1
|
Liu L, Liang Z, Zhang L, Feng Z, Cao F, Zhang Y, Yang X, Zhang L, Wang J, Zhu Q. Corticothalamic input derived from corticospinal neurons contributes to chronic neuropathic pain after spinal cord injury. Exp Neurol 2024; 381:114923. [PMID: 39142366 DOI: 10.1016/j.expneurol.2024.114923] [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: 03/08/2024] [Revised: 08/01/2024] [Accepted: 08/12/2024] [Indexed: 08/16/2024]
Abstract
Neuropathic pain is a significant and persistent issue for individuals with spinal cord injuries (SCI), severely impacting their quality of life. While changes at the peripheral and spinal levels are known to contribute to SCI-related pain, whether and how supraspinal centers contribute to post SCI chronic neuropathic pain is poorly understood. Here, we first validated delayed development of chronic neuropathic pain in mice with moderate contusion SCI. To identify supraspinal regions involved in the pathology of neuropathic pain after SCI, we next performed an activity dependent genetic screening and identified multiple cortical and subcortical regions that were activated by innocuous tactile stimuli at a late stage following contusion SCI. Notably, chemogenetic inactivation of pain trapped neurons in the lateral thalamus alleviated neuropathic pain and reduced tactile stimuli evoked cortical overactivation. Retrograde tracing showed that contusion SCI led to enhanced corticothalamic axonal sprouting and over-activation of corticospinal neurons. Mechanistically, ablation or silencing of corticospinal neurons prevented the establishment or maintenance of chronic neuropathic pain following contusion SCI. These results highlighted a corticospinal-lateral thalamic feed-forward loop whose activation is required for the development and maintenance of chronic neuropathic pain after SCI. Our data thus shed lights into the central mechanisms underlying chronic neuropathic pain associated with SCI and the development of novel therapeutic avenues to treat refractory pain caused by traumatic brain or spinal cord injuries.
Collapse
Affiliation(s)
- Ling Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhou Feng
- Department of Rehabilitation, Southwest Hospital, Army Medical University, Chongqing, China
| | - Fei Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunjian Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lijie Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qing Zhu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
2
|
Krsek A, Ostojic L, Zivalj D, Baticic L. Navigating the Neuroimmunomodulation Frontier: Pioneering Approaches and Promising Horizons-A Comprehensive Review. Int J Mol Sci 2024; 25:9695. [PMID: 39273641 PMCID: PMC11396210 DOI: 10.3390/ijms25179695] [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: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The research in neuroimmunomodulation aims to shed light on the complex relationships that exist between the immune and neurological systems and how they affect the human body. This multidisciplinary field focuses on the way immune responses are influenced by brain activity and how neural function is impacted by immunological signaling. This provides important insights into a range of medical disorders. Targeting both brain and immunological pathways, neuroimmunomodulatory approaches are used in clinical pain management to address chronic pain. Pharmacological therapies aim to modulate neuroimmune interactions and reduce inflammation. Furthermore, bioelectronic techniques like vagus nerve stimulation offer non-invasive control of these systems, while neuromodulation techniques like transcranial magnetic stimulation modify immunological and neuronal responses to reduce pain. Within the context of aging, neuroimmunomodulation analyzes the ways in which immunological and neurological alterations brought on by aging contribute to cognitive decline and neurodegenerative illnesses. Restoring neuroimmune homeostasis through strategies shows promise in reducing age-related cognitive decline. Research into mood disorders focuses on how immunological dysregulation relates to illnesses including anxiety and depression. Immune system fluctuations are increasingly recognized for their impact on brain function, leading to novel treatments that target these interactions. This review emphasizes how interdisciplinary cooperation and continuous research are necessary to better understand the complex relationship between the neurological and immune systems.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Leona Ostojic
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Dorotea Zivalj
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
3
|
Zhang ZH, Wu TY, Ju C, Zuo XS, Wang XK, Ma YG, Luo L, Zhu ZJ, Song ZW, Yao Z, Zhou J, Wang Z, Hu XY. Photobiomodulation Increases M2-Type Polarization of Macrophages by Inhibiting Versican Production After Spinal Cord Injury. Mol Neurobiol 2024; 61:6950-6967. [PMID: 38363534 DOI: 10.1007/s12035-024-03980-5] [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: 07/17/2023] [Accepted: 01/21/2024] [Indexed: 02/17/2024]
Abstract
Spinal cord injury (SCI) is a catastrophic accidence with little effective treatment, and inflammation played an important role in that. Previous studies showed photobiomodulation (PBM) could effectively downregulate the process of inflammation with modification of macrophage polarization after SCI; however, the potential mechanism behind that is still unclear. In the presented study, we aimed to investigate the effect of PBM on the expression level of versican, a matrix molecular believed to be associated with inflammation, and tried to find the mechanism on how that could regulate the inflammation process. Using immunofluorescence technique and western blot, we found the expression level of versican is increased after injury and markedly downregulated by irradiation treatment. Using virus intrathecal injection, we found the knock-down of versican could produce the effect similar to that of PBM and might have an effect on inflammation and macrophage polarization after SCI. To further verify the deduction, we peptide the supernatant of astrocytes to induce M0, M1, and M2 macrophages. We found that the versican produced by astrocytes might have a role on the promotion of M2 macrophages to inflammatory polarization. Finally, we investigated the potential pathway in the regulation of M2 polarization with the induction of versican. This study tried to give an interpretation on the mechanism of inflammation inhibition for PBM in the perspective of matrix regulation. Our results might provide light on the inflammation regulation after SCI.
Collapse
Affiliation(s)
- Zhi-Hao Zhang
- General Hospital of Northern Theater Command, Shenyang, 110000, Liaoning Province, China
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Ting-Yu Wu
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Cheng Ju
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Xiao-Shuang Zuo
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Xuan-Kang Wang
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Yang-Guang Ma
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Liang Luo
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Zhi-Jie Zhu
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Zhi-Wen Song
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Zhou Yao
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Jie Zhou
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China
| | - Zhe Wang
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| | - Xue-Yu Hu
- Department of Orthopedics, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi Province, China.
| |
Collapse
|
4
|
Niazi SK, Mariam Z, Magoola M. Engineered Antibodies to Improve Efficacy against Neurodegenerative Disorders. Int J Mol Sci 2024; 25:6683. [PMID: 38928395 PMCID: PMC11203520 DOI: 10.3390/ijms25126683] [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: 05/11/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antibodies that can selectively remove rogue proteins in the brain are an obvious choice to treat neurodegenerative disorders (NDs), but after decades of efforts, only two antibodies to treat Alzheimer's disease are approved, dozens are in the testing phase, and one was withdrawn, and the other halted, likely due to efficacy issues. However, these outcomes should have been evident since these antibodies cannot enter the brain sufficiently due to the blood-brain barrier (BBB) protectant. However, all products can be rejuvenated by binding them with transferrin, preferably as smaller fragments. This model can be tested quickly and at a low cost and should be applied to bapineuzumab, solanezumab, crenezumab, gantenerumab, aducanumab, lecanemab, donanemab, cinpanemab, and gantenerumab, and their fragments. This paper demonstrates that conjugating with transferrin does not alter the binding to brain proteins such as amyloid-β (Aβ) and α-synuclein. We also present a selection of conjugate designs that will allow cleavage upon entering the brain to prevent their exocytosis while keeping the fragments connected to enable optimal binding to proteins. The identified products can be readily tested and returned to patients with the lowest regulatory cost and delays. These engineered antibodies can be manufactured by recombinant engineering, preferably by mRNA technology, as a more affordable solution to meet the dire need to treat neurodegenerative disorders effectively.
Collapse
Affiliation(s)
| | - Zamara Mariam
- Centre for Health and Life Sciences, Coventry University, Coventry City CV1 5FB, UK;
| | | |
Collapse
|
5
|
Cohen J, Mathew A, Dourvetakis KD, Sanchez-Guerrero E, Pangeni RP, Gurusamy N, Aenlle KK, Ravindran G, Twahir A, Isler D, Sosa-Garcia SR, Llizo A, Bested AC, Theoharides TC, Klimas NG, Kempuraj D. Recent Research Trends in Neuroinflammatory and Neurodegenerative Disorders. Cells 2024; 13:511. [PMID: 38534355 PMCID: PMC10969521 DOI: 10.3390/cells13060511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/03/2024] [Accepted: 03/12/2024] [Indexed: 03/28/2024] Open
Abstract
Neuroinflammatory and neurodegenerative disorders including Alzheimer's disease (AD), Parkinson's disease (PD), traumatic brain injury (TBI) and Amyotrophic lateral sclerosis (ALS) are chronic major health disorders. The exact mechanism of the neuroimmune dysfunctions of these disease pathogeneses is currently not clearly understood. These disorders show dysregulated neuroimmune and inflammatory responses, including activation of neurons, glial cells, and neurovascular unit damage associated with excessive release of proinflammatory cytokines, chemokines, neurotoxic mediators, and infiltration of peripheral immune cells into the brain, as well as entry of inflammatory mediators through damaged neurovascular endothelial cells, blood-brain barrier and tight junction proteins. Activation of glial cells and immune cells leads to the release of many inflammatory and neurotoxic molecules that cause neuroinflammation and neurodegeneration. Gulf War Illness (GWI) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are chronic disorders that are also associated with neuroimmune dysfunctions. Currently, there are no effective disease-modifying therapeutic options available for these diseases. Human induced pluripotent stem cell (iPSC)-derived neurons, astrocytes, microglia, endothelial cells and pericytes are currently used for many disease models for drug discovery. This review highlights certain recent trends in neuroinflammatory responses and iPSC-derived brain cell applications in neuroinflammatory disorders.
Collapse
Affiliation(s)
- Jessica Cohen
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Annette Mathew
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kirk D Dourvetakis
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Estella Sanchez-Guerrero
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Rajendra P Pangeni
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Narasimman Gurusamy
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Kristina K Aenlle
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Geeta Ravindran
- Cell Therapy Institute, Dr. Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Assma Twahir
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Dylan Isler
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Sara Rukmini Sosa-Garcia
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Axel Llizo
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Alison C Bested
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| | - Theoharis C Theoharides
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Laboratory of Molecular Immunopharmacology and Drug Discovery, Department of Immunology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Nancy G Klimas
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
- Miami VA Geriatric Research Education and Clinical Center (GRECC), Miami Veterans Affairs Healthcare System, Miami, FL 33125, USA
| | - Duraisamy Kempuraj
- Institute for Neuro-Immune Medicine, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Ft. Lauderdale, FL 33328, USA
| |
Collapse
|
6
|
Laricchiuta D, Papi M, Decandia D, Panuccio A, Cutuli D, Peciccia M, Mazzeschi C, Petrosini L. The role of glial cells in mental illness: a systematic review on astroglia and microglia as potential players in schizophrenia and its cognitive and emotional aspects. Front Cell Neurosci 2024; 18:1358450. [PMID: 38419655 PMCID: PMC10899480 DOI: 10.3389/fncel.2024.1358450] [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: 12/19/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024] Open
Abstract
Schizophrenia is a complex and severe mental disorder that affects approximately 1% of the global population. It is characterized by a wide range of symptoms, including delusions, hallucinations, disorganized speech and behavior, and cognitive impairment. Recent research has suggested that the immune system dysregulation may play a significant role in the pathogenesis of schizophrenia, and glial cells, such as astroglia and microglia known to be involved in neuroinflammation and immune regulation, have emerged as potential players in this process. The aim of this systematic review is to summarize the glial hallmarks of schizophrenia, choosing as cellular candidate the astroglia and microglia, and focusing also on disease-associated psychological (cognitive and emotional) changes. We conducted a systematic review following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We searched PubMed, Scopus, and Web of Science for articles that investigated the differences in astroglia and microglia in patients with schizophrenia, published in the last 5 years. The present systematic review indicates that changes in the density, morphology, and functioning of astroglia and microglia may be involved in the development of schizophrenia. The glial alterations may contribute to the pathogenesis of schizophrenia by dysregulating neurotransmission and immune responses, worsening cognitive capabilities. The complex interplay of astroglial and microglial activation, genetic/epigenetic variations, and cognitive assessments underscores the intricate relationship between biological mechanisms, symptomatology, and cognitive functioning in schizophrenia.
Collapse
Affiliation(s)
- Daniela Laricchiuta
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Martina Papi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Davide Decandia
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Anna Panuccio
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Debora Cutuli
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Psychology, University Sapienza of Rome, Rome, Italy
| | - Maurizio Peciccia
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Claudia Mazzeschi
- Department of Philosophy, Social Sciences and Education, University of Perugia, Perugia, Italy
| | - Laura Petrosini
- Laboratory of Experimental and Behavioral Neurophysiology, IRCCS Santa Lucia Foundation, Rome, Italy
| |
Collapse
|
7
|
Estera LA, Walsh SP, Headen JA, Williamson RE, Kalinski AL. Neuroinflammation: Breaking barriers and bridging gaps. Neurosci Res 2023; 197:9-17. [PMID: 34748905 DOI: 10.1016/j.neures.2021.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 01/04/2023]
Abstract
Neurons are the cells of the nervous system and are responsible for every thought, movement and perception. Immune cells are the cells of the immune system, constantly protecting from foreign pathogens. Understanding the interaction between the two systems is especially important in disease states such as autoimmune or neurodegenerative disease. Unfortunately, this interaction is typically detrimental to the host. However, recent efforts have focused on how neurons and immune cells interact, either directly or indirectly, following traumatic injury to the nervous system. The outcome of this interaction can be beneficial - leading to successful neural repair, or detrimental - leading to functional deficits, depending on where the injury occurs. This review will discuss our understanding of neuron-immune cell interactions after traumatic injury to both the peripheral and central nervous system.
Collapse
Affiliation(s)
- Lora A Estera
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Sam P Walsh
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | - Jordan A Headen
- Department of Biology, Ball State University, Muncie, IN 47306, USA
| | | | - Ashley L Kalinski
- Department of Biology, Ball State University, Muncie, IN 47306, USA.
| |
Collapse
|
8
|
Schoenberg PLA, Song AK, Mohr EM, Rogers BP, Peterson TE, Murphy BA. Increased microglia activation in late non-central nervous system cancer survivors links to chronic systemic symptomatology. Hum Brain Mapp 2023; 44:6001-6019. [PMID: 37751068 PMCID: PMC10619383 DOI: 10.1002/hbm.26491] [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: 02/03/2023] [Revised: 08/21/2023] [Accepted: 09/06/2023] [Indexed: 09/27/2023] Open
Abstract
Prolonged inflammatory expression within the central nervous system (CNS) is recognized by the brain as a molecular signal of "sickness", that has knock-on effects to the blood-brain barrier, brain-spinal barrier, blood-cerebrospinal fluid barrier, neuro-axonal structures, neurotransmitter activity, synaptic plasticity, neuroendocrine function, and resultant systemic symptomatology. It is concurred that the inflammatory process associated with cancer and cancer treatments underline systemic symptoms present in a large portion of survivors, although this concept is largely theoretical from disparate and indirect evidence and/or clinical anecdotal reports. We conducted a proof-of-concept study to link for the first time late non-CNS cancer survivors presenting chronic systemic symptoms and the presence of centralized inflammation, or neuroinflammation, using TSPO-binding PET tracer [11 C]-PBR28 to visualize microglial activation. We compared PBR28 SUVR in 10 non-CNS cancer survivors and 10 matched healthy controls. Our data revealed (1) microglial activation was significantly higher in caudate, temporal, and occipital regions in late non-central nervous system/CNS cancer survivors compared to healthy controls; (2) increased neuroinflammation in cancer survivors was not accompanied by significant differences in plasma cytokine markers of peripheral inflammation; (3) increased neuroinflammation was not accompanied by reduced fractional anisotropy, suggesting intact white matter microstructural integrity, a marker of neurovascular fiber tract organization; and (4) the presentation of chronic systemic symptoms in cancer survivors was significantly connected with microglial activation. We present the first data empirically supporting the concept of a peripheral-to-centralized inflammatory response in non-CNS cancer survivors, specifically those previously afflicted with head and neck cancer. Following resolution of the initial peripheral inflammation from the cancer/its treatments, in some cases damage/toxification to the central nervous system occurs, ensuing chronic systemic symptoms.
Collapse
Affiliation(s)
- Poppy L. A. Schoenberg
- Department of Physical Medicine and RehabilitationVanderbilt University Medical CenterNashvilleTennesseeUSA
- Osher Center for Integrative HealthVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Alexander K. Song
- Department of NeurologyVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Brain InstituteVanderbilt UniversityNashvilleTennesseeUSA
| | - Emily M. Mohr
- Osher Center for Integrative HealthVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Baxter P. Rogers
- Vanderbilt Brain InstituteVanderbilt UniversityNashvilleTennesseeUSA
- Department of Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Todd E. Peterson
- Vanderbilt Brain InstituteVanderbilt UniversityNashvilleTennesseeUSA
- Department of Radiology and Radiological SciencesVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Barbara A. Murphy
- Division of Hematology and OncologyVanderbilt‐Ingram Cancer CenterNashvilleTennesseeUSA
| |
Collapse
|
9
|
Wu X, Liu J, Li W, Khan MF, Dai H, Tian J, Priya R, Tian DJ, Wu W, Yaacoub A, Gu J, Syed F, Yu CH, Gao X, Yu Q, Xu XM, Brutkiewicz RR. CD1d-dependent neuroinflammation impairs tissue repair and functional recovery following a spinal cord injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.13.562047. [PMID: 37905092 PMCID: PMC10614755 DOI: 10.1101/2023.10.13.562047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Tissue damage resulting from a spinal cord injury (SCI) is primarily driven by a robust neuroimmune/neuroinflammatory response. This intricate process is mainly governed by a multitude of cytokines and cell surface proteins in the central nervous system (CNS). However, the critical components of the neuroimmune/neuroinflammatory response during SCI are still not well-defined. In this study, we investigated the impact of CD1d, an MHC class I-like molecule mostly known for presenting lipid antigens to natural killer T (NKT) cells and regulating immune/inflammatory responses, on neuroimmune/neuroinflammatory responses induced by SCI. We observed an increased expression of CD1d on various cell types within the spinal cord, including microglia/macrophages, oligodendrocytes (ODCs), and endothelial cells (DCs), but not on neurons or astrocytes post-SCI. In comparison to wildtype (WT) mice, a T10 contusive SCI in CD1d knockout (CD1dKO or Cd1d -/- ) mice resulted in markedly reduced proinflammatory cytokine release, microglia/macrophage activation and proliferation. Following SCI, the levels of inflammatory cytokines and activation/proliferation of microglia/macrophages were dramatically reduced, while anti-inflammatory cytokines such as IL-4 and growth factors like VEGF were substantially increased in the spinal cord tissues of CD1dKO mice when compared to WT mice. In the post-acute phase of SCI (day 7 post-SCI), CD1dKO mice had a significantly higher frequency of tissue-repairing macrophages, but not other types of immune cells, in the injured spinal cord tissues compared to WT mice. Moreover, CD1d-deficiency protected spinal cord neuronal cells and tissue, promoting functional recovery after a SCI. However, the neuroinflammation in WT mouse spinal cords was independent of the canonical CD1d/NKT cell axis. Finally, treatment of injured mice with a CD1d-specific monoclonal antibody significantly enhanced neuroprotection and improved functional recovery. Therefore, CD1d promotes the proinflammatory response following a SCI and represents a potential therapeutic target for spinal cord repair. Significance Statement The cell surface molecule, CD1d, is known to be recognized by cells of the immune system. To our knowledge, this is the first observation that the CD1d molecule significantly contributes to neuroinflammation following a spinal cord injury (SCI) in a manner independent of the CD1d/NKT cell axis. This is important, because this work reveals CD1d as a potential therapeutic target following an acute SCI for which there are currently no effective treatments.
Collapse
|
10
|
Ahmed RU, Medina‐Aguinaga D, Adams S, Knibbe CA, Morgan M, Gibson D, Kim J, Sharma M, Chopra M, Davison S, Sherwood LC, Negahdar M, Bert R, Ugiliweneza B, Hubscher C, Budde MD, Xu J, Boakye M. Predictive values of spinal cord diffusion magnetic resonance imaging to characterize outcomes after contusion injury. Ann Clin Transl Neurol 2023; 10:1647-1661. [PMID: 37501362 PMCID: PMC10502634 DOI: 10.1002/acn3.51855] [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: 04/14/2023] [Revised: 06/21/2023] [Accepted: 07/09/2023] [Indexed: 07/29/2023] Open
Abstract
OBJECTIVES To explore filtered diffusion-weighted imaging (fDWI), in comparison with conventional magnetic resonance imaging (MRI) and diffusion tensor imaging (DTI), as a predictor for long-term locomotor and urodynamic (UD) outcomes in Yucatan minipig model of spinal cord injury (SCI). Additionally, electrical conductivity of neural tissue using D-waves above and below the injury was measured to assess correlations between fDWI and D-waves data. METHODS Eleven minipigs with contusion SCI at T8-T10 level underwent MRI at 3T 4 h. post-SCI. Parameters extracted from region of interest analysis included Daxial from fDWI at injury site, fractional anisotropy and radial diffusivity from DTI above the injury site along with measures of edema length and cord width at injury site from T2 -weighted images. Locomotor recovery was assessed pre- and weekly post-SCI through porcine thoracic injury behavior scale (PTIBS) and UD were performed pre- and at 12 weeks of SCI. D-waves latency and amplitude differences were recorded before and immediately after SCI. RESULTS Two groups of pigs were found based on the PTIBS at week 12 (p < 0.0001) post-SCI and were labeled "poor" and "good" recovery. D-waves amplitude decreased below injury and increased above injury. UD outcomes pre/post SCI changed significantly. Conventional MRI metrics from T2 -weighted images were significantly correlated with diffusion MRI metrics. Daxial at injury epicenter was diminished by over 50% shortly after SCI, and it differentiated between good and poor locomotor recovery and UD outcomes. INTERPRETATION Similar to small animal studies, fDWI from acute imaging after SCI is a promising predictor for functional outcomes in large animals.
Collapse
Affiliation(s)
- Rakib Uddin Ahmed
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Daniel Medina‐Aguinaga
- Department of Anatomical Sciences and NeurobiologyUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Shawns Adams
- Department of NeurosurgeryDuke UniversityRaleighNorth CarolinaUSA
| | - Chase A. Knibbe
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Monique Morgan
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Destiny Gibson
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Joo‐won Kim
- Department of RadiologyBaylor College of MedicineHoustonTexasUSA
- Department of PsychiatryBaylor College of MedicineHoustonTexasUSA
| | - Mayur Sharma
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Manpreet Chopra
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Steven Davison
- Comparative Medicine Research UnitUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Leslie C. Sherwood
- Comparative Medicine Research UnitUniversity of LouisvilleLouisvilleKentuckyUSA
| | - M.J. Negahdar
- Department of RadiologyUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Robert Bert
- Department of RadiologyUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Beatrice Ugiliweneza
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Charles Hubscher
- Department of Anatomical Sciences and NeurobiologyUniversity of LouisvilleLouisvilleKentuckyUSA
| | - Matthew D. Budde
- Department of NeurosurgeryMedical College of WisconsinMilwaukeeWisconsinUSA
- Clement J. Zablocki Veterans Affairs Medical CenterMilwaukeeWisconsinUSA
| | - Junqian Xu
- Department of RadiologyBaylor College of MedicineHoustonTexasUSA
- Department of PsychiatryBaylor College of MedicineHoustonTexasUSA
| | - Maxwell Boakye
- Department of Neurological Surgery and Kentucky Spinal Cord Injury Research CenterUniversity of LouisvilleLouisvilleKentuckyUSA
| |
Collapse
|
11
|
Wulf MJ, Tom VJ. Consequences of spinal cord injury on the sympathetic nervous system. Front Cell Neurosci 2023; 17:999253. [PMID: 36925966 PMCID: PMC10011113 DOI: 10.3389/fncel.2023.999253] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 02/09/2023] [Indexed: 03/06/2023] Open
Abstract
Spinal cord injury (SCI) damages multiple structures at the lesion site, including ascending, descending, and propriospinal axons; interrupting the conduction of information up and down the spinal cord. Additionally, axons associated with the autonomic nervous system that control involuntary physiological functions course through the spinal cord. Moreover, sympathetic, and parasympathetic preganglionic neurons reside in the spinal cord. Thus, depending on the level of an SCI, autonomic function can be greatly impacted by the trauma resulting in dysfunction of various organs. For example, SCI can lead to dysregulation of a variety of organs, such as the pineal gland, the heart and vasculature, lungs, spleen, kidneys, and bladder. Indeed, it is becoming more apparent that many disorders that negatively affect quality-of-life for SCI individuals have a basis in dysregulation of the sympathetic nervous system. Here, we will review how SCI impacts the sympathetic nervous system and how that negatively impacts target organs that receive sympathetic innervation. A deeper understanding of this may offer potential therapeutic insight into how to improve health and quality-of-life for those living with SCI.
Collapse
Affiliation(s)
| | - Veronica J. Tom
- Marion Murray Spinal Cord Research Center, Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, PA, United States
| |
Collapse
|
12
|
Liu Y, Tan Y, Zhang Z, Li H, Yi M, Zhang Z, Hui S, Peng W. Neuroimmune mechanisms underlying Alzheimer's disease: Insights into central and peripheral immune cell crosstalk. Ageing Res Rev 2023; 84:101831. [PMID: 36565960 DOI: 10.1016/j.arr.2022.101831] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a highly life-threatening neurodegenerative disease. Dysregulation of the immune system plays a critical role in promoting AD, which has attracted extensive attention recently. Central and peripheral immune responses are involved in the pathogenesis of AD. Immune changes precede Aβ-associated senile plaque formation and tau-related neurofibrillary tangles, which are the recognised pathological features of AD. Therefore, elucidating immune-related mechanisms underlying the development of AD can help to prevent and treat AD at the source by blocking its progression before the development of pathological changes. To understand the specific pathogenesis of AD, it is important to examine the role of central and peripheral immunity in AD. This review summarises immune-related mechanisms underlying the pathogenesis of AD, focusing on the effect of various central and peripheral immune cells, and describes the possible crosstalk between central and peripheral immunity during the development of AD. This review provides novel insights into the treatment of AD and offers a new direction for immune-related research on AD in the future.
Collapse
Affiliation(s)
- Yuqing Liu
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Yejun Tan
- School of Mathematics, University of Minnesota Twin Cities, Minneapolis, MN, USA.
| | - Zheyu Zhang
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Hongli Li
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Min Yi
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| | - Zhen Zhang
- YangSheng College of Traditional Chinese Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China.
| | - Shan Hui
- Department of Geratology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha 410005, China.
| | - Weijun Peng
- Department of Integrated Traditional Chinese & Western Medicine, The Second Xiangya Hospital, Central South University, Changsha 410011, China; National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.
| |
Collapse
|
13
|
Owoyele BV, Bakare AO, Olaseinde OF, Ochu MJ, Yusuff AM, Ekebafe F, Fogabi OL, Roi T. Synergistic interaction between acetaminophen and L-carnosine improved neuropathic pain via NF-κB pathway and antioxidant properties in chronic constriction injury model. Korean J Pain 2022; 35:271-279. [PMID: 35768982 PMCID: PMC9251391 DOI: 10.3344/kjp.2022.35.3.271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/28/2022] Open
Abstract
Background Inflammation is known to underlie the pathogenesis in neuropathic pain. This study investigated the anti-inflammatory and neuroprotective mechanisms involved in antinociceptive effects of co-administration of acetaminophen and L-carnosine in chronic constriction injury (CCI)-induced peripheral neuropathy in male Wistar rats. Methods Fifty-six male Wistar rats were randomly divided into seven experimental groups (n = 8) treated with normal saline/acetaminophen/acetaminophen + L-carnosine. CCI was used to induce neuropathic pain in rats. Hyperalgesia and allodynia were assessed using hotplate and von Frey tests, respectively. Investigation of spinal proinflammatory cytokines and antioxidant system were carried out after twenty-one days of treatment. Results The results showed that the co-administration of acetaminophen and L-carnosine significantly (P < 0.001) increased the paw withdrawal threshold to thermal and mechanical stimuli in ligated rats compared to the ligated naïve group. There was a significant (P < 0.001) decrease in the levels of nuclear factor kappa light chain enhancer B cell inhibitor, calcium ion, interleukin-1-beta, and tumour necrotic factor-alpha in the spinal cord of the group coadministered with acetaminophen and L-carnosine compared to the ligated control group. Co-administration with acetaminophen and L-carnosine increased the antioxidant enzymatic activities and reduced the lipid peroxidation in the spinal cord. Conclusions Co-administration of acetaminophen and L-carnosine has anti-inflammatory effects as a mechanism that mediate its antinociceptive effects in CCI-induced peripheral neuropathy in Wistar rat.
Collapse
Affiliation(s)
- Bamidele Victor Owoyele
- Neuroscience and Inflammation Unit, Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Ahmed Olalekan Bakare
- Neuroscience and Inflammation Unit, Department of Physiology, Adeleke University, Ede, Osun State, Nigeria
| | - Olutayo Folajimi Olaseinde
- Neuroscience and Inflammation Unit, Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Mohammed Jelil Ochu
- Neuroscience and Inflammation Unit, Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Akorede Munirdeen Yusuff
- Neuroscience and Inflammation Unit, Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Favour Ekebafe
- Neuroscience and Inflammation Unit, Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Oluwadamilare Lanre Fogabi
- Neuroscience and Inflammation Unit, Department of Physiology, Faculty of Basic Medical Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Treister Roi
- Department of Nursing, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
14
|
Jung O, Thomas A, Burks SR, Dustin ML, Frank JA, Ferrer M, Stride E. Neuroinflammation associated with ultrasound-mediated permeabilization of the blood-brain barrier. Trends Neurosci 2022; 45:459-470. [PMID: 35461727 PMCID: PMC9117477 DOI: 10.1016/j.tins.2022.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/17/2022] [Accepted: 03/10/2022] [Indexed: 12/30/2022]
Abstract
The blood-brain barrier (BBB) continues to represent one of the most significant challenges for successful drug-based treatments of neurological disease. Mechanical modulation of the BBB using focused ultrasound (FUS) and microbubbles (MBs) has shown considerable promise in enhancing the delivery of therapeutics to the brain, but questions remain regarding possible long-term effects of such forced disruption. This review examines the evidence for inflammation associated with ultrasound-induced BBB disruption and potential strategies for managing such inflammatory effects to improve both the efficacy and safety of therapeutic ultrasound in neurological applications.
Collapse
Affiliation(s)
- Olive Jung
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK; 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Alec Thomas
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Scott R Burks
- The Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Michael L Dustin
- Nuffield Department of Orthopedics, Rheumatology, and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Joseph A Frank
- The Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA; Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Marc Ferrer
- 3D Tissue Bioprinting Laboratory, Department of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Eleanor Stride
- Biomedical Ultrasonics, Biotherapy, and Biopharmaceuticals Laboratory, Institute of Biomedical Engineering, University of Oxford, Oxford, UK.
| |
Collapse
|
15
|
Chondroitin sulfate proteoglycans prevent immune cell phenotypic conversion and inflammation resolution via TLR4 in rodent models of spinal cord injury. Nat Commun 2022; 13:2933. [PMID: 35614038 PMCID: PMC9133109 DOI: 10.1038/s41467-022-30467-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 05/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs) act as potent inhibitors of axonal growth and neuroplasticity after spinal cord injury (SCI). Here we reveal that CSPGs also play a critical role in preventing inflammation resolution by blocking the conversion of pro-inflammatory immune cells to a pro-repair phenotype in rodent models of SCI. We demonstrate that enzymatic digestion of CSPG glycosaminoglycans enhances immune cell clearance and reduces pro-inflammatory protein and gene expression profiles at key resolution time points. Analysis of phenotypically distinct immune cell clusters revealed CSPG-mediated modulation of macrophage and microglial subtypes which, together with T lymphocyte infiltration and composition changes, suggests a role for CSPGs in modulating both innate and adaptive immune responses after SCI. Mechanistically, CSPG activation of a pro-inflammatory phenotype in pro-repair immune cells was found to be TLR4-dependent, identifying TLR4 signalling as a key driver of CSPG-mediated immune modulation. These findings establish CSPGs as critical mediators of inflammation resolution failure after SCI in rodents, which leads to prolonged inflammatory pathology and irreversible tissue destruction.
Collapse
|
16
|
Role of Inflammation in Traumatic Brain Injury-Associated Risk for Neuropsychiatric Disorders: State of the Evidence and Where Do We Go From Here. Biol Psychiatry 2022; 91:438-448. [PMID: 34955170 DOI: 10.1016/j.biopsych.2021.11.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/01/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, there has been an increasing awareness that traumatic brain injury (TBI) and concussion substantially increase the risk for developing psychiatric disorders. Even mild TBI increases the risk for depression and anxiety disorders such as posttraumatic stress disorder by two- to threefold, predisposing patients to further functional impairment. This strong epidemiological link supports examination of potential mechanisms driving neuropsychiatric symptom development after TBI. One potential mechanism for increased neuropsychiatric symptoms after TBI is via inflammatory processes, as central nervous system inflammation can last years after initial injury. There is emerging preliminary evidence that TBI patients with posttraumatic stress disorder or depression exhibit increased central and peripheral inflammatory markers compared with TBI patients without these comorbidities. Growing evidence has demonstrated that immune signaling in animals plays an integral role in depressive- and anxiety-like behaviors after severe stress or brain injury. In this review, we will 1) discuss current evidence for chronic inflammation after TBI in the development of neuropsychiatric symptoms, 2) highlight potential microglial activation and cytokine signaling contributions, and 3) discuss potential promise and pitfalls for immune-targeted interventions and biomarker strategies to identify and treat TBI patients with immune-related neuropsychiatric symptoms.
Collapse
|
17
|
Walker JR, Detloff MR. Plasticity in Cervical Motor Circuits following Spinal Cord Injury and Rehabilitation. BIOLOGY 2021; 10:biology10100976. [PMID: 34681075 PMCID: PMC8533179 DOI: 10.3390/biology10100976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/17/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022]
Abstract
Simple Summary Spinal cord injury results in a decreased quality of life and impacts hundreds of thousands of people in the US alone. This review discusses the underlying cellular mechanisms of injury and the concurrent therapeutic hurdles that impede recovery. It then describes the phenomena of neural plasticity—the nervous system’s ability to change. The primary focus of the review is on the impact of cervical spinal cord injury on control of the upper limbs. The neural plasticity that occurs without intervention is discussed, which shows new connections growing around the injury site and the involvement of compensatory movements. Rehabilitation-driven neural plasticity is shown to have the ability to guide connections to create more normal functions. Various novel stimulation and recording technologies are outlined for their role in further improving rehabilitative outcomes and gains in independence. Finally, the importance of sensory input, an often-overlooked aspect of motor control, is shown in driving neural plasticity. Overall, this review seeks to delineate the historical and contemporary research into neural plasticity following injury and rehabilitation to guide future studies. Abstract Neuroplasticity is a robust mechanism by which the central nervous system attempts to adapt to a structural or chemical disruption of functional connections between neurons. Mechanical damage from spinal cord injury potentiates via neuroinflammation and can cause aberrant changes in neural circuitry known as maladaptive plasticity. Together, these alterations greatly diminish function and quality of life. This review discusses contemporary efforts to harness neuroplasticity through rehabilitation and neuromodulation to restore function with a focus on motor recovery following cervical spinal cord injury. Background information on the general mechanisms of plasticity and long-term potentiation of the nervous system, most well studied in the learning and memory fields, will be reviewed. Spontaneous plasticity of the nervous system, both maladaptive and during natural recovery following spinal cord injury is outlined to provide a baseline from which rehabilitation builds. Previous research has focused on the impact of descending motor commands in driving spinal plasticity. However, this review focuses on the influence of physical therapy and primary afferent input and interneuron modulation in driving plasticity within the spinal cord. Finally, future directions into previously untargeted primary afferent populations are presented.
Collapse
|
18
|
O'Reilly ML, Mironets E, Shapiro TM, Crowther K, Collyer E, Bethea JR, Tom VJ. Pharmacological Inhibition of Soluble Tumor Necrosis Factor-Alpha Two Weeks after High Thoracic Spinal Cord Injury Does Not Affect Sympathetic Hyperreflexia. J Neurotrauma 2021; 38:2186-2191. [PMID: 33397170 PMCID: PMC8309421 DOI: 10.1089/neu.2020.7504] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
After a severe, high-level spinal cord injury (SCI), plasticity to intraspinal circuits below injury results in heightened spinal sympathetic reflex activity and detrimentally impacts peripheral organ systems. Such sympathetic hyperreflexia is immediately apparent as an episode of autonomic dysreflexia (AD), a life-threatening condition characterized by sudden hypertension and reflexive bradycardia following below-level sensory inputs; for example, pressure sores or impacted fecal matter. Over time, plasticity within the spinal sympathetic reflex (SSR) circuit contributes to the progressive intensification of AD events, as the frequency and severity of AD events increase greatly beginning ∼2 weeks post-injury (wpi). The neuroimmune system has been implicated in driving sympathetic hyperreflexia, as inhibition of the cytokine soluble tumor necrosis factor-alpha (sTNFα) using the biological mimetic XPro1595 beginning within days post-SCI has been shown to attenuate the development of AD. Here, we sought to further understand the effective therapeutic time window of XPro1595 to diminish sympathetic hyperreflexia, as indicated by AD. We delayed the commencement of continuous intrathecal administration of XPro1595 until 2 weeks after a complete, thoracic level 3 injury in adult rats. We examined the severity of colorectal distension-induced AD biweekly. We found that initiation of sTNFα inhibition at 2 wpi does not attenuate the severity or intensification of sympathetic hyperreflexia compared with saline-treated controls. Coupled with previous data from our group, these findings suggest that central sTNFα signaling must be targeted prior to 2 weeks post-SCI in order to decrease sympathetic hyperreflexia.
Collapse
Affiliation(s)
- Micaela L. O'Reilly
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Eugene Mironets
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Tatiana M. Shapiro
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Kallon Crowther
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Eileen Collyer
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - John R. Bethea
- Department of Biology, Drexel University, Philadelphia, Pennsylvania, USA
| | - Veronica J. Tom
- Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
19
|
Passaro AP, Lebos AL, Yao Y, Stice SL. Immune Response in Neurological Pathology: Emerging Role of Central and Peripheral Immune Crosstalk. Front Immunol 2021; 12:676621. [PMID: 34177918 PMCID: PMC8222736 DOI: 10.3389/fimmu.2021.676621] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
Neuroinflammation is a key component of neurological disorders and is an important therapeutic target; however, immunotherapies have been largely unsuccessful. In cases where these therapies have succeeded, particularly multiple sclerosis, they have primarily focused on one aspect of the disease and leave room for improvement. More recently, the impact of the peripheral immune system is being recognized, since it has become evident that the central nervous system is not immune-privileged, as once thought. In this review, we highlight key interactions between central and peripheral immune cells in neurological disorders. While traditional approaches have examined these systems separately, the immune responses and processes in neurological disorders consist of substantial crosstalk between cells of the central and peripheral immune systems. Here, we provide an overview of major immune effector cells and the role of the blood-brain barrier in regard to neurological disorders and provide examples of this crosstalk in various disorders, including stroke and traumatic brain injury, multiple sclerosis, neurodegenerative diseases, and brain cancer. Finally, we propose targeting central-peripheral immune interactions as a potential improved therapeutic strategy to overcome failures in clinical translation.
Collapse
Affiliation(s)
- Austin P. Passaro
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA, United States
| | - Abraham L. Lebos
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Biochemistry and Microbiology, University of Georgia, Athens, GA, United States
| | - Yao Yao
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| | - Steven L. Stice
- Regenerative Bioscience Center, University of Georgia, Athens, GA, United States
- Division of Neuroscience, Biomedical Health and Sciences Institute, University of Georgia, Athens, GA, United States
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, United States
| |
Collapse
|
20
|
Guo YS, Yuan M, Han Y, Shen XY, Gao ZK, Bi X. Therapeutic Potential of Cytokines in Demyelinating Lesions After Stroke. J Mol Neurosci 2021; 71:2035-2052. [PMID: 33970426 DOI: 10.1007/s12031-021-01851-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022]
Abstract
White matter damage is a component of most human stroke and usually accounts for at least half of the lesion volume. Subcortical white matter stroke (WMS) accounts for 25% of all strokes and causes severe motor and cognitive dysfunction. The adult brain has a very limited ability to repair white matter damage. Pathological analysis shows that demyelination or myelin loss is the main feature of white matter injury and plays an important role in long-term sensorimotor and cognitive dysfunction. This suggests that demyelination is a major therapeutic target for ischemic stroke injury. An acute inflammatory reaction is triggered by brain ischemia, which is accompanied by cytokine production. The production of cytokines is an important factor affecting demyelination and myelin regeneration. Different cytokines have different effects on myelin damage and myelin regeneration. Exploring the role of cytokines in demyelination and remyelination after stroke and the underlying molecular mechanisms of demyelination and myelin regeneration after ischemic injury is very important for the development of rehabilitation treatment strategies. This review focuses on recent findings on the effects of cytokines on myelin damage and remyelination as well as the progress of research on the role of cytokines in ischemic stroke prognosis to provide a new treatment approach for amelioration of white matter damage after stroke.
Collapse
Affiliation(s)
- Yi-Sha Guo
- Shanghai University of Sport, Shanghai, 200438, China
| | - Mei Yuan
- Shanghai University of Sport, Shanghai, 200438, China
| | - Yu Han
- Shanghai University of Sport, Shanghai, 200438, China
| | - Xin-Ya Shen
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Zhen-Kun Gao
- Shanghai University of Traditional Chinese Medicine, Shanghai, 200438, China
| | - Xia Bi
- Department of Rehabilitation Medicine, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, 201318, China.
| |
Collapse
|
21
|
Conditional RAC1 knockout in motor neurons restores H-reflex rate-dependent depression after spinal cord injury. Sci Rep 2021; 11:7838. [PMID: 33837249 PMCID: PMC8035187 DOI: 10.1038/s41598-021-87476-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/30/2021] [Indexed: 12/27/2022] Open
Abstract
A major complication with spinal cord injury (SCI) is the development of spasticity, a clinical symptom of hyperexcitability within the spinal H-reflex pathway. We have previously demonstrated a common structural motif of dendritic spine dysgenesis associated with hyperexcitability disorders after injury or disease insults to the CNS. Here, we used an adeno-associated viral (AAV)-mediated Cre-Lox system to knockout Rac1 protein expression in motor neurons after SCI. Three weeks after AAV9-Cre delivery into the soleus/gastrocnemius of Rac1-“floxed” adult mice to retrogradely infect spinal alpha-motor neurons, we observed significant restoration of RDD and reduced H-reflex excitability in SCI animals. Additionally, viral-mediated Rac1 knockdown reduced presence of dendritic spine dysgenesis on motor neurons. In control SCI animals without Rac1 knockout, we continued to observe abnormal dendritic spine morphology associated with hyperexcitability disorder, including an increase in mature, mushroom dendritic spines, and an increase in overall spine length and spine head size. Taken together, our results demonstrate that viral-mediated disruption of Rac1 expression in ventral horn motor neurons can mitigate dendritic spine morphological correlates of neuronal hyperexcitability, and reverse hyperreflexia associated with spasticity after SCI. Finally, our findings provide evidence of a putative mechanistic relationship between motor neuron dendritic spine dysgenesis and SCI-induced spasticity.
Collapse
|
22
|
Kempuraj D, Thangavel R, Kempuraj DD, Ahmed ME, Selvakumar GP, Raikwar SP, Zaheer SA, Iyer SS, Govindarajan R, Chandrasekaran PN, Zaheer A. Neuroprotective effects of flavone luteolin in neuroinflammation and neurotrauma. Biofactors 2021; 47:190-197. [PMID: 33098588 DOI: 10.1002/biof.1687] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/27/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
Abstract
Neuroinflammation leads to neurodegeneration, cognitive defects, and neurodegenerative disorders. Neurotrauma/traumatic brain injury (TBI) can cause activation of glial cells, neurons, and neuroimmune cells in the brain to release neuroinflammatory mediators. Neurotrauma leads to immediate primary brain damage (direct damage), neuroinflammatory responses, neuroinflammation, and late secondary brain damage (indirect) through neuroinflammatory mechanism. Secondary brain damage leads to chronic inflammation and the onset and progression of neurodegenerative diseases. Currently, there are no effective and specific therapeutic options to treat these brain damages or neurodegenerative diseases. Flavone luteolin is an important natural polyphenol present in several plants that show anti-inflammatory, antioxidant, anticancer, cytoprotective, and macrophage polarization effects. In this short review article, we have reviewed the neuroprotective effects of luteolin in neurotrauma and neurodegenerative disorders and pathways involved in this mechanism. We have collected data for this study from publications in the PubMed using the keywords luteolin and mast cells, neuroinflammation, neurodegenerative diseases, and TBI. Recent reports suggest that luteolin suppresses systemic and neuroinflammatory responses in Coronavirus disease 2019 (COVID-19). Studies have shown that luteolin exhibits neuroprotective effects through various mechanisms, including suppressing immune cell activation, such as mast cells, and inflammatory mediators released from these cells. In addition, luteolin can suppress neuroinflammatory response, activation of microglia and astrocytes, oxidative stress, neuroinflammation, and the severity of neuroinflammatory diseases such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, and TBI pathogenesis. In conclusion, luteolin can improve cognitive decline and enhance neuroprotection in neurodegenerative diseases, TBI, and stroke.
Collapse
Affiliation(s)
- Duraisamy Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Ramasamy Thangavel
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Deepak D Kempuraj
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- David H. Hickman High School, Columbia Public Schools, Columbia, Missouri, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Govindhasamy Pushpavathi Selvakumar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Sudhanshu P Raikwar
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Smita A Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | - Shankar S Iyer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| | - Raghav Govindarajan
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
| | | | - Asgar Zaheer
- Department of Neurology, School of Medicine, University of Missouri, Columbia, Missouri, USA
- The Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Memorial Veterans Hospital, U.S. Department of Veterans Affairs, Columbia, Missouri, USA
| |
Collapse
|
23
|
Tsuchiyagaito A, Smith JL, El-Sabbagh N, Zotev V, Misaki M, Al Zoubi O, Kent Teague T, Paulus MP, Bodurka J, Savitz J. Real-time fMRI neurofeedback amygdala training may influence kynurenine pathway metabolism in major depressive disorder. NEUROIMAGE-CLINICAL 2021; 29:102559. [PMID: 33516062 PMCID: PMC7847971 DOI: 10.1016/j.nicl.2021.102559] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/30/2020] [Accepted: 01/09/2021] [Indexed: 12/15/2022]
Abstract
rtfMRI-nf LA emotional training reduces depressive symptoms. rtfMRI-nf LA training increases KynA/3-HK, a neuroprotective index. Baseline KynA/QA is associated with the ability to upregulate the LA. In partial responder group LA upregulation positively correlates with KynA/QA. In partial responder group LA upregulation inversely correlates with MADRS. Modulation of the KP may drive rtfMRI-nf-induced changes in neuroplasticity. Non-specific effects cannot be ruled out due to the lack of a sham control.
Real-time fMRI neurofeedback (rtfMRI-nf) left amygdala (LA) training is a promising intervention for major depressive disorder (MDD). We have previously proposed that rtfMRI-nf LA training may reverse depression-associated regional impairments in neuroplasticity and restore information flow within emotion-regulating neural circuits. Inflammatory cytokines as well as the neuroactive metabolites of an immunoregulatory pathway, i.e. the kynurenine pathway (KP), have previously been implicated in neuroplasticity. Therefore, in this proof-of-principle study, we investigated the association between rtfMRI-nf LA training and circulating inflammatory mediators and KP metabolites. Based on our previous work, the primary variable of interest was the ratio of the NMDA-receptor antagonist, kynurenic acid to the NMDA receptor agonist, quinolinic acid (KynA/QA), a putative neuroprotective index. We tested two main hypotheses. i. Whether rtfMRI-nf acutely modulates KynA/QA, and ii. whether baseline KynA/QA predicts response to rtfMRI-nf. Twenty-nine unmedicated participants who met DSM-5 criteria for MDD based on the Mini-International Neuropsychiatric Interview and had current depressive symptoms (Montgomery-Åsberg Depression Rating Scale (MADRS) score > 6) completed two rtfMRI-nf sessions to upregulate LA activity (Visit1 and 2), as well as a follow-up (Visit3) without rtfMRI-nf. All visits occurred at two-week intervals. At all three visits, the MADRS was administered to participants and serum samples for the quantification of inflammatory cytokines and KP metabolites were obtained. First, the longitudinal changes in the MADRS score and immune markers were tested by linear mixed effect model analysis. Further, utilizing a linear regression model, we investigated the relationship between rtfMRI-nf performance and immune markers. After two sessions of rtfMRI-nf, MADRS scores were significantly reduced (t[58] = −4.07, p = 0.009, d = 0.56). Thirteen participants showed a ≥ 25% reduction in the MADRS score (the partial responder group). There was a significant effect of visit (F[2,58] = 3.17, p = 0.05) for the neuroprotective index, KynA to 3-hydroxykynurenine (3-HK), that was driven by a significant increase in KynA/3-HK between Visit1 and Visit3 (t[58] = 2.50, p = 0.03, d = 0.38). A higher baseline level of KynA/QA (β = 5.23, p = 0.06; rho = 0.49, p = 0.02) was associated with greater ability to upregulate the LA. Finally, for exploratory purposes correlation analyses were performed between the partial responder and the non-responder groups as well as in the whole sample including all KP metabolites and cytokines. In the partial responder group, greater ability to upregulate the LA was correlated with an increase in KynA/QA after rtfMRI-nf (rho = 0.75, p = 0.03). The results are consistent with the possibility that rtfMRI-nf decreases metabolism down the so-called neurotoxic branch of the KP. Nevertheless, non-specific effects cannot be ruled out due to the lack of a sham control. Future, controlled studies are needed to determine whether the increase in KynA/3HK and KynA/QA is specific to rtfMRI-nf or whether it is a non-specific correlate of the resolution of depressive symptoms. Similarly, replication studies are needed to determine whether KynA/QA has clinical utility as a treatment response biomarker.
Collapse
Affiliation(s)
- Aki Tsuchiyagaito
- Laureate Institute for Brain Research, Tulsa, OK, USA; Research Center for Child Mental Development, Chiba University, Chiba, Japan
| | - Jared L Smith
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | | | - Vadim Zotev
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | - Masaya Misaki
- Laureate Institute for Brain Research, Tulsa, OK, USA
| | | | - T Kent Teague
- Department of Surgery, University of Oklahoma School of Community Medicine, Tulsa, OK, USA; Department of Psychiatry, University of Oklahoma School of Community Medicine, Tulsa, OK, USA; Department of Biochemistry and Microbiology, Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
| | | | - Jerzy Bodurka
- Laureate Institute for Brain Research, Tulsa, OK, USA; Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA.
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA; Department of Community Medicine, Oxley Health Sciences, University of Tulsa, Tulsa, OK, USA.
| |
Collapse
|
24
|
Bangma JT, Hartwell H, Santos HP, O'Shea TM, Fry RC. Placental programming, perinatal inflammation, and neurodevelopment impairment among those born extremely preterm. Pediatr Res 2021; 89:326-335. [PMID: 33184498 PMCID: PMC7658618 DOI: 10.1038/s41390-020-01236-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/02/2020] [Accepted: 10/08/2020] [Indexed: 01/30/2023]
Abstract
Individuals born extremely preterm are at significant risk for impaired neurodevelopment. After discharge from the neonatal intensive care, associations between the child's well-being and factors in the home and social environment become increasingly apparent. Mothers' prenatal health and socioeconomic status are associated with neurodevelopmental outcomes, and emotional and behavioral problems. Research on early life risk factors and on mechanisms underlying inter-individual differences in neurodevelopment later in life can inform the design of personalized approaches to prevention. Here, we review early life predictors of inter-individual differences in later life neurodevelopment among those born extremely preterm. Among biological mechanisms that mediate relationships between early life predictors and later neurodevelopmental outcomes, we highlight evidence for disrupted placental processes and regulated at least in part via epigenetic mechanisms, as well as perinatal inflammation. In relation to these mechanisms, we focus on four prenatal antecedents of impaired neurodevelopment, namely, (1) fetal growth restriction, (2) maternal obesity, (3) placental microorganisms, and (4) socioeconomic adversity. In the future, this knowledge may inform efforts to detect and prevent adverse outcomes in infants born extremely preterm. IMPACT: This review highlights early life risk factors and mechanisms underlying inter-individual differences in neurodevelopment later in life. The review emphasizes research on early life risk factors (fetal growth restriction, maternal obesity, placental microorganisms, and socioeconomic adversity) and on mechanisms (disrupted placental processes and perinatal inflammation) underlying inter-individual differences in neurodevelopment later in life. The findings highlighted here may inform efforts to detect and prevent adverse outcomes in infants born extremely preterm.
Collapse
Affiliation(s)
- Jacqueline T Bangma
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hadley Hartwell
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hudson P Santos
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - T Michael O'Shea
- Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rebecca C Fry
- Biobehavioral Laboratory, School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Institute for Environmental Health Solutions, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Curriculum in Toxicology and Environmental Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
25
|
Young G. Thirty Complexities and Controversies in Mild Traumatic Brain Injury and Persistent Post-concussion Syndrome: a Roadmap for Research and Practice. PSYCHOLOGICAL INJURY & LAW 2020. [DOI: 10.1007/s12207-020-09395-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Wieters F, Weiss Lucas C, Gruhn M, Büschges A, Fink GR, Aswendt M. Introduction to spasticity and related mouse models. Exp Neurol 2020; 335:113491. [PMID: 33007294 DOI: 10.1016/j.expneurol.2020.113491] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 12/22/2022]
Abstract
Although spasticity is one of the most common causes of motor disability worldwide, its precise definition and pathophysiology remain elusive, which to date renders its experimental targeting tricky. At least in part, this difficulty is caused by heterogeneous phenotypes of spasticity-causing neurological disorders, all causing spasticity by involving upper motor neurons. The most common clinical symptoms are a series of rapid muscle contractions (clonus), an increased muscle tone (hypertonia), and augmented tendon reflex activity (hyperreflexia). This muscle overactivity is due to disturbed inhibition of spinal reflexes following upper motor neuron dysfunction. Despite a range of physical and pharmacological therapies ameliorating the symptoms, their targeted application remains difficult. Therefore, to date, spasticity impacts rehabilitative therapy, and no therapy exists that reverses the pathology completely. In contrast to the incidence and importance of spasticity, only very little pre-clinical work in animal models exists, and this research is focused on the cat or the rat spastic tail model to decipher altered reflexes and excitability of the motor neurons in the spinal cord. Meanwhile, the characterization of spasticity in clinically more relevant mouse models of neurological disorders, such as stroke, remains understudied. Here, we provide a brief introduction into the clinical knowledge and therapy of spasticity and an in-depth review of pre-clinical studies of spasticity in mice including the current experimental challenges for clinical translation.
Collapse
Affiliation(s)
- Frederique Wieters
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Neurology, Cologne, Germany
| | - Carolin Weiss Lucas
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Center of Neurosurgery, Cologne, Germany
| | - Matthias Gruhn
- Department for Animal Physiology, Institute for Zoology, Biocenter Cologne, University of Cologne
| | - Ansgar Büschges
- Department for Animal Physiology, Institute for Zoology, Biocenter Cologne, University of Cologne
| | - Gereon R Fink
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Neurology, Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Germany
| | - Markus Aswendt
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Neurology, Cologne, Germany; Cognitive Neuroscience, Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Germany.
| |
Collapse
|