Saatman KE. Sex-based differences in the long-term fate of hippocampal neurons born after a traumatic brain injury. Front Behav Neurosci 2025; 19:1523969. [PMID: 39974293 PMCID: PMC11836013 DOI: 10.3389/fnbeh.2025.1523969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/15/2025] [Indexed: 02/21/2025] Open
Abstract
Introduction Moderate-to-severe traumatic brain injury (TBI) results in an early loss of immature hippocampal granule cells and the activation of typically quiescent neural stem cells (NSCs) in the dentate gyrus. Activation of NSCs leads to a robust increase in proliferation and generation of neural progenitor cells (NPCs), supporting restoration of the immature neuron population of over a period of 1-2 weeks. However, it is unclear if neurons born early after injury develop normally, survive long-term and functionally integrate into the hippocampal network. Although adult hippocampal neurogenesis is regulated in a sex-dependent manner, the majority of pre-clinical TBI studies lack the inclusion of both sexes. The goal of this study was to examine sex differences in hippocampal neurogenesis in response to a moderate controlled cortical impact brain injury. Methods In-vivo labeling of NPCs and tracking of their morphological development into a granule cell was achieved using an inducible Cre recombinase driven by the Ascl1 promoter in a CAG-floxStopTom reporter mouse. Ascl1 is a basic helix-loop-helix transcription factor transiently expressed in NPCs and activated NSCs in the dentate gyrus of the adult mammalian brain. To specifically label NPCs born acutely after TBI, tamoxifen was delivered to mice on days 2 and 3 postinjury. Mice survived to 6 weeks after TBI to allow for full neuronal maturation of tdTomato-labeled NPCs. Results At 6 weeks postinjury, numbers of tdTomato-positive granule cells were significantly reduced in the ipsilateral hippocampus of brain-injured mice compared to controls, with a more pronounced decrease in males. Further, posttrauma-born neurons in males, but not females, exhibited impaired dendritic development. Neurons born after injury extended axons which formed synaptic terminals within the CA3 region. Numbers of mossy fiber boutons were significantly decreased in injured males compared to naïve males or to injured females. Potential forms of plasticity were observed in brain-injured females, including increased neurogenesis in the contralateral hippocampus and increased mossy fiber bouton volume. Together these data suggest a neurogenic advantage in females after injury. Discussion This study is the first to report sex differences in posttraumatic hippocampal neurogenesis and to demonstrate modification of synaptic terminals formed by neurons born after TBI.
Collapse
Affiliation(s)
- Hannah C. Downing
- Department of Physiology, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Ashley B. Glover
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
| | - Jessica E. Gebhardt
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| | - Katherine L. Thompson
- Dr. Bing Zhang Department of Statistics, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Kathryn E. Saatman
- Department of Physiology, University of Kentucky, Lexington, KY, United States
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States
- Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
6
|
Garcia JP, Armbruster M, Sommer M, Nunez-Beringer A, Dulla CG. Glutamate uptake is transiently compromised in the perilesional cortex following controlled cortical impact. Cereb Cortex 2025; 35:bhaf031. [PMID: 40007051 DOI: 10.1093/cercor/bhaf031] [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: 08/28/2024] [Revised: 12/03/2024] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Glutamate, the primary excitatory neurotransmitter in the central nervous system (CNS), is regulated by the excitatory amino acid transporters glutamate transporter 1 (GLT-1) and glutamate aspartate transporter (GLAST). Following traumatic brain injury, extracellular glutamate levels increase, contributing to excitotoxicity, circuit dysfunction, and morbidity. Increased neuronal glutamate release and compromised astrocyte-mediated uptake contribute to elevated glutamate, but the mechanistic and spatiotemporal underpinnings of these changes are not well established. Using the controlled cortical impact model of TBI and iGluSnFR glutamate imaging, we quantified extracellular glutamate dynamics after injury. Three days postinjury, glutamate release was increased, and glutamate uptake and GLT-1 expression were reduced. Seven and 14 days postinjury, glutamate dynamics were comparable between sham and controlled cortical impact animals. Changes in peak glutamate response were unique to specific cortical layers and proximity to injury. This was likely driven by increases in glutamate release, which was spatially heterogeneous, rather than reduced uptake, which was spatially uniform. The astrocyte K+ channel, Kir4.1, regulates activity-dependent slowing of glutamate uptake. Surprisingly, Kir4.1 was unchanged after controlled cortical impact and accordingly, activity-dependent slowing of glutamate uptake was unaltered. This dynamic glutamate dysregulation after traumatic brain injury underscores a brief period in which disrupted glutamate uptake may contribute to dysfunction and highlights a potential therapeutic window to restore glutamate homeostasis.
Collapse
Affiliation(s)
- Jacqueline P Garcia
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
- Cellular, Molecular, and Developmental Biology Program, Tufts University School of Medicine, Boston, MA, United States
| | - Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Mary Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Aliana Nunez-Beringer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
7
|
Barnes RC, Guindon J. Exploration of a novel therapeutic option: Use of a β 2 agonist to prevent neuropathic pain development secondary to spinal cord injury in a mouse model. J Pharmacol Exp Ther 2025; 392:100038. [PMID: 40023590 DOI: 10.1016/j.jpet.2024.100038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/03/2024] [Indexed: 03/04/2025] Open
Affiliation(s)
- Robert C Barnes
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas
| | - Josée Guindon
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, Texas.
| |
Collapse
|
8
|
Alves M, de Diego-Garcia L, Vegliante G, Moreno O, Gil B, Ramos-Cabrer P, Mitra M, Martin AF, Menéndez-Méndez A, Wang Y, Strogulski NR, Sun MJ, Melia C, Conte G, Plaza-García S, Khalin I, Teng X, Plesnila N, Klebl B, Dinkel K, Hamacher M, Bhattacharya A, Ceusters M, Palmer J, Loane DJ, Llop J, Henshall DC, Engel T. P2X7R antagonism suppresses long-lasting brain hyperexcitability following traumatic brain injury in mice. Theranostics 2025; 15:1399-1419. [PMID: 39886340 PMCID: PMC11780721 DOI: 10.7150/thno.97254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/01/2024] [Indexed: 02/01/2025] Open
Abstract
Purpose: Post-traumatic epilepsy (PTE) is one of the most common life-quality reducing consequences of traumatic brain injury (TBI). However, to date there are no pharmacological approaches to predict or to prevent the development of PTE. The P2X7 receptor (P2X7R) is a cationic ATP-dependent membrane channel that is expressed throughout the brain. While increasing evidence suggests a role for the P2X7R during seizures and epilepsy, it is unclear if changes in P2X7R expression can predict TBI-induced epilepsy development, and whether P2X7R antagonism can protect against long-lasting brain hyperexcitability caused by TBI. Methods: TBI was induced in adult male mice using the controlled cortical impact model (CCI). To test the anti-epileptogenic effects of P2X7R antagonism, mice were treated with brain-penetrant P2X7R antagonists JNJ-54175446 (30 mg/kg) or AFC-5128 (30 mg/kg) for 7 days post-CCI. The cell-type specific effects of P2X7Rs on TBI-induced hyperexcitability were analyzed in mice lacking exon 2 of the P2rx7 gene selectively in microglia (P2rx7:Cx3cr1-Cre). Static positron emission tomography (PET) via an intravenous injection of the P2X7R radioligand 18F-JNJ-64413739 and magnetic resonance imaging (MRI) were conducted twice during the first- and third-week post-injury. Results: Following TBI, while there were no obvious changes in P2X7R protein levels in the ipsilateral hippocampus post-injury, there was a delayed increase in P2X7R protein levels in the ipsilateral cortex at 3 months post-injury. Treatment with P2X7R antagonists shortly after TBI reduced long-lasting brain hyperexcitability, reduced cortical contusion volume, and normalized injury-induced hyperactivity to control sham-levels at 3 weeks post-TBI. Notably, mice lacking P2rx7 in microglia had an increased seizure threshold after TBI, suggesting that P2X7R contributed to brain hyperexcitability via its effects on microglia. Finally, P2X7R radioligand uptake after TBI correlated with seizure threshold at 3 weeks post-injury. Conclusions: Our results demonstrate the antiepileptogenic potential of P2X7R antagonism to prevent TBI-induced epilepsy and indicate that P2X7R-based PET imaging may be a useful diagnostic tool to identify people at risk of developing PTE.
Collapse
MESH Headings
- Animals
- Brain Injuries, Traumatic/drug therapy
- Brain Injuries, Traumatic/complications
- Brain Injuries, Traumatic/metabolism
- Brain Injuries, Traumatic/physiopathology
- Receptors, Purinergic P2X7/metabolism
- Receptors, Purinergic P2X7/genetics
- Mice
- Purinergic P2X Receptor Antagonists/pharmacology
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
- Brain/metabolism
- Brain/diagnostic imaging
- Brain/drug effects
- Epilepsy, Post-Traumatic/drug therapy
- Epilepsy, Post-Traumatic/metabolism
- Mice, Knockout
- Hippocampus/metabolism
- Hippocampus/drug effects
Collapse
Affiliation(s)
- Mariana Alves
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Laura de Diego-Garcia
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
- Department of Optometry, Faculty of Optics and Optometry, Universidad Complutense de Madrid, Avda. Arcos de Jalon 118, 28040 Madrid, Spain
| | - Gloria Vegliante
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Oscar Moreno
- CIC biomaGUNE, Basque research and Technology Alliance (BRTA), P° Miramon 182, 20014 San Sebastian, Gipuzkoa, Spain
| | - Beatriz Gil
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Pedro Ramos-Cabrer
- CIC biomaGUNE, Basque research and Technology Alliance (BRTA), P° Miramon 182, 20014 San Sebastian, Gipuzkoa, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Meghma Mitra
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Ana Fernandez Martin
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Aida Menéndez-Méndez
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
- Department of Medicine, Faculty of Biomedical Sciences and Health, Universidad Europea de Madrid, C. Tajo, s/n, 28670 Villaviciosa de Odón, Madrid, Spain
| | - Yitao Wang
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Nathan Ryzewski Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Meng-Juan Sun
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Ciara Melia
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Giorgia Conte
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
| | - Sandra Plaza-García
- CIC biomaGUNE, Basque research and Technology Alliance (BRTA), P° Miramon 182, 20014 San Sebastian, Gipuzkoa, Spain
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Xinchen Teng
- College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), LMU University Hospital, Ludwig-Maximilians-University (LMU) Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Bert Klebl
- KHAN Technology Transfer Fund I GmbH & Co. KG, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Klaus Dinkel
- Lead Discovery Center GmbH, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | - Michael Hamacher
- Affectis Pharmaceuticals AG, Otto-Hahn-Straße 15, 44227 Dortmund, Germany
| | | | - Marc Ceusters
- Janssen Pharmaceutica NV, Beerse, Belgium
- The Marc Ceusters Company, BV, Diest, Belgium
| | - James Palmer
- Janssen Research and Development LLC, San Diego, California, USA
| | - David J. Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Jordi Llop
- CIC biomaGUNE, Basque research and Technology Alliance (BRTA), P° Miramon 182, 20014 San Sebastian, Gipuzkoa, Spain
| | - David C. Henshall
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
- FutureNeuro Research Ireland Centre for Translational Brain Science, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| | - Tobias Engel
- Department of Physiology & Medical Physics, RCSI University of Medicine & Health Sciences, Dublin D02 YN77, Ireland
- FutureNeuro Research Ireland Centre for Translational Brain Science, RCSI University of Medicine and Health Sciences, Dublin D02 YN77, Ireland
| |
Collapse
|
9
|
Henry RJ, Loane DJ. Unraveling the complexity of microglial responses in traumatic brain and spinal cord injury. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:113-132. [PMID: 40148040 DOI: 10.1016/b978-0-443-19102-2.00015-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Microglia, the resident innate immune cells of the central nervous system (CNS), play an important role in neuroimmune signaling, neuroprotection, and neuroinflammation. In the healthy CNS, microglia adopt a surveillant and antiinflammatory phenotype characterized by a ramified scanning morphology that maintains CNS homeostasis. In response to acquired insults, such as traumatic brain injury (TBI) or spinal cord injury (SCI), microglia undergo a dramatic morphologic and functional switch to that of a reactive state. This microglial switch is initially protective and supports the return of the injured tissue to a physiologic homeostatic state. However, there is now a significant body of evidence that both TBI and SCI can result in a chronic state of microglial activation, which contributes to neurodegeneration and impairments in long-term neurologic outcomes in humans and animal models. In this review, we discuss the complex role of microglia in the pathophysiology of TBI and SCI, and recent advancements in knowledge of microglial phenotypic states in the injured CNS. Furthermore, we highlight novel therapeutic strategies targeting chronic microglial responses in experimental models and discuss how they may ultimately be translated to the clinic for human brain and SCI.
Collapse
Affiliation(s)
- Rebecca J Henry
- Department of Pharmacology, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland.
| | - David J Loane
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
10
|
Angolano C, Hansen E, Ajjawi H, Nowlin P, Zhang Y, Thunemann N, Ferran C, Todd N. Characterization of focused ultrasound blood-brain barrier disruption effect on inflammation as a function of treatment parameters. Biomed Pharmacother 2025; 182:117762. [PMID: 39719739 PMCID: PMC11803570 DOI: 10.1016/j.biopha.2024.117762] [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: 09/10/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/26/2024] Open
Abstract
The technology of focused ultrasound-mediated disruption of the blood-brain barrier (FUS-BBB opening) has now been used in over 20 Phase 1 clinical trials to validate the safety and feasibility of BBB opening for drug delivery in patients with brain tumors and neurodegenerative diseases. The primary treatment parameters, FUS intensity and microbubble dose, are chosen to balance sufficient BBB disruption to achieve drug delivery against potential acute vessel damage leading to microhemorrhage. However, other safety considerations due to second order effects caused by BBB disruption, such as inflammation and alteration of neurovascular function, are only beginning to be understood. This study builds on previous work that has investigated the inflammatory response following FUS-BBB opening. In this study, we characterize the effect of FUS intensity, microbubble dose and single vs multiple treatments on the extent of BBB disruption, observed level of microhemorrhage, and degree of inflammatory response at acute post-treatment time points in the wild-type mouse brain. Results show that upregulation of pro-inflammatory markers is primarily driven by microbubble dose, with peak effects seen at 24 hours post-treatment. We additionally saw significantly elevated levels of cytokine and chemokine markers in female vs male mice, despite no sex differences in level of BBB disruption or microglia activation. Multiple treatments did not result in increased levels of pro-inflammatory markers compared to single treatment baseline. However, we did see an interesting elevation of the anti-inflammatory molecule eNOS after multiple treatments, indicating active mechanisms were at work to restore homeostasis in the brain environment.
Collapse
Affiliation(s)
- Cleide Angolano
- Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Emily Hansen
- Harvard University, Cambridge, MA, United States
| | - Hala Ajjawi
- Harvard University, Cambridge, MA, United States
| | - Paige Nowlin
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Yongzhi Zhang
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Natalie Thunemann
- Department of Biomedical Engineering, Boston University, Boston, MA, United States
| | - Christiane Ferran
- Division of Vascular and Endovascular Surgery, Center for Vascular Biology Research, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States; Division of Nephrology and the Transplant Institute, Department of Nephrology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Nick Todd
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
11
|
Rigo YR, Benvenutti R, Portela LV, Strogulski NR. Neurogenic potential of NG2 in neurotrauma: a systematic review. Neural Regen Res 2024; 19:2673-2683. [PMID: 38595286 PMCID: PMC11168526 DOI: 10.4103/nrr.nrr-d-23-01031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 12/20/2023] [Accepted: 02/07/2024] [Indexed: 04/11/2024] Open
Abstract
Regenerative approaches towards neuronal loss following traumatic brain or spinal cord injury have long been considered a dogma in neuroscience and remain a cutting-edge area of research. This is reflected in a large disparity between the number of studies investigating primary and secondary injury as therapeutic targets in spinal cord and traumatic brain injuries. Significant advances in biotechnology may have the potential to reshape the current state-of-the-art and bring focus to primary injury neurotrauma research. Recent studies using neural-glial factor/antigen 2 (NG2) cells indicate that they may differentiate into neurons even in the developed brain. As these cells show great potential to play a regenerative role, studies have been conducted to test various manipulations in neurotrauma models aimed at eliciting a neurogenic response from them. In the present study, we systematically reviewed the experimental protocols and findings described in the scientific literature, which were peer-reviewed original research articles (1) describing preclinical experimental studies, (2) investigating NG2 cells, (3) associated with neurogenesis and neurotrauma, and (4) in vitro and/or in vivo, available in PubMed/MEDLINE, Web of Science or SCOPUS, from 1998 to 2022. Here, we have reviewed a total of 1504 papers, and summarized findings that ultimately suggest that NG2 cells possess an inducible neurogenic potential in animal models and in vitro. We also discriminate findings of NG2 neurogenesis promoted by different pharmacological and genetic approaches over functional and biochemical outcomes of traumatic brain injury and spinal cord injury models, and provide mounting evidence for the potential benefits of manipulated NG2 cell ex vivo transplantation in primary injury treatment. These findings indicate the feasibility of NG2 cell neurogenesis strategies and add new players in the development of therapeutic alternatives for neurotrauma.
Collapse
Affiliation(s)
- Yuri R. Rigo
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Radharani Benvenutti
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Luis V. Portela
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Nathan R. Strogulski
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, University of Dublin, Dublin, Ireland
| |
Collapse
|
12
|
Bhat AR, Arya AK, Bhopale VM, Imtiyaz Z, Thom SR. Recombinant human plasma gelsolin suppresses persistent neuroinflammation and restores hippocampal neurogenesis in murine model of decompression sickness. J Neurophysiol 2024; 132:1877-1886. [PMID: 39475490 PMCID: PMC11687844 DOI: 10.1152/jn.00332.2024] [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: 07/29/2024] [Revised: 09/19/2024] [Accepted: 10/23/2024] [Indexed: 12/17/2024] Open
Abstract
Loss of plasma gelsolin (pGSN), a protein that lyses actin filaments, is implicated in the pathology of inflammatory and neurodegenerative diseases. We hypothesized that because pGSN is depleted in a murine model of decompression sickness (DCS), supplementation by administration of human recombinant (rhu-) pGSN would ameliorate inflammatory events. We observed that pGSN levels were persistently decreased in mice for at least 12 days postexposure to 790 kPa of air for 2 h. This decline was associated with elevated levels of inflammatory microparticles (MPs) in the blood and cervical lymph nodes, which previously were shown to cause neuroinflammation. In addition, these mice exhibited reduced expression of synaptic proteins, impaired neurogenesis and impaired cognitive and motor functions. Rhu-pGSN ameliorated the inflammatory changes and resulted in restored synaptic protein expression, neurogenesis, and neurological function. These findings demonstrate that neuronal dysfunction in our murine model of DCS is mediated by MPs and that rhu-pGSN can ameliorate injury even when administered in a delayed fashion.NEW & NOTEWORTHY A decrease in plasma gelsolin levels and the release of inflammatory microparticles (MPs) occur in response to high pressure followed by decompression, with expression of filamentous (F)-actin leading to persistent neuroinflammation and functional deficits lasting at least 12 days. The infusion of recombinant human plasma gelsolin lyses these MPs in decompressed mice, thereby alleviating particle-associated neuronal dysfunction. Rhu-gelsolin infusion may be beneficial as a prophylactic or treatment for decompression sickness.
Collapse
Affiliation(s)
- Abid R Bhat
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Awadhesh K Arya
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Veena M Bhopale
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Zuha Imtiyaz
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Stephen R Thom
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
13
|
Andrew PM, MacMahon JA, Bernardino PN, Tsai YH, Hobson BA, Porter VA, Huddleston SL, Luo AS, Bruun DA, Saito NH, Harvey DJ, Brooks-Kayal A, Chaudhari AJ, Lein PJ. Shifts in the spatiotemporal profile of inflammatory phenotypes of innate immune cells in the rat brain following acute intoxication with the organophosphate diisopropylfluorophosphate. J Neuroinflammation 2024; 21:285. [PMID: 39497181 PMCID: PMC11533402 DOI: 10.1186/s12974-024-03272-8] [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: 09/01/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024] Open
Abstract
Acute intoxication with cholinesterase inhibiting organophosphates (OP) can produce life-threatening cholinergic crisis and status epilepticus (SE). Survivors often develop long-term neurological consequences, including spontaneous recurrent seizures (SRS) and impaired cognition. Numerous studies implicate OP-induced neuroinflammation as a pathogenic mechanism contributing to these chronic sequelae; however, little is known about the inflammatory phenotype of innate immune cells in the brain following acute OP intoxication. Thus, the aim of this study was to characterize the natural history of microglial and astrocytic inflammatory phenotypes following acute intoxication with the OP, diisopropylfluorophosphate (DFP). Adult male and female Sprague-Dawley rats were administered a single dose of DFP (4 mg/kg, sc) followed by standard medical countermeasures. Within minutes, animals developed benzodiazepine-resistant SE as determined by monitoring seizures using a modified Racine scale. At 1, 3, 7, 14, and 28 d post-exposure (DPE), neuroinflammation was assessed using translocator protein (TSPO) positron emission tomography (PET) and magnetic resonance imaging (MRI). In both sexes, we observed consistently elevated radiotracer uptake across all examined brain regions and time points. A separate group of animals was euthanized at these same time points to collect tissues for immunohistochemical analyses. Colocalization of IBA-1, a marker for microglia, with iNOS or Arg1 was used to identify pro- and anti-inflammatory microglia, respectively; colocalization of GFAP, a marker for astrocytes, with C3 or S100A10, pro- and anti-inflammatory astrocytes, respectively. We observed shifts in the inflammatory profiles of microglia and astrocyte populations during the first month post-intoxication, largely in hyperintense inflammatory lesions in the piriform cortex and amygdala regions. In these areas, iNOS+ proinflammatory microglial cell density peaked at 3 and 7 DPE, while anti-inflammatory Arg1+ microglia cell density peaked at 14 DPE. Pro- and anti-inflammatory astrocytes emerged within 7 DPE, and roughly equal ratios of C3+ pro-inflammatory and S100A10+ anti-inflammatory astrocytes persisted at 28 DPE. In summary, microglia and astrocytes adopted mixed inflammatory phenotypes post-OP intoxication, which evolved over one month post exposure. These activated cell populations were most prominent in the piriform and amygdala areas and were more abundant in males compared to females. The temporal relationship between microglial and astrocytic responses suggests that initial microglial activity may influence delayed, persistent astrocytic responses. Further, our findings identify putative windows for inhibition of OP-induced neuroinflammatory responses in both sexes to evaluate the therapeutic benefit of anti-inflammation in this context.
Collapse
Affiliation(s)
- Peter M Andrew
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Jeremy A MacMahon
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Pedro N Bernardino
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Yi-Hua Tsai
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Brad A Hobson
- Center for Molecular and Genomic Imaging, College of Engineering, University of California, DavisDavis, CA, 95616, USA
| | - Valerie A Porter
- Department of Biomedical Engineering, College of Engineering, University of California, DavisDavis, CA, 95616, USA
| | - Sydney L Huddleston
- Center for Molecular and Genomic Imaging, College of Engineering, University of California, DavisDavis, CA, 95616, USA
| | - Audrey S Luo
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Donald A Bruun
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Naomi H Saito
- Department of Public Health Sciences, Davis, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Danielle J Harvey
- Department of Public Health Sciences, Davis, School of Medicine, University of California, Davis, CA, 95616, USA
| | - Amy Brooks-Kayal
- Department of Neurology, Davis, School of Medicine, University of California, Sacramento, CA, 95817, USA
| | - Abhijit J Chaudhari
- Center for Molecular and Genomic Imaging, College of Engineering, University of California, DavisDavis, CA, 95616, USA
- Department of Radiology, Davis, School of Medicine, University of California, Sacramento, CA, 95817, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, Davis, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
14
|
Wu J, Ren R, Chen T, Su LD, Tang T. Neuroimmune and neuroinflammation response for traumatic brain injury. Brain Res Bull 2024; 217:111066. [PMID: 39241894 DOI: 10.1016/j.brainresbull.2024.111066] [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: 06/15/2024] [Revised: 08/18/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Traumatic brain injury (TBI) is one of the major diseases leading to mortality and disability, causing a serious disease burden on individuals' ordinary lives as well as socioeconomics. In primary injury, neuroimmune and neuroinflammation are both responsible for the TBI. Besides, extensive and sustained injury induced by neuroimmune and neuroinflammation also prolongs the course and worsens prognosis of TBI. Therefore, this review aims to explore the role of neuroimmune, neuroinflammation and factors associated them in TBI as well as the therapies for TBI. Thus, we conducted by searching PubMed, Scopus, and Web of Science databases for articles published between 2010 and 2023. Keywords included "traumatic brain injury," "neuroimmune response," "neuroinflammation," "astrocytes," "microglia," and "NLRP3." Articles were selected based on relevance and quality of evidence. On this basis, we provide the cellular and molecular mechanisms of TBI-induced both neuroimmune and neuroinflammation response, as well as the different factors affecting them, are introduced based on physiology of TBI, which supply a clear overview in TBI-induced chain-reacting, for a better understanding of TBI and to offer more thoughts on the future therapies for TBI.
Collapse
Affiliation(s)
- Junyun Wu
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Reng Ren
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Tao Chen
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China
| | - Li-Da Su
- Neuroscience Care Unit, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| | - Tianchi Tang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
15
|
Culkin MC, Bele P, Georges AP, Santos P, Niziolek G, Kaplan LJ, Smith DH, Pascual JL. Dose-Dependent Tranexamic Acid Blunting of Penumbral Leukocyte Mobilization and Blood-Brain Barrier Permeability Following Traumatic Brain Injury: An In Vivo Murine Study. Neurocrit Care 2024; 41:469-478. [PMID: 38443709 DOI: 10.1007/s12028-024-01952-0] [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: 10/26/2023] [Accepted: 01/29/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Early posttraumatic brain injury (TBI) tranexamic acid (TXA) may reduce blood-brain barrier (BBB) permeability, but it is unclear if this effect is fixed regardless of dose. We hypothesized that post-TBI TXA demonstrates a dose-dependent reduction of in vivo penumbral leukocyte mobilization, BBB microvascular permeability, and enhancement of neuroclinical recovery. METHODS CD1 male mice (n = 40) were randomly assigned to TBI by controlled cortical impact (injury [I]) or sham TBI (S), followed by intravenous bolus of either saline (placebo [P]) or TXA (15, 30, or 60 mg/kg). At 48 h, in vivo pial intravital microscopy visualized live penumbral BBB microvascular leukocytes and albumin leakage. Neuroclinical recovery was assessed by Garcia Neurological Test scores and animal weight changes at 24 h and 48 h after injury. RESULTS I + TXA60 reduced live penumbral leukocyte rolling compared with I + P (p < 0.001) and both lower TXA doses (p = 0.017 vs. I + TXA15, p = 0.012 vs. I + TXA30). Leukocyte adhesion was infrequent and similar across groups. Only I + TXA60 significantly reduced BBB permeability compared with that in the I + P (p = 0.004) group. All TXA doses improved Garcia Test scores relative to I + P at both 24 h and 48 h (p < 0.001 vs. I + P for all at both time points). Mean 24-h body weight loss was greatest in the I + P (- 8.7 ± 1.3%) group and lowest in the I + TXA15 (- 4.4 ± 1.0%, p = 0.051 vs. I + P) group. CONCLUSIONS Only higher TXA dosing definitively abrogates penumbral leukocyte mobilization, preserving BBB integrity post TBI. Some neuroclinical recovery is observed, even with lower TXA dosing. Better outcomes with higher dose TXA after TBI may occur secondary to blunting of leukocyte-mediated penumbral cerebrovascular inflammation.
Collapse
Affiliation(s)
- Matthew C Culkin
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Priyanka Bele
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anastasia P Georges
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Patricia Santos
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Grace Niziolek
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Lewis J Kaplan
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Douglas H Smith
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jose L Pascual
- Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery, Perelman School of Medicine, University of Pennsylvania, SICU Administration Office - 5 Founders Pavilion, 3400 Spruce Street, Philadelphia, PA, 19104, USA.
- Center for Brain Injury and Repair, Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
16
|
Logan-Wesley AL, Gorse KM, Lafrenaye AD. Microglial process convergence onto injured axonal swellings, a human postmortem brain tissue study. Sci Rep 2024; 14:21369. [PMID: 39266604 PMCID: PMC11392954 DOI: 10.1038/s41598-024-71312-7] [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: 07/09/2024] [Accepted: 08/27/2024] [Indexed: 09/14/2024] Open
Abstract
Traumatic brain injury (TBI) affects millions globally, with a majority of TBI cases being classified as mild, in which diffuse pathologies prevail. Two of the pathological hallmarks of TBI are diffuse axonal injury (DAI) and microglial activation. While progress has been made investigating the breadth of TBI-induced axonal injury and microglial changes in rodents, the neuroinflammatory progression and interaction between microglia and injured axons in humans is less well understood. Our group previously investigated microglial process convergence (MPC), in which processes of non-phagocytic microglia directly contact injured proximal axonal swellings, in rats and micropigs acutely following TBI. These studies demonstrated that MPC occurred on injured axons in the micropig, but not in the rat, following diffuse TBI. While it has been shown that microglia co-exist and interact with injured axons in humans post-TBI, the occurrence of MPC has not been quantitatively measured in the human brain. Therefore, in the current study we sought to validate our pig findings in human postmortem tissue. We investigated MPC onto injured axonal swellings and intact myelinated fibers in cases from individuals with confirmed DAI and control human brain tissue using multiplex immunofluorescent histochemistry. We found an increase in MPC onto injured axonal swellings, consistent with our previous findings in micropigs, indicating that MPC is a clinically relevant phenomenon that warrants further investigation.
Collapse
Affiliation(s)
| | - Karen M Gorse
- Virginia Commonwealth University, BOX 980709, Richmond, VA, 23298, USA
| | | |
Collapse
|
17
|
Garcia JP, Armbruster M, Sommer M, Nunez-Beringer A, Dulla CG. Glutamate uptake is transiently compromised in the perilesional cortex following controlled cortical impact. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610143. [PMID: 39257826 PMCID: PMC11383988 DOI: 10.1101/2024.08.28.610143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Glutamate, the primary excitatory neurotransmitter in the CNS, is regulated by the excitatory amino acid transporters (EAATs) GLT-1 and GLAST. Following traumatic brain injury (TBI), extracellular glutamate levels increase, contributing to excitotoxicity, circuit dysfunction, and morbidity. Increased neuronal glutamate release and compromised astrocyte-mediated uptake contribute to elevated glutamate, but the mechanistic and spatiotemporal underpinnings of these changes are not well established. Using the controlled cortical impact (CCI) model of TBI and iGluSnFR glutamate imaging, we quantified extracellular glutamate dynamics after injury. Three days post-injury, glutamate release was increased, and glutamate uptake and GLT-1 expression were reduced. 7- and 14-days post-injury, glutamate dynamics were comparable between sham and CCI animals. Changes in peak glutamate response were unique to specific cortical layers and proximity to injury. This was likely driven by increases in glutamate release, which was spatially heterogenous, rather than reduced uptake, which was spatially uniform. The astrocyte K + channel, Kir4.1, regulates activity-dependent slowing of glutamate uptake. Surprisingly, Kir4.1 was unchanged after CCI and accordingly, activity-dependent slowing of glutamate uptake was unaltered. This dynamic glutamate dysregulation after TBI underscores a brief period in which disrupted glutamate uptake may contribute to dysfunction and highlights a potential therapeutic window to restore glutamate homeostasis.
Collapse
|
18
|
Logan-Wesley AL, Gorse KM, Lafrenaye AD. Microglial process convergence onto injured axonal swellings, a human postmortem brain tissue study. RESEARCH SQUARE 2024:rs.3.rs-4713316. [PMID: 39149456 PMCID: PMC11326398 DOI: 10.21203/rs.3.rs-4713316/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Traumatic brain injury (TBI) affects millions globally, with a majority of TBI cases being classified as mild, in which diffuse pathologies prevail. Two of the pathological hallmarks of TBI are diffuse axonal injury and microglial activation. While progress has been made investigating the breadth of TBI-induced axonal injury and microglial changes in rodents, the neuroinflammatory progression and interaction between microglia and injured axons following brain injury in humans is less well understood. Our group previously investigated microglial process convergence (MPC), in which processes of non-phagocytic microglia directly contact injured proximal axonal segments, in rats and micropigs acutely following TBI. These studies demonstrated that MPC occurred on injured axons in the micropig, but not in the rat, following diffuse TBI. While it has been shown that microglia co-exist and interact with injured axons in humans post-TBI, the occurrence of MPC has not been quantitatively measured in the human brain. Therefore, in the current study we sought to validate our pig findings in human postmortem tissue. We investigated MPC onto injured axonal swellings and intact myelinated fibers in cases from individuals that sustained a TBI and control human brain tissue using multiplex immunofluorescent histochemistry. We found an increase in MPC onto injured axonal swellings, consistent with our previous findings in micropigs, indicating that MPC is a clinically relevant phenomenon that warrants further investigation.
Collapse
|
19
|
Angolano C, Hansen E, Ajjawi H, Nowlin P, Zhang Y, Thunemann N, Ferran C, Todd N. Characterization of focused ultrasound blood-brain barrier disruption effect on inflammation as a function of treatment parameters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.10.602776. [PMID: 39071338 PMCID: PMC11275883 DOI: 10.1101/2024.07.10.602776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
The technology of focused ultrasound-mediated disruption of the blood-brain barrier (FUS- BBB opening) has now been used in over 20 Phase 1 clinical trials to validate the safety and feasibility of BBB opening for drug delivery in patients with brain tumors and neurodegenerative diseases. The primary treatment parameters, FUS intensity and microbubble dose, are chosen to balance sufficient BBB disruption to achieve drug delivery against potential acute vessel damage leading to microhemorrhage. This can largely be achieved based on both empirical results from animal studies and by monitoring the microbubble cavitation signal in real time during the treatment. However, other safety considerations due to second order effects caused by BBB disruption, such as inflammation and alteration of neurovascular function, are not as easily measurable, may take longer to manifest and are only beginning to be understood. This study builds on previous work that has investigated the inflammatory response following FUS-BBB opening. In this study, we characterize the effect of FUS intensity and microbubble dose on the extent of BBB disruption, observed level of microhemorrhage, and degree of inflammatory response at three acute post-treatment time points in the wild-type mouse brain. Additionally, we evaluate differences related to biological sex, presence and degree of the anti- inflammatory response that develops to restore homeostasis in the brain environment, and the impact of multiple FUS-BBB opening treatments on this inflammatory response.
Collapse
|
20
|
Davis CK, Bathula S, Jeong S, Arruri V, Choi J, Subramanian S, Ostrom CM, Vemuganti R. An antioxidant and anti-ER stress combination therapy elevates phosphorylation of α-Syn at serine 129 and alleviates post-TBI PD-like pathology in a sex-specific manner in mice. Exp Neurol 2024; 377:114795. [PMID: 38657855 DOI: 10.1016/j.expneurol.2024.114795] [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: 02/22/2024] [Revised: 04/13/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
Clinical studies have shown that traumatic brain injury (TBI) increases the onset of Parkinson's disease (PD) in later life by >50%. Oxidative stress, endoplasmic reticulum (ER) stress, and inflammation are the major drivers of both TBI and PD pathologies. We presently evaluated if curtailing oxidative stress and ER stress concomitantly using a combination of apocynin and tert-butylhydroquinone and salubrinal during the acute stage after TBI in mice reduces the severity of late-onset PD-like pathology. The effect of multiple low doses of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) on post-TBI neurodegeneration was also evaluated. The combo therapy elevated the level of phosphorylation at serine 129 (pS129) of α-Syn in the pericontusional cortex of male mice at 72 h post-TBI. Motor and cognitive deficits induced by TBI lasted at least 3 months and the combo therapy curtailed these deficits in both sexes. At 3 months post-TBI, male mice given combo therapy exhibited significantly lesser α-Syn aggregates in the SN and higher TH+ cells in the SNpc, compared to vehicle control. However, the aggregate number was not significantly different between groups of female mice. Moreover, TBI-induced loss of TH+ cells was negligible in female mice irrespective of treatment. The MPTP treatment aggravated PD-like pathology in male mice but had a negligible effect on the loss of TH+ cells in female mice. Thus, the present study indicates that mitigation of TBI-induced oxidative stress and ER stress at the acute stage could potentially reduce the risk of post-TBI PD-like pathology at least in male mice, plausibly by elevating pS129-α-Syn level.
Collapse
Affiliation(s)
- Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | | | - Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Vijay Arruri
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Jeongwoo Choi
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Shruti Subramanian
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Carlie M Ostrom
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin, Madison, WI, USA; William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
21
|
Bhat AR, Arya AK, Bhopale VM, Imtiyaz Z, Xu S, Bedir D, Thom SR. Persistent neuroinflammation and functional deficits in a murine model of decompression sickness. J Appl Physiol (1985) 2024; 137:63-73. [PMID: 38660728 PMCID: PMC11389893 DOI: 10.1152/japplphysiol.00097.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 04/11/2024] [Accepted: 04/22/2024] [Indexed: 04/26/2024] Open
Abstract
We hypothesized that early intra-central nervous system (CNS) responses in a murine model of decompression sickness (DCS) would be reflected by changes in the microparticles (MPs) that exit the brain via the glymphatic system, and due to systemic responses the MPs would cause inflammatory changes lasting for many days leading to functional neurological deficits. Elevations on the order of threefold of blood-borne inflammatory MPs, neutrophil activation, glymphatic flow, and neuroinflammation in cerebral cortex and hippocampus were found in mice at 12 days after exposure to 760 kPa of air for 2 h. Mice also exhibited a significant decline in memory and locomotor activity, as assessed by novel object recognition and rotarod testing. Similar inflammatory changes in blood, neuroinflammation, and functional impairments were initiated in naïve mice by injection of filamentous (F-) actin-positive MPs, but not F-actin-negative MPs, obtained from decompressed mice. We conclude that high pressure/decompression stress establishes a systemic inflammatory process that results in prolonged neuroinflammation and functional impairments in the mouse decompression model.NEW & NOTEWORTHY Elevated glymphatic flow due to astrocyte and microglial activation from high-pressure exposure triggers release of microparticles (MPs) to the circulation where neutrophil activation and production of filamentous (F)-actin expressing MPs result in a persistent feed-forward neuroinflammatory cycle and functional deficits lasting for at least 12 days.
Collapse
Affiliation(s)
- Abid R Bhat
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Awadhesh K Arya
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Veena M Bhopale
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Zuha Imtiyaz
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Dilara Bedir
- Department of Undersea and Hyperbaric Medicine, Gulhane Education and Research Hospital, Istanbul, Turkey
| | - Stephen R Thom
- Department of Emergency Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
22
|
Fresquez AM, Hogan JO, Rivera P, Patterson KM, Singer K, Reynolds JM, White C. STIM1-dependent store-operated calcium entry mediates sex differences in macrophage chemotaxis and monocyte recruitment. J Biol Chem 2024; 300:107422. [PMID: 38815866 PMCID: PMC11231831 DOI: 10.1016/j.jbc.2024.107422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 05/09/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024] Open
Abstract
Infiltration of monocyte-derived cells to sites of infection and injury is greater in males than in females, due in part, to increased chemotaxis, the process of directed cell movement toward a chemical signal. The mechanisms governing sexual dimorphism in chemotaxis are not known. We hypothesized a role for the store-operated calcium entry (SOCE) pathway in regulating chemotaxis by modulating leading and trailing edge membrane dynamics. We measured the chemotactic response of bone marrow-derived macrophages migrating toward complement component 5a (C5a). Chemotactic ability was dependent on sex and inflammatory phenotype (M0, M1, and M2), and correlated with SOCE. Notably, females exhibited a significantly lower magnitude of SOCE than males. When we knocked out the SOCE gene, stromal interaction molecule 1 (STIM1), it eliminated SOCE and equalized chemotaxis across both sexes. Analysis of membrane dynamics at the leading and trailing edges showed that STIM1 influences chemotaxis by facilitating retraction of the trailing edge. Using BTP2 to pharmacologically inhibit SOCE mirrored the effects of STIM1 knockout, demonstrating a central role of STIM/Orai-mediated calcium signaling. Importantly, by monitoring the recruitment of adoptively transferred monocytes in an in vivo model of peritonitis, we show that increased infiltration of male monocytes during infection is dependent on STIM1. These data support a model in which STIM1-dependent SOCE is necessary and sufficient for mediating the sex difference in monocyte recruitment and macrophage chemotactic ability by regulating trailing edge dynamics.
Collapse
Affiliation(s)
- Adriana M Fresquez
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - James O Hogan
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Patricia Rivera
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Kristen M Patterson
- Microbiology and Immunology, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Joseph M Reynolds
- Microbiology and Immunology, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA
| | - Carl White
- Physiology & Biophysics, Center for Cancer Cell Biology, Immunology, and Infection, Rosalind Franklin University of Medicine & Science, North Chicago, Illinois, USA.
| |
Collapse
|
23
|
Gu N, Yan J, Tang W, Zhang Z, Wang L, Li Z, Wang Y, Zhu Y, Tang S, Zhong J, Cheng C, Sun X, Huang Z. Prevotella copri transplantation promotes neurorehabilitation in a mouse model of traumatic brain injury. J Neuroinflammation 2024; 21:147. [PMID: 38835057 DOI: 10.1186/s12974-024-03116-5] [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: 01/30/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND The gut microbiota plays a critical role in regulating brain function through the microbiome-gut-brain axis (MGBA). Dysbiosis of the gut microbiota is associated with neurological impairment in Traumatic brain injury (TBI) patients. Our previous study found that TBI results in a decrease in the abundance of Prevotella copri (P. copri). P. copri has been shown to have antioxidant effects in various diseases. Meanwhile, guanosine (GUO) is a metabolite of intestinal microbiota that can alleviate oxidative stress after TBI by activating the PI3K/Akt pathway. In this study, we investigated the effect of P. copri transplantation on TBI and its relationship with GUO-PI3K/Akt pathway. METHODS In this study, a controlled cortical impact (CCI) model was used to induce TBI in adult male C57BL/6J mice. Subsequently, P. copri was transplanted by intragastric gavage for 7 consecutive days. To investigate the effect of the GUO-PI3K/Akt pathway in P. copri transplantation therapy, guanosine (GUO) was administered 2 h after TBI for 7 consecutive days, and PI3K inhibitor (LY294002) was administered 30 min before TBI. Various techniques were used to assess the effects of these interventions, including quantitative PCR, neurological behavior tests, metabolite analysis, ELISA, Western blot analysis, immunofluorescence, Evans blue assays, transmission electron microscopy, FITC-dextran permeability assay, gastrointestinal transit assessment, and 16 S rDNA sequencing. RESULTS P. copri abundance was significantly reduced after TBI. P. copri transplantation alleviated motor and cognitive deficits tested by the NSS, Morris's water maze and open field test. P. copri transplantation attenuated oxidative stress and blood-brain barrier damage and reduced neuronal apoptosis after TBI. In addition, P. copri transplantation resulted in the reshaping of the intestinal flora, improved gastrointestinal motility and intestinal permeability. Metabolomics and ELISA analysis revealed a significant increase in GUO levels in feces, serum and injured brain after P. copri transplantation. Furthermore, the expression of p-PI3K and p-Akt was found to be increased after P. copri transplantation and GUO treatment. Notably, PI3K inhibitor LY294002 treatment attenuated the observed improvements. CONCLUSIONS We demonstrate for the first time that P. copri transplantation can improve GI functions and alter gut microbiota dysbiosis after TBI. Additionally, P. copri transplantation can ameliorate neurological deficits, possibly via the GUO-PI3K/Akt signaling pathway after TBI.
Collapse
Affiliation(s)
- Nina Gu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jin Yan
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Tang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhaosi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lin Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurosurgery, The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, China
| | - Zhao Li
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Emergency Department, Chengdu First People's Hospital, Chengdu, China
| | - Yingwen Wang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yajun Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Shuang Tang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Department of Neurosurgery, Suining Central Hospital, Suining, China
| | - Jianjun Zhong
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Zhijian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
24
|
Li C, Ajmal E, Alok K, Powell K, Wadolowski S, Tambo W, Turpin J, Barthélemy E, Al-Abed Y, LeDoux D. CGRP as a potential mediator for the sexually dimorphic responses to traumatic brain injury. Biol Sex Differ 2024; 15:44. [PMID: 38816868 PMCID: PMC11138127 DOI: 10.1186/s13293-024-00619-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/15/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND The outcomes of traumatic brain injury (TBI) exhibit variance contingent upon biological sex. Although female sex hormones exert neuroprotective effects, the administration of estrogen and progesterone has not yielded conclusive results. Hence, it is conceivable that additional mediators, distinct from female sex hormones, merit consideration due to their potential differential impact on TBI outcomes. Calcitonin gene-related peptide (CGRP) exhibits sexually dimorphic expression and demonstrates neuroprotective effects in acute brain injuries. In this study, we aimed to examine sex-based variations in TBI structural and functional outcomes with respect to CGRP expression. METHODS Male and female Sprague Dawley rats were exposed to controlled cortical impact to induce severe TBI, followed by interventions with and without CGRP inhibition. In the acute phase of TBI, the study centered on elucidating the influence of CGRP on oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) and endothelial nitric oxide synthase (eNOS) signaling in the peri-impact tissue. Subsequently, during the chronic phase of TBI, the investigation expanded to evaluate CGRP expression in relation to lesion volume, microvascular dysfunction, and white matter injury, as well as working and spatial memory, anxiety-like, and depression-like behaviors in subjects of both sexes. RESULTS Female rats exhibited elevated levels of CGRP in the peri-impact brain tissue during both baseline conditions and in the acute and chronic phases of TBI, in comparison to age-matched male counterparts. Enhanced CGRP levels in specific brain sub-regions among female rats correlated with superior structural and functional outcomes following TBI compared to their male counterparts. CGRP inhibition induced heightened oxidative stress and a reduction in the expression of Nrf2 and eNOS in both male and female rats, with the observed alteration being more pronounced in females than in males. CONCLUSIONS This study marks the inaugural identification of CGRP as a downstream mediator contributing to the sexually dimorphic response observed in TBI outcomes.
Collapse
Affiliation(s)
- Chunyan Li
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA.
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA.
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA.
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA.
| | - Erum Ajmal
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Khaled Alok
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Keren Powell
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Steven Wadolowski
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Willians Tambo
- Translational Brain Research Laboratory, The Feinstein Institutes for Medical Research, 350 Community Dr, Manhasset, NY, 11030, USA
- Elmezzi Graduate School of Molecular Medicine at Northwell Health, Manhasset, NY, 11030, USA
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - Justin Turpin
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| | - Ernest Barthélemy
- Division of Neurosurgery, SUNY Downstate College of Medicine, Brooklyn, NY, 11203, USA
| | - Yousef Al-Abed
- Institute of Bioelectronic Medicine, The Feinstein Institutes for Medical Research, Manhasset, NY, 11030, USA
| | - David LeDoux
- Department of Neurosurgery, North Shore University Hospital, Manhasset, NY, 11030, USA
| |
Collapse
|
25
|
Caldarelli M, Rio P, Marrone A, Ocarino F, Chiantore M, Candelli M, Gasbarrini A, Gambassi G, Cianci R. Gut-Brain Axis: Focus on Sex Differences in Neuroinflammation. Int J Mol Sci 2024; 25:5377. [PMID: 38791415 PMCID: PMC11120930 DOI: 10.3390/ijms25105377] [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: 03/30/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
In recent years, there has been a growing interest in the concept of the "gut-brain axis". In addition to well-studied diseases associated with an imbalance in gut microbiota, such as cancer, chronic inflammation, and cardiovascular diseases, research is now exploring the potential role of gut microbial dysbiosis in the onset and development of brain-related diseases. When the function of the intestinal barrier is altered by dysbiosis, the aberrant immune system response interacts with the nervous system, leading to a state of "neuroinflammation". The gut microbiota-brain axis is mediated by inflammatory and immunological mechanisms, neurotransmitters, and neuroendocrine pathways. This narrative review aims to illustrate the molecular basis of neuroinflammation and elaborate on the concept of the gut-brain axis by virtue of analyzing the various metabolites produced by the gut microbiome and how they might impact the nervous system. Additionally, the current review will highlight how sex influences these molecular mechanisms. In fact, sex hormones impact the brain-gut microbiota axis at different levels, such as the central nervous system, the enteric nervous one, and enteroendocrine cells. A deeper understanding of the gut-brain axis in human health and disease is crucial to guide diagnoses, treatments, and preventive interventions.
Collapse
Affiliation(s)
- Mario Caldarelli
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Pierluigi Rio
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Andrea Marrone
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Francesca Ocarino
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Monica Chiantore
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Marcello Candelli
- Department of Emergency, Anesthesiological and Reanimation Sciences, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giovanni Gambassi
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Rossella Cianci
- Department of Translational Medicine and Surgery, Catholic University of Rome, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
26
|
Shad A, Rewell SSJ, Macowan M, Gandasasmita N, Wang J, Chen K, Marsland B, O'Brien TJ, Li J, Semple BD. Modelling lung infection with Klebsiella pneumoniae after murine traumatic brain injury. J Neuroinflammation 2024; 21:122. [PMID: 38720343 PMCID: PMC11080247 DOI: 10.1186/s12974-024-03093-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
Pneumonia is a common comorbidity in patients with severe traumatic brain injury (TBI), and is associated with increased morbidity and mortality. In this study, we established a model of intratracheal Klebsiella pneumoniae administration in young adult male and female mice, at 4 days following an experimental TBI, to investigate how K. pneumoniae infection influences acute post-TBI outcomes. A dose-response curve determined the optimal dose of K. pneumoniae for inoculation (1 x 10^6 colony forming units), and administration at 4 days post-TBI resulted in transient body weight loss and sickness behaviors (hypoactivity and acute dyspnea). K. pneumoniae infection led to an increase in pro-inflammatory cytokines in serum and bronchoalveolar lavage fluid at 24 h post-infection, in both TBI and sham (uninjured) mice. By 7 days, when myeloperoxidase + neutrophil numbers had returned to baseline in all groups, lung histopathology was observed with an increase in airspace size in TBI + K. pneumoniae mice compared to TBI + vehicle mice. In the brain, increased neuroinflammatory gene expression was observed acutely in response to TBI, with an exacerbated increase in Ccl2 and Hmox1 in TBI + K. pneumoniae mice compared to either TBI or K. pneumoniae alone. However, the presence of neuroinflammatory immune cells in the injured brain, and the extent of damage to cortical and hippocampal brain tissue, was comparable between K. pneumoniae and vehicle-treated mice by 7 days. Examination of the fecal microbiome across a time course did not reveal any pronounced effects of either injury or K. pneumoniae on bacterial diversity or abundance. Together, these findings demonstrate that K. pneumoniae lung infection after TBI induces an acute and transient inflammatory response, primarily localized to the lungs with some systemic effects. However, this infection had minimal impact on secondary injury processes in the brain following TBI. Future studies are needed to evaluate the potential longer-term consequences of this dual-hit insult.
Collapse
Affiliation(s)
- Ali Shad
- Department of Neuroscience, The School of Translational Medicine, Monash University, Level 6 Alfred Centre, 99 Commercial Rd, Melbourne, VIC, 3004 VIC, Australia
- Alfred Health, Prahran, VIC, Australia
| | - Sarah S J Rewell
- Department of Neuroscience, The School of Translational Medicine, Monash University, Level 6 Alfred Centre, 99 Commercial Rd, Melbourne, VIC, 3004 VIC, Australia
- Alfred Health, Prahran, VIC, Australia
| | - Matthew Macowan
- Department of Immunology, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- GIN Discovery Program, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Natasha Gandasasmita
- Department of Neuroscience, The School of Translational Medicine, Monash University, Level 6 Alfred Centre, 99 Commercial Rd, Melbourne, VIC, 3004 VIC, Australia
| | - Jiping Wang
- Department of Microbiology, Monash Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Ke Chen
- Department of Microbiology, Monash Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Ben Marsland
- Department of Immunology, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- GIN Discovery Program, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, The School of Translational Medicine, Monash University, Level 6 Alfred Centre, 99 Commercial Rd, Melbourne, VIC, 3004 VIC, Australia
- Alfred Health, Prahran, VIC, Australia
- GIN Discovery Program, The School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, Australia
| | - Jian Li
- Department of Microbiology, Monash Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Bridgette D Semple
- Department of Neuroscience, The School of Translational Medicine, Monash University, Level 6 Alfred Centre, 99 Commercial Rd, Melbourne, VIC, 3004 VIC, Australia.
- Alfred Health, Prahran, VIC, Australia.
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
27
|
Al Yacoub ON, Awwad HO, Standifer KM. Recovery from Traumatic Brain Injury Is Nociceptin/Orphanin FQ Peptide Receptor Genotype-, Sex-, and Injury Severity-Dependent. J Pharmacol Exp Ther 2024; 389:136-149. [PMID: 37442620 DOI: 10.1124/jpet.123.001664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/24/2023] [Accepted: 06/12/2023] [Indexed: 07/15/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of death and disability in the United States, and survivors often experience mental and physical health consequences that reduce quality of life. We previously reported that blockade of the nociceptin/orphanin FQ (N/OFQ) peptide (NOP) receptor reduced tissue damage markers produced by blast TBI. The goal of this study was to determine the extent to which N/OFQ and NOP receptor levels change following mild (mTBI) and moderate TBI (modTBI) and whether the absence of the NOP receptor attenuates TBI-induced sequelae. Male and female NOP receptor knockout (KO) or wild-type (WT) rats received craniotomy-only (sham) or craniotomy plus mTBI, or modTBI impact to the left cerebral hemisphere. Neurologic and vestibulomotor deficits and nociceptive hyperalgesia and allodynia found in WT male and female rats following mTBI and modTBI were greatly reduced or absent in NOP receptor KO rats. NOP receptor levels increased in brain tissue from injured males but remained unchanged in females. Neurofilament light chain (NF-L) and glial fibrillary acidic protein (GFAP) expression were reduced in NOP receptor KO rats compared with WT following TBI. Levels of N/OFQ in injured brain tissue correlated with neurobehavioral outcomes and GFAP in WT males, but not with KO male or WT and KO female rats. This study reveals a significant contribution of the N/OFQ-NOP receptor system to TBI-induced deficits and suggests that the NOP receptor should be regarded as a potential therapeutic target for TBI. SIGNIFICANCE STATEMENT: This study revealed that nociceptin/orphanin FQ peptide (NOP) receptor knockout animals experienced fewer traumatic brain injury (TBI)-induced deficits than their wild-type counterparts in a sex- and injury severity-dependent manner, suggesting that NOP receptor antagonists may be a potential therapy for TBI.
Collapse
Affiliation(s)
- Omar N Al Yacoub
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy (O.N.A., H.O.A., K.M.S.), and the Neuroscience Program (K.M.S., H.O.A.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hibah O Awwad
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy (O.N.A., H.O.A., K.M.S.), and the Neuroscience Program (K.M.S., H.O.A.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kelly M Standifer
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy (O.N.A., H.O.A., K.M.S.), and the Neuroscience Program (K.M.S., H.O.A.), University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
28
|
Dean T, Mendiola AS, Yan Z, Meza-Acevedo R, Cabriga B, Akassoglou K, Ryu JK. Fibrin promotes oxidative stress and neuronal loss in traumatic brain injury via innate immune activation. J Neuroinflammation 2024; 21:94. [PMID: 38622640 PMCID: PMC11017541 DOI: 10.1186/s12974-024-03092-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/05/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) causes significant blood-brain barrier (BBB) breakdown, resulting in the extravasation of blood proteins into the brain. The impact of blood proteins, especially fibrinogen, on inflammation and neurodegeneration post-TBI is not fully understood, highlighting a critical gap in our comprehension of TBI pathology and its connection to innate immune activation. METHODS We combined vascular casting with 3D imaging of solvent-cleared organs (uDISCO) to study the spatial distribution of the blood coagulation protein fibrinogen in large, intact brain volumes and assessed the temporal regulation of the fibrin(ogen) deposition by immunohistochemistry in a murine model of TBI. Fibrin(ogen) deposition and innate immune cell markers were co-localized by immunohistochemistry in mouse and human brains after TBI. We assessed the role of fibrinogen in TBI using unbiased transcriptomics, flow cytometry and immunohistochemistry for innate immune and neuronal markers in Fggγ390-396A knock-in mice, which express a mutant fibrinogen that retains normal clotting function, but lacks the γ390-396 binding motif to CD11b/CD18 integrin receptor. RESULTS We show that cerebral fibrinogen deposits were associated with activated innate immune cells in both human and murine TBI. Genetic elimination of fibrin-CD11b interaction reduced peripheral monocyte recruitment and the activation of inflammatory and reactive oxygen species (ROS) gene pathways in microglia and macrophages after TBI. Blockade of the fibrin-CD11b interaction was also protective from oxidative stress damage and cortical loss after TBI. CONCLUSIONS These data suggest that fibrinogen is a regulator of innate immune activation and neurodegeneration in TBI. Abrogating post-injury neuroinflammation by selective blockade of fibrin's inflammatory functions may have implications for long-term neurologic recovery following brain trauma.
Collapse
Affiliation(s)
- Terry Dean
- Gladstone Institute for Neurological Disease, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone, University of California San Francisco, San Francisco, CA, USA
- Center for Neuroscience Research, Children's National Hospital, Washington, DC, USA
| | - Andrew S Mendiola
- Gladstone Institute for Neurological Disease, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone, University of California San Francisco, San Francisco, CA, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Zhaoqi Yan
- Gladstone Institute for Neurological Disease, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone, University of California San Francisco, San Francisco, CA, USA
| | - Rosa Meza-Acevedo
- Gladstone Institute for Neurological Disease, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone, University of California San Francisco, San Francisco, CA, USA
| | - Belinda Cabriga
- Gladstone Institute for Neurological Disease, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone, University of California San Francisco, San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institute for Neurological Disease, San Francisco, CA, USA
- Center for Neurovascular Brain Immunology at Gladstone, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jae Kyu Ryu
- Gladstone Institute for Neurological Disease, San Francisco, CA, USA.
- Center for Neurovascular Brain Immunology at Gladstone, University of California San Francisco, San Francisco, CA, USA.
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
29
|
Ghosh M, Lee J, Burke AN, Strong TA, Sagen J, Pearse DD. Sex Dependent Disparities in the Central Innate Immune Response after Moderate Spinal Cord Contusion in Rat. Cells 2024; 13:645. [PMID: 38607084 PMCID: PMC11011714 DOI: 10.3390/cells13070645] [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: 01/31/2024] [Revised: 03/12/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Subacute spinal cord injury (SCI) displays a complex pathophysiology associated with pro-inflammation and ensuing tissue damage. Microglia, the resident innate immune cells of the CNS, in concert with infiltrating macrophages, are the primary contributors to SCI-induced inflammation. However, subpopulations of activated microglia can also possess immunomodulatory activities that are essential for tissue remodeling and repair, including the production of anti-inflammatory cytokines and growth factors that are vital for SCI recovery. Recently, reports have provided convincing evidence that sex-dependent differences exist in how microglia function during CNS pathologies and the extent to which these cells contribute to neurorepair and endogenous recovery. Herein we employed flow cytometry and immunohistochemical methods to characterize the phenotype and population dynamics of activated innate immune cells within the injured spinal cord of age-matched male and female rats within the first week (7 days) following thoracic SCI contusion. This assessment included the analysis of pro- and anti-inflammatory markers, as well as the expression of critical immunomodulatory kinases, including P38 MAPK, and transcription factors, such as NFκB, which play pivotal roles in injury-induced inflammation. We demonstrate that activated microglia from the injured spinal cord of female rats exhibited a significantly diminutive pro-inflammatory response, but enhanced anti-inflammatory activity compared to males. These changes included lower levels of iNOS and TLR4 expression but increased levels of ARG-1 and CD68 in females after SCI. The altered expression of these markers is indicative of a disparate secretome between the microglia of males and females after SCI and that the female microglia possesses higher phagocytic capabilities (increased CD68). The examination of immunoregulatory kinases and transcription factors revealed that female microglia had higher levels of phosphorylated P38Thr180/Tyr182 MAPK and nuclear NFκB pp50Ser337 but lower amounts of nuclear NFκB pp65Ser536, suggestive of an attenuated pro-inflammatory phenotype in females compared to males after SCI. Collectively, this work provides novel insight into some of the sex disparities that exist in the innate immune response after SCI and indicates that sex is an important variable when designing and testing new therapeutic interventions or interpretating positive or negative responses to an intervention.
Collapse
Affiliation(s)
- Mousumi Ghosh
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
| | - Jinyoung Lee
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Ashley N. Burke
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Thomas A. Strong
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
| | - Jacqueline Sagen
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Damien D. Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (J.L.); (A.N.B.); (T.A.S.); (J.S.); (D.D.P.)
- The Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Veterans Affairs, Veterans Affairs Medical Center, Miami, FL 33136, USA
- The Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- The Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
30
|
Davila-Valencia I, Saad M, Olthoff G, Faulkner M, Charara M, Farnum A, Dysko RC, Zhang Z. Sex specific effects of buprenorphine on adult hippocampal neurogenesis and behavioral outcomes during the acute phase after pediatric traumatic brain injury in mice. Neuropharmacology 2024; 245:109829. [PMID: 38159797 DOI: 10.1016/j.neuropharm.2023.109829] [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: 10/05/2023] [Revised: 12/15/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Traumatic brain injury (TBI) in children often causes cognitive and mental dysfunctions, as well as acute and chronic pain. Adult hippocampal neurogenesis plays a key role in cognition, depression, and pain. Adult hippocampal neurogenesis can be modulated by genetic and environmental factors, such as TBI and opioids. Buprenorphine (BPN), a semisynthetic opioid, is commonly used for pain management in children, however, the effects of BPN on adult hippocampal neurogenesis after pediatric TBI are still unclear. This study investigated the sex-specific effects of BPN on adult hippocampal neurogenesis during acute phase after pediatric TBI. Male and female littermates were randomized on postnatal day 20-21(P20-21) into Sham, TBI+saline and TBI+BPN groups. BPN was administered intraperitoneally to the TBI+BPN mice at 30 min after injury, and then every 6-12 h (h) for 2 days (d). Bromodeoxyuridine (BrdU) was administered intraperitoneally to all groups at 2, 4, 6, and 8-h post-injury. All outcomes were evaluated at 3-d post-BrdU administration. We found that TBI induced significant cognitive impairment, depression, and reduced adult hippocampal neurogenesis in both male and female mice, with more prominent effects in females. BPN significantly improved adult hippocampal neurogenesis and depression in males, but not in females. We further demonstrated that differential expressions of opioid receptors, transcription factors and neuroinflammatory markers at the neurogenic niche might be responsible for the differential effects of BPN in males and females. In conclusion, this study elucidates the effects of BPN on adult hippocampal neurogenesis and behavioral outcomes at the acute phase after pediatric TBI.
Collapse
Affiliation(s)
- Ivan Davila-Valencia
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Mark Saad
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Grace Olthoff
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Megan Faulkner
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Maysoun Charara
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Abigail Farnum
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Robert C Dysko
- Unit for Laboratory Animal Medicine, University of Michigan-Ann Arbor, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Zhi Zhang
- Department of Natural Sciences, College of Arts, Sciences, and Letters, University of Michigan-Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| |
Collapse
|
31
|
Boland R, Kokiko-Cochran ON. Deplete and repeat: microglial CSF1R inhibition and traumatic brain injury. Front Cell Neurosci 2024; 18:1352790. [PMID: 38450286 PMCID: PMC10915023 DOI: 10.3389/fncel.2024.1352790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Traumatic brain injury (TBI) is a public health burden affecting millions of people. Sustained neuroinflammation after TBI is often associated with poor outcome. As a result, increased attention has been placed on the role of immune cells in post-injury recovery. Microglia are highly dynamic after TBI and play a key role in the post-injury neuroinflammatory response. Therefore, microglia represent a malleable post-injury target that could substantially influence long-term outcome after TBI. This review highlights the cell specific role of microglia in TBI pathophysiology. Microglia have been manipulated via genetic deletion, drug inhibition, and pharmacological depletion in various pre-clinical TBI models. Notably, colony stimulating factor 1 (CSF1) and its receptor (CSF1R) have gained much traction in recent years as a pharmacological target on microglia. CSF1R is a transmembrane tyrosine kinase receptor that is essential for microglia proliferation, differentiation, and survival. Small molecule inhibitors targeting CSF1R result in a swift and effective depletion of microglia in rodents. Moreover, discontinuation of the inhibitors is sufficient for microglia repopulation. Attention is placed on summarizing studies that incorporate CSF1R inhibition of microglia. Indeed, microglia depletion affects multiple aspects of TBI pathophysiology, including neuroinflammation, oxidative stress, and functional recovery with measurable influence on astrocytes, peripheral immune cells, and neurons. Taken together, the data highlight an important role for microglia in sustaining neuroinflammation and increasing risk of oxidative stress, which lends to neuronal damage and behavioral deficits chronically after TBI. Ultimately, the insights gained from CSF1R depletion of microglia are critical for understanding the temporospatial role that microglia develop in mediating TBI pathophysiology and recovery.
Collapse
Affiliation(s)
- Rebecca Boland
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| | - Olga N Kokiko-Cochran
- Department of Neuroscience, College of Medicine, Chronic Brain Injury Program, Institute for Behavioral Medicine Research, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
32
|
Laaksonen S, Saraste M, Nylund M, Hinz R, Snellman A, Rinne J, Matilainen M, Airas L. Sex-driven variability in TSPO-expressing microglia in MS patients and healthy individuals. Front Neurol 2024; 15:1352116. [PMID: 38445263 PMCID: PMC10913932 DOI: 10.3389/fneur.2024.1352116] [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: 12/07/2023] [Accepted: 01/31/2024] [Indexed: 03/07/2024] Open
Abstract
Background Males with multiple sclerosis (MS) have a higher risk for disability progression than females, but the reasons for this are unclear. Objective We hypothesized that potential differences in TSPO-expressing microglia between female and male MS patients could contribute to sex differences in clinical disease progression. Methods The study cohort consisted of 102 MS patients (mean (SD) age 45.3 (9.7) years, median (IQR) disease duration 12.1 (7.0-17.2) years, 72% females, 74% relapsing-remitting MS) and 76 age- and sex-matched healthy controls. TSPO-expressing microglia were measured using the TSPO-binding radioligand [11C](R)-PK11195 and brain positron emission tomography (PET). TSPO-binding was quantified as distribution volume ratio (DVR) in normal-appearing white matter (NAWM), thalamus, whole brain and cortical gray matter (cGM). Results Male MS patients had higher DVRs compared to female patients in the whole brain [1.22 (0.04) vs. 1.20 (0.02), p = 0.002], NAWM [1.24 (0.06) vs. 1.21 (0.05), p = 0.006], thalamus [1.37 (0.08) vs. 1.32 (0.02), p = 0.008] and cGM [1.25 (0.04) vs. 1.23 (0.04), p = 0.028]. Similarly, healthy men had higher DVRs compared to healthy women except for cGM. Of the studied subgroups, secondary progressive male MS patients had the highest DVRs in all regions, while female controls had the lowest DVRs. Conclusion We observed higher TSPO-binding in males compared to females among people with MS and in healthy individuals. This sex-driven inherent variability in TSPO-expressing microglia may predispose male MS patients to greater likelihood of disease progression.
Collapse
Affiliation(s)
- Sini Laaksonen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Maija Saraste
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Marjo Nylund
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Rainer Hinz
- Wolfson Molecular Imaging Centre, University of Manchester, Manchester, United Kingdom
| | - Anniina Snellman
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Juha Rinne
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| | - Markus Matilainen
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
| | - Laura Airas
- Turku PET Centre, Turku University Hospital, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
- Clinical Neurosciences, University of Turku, Turku, Finland
- InFLAMES Research Flagship, University of Turku, Turku, Finland
| |
Collapse
|
33
|
Simmons A, Mihalek O, Bimonte Nelson HA, Sirianni RW, Stabenfeldt SE. Acute brain injury and nanomedicine: sex as a biological variable. FRONTIERS IN BIOMATERIALS SCIENCE 2024; 3:1348165. [PMID: 39450372 PMCID: PMC11500709 DOI: 10.3389/fbiom.2024.1348165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Sex as a biological variable has been recognized for decades to be a critical aspect of the drug development process, as differences in drug pharmacology and toxicity in female versus male subjects can drive the success or failure of new therapeutics. These concepts in development of traditional drug systems have only recently begun to be applied for advancing nanomedicine systems that are designed for drug delivery or imaging in the central nervous system (CNS). This review provides a comprehensive overview of the current state of two fields of research - nanomedicine and acute brain injury-centering on sex as a biological variable. We highlight areas of each field that provide foundational understanding of sex as a biological variable in nanomedicine, brain development, immune response, and pathophysiology of traumatic brain injury and stroke. We describe current knowledge on female versus male physiology as well as a growing number of empirical reports that directly address sex as a biological variable in these contexts. In sum, the data make clear two key observations. First, the manner in which sex affects nanomedicine distribution, toxicity, or efficacy is important, complex, and depends on the specific nanoparticle system under considerations; second, although field knowledge is accumulating to enable us to understand sex as a biological variable in the fields of nanomedicine and acute brain injury, there are critical gaps in knowledge that will need to be addressed. We anticipate that understanding sex as a biological variable in the development of nanomedicine systems to treat acute CNS injury will be an important determinant of their success.
Collapse
Affiliation(s)
- Amberlyn Simmons
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Olivia Mihalek
- Department of Neurological Surgery, UMass Chan Medical School, Worcester, MA, United States
| | | | - Rachael W. Sirianni
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
- Department of Neurological Surgery, UMass Chan Medical School, Worcester, MA, United States
| | - Sarah E. Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
34
|
Bahader GA, Naghavi F, Alotaibi A, Dehghan A, Swain CC, Burkett JP, Shah ZA. Neurobehavioral and inflammatory responses following traumatic brain injury in male and female mice. Behav Brain Res 2024; 456:114711. [PMID: 37827252 PMCID: PMC10615863 DOI: 10.1016/j.bbr.2023.114711] [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: 06/30/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and is associated with a high rate of functional comorbidities, including motor, cognitive, anxiety, depression, and emotional disorders. TBI pathophysiology and recovery are complicated and involve several mechanistic pathways that control neurobehavioral outcomes. In this study, male and female C57Bl/6 J mice were subjected to a controlled cortical impact model of TBI or sham injury and evaluated for different neurobehavioral and inflammatory outcomes over a month. We demonstrate that TBI mice have increased motor dysfunction at early and late time points following the injury as compared to the sham group. Anxiety-like symptoms were time- and task-dependent, with both sexes having increased anxiety-like behavior 2 weeks post-injury. Cognitive functions measured by T-maze presented greater deficits in TBI mice, while there was no sex or injury-related difference in depressive-like behaviors. Notably, a significant effect of sex was found in empathy-like behavior, with females showing more allogrooming and freezing behavior in the consoling and fear observational tests, respectively. Evaluating the impact of the injury-induced brain damage demonstrated a greater injury volume and neuronal degeneration in males compared to females one month after TBI. Moreover, male mice showed higher peripheral inflammatory responses, as represented by elevated serum levels of peripheral leukocytes and inflammatory markers. These results will have significant implications for understanding TBI's long-term consequences on neurobehavioral and inflammatory responses, which are sex-specific and can be considered for individualized therapeutic strategies in treating TBI.
Collapse
Affiliation(s)
- Ghaith A Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Farzaneh Naghavi
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Ahmed Alotaibi
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Amir Dehghan
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Caroline C Swain
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - James P Burkett
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA.
| |
Collapse
|
35
|
Yatoo MI, Bahader GA, Beigh SA, Khan AM, James AW, Asmi MR, Shah ZA. Neuroprotection or Sex Bias: A Protective Response to Traumatic Brain Injury in the Females. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:906-916. [PMID: 37592792 DOI: 10.2174/1871527323666230817102125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/29/2023] [Accepted: 07/14/2023] [Indexed: 08/19/2023]
Abstract
Traumatic brain injury (TBI) is a major healthcare problem and a common cause of mortality and morbidity. Clinical and preclinical research suggests sex-related differences in short- and longterm outcomes following TBI; however, males have been the main focus of TBI research. Females show a protective response against TBI. Female animals in preclinical studies and women in clinical trials have shown comparatively better outcomes against mild, moderate, or severe TBI. This reflects a favorable protective nature of the females compared to the males, primarily attributed to various protective mechanisms that provide better prognosis and recovery in the females after TBI. Understanding the sex difference in the TBI pathophysiology and the underlying mechanisms remains an elusive goal. In this review, we provide insights into various mechanisms related to the anatomical, physiological, hormonal, enzymatic, inflammatory, oxidative, genetic, or mitochondrial basis that support the protective nature of females compared to males. Furthermore, we sought to outline the evidence of multiple biomarkers that are highly potential in the investigation of TBI's prognosis, pathophysiology, and treatment and which can serve as objective measures and novel targets for individualized therapeutic interventions in TBI treatment. Implementations from this review are important for the understanding of the effect of sex on TBI outcomes and possible mechanisms behind the favorable response in females. It also emphasizes the critical need to include females as a biological variable and in sufficient numbers in future TBI studies.
Collapse
Affiliation(s)
- Mohammad I Yatoo
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Ghaith A Bahader
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Shafayat A Beigh
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Adil M Khan
- Division of Veterinary Clinical Complex, Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Shalimar, Shuhama, Alusteng, Srinagar, 190006, Jammu and Kashmir, India
| | - Antonisamy William James
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Maleha R Asmi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Zahoor A Shah
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
36
|
Weyer MP, Strehle J, Schäfer MKE, Tegeder I. Repurposing of pexidartinib for microglia depletion and renewal. Pharmacol Ther 2024; 253:108565. [PMID: 38052308 DOI: 10.1016/j.pharmthera.2023.108565] [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: 09/28/2023] [Revised: 11/20/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2023]
Abstract
Pexidartinib (PLX3397) is a small molecule receptor tyrosine kinase inhibitor of colony stimulating factor 1 receptor (CSF1R) with moderate selectivity over other members of the platelet derived growth factor receptor family. It is approved for treatment of tenosynovial giant cell tumors (TGCT). CSF1R is highly expressed by microglia, which are macrophages of the central nervous system (CNS) that defend the CNS against injury and pathogens and contribute to synapse development and plasticity. Challenged by pathogens, apoptotic cells, debris, or inflammatory molecules they adopt a responsive state to propagate the inflammation and eventually return to a homeostatic state. The phenotypic switch may fail, and disease-associated microglia contribute to the pathophysiology in neurodegenerative or neuropsychiatric diseases or long-lasting detrimental brain inflammation after brain, spinal cord or nerve injury or ischemia/hemorrhage. Microglia also contribute to the growth permissive tumor microenvironment of glioblastoma (GBM). In rodents, continuous treatment for 1-2 weeks via pexidartinib food pellets leads to a depletion of microglia and subsequent repopulation from the remaining fraction, which is aided by peripheral monocytes that search empty niches for engraftment. The putative therapeutic benefit of such microglia depletion or forced renewal has been assessed in almost any rodent model of CNS disease or injury or GBM with heterogeneous outcomes, but a tendency of partial beneficial effects. So far, microglia monitoring e.g. via positron emission imaging is not standard of care for patients receiving Pexidartinib (e.g. for TGCT), so that the depletion and repopulation efficiency in humans is still largely unknown. Considering the virtuous functions of microglia, continuous depletion is likely no therapeutic option but short-lasting transient partial depletion to stimulate microglia renewal or replace microglia in genetic disease in combination with e.g. stem cell transplantation or as part of a multimodal concept in treatment of glioblastoma appears feasible. The present review provides an overview of the preclinical evidence pro and contra microglia depletion as a therapeutic approach.
Collapse
Affiliation(s)
- Marc-Philipp Weyer
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany
| | - Jenny Strehle
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center Johannes Gutenberg-University Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Frankfurt, Faculty of Medicine, Frankfurt, Germany.
| |
Collapse
|
37
|
Culkin MC, Coons M, Bele P, Thaploo A, Georges AP, Anderson E, Browne KD, Jacovides C, Santos P, Kaplan LJ, Meaney DF, Smith DH, Pascual JL. Delayed tranexamic acid after traumatic brain injury impedes learning and memory: Early tranexamic acid is favorable but not in sham animals. J Trauma Acute Care Surg 2024; 96:26-34. [PMID: 37853567 DOI: 10.1097/ta.0000000000004155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
BACKGROUND Early but not late tranexamic acid (TXA) after TBI preserves blood-brain-barrier integrity, but it is unclear if and how dose timing affects cognitive recovery beyond hours postinjury. We hypothesized that early (1 hour post-TBI) but not late (24 hours post-TBI) TXA administration improves cognitive recovery for 14 days. METHODS CD1 male mice (n = 25) were randomized to severe TBI (injury [I], by controlled cortical impact) or sham craniotomy (S) followed by intravenous saline at 1 hour (placebo [P1]) or 30 mg/kg TXA at 1 hour (TXA1) or 24 hours (TXA24). Daily body weights, Garcia Neurological Test scores, brain/lung water content, and Morris water maze exercises quantifying swimming traffic in the platform quadrant (zone [Z] 1) and platform area (Z5) were recorded for up to 14 days. RESULTS Among injured groups, I-TXA1 demonstrated fastest weight gain for 14 days and only I-TXA1 showed rapid (day 1) normalization of Garcia Neurological Test ( p = 0.01 vs. I-P1, I-TXA24). In cumulative spatial trials, compared with I-TXA1, I-TXA24 hindered learning (distance to Z5 and % time in Z1, p < 0.05). Compared with I-TXA1, I-TXA24 showed poorer memory with less Z5 time (0.51 vs. 0.16 seconds, p < 0.01) and Z5 crossing frequency. Unexpectedly, TXA in uninjured animals (S-TXA1) displayed faster weight gain but inferior learning and memory. CONCLUSION Early TXA appears beneficial for cognitive and behavioral outcomes following TBI, although administration 24 hours postinjury consistently impairs cognitive recovery. Tranexamic acid in sham animals may lead to adverse effects on cognition.
Collapse
Affiliation(s)
- Matthew C Culkin
- From the Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery (M.C.C., M.C., P.B., A.T., C.J., P.S., L.J.K., J.L.P.), and Center for Brain Injury and Repair, Department of Neurosurgery (M.C.C., M.C., P.B., A.T., A.P.G., E.A., K.D.B., C.J., P.S., L.J.K., D.F.M., D.H.S., J.L.P.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Tarudji AW, Gee CC, Miller HA, Steffen R, Curtis ET, Priester AM, Convertine AJ, Kievit FM. Antioxidant theranostic copolymer-mediated reduction in oxidative stress following traumatic brain injury improves outcome in a mouse model. ADVANCED THERAPEUTICS 2023; 6:2300147. [PMID: 38464558 PMCID: PMC10923536 DOI: 10.1002/adtp.202300147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Indexed: 03/12/2024]
Abstract
Following a traumatic brain injury (TBI), excess reactive oxygen species (ROS) and lipid peroxidation products (LPOx) are generated and lead to secondary injury beyond the primary insult. A major limitation of current treatments is poor target engagement, which has prevented success in clinical trials. Thus, nanoparticle-based treatments have received recent attention because of their ability to increase accumulation and retention in damaged brain. Theranostic neuroprotective copolymers (NPC3) containing thiol functional groups can neutralize ROS and LPOx. Immediate administration of NPC3 following injury in a controlled cortical impact (CCI) mouse model provides a therapeutic window in reducing ROS levels at 2.08-20.83 mg/kg in males and 5.52-27.62 mg/kg in females. This NPC3-mediated reduction in oxidative stress improves spatial learning and memory in males, while females show minimal improvement. Notably, NPC3-mediated reduction in oxidative stress prevents the bilateral spread of necrosis in male mice, which was not observed in female mice and likely accounts for the sex-based spatial learning and memory differences. Overall, these findings suggest sex-based differences to oxidative stress scavenger nanoparticle treatments, and a possible upper threshold of antioxidant activity that provides therapeutic benefit in injured brain since female mice benefit from NPC3 treatment to a lesser extent than male mice.
Collapse
Affiliation(s)
- Aria W Tarudji
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 262 Morrison Center, Lincoln, NE, 68583, USA
| | - Connor C Gee
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 262 Morrison Center, Lincoln, NE, 68583, USA
| | - Hunter A Miller
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 262 Morrison Center, Lincoln, NE, 68583, USA
| | - Rylie Steffen
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 262 Morrison Center, Lincoln, NE, 68583, USA
| | - Evan T Curtis
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 262 Morrison Center, Lincoln, NE, 68583, USA
| | - Aaron M Priester
- Department of Materials Science and Engineering, Missouri University of Science and Technology, 223 McNutt Hall, Rolla, MO, 65409, USA
| | - Anthony J Convertine
- Department of Materials Science and Engineering, Missouri University of Science and Technology, 223 McNutt Hall, Rolla, MO, 65409, USA
| | - Forrest M Kievit
- Department of Biological Systems Engineering, University of Nebraska - Lincoln, 262 Morrison Center, Lincoln, NE, 68583, USA
| |
Collapse
|
39
|
Svirsky SE, Li Y, Henchir J, Rodina A, Carlson SW, Chiosis G, Dixon CE. Experimental traumatic brain injury increases epichaperome formation. Neurobiol Dis 2023; 188:106331. [PMID: 37863370 PMCID: PMC10698712 DOI: 10.1016/j.nbd.2023.106331] [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: 07/24/2023] [Revised: 09/13/2023] [Accepted: 10/17/2023] [Indexed: 10/22/2023] Open
Abstract
Under normal conditions, heat shock proteins work in unison through dynamic protein interactions collectively referred to as the "chaperome." Recent work revealed that during cellular stress, the functional interactions of the chaperome are modified to form the "epichaperome," which results in improper protein folding, degradation, aggregation, and transport. This study is the first to investigate this novel mechanism of protein dishomeostasis in traumatic brain injury (TBI). Male and female adult, Sprague-Dawley rats received a lateral controlled cortical impact (CCI) and the ipsilateral hippocampus was collected 24 h 1, 2, and 4 weeks after injury. The epichaperome complex was visualized by measuring HSP90, HSC70 and HOP expression in native-PAGE and normalized to monomeric protein expression. A two-way ANOVA examined the effect of injury and sex at each time-point. Native HSP90, HSC70 and HOP protein expression showed a significant effect of injury effect across all time-points. Additionally, HSC70 and HOP showed significant sex effects at 24 h and 4 weeks. Altogether, controlled cortical impact significantly increased formation of the epichaperome across all proteins measured. Further investigation of this pathological mechanism can lead to a greater understanding of the link between TBI and increased risk of neurodegenerative disease and targeting the epichaperome for therapeutics.
Collapse
Affiliation(s)
- Sarah E Svirsky
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | - Youming Li
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jeremy Henchir
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Anna Rodina
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Shaun W Carlson
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Gabriela Chiosis
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - C Edward Dixon
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurological Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA; V.A. Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
40
|
Ince LM, Darling JS, Sanchez K, Bell KS, Melbourne JK, Davis LK, Nixon K, Gaudet AD, Fonken LK. Sex differences in microglia function in aged rats underlie vulnerability to cognitive decline. Brain Behav Immun 2023; 114:438-452. [PMID: 37709153 PMCID: PMC10790303 DOI: 10.1016/j.bbi.2023.09.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/07/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023] Open
Abstract
Aging is associated with a significant shift in immune system reactivity ("inflammaging"), as basal inflammation increases but protective responses to infection are compromised. The immune system exhibits considerable sex differences, which may influence the process of inflammaging, including immune cell activation and behavioral consequences of immune signaling (i.e., impaired memory). Here, we test the hypothesis that sex differences in immune aging may mediate sex differences in cognitive decline. Aged male and female rats received peripheral immune stimulation using lipopolysaccharide (LPS), then molecular, cellular, and behavioral outcomes were assessed. We observed that LPS-treated aged male rats showed cognitive impairment and increased neuroinflammatory responses relative to adult males. In contrast, aged female rats did not display these aging-related deficits. Using transcriptomic and flow cytometry analyses, we further observed significant age- and sex- dependent changes in immune cell populations in the brain parenchyma and meninges, indicating a broad shift in the neuroinflammatory environment that may potentiate these behavioral effects. Ovariectomized aged female rats were also resistant to inflammation-induced memory deficits, indicating that ovarian hormones are not required for the attenuated neuroinflammation in aged females. Overall, our results indicate that males have amplified inflammatory priming with age, which contributes to age-associated cognitive decline. Our findings highlight sexual dimorphism in mechanisms of aging, and suggest that sex is a crucial consideration for identifying therapies for aging and neuroinflammation.
Collapse
Affiliation(s)
- Louise M Ince
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jeffrey S Darling
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Kevin Sanchez
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Kiersten S Bell
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Jennifer K Melbourne
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Lourdes K Davis
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA
| | - Kimberly Nixon
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA
| | - Andrew D Gaudet
- Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA; Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA; Department of Neurology, Dell Medical School, University of Texas at Austin, Austin, TX 78712, USA
| | - Laura K Fonken
- Division of Pharmacology & Toxicology, College of Pharmacy, University of Texas at Austin, Austin, TX 78712, USA; Institute for Neuroscience, University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
41
|
Neale KJ, Reid HMO, Sousa B, McDonagh E, Morrison J, Shultz S, Eyolfson E, Christie BR. Repeated mild traumatic brain injury causes sex-specific increases in cell proliferation and inflammation in juvenile rats. J Neuroinflammation 2023; 20:250. [PMID: 37907981 PMCID: PMC10617072 DOI: 10.1186/s12974-023-02916-5] [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: 06/14/2023] [Accepted: 09/29/2023] [Indexed: 11/02/2023] Open
Abstract
Childhood represents a period of significant growth and maturation for the brain, and is also associated with a heightened risk for mild traumatic brain injuries (mTBI). There is also concern that repeated-mTBI (r-mTBI) may have a long-term impact on developmental trajectories. Using an awake closed head injury (ACHI) model, that uses rapid head acceleration to induce a mTBI, we investigated the acute effects of repeated-mTBI (r-mTBI) on neurological function and cellular proliferation in juvenile male and female Long-Evans rats. We found that r-mTBI did not lead to cumulative neurological deficits with the model. R-mTBI animals exhibited an increase in BrdU + (bromodeoxyuridine positive) cells in the dentate gyrus (DG), and that this increase was more robust in male animals. This increase was not sustained, and cell proliferation returning to normal by PID3. A greater increase in BrdU + cells was observed in the dorsal DG in both male and female r-mTBI animals at PID1. Using Ki-67 expression as an endogenous marker of cellular proliferation, a robust proliferative response following r-mTBI was observed in male animals at PID1 that persisted until PID3, and was not constrained to the DG alone. Triple labeling experiments (Iba1+, GFAP+, Brdu+) revealed that a high proportion of these proliferating cells were microglia/macrophages, indicating there was a heightened inflammatory response. Overall, these findings suggest that rapid head acceleration with the ACHI model produces an mTBI, but that the acute neurological deficits do not increase in severity with repeated administration. R-mTBI transiently increases cellular proliferation in the hippocampus, particularly in male animals, and the pattern of cell proliferation suggests that this represents a neuroinflammatory response that is focused around the mid-brain rather than peripheral cortical regions. These results add to growing literature indicating sex differences in proliferative and inflammatory responses between females and males. Targeting proliferation as a therapeutic avenue may help reduce the short term impact of r-mTBI, but there may be sex-specific considerations.
Collapse
Affiliation(s)
- Katie J Neale
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Barbara Sousa
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Erin McDonagh
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Jamie Morrison
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Sandy Shultz
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
- Vancouver Island University, 900 Fifth Street, Nanaimo, BC, V9R 5S5, Canada
- Monash Trauma Group, Monash University, Melbourne, Australia
| | - Eric Eyolfson
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Medical Sciences Building,3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada.
- Institute for Aging and Life Long Health, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada.
- Island Medical Program, Cellular and Physiological Sciences, University of British Columbia, 3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, 3800 Finnerty Road, Victoria, BC, V8P 5C2, Canada.
| |
Collapse
|
42
|
Chu E, Mychasiuk R, Green TRF, Zamani A, Dill LK, Sharma R, Raftery AL, Tsantikos E, Hibbs ML, Semple BD. Regulation of microglial responses after pediatric traumatic brain injury: exploring the role of SHIP-1. Front Neurosci 2023; 17:1276495. [PMID: 37901420 PMCID: PMC10603304 DOI: 10.3389/fnins.2023.1276495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Severe traumatic brain injury (TBI) is the world's leading cause of permanent neurological disability in children. TBI-induced neurological deficits may be driven by neuroinflammation post-injury. Abnormal activity of SH2 domain-containing inositol 5' phosphatase-1 (SHIP-1) has been associated with dysregulated immunological responses, but the role of SHIP-1 in the brain remains unclear. The current study investigated the immunoregulatory role of SHIP-1 in a mouse model of moderate-severe pediatric TBI. Methods SHIP-1+/- and SHIP-1-/- mice underwent experimental TBI or sham surgery at post-natal day 21. Brain gene expression was examined across a time course, and immunofluorescence staining was evaluated to determine cellular immune responses, alongside peripheral serum cytokine levels by immunoassays. Brain tissue volume loss was measured using volumetric analysis, and behavior changes both acutely and chronically post-injury. Results Acutely, inflammatory gene expression was elevated in the injured cortex alongside increased IBA-1 expression and altered microglial morphology; but to a similar extent in SHIP-1-/- mice and littermate SHIP-1+/- control mice. Similarly, the infiltration and activation of CD68-positive macrophages, and reactivity of GFAP-positive astrocytes, was increased after TBI but comparable between genotypes. TBI increased anxiety-like behavior acutely, whereas SHIP-1 deficiency alone reduced general locomotor activity. Chronically, at 12-weeks post-TBI, SHIP-1-/- mice exhibited reduced body weight and increased circulating cytokines. Pro-inflammatory gene expression in the injured hippocampus was also elevated in SHIP-1-/- mice; however, GFAP immunoreactivity at the injury site in TBI mice was lower. TBI induced a comparable loss of cortical and hippocampal tissue in both genotypes, while SHIP-1-/- mice showed reduced general activity and impaired working memory, independent of TBI. Conclusion Together, evidence does not support SHIP-1 as an essential regulator of brain microglial morphology, brain immune responses, or the extent of tissue damage after moderate-severe pediatric TBI in mice. However, our data suggest that reduced SHIP-1 activity induces a greater inflammatory response in the hippocampus chronically post-TBI, warranting further investigation.
Collapse
Affiliation(s)
- Erskine Chu
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Deparment of Neurology, Alfred Health, Prahran, VIC, Australia
| | - Tabitha R. F. Green
- Department of Integrative Physiology, The University of Colorado Boulder, Boulder, CO, United States
| | - Akram Zamani
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - Larissa K. Dill
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Alfred Health, Prahran, VIC, Australia
| | - Rishabh Sharma
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
| | - April L. Raftery
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Evelyn Tsantikos
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Margaret L. Hibbs
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Bridgette D. Semple
- Department of Neuroscience, Monash University, Melbourne, VIC, Australia
- Deparment of Neurology, Alfred Health, Prahran, VIC, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
43
|
Terstege DJ, Epp JR. Parvalbumin as a sex-specific target in Alzheimer's disease research - A mini-review. Neurosci Biobehav Rev 2023; 153:105370. [PMID: 37619647 DOI: 10.1016/j.neubiorev.2023.105370] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/14/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, and both the incidence of this disease and its associated cognitive decline disproportionally effect women. While the etiology of AD is unknown, recent work has demonstrated that the balance of excitatory and inhibitory activity across the brain may serve as a strong predictor of cognitive impairments in AD. Across the cortex, the most prominent source of inhibitory signalling is from a class of parvalbumin-expressing interneurons (PV+). In this mini-review, the impacts of sex- and age-related factors on the function of PV+ neurons are examined within the context of vulnerability to AD pathology. These primary factors of influence include changes in brain metabolism, circulating sex hormone levels, and inflammatory response. In addition to positing the increased vulnerability of PV+ neurons to dysfunction in AD, this mini-review highlights the critical importance of presenting sex stratified data in the study of AD.
Collapse
Affiliation(s)
- Dylan J Terstege
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada
| | - Jonathan R Epp
- Department of Cell Biology and Anatomy, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
44
|
Scieszka D, Jin Y, Noor S, Barr E, Garcia M, Begay J, Herbert G, Hunter RP, Bhaskar K, Kumar R, Gullapalli R, Bolt A, McCormick MA, Bleske B, Gu H, Campen MJ. Biomass smoke inhalation promotes neuroinflammatory and metabolomic temporal changes in the hippocampus of female mice. J Neuroinflammation 2023; 20:192. [PMID: 37608305 PMCID: PMC10464132 DOI: 10.1186/s12974-023-02874-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/15/2023] [Indexed: 08/24/2023] Open
Abstract
Smoke from wildland fires has been shown to produce neuroinflammation in preclinical models, characterized by neural infiltrations of neutrophils and monocytes, as well as altered neurovascular endothelial phenotypes. To address the longevity of such outcomes, the present study examined the temporal dynamics of neuroinflammation and metabolomics after inhalation exposures from biomass-derived smoke. 2-month-old female C57BL/6 J mice were exposed to wood smoke every other day for 2 weeks at an average exposure concentration of 0.5 mg/m3. Subsequent serial euthanasia occurred at 1-, 3-, 7-, 14-, and 28-day post-exposure. Flow cytometry of right hemispheres revealed two endothelial populations of CD31Hi and CD31Med expressors, with wood smoke inhalation causing an increased proportion of CD31Hi. These populations of CD31Hi and CD31Med were associated with an anti-inflammatory and pro-inflammatory response, respectively, and their inflammatory profiles were largely resolved by the 28-day mark. However, activated microglial populations (CD11b+/CD45low) remained higher in wood smoke-exposed mice than controls at day 28. Infiltrating neutrophil populations decreased to levels below controls by day 28. However, the MHC-II expression of the peripheral immune infiltrate remained high, and the population of neutrophils retained an increased expression of CD45, Ly6C, and MHC-II. Utilizing an unbiased approach examining the metabolomic alterations, we observed notable hippocampal perturbations in neurotransmitter and signaling molecules, such as glutamate, quinolinic acid, and 5-α-dihydroprogesterone. Utilizing a targeted panel designed to explore the aging-associated NAD+ metabolic pathway, wood smoke exposure drove fluctuations and compensations across the 28-day time course, ending with decreased hippocampal NAD+ abundance on day 28. Summarily, these results indicate a highly dynamic neuroinflammatory environment, with potential resolution extending past 28 days, the implications of which may include long-term behavioral changes, systemic and neurological sequalae directly associated with wildfire smoke exposure.
Collapse
Affiliation(s)
- David Scieszka
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Yan Jin
- Florida International University Center for Translational Sciences, Port St. Lucie, FL, 34987, USA
| | - Shahani Noor
- Department of Molecular Genetics and Microbiology, Department of Neurology, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Ed Barr
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Marcus Garcia
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Jessica Begay
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Guy Herbert
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Russell P Hunter
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, Department of Neurology, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Rahul Kumar
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Rama Gullapalli
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Alicia Bolt
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA
| | - Mark A McCormick
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131, USA
| | - Barry Bleske
- Department of Pharmacy Practice and Administrative Sciences, College of Pharmacy, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Haiwei Gu
- Florida International University Center for Translational Sciences, Port St. Lucie, FL, 34987, USA
| | - Matthew J Campen
- Department of Pharmaceutical Sciences College of Pharmacy, University of New Mexico, MSC09 5360; 1, Albuquerque, NM, 87131-0001, USA.
| |
Collapse
|
45
|
Todd BP, Luo Z, Gilkes N, Chimenti MS, Peterson Z, Mix MR, Harty JT, Nickl-Jockschat T, Ferguson PJ, Bassuk AG, Newell EA. Selective neuroimmune modulation by type I interferon drives neuropathology and neurologic dysfunction following traumatic brain injury. Acta Neuropathol Commun 2023; 11:134. [PMID: 37596685 PMCID: PMC10436463 DOI: 10.1186/s40478-023-01635-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/05/2023] [Indexed: 08/20/2023] Open
Abstract
Accumulating evidence suggests that type I interferon (IFN-I) signaling is a key contributor to immune cell-mediated neuropathology in neurodegenerative diseases. Recently, we demonstrated a robust upregulation of type I interferon-stimulated genes in microglia and astrocytes following experimental traumatic brain injury (TBI). The specific molecular and cellular mechanisms by which IFN-I signaling impacts the neuroimmune response and neuropathology following TBI remains unknown. Using the lateral fluid percussion injury model (FPI) in adult male mice, we demonstrated that IFN α/β receptor (IFNAR) deficiency resulted in selective and sustained blockade of type I interferon-stimulated genes following TBI as well as decreased microgliosis and monocyte infiltration. Molecular alteration of reactive microglia also occurred with diminished expression of genes needed for MHC class I antigen processing and presentation following TBI. This was associated with decreased accumulation of cytotoxic T cells in the brain. The IFNAR-dependent modulation of the neuroimmune response was accompanied by protection from secondary neuronal death, white matter disruption, and neurobehavioral dysfunction. These data support further efforts to leverage the IFN-I pathway for novel, targeted therapy of TBI.
Collapse
Affiliation(s)
- Brittany P Todd
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
- Interdisciplinary Graduate Program in Neuroscience, University of Iowa, Iowa City, USA
| | - Zili Luo
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Noah Gilkes
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Michael S Chimenti
- Bioinformatics Division, Iowa Institute of Human Genetics, University of Iowa, Iowa City, IA, USA
| | - Zeru Peterson
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Madison R Mix
- Medical Scientist Training Program, University of Iowa, Iowa City, IA, USA
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - John T Harty
- Department of Pathology and Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA, USA
| | - Thomas Nickl-Jockschat
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Department of Psychiatry, University of Iowa, Iowa City, IA, USA
| | - Polly J Ferguson
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Alexander G Bassuk
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA
| | - Elizabeth A Newell
- Department of Pediatrics, University of Iowa, 200 Hawkins Drive, Iowa City, IA, 52242, USA.
| |
Collapse
|
46
|
Askarifirouzjaei H, Khajoueinejad L, Wei E, Cheruvu S, Ayala C, Chiang N, Theis T, Sun D, Fazeli M, Young W. Sex Differences in Immune Cell Infiltration and Hematuria in SCI-Induced Hemorrhagic Cystitis. PATHOPHYSIOLOGY 2023; 30:275-295. [PMID: 37489403 PMCID: PMC10366728 DOI: 10.3390/pathophysiology30030023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/07/2023] [Accepted: 07/08/2023] [Indexed: 07/26/2023] Open
Abstract
Rats manifest a condition called hemorrhagic cystitis after spinal cord injury (SCI). The mechanism of this condition is unknown, but it is more severe in male rats than in female rats. We assessed the role of sex regarding hemorrhagic cystitis and pathological chronic changes in the bladder. We analyzed the urine of male and female Sprague-Dawley and Fischer 344 rats after experimental spinal cord contusion, including unstained microscopic inspections of the urine, differential white blood cell counts colored by the Wright stain, and total leukocyte counts using fluorescent nuclear stains. We examined bladder histological changes in acute and chronic phases of SCI, using principal component analysis (PCA) and clustered heatmaps of Pearson correlation coefficients to interpret how measured variables correlated with each other. Male rats showed a distinct pattern of macroscopic hematuria after spinal cord injury. They had higher numbers of red blood cells with significantly more leukocytes and neutrophils than female rats, particularly hypersegmented neutrophils. The histological examination of the bladders revealed a distinct line of apoptotic umbrella cells and disrupted bladder vessels early after SCI and progressive pathological changes in multiple bladder layers in the chronic phase. Multivariate analyses indicated immune cell infiltration in the bladder, especially hypersegmented neutrophils, that correlated with red blood cell counts in male rats. Our study highlights a hitherto unreported sex difference of hematuria and pathological changes in males and females' bladders after SCI, suggesting an important role of immune cell infiltration, especially neutrophils, in SCI-induced hemorrhagic cystitis.
Collapse
Affiliation(s)
- Hadi Askarifirouzjaei
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Leila Khajoueinejad
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
- Department of Pharmacology, School of Veterinary Medicine, Shiraz University, Shiraz 71345, Iran
- Weill Cornell Medical College, New York, NY 10065, USA
| | - Elena Wei
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Sruti Cheruvu
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Carlos Ayala
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Ning Chiang
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Dongming Sun
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| | - Mehdi Fazeli
- Department of Pharmacology, School of Veterinary Medicine, Shiraz University, Shiraz 71345, Iran
| | - Wise Young
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA
| |
Collapse
|
47
|
Kobori N, Moore AN, Redell JB, Dash PK. Caudal DMN neurons innervate the spleen and release CART peptide to regulate neuroimmune function. J Neuroinflammation 2023; 20:158. [PMID: 37403174 PMCID: PMC10318820 DOI: 10.1186/s12974-023-02838-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/19/2023] [Indexed: 07/06/2023] Open
Abstract
BACKGROUND Inflammation is a fundamental biological response to injury and infection, which if unregulated can contribute to the pathophysiology of many diseases. The vagus nerve, which primarily originates from the dorsal motor nucleus (DMN), plays an important role in rapidly dampening inflammation by regulating splenic function. However, direct vagal innervation of the spleen, which houses the majority of immune and inflammatory cells, has not been established. As an alternative to direct innervation, an anti-inflammatory reflex pathway has been proposed which involves the vagus nerve, the sympathetic celiac ganglion, and the neurotransmitter norepinephrine. Although sympathetic regulation of inflammation has been shown, the interaction of the vagus nerve and the celiac ganglia requires a unique interaction of parasympathetic and sympathetic inputs, making this putative mechanism of brain-spleen interaction controversial. BODY: As neuropeptides can be expressed at relatively high levels in neurons, we reasoned that DMN neuropeptide immunoreactivity could be used to determine their target innervation. Employing immunohistochemistry, subdiaphragmatic vagotomy, viral tract tracing, CRISPR-mediated knock-down, and functional assays, we show that cocaine and amphetamine-regulated transcript (CART) peptide-expressing projection neurons in the caudal DMN directly innervate the spleen. In response to lipopolysaccharide (LPS) stimulation, CART acts to reduce inflammation, an effect that can be augmented by intrasplenic administration of a synthetic CART peptide. These in vivo effects could be recapitulated in cultured splenocytes, suggesting that these cells express the as yet unidentified CART receptor(s). CONCLUSION Our results provide evidence for direct connections between the caudal DMN and spleen. In addition to acetylcholine, these neurons express the neuropeptide CART that, once released, acts to suppress inflammation by acting directly upon splenocytes.
Collapse
Affiliation(s)
- Nobuhide Kobori
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Anthony N Moore
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - John B Redell
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA
| | - Pramod K Dash
- Department of Neurobiology and Anatomy, The University of Texas McGovern Medical School, P.O. Box 20708, Houston, TX, 77225, USA.
| |
Collapse
|
48
|
Culkin MC, Bele P, Georges AP, Lopez AJ, Niziolek G, Jacovides CL, Song H, Johnson VE, Kaplan LJ, Smith DH, Pascual JL. Early posttraumatic brain injury tranexamic acid prevents blood-brain barrier hyperpermeability and improves surrogates of neuroclinical recovery. J Trauma Acute Care Surg 2023; 95:47-54. [PMID: 37038259 DOI: 10.1097/ta.0000000000003971] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
BACKGROUND Tranexamic acid (TXA) given early, but not late, after traumatic brain injury (TBI) appears to improve survival. This may be partly related to TXA-driven profibrinolysis and increased leukocyte (LEU)-mediated inflammation when administered late post-injury. We hypothesized that early TXA (1 hour post-TBI), blunts penumbral, blood-brain barrier (BBB) leukocyte-endothelial cell (LEU-EC) interactions and microvascular permeability, in vivo when compared with late administration (24 hours post-TBI). METHODS CD1 male mice (n = 35) were randomized to severe TBI (injury by controlled cortical impact; injury: velocity, 6 m/s; depth, 1 mm; diameter, 3 mm) or sham craniotomy followed by intravenous saline (placebo) at 1 hour, or TXA (30 mg/kg) at 1 hour or 24 hours. At 48 hours, in vivo pial intravital microscopy visualized live penumbral LEU-EC interactions and BBB microvascular fluorescent albumin leakage. Neuroclinical recovery was assessed by the Garcia Neurological Test (motor, sensory, reflex, and balance assessments) and body weight loss recovery at 1 and 2 days after injury. Analysis of variance with Bonferroni correction assessed intergroup differences ( p < 0.05). RESULTS One-hour, but not 24-hour, TXA improved Garcia Neurological Test performance on day 1 post-TBI compared with placebo. Both 1 hour and 24 hours TXA similarly improved day 1 weight loss recovery, but only 1 hour TXA significantly improved weight loss recovery on day 2 compared with placebo ( p = 0.04). No intergroup differences were found in LEU rolling or adhesion between injured animal groups. Compared with untreated injured animals, only TXA at 1 hour reduced BBB permeability. CONCLUSION Only early post-TBI TXA consistently improves murine neurological recovery. Tranexamic acid preserves BBB integrity but only when administered early. This effect appears independent of LEU-EC interactions and demonstrates a time-sensitive effect that supports only early TXA administration.
Collapse
Affiliation(s)
- Matthew C Culkin
- From the Division of Traumatology, Surgical Critical Care and Emergency Surgery, Department of Surgery (M.C.C., P.B., A.J.L., G.N., C.L.J., L.J.K., J.L.P.), and Center for Brain Injury and Repair, Department of Neurosurgery (M.C.C., P.B., A.P.G., A.J.L., G.N., C.L.J., H.S., V.E.J., L.J.K., D.H.S., J.L.P.), Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Todd BP, Luo Z, Gilkes N, Chimenti MS, Peterson Z, Mix M, Harty JT, Nickl-Jockschat T, Ferguson PJ, Bassuk AG, Newell EA. Selective neuroimmune modulation by type I interferon drives neuropathology and neurologic dysfunction following traumatic brain injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543774. [PMID: 37333385 PMCID: PMC10274693 DOI: 10.1101/2023.06.06.543774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Accumulating evidence suggests that type I interferon (IFN-I) signaling is a key contributor to immune cell-mediated neuropathology in neurodegenerative diseases. Recently, we demonstrated a robust upregulation of type I interferon-stimulated genes in microglia and astrocytes following experimental traumatic brain injury (TBI). The specific molecular and cellular mechanisms by which IFN-I signaling impacts the neuroimmune response and neuropathology following TBI remains unknown. Using the lateral fluid percussion injury model (FPI) in adult male mice, we demonstrated that IFN α/β receptor (IFNAR) deficiency resulted in selective and sustained blockade of type I interferon-stimulated genes following TBI as well as decreased microgliosis and monocyte infiltration. Phenotypic alteration of reactive microglia also occurred with diminished expression of molecules needed for MHC class I antigen processing and presentation following TBI. This was associated with decreased accumulation of cytotoxic T cells in the brain. The IFNAR-dependent modulation of the neuroimmune response was accompanied by protection from secondary neuronal death, white matter disruption, and neurobehavioral dysfunction. These data support further efforts to leverage the IFN-I pathway for novel, targeted therapy of TBI.
Collapse
|
50
|
Scieszka D, Jin Y, Noor S, Barr E, Garcia M, Begay J, Herbert G, Hunter RP, Bhaskar K, Kumar R, Gullapalli R, Bolt A, McCormick MA, Bleske B, Gu H, Campen M. Neuroinflammatory and Metabolomic Temporal Dynamics Following Wood Smoke Inhalation. RESEARCH SQUARE 2023:rs.3.rs-3002040. [PMID: 37333410 PMCID: PMC10275049 DOI: 10.21203/rs.3.rs-3002040/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Smoke from wildland fires has been shown to produce neuroinflammation in preclinical models, characterized by neural infiltrations of neutrophils and monocytes, as well as altered neurovascular endothelial phenotypes. To address the longevity of such outcomes, the present study examined the neuroinflammatory and metabolomic temporal dynamics after inhalation exposures from biomass-derived smoke. 2-month-old female C57BL/6J mice were exposed to wood smoke every other day for two weeks at an average exposure concentration of 0.5mg/m 3 . Subsequent serial euthanasia occurred at 1-, 3-, 7-, 14-, and 28-days post-exposure. Flow cytometry of right hemispheres revealed two endothelial populations of PECAM (CD31), high and medium expressors, with wood smoke inhalation causing an increased proportion of PECAM Hi . These populations of PECAM Hi and PECAM Med were associated with an anti-inflammatory and pro-inflammatory response, respectively, and their inflammatory profiles were largely resolved by the 28-day mark. However, activated microglial populations (CD11b + /CD45 low ) remained higher in wood smoke-exposed mice than controls at day 28. Infiltrating neutrophil populations decreased to levels below controls by day 28. However, the MHC-II expression of the peripheral immune infiltrate remained high, and the population of neutrophils retained an increased expression of CD45, Ly6C, and MHC-II. Utilizing an unbiased approach examining the metabolomic alterations, we observed notable hippocampal perturbations in neurotransmitter and signaling molecules like glutamate, quinolinic acid, and 5-α-dihydroprogesterone. Utilizing a targeted panel designed to explore the aging-associated NAD + metabolic pathway, wood smoke exposure drove fluctuations and compensations across the 28-day time course, ending with decreased hippocampal NAD + abundance at day 28. Summarily, these results indicate a highly dynamic neuroinflammatory environment, with potential resolution extending past 28 days, the implications of which may include long-term behavioral changes, systemic and neurological sequalae directly associated wtith wildfire smoke exposure.
Collapse
Affiliation(s)
| | - Yan Jin
- Florida International University, Center for Translational Sciences
| | - Shahani Noor
- University of New Mexico, Department of Molecular Genetics and Microbiology
| | - Ed Barr
- University of New Mexico, College of Pharmacy
| | | | | | - Guy Herbert
- University of New Mexico, College of Pharmacy
| | | | - Kiran Bhaskar
- University of New Mexico, Department of Molecular Genetics and Microbiology
| | - Rahul Kumar
- University of New Mexico, Department of Pathology
| | | | - Alicia Bolt
- University of New Mexico, College of Pharmacy
| | - Mark A McCormick
- University of New Mexico, Department of Biochemistry and Molecular Biology
| | - Barry Bleske
- University of New Mexico, Department of Pharmacy Practice and Administrative Science
| | - Haiwei Gu
- Florida International University, Center for Translational Sciences
| | | |
Collapse
|