1
|
Arizanovska D, Dallera CA, Folorunso OO, Bush GF, Frye JB, Doyle KP, Jagid JR, Wolosker H, Monaco BA, Cordeiro JG, Atkins CM, Griswold AJ, Liebl DJ. Cognitive dysfunction following brain trauma results from sex-specific reactivation of the developmental pruning processes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607610. [PMID: 39211262 PMCID: PMC11360988 DOI: 10.1101/2024.08.13.607610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cognitive losses resulting from severe brain trauma have long been associated with the focal region of tissue damage, leading to devastating functional impairment. For decades, researchers have focused on the sequelae of cellular alterations that exist within the perilesional tissues; however, few clinical trials have been successful. Here, we employed a mouse brain injury model that resulted in expansive synaptic damage to regions outside the focal injury. Our findings demonstrate that synaptic damage results from the prolonged increase in D-serine release from activated microglia and astrocytes, which leads to hyperactivation of perisynaptic NMDARs, tagging of damaged synapses by complement components, and the reactivation of developmental pruning processes. We show that this mechanistic pathway is reversible at several stages within a prolonged and progressive period of synaptic loss. Importantly, these key factors are present in acutely injured brain tissue acquired from patients with brain injury, which supports a therapeutic neuroprotective strategy.
Collapse
|
2
|
Strong TA, Esquivel J, Wang Q, Ledon PJ, Wang H, Gaidosh G, Tse D, Pelaez D. Activation of multiple Eph receptors on neuronal membranes correlates with the onset of optic neuropathy. EYE AND VISION (LONDON, ENGLAND) 2023; 10:42. [PMID: 37779186 PMCID: PMC10544557 DOI: 10.1186/s40662-023-00359-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023]
Abstract
BACKGROUND Optic neuropathy is a major cause of irreversible blindness, yet the molecular determinants that contribute to neuronal demise have not been fully elucidated. Several studies have identified 'ephrin signaling' as one of the most dysregulated pathways in the early pathophysiology of optic neuropathy with varied etiologies. Developmentally, gradients in ephrin signaling coordinate retinotopic mapping via repulsive modulation of cytoskeletal dynamics in neuronal membranes. Little is known about the role ephrin signaling plays in the post-natal visual system and its correlation with the onset of optic neuropathy. METHODS Postnatal mouse retinas were collected for mass spectrometry analysis for erythropoietin-producing human hepatocellular (Eph) receptors. Optic nerve crush (ONC) model was employed to induce optic neuropathy, and proteomic changes during the acute phase of neuropathic onset were analyzed. Confocal and super-resolution microscopy determined the cellular localization of activated Eph receptors after ONC injury. Eph receptor inhibitors assessed the neuroprotective effect of ephrin signaling modulation. RESULTS Mass spectrometry revealed expression of seven Eph receptors (EphA2, A4, A5, B1, B2, B3, and B6) in postnatal mouse retinal tissue. Immunoblotting analysis indicated a significant increase in phosphorylation of these Eph receptors 48 h after ONC. Confocal microscopy demonstrated the presence of both subclasses of Eph receptors within the retina. Stochastic optical reconstruction microscopy (STORM) super-resolution imaging combined with optimal transport colocalization analysis revealed a significant co-localization of activated Eph receptors with injured neuronal cells, compared to uninjured neuronal and/or injured glial cells, 48 h post-ONC. Eph receptor inhibitors displayed notable neuroprotective effects for retinal ganglion cells (RGCs) after six days of ONC injury. CONCLUSIONS Our findings demonstrate the functional presence of diverse Eph receptors in the postnatal mammalian retina, capable of modulating multiple biological processes. Pan-Eph receptor activation contributes to the onset of neuropathy in optic neuropathies, with preferential activation of Eph receptors on neuronal processes in the inner retina following optic nerve injury. Notably, Eph receptor activation precedes neuronal loss. We observed a neuroprotective effect on RGCs upon inhibiting Eph receptors. Our study highlights the importance of investigating this repulsive pathway in early optic neuropathies and provides a comprehensive characterization of the receptors present in the developed retina of mice, relevant to both homeostasis and disease processes.
Collapse
Affiliation(s)
- Thomas A Strong
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL, 33136, USA
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, USA
| | - Juan Esquivel
- Department of Physics, University of Florida College of Liberal Arts and Sciences, Gainesville, FL, USA
| | - Qikai Wang
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL, 33136, USA
| | - Paul J Ledon
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL, 33136, USA
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Hua Wang
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL, 33136, USA
| | - Gabriel Gaidosh
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David Tse
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL, 33136, USA
| | - Daniel Pelaez
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, 1638 NW 10th Avenue, Miami, FL, 33136, USA.
- Department of Biomedical Engineering, University of Miami College of Engineering, University of Miami, Coral Gables, FL, USA.
- Department of Cell Biology, University of Miami Miller School of Medicine, Miami, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
3
|
Strong TA, Esquivel J, Wang Q, Ledon PJ, Wang H, Gaidosh G, Tse D, Pelaez D. Activation of Multiple Eph Receptors on Neuronal Membranes Correlates with The Onset of Traumatic Optic Neuropathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.05.543735. [PMID: 37333178 PMCID: PMC10274644 DOI: 10.1101/2023.06.05.543735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Background Optic neuropathy (ON) is a major cause of irreversible blindness, yet the molecular determinants that contribute to neuronal demise have not been fully elucidated. Several studies have identified 'ephrin signaling' as one of the most dysregulated pathways in the early pathophysiology of ON with varied etiologies. Developmentally, gradients in ephrin signaling coordinate retinotopic mapping via repulsive modulation of cytoskeletal dynamics in neuronal membranes. Little is known about the role ephrin signaling played in the post-natal visual system and its correlation with the onset of optic neuropathy. Methods Postnatal mouse retinas were collected for mass spectrometry analysis for Eph receptors. Optic nerve crush (ONC) model was employed to induce optic neuropathy, and proteomic changes during the acute phase of neuropathic onset were analyzed. Confocal and super-resolution microscopy determined the cellular localization of activated Eph receptors after ONC injury. Eph receptor inhibitors assessed the neuroprotective effect of ephrin signaling modulation. Results Mass spectrometry revealed expression of seven Eph receptors (EphA2, A4, A5, B1, B2, B3, and B6) in postnatal mouse retinal tissue. Immunoblotting analysis indicated a significant increase in phosphorylation of these Eph receptors 48 hours after ONC. Confocal microscopy demonstrated the presence of both subclasses of Eph receptors in the inner retinal layers. STORM super-resolution imaging combined with optimal transport colocalization analysis revealed a significant co-localization of activated Eph receptors with injured neuronal processes, compared to uninjured neuronal and/or injured glial cells, 48 hours post-ONC. Eph receptor inhibitors displayed notable neuroprotective effects after 6 days of ONC injury. Conclusions Our findings demonstrate the functional presence of diverse Eph receptors in the postnatal mammalian retina, capable of modulating multiple biological processes. Pan-Eph receptor activation contributes to the onset of neuropathy in ONs, with preferential activation of Eph receptors on neuronal processes in the inner retina following optic nerve injury. Notably, Eph receptor activation precedes neuronal loss. We observed neuroprotective effects upon inhibiting Eph receptors. Our study highlights the importance of investigating this repulsive pathway in early optic neuropathies and provides a comprehensive characterization of the receptors present in the developed retina of mice, relevant to both homeostasis and disease processes.
Collapse
Affiliation(s)
- Thomas A. Strong
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Cell Biology, University of Miami Miller School of Medicine
| | - Juan Esquivel
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Qikai Wang
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Paul J. Ledon
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Hua Wang
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Gabriel Gaidosh
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - David Tse
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Daniel Pelaez
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Dr. Nasser Al-Rashid Orbital Vision Research Center, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL, United States of America
- Department of Biomedical Engineering, University of Miami College of Engineering, University of Miami, Coral Gables, FL, United States of America
- Department of Cell Biology, University of Miami Miller School of Medicine
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States of America
| |
Collapse
|
4
|
Yang M, Wu Y, Yang XB, Liu T, Zhang Y, Zhuo Y, Luo Y, Zhang N. Establishing a prediction model of severe acute mountain sickness using machine learning of support vector machine recursive feature elimination. Sci Rep 2023; 13:4633. [PMID: 36944699 PMCID: PMC10030784 DOI: 10.1038/s41598-023-31797-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023] Open
Abstract
Severe acute mountain sickness (sAMS) can be life-threatening, but little is known about its genetic basis. The study was aimed to explore the genetic susceptibility of sAMS for the purpose of prediction, using microarray data from 112 peripheral blood mononuclear cell (PBMC) samples of 21 subjects, who were exposed to very high altitude (5260 m), low barometric pressure (406 mmHg), and hypobaric hypoxia (VLH) at various timepoints. We found that exposure to VLH activated gene expression in leukocytes, resulting in an inverted CD4/CD8 ratio that interacted with other phenotypic risk factors at the genetic level. A total of 2286 underlying risk genes were input into the support vector machine recursive feature elimination (SVM-RFE) system for machine learning, and a model with satisfactory predictive accuracy and clinical applicability was established for sAMS screening using ten featured genes with significant predictive power. Five featured genes (EPHB3, DIP2B, RHEBL1, GALNT13, and SLC8A2) were identified upstream of hypoxia- and/or inflammation-related pathways mediated by microRNAs as potential biomarkers for sAMS. The established prediction model of sAMS holds promise for clinical application as a genetic screening tool for sAMS.
Collapse
Affiliation(s)
- Min Yang
- Department of Traditional Chinese Medicine, Rheumatology Center of Integrated Medicine, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China.
| | - Yang Wu
- Department of Traditional Chinese Medicine, Rheumatology Center of Integrated Medicine, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China
| | - Xing-Biao Yang
- Department of Traditional Chinese Medicine, Rheumatology Center of Integrated Medicine, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China
| | - Tao Liu
- Department of Traditional Chinese Medicine, Rheumatology Center of Integrated Medicine, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China
| | - Ya Zhang
- Department of Traditional Chinese Medicine, Rheumatology Center of Integrated Medicine, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China
| | - Yue Zhuo
- Department of Traditional Chinese Medicine, Rheumatology Center of Integrated Medicine, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China
| | - Yong Luo
- Department of Traditional Chinese Medicine, Rheumatology Center of Integrated Medicine, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China
| | - Nan Zhang
- Department of Hematology, The General Hospital of Western Theater Command, PLA, Chengdu, 610083, China
| |
Collapse
|
5
|
Fessel J. Supplementary Pharmacotherapy for the Behavioral Abnormalities Caused by Stressors in Humans, Focused on Post-Traumatic Stress Disorder (PTSD). J Clin Med 2023; 12:1680. [PMID: 36836215 PMCID: PMC9967886 DOI: 10.3390/jcm12041680] [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: 01/14/2023] [Revised: 02/17/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023] Open
Abstract
Used as a supplement to psychotherapy, pharmacotherapy that addresses all of the known metabolic and genetic contributions to the pathogenesis of psychiatric conditions caused by stressors would require an inordinate number of drugs. Far simpler is to address the abnormalities caused by those metabolic and genetic changes in the cell types of the brain that mediate the behavioral abnormality. Relevant data regarding the changed brain cell types are described in this article and are derived from subjects with the paradigmatic behavioral abnormality of PTSD and from subjects with traumatic brain injury or chronic traumatic encephalopathy. If this analysis is correct, then therapy is required that benefits all of the affected brain cell types; those are astrocytes, oligodendrocytes, synapses and neurons, endothelial cells, and microglia (the pro-inflammatory (M1) subtype requires switching to the anti-inflammatory (M2) subtype). Combinations are advocated using several drugs, erythropoietin, fluoxetine, lithium, and pioglitazone, that benefit all of the five cell types, and that should be used to form a two-drug combination, suggested as pioglitazone with either fluoxetine or lithium. Clemastine, fingolimod, and memantine benefit four of the cell types, and one chosen from those could be added to the two-drug combination to form a three-drug combination. Using low doses of chosen drugs will limit both toxicity and drug-drug interactions. A clinical trial is required to validate both the advocated concept and the choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA 94123, USA
| |
Collapse
|
6
|
Díaz MM, Tsenkina Y, Arizanovska D, Mehlen P, Liebl DJ. DCC/netrin-1 regulates cell death in oligodendrocytes after brain injury. Cell Death Differ 2023; 30:397-406. [PMID: 36456775 PMCID: PMC9950151 DOI: 10.1038/s41418-022-01091-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Hallmark pathological features of brain trauma are axonal degeneration and demyelination because myelin-producing oligodendrocytes (OLs) are particularly vulnerable to injury-induced death signals. To reveal mechanisms responsible for this OL loss, we examined a novel class of "death receptors" called dependence receptors (DepRs). DepRs initiate pro-death signals in the absence of their respective ligand(s), yet little is known about their role after injury. Here, we investigated whether the deleted in colorectal cancer (DCC) DepR contributes to OL loss after brain injury. We found that administration of its netrin-1 ligand is sufficient to block OL cell death. We also show that upon acute injury, DCC is upregulated while netrin-1 is downregulated in perilesional tissues. Moreover, after genetically silencing pro-death activity using DCCD1290N mutant mice, we observed greater OL survival, greater myelin integrity, and improved motor function. Our findings uncover a novel role for the netrin-1/DCC pathway in regulating OL loss in the traumatically injured brain.
Collapse
Affiliation(s)
- Madelen M Díaz
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Yanina Tsenkina
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dena Arizanovska
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory - Equipe labellisée 'La Ligue', LabEx DEVweCAN, Centre de Recherche en Cancérologie de Lyon, INSERM U1052-CNRS UMR5286, Centre Léon Bérard, Université de Lyon, Université de Lyon1, Lyon, France.
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
7
|
Fessel J. Formulating treatment of major psychiatric disorders: algorithm targets the dominantly affected brain cell-types. DISCOVER MENTAL HEALTH 2023; 3:3. [PMID: 37861813 PMCID: PMC10501034 DOI: 10.1007/s44192-022-00029-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 12/21/2022] [Indexed: 10/21/2023]
Abstract
BACKGROUND Pharmacotherapy for most psychiatric conditions was developed from serendipitous observations of benefit from drugs prescribed for different reasons. An algorithmic approach to formulating pharmacotherapy is proposed, based upon which combination of changed activities by brain cell-types is dominant for any particular condition, because those cell-types contain and surrogate for genetic, metabolic and environmental information, that has affected their function. The algorithm performs because functions of some or all the affected cell-types benefit from several available drugs: clemastine, dantrolene, erythropoietin, fingolimod, fluoxetine, lithium, memantine, minocycline, pioglitazone, piracetam, and riluzole PROCEDURES/FINDINGS: Bipolar disorder, major depressive disorder, schizophrenia, Alzheimer's disease, and post-traumatic stress disorder, illustrate the algorithm; for them, literature reviews show that no single combination of altered cell-types accounts for all cases; but they identify, for each condition, which combination occurs most frequently, i.e., dominates, as compared with other possible combinations. Knowing the dominant combination of altered cell-types in a particular condition, permits formulation of therapy with combinations of drugs taken from the above list. The percentage of patients who might benefit from that therapy, depends upon the frequency with which the dominant combination occurs in patients with that particular condition. CONCLUSIONS Knowing the dominant combination of changed cell types in psychiatric conditions, permits an algorithmically formulated, rationally-based treatment. Different studies of the same condition often produce discrepant results; all might be correct, because identical clinical phenotypes result from different combinations of impaired cell-types, thus producing different results. Clinical trials would validate both the proposed concept and choice of drugs.
Collapse
Affiliation(s)
- Jeffrey Fessel
- Department of Medicine, University of California, 2069 Filbert Street, San Francisco, CA, 94123, USA.
| |
Collapse
|
8
|
Wu T, Kou J, Li X, Diwu Y, Li Y, Cao DY, Wang R. Electroacupuncture alleviates traumatic brain injury by inhibiting autophagy via increasing IL-10 production and blocking the AMPK/mTOR signaling pathway in rats. Metab Brain Dis 2022; 38:921-932. [PMID: 36517637 DOI: 10.1007/s11011-022-01133-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/24/2022] [Indexed: 12/23/2022]
Abstract
Autophagy, switched by the AMPK/mTOR signaling, has been revealed to contribute greatly to traumatic brain injury (TBI). Electroacupuncture (EA) is a promising therapeutic method for TBI, however, the underlying mechanism is still unclear. Herein, we hypothesize that the therapeutic effect of EA on TBI is associated with its inhibition on AMPK/mTOR-mediated autophagy. Sprague-Dawley rats were randomly divided into three groups: sham, TBI, and TBI + EA. TBI model was established by using an electronic controlled cortical impactor. Rats were treated with EA at 12 h after modeling, 15 min daily for 14 consecutive days. EA was applied at the acupuncture points Quchi (LI 11), Hegu (LI4), Baihui (GV20), Guanyuan (CV4), Zusanli (ST36) and Yongquan (KI1), using dense-sparse wave, at frequencies of 1 Hz, and an amplitude of 1 mA. After 3, 7 and 14 days of modeling, the modified neurological severity scale (mNSS), rota rod system, and Morris Water Maze (MWM) test showed that EA treatment promoted neurological function recovery in TBI rats. Moreover, EA treatment alleviated brain edema, pathological damage, neuronal apoptosis in TBI rats. EA improved abnormal ultrastructure, including abnormal mitochondrial morphology and increased autophagosomes, in the brain neurons of TBI rats, as measured by transmission electron microscopy, and the concentration of adenosine triphosphate (ATP), adenosine diphosphate (ADP), and adenosine monophosphate (AMP). Western blot and immunohistochemistry (IHC) assays were performed to measure the protein levels of interleukin 10 (IL-10), autophagy-related proteins and key proteins in the AMPK/mTOR signaling pathway. EA treatment increased IL-10 production, inhibited the AMPK/mTOR signaling, and inhibited excessive autophagy in TBI rats. Additionally, AMPK inhibitor Compound C treatment had similar effects to EA. Both AMPK agonist AICAR and IL-10 neutralizing antibody treatments reversed the effects of EA on the related protein levels of autophagy and the AMPK/mTOR signaling pathway, and abolished the protective effects of EA on TBI rats. In conclusion, EA treatment promoted neurological function recovery and alleviated pathological damage and neuronal apoptosis in TBI rats through inhibiting excessive autophagy via increasing IL-10 production and blocking the AMPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Tao Wu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China
- College of Acupuncture and Tuina, Shaanxi University of Chinese Medicine, Xixian New Area, Shaanxi, 712046, People's Republic of China
| | - Jiushe Kou
- Pain Department, The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xixian New Area, Shaanxi, 712000, People's Republic of China
| | - Xuemei Li
- Orthopedics Department, The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xixian New Area, Shaanxi, 712000, People's Republic of China
| | - Yongchang Diwu
- Department of Clinical Medicine, The Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xixian New Area, Shaanxi Province, 712046, People's Republic of China
| | - Yuanyuan Li
- Scientific Research Department, The Second Affiliated Hospital, Shaanxi University of Chinese Medicine, Xixian New Area, Shaanxi, 712000, People's Republic of China
| | - Dong-Yuan Cao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi'an Jiaotong University College of Stomatology, 98 West 5th Road, Xi'an, Shaanxi, 710004, People's Republic of China.
| | - Ruihui Wang
- College of Acupuncture and Tuina, Shaanxi University of Chinese Medicine, Xixian New Area, Shaanxi, 712046, People's Republic of China.
| |
Collapse
|
9
|
Zhang S, Gan L, Cao F, Wang H, Gong P, Ma C, Ren L, Lin Y, Lin X. The barrier and interface mechanisms of the brain barrier, and brain drug delivery. Brain Res Bull 2022; 190:69-83. [PMID: 36162603 DOI: 10.1016/j.brainresbull.2022.09.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/25/2022] [Accepted: 09/20/2022] [Indexed: 11/26/2022]
Abstract
Three different barriers are formed between the cerebrovascular and the brain parenchyma: the blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the cerebrospinal fluid-brain barrier (CBB). The BBB is the main regulator of blood and central nervous system (CNS) material exchange. The semipermeable nature of the BBB limits the passage of larger molecules and hydrophilic small molecules, Food and Drug Administration (FDA)-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Although the complexity of the BBB affects CNS drug delivery, understanding the composition and function of the BBB can provide a platform for the development of new methods for CNS drug delivery. This review summarizes the classification of the brain barrier, the composition and role of the basic structures of the BBB, and the transport, barrier, and destruction mechanisms of the BBB; discusses the advantages and disadvantages of different drug delivery methods and prospects for future drug delivery strategies.
Collapse
Affiliation(s)
- Shanshan Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| | - Lin Gan
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Fengye Cao
- Yiyang The First Hospital of Traditional Chinese Medicine, Yiyang, Hunan Province, 413000, China
| | - Hao Wang
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Peng Gong
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Congcong Ma
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Li Ren
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Yubo Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China
| | - Xianming Lin
- Department of Neurobiology and Acupuncture Research, The Third Clinical Medical College, Zhejiang Chinese Medical University, Key Laboratory of Acupuncture and Neurology of Zhejiang Province, Hangzhou 310053, China.
| |
Collapse
|
10
|
Nishimura K, Cordeiro JG, Ahmed AI, Yokobori S, Gajavelli S. Advances in Traumatic Brain Injury Biomarkers. Cureus 2022; 14:e23804. [PMID: 35392277 PMCID: PMC8978594 DOI: 10.7759/cureus.23804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2022] [Indexed: 11/05/2022] Open
Abstract
Traumatic brain injury (TBI) is increasingly a major cause of disability across the globe. The current methods of diagnosis are inadequate at classifying patients and prognosis. TBI is a diagnostic and therapeutic challenge. There is no Food and Drug Administration (FDA)-approved treatment for TBI yet. It took about 16 years of preclinical research to develop accurate and objective diagnostic measures for TBI. Two brain-specific protein biomarkers, namely, ubiquitin C-terminal hydrolase-L1 and glial fibrillary acidic protein, have been extensively characterized. Recently, the two biomarkers were approved by the FDA as the first blood-based biomarker, Brain Trauma Indicator™ (BTI™), via the Breakthrough Devices Program. This scoping review presents (i) TBI diagnosis challenges, (ii) the process behind the FDA approval of biomarkers, and (iii) known unknowns in TBI biomarker biology. The current lag in TBI incidence and hospitalization can be reduced if digital biomarkers such as hard fall detection are standardized and used as a mechanism to alert paramedics to an unresponsive trauma patient.
Collapse
|
11
|
Lu Q, Xiong J, Yuan Y, Ruan Z, Zhang Y, Chai B, Li L, Cai S, Xiao J, Wu Y, Huang P, Zhang H. Minocycline improves the functional recovery after traumatic brain injury via inhibition of aquaporin-4. Int J Biol Sci 2022; 18:441-458. [PMID: 34975343 PMCID: PMC8692149 DOI: 10.7150/ijbs.64187] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/04/2021] [Indexed: 12/28/2022] Open
Abstract
Traumatic brain injury (TBI) is one of the main concerns worldwide as there is still no comprehensive therapeutic intervention. Astrocytic water channel aquaporin-4 (AQP-4) system is closely related to the brain edema, water transport at blood-brain barrier (BBB) and astrocyte function in the central nervous system (CNS). Minocycline, a broad-spectrum semisynthetic tetracycline antibiotic, has shown anti-inflammation, anti-apoptotic, vascular protection and neuroprotective effects on TBI models. Here, we tried to further explore the underlying mechanism of minocycline treatment for TBI, especially the relationship of minocycline and AQP4 during TBI treatment. In present study, we observed that minocycline efficaciously reduces the elevation of AQP4 in TBI mice. Furthermore, minocycline significantly reduced neuronal apoptosis, ameliorated brain edema and BBB disruption after TBI. In addition, the expressions of tight junction protein and astrocyte morphology alteration were optimized by minocycline administration. Similar results were found after treating with TGN-020 (an inhibitor of AQP4) in TBI mice. Moreover, these effects were reversed by cyanamide (CYA) treatment, which notably upregulated AQP4 expression level in vivo. In primary cultured astrocytes, small-interfering RNA (siRNA) AQP4 treatment prevented glutamate-induced astrocyte swelling. To sum up, our study suggests that minocycline improves the functional recovery of TBI through reducing AQP4 level to optimize BBB integrity and astrocyte function, and highlights that the AQP4 may be an important therapeutic target during minocycline treating for TBI.
Collapse
Affiliation(s)
- Qi Lu
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Jun Xiong
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yuan Yuan
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China.,Department of pharmacy, Hangzhou Red Cross Hospital, Zhejiang Province Hospital of Integrated Traditional Chinese and Western Medicine, 310003, Hangzhou, Zhejiang, China
| | - Zhanwei Ruan
- Department of Emergency, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yu Zhang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Bo Chai
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Lei Li
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Shufang Cai
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, 325035, Wenzhou, Zhejiang, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China
| | - Yanqing Wu
- The Institute of Life Sciences, Engineering Laboratory of Zhejiang Province for Pharmaceutical Development of Growth Factors, Biomedical Collaborative Innovation Center of Wenzhou, Wenzhou University, 325035, Wenzhou, Zhejiang, China
| | - Peng Huang
- Department of Pharmacy, Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, 325200, Wenzhou, Zhejiang, China
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Wound Repair and Regeneration Key Laboratory, Cixi Biomedical Research Institute, Wenzhou Medical University, 325000, Wenzhou, Zhejiang, China.,Department of Pharmacy, Zhuji People's Hospital, The Affiliated Hospital of Wenzhou Medical University, 311899, Shaoxing, Zhejiang, China
| |
Collapse
|
12
|
Yuan M, Wu H. Astrocytes in the Traumatic Brain Injury: the Good and the Bad. Exp Neurol 2021; 348:113943. [PMID: 34863998 DOI: 10.1016/j.expneurol.2021.113943] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 11/08/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022]
Abstract
Astrocytes control many processes of the nervous system in health and disease, and respond to injury quickly. Astrocytes produce neuroprotective factors in the injured brain to clear cellular debris and to orchestrate neurorestorative processes that are beneficial for neurological recovery after traumatic brain injury (TBI). However, astrocytes also become dysregulated and produce cytotoxic mediators that hinder CNS repair by induction of neuronal dysfunction and cell death. Hence, we discuss the potential role of astrocytes in neuropathological processes such as neuroinflammation, neurogenesis, synaptogenesis and blood-brain barrier repair after TBI. Thus, an improved understanding of the dual role of astrocytes may advance our knowledge of post-brain injury recovery, and provide opportunities for the development of novel therapeutic strategies for TBI.
Collapse
Affiliation(s)
- Mengqi Yuan
- Institute of Neuroscience, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Haitao Wu
- Beijing Institute of Basic Medical Sciences, 100850 Beijing, China; Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, Jiangsu, China; Chinese Institute for Brain Research (CIBR), 102206 Beijing, China.
| |
Collapse
|
13
|
Guo P, Jin Z, Wang J, Sang A, Wu H. Irisin Rescues Blood-Brain Barrier Permeability following Traumatic Brain Injury and Contributes to the Neuroprotection of Exercise in Traumatic Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:1118981. [PMID: 34697562 PMCID: PMC8541859 DOI: 10.1155/2021/1118981] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 08/21/2021] [Accepted: 09/28/2021] [Indexed: 11/17/2022]
Abstract
Traumatic brain injury (TBI) has a high incidence, mortality, and morbidity all over the world. One important reason for its poor clinical prognosis is brain edema caused by blood-brain barrier (BBB) dysfunction after TBI. The mechanism may be related to the disorder of mitochondrial morphology and function of neurons in damaged brain tissue, the decrease of uncoupling protein 2 (UCP2) activity, and the increase of inflammatory reaction and oxidative stress. In this study, we aimed to investigate the effects of exogenous irisin on BBB dysfunction after TBI and its role in the neuroprotective effects of endurance exercise (EE) in mice. The concentrations of irisin in cerebrospinal fluid (CSF) and plasma of patients with mild to severe TBI were measured by ELISA. Then, male C57BL/6J mice and UCP2 knockout mice with C57BL/6J background were used to establish the TBI model. The BBB structure and permeability were examined by transmission electron microscopy and Evans blue extravasation, respectively. The protein expressions of irisin, occludin, claudin-5, zonula occludens-1 (ZO-1), nuclear factor E2-related factor 2(Nrf2), quinine oxidoreductase (NQO-1), hemeoxygenase-1 (HO-1), cytochrome C (Cyt-C), cytochrome C oxidase (COX IV), BCL2-associated X protein (Bax), cleaved caspase-3, and UCP2 were detected by western blot. The production of reactive oxygen species (ROS) was evaluated by the dihydroethidium (DHE) staining. The levels of inflammatory factors were detected by ELISA. In this study, we found that the CSF irisin levels were positively correlated with the severity of disease in patients with TBI and both EE and exogenous irisin could reduce BBB damage in a mouse model of TBI. In addition, we used UCP2-/- mice and further found that irisin could improve the dysfunction of BBB after TBI by promoting the expression of UCP2 on the mitochondrial membrane of neurons, reducing the damage of mitochondrial structure and function, thus alleviating the inflammatory response and oxidative stress. In conclusion, the results of this study suggested that irisin might alleviate brain edema after TBI by promoting the expression of UCP2 on the mitochondrial membrane of neurons and contribute to the neuroprotection of EE against TBI.
Collapse
Affiliation(s)
- Peipei Guo
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province 230022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan City, Hubei Province 430071, China
| | - Zhao Jin
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan City, Hubei Province 430071, China
| | - Jin Wang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan City, Hubei Province 430071, China
| | - Aming Sang
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan City, Hubei Province 430071, China
| | - Huisheng Wu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province 230022, China
- Department of Anesthesiology, Zhongnan Hospital of Wuhan University, Wuhan City, Hubei Province 430071, China
| |
Collapse
|
14
|
Clark IC, Gutiérrez-Vázquez C, Wheeler MA, Li Z, Rothhammer V, Linnerbauer M, Sanmarco LM, Guo L, Blain M, Zandee SEJ, Chao CC, Batterman KV, Schwabenland M, Lotfy P, Tejeda-Velarde A, Hewson P, Manganeli Polonio C, Shultis MW, Salem Y, Tjon EC, Fonseca-Castro PH, Borucki DM, Alves de Lima K, Plasencia A, Abate AR, Rosene DL, Hodgetts KJ, Prinz M, Antel JP, Prat A, Quintana FJ. Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science 2021; 372:372/6540/eabf1230. [PMID: 33888612 DOI: 10.1126/science.abf1230] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/27/2021] [Accepted: 03/12/2021] [Indexed: 12/11/2022]
Abstract
Cell-cell interactions control the physiology and pathology of the central nervous system (CNS). To study astrocyte cell interactions in vivo, we developed rabies barcode interaction detection followed by sequencing (RABID-seq), which combines barcoded viral tracing and single-cell RNA sequencing (scRNA-seq). Using RABID-seq, we identified axon guidance molecules as candidate mediators of microglia-astrocyte interactions that promote CNS pathology in experimental autoimmune encephalomyelitis (EAE) and, potentially, multiple sclerosis (MS). In vivo cell-specific genetic perturbation EAE studies, in vitro systems, and the analysis of MS scRNA-seq datasets and CNS tissue established that Sema4D and Ephrin-B3 expressed in microglia control astrocyte responses via PlexinB2 and EphB3, respectively. Furthermore, a CNS-penetrant EphB3 inhibitor suppressed astrocyte and microglia proinflammatory responses and ameliorated EAE. In summary, RABID-seq identified microglia-astrocyte interactions and candidate therapeutic targets.
Collapse
Affiliation(s)
- Iain C Clark
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Bioengineering, University of California, Berkeley, California Institute for Quantitative Biosciences, Berkeley, CA 94720, USA
| | - Cristina Gutiérrez-Vázquez
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Zhaorong Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Veit Rothhammer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Mathias Linnerbauer
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Neurology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Liliana M Sanmarco
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Lydia Guo
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Manon Blain
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Stephanie E J Zandee
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Chun-Cheih Chao
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Katelyn V Batterman
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Marius Schwabenland
- Institute of Neuropathology, University of Freiburg, D-79106 Freiburg, Germany
| | - Peter Lotfy
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amalia Tejeda-Velarde
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Patrick Hewson
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Carolina Manganeli Polonio
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Michael W Shultis
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yasmin Salem
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Emily C Tjon
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro H Fonseca-Castro
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Davis M Borucki
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kalil Alves de Lima
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Agustin Plasencia
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Adam R Abate
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California Institute for Quantitative Biosciences, San Francisco, CA 94158, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Douglas L Rosene
- Department of Anatomy and Neurobiology, Boston University School of Medicine, Boston, MA 02118, USA
| | - Kevin J Hodgetts
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, D-79106 Freiburg, Germany.,Signaling Research Centres BIOSS and CIBSS, University of Freiburg, D-79106 Freiburg, Germany.,Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, D-79106 Freiburg, Germany
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Alexandre Prat
- Neuroimmunology Research Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC H2X 0A9, Canada
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
15
|
Brain microvascular damage linked to a moderate level of strain induced by controlled cortical impact. J Biomech 2021; 122:110452. [PMID: 33901935 DOI: 10.1016/j.jbiomech.2021.110452] [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: 12/30/2020] [Revised: 03/30/2021] [Accepted: 04/09/2021] [Indexed: 01/10/2023]
Abstract
Cerebral blood vessels play an important role in brain metabolic activity in general and following traumatic brain injury (TBI) in particular. However, the extent to which TBI alters microvessel structure is not well understood. Specifically, how intracranial mechanical responses produced during impacts relate to vascular damage needs to be better studied. Therefore, the objective of this study was to investigate the biomechanical mechanisms and thresholds of brain microvascular injury. Detailed microvascular damage of mouse brain was quantified using Arterial Spin Labeling (ASL) magnetic resonance imaging (MRI) and ex vivo Serial Two-Photon Tomography (STPT) in seven mice that had undergone controlled cortical impact. Mechanical strains were investigated through finite element (FE) modeling of the mouse brain. We then compared the post-injury vessel density map with FE-predicted strain and found a moderate correlation between the vessel length density and the predicted peak maximum principal strains (MPS) (R2 = 0.52). High MPS was observed at the impact regions with low vessel length density, supporting the mechanism of strain-triggered microvascular damage. Using logistic regression, the MPS corresponding to a 50% probability of injury was found to be 0.17. Given the literature reporting MPS of over 0.2 in the human brain for mild TBI/concussion cases, it is highly recommended to consider microvascular damage when investigating mild TBI/concussion in the future.
Collapse
|
16
|
Tsenkina Y, Tapanes SA, Díaz MM, Titus DJ, Gajavelli S, Bullock R, Atkins CM, Liebl DJ. EphB3 interacts with initiator caspases and FHL-2 to activate dependence receptor cell death in oligodendrocytes after brain injury. Brain Commun 2020; 2:fcaa175. [PMID: 33305261 PMCID: PMC7713998 DOI: 10.1093/braincomms/fcaa175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 09/09/2020] [Accepted: 09/17/2020] [Indexed: 11/16/2022] Open
Abstract
Clinical trials examining neuroprotective strategies after brain injury, including those targeting cell death mechanisms, have been underwhelming. This may be in part due to an incomplete understanding of the signalling mechanisms that induce cell death after traumatic brain injury. The recent identification of a new family of death receptors that initiate pro-cell death signals in the absence of their ligand, called dependence receptors, provides new insight into the factors that contribute to brain injury. Here, we show that blocking the dependence receptor signalling of EphB3 improves oligodendrocyte cell survival in a murine controlled cortical impact injury model, which leads to improved myelin sparing, axonal conductance and behavioural recovery. EphB3 also functions as a cysteine-aspartic protease substrate, where the recruitment of injury-dependent adaptor protein Dral/FHL-2 together with capsase-8 or -9 leads to EphB3 cleavage to initiate cell death signals in murine and human traumatic brain-injured patients, supporting a conserved mechanism of cell death. These pro-apoptotic responses can be blocked via exogenous ephrinB3 ligand administration leading to improved oligodendrocyte survival. In short, our findings identify a novel mechanism of oligodendrocyte cell death in the traumatically injured brain that may reflect an important neuroprotective strategy in patients.
Collapse
Affiliation(s)
- Yanina Tsenkina
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Stephen A Tapanes
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Madelen M Díaz
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David J Titus
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Shyam Gajavelli
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Ross Bullock
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Coleen M Atkins
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Daniel J Liebl
- The Miami Project to Cure Paralysis, Department of Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
17
|
Liu KL, Yu XJ, Sun TZ, Wang YC, Chen MX, Su YW, Zhang HC, Chen YM, Gao HL, Shi XL, Qi J, Li Y, Li HB, Dong WJ, He JK, Kang YM. Effects of seawater immersion on open traumatic brain injury in rabbit model. Brain Res 2020; 1743:146903. [PMID: 32445716 DOI: 10.1016/j.brainres.2020.146903] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 05/10/2020] [Accepted: 05/19/2020] [Indexed: 11/19/2022]
Abstract
We emulated instances of open traumatic brain injuries (TBI) in a maritime disaster. New Zealand rabbit animal models were used to evaluate the pathophysiological changes in open TBI with and without the influence of artificial seawater. New Zealand rabbits were randomly divided into 3 groups. Control group consisted of only normal animals. Animals in TBI and TBI + Seawater groups underwent craniotomy with dura mater incised and brain tissue exposed to free-fall impact. Afterward, only TBI + Seawater group received on-site artificial seawater infusion. Brain water content (BWC) and permeability of blood-brain barrier (BBB) were assessed. Reactive oxygen species levels were measured. Western blotting and immunofluorescence were employed to detect: apoptosis-related factors Caspase-3, Bax and Bcl-2; angiogenesis-related factors CD31 and CD34; astrogliosis-related factor glial fibrillary acidic protein (GFAP); potential neuron injury indicator neuron-specific enolase (NSE). Hematoxylin & eosin, Masson-trichrome and Nissl stainings were performed for pathological observations. Comparing to Control group, TBI group manifested abnormal neuronal morphology; increased BWC; compromised BBB integrity; increased ROS, Bax, CD31, CD34, Caspase-3 and GFAP expressions; decreased Bcl-2 and NSE expression. Seawater immersion caused all changes, except BWC, to become more significant. Seawater immersion worsens the damage inflicted to brain tissue by open TBI. It aggravates hypoxia in brain tissue, upregulates ROS expression, increases neuron sensitivity to apoptosis-inducing factors, and promotes angiogenesis as well as astrogliosis.
Collapse
Affiliation(s)
- Kai-Li Liu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Tian-Ze Sun
- Department of Human Anatomy and Histology and Embryology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi an 710061, China
| | - Yi-Chang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Meng-Xuan Chen
- College of Stomatology, Xi'an Jiaotong University, Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, 98 XiWu Road, Xi'an, Shaanxi 710004, People's Republic of China
| | - Yan-Wen Su
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China
| | - Hao-Chen Zhang
- School of Clinical Medicine, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yan-Mei Chen
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Hong-Li Gao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Xiao-Lian Shi
- Department of Pharmacology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an 710061, China
| | - Jie Qi
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Ying Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Hong-Bao Li
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China
| | - Wei-Jiang Dong
- Department of Human Anatomy and Histology and Embryology, Xi'an Jiaotong University School of Basic Medical Sciences, Xi an 710061, China.
| | - Jian-Kang He
- State Key Laboratory for Manufacturing Systems Engineering, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China, Xi an 710061, China.
| |
Collapse
|
18
|
Vreeken D, Zhang H, van Zonneveld AJ, van Gils JM. Ephs and Ephrins in Adult Endothelial Biology. Int J Mol Sci 2020; 21:ijms21165623. [PMID: 32781521 PMCID: PMC7460586 DOI: 10.3390/ijms21165623] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/19/2022] Open
Abstract
Eph receptors and their ephrin ligands are important guidance molecules during neurological and vascular development. In recent years, it has become clear that the Eph protein family remains functional in adult physiology. A subset of Ephs and ephrins is highly expressed by endothelial cells. As endothelial cells form the first barrier between the blood and surrounding tissues, maintenance of a healthy endothelium is crucial for tissue homeostasis. This review gives an overview of the current insights of the role of ephrin ligands and receptors in endothelial function and leukocyte recruitment in the (patho)physiology of adult vascular biology.
Collapse
|
19
|
Cash A, Theus MH. Mechanisms of Blood-Brain Barrier Dysfunction in Traumatic Brain Injury. Int J Mol Sci 2020; 21:ijms21093344. [PMID: 32397302 PMCID: PMC7246537 DOI: 10.3390/ijms21093344] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/02/2020] [Accepted: 05/04/2020] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injuries (TBIs) account for the majority of injury-related deaths in the United States with roughly two million TBIs occurring annually. Due to the spectrum of severity and heterogeneity in TBIs, investigation into the secondary injury is necessary in order to formulate an effective treatment. A mechanical consequence of trauma involves dysregulation of the blood–brain barrier (BBB) which contributes to secondary injury and exposure of peripheral components to the brain parenchyma. Recent studies have shed light on the mechanisms of BBB breakdown in TBI including novel intracellular signaling and cell–cell interactions within the BBB niche. The current review provides an overview of the BBB, novel detection methods for disruption, and the cellular and molecular mechanisms implicated in regulating its stability following TBI.
Collapse
Affiliation(s)
- Alison Cash
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA;
| | - Michelle H. Theus
- The Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA;
- The Center for Regenerative Medicine, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA 24061, USA
- Correspondence: ; Tel.: 1-540-231-0909; Fax: 1-540-231-7425
| |
Collapse
|
20
|
Withaferin A alleviates traumatic brain injury induced secondary brain injury via suppressing apoptosis in endothelia cells and modulating activation in the microglia. Eur J Pharmacol 2020; 874:172988. [PMID: 32032599 DOI: 10.1016/j.ejphar.2020.172988] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 02/01/2020] [Accepted: 02/03/2020] [Indexed: 01/06/2023]
Abstract
Traumatic brain injury (TBI) is a major public health concern with high rates of morbidity and mortality worldwide. Currently used medications, though effective, are also associated with several adverse effects. Development of effective neuroprotective agents with fewer side-effects would be of clinical value. Previous studies have shown that withaferin compounds have a potential neuroprotective effect in nervous system disorders. However, the effect of withaferin compounds, especially withaferin A (WFA), on traumatic brain injury is unclear. In the present study, both in vivo and in vitro models were used to assess whether WFA could exert a neuroprotective effect after TBI and were used to explore the associated mechanisms. The results showed that WFA significantly improved neurobehavioral function in a dose-dependent fashion and alleviated histological alteration of injury to tissues in TBI mice. In vitro models of TBI revealed that dose-dependent WFA treatment increased the viability of SH-SY5Y cells. In addition, WFA treatment could attenuate blood-brain barrier disruption and brain edema via suppressing apoptosis in endothelial cells. Furthermore, both our in vivo and in vitro results reveal that WFA treatment could significantly reduce levels of several neuroinflammation cytokines (IL-1β, IL-6, and TNF-α), which correlate with an overall reduction in microglial activation. These data suggest that the neuroprotection by WFA is, at least in part, related to regulation of microglial activation and inhibition of vascular endothelial cell apoptosis. Taken together, these findings support further investigation of WFA as a promising therapeutic agent for promoting functional recovery after traumatic brain injury.
Collapse
|
21
|
Acute and chronic stage adaptations of vascular architecture and cerebral blood flow in a mouse model of TBI. Neuroimage 2019; 202:116101. [DOI: 10.1016/j.neuroimage.2019.116101] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/12/2019] [Accepted: 08/14/2019] [Indexed: 11/18/2022] Open
|
22
|
Rui Q, Ni H, Lin X, Zhu X, Li D, Liu H, Chen G. Astrocyte-derived fatty acid-binding protein 7 protects blood-brain barrier integrity through a caveolin-1/MMP signaling pathway following traumatic brain injury. Exp Neurol 2019; 322:113044. [PMID: 31454490 DOI: 10.1016/j.expneurol.2019.113044] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 08/15/2019] [Accepted: 08/23/2019] [Indexed: 11/19/2022]
Abstract
The astrocyte-endothelial cell interaction is crucial for normal brain homeostasis and blood-brain barrier (BBB) disruption in pathological conditions. However, the mechanism by which astrocytes control BBB integrity, especially after traumatic brain injury (TBI), remains unclear. Here, we present evidence that astrocyte-derived fatty acid-binding protein 7 (FABP7), a differentiation- and migration-associated molecule, may function as a modulator of BBB permeability in a rat weight-drop model of TBI. Immunohistochemical analysis revealed that TBI induced increased expression of FABP7 in astrocytes, accompanied by caveolin-1 (Cav-1) upregulation in endothelial cells. Administration of recombinant FABP7 significantly ameliorated TBI-induced neurological deficits, brain edema, and BBB permeability, concomitant with upregulation of endothelial Cav-1 and tight junction protein expression, while FABP7 knockdown resulted in the opposite effects. Furthermore, pretreatment with daidzein, a specific inhibitor of Cav-1, reversed the inhibitory effects of recombinant FABP7 on matrix metalloproteinase (MMP)-2/9 expression and abolished its BBB protection after TBI. Altogether, these findings suggest that astrocyte-derived FABP7 upregulation may represent an endogenous protective response to BBB disruption partly mediated through a Cav-1/MMP signaling pathway following TBI.
Collapse
Affiliation(s)
- Qin Rui
- Department of Laboratory, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215006, China
| | - Haibo Ni
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215006, China
| | - Xiaolong Lin
- Department of Orthopaedics, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215006, China
| | - Xiaojue Zhu
- Department of Laboratory, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215006, China
| | - Di Li
- Department of Translational Medicine Center, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215006, China
| | - Huixiang Liu
- Department of Neurosurgery, The Affiliated Zhangjiagang Hospital of Soochow University, Suzhou 215006, China.
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| |
Collapse
|
23
|
Angiopoietin/Tie2 Axis Regulates the Age-at-Injury Cerebrovascular Response to Traumatic Brain Injury. J Neurosci 2018; 38:9618-9634. [PMID: 30242049 DOI: 10.1523/jneurosci.0914-18.2018] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/15/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022] Open
Abstract
Although age-at-injury influences chronic recovery from traumatic brain injury (TBI), the differential effects of age on early outcome remain understudied. Using a male murine model of moderate contusion injury, we investigated the underlying mechanism(s) regulating the distinct response between juvenile and adult TBI. We demonstrate similar biomechanical and physical properties of naive juvenile and adult brains. However, following controlled cortical impact (CCI), juvenile mice displayed reduced cortical lesion formation, cell death, and behavioral deficits at 4 and 14 d. Analysis of high-resolution laser Doppler imaging showed a similar loss of cerebral blood flow (CBF) in the ipsilateral cortex at 3 and 24 h post-CCI, whereas juvenile mice showed enhanced subsequent restoration at 2-4 d compared with adults. These findings correlated with reduced blood-brain barrier (BBB) disruption and increased perilesional vessel density. To address whether an age-dependent endothelial cell (EC) response affects vessel stability and tissue outcome, we magnetically isolated CD31+ ECs from sham and injured cortices and evaluated mRNA expression. Interestingly, we found increased transcripts for BBB stability-related genes and reduced expression of BBB-disrupting genes in juveniles compared with adults. These differences were concomitant with significant changes in miRNA-21-5p and miR-148a levels. Accompanying these findings was robust GFAP immunoreactivity, which was not resolved by day 35. Importantly, pharmacological inhibition of EC-specific Tie2 signaling abolished the juvenile protective effects. These findings shed new mechanistic light on the divergent effects that age plays on acute TBI outcome that are both spatial and temporal dependent.SIGNIFICANCE STATEMENT Although a clear "window of susceptibility" exists in the developing brain that could deter typical developmental trajectories if exposed to trauma, a number of preclinical models have demonstrated evidence of early recovery in younger patients. Our findings further demonstrate acute neuroprotection and improved restoration of cerebral blood flow in juvenile mice subjected to cortical contusion injury compared with adults. We also demonstrate a novel role for endothelial cell-specific Tie2 signaling in this age-related response, which is known to promote barrier stability, is heightened in the injured juvenile vasculature, and may be exploited for therapeutic interventions across the age spectrum following traumatic brain injury.
Collapse
|