1
|
Hasanpour-Segherlou Z, Masheghati F, Shakeri-Darzehkanani M, Hosseini-Siyanaki MR, Lucke-Wold B. Neurodegenerative Disorders in the Context of Vascular Changes after Traumatic Brain Injury. JOURNAL OF VASCULAR DISEASES 2024; 3:319-332. [DOI: 10.3390/jvd3030025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2025]
Abstract
Traumatic brain injury (TBI) results from external biomechanical forces that cause structural and physiological disturbances in the brain, leading to neuronal, axonal, and vascular damage. TBIs are predominantly mild (65%), with moderate (10%) and severe (25%) cases also prevalent. TBI significantly impacts health, increasing the risk of neurodegenerative diseases such as dementia, post injury. The initial phase of TBI involves acute disruption of the blood–brain barrier (BBB) due to vascular shear stress, leading to ischemic damage and amyloid-beta accumulation. Among the acute cerebrovascular changes after trauma are early progressive hemorrhage, micro bleeding, coagulopathy, neurovascular unit (NVU) uncoupling, changes in the BBB, changes in cerebral blood flow (CBF), and cerebral edema. The secondary phase is characterized by metabolic dysregulation and inflammation, mediated by oxidative stress and reactive oxygen species (ROS), which contribute to further neurodegeneration. The cerebrovascular changes and neuroinflammation include excitotoxicity from elevated extracellular glutamate levels, coagulopathy, NVU, immune responses, and chronic vascular changes after TBI result in neurodegeneration. Severe TBI often leads to dysfunction in organs outside the brain, which can significantly impact patient care and outcomes. The vascular component of systemic inflammation after TBI includes immune dysregulation, hemodynamic dysfunction, coagulopathy, respiratory failure, and acute kidney injury. There are differences in how men and women acquire traumatic brain injuries, how their brains respond to these injuries at the cellular and molecular levels, and in their brain repair and recovery processes. Also, the patterns of cerebrovascular dysfunction and stroke vulnerability after TBI are different in males and females based on animal studies.
Collapse
Affiliation(s)
| | | | | | | | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
2
|
Singh A, Gong S, Vu A, Li S, Obenaus A. Social deficits mirror delayed cerebrovascular dysfunction after traumatic brain injury. Acta Neuropathol Commun 2024; 12:126. [PMID: 39107831 PMCID: PMC11304659 DOI: 10.1186/s40478-024-01840-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/28/2024] [Indexed: 08/10/2024] Open
Abstract
Traumatic brain injury (TBI) survivors face debilitating long-term psychosocial consequences, including social isolation and depression. TBI modifies neurovascular physiology and behavior but the chronic physiological implications of altered brain perfusion on social interactions are unknown. Adult C57/BL6 male mice received a moderate cortical TBI, and social behaviors were assessed at baseline, 3-, 7-, 14-, 30-, and 60-days post injury (dpi). Magnetic resonance imaging (MRI, 9.4T) using dynamic susceptibility contrast perfusion weighted MRI were acquired. At 60dpi mice underwent histological angioarchitectural mapping. Analysis utilized standardized protocols followed by cross-correlation metrics. Social behavior deficits at 60dpi emerged as reduced interactions with a familiar cage-mate (partner) that mirrored significant reductions in cerebral blood flow (CBF) at 60dpi. CBF perturbations were dynamic temporally and across brain regions including regions known to regulate social behavior such as hippocampus, hypothalamus, and rhinal cortex. Social isolation in TBI-mice emerged with a significant decline in preference to spend time with a cage mate. Cortical vascular density was also reduced corroborating the decline in brain perfusion and social interactions. Thus, the late emergence of social interaction deficits mirrored the reduced vascular density and CBF in regions known to be involved in social behaviors. Vascular morphology and function improved prior to the late decrements in social function and our correlations strongly implicate a linkage between vascular density, cerebral perfusion, and social interactions. Our study provides a clinically relevant timeline of alterations in social deficits alongside functional vascular recovery that can guide future therapeutics.
Collapse
Affiliation(s)
- Aditya Singh
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
- Department of Neurology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, 120 Walter P Martin Research Center, Torrance, California, 90502, USA
- Department of Neurosurgery, University of California Los Angeles, Los Angeles, California, 90095, USA
| | - Steven Gong
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
| | - Anh Vu
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
| | - Scott Li
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA
| | - Andre Obenaus
- Department of Pediatrics, School of Medicine, University of California Irvine, Hewitt Hall Rm. 2066, Irvine, CA, 92697, USA.
- Division of Biomedical Sciences, 206 SOM Research Bldg, University of California Riverside, Riverside, CA, 92521, USA.
| |
Collapse
|
3
|
Jahromi HM, Rafati A, Karbalay-Doust S, Keshavarz S, Naseh M. The combination treatment of hypothermia and intranasal insulin ameliorates the structural and functional changes in a rat model of traumatic brain injury. Brain Struct Funct 2024; 229:947-957. [PMID: 38498064 DOI: 10.1007/s00429-024-02769-5] [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: 04/19/2023] [Accepted: 01/26/2024] [Indexed: 03/19/2024]
Abstract
The present study aimed to investigate the combination effects of hypothermia (HT) and intranasal insulin (INS) on structural changes of the hippocampus and cognitive impairments in the traumatic brain injury (TBI) rat model. The rats were divided randomly into the following five groups (n = 10): Sham, TBI, TBI with HT treatment for 3 h (TBI + HT), TBI with INS (ten microliters of insulin) treatment daily for 7 days (TBI + INS), and TBI with combining HT and INS (TBI + HT + INS). At the end of the 7th day, the open field and the Morris water maze tests were done for evaluation of anxiety-like behavior and memory performance. Then, after sacrificing, the brain was removed for stereological study. TBI led to an increase in the total volume of hippocampal subfields CA1 and DG and a decrease in the total number of neurons and non-neuronal cells in both sub-regions, which was associated with anxiety-like behavior and memory impairment. Although, the combination of HT and INS prevented the increased hippocampal volume and cell loss and improved behavioral performances in the TBI group. Our study suggests that the combined treatment of HT and INS could prevent increased hippocampal volume and cell loss in CA1 and DG sub-regions and consequently improve anxiety-like behaviors and memory impairment following TBI.
Collapse
Affiliation(s)
- Hadi Moatamed Jahromi
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Rafati
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saied Karbalay-Doust
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Anatomy Department, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somaye Keshavarz
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Physiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Maryam Naseh
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
4
|
Kalyani P, Lippa SM, Werner JK, Amyot F, Moore CB, Kenney K, Diaz-Arrastia R. Phosphodiesterase-5 (PDE-5) Inhibitors as Therapy for Cerebrovascular Dysfunction in Chronic Traumatic Brain Injury. Neurotherapeutics 2023; 20:1629-1640. [PMID: 37697134 PMCID: PMC10684467 DOI: 10.1007/s13311-023-01430-z] [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] [Accepted: 08/22/2023] [Indexed: 09/13/2023] Open
Abstract
Multiple phase III randomized controlled trials (RCTs) for pharmacologic interventions in traumatic brain injury (TBI) have failed despite promising results in experimental models. The heterogeneity of TBI, in terms of pathomechanisms and impacted brain structures, likely contributes to these failures. Biomarkers have been recommended to identify patients with relevant pathology (predictive biomarkers) and confirm target engagement and monitor therapy response (pharmacodynamic biomarkers). Our group focuses on traumatic cerebrovascular injury as an understudied endophenotype of TBI and is validating a predictive and pharmacodynamic imaging biomarker (cerebrovascular reactivity; CVR) in moderate-severe TBI. We aim to extend these studies to milder forms of TBI to determine the optimal dose of sildenafil for maximal improvement in CVR. We will conduct a phase II dose-finding study involving 160 chronic TBI patients (mostly mild) using three doses of sildenafil, a phosphodiesterase-5 (PDE-5) inhibitor. The study measures baseline CVR and evaluates the effect of escalating sildenafil doses on CVR improvement. A 4-week trial of thrice daily sildenafil will assess safety, tolerability, and clinical efficacy. This dual-site 4-year study, funded by the Department of Defense and registered in ClinicalTrials.gov (NCT05782244), plans to launch in June 2023. Biomarker-informed RCTs are essential for developing effective TBI interventions, relying on an understanding of underlying pathomechanisms. Traumatic microvascular injury (TMVI) is an attractive mechanism which can be targeted by vaso-active drugs such as PDE-5 inhibitors. CVR is a potential predictive and pharmacodynamic biomarker for targeted interventions aimed at TMVI. (Trial registration: NCT05782244, ClinicalTrials.gov ).
Collapse
Affiliation(s)
- Priyanka Kalyani
- Department of Neurology, University of Pennsylvania, 3400 Spruce St, Philadelphia, PA, 19104, USA.
| | - Sara M Lippa
- Walter Reed National Military Medical Center, The National Intrepid Center of Excellence, Palmer Rd S, Bethesda, MD, 20814, USA
- Department of Neuroscience, Uniformed Services University Health Sciences, 4301, Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - J Kent Werner
- Walter Reed National Military Medical Center, The National Intrepid Center of Excellence, Palmer Rd S, Bethesda, MD, 20814, USA
- Department of Neuroscience, Uniformed Services University Health Sciences, 4301, Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Franck Amyot
- Walter Reed National Military Medical Center, The National Intrepid Center of Excellence, Palmer Rd S, Bethesda, MD, 20814, USA
| | - Carol B Moore
- Department of Neuroscience, Uniformed Services University Health Sciences, 4301, Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Kimbra Kenney
- Department of Neuroscience, Uniformed Services University Health Sciences, 4301, Jones Bridge Rd, Bethesda, MD, 20814, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, 3400 Spruce St, Philadelphia, PA, 19104, USA
| |
Collapse
|
5
|
Reddy P, Izzetoglu K, Shewokis PA, Sangobowale M, Diaz-Arrastia R. Differences in time-frequency characteristics between healthy controls and TBI patients during hypercapnia assessed via fNIRS. Neuroimage Clin 2023; 40:103504. [PMID: 37734166 PMCID: PMC10518610 DOI: 10.1016/j.nicl.2023.103504] [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: 03/10/2023] [Revised: 07/24/2023] [Accepted: 09/01/2023] [Indexed: 09/23/2023]
Abstract
Damage to the cerebrovascular network is a universal feature of traumatic brain injury (TBI). This damage is present during different phases of the injury and can be non-invasively assessed using functional near infrared spectroscopy (fNIRS). fNIRS signals are influenced by partial arterial carbon dioxide (PaCO2), neurogenic, Mayer waves, respiratory and cardiac oscillations, whose characteristics vary in time and frequency and may differ in the presence of TBI. Therefore, this study aims to investigate differences in time-frequency characteristics of these fNIRS signal components between healthy controls and TBI patients and characterize the changes in their characteristics across phases of the injury. Data from 11 healthy controls and 21 TBI patients were collected during the hypercapnic protocol. Results demonstrated significant differences in low-frequency oscillations between healthy controls and TBI patients, with the largest differences observed in Mayer wave band (0.06 to 0.15 Hz), followed by the PaCO2 band (0.012 to 0.02 Hz). The effects within these bands were opposite, with (i) Mayer wave activity being lower in TBI patients during acute phase of the injury (d = 0.37 [0.16, 0.57]) and decreasing further during subacute (d = 0.66 [0.44, 0.87]) and postacute (d = 0.75 [0.50, 0.99]) phases; (ii) PaCO2 activity being lower in TBI patients only during acute phase of the injury (d = 0.36 [0.15, 0.56]) and stabilizing to healthy levels by the subacute phase. These findings demonstrate that TBI patients have impairments in low frequency oscillations related to different mechanisms and that these impairments evolve differently over the course of injury.
Collapse
Affiliation(s)
- Pratusha Reddy
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA.
| | - Kurtulus Izzetoglu
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA.
| | - Patricia A Shewokis
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA; Nutrition Sciences Department, Health Sciences Division of College of Nursing and Health Professions, Drexel University, Philadelphia, PA 19104, USA
| | - Michael Sangobowale
- Clinical TBI Research Center and Department of Neurology at University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ramon Diaz-Arrastia
- Clinical TBI Research Center and Department of Neurology at University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
6
|
Amyot F, Davis C, Sangobowale M, Moore C, Silverman E, Gandjbakhche A, Diaz-Arrastia R, Kenney K. Cerebrovascular Reactivity Measurement with Functional Near Infrared Spectroscopy. J Vis Exp 2022:10.3791/61284. [PMID: 35723463 PMCID: PMC11189107 DOI: 10.3791/61284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2024] Open
Abstract
Cerebrovascular reactivity (CVR) is the capacity of blood vessels in the brain to alter cerebral blood flow (either with dilation or constriction) in response to chemical or physical stimuli. The amount of reactivity in the cerebral microvasculature depends on the integrity of the capacitance vasculature and is the primary function of endothelial cells. CVR is, therefore, an indicator of the microvasculature's physiology and overall health. Imaging methods that can measure CVR are available but can be costly, and require magnetic resonance imaging centers and technical expertise. In this study, we used fNIRS technology to monitor changes of oxyhemoglobin (HbO) and deoxyhemoglobin (HbR) in the cerebral microvasculature to assess the CVR of 15 healthy controls (HC) in response to a vasoactive stimulus (inhaled 5% carbon dioxide or CO2). Our results suggest that this is a promising imaging technology that offers a non-invasive, accurate, portable, and cost-effective method of mapping cortical CVR and associated microvasculature function, resulting from a traumatic brain injury or other conditions associated with cerebral microvasculopathy.
Collapse
Affiliation(s)
- Franck Amyot
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center;
| | - Cora Davis
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center
| | | | - Carol Moore
- Uniformed Services University of the Health Sciences
| | | | - Amir Gandjbakhche
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | | | - Kimbra Kenney
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center; Uniformed Services University of the Health Sciences
| |
Collapse
|
7
|
Thomas BP, Tarumi T, Wang C, Zhu DC, Tomoto T, Munro Cullum C, Dieppa M, Diaz-Arrastia R, Bell K, Madden C, Zhang R, Ding K. Hippocampal and rostral anterior cingulate blood flow is associated with affective symptoms in chronic traumatic brain injury. Brain Res 2021; 1771:147631. [PMID: 34464600 DOI: 10.1016/j.brainres.2021.147631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/02/2021] [Accepted: 08/21/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVE The purpose of this study was to assess cerebral blood flow (CBF) and its association with self-reported symptoms in chronic traumatic brain injury (TBI). PARTICIPANTS Sixteen participants with mild to severe TBI and persistent self-reported neurological symptoms, 6 to 72 months post-injury were included. For comparison, 16 age- and gender-matched healthy normal control participants were also included. MAIN MEASURES Regional CBF and brain volume were assessed using pseudo-continuous Arterial Spin Labeling (PCASL) and T1-weighted data respectively. Cognitive function and self-reported symptoms were assessed in TBI participants using the national institutes of health (NIH) Toolbox Cognition Battery and Patient-Reported Outcome Measurement Information System respectively. Associations between CBF and cognitive function, symptoms were assessed. RESULTS Global CBF and regional brain volumes were similar between groups, but region of interest (ROI) analysis revealed lower CBF bilaterally in the thalamus, hippocampus, left caudate, and left amygdala in the TBI group. Voxel-wise analysis revealed that CBF in the hippocampus, parahippocampus, rostral anterior cingulate, inferior frontal gyrus, and other temporal regions were negatively associated with self-reported anger, anxiety, and depression symptoms. Furthermore, region of interest (ROI) analysis revealed that hippocampal and rostral anterior cingulate CBF were negatively associated with symptoms of fatigue, anxiety, depression, and sleep issues. CONCLUSION Regional CBF deficit was observed in the group with chronic TBI compared to the normal control (NC) group despite similar volume of cerebral structures. The observed negative correlation between regional CBF and affective symptoms suggests that CBF-targeted intervention may potentially improve affective symptoms and quality of life after TBI, which needs to be assessed in future studies.
Collapse
Affiliation(s)
- Binu P Thomas
- Advanced Imaging Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA; Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA; Department of Bioengineering, University of Texas at Arlington, 500 UTA Blvd., Arlington, TX 76010, USA.
| | - Takashi Tarumi
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA; Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, 8200 Walnut Hill Ln, Dallas, TX 75231, USA.
| | - Ciwen Wang
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA
| | - David C Zhu
- Department of Radiology and Cognitive Imaging Research Center, Michigan State University, 86 Service Road, East Lansing, MI 48824, USA
| | - Tsubasa Tomoto
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA; Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, 8200 Walnut Hill Ln, Dallas, TX 75231, USA
| | - C Munro Cullum
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA; Department of Radiology and Cognitive Imaging Research Center, Michigan State University, 86 Service Road, East Lansing, MI 48824, USA; Department of Psychiatry, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA; Department of Neurological Surgery, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA
| | - Marisara Dieppa
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, 51 North 39(th) St, Philadelphia, PA 19104, USA
| | - Kathleen Bell
- Department of Physical Medicine and Rehabilitation, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA
| | - Christopher Madden
- Department of Radiology and Cognitive Imaging Research Center, Michigan State University, 86 Service Road, East Lansing, MI 48824, USA
| | - Rong Zhang
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA; Institute for Exercise and Environmental Medicine, Texas Health Presbyterian Hospital Dallas, 8200 Walnut Hill Ln, Dallas, TX 75231, USA
| | - Kan Ding
- Department of Neurology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, TX 75390, USA
| |
Collapse
|
8
|
Haber M, Amyot F, Lynch CE, Sandsmark DK, Kenney K, Werner JK, Moore C, Flesher K, Woodson S, Silverman E, Chou Y, Pham D, Diaz-Arrastia R. Imaging biomarkers of vascular and axonal injury are spatially distinct in chronic traumatic brain injury. J Cereb Blood Flow Metab 2021; 41:1924-1938. [PMID: 33444092 PMCID: PMC8327117 DOI: 10.1177/0271678x20985156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 10/07/2020] [Accepted: 12/06/2020] [Indexed: 11/17/2022]
Abstract
Traumatic Brain Injury (TBI) is associated with both diffuse axonal injury (DAI) and diffuse vascular injury (DVI), which result from inertial shearing forces. These terms are often used interchangeably, but the spatial relationships between DAI and DVI have not been carefully studied. Multimodal magnetic resonance imaging (MRI) can help distinguish these injury mechanisms: diffusion tensor imaging (DTI) provides information about axonal integrity, while arterial spin labeling (ASL) can be used to measure cerebral blood flow (CBF), and the reactivity of the Blood Oxygen Level Dependent (BOLD) signal to a hypercapnia challenge reflects cerebrovascular reactivity (CVR). Subjects with chronic TBI (n = 27) and healthy controls (n = 14) were studied with multimodal MRI. Mean values of mean diffusivity (MD), fractional anisotropy (FA), CBF, and CVR were extracted for pre-determined regions of interest (ROIs). Normalized z-score maps were generated from the pool of healthy controls. Abnormal ROIs in one modality were not predictive of abnormalities in another. Approximately 9-10% of abnormal voxels for CVR and CBF also showed an abnormal voxel value for MD, while only 1% of abnormal CVR and CBF voxels show a concomitant abnormal FA value. These data indicate that DAI and DVI represent two distinct TBI endophenotypes that are spatially independent.
Collapse
Affiliation(s)
- Margalit Haber
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Franck Amyot
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Cillian E Lynch
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Danielle K Sandsmark
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Kimbra Kenney
- National Intrepid Center of Excellence, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - John K Werner
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Carol Moore
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kelley Flesher
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Sarah Woodson
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Erika Silverman
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Yiyu Chou
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Dzung Pham
- Center for Neuroscience and Regenerative Medicine, Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
9
|
Lynch CE, Eisenbaum M, Algamal M, Balbi M, Ferguson S, Mouzon B, Saltiel N, Ojo J, Diaz-Arrastia R, Mullan M, Crawford F, Bachmeier C. Impairment of cerebrovascular reactivity in response to hypercapnic challenge in a mouse model of repetitive mild traumatic brain injury. J Cereb Blood Flow Metab 2021; 41:1362-1378. [PMID: 33050825 PMCID: PMC8142124 DOI: 10.1177/0271678x20954015] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Incidences of repetitive mild TBI (r-mTBI), like those sustained by contact sports athletes and military personnel, are thought to be a risk factor for development of neurodegenerative disorders. Those suffering from chronic TBI-related illness demonstrate deficits in cerebrovascular reactivity (CVR), the ability of the cerebral vasculature to respond to a vasoactive stimulus. CVR is thus an important measure of traumatic cerebral vascular injury (TCVI), and a possible in vivo endophenotype of TBI-related neuropathogenesis. We combined laser speckle imaging of CVR in response to hypercapnic challenge with neurobehavioral assessment of learning and memory, to investigate if decreased cerebrovascular responsiveness underlies impaired cognitive function in our mouse model of chronic r-mTBI. We demonstrate a profile of blunted hypercapnia-evoked CVR in the cortices of r-mTBI mice like that of human TBI, alongside sustained memory and learning impairment, without biochemical or immunohistopathological signs of cerebral vessel laminar or endothelium constituent loss. Transient decreased expression of alpha smooth muscle actin and platelet-derived growth factor receptor β, indicative of TCVI, is obvious only at the time of the most pronounced CVR deficit. These findings implicate CVR as a valid preclinical measure of TCVI, perhaps useful for developing therapies targeting TCVI after recurrent mild head trauma.
Collapse
Affiliation(s)
- Cillian E Lynch
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA.,Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Maxwell Eisenbaum
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Moustafa Algamal
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Matilde Balbi
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Scott Ferguson
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Benoit Mouzon
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA
| | | | - Joseph Ojo
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mike Mullan
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK
| | - Fiona Crawford
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,James A. Haley Veteran's Administration, Tampa, FL, USA
| | - Corbin Bachmeier
- The Roskamp Institute, Sarasota, FL, USA.,Department of Life Sciences, The Open University, Milton Keynes, UK.,Bay Pines VA Healthcare System, Bay Pines, FL, USA
| |
Collapse
|
10
|
Han X, Chai Z, Ping X, Song LJ, Ma C, Ruan Y, Jin X. In vivo Two-Photon Imaging Reveals Acute Cerebral Vascular Spasm and Microthrombosis After Mild Traumatic Brain Injury in Mice. Front Neurosci 2020; 14:210. [PMID: 32210758 PMCID: PMC7077429 DOI: 10.3389/fnins.2020.00210] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/25/2020] [Indexed: 12/23/2022] Open
Abstract
Mild traumatic brain injury (mTBI), or concussion, is reported to interfere with cerebral blood flow and microcirculation in patients, but our current understanding is quite limited and the results are often controversial. Here we used longitudinal in vivo two-photon imaging to investigate dynamic changes in cerebral vessels and velocities of red blood cells (RBC) following mTBI. Closed-head mTBI induced using a controlled cortical impact device resulted in a significant reduction of dwell time in a Rotarod test but no significant change in water maze test. Cerebral blood vessels were repeatedly imaged through a thinned skull window at baseline, 0.5, 1, 6 h, and 1 day following mTBI. In both arterioles and capillaries, their diameters and RBC velocities were significantly decreased at 0.5, 1, and 6 h after injury, and recovered in 1 day post-mTBI. In contrast, decreases in the diameter and RBC velocity of venules occurred only in 0.5–1 h after mTBI. We also observed formation and clearance of transient microthrombi in capillaries within 1 h post-mTBI. We concluded that in vivo two-photon imaging is useful for studying earlier alteration of vascular dynamics after mTBI and that mTBI induced reduction of cerebral blood flow, vasospasm, and formation of microthrombi in the acute stage following injury. These changes may contribute to early brain functional deficits of mTBI.
Collapse
Affiliation(s)
- Xinjia Han
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Obstetrics and Gynecology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China
| | - Zhi Chai
- Neurobiology Research Center, Shanxi Key Laboratory of Innovative Drugs for Serious Illness, College of Basic Medicine, Shaanxi University of Chinese Medicine, Jinzhong, China
| | - Xingjie Ping
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Li-Juan Song
- Neurobiology Research Center, Shanxi Key Laboratory of Innovative Drugs for Serious Illness, College of Basic Medicine, Shaanxi University of Chinese Medicine, Jinzhong, China
| | - Cungen Ma
- Neurobiology Research Center, Shanxi Key Laboratory of Innovative Drugs for Serious Illness, College of Basic Medicine, Shaanxi University of Chinese Medicine, Jinzhong, China
| | - Yiwen Ruan
- GHM Institute of CNS Regeneration (GHMICR), Jinan University, Guangzhou, China.,Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaoming Jin
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States.,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
11
|
Abutarboush R, Gu M, Kawoos U, Mullah SH, Chen Y, Goodrich SY, Lashof-Sullivan M, McCarron RM, Statz JK, Bell RS, Stone JR, Ahlers ST. Exposure to Blast Overpressure Impairs Cerebral Microvascular Responses and Alters Vascular and Astrocytic Structure. J Neurotrauma 2019; 36:3138-3157. [PMID: 31210096 PMCID: PMC6818492 DOI: 10.1089/neu.2019.6423] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Exposure to blast overpressure may result in cerebrovascular impairment, including cerebral vasospasm. The mechanisms contributing to this vascular response are unclear. The aim of this study was to evaluate the relationship between blast and functional alterations of the cerebral microcirculation and to investigate potential underlying changes in vascular microstructure. Cerebrovascular responses were assessed in sham- and blast-exposed male rats at multiple time points from 2 h through 28 days after a single 130-kPa (18.9-psi) exposure. Pial microcirculation was assessed through a cranial window created in the parietal bone of anesthetized rats. Pial arteriolar reactivity was evaluated in vivo using hypercapnia, barium chloride, and serotonin. We found that exposure to blast leads to impairment of arteriolar reactivity >24 h after blast exposure, suggesting delayed injury mechanisms that are not simply attributed to direct mechanical deformation. Observed vascular impairment included a reduction in hypercapnia-induced vasodilation, increase in barium-induced constriction, and reversal of the serotonin effect from constriction to dilation. A reduction in vascular smooth muscle contractile proteins consistent with vascular wall proliferation was observed, as well as delayed reduction in nitric oxide synthase and increase in endothelin-1 B receptors, mainly in astrocytes. Collectively, the data show that exposure to blast results in delayed and prolonged alterations in cerebrovascular reactivity that are associated with changes in the microarchitecture of the vessel wall and astrocytes. These changes may contribute to long-term pathologies involving dysfunction of the neurovascular unit, including cerebral vasospasm.
Collapse
Affiliation(s)
- Rania Abutarboush
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Ming Gu
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Usmah Kawoos
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Saad H Mullah
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Ye Chen
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Samantha Y Goodrich
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Margaret Lashof-Sullivan
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Richard M McCarron
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | - Jonathan K Statz
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland.,The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Bethesda, Maryland
| | - Randy S Bell
- Neurosurgery Department, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - James R Stone
- Department of Radiology and Medical Imaging, University of Virginia Medical Center, Charlottesville, Virginia
| | - Stephen T Ahlers
- Neurotrauma Department, Naval Medical Research Center, Silver Spring, Maryland
| |
Collapse
|
12
|
Monson KL, Converse MI, Manley GT. Cerebral blood vessel damage in traumatic brain injury. Clin Biomech (Bristol, Avon) 2019; 64:98-113. [PMID: 29478776 DOI: 10.1016/j.clinbiomech.2018.02.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 01/12/2018] [Accepted: 02/13/2018] [Indexed: 02/07/2023]
Abstract
Traumatic brain injury is a devastating cause of death and disability. Although injury of brain tissue is of primary interest in head trauma, nearly all significant cases include damage of the cerebral blood vessels. Because vessels are critical to the maintenance of the healthy brain, any injury or dysfunction of the vasculature puts neural tissue at risk. It is well known that these vessels commonly tear and bleed as an immediate consequence of traumatic brain injury. It follows that other vessels experience deformations that are significant though not severe enough to produce bleeding. Recent data show that such subfailure deformations damage the microstructure of the cerebral vessels, altering both their structure and function. Little is known about the prognosis of these injured vessels and their potential contribution to disease development. The objective of this review is to describe the current state of knowledge on the mechanics of cerebral vessels during head trauma and how they respond to the applied loads. Further research on these topics will clarify the role of blood vessels in the progression of traumatic brain injury and is expected to provide insight into improved strategies for treatment of the disease.
Collapse
Affiliation(s)
- Kenneth L Monson
- Department of Mechanical Engineering, University of Utah, USA; Department of Bioengineering, University of Utah, USA.
| | | | - Geoffrey T Manley
- Department of Neurological Surgery, University of California, San Francisco, USA
| |
Collapse
|
13
|
Kenney K, Amyot F, Moore C, Haber M, Turtzo LC, Shenouda C, Silverman E, Gong Y, Qu BX, Harburg L, Wassermann EM, Lu H, Diaz‐Arrastia R. Phosphodiesterase-5 inhibition potentiates cerebrovascular reactivity in chronic traumatic brain injury. Ann Clin Transl Neurol 2018; 5:418-428. [PMID: 29687019 PMCID: PMC5899908 DOI: 10.1002/acn3.541] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 12/12/2017] [Accepted: 12/26/2017] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Traumatic cerebrovascular injury (TCVI), a common consequence of traumatic brain injury (TBI), presents an attractive therapeutic target. Because phosphodiesterase-5 (PDE5) inhibitors potentiate the action of nitric oxide (NO) produced by endothelial cells, they are candidate therapies for TCVI. This study aims to: (1) measure cerebral blood flow (CBF), cerebrovascular reactivity (CVR), and change in CVR after a single dose of sildenafil (ΔCVR) in chronic TBI compared to uninjured controls; (2) examine the safety and tolerability of 8-week sildenafil administration in chronic symptomatic moderate/severe TBI patients; and as an exploratory aim, (3) assess the effect of an 8-week course of sildenafil on chronic TBI symptoms. METHODS Forty-six subjects (31 chronic TBI, 15 matched healthy volunteers) were enrolled. Baseline CBF and CVR before and after administration of sildenafil were measured. Symptomatic TBI subjects then completed an 8-week double-blind, placebo-controlled, crossover trial of sildenafil. A neuropsychological battery and neurobehavioral symptom questionnaires were administered at each study visit. RESULTS After a single dose of sildenafil, TBI subjects showed a significant increase in global CVR compared to healthy controls (P < 0.001, d = 0.9). Post-sildenafil CVR maps showed near-normalization of CVR in many regions where baseline CVR was low, predominantly within areas without structural abnormalities. Sildenafil was well tolerated. Clinical Global Impression (CGI) scale showed a trend toward clinical improvement while on sildenafil treatment. FINDINGS Single-dose sildenafil improves regional CVR deficits in chronic TBI patients. CVR and ΔCVR are potential predictive and pharmacodynamic biomarkers of PDE5 inhibitor therapy for TCVI. Sildenafil is a potential therapy for TCVI.
Collapse
Affiliation(s)
- Kimbra Kenney
- Department of NeurologyUniformed Services University of the Health SciencesBethesdaMaryland
| | - Franck Amyot
- Department of NeurologyUniformed Services University of the Health SciencesBethesdaMaryland
| | - Carol Moore
- Department of NeurologyUniformed Services University of the Health SciencesBethesdaMaryland
| | - Margalit Haber
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | | | - Christian Shenouda
- Department of Physical Medicine and RehabilitationNational Institutes of Health Clinical CenterBethesdaMaryland
| | - Erika Silverman
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Yunhua Gong
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Bao‐ Xi Qu
- Department of NeurologyUniformed Services University of the Health SciencesBethesdaMaryland
| | - Leah Harburg
- Department of NeurologyUniformed Services University of the Health SciencesBethesdaMaryland
| | - Eric M. Wassermann
- Behavioral Neurology UnitNational Institute of Neurological Diseases and StrokeNational Institutes of HealthBethesdaMaryland
| | | | - Ramon Diaz‐Arrastia
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| |
Collapse
|
14
|
Amyot F, Kenney K, Moore C, Haber M, Turtzo LC, Shenouda C, Silverman E, Gong Y, Qu BX, Harburg L, Lu HY, Wassermann EM, Diaz-Arrastia R. Imaging of Cerebrovascular Function in Chronic Traumatic Brain Injury. J Neurotrauma 2018; 35:1116-1123. [PMID: 29065769 DOI: 10.1089/neu.2017.5114] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic cerebrovascular injury (TCVI) is a common pathologic mechanism of traumatic brain injury (TBI) and presents an attractive target for intervention. The aims of this study were to assess cerebral blood flow (CBF) and cerebrovascular reactivity (CVR) using magnetic resonance imaging (MRI) to assess their value as biomarkers of TCVI in chronic TBI, characterize the spatial distribution of TCVI, and assess the relationships between each biomarker and neuropsychological and clinical assessments. Forty-two subjects (27 chronic TBI, 15 age- and gender-matched healthy volunteers) were studied cross-sectionally. CBF was measured by arterial spin labeling and CVR by assessing the MRI-blood oxygen level-dependent signal with hypercapnia challenge. A focused neuropsychological battery adapted from the TBI Common Data Elements and neurobehavioral symptom questionnaires were administered at the time of the imaging session. Chronic TBI subjects showed a significant reduction in mean global, gray matter (GM), and white matter (WM) CVR, compared with healthy volunteers (p < 0.001). Mean GM CVR had the greatest effect size (Cohen's d = 0.9). CVR maps in chronic TBI subjects showed patchy, multifocal CVR deficits. CBF discriminated poorly between TBI subjects and healthy volunteers and did not correlate with CVR. Mean global CVR correlated best with chronic neurobehavioral symptoms among TBI subjects. Global, GM, and WM CVR are reliable and potentially useful biomarkers of TCVI in the chronic stage after moderate-to-severe TBI. CBF is less useful as biomarker of TCVI. CVR correlates best with chronic TBI symptoms. CVR has potential as a predictive and pharmacodynamic biomarker for interventions targeting TCVI.
Collapse
Affiliation(s)
- Franck Amyot
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Kimbra Kenney
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Carol Moore
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Margalit Haber
- 2 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - L Christine Turtzo
- 3 Center for Neuroscience and Regenerative Medicine, National Institute of Neurological Disorder and Stroke, National Institutes of Health , Bethesda, Maryland
| | - Christian Shenouda
- 3 Center for Neuroscience and Regenerative Medicine, National Institute of Neurological Disorder and Stroke, National Institutes of Health , Bethesda, Maryland
| | - Erika Silverman
- 2 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Yunhua Gong
- 2 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| | - Bao-Xi Qu
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Leah Harburg
- 1 Department of Neurology, Uniformed Services University of the Health Sciences , Bethesda, Maryland
| | - Hanzhang Y Lu
- 4 Department of Radiology and Radiological Science, Johns Hopkins University Baltimore , Maryland
| | - Eric M Wassermann
- 5 National Institute of Neurological Disorders and Stroke, National Institutes of Health , Bethesda, Maryland
| | - Ramon Diaz-Arrastia
- 2 Department of Neurology, University of Pennsylvania Perelman School of Medicine , Philadelphia, Pennsylvania
| |
Collapse
|
15
|
Pitkänen A, Löscher W, Vezzani A, Becker AJ, Simonato M, Lukasiuk K, Gröhn O, Bankstahl JP, Friedman A, Aronica E, Gorter JA, Ravizza T, Sisodiya SM, Kokaia M, Beck H. Advances in the development of biomarkers for epilepsy. Lancet Neurol 2017; 15:843-856. [PMID: 27302363 DOI: 10.1016/s1474-4422(16)00112-5] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 02/16/2016] [Accepted: 03/02/2016] [Indexed: 12/13/2022]
Abstract
Over 50 million people worldwide have epilepsy. In nearly 30% of these cases, epilepsy remains unsatisfactorily controlled despite the availability of over 20 antiepileptic drugs. Moreover, no treatments exist to prevent the development of epilepsy in those at risk, despite an increasing understanding of the underlying molecular and cellular pathways. One of the major factors that have impeded rapid progress in these areas is the complex and multifactorial nature of epilepsy, and its heterogeneity. Therefore, the vision of developing targeted treatments for epilepsy relies upon the development of biomarkers that allow individually tailored treatment. Biomarkers for epilepsy typically fall into two broad categories: diagnostic biomarkers, which provide information on the clinical status of, and potentially the sensitivity to, specific treatments, and prognostic biomarkers, which allow prediction of future clinical features, such as the speed of progression, severity of epilepsy, development of comorbidities, or prediction of remission or cure. Prognostic biomarkers are of particular importance because they could be used to identify which patients will develop epilepsy and which might benefit from preventive treatments. Biomarker research faces several challenges; however, biomarkers could substantially improve the management of people with epilepsy and could lead to prevention in the right person at the right time, rather than just symptomatic treatment.
Collapse
Affiliation(s)
- Asla Pitkänen
- Department of Neurobiology, A I Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Hannover, Germany; Center for Systems Neuroscience, Hannover, Germany
| | - Annamaria Vezzani
- Department of Neuroscience, Experimental Neurology, IRCCS-Istituto di Recerche Farmacologiche "Mario Negri", Milan, Italy
| | - Albert J Becker
- Section for Translational Epilepsy Research, Department of Neuropathology, University of Bonn Medical Center, University of Bonn, Bonn, Germany
| | - Michele Simonato
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy; Unit of Gene Therapy of Neurodegenerative Diseases, Division of Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | - Katarzyna Lukasiuk
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Olli Gröhn
- Department of Neurobiology, A I Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jens P Bankstahl
- Preclinical Molecular Imaging, Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Alon Friedman
- Department of Brain and Cognitive Sciences, Zlotowski Center for Neuroscience, Ben-Gurion University of the Negev, Israel; Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - Eleonora Aronica
- Department of Neuropathology, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands; Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, Netherlands
| | - Jan A Gorter
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| | - Teresa Ravizza
- Department of Neuroscience, Experimental Neurology, IRCCS-Istituto di Recerche Farmacologiche "Mario Negri", Milan, Italy
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Institute of Neurology, London, UK; Epilepsy Society, Chalfont St Peter, Buckinghamshire, UK
| | - Merab Kokaia
- Epilepsy Center, Experimental Epilepsy Group, Division of Neurology, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Heinz Beck
- Laboratory for Experimental Epileptology and Cognition Research, Department of Epileptology, University of Bonn, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
16
|
Abstract
Traumatic brain injury (TBI) has become the signature injury of the military conflict in Iraq and Afghanistan and also has a high rate of occurrence in civilian populations in the United States. Although the effects of a moderate to severe brain injury have been investigated for decades, the chronic effects of single and repetitive mild TBI are just beginning to be investigated. Data suggest that the different types and severities of TBI have unique long-term outcomes and thus may represent different types of diseases. Therefore, this review outlines the causes, incidence, symptoms, and pathophysiology of mild, moderate, and severe TBI.
Collapse
|
17
|
Szczygielski J, Müller A, Mautes AE, Sippl C, Glameanu C, Schwerdtfeger K, Steudel WI, Oertel J. Selective Brain Hypothermia Mitigates Brain Damage and Improves Neurological Outcome after Post-Traumatic Decompressive Craniectomy in Mice. J Neurotrauma 2017; 34:1623-1635. [PMID: 27799012 DOI: 10.1089/neu.2016.4615] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hypothermia and decompressive craniectomy (DC) have been considered as treatment for traumatic brain injury. The present study investigates whether selective brain hypothermia added to craniectomy could improve neurological outcome after brain trauma. Male CD-1 mice were assigned into the following groups: sham; DC; closed head injury (CHI); CHI followed by craniectomy (CHI+DC); and CHI+DC followed by focal hypothermia (CHI+DC+H). At 24 h post-trauma, animals were subjected to Neurological Severity Score (NSS) test and Beam Balance Score test. At the same time point, magnetic resonance imaging using a 9.4 Tesla scanner and subsequent volumetric evaluation of edema and contusion were performed. Thereafter, the animals were sacrificed and subjected to histopathological analysis. According to NSS, there was a significant impairment among all the groups subjected to trauma. Animals with both trauma and craniectomy performed significantly worse than animals with craniectomy alone. This deleterious effect disappeared when additional hypothermia was applied. BBS was significantly worse in the CHI and CHI+DC groups, but not in the CHI+DC+H group, compared to the sham animals. Edema and contusion volumes were significantly increased in CHI+DC animals, but not in the CHI+DC+H group, compared to the DC group. Histopathological analysis showed that neuronal loss and contusional blossoming could be attenuated by application of selective brain hypothermia. Selective brain cooling applied post-trauma and craniectomy improved neurological function and reduced structural damage and may be therefore an alternative to complication-burdened systemic hypothermia. Clinical studies are recommended in order to explore the potential of this treatment.
Collapse
Affiliation(s)
- Jacek Szczygielski
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Andreas Müller
- 2 Department of Radiology, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Angelika E Mautes
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Christoph Sippl
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Cosmin Glameanu
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Karsten Schwerdtfeger
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Wolf-Ingo Steudel
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| | - Joachim Oertel
- 1 Department of Neurosurgery, Saarland University Medical Center and Saarland University Faculty of Medicine , Homburg/Saar, Germany
| |
Collapse
|
18
|
Muradashvili N, Tyagi R, Tyagi N, Tyagi SC, Lominadze D. Cerebrovascular disorders caused by hyperfibrinogenaemia. J Physiol 2016; 594:5941-5957. [PMID: 27121987 DOI: 10.1113/jp272558] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/25/2016] [Indexed: 01/18/2023] Open
Abstract
KEY POINTS Hyperfibrinogenaemia (HFg) results in vascular remodelling, and fibrinogen (Fg) and amyloid β (Aβ) complex formation is a hallmark of Alzheimer's disease. However, the interconnection of these effects, their mechanisms and implications in cerebrovascular diseases are not known. Using a mouse model of HFg, we showed that at an elevated blood level, Fg increases cerebrovascular permeability via mainly caveolar protein transcytosis. This enhances deposition of Fg in subendothelial matrix and interstitium making the immobilized Fg a readily accessible substrate for binding Aβ and cellular prion protein (PrPC ), the protein that is thought to have a greater effect on memory than Aβ. We showed that enhanced formation of Fg-Aβ and Fg-PrPC complexes are associated with reduction in short-term memory. The present study delineates a new mechanistic pathway for vasculo-neuronal dysfunctions found in inflammatory cardiovascular and cerebrovascular diseases associated with an elevated blood level of Fg. ABSTRACT Many cardiovascular diseases are associated with inflammation and as such are accompanied by an increased blood level of fibrinogen (Fg). Besides its well-known prothrombotic effects Fg seems to have other destructive roles in developing microvascular dysfunction that include changes in vascular reactivity and permeability. Increased permeability of brain microvessels has the most profound effects as it may lead to cerebrovascular remodelling and result in memory reduction. The goal of the present study was to define mechanisms of cerebrovascular permeability and associated reduction in memory induced by elevated blood content of Fg. Genetically modified, transgenic hyperfibrinogenic (HFg) mice were used to study cerebrovascular transcellular and paracellular permeability in vivo. The extent of caveolar formation and the role of caveolin-1 signalling were evaluated by immunohistochemistry (IHC) and Western blot (WB) analysis in brain samples from experimental animals. Formation of Fg complexes with amyloid β (Aβ) and with cellular prion protein (PrPC ) were also assessed with IHC and WB analysis. Short-term memory of mice was assessed by novel object recognition and Y-maze tests. Caveolar protein transcytosis was found to have a prevailing role in overall increased cerebrovascular permeability in HFg mice. These results were associated with enhanced formation of caveolae. Increased formation of Fg-PrPC and Fg-Aβ complexes were correlated with reduction in short-term memory in HFg mice. Using the model of hyperfibrinogenaemia, the present study shows a novel mechanistic pathway of inflammation-induced and Fg-mediated reduction in short-term memory.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Reeta Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Neetu Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
19
|
Kenney K, Amyot F, Haber M, Pronger A, Bogoslovsky T, Moore C, Diaz-Arrastia R. Cerebral Vascular Injury in Traumatic Brain Injury. Exp Neurol 2016; 275 Pt 3:353-366. [DOI: 10.1016/j.expneurol.2015.05.019] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/19/2015] [Accepted: 05/26/2015] [Indexed: 12/14/2022]
|
20
|
Ziebell JM, Rowe RK, Harrison JL, Eakin KC, Colburn T, Willyerd FA, Lifshitz J. Experimental diffuse brain injury results in regional alteration of gross vascular morphology independent of neuropathology. Brain Inj 2015; 30:217-24. [PMID: 26646974 DOI: 10.3109/02699052.2015.1090012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PRIMARY OBJECTIVE A dynamic relationship exists between diffuse traumatic brain injury and changes to the neurovascular unit. The purpose of this study was to evaluate vascular changes during the first week following diffuse TBI. It was hypothesized that pathology is associated with modification of the vasculature. METHODS Male Sprague-Dawley rats underwent either midline fluid percussion injury or sham-injury. Brain tissue was collected 1, 2 or 7 days post-injury or sham-injury (n = 3/time point). Tissue was collected and stained by de Olmos amino-cupric silver technique to visualize neuropathology or animals were perfused with AltaBlue casting resin before high-resolution vascular imaging. The average volume, surface area, radius, branching and tortuosity of the vessels were evaluated across three regions of interest. RESULTS In M2, average vessel volume (p < 0.01) and surface area (p < 0.05) were significantly larger at 1 day relative to 2 days, 7 days and sham. In S1BF and VPM, no significant differences in the average vessel volume or surface area at any of the post-injury time points were observed. No significant changes in average radius, branching or tortuosity were observed. CONCLUSIONS Preliminary findings suggest gross morphological changes within the vascular network likely represent an acute response to mechanical forces of injury, rather than delayed or chronic pathological processes.
Collapse
Affiliation(s)
- Jenna M Ziebell
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Rachel K Rowe
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA.,c Phoenix VA Healthcare System , Phoenix , AZ , USA
| | - Jordan L Harrison
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Katharine C Eakin
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - Taylor Colburn
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA
| | - F Anthony Willyerd
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA.,d Critical Care, Phoenix Children's Hospital , Phoenix , AZ , USA
| | - Jonathan Lifshitz
- a BARROW Neurological Institute at Phoenix Children's Hospital , Phoenix , AZ , USA.,b Department of Child Health , University of Arizona College of Medicine - Phoenix , Phoenix , AZ , USA.,c Phoenix VA Healthcare System , Phoenix , AZ , USA.,e Psychology , Arizona State University , Tempe , AZ , USA
| |
Collapse
|
21
|
Muradashvili N, Benton RL, Saatman KE, Tyagi SC, Lominadze D. Ablation of matrix metalloproteinase-9 gene decreases cerebrovascular permeability and fibrinogen deposition post traumatic brain injury in mice. Metab Brain Dis 2015; 30:411-26. [PMID: 24771110 PMCID: PMC4213324 DOI: 10.1007/s11011-014-9550-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/15/2014] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is accompanied with enhanced matrix metalloproteinase-9 (MMP-9) activity and elevated levels of plasma fibrinogen (Fg), which is a known inflammatory agent. Activation of MMP-9 and increase in blood content of Fg (i.e. hyperfibrinogenemia, HFg) both contribute to cerebrovascular disorders leading to blood brain barrier disruption. It is well-known that activation of MMP-9 contributes to vascular permeability. It has been shown that at an elevated level (i.e. HFg) Fg disrupts blood brain barrier. However, mechanisms of their actions during TBI are not known. Mild TBI was induced in wild type (WT, C57BL/6 J) and MMP-9 gene knockout (Mmp9(-/-)) homozygous, mice. Pial venular permeability to fluorescein isothiocyanate-conjugated bovine serum albumin in pericontusional area was observed 14 days after injury. Mice memory was tested with a novel object recognition test. Increased expression of Fg endothelial receptor intercellular adhesion protein-1 and formation of caveolae were associated with enhanced activity of MMP-9 causing an increase in pial venular permeability. As a result, an enhanced deposition of Fg and cellular prion protein (PrP(C)) were found in pericontusional area. These changes were attenuated in Mmp9(-/-) mice and were associated with lesser loss of short-term memory in these mice than in WT mice. Our data suggest that mild TBI-induced increased cerebrovascular permeability enhances deposition of Fg-PrP(C) and loss of memory, which is ameliorated in the absence of MMP-9 activity. Thus, targeting MMP-9 activity and blood level of Fg can be a possible therapeutic remedy to diminish vasculo-neuronal damage after TBI.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - Richard L. Benton
- Department of Anatomical Sciences and Neurobiology and Kentucky Spinal Cord Injury Research Center (KSCIRC), University of Louisville, School of Medicine, Louisville, KY
| | - Kathryn E. Saatman
- Department of Physiology and Neurosurgery and Spinal Cord & Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY, USA
| | - Suresh C. Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - David Lominadze
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
- Corresponding Author: David Lominadze, Ph. D., University of Louisville, Dept. of Physiology & Biophysics, School of Medicine, Bldg. A, Room 1115, 500 South Preston Street, Louisville, KY 40202, Phone (502) 852-4902, Fax (502) 852-6239,
| |
Collapse
|
22
|
Jullienne A, Roberts JM, Pop V, Paul Murphy M, Head E, Bix GJ, Badaut J. Juvenile traumatic brain injury induces long-term perivascular matrix changes alongside amyloid-beta accumulation. J Cereb Blood Flow Metab 2014; 34:1637-45. [PMID: 25052558 PMCID: PMC4269722 DOI: 10.1038/jcbfm.2014.124] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/28/2014] [Accepted: 06/05/2014] [Indexed: 02/08/2023]
Abstract
In our juvenile traumatic brain injury (jTBI) model, emergence of cognitive dysfunctions was observed up to 6 months after trauma. Here we hypothesize that early brain injury induces changes in the neurovascular unit (NVU) that would be associated with amyloid-beta (Aβ) accumulation. We investigated NVU changes for up to 6 months in a rat jTBI model, with a focus on the efflux protein P-glycoprotein (P-gp) and on the basement membrane proteins perlecan and fibronectin, all known to be involved in Aβ clearance. Rodent-Aβ staining is present and increased after jTBI around cerebral blood microvessels, and the diameter of those is decreased by 25% and 34% at 2 and 6 months, respectively, without significant angiogenesis. P-glycoprotein staining in endothelium is decreased by 22% and parallels an increase of perlecan and fibronectin staining around cerebral blood vessels. Altogether, these results strongly suggest that the emergence of long-term behavioral dysfunctions observed in rodent jTBI may be related to endothelial remodeling at the blood-brain barrier alongside vascular dysfunction and altered Aβ trafficking. This study shows that it is important to consider jTBI as a vascular disorder with long-term consequences on cognitive functions.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, Loma Linda University, Loma Linda, California, USA
| | - Jill M Roberts
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Viorela Pop
- Department of Pediatrics, Loma Linda University, Loma Linda, California, USA
| | - M Paul Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Gregory J Bix
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Jérôme Badaut
- 1] Department of Pediatrics, Loma Linda University, Loma Linda, California, USA [2] Department of Physiology, Loma Linda University, Loma Linda, California, USA [3] CNRS UMR 5287, Bordeaux University, Bordeaux, France
| |
Collapse
|
23
|
Glushakova OY, Johnson D, Hayes RL. Delayed Increases in Microvascular Pathology after Experimental Traumatic Brain Injury Are Associated with Prolonged Inflammation, Blood–Brain Barrier Disruption, and Progressive White Matter Damage. J Neurotrauma 2014; 31:1180-93. [DOI: 10.1089/neu.2013.3080] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
24
|
Muradashvili N, Benton RL, Tyagi R, Tyagi SC, Lominadze D. Elevated level of fibrinogen increases caveolae formation; role of matrix metalloproteinase-9. Cell Biochem Biophys 2014; 69:283-94. [PMID: 24307281 PMCID: PMC4020992 DOI: 10.1007/s12013-013-9797-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The role of the inflammatory agent fibrinogen (Fg) in increased pial venular permeability has been shown previously. It was suggested that an activation of matrix metalloproteinase-9 (MMP-9) is involved in Fg-induced enhanced transcytosis through endothelial cells (ECs). However, direct link between Fg, caveolae formation, and MMP-9 activity has never been shown. We hypothesized that at an elevated level, Fg enhances formation of functional caveolae through activation of MMP-9. Male wild-type (WT, C57BL/6J) or MMP-9 gene knockout (MMP9-/-) mice were infused with Fg (4 mg/ml, final blood concentration) or equal volume of phosphate buffered saline (PBS). After 2 h, mice were sacrificed and brains were collected for immunohistochemical analyses. Mouse brain ECs were treated with 4 mg/ml of Fg or PBS in the presence or absence of MMP-9 activity inhibitor, tissue inhibitor of metalloproteinases-4 (TIMP-4, 12 ng/ml). Formation of functional caveolae was assessed by confocal microscopy. Fg-induced increased formation of caveolae, which was defined by an increased co-localization of caveolin-1 (Cav-1) and plasmalemmal vesicle-associated protein-1 and was associated with an increased phosphorylation of Cav-1, was ameliorated in the presence of TIMP-4. These results suggest that at high levels, Fg enhances formation of functional caveolae that may involve Cav-1 signaling and MMP-9 activation.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - Richard L. Benton
- Department of Anatomical Sciences and Neurobiology and Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, KY
| | - Reeta Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - Suresh C. Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - David Lominadze
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| |
Collapse
|
25
|
Badaut J, Bix GJ. Vascular neural network phenotypic transformation after traumatic injury: potential role in long-term sequelae. Transl Stroke Res 2013; 5:394-406. [PMID: 24323723 DOI: 10.1007/s12975-013-0304-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 01/11/2023]
Abstract
The classical neurovascular unit (NVU), composed primarily of endothelium, astrocytes, and neurons, could be expanded to include smooth muscle and perivascular nerves present in both the up- and downstream feeding blood vessels (arteries and veins). The extended NVU, which can be defined as the vascular neural network (VNN), may represent a new physiological unit to consider for therapeutic development in stroke, traumatic brain injury, and other brain disorders (Zhang et al., Nat Rev Neurol 8(12):711-716, 2012). This review is focused on traumatic brain injury and resultant post-traumatic changes in cerebral blood flow, smooth muscle cells, matrix, blood-brain barrier structures and function, and the association of these changes with cognitive outcomes as described in clinical and experimental reports. We suggest that studies characterizing TBI outcomes should increase their focus on changes to the VNN, as this may yield meaningful therapeutic targets to resolve posttraumatic dysfunction.
Collapse
Affiliation(s)
- J Badaut
- Department of Pediatrics, Loma Linda University School of Medicine, Coleman Pavilion, Room A1120, 11175 Campus Street, Loma Linda, CA, 92354, USA,
| | | |
Collapse
|
26
|
Abstract
Traumatic brain injury (TBI) has been associated with various neurological disorders. However, the role of cerebrovascular dysfunction and its mechanisms associated with TBI are still not well understood. Inflammation is the main cause of vascular dysfunction. It affects properties of blood components and the vascular wall leading to changes in blood flow and in interaction of blood components and vascular endothelium exacerbating microcirculatory complications during inflammatory diseases. One of the markers of inflammation is a plasma adhesion protein, fibrinogen (Fg). At elevated levels, Fg can also cause inflammatory responses. One of the manifestations of inflammatory responses is an increase in microvascular permeability leading to accumulation of plasma proteins in the subendothelial matrix and causing vascular remodelling. This has a most devastating effect on cerebral circulation after TBI that is accompanied with an elevation of plasma level of Fg and with an increased cerebrovascular permeability in injury penumbra impairing the normal healing process. This study reviews cerebrovascular alterations after TBI, considers the consequences of increased blood-brain barrier permeability, defines the role of elevated level of Fg and discusses the potential mechanisms of its action leading to vascular dysfunction, which subsequently can cause impairment in neuronal function. Thus, possible mechanisms of vasculo-neuronal dysfunction after TBI are considered.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine , Louisville, KY , USA
| | | |
Collapse
|
27
|
Fujita M, Oda Y, Wei EP, Povlishock JT. The combination of either tempol or FK506 with delayed hypothermia: implications for traumatically induced microvascular and axonal protection. J Neurotrauma 2012; 28:1209-18. [PMID: 21521034 DOI: 10.1089/neu.2011.1852] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Following traumatic brain injury (TBI), inhibition of reactive oxygen species and/or calcineurin can exert axonal and vascular protection. This protection proves optimal when these strategies are used early post-injury. Recent work has shown that the combination of delayed drug administration and delayed hypothermia extends this protection. Here we revisit this issue in TBI using the nitroxide antioxidant Tempol, or the immunophilin ligand FK506, together with delayed hypothermia, to determine their effects upon cerebral vascular reactivity and axonal damage. Animals were subjected to TBI and treated with Tempol at 30 or 90 min post-injury, or 90 min post-injury with concomitant mild hypothermia (33°C). Another group of animals were treated in the same fashion with the exception that they received FK506. Cranial windows were placed to assess vascular reactivity over 6 h post-injury, when the animals were assessed for traumatically induced axonal damage. Vasoreactivity was preserved by early Tempol administration; however, this benefit declined with time. The coupling of hypothermia and delayed Tempol, however, exerted significant vascular protection. The use of early and delayed FK506 provided significant vascular protection which was not augmented by hypothermia. The early administration of Tempol provided dramatic axonal protection that was not enhanced with hypothermia. Early and delayed FK506 provided significant axonal protection, although this protection was not enhanced by delayed hypothermia. The current investigation supports the premise that Tempol coupled with hypothermia extends its benefits. While FK506 proved efficacious with early and delayed administration, it did not provide either increased vascular or axonal benefit with hypothermia. These studies illustrate the potential benefits of Tempol coupled to delayed hypothermia. However, these findings do not transfer to the use of FK506, which in previous studies proved beneficial when coupled with hypothermia. These divergent results may be a reflection of the different animal models used and/or their associated injury severity.
Collapse
Affiliation(s)
- Motoki Fujita
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, VA 23298, USA
| | | | | | | |
Collapse
|
28
|
Fujita M, Wei EP, Povlishock JT. Effects of hypothermia on cerebral autoregulatory vascular responses in two rodent models of traumatic brain injury. J Neurotrauma 2012; 29:1491-8. [PMID: 22364620 DOI: 10.1089/neu.2011.2278] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) can trigger disturbances of cerebral pressure autoregulation that can translate into the generation of secondary insults and increased morbidity/mortality. Few therapies have been developed to attenuate the damaging consequences of disturbed autoregulatory control, although some suggest that hypothermia may exert such protection. Here we reexamine this issue of traumatically induced autoregulatory disturbances and their modulation by hypothermic intervention, examining these phenomena in two different models of TBI. Adult rats were subjected to either impact acceleration injury (IAI) or lateral fluid percussion injury (LFPI) followed by the insertion of cranial windows to assess the pial arteriolar cerebral autoregulatory vascular response to the post-traumatic induction of sequential reductions of arterial blood pressure. The potential for continued pial vasodilation in response to declining blood pressure was directly measured post-injury and compared with that in injured groups subjected to 33° C of hypothermia of 1-2 h duration initiated 1 h post-injury. We observed that the TBI resulted in either impaired or abolished cerebral vascular dilation in response to the sequential declines in blood pressure. Following IAI there was a 50% reduction in the vasculature's ability to dilate in response to the induced hypotension. In contrast, following LFPI, the vascular response to hypotension was abolished both ipsilateral and contralateral to the LFPI. In animals sustaining IAI, the use of 1 h post-traumatic hypothermia preserved vascular dilation in response to declines in blood pressure in contrast to the LFPI in which the use of the same strategy afforded no improvement. However, with LFPI, the use of 2 h of hypothermia provided partial vascular protection. These results clearly illustrate that TBI can alter the cerebral autoregulatory vascular response to sequentially induced hypotensive insult, whereas the use of post-traumatic hypothermia provides benefit. Collectively, these studies also demonstrate that different animal models of TBI can evoke different biological responses to injury.
Collapse
Affiliation(s)
- Motoki Fujita
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| | | | | |
Collapse
|
29
|
Schmutzhard E, Fischer M, Dietmann A, Helbok R, Broessner G. Rewarming: facts and myths from the neurological perspectives. Crit Care 2012. [PMCID: PMC3389484 DOI: 10.1186/cc11282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
30
|
Frydrychowski AF, Wszedybyl-Winklewska M, Guminski W, Lass P, Bandurski T, Winklewski PJ. Effects of acute hypercapnia on the amplitude of cerebrovascular pulsation in humans registered with a non-invasive method. Microvasc Res 2012; 83:229-36. [DOI: 10.1016/j.mvr.2011.08.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/07/2011] [Accepted: 08/13/2011] [Indexed: 11/15/2022]
|
31
|
Oda Y, Gao G, Wei EP, Povlishock JT. Combinational therapy using hypothermia and the immunophilin ligand FK506 to target altered pial arteriolar reactivity, axonal damage, and blood-brain barrier dysfunction after traumatic brain injury in rat. J Cereb Blood Flow Metab 2011; 31:1143-54. [PMID: 21157473 PMCID: PMC3070975 DOI: 10.1038/jcbfm.2010.208] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
This study evaluated the utility of combinational therapy, coupling delayed posttraumatic hypothermia with delayed FK506 administration, on altered cerebral vascular reactivity, axonal injury, and blood-brain barrier (BBB) disruption seen following traumatic brain injury (TBI). Animals were injured, subjected to various combinations of hypothermic/FK506 intervention, and equipped with cranial windows to assess pial vascular reactivity to acetylcholine. Animals were then processed with antibodies to the amyloid precursor protein and immunoglobulin G to assess axonal injury and BBB disruption, respectively. Animals were assigned to five groups: (1) sham injury plus delayed FK506, (2) TBI, (3) TBI plus delayed hypothermia, (4) TBI plus delayed FK506, and (5) TBI plus delayed hypothermia with FK506. Sham injury plus FK506 had no impact on vascular reactivity, axonal injury, or BBB disruption. Traumatic brain injury induced dramatic axonal injury and altered pial vascular reactivity, while triggering local BBB disruption. Delayed hypothermia or FK506 after TBI provided limited protection. However, TBI with combinational therapy achieved significantly enhanced vascular and axonal protection, with no BBB protection. This study shows the benefits of combinational therapy, using posttraumatic hypothermia with FK506 to attenuate important features of TBI. This suggests that hypothermia not only protects but also extends the therapeutic window for improved FK506 efficacy.
Collapse
Affiliation(s)
- Yasutaka Oda
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| | | | | | | |
Collapse
|
32
|
Schwarzmaier SM, Kim SW, Trabold R, Plesnila N. Temporal profile of thrombogenesis in the cerebral microcirculation after traumatic brain injury in mice. J Neurotrauma 2010; 27:121-30. [PMID: 19803784 DOI: 10.1089/neu.2009.1114] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Traumatic brain injury (TBI) is associated with an almost immediate reduction in cerebral blood flow (CBF). Because cerebral perfusion pressure is often normal under these circumstances it was hypothesized that the reduction of post-traumatic CBF has to occur at the level of the microcirculation. The aim of the current study was to investigate whether cerebral microvessels are involved in the development of blood flow disturbances following experimental TBI. C57/BL6 mice (n = 12) were intubated and ventilated under control of end-tidal Pco(2) ((ET)P(CO2)). After preparation of a cranial window and baseline recordings, the animals were subjected to experimental TBI by controlled cortical impact (CCI; 6 m/sec, 0.5 mm). Vessel lumina and intravascular cells were visualized by in vivo fluorescence microscopy (IVM) using the fluorescent dyes FITC-dextran and rhodamine 6G, respectively. Vessel diameter, cell-endothelial interactions, and thrombus formation were quantified within the traumatic penumbra by IVM up to 2 h after CCI. Arteriolar diameters increased after CCI by 26.2 +/- 2.5% (mean +/- SEM, p < 0.01 versus baseline), and remained at this level until the end of the observation period. Rolling of leukocytes on the cerebrovascular endothelium was observed both in arterioles and venules, while leukocyte-platelet aggregates were found only in venules. Microthrombi occluded up to 70% of venules and 33% of arterioles. The current data suggest that the immediate post-traumatic decrease in peri-contusional blood flow is not caused by arteriolar vasoconstriction, but by platelet activation and the subsequent formation of thrombi in the cerebral microcirculation.
Collapse
Affiliation(s)
- Susanne M Schwarzmaier
- Institute for Surgical Research in the Walter Brendel Center for Experimental Medicine, Department of Neurosurgery, University of Munich Medical Center-Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | | | | | | |
Collapse
|
33
|
Effect of temperature on spinal cord regeneration in the weakly electric fish, Apteronotus leptorhynchus. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2010; 196:359-68. [DOI: 10.1007/s00359-010-0521-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2009] [Revised: 03/03/2010] [Accepted: 03/04/2010] [Indexed: 11/27/2022]
|
34
|
Rubovitch V, Edut S, Sarfstein R, Werner H, Pick CG. The intricate involvement of the Insulin-like growth factor receptor signaling in mild traumatic brain injury in mice. Neurobiol Dis 2010; 38:299-303. [PMID: 20138993 DOI: 10.1016/j.nbd.2010.01.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Revised: 01/26/2010] [Accepted: 01/27/2010] [Indexed: 12/28/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) was suggested as a potential neuroprotective treatment for traumatic brain injury (TBI) induced damage (cognitive as well as cellular). The main goal of the present study was to evaluate the role of the IGF-1R activation in spatial memory outcome following mild traumatic brain injury. mTBI-induced phosphorylation of IGF-1R, AKT and ERK1/2, in mice hippocampus, which was inhibited when mice were pretreated with the selective IGF-1R inhibitor AG1024. IGF-1 administration prevented spatial memory deficits following mTBI. Surprisingly, blocking the IGF-1R signaling in mTBI mice did not augment the spatial memory deficit. In addition, this data imply an intriguing and complex role of the IGF-1 signaling axis in the cellular and behavioral events following mTBI.
Collapse
Affiliation(s)
- Vardit Rubovitch
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Israel
| | | | | | | | | |
Collapse
|