1
|
Pearson N, Boiczyk GM, Anderl WJ, Marino M, Yu SM, Monson KL. Softening of elastic and viscoelastic properties is independent of overstretch rate in cerebral arteries. J Mech Behav Biomed Mater 2025; 166:106957. [PMID: 40014942 DOI: 10.1016/j.jmbbm.2025.106957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 03/01/2025]
Abstract
Collagenous soft tissues are frequently injured by supraphysiologic mechanical deformation, leading to measurable changes in both extra-cellular matrix (ECM) structure and mechanical properties. While each of these alterations has been well studied following quasi-static deformation, little is known about the influence of high strain rate. Previous investigations of high-rate ECM alterations found tropocollagen denaturation and fibrillar kinking to be rate dependent. Given these observations of rate dependence in microstructure alterations, the present work evaluated if the rate and magnitude of overstretch affect the baseline viscoelastic properties of porcine middle cerebral arteries (MCAs). Changes in tissue response were assessed using a series of harmonic oscillations before and after sub-failure overstretches across a large range of rates and magnitudes. We used collagen-hybridizing peptide (CHP) to evaluate the role of tropocollagen denaturation in mechanical softening. Experiments show that softening is dependent on overstretch magnitude but is independent of overstretch rate. We also note that softening progresses at the same rate for both equilibrium (quasi-static) and non-equilibrium (high-rate) properties. Finally, results suggest that tropocollagen denaturation is not the source of the observed sub-yield softening behavior. This study expands fundamental knowledge on the form-function relationship of constituents in collagen fibrils and clarifies material behavior following sub-failure overstretch across a range of strain rates.
Collapse
Affiliation(s)
- Noah Pearson
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Gregory M Boiczyk
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - William J Anderl
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA
| | - Michele Marino
- Department of Civil Engineering and Computer Science Engineering, University of Rome Tor Vergata, Rome, Italy
| | - S Michael Yu
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Kenneth L Monson
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, 84112, USA.
| |
Collapse
|
2
|
Barbour MA, Whitehead B, Gumbo C, Karelina K, Weil ZM. Traumatic brain injury persistently increases the incidence of both ischemic and hemorrhagic strokes: Potential mechanisms. Prog Neurobiol 2025; 248:102749. [PMID: 40113130 PMCID: PMC12021558 DOI: 10.1016/j.pneurobio.2025.102749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Traumatic brain injuries (TBI) significantly increase the risk of both ischemic and hemorrhagic strokes, with effects persisting for years after the initial injury. The mechanisms underlying this increased stroke risk are complex, multifactorial, and incompletely understood but likely include chronic cerebrovascular dysfunction, blood-brain barrier disruption, and inflammatory responses. Epidemiological studies consistently show that TBI is an independent risk factor for stroke, with more severe injuries associated with greater risk, especially for hemorrhagic strokes. Traditional risk factors for stroke, such as hypertension, poor diet, and sedentary lifestyle, further elevate the risk in TBI survivors. Modifiable lifestyle factors, such as improving sleep, increasing physical activity, and adopting heart-healthy diets, offer potential intervention points to mitigate stroke risk. Pharmacological considerations, including the use of antidepressants, anticoagulants, and statins, also influence stroke risk, particularly with regard to hemorrhagic complications. This review explores the pathophysiological mechanisms linking TBI and stroke, emphasizing the need for future research to identify specific biomarkers and imaging techniques to predict stroke vulnerability in TBI patients. Addressing the gaps in understanding, particularly regarding small vessel pathology, will be essential to developing targeted therapies for reducing stroke incidence in TBI survivors.
Collapse
Affiliation(s)
- Mikaela A Barbour
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA.
| | - Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| | - Claymore Gumbo
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| | - Zachary M Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, 108 Biomedical Road, 313 BMRC, Morgantown, WV 26506, USA
| |
Collapse
|
3
|
Sassani M, Ghafari T, Arachchige PRW, Idrees I, Gao Y, Waitt A, Weaver SRC, Mazaheri A, Lyons HS, Grech O, Thaller M, Witton C, Bagshaw AP, Wilson M, Park H, Brookes M, Novak J, Mollan SP, Hill LJ, Lucas SJE, Mitchell JL, Sinclair AJ, Mullinger K, Fernández-Espejo D. Current and prospective roles of magnetic resonance imaging in mild traumatic brain injury. Brain Commun 2025; 7:fcaf120. [PMID: 40241788 PMCID: PMC12001801 DOI: 10.1093/braincomms/fcaf120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 11/26/2024] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
There is unmet clinical need for biomarkers to predict recovery or the development of long-term sequelae of mild traumatic brain injury, a highly prevalent condition causing a constellation of disabling symptoms. A substantial proportion of patients live with long-lasting sequelae affecting their quality of life and ability to work. At present, symptoms can be assessed through clinical tests; however, there are no imaging or laboratory tests fully reflective of pathophysiology routinely used by clinicians to characterize post-concussive symptoms. Magnetic resonance imaging has potential to link subtle pathophysiological alterations to clinical outcomes. Here, we review the state of the art of MRI research in adults with mild traumatic brain injury and provide recommendations to facilitate transition into clinical practice. Studies utilizing MRI can inform on pathophysiology of mild traumatic brain injury. They suggest presence of early cytotoxic and vasogenic oedema. They also show that mild traumatic brain injury results in cellular injury and microbleeds affecting the integrity of myelin and white matter tracts, all processes that appear to induce delayed vascular reactions and functional changes. Crucially, correlates between MRI parameters and post-concussive symptoms are emerging. Clinical sequences such as T1-weighted MRI, susceptibility-weighted MRI or fluid attenuation inversion recovery could be easily implementable in clinical practice, but are not sufficient, in isolation for prognostication. Diffusion sequences have shown promises and, although in need of analysis standardization, are a research priority. Lastly, arterial spin labelling is emerging as a high-utility research as it could become useful to assess delayed neurovascular response and possible long-term symptoms.
Collapse
Affiliation(s)
- Matilde Sassani
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Tara Ghafari
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Pradeepa R W Arachchige
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Iman Idrees
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Yidian Gao
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Alice Waitt
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Samuel R C Weaver
- Centre for Human Brain Health and School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Ali Mazaheri
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Hannah S Lyons
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Olivia Grech
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
| | - Mark Thaller
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Caroline Witton
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Andrew P Bagshaw
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Martin Wilson
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Hyojin Park
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| | - Matthew Brookes
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jan Novak
- College of Health and Life Sciences, Aston Institute of Health and Neurodevelopment, Aston University, Birmingham B4 7ET, UK
| | - Susan P Mollan
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Birmingham Neuro-ophthalmology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust Birmingham, Birmingham B15 2WB, UK
| | - Lisa J Hill
- Department of Biomedical Sciences, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Samuel J E Lucas
- Centre for Human Brain Health and School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - James L Mitchell
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Alexandra J Sinclair
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham B15 2TH, UK
- Department of Neurology, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham B15 2WB, UK
| | - Karen Mullinger
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
- Sir Peter Mansfield Imaging Centre, School of Physics and Astronomy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Davinia Fernández-Espejo
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham B15 2TT, UK
| |
Collapse
|
4
|
Brengel EK, Axe B, Maheswari A, Abeer MI, Ortiz RJ, Woodward TJ, Walhof R, Utama R, Sawada C, Balaji S, Kulkarni PP, Bradshaw HB, Gitcho MA, Ferris CF. Psilocybin as a Treatment for Repetitive Mild Head Injury: Evidence from Neuroradiology and Molecular Biology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636248. [PMID: 39975204 PMCID: PMC11838531 DOI: 10.1101/2025.02.03.636248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Repetitive mild head injuries incurred while playing organized sports, during car accidents and falls, or in active military service are a major health problem. These head injuries induce cognitive, motor, and behavioral deficits that can last for months and even years with an increased risk of dementia, Parkinson's disease, and chronic traumatic encephalopathy. There is no approved medical treatment for these types of head injuries. To this end, we tested the healing effects of the psychedelic psilocybin, as it is known to reduce neuroinflammation and enhance neuroplasticity. Using a model of mild repetitive head injury in adult female rats, we provide unprecedented data that psilocybin can reduce vasogenic edema, restore normal vascular reactivity and functional connectivity, reduce phosphorylated tau buildup, enhance levels of brain-derived neurotrophic factor and its receptor TrkB, and modulate lipid signaling molecules.
Collapse
Affiliation(s)
- Eric K Brengel
- Dept Psychology, Northeastern Univ, Boston, MA, USA
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Bryce Axe
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Ashwath Maheswari
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | | | | | - Taylor J Woodward
- Psychological & Brain Sciences, Program in Neuroscience, Indiana Univ, Bloomington, IN
| | - Reagan Walhof
- Psychological & Brain Sciences, Program in Neuroscience, Indiana Univ, Bloomington, IN
| | - Rachel Utama
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Courtney Sawada
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Shreyas Balaji
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Praveen P Kulkarni
- Dept Psychology, Northeastern Univ, Boston, MA, USA
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
| | - Heather B Bradshaw
- Psychological & Brain Sciences, Program in Neuroscience, Indiana Univ, Bloomington, IN
| | | | - Craig F Ferris
- Dept Psychology, Northeastern Univ, Boston, MA, USA
- Center for Translational Neuroimaging, Northeastern Univ, Boston, MA
- Dept Pharmaceutical Sciences, Northeastern Univ, Boston, MA
| |
Collapse
|
5
|
Özen I, Hamdeh SA, Ruscher K, Marklund N. Traumatic brain injury causes early aggregation of beta-amyloid peptides and NOTCH3 reduction in vascular smooth muscle cells of leptomeningeal arteries. Acta Neuropathol 2025; 149:10. [PMID: 39841284 PMCID: PMC11754316 DOI: 10.1007/s00401-025-02848-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/28/2024] [Accepted: 01/11/2025] [Indexed: 01/23/2025]
Abstract
Traumatic brain injury (TBI) often leads to impaired regulation of cerebral blood flow, which may be caused by pathological changes of the vascular smooth muscle cells (VSMCs) in the arterial wall. Moreover, these cerebrovascular changes may contribute to the development of various neurodegenerative disorders such as Alzheimer's-like pathologies that include amyloid beta aggregation. Despite its importance, the pathophysiological mechanisms responsible for VSMC dysfunction after TBI have rarely been evaluated. Here, we show that acute human TBI resulted in early pathological changes in leptomeningeal arteries, closely associated with a decrease in VSMC markers such as NOTCH3 and alpha smooth muscle actin (α-SMA).These changes coincided with increased aggregation of variable-length amyloid peptides including Aβ1-40/42, Aβ1-16, and β-secretase-derived fragment (βCTF) (C99) caused by altered processing of amyloid precursor protein (APP) in VSMCs. The aggregation of Aβ1-40/42 peptides were also observed in the leptomeningeal arteries of young TBI patients. These pathological changes also included higher β-secretase (BACE1) when compared to α-secretase A Disintegrin And Metalloprotease 10 (ADAM10) expression in the leptomeningeal arteries, plausibly caused by hypoxia and oxidative stress as shown using human VSMCs in vitro. Importantly, BACE1 inhibition not only restored NOTCH3 signalling but also normalized ADAM10 levels in vitro. Furthermore, we found reduced ADAM10 activity and decreased NOTCH3, along with increased βCTF (C99) levels in mice subjected to an experimental model of TBI. This study provides evidence of early post-injury changes in VSMCs of leptomeningeal arteries that can contribute to vascular dysfunction and exacerbate secondary injury mechanisms following TBI.
Collapse
MESH Headings
- Receptor, Notch3/metabolism
- Amyloid beta-Peptides/metabolism
- Animals
- Humans
- Brain Injuries, Traumatic/pathology
- Brain Injuries, Traumatic/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Male
- Mice
- Meningeal Arteries/pathology
- Meningeal Arteries/metabolism
- Female
- Mice, Inbred C57BL
- Adult
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Middle Aged
- Amyloid Precursor Protein Secretases/metabolism
- Young Adult
- Peptide Fragments/metabolism
Collapse
Affiliation(s)
- Ilknur Özen
- Department of Clinical Sciences, Lund Brain Injury Laboratory for Neurosurgical Research, Lund University, 222 20, Lund, Sweden
| | - Sami Abu Hamdeh
- Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden
| | - Karsten Ruscher
- Department of Clinical Sciences, Laboratory for Experimental Brain Research, Lund University, Lund, Sweden
| | - Niklas Marklund
- Department of Clinical Sciences, Lund Brain Injury Laboratory for Neurosurgical Research, Lund University, 222 20, Lund, Sweden.
- Department of Clinical Sciences Lund, Neurosurgery, Lund University, Skåne University Hospital, Lund, Sweden.
| |
Collapse
|
6
|
Monti DA, Faezeh V, Zabrecky G, Alizadeh M, Wintering N, Bazzan AJ, Mohamed FB, Newberg AB. Changes in Resting-State Functional Connectivity and Cognitive-Affective Symptoms in Patients With Post-Concussion Syndrome Treated With N-Acetyl Cysteine. J Head Trauma Rehabil 2024:00001199-990000000-00211. [PMID: 39531327 DOI: 10.1097/htr.0000000000000976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
OBJECTIVE Concussion accounts for more than 80% of people experiencing traumatic brain injury. Acute concussion is associated with characteristic cognitive and functional deficits that may persist for weeks to months. A subgroup of these patients (from 10% to 50%) have persistent symptoms referred to as chronic post-concussion syndrome (PCS). There are limited treatment options for these patients and the pathophysiology is poorly understood, though oxidative stress is thought to be a contributing factor. The purpose of this study was to evaluate whether an antioxidant, N-acetylcysteine (NAC), might be beneficial in patients with PCS. SETTING Outpatient medicine center. PARTICIPANTS Fifty patients with chronic PCS for at least 3 months post injury. DESIGN The patients with PCS were enrolled in this randomized unblinded clinical trial to receive the antioxidant NAC as a combination of daily oral and weekly intravenous infusions, or assigned to a waitlist control group where they would continue to receive standard of care. MAIN MEASURES Resting-state functional connectivity (FC) magnetic resonance imaging (rsFC-MRI) was performed pre and post either NAC or the waitlist period along with cognitive, emotional, and sensory symptom assessments. RESULTS The results demonstrated significant (P < .05) improvements in symptoms as determined by the Rivermead Post-Concussion Symptoms Questionnaire, Spielberger State-Trait Anxiety Inventory, and Profile of Mood Scale in the PCS group receiving NAC as compared to patients receiving ongoing standard care. Importantly, there were significant (P< .01) changes in FC in the NAC group, particularly in networks such as the default mode network, salience network, and executive control network. These changes in FC also correlated with improvements in symptoms. CONCLUSIONS In patients with chronic PCS, NAC treatment was associated with significant changes in resting state FC and improvement in a variety of symptoms, particularly cognitive and affective symptoms.
Collapse
Affiliation(s)
- Daniel A Monti
- Author Affiliations: Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA (Drs Monti and Zabrecky, Ms Wintering, and Drs Bazzan and Newberg); and Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, PA (Ms Vedaei and Drs Alizadeh, Mohamed, and Newberg)
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Caldas J, Cardim D, Edmundson P, Morales J, Feng A, Ashley JD, Park C, Valadka A, Foreman M, Cullum M, Sharma K, Liu Y, Zhu D, Zhang R, Ding K. Study protocol: Cerebral autoregulation, brain perfusion, and neurocognitive outcomes after traumatic brain injury -CAPCOG-TBI. Front Neurol 2024; 15:1465226. [PMID: 39479003 PMCID: PMC11521900 DOI: 10.3389/fneur.2024.1465226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 09/27/2024] [Indexed: 11/02/2024] Open
Abstract
Background Moderate-severe traumatic brain injury (msTBI) stands as a prominent etiology of adult disability, with increased risk for cognitive impairment and dementia. Although some recovery often occurs within the first year post-injury, predicting long-term cognitive outcomes remains challenging, partly due to the significant pathophysiological heterogeneity of TBI, including acute cerebrovascular injury. The primary aim of our recently funded study, cerebral autoregulation, brain perfusion, and neurocognitive outcomes after traumatic brain injury (CAPCOG-TBI), is to determine if acute cerebrovascular dysfunction after msTBI measured using multimodal non-invasive neuromonitoring is associated with cognitive outcome at 1-year post-injury. Methods This longitudinal observational study will be conducted at two Level 1 trauma centers in Texas, USA, and will include adult patients with msTBI, and/or mild TBI with neuroimaging abnormalities. Multimodal cerebral vascular assessment using transcranial Doppler and cerebral near-infrared spectroscopy (NIRS) will be conducted within 7-days of onset of TBI. Longitudinal outcomes, including cognitive/functional assessments (Glasgow Outcome Scale and Patient-Reported Outcomes Measurement Information System), cerebral vascular assessment, and imaging will be performed at follow-ups 3-, 6-, and 12-months post-injury. We aim to recruit 100 subjects with msTBI along with 30 orthopedic trauma controls (OTC). This study is funded by National Institute of Neurological Disease and Stroke (NINDS) and is registered on Clinicaltrial.org (NCT06480838). Expected results We anticipate that msTBI patients will exhibit impaired cerebrovascular function in the acute phase compared to the OTC group. The severity of cerebrovascular dysfunction during this stage is expected to inversely correlate with cognitive and functional outcomes at 1-year post-injury. Additionally, recovery from cerebrovascular dysfunction is expected to be linked to cognitive recovery. Conclusion The results of this study could help to understand the contribution of cerebrovascular dysfunction to cognitive outcomes after TBI and pave the way for innovative vascular-focused interventions aimed at enhancing cognitive recovery and mitigating neurodegeneration following msTB. In addition, its focus toward personalized medicine to aid in the management and prognosis of TBI patients.
Collapse
Affiliation(s)
- Juliana Caldas
- University of Texas Southwestern Medical Center, Dallas, TX, United States
- Bahiana School of Medicine and Public Health, Salvador, Bahia, Brazil
- D'or Institute for Research and Teaching, Salvador, Bahia, Brazil
| | - Danilo Cardim
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | | | - Jill Morales
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Aaron Feng
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | | | - Caroline Park
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Alex Valadka
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | | | - Munro Cullum
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kartavya Sharma
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Yulun Liu
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - David Zhu
- Albert Einstein College of Medicine, New York, NY, United States
| | - Rong Zhang
- University of Texas Southwestern Medical Center, Dallas, TX, United States
- Texas Health Resources, Dallas, TX, United States
| | - Kan Ding
- University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
8
|
Liu L, Huang R, Fan C, Chen X. Diagnostic and prognostic utility of plasma thrombospondin-1 levels in traumatic brain injury. Eur J Trauma Emerg Surg 2024; 50:2229-2237. [PMID: 39112761 DOI: 10.1007/s00068-024-02605-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 07/09/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE Thrombospondin-1 (TSP-1), a powerful antiangiogenic agent, is increasingly expressed in mice brain tissues after traumatic brain injury (TBI). However, in the peripheral blood of TBI patients, TSP-1 concentrations have not been identified. This study aimed to determine if TSP-1 measured in the plasma of patients relates to TBI diagnosis and injury severity. METHODS Plasma TSP-1 levels were assessed in 75 patients with mild to severe TBI and 60 healthy volunteers. Glasgow Coma Scale (GCS) score was recorded to assess traumatic severity. Other relevant clinical characters and laboratory tests were collected to evaluate the diagnostic efficiency of TSP-1. Glasgow outcome scale (GOSE) 3 months after trauma was dichotomized into unfavorable (GOSE1-4) and favorable (GOSE5-8) outcomes. RESULTS TSP-1 levels were significantly higher in TBI patients than in controls (median 530.4 ng/l, the upper- lower quartiles 373.2-782.1 vs. median 201.5 mg/l, the upper - lower quartiles 83.1-351.4, P < 0.001). Plasma TSP-1 was able to differentiate patients with mild, moderate, and severe TBI from healthy controls with Area Under the Receiver-Operating Characteristic Curve (AUROC) of 0.8089, 0.9312, and 0.9189, respectively. TSP-1 levels were closely and negatively correlated with GCS score (r = -0.41). TSP-1 levels > 624.4 ng/ml independently predicted a 3-month unfavorable outcome with an odds ratio value of 9.666 (95% confidence interval (CI),1.393-69.072). TSP-1 levels significantly discriminated 3-month unfavorable outcome with AUROC of 0.7445 (95%CI, 0.6152-0.8739). CONCLUSION The results of this study indicate that plasma TSP-1 should be further investigated as a diagnostic and prognostic marker for patients with TBI.
Collapse
Affiliation(s)
- Lei Liu
- Department of Laboratory Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Rongfu Huang
- Department of Laboratory Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Chunmei Fan
- Department of Laboratory Medicine, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| | - Xiangrong Chen
- Department of Neurosurgery, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
9
|
Bens N, Kulkarni P, Ferris CF. Changes in cerebral vascular reactivity following mild repetitive head injury in awake rats: modeling the human experience. Exp Brain Res 2024; 242:2433-2442. [PMID: 39162729 PMCID: PMC11422282 DOI: 10.1007/s00221-024-06907-7] [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: 03/29/2024] [Accepted: 08/01/2024] [Indexed: 08/21/2024]
Abstract
The changes in brain function in response to mild head injury are usually subtle and go undetected. Physiological biomarkers would aid in the early diagnosis of mild head injury. In this study we used hypercapnia to follow changes in cerebral vascular reactivity after repetitive mild head injury. We hypothesized head injury would reduce vascular reactivity. Rats were maintained on a reverse light-dark cycle and head impacted daily at 24 h intervals over three days. All head impacts were delivered while rats were fully awake under red light illumination. There was no neuroradiological evidence of brain damage. After the 3rd impact rats were exposed to 5% CO2 and imaged for changes in BOLD signal. All imaging was done while rats were awake without the confound of anesthesia. The data were registered to a 3D MRI rat atlas with 171 segmented brain areas providing site specific information on vascular reactivity. The changes in vascular reactivity were not uniform across the brain. The prefrontal cortex, somatosensory cortex and basal ganglia showed the hypothesized decrease in vascular reactivity while the cerebellum, thalamus, brainstem, and olfactory system showed an increase in BOLD signal to hypercapnia.
Collapse
Affiliation(s)
- Nicole Bens
- Center for Translational Neuroimaging, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Praveen Kulkarni
- Center for Translational Neuroimaging, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA
| | - Craig F Ferris
- Center for Translational Neuroimaging, Northeastern University, 360 Huntington Ave, Boston, MA, 02115, USA.
- Departments of Psychology and Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Whitehead BJ, Corbin D, Alexander ML, Bumgarner J, Zhang N, Karelina K, Weil ZM. Cerebral hypoperfusion exacerbates traumatic brain injury in male but not female mice. Eur J Neurosci 2024; 60:4346-4361. [PMID: 38858126 PMCID: PMC11533132 DOI: 10.1111/ejn.16439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/07/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Mild-moderate traumatic brain injuries (TBIs) are prevalent, and while many individuals recover, there is evidence that a significant number experience long-term health impacts, including increased vulnerability to neurodegenerative diseases. These effects are influenced by other risk factors, such as cardiovascular disease. Our study tested the hypothesis that a pre-injury reduction in cerebral blood flow (CBF), mimicking cardiovascular disease, worsens TBI recovery. We induced bilateral carotid artery stenosis (BCAS) and a mild-moderate closed-head TBI in male and female mice, either alone or in combination, and analyzed CBF, spatial learning, memory, axonal damage, and gene expression. Findings showed that BCAS and TBI independently caused a ~10% decrease in CBF. Mice subjected to both BCAS and TBI experienced more significant CBF reductions, notably affecting spatial learning and memory, particularly in males. Additionally, male mice showed increased axonal damage with both BCAS and TBI compared to either condition alone. Females exhibited spatial memory deficits due to BCAS, but these were not worsened by subsequent TBI. Gene expression analysis in male mice highlighted that TBI and BCAS individually altered neuronal and glial profiles. However, the combination of BCAS and TBI resulted in markedly different transcriptional patterns. Our results suggest that mild cerebrovascular impairments, serving as a stand-in for preexisting cardiovascular conditions, can significantly worsen TBI outcomes in males. This highlights the potential for mild comorbidities to modify TBI outcomes and increase the risk of secondary diseases.
Collapse
Affiliation(s)
- Bailey J. Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Deborah Corbin
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Megan L. Alexander
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Jacob Bumgarner
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Ning Zhang
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| | - Zachary M. Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown WV USA
| |
Collapse
|
13
|
Irastorza-Valera L, Soria-Gómez E, Benitez JM, Montáns FJ, Saucedo-Mora L. Review of the Brain's Behaviour after Injury and Disease for Its Application in an Agent-Based Model (ABM). Biomimetics (Basel) 2024; 9:362. [PMID: 38921242 PMCID: PMC11202129 DOI: 10.3390/biomimetics9060362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/28/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
The brain is the most complex organ in the human body and, as such, its study entails great challenges (methodological, theoretical, etc.). Nonetheless, there is a remarkable amount of studies about the consequences of pathological conditions on its development and functioning. This bibliographic review aims to cover mostly findings related to changes in the physical distribution of neurons and their connections-the connectome-both structural and functional, as well as their modelling approaches. It does not intend to offer an extensive description of all conditions affecting the brain; rather, it presents the most common ones. Thus, here, we highlight the need for accurate brain modelling that can subsequently be used to understand brain function and be applied to diagnose, track, and simulate treatments for the most prevalent pathologies affecting the brain.
Collapse
Affiliation(s)
- Luis Irastorza-Valera
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040 Madrid, Spain; (L.I.-V.); (J.M.B.); (F.J.M.)
- PIMM Laboratory, ENSAM–Arts et Métiers ParisTech, 151 Bd de l’Hôpital, 75013 Paris, France
| | - Edgar Soria-Gómez
- Achúcarro Basque Center for Neuroscience, Barrio Sarriena, s/n, 48940 Leioa, Spain;
- Ikerbasque, Basque Foundation for Science, Plaza Euskadi, 5, 48009 Bilbao, Spain
- Department of Neurosciences, University of the Basque Country UPV/EHU, Barrio Sarriena, s/n, 48940 Leioa, Spain
| | - José María Benitez
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040 Madrid, Spain; (L.I.-V.); (J.M.B.); (F.J.M.)
| | - Francisco J. Montáns
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040 Madrid, Spain; (L.I.-V.); (J.M.B.); (F.J.M.)
- Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Luis Saucedo-Mora
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040 Madrid, Spain; (L.I.-V.); (J.M.B.); (F.J.M.)
- Department of Materials, University of Oxford, Parks Road, Oxford OX1 3PJ, UK
- Department of Nuclear Science and Engineering, Massachusetts Institute of Technology (MIT), 77 Massachusetts Ave, Cambridge, MA 02139, USA
| |
Collapse
|
14
|
Elser H, Pappalardo LW, Gottesman RF, Coresh J, Diaz-Arrastia R, Mosley TH, Kasner SE, Koton S, Schneider ALC. Head Injury and Risk of Incident Ischemic Stroke in Community-Dwelling Adults. Stroke 2024; 55:1562-1571. [PMID: 38716662 PMCID: PMC11126353 DOI: 10.1161/strokeaha.123.046443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 03/29/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND While stroke is a recognized short-term sequela of traumatic brain injury, evidence about long-term ischemic stroke risk after traumatic brain injury remains limited. METHODS The Atherosclerosis Risk in Communities Study is an ongoing prospective cohort comprised of US community-dwelling adults enrolled in 1987 to 1989 followed through 2019. Head injury was defined using self-report and hospital-based diagnostic codes and was analyzed as a time-varying exposure. Incident ischemic stroke events were physician-adjudicated. We used Cox regression adjusted for sociodemographic and cardiovascular risk factors to estimate the hazard of ischemic stroke as a function of head injury. Secondary analyses explored the number and severity of head injuries; the mechanism and severity of incident ischemic stroke; and heterogeneity within subgroups defined by race, sex, and age. RESULTS Our analysis included 12 813 participants with no prior head injury or stroke. The median follow-up age was 27.1 years (25th-75th percentile=21.1-30.5). Participants were of median age 54 years (25th-75th percentile=49-59) at baseline; 57.7% were female and 27.8% were Black. There were 2158 (16.8%) participants with at least 1 head injury and 1141 (8.9%) participants with an incident ischemic stroke during follow-up. For those with head injuries, the median age to ischemic stroke was 7.5 years (25th-75th percentile=2.2-14.0). In adjusted models, head injury was associated with an increased hazard of incident ischemic stroke (hazard ratio [HR], 1.34 [95% CI, 1.12-1.60]). We observed evidence of dose-response for the number of head injuries (1: HR, 1.16 [95% CI, 0.97-1.40]; ≥2: HR, 1.94 [95% CI, 1.39-2.71]) but not for injury severity. We observed evidence of stronger associations between head injury and more severe stroke (National Institutes of Health Stroke Scale score ≤5: HR, 1.31 [95% CI, 1.04-1.64]; National Institutes of Health Stroke Scale score 6-10: HR, 1.64 [95% CI, 1.06-2.52]; National Institutes of Health Stroke Scale score ≥11: HR, 1.80 [95% CI, 1.18-2.76]). Results were similar across stroke mechanism and within strata of race, sex, and age. CONCLUSIONS In this community-based cohort, head injury was associated with subsequent ischemic stroke. These results suggest the importance of public health interventions aimed at preventing head injuries and primary stroke prevention among individuals with prior traumatic brain injuries.
Collapse
Affiliation(s)
- Holly Elser
- Department of Neurology (H.E., L.W.P., R.D.-A., S.E.K., A.L.C.S.), University of Pennsylvania, Philadelphia
| | - Laura W Pappalardo
- Department of Neurology (H.E., L.W.P., R.D.-A., S.E.K., A.L.C.S.), University of Pennsylvania, Philadelphia
| | - Rebecca F Gottesman
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD (R.F.G.)
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (J.C., S.K.)
| | - Ramon Diaz-Arrastia
- Department of Neurology (H.E., L.W.P., R.D.-A., S.E.K., A.L.C.S.), University of Pennsylvania, Philadelphia
| | - Thomas H Mosley
- The Memory Impairment and Neurodegenerative Dementia Center, University of Mississippi Medical Center, Jackson (T.H.M.)
| | - Scott E Kasner
- Department of Neurology (H.E., L.W.P., R.D.-A., S.E.K., A.L.C.S.), University of Pennsylvania, Philadelphia
| | - Silvia Koton
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (J.C., S.K.)
- School of Health Professions, Tel Aviv University, Israel (S.K.)
| | - Andrea L C Schneider
- Department of Neurology (H.E., L.W.P., R.D.-A., S.E.K., A.L.C.S.), University of Pennsylvania, Philadelphia
- Department of Biostatistics, Epidemiology, and Informatics, School of Medicine (A.L.C.S.), University of Pennsylvania, Philadelphia
| |
Collapse
|
15
|
Mokbel AY, Burns MP, Main BS. The contribution of the meningeal immune interface to neuroinflammation in traumatic brain injury. J Neuroinflammation 2024; 21:135. [PMID: 38802931 PMCID: PMC11131220 DOI: 10.1186/s12974-024-03122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Traumatic brain injury (TBI) is a major cause of disability and mortality worldwide, particularly among the elderly, yet our mechanistic understanding of what renders the post-traumatic brain vulnerable to poor outcomes, and susceptible to neurological disease, is incomplete. It is well established that dysregulated and sustained immune responses elicit negative consequences after TBI; however, our understanding of the neuroimmune interface that facilitates crosstalk between central and peripheral immune reservoirs is in its infancy. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in both healthy and disease settings. It has been previously shown that disruption of this system exacerbates neuroinflammation in age-related neurodegenerative disorders such as Alzheimer's disease; however, we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. In this manuscript, we will offer a detailed overview of the holistic nature of neuroinflammatory responses in TBI, including hallmark features observed across clinical and animal models. We will highlight the structure and function of the meningeal lymphatic system, including its role in immuno-surveillance and immune responses within the meninges and the brain. We will provide a comprehensive update on our current knowledge of meningeal-derived responses across the spectrum of TBI, and identify new avenues for neuroimmune modulation within the neurotrauma field.
Collapse
Affiliation(s)
- Alaa Y Mokbel
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Mark P Burns
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Bevan S Main
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
16
|
Yu J, Joo IL, Bazzigaluppi P, Koletar MM, Cherin E, Stanisz AG, Graham JWC, Demore C, Stefanovic B. Micro-ultrasound based characterization of cerebrovasculature following focal ischemic stroke and upon short-term rehabilitation. J Cereb Blood Flow Metab 2024; 44:461-476. [PMID: 37974304 PMCID: PMC10981404 DOI: 10.1177/0271678x231215004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 08/21/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023]
Abstract
Notwithstanding recanalization treatments in the acute stage of stroke, many survivors suffer long-term impairments. Physical rehabilitation is the only widely available strategy for chronic-stage recovery, but its optimization is hindered by limited understanding of its effects on brain structure and function. Using micro-ultrasound, behavioral testing, and electrophysiology, we investigated the impact of skilled reaching rehabilitation on cerebral hemodynamics, motor function, and neuronal activity in a rat model of focal ischemic stroke. A 50 MHz micro-ultrasound transducer and intracortical electrophysiology were utilized to characterize neurovascular changes three weeks following focal ischemia elicited by endothelin-1 injection into the sensorimotor cortex. Sprague-Dawley rats were rehabilitated through tray reaching, and their fine skilled reaching was assessed via the Montoya staircase. Focal ischemia led to a sustained deficit in forelimb reaching; and increased tortuosity of the penetrating vessels in the perilesional cortex; with no lateralization of spontaneous neuronal activity. Rehabilitation improved skilled reaching; decreased cortical vascularity; was associated with elevated peri- vs. contralesional hypercapnia-induced flow homogenization and increased perilesional spontaneous cortical neuronal activity. Our study demonstrated neurovascular plasticity accompanying rehabilitation-elicited functional recovery in the subacute stage following stroke, and multiple micro-ultrasound-based markers of cerebrovascular structure and function modified in recovery from ischemia and upon rehabilitation.
Collapse
Affiliation(s)
- Johnson Yu
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Illsung L Joo
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Paolo Bazzigaluppi
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
- MetaCell, Cagliari, Italy
| | - Margaret M Koletar
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Emmanuel Cherin
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Andrew G Stanisz
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - James WC Graham
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Christine Demore
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Bojana Stefanovic
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Chen Y, Gu M, Patterson J, Zhang R, Statz JK, Reed E, Abutarboush R, Ahlers ST, Kawoos U. Temporal Alterations in Cerebrovascular Glycocalyx and Cerebral Blood Flow after Exposure to a High-Intensity Blast in Rats. Int J Mol Sci 2024; 25:3580. [PMID: 38612392 PMCID: PMC11011510 DOI: 10.3390/ijms25073580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/09/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
The glycocalyx is a proteoglycan-glycoprotein structure lining the luminal surface of the vascular endothelium and is susceptible to damage due to blast overpressure (BOP) exposure. The glycocalyx is essential in maintaining the structural and functional integrity of the vasculature and regulation of cerebral blood flow (CBF). Assessment of alterations in the density of the glycocalyx; its components (heparan sulphate proteoglycan (HSPG/syndecan-2), heparan sulphate (HS), and chondroitin sulphate (CS)); CBF; and the effect of hypercapnia on CBF was conducted at 2-3 h, 1, 3, 14, and 28 days after a high-intensity (18.9 PSI/131 kPa peak pressure, 10.95 ms duration, and 70.26 PSI·ms/484.42 kPa·ms impulse) BOP exposure in rats. A significant reduction in the density of the glycocalyx was observed 2-3 h, 1-, and 3 days after the blast exposure. The glycocalyx recovered by 28 days after exposure and was associated with an increase in HS (14 and 28 days) and in HSPG/syndecan-2 and CS (28 days) in the frontal cortex. In separate experiments, we observed significant decreases in CBF and a diminished response to hypercapnia at all time points with some recovery at 3 days. Given the role of the glycocalyx in regulating physiological function of the cerebral vasculature, damage to the glycocalyx after BOP exposure may result in the onset of pathogenesis and progression of cerebrovascular dysfunction leading to neuropathology.
Collapse
Affiliation(s)
- Ye Chen
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Ming Gu
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Jacob Patterson
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- Parsons Corporation, Columbia, MD 21046, USA
| | - Ruixuan Zhang
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Jonathan K. Statz
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Eileen Reed
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- Parsons Corporation, Columbia, MD 21046, USA
| | - Rania Abutarboush
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Stephen T. Ahlers
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
| | - Usmah Kawoos
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (Y.C.); (M.G.)
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| |
Collapse
|
18
|
Arndt P, Chahem C, Luchtmann M, Kuschel JN, Behme D, Pfister M, Neumann J, Görtler M, Dörner M, Pawlitzki M, Jansen R, Meuth SG, Vielhaber S, Henneicke S, Schreiber S. Risk factors for intracerebral hemorrhage in small-vessel disease and non-small-vessel disease etiologies-an observational proof-of-concept study. Front Neurol 2024; 15:1322442. [PMID: 38515448 PMCID: PMC10954881 DOI: 10.3389/fneur.2024.1322442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
Background Sporadic cerebral small-vessel disease (CSVD), i.e., hypertensive arteriopathy (HA) and cerebral amyloid angiopathy (CAA), is the main cause of spontaneous intracerebral hemorrhage (ICH). Nevertheless, a substantial portion of ICH cases arises from non-CSVD etiologies, such as trauma, vascular malformations, and brain tumors. While studies compared HA- and CAA-related ICH, non-CSVD etiologies were excluded from these comparisons and are consequently underexamined with regard to additional factors contributing to increased bleeding risk beyond their main pathology. Methods As a proof of concept, we conducted a retrospective observational study in 922 patients to compare HA, CAA, and non-CSVD-related ICH with regard to factors that are known to contribute to spontaneous ICH onset. Medical records (available for n = 861) were screened for demographics, antithrombotic medication, and vascular risk profile, and CSVD pathology was rated on magnetic resonance imaging (MRI) in a subgroup of 185 patients. The severity of CSVD was assessed with a sum score ranging from 0 to 6, where a score of ≥2 was defined as advanced pathology. Results In 922 patients with ICH (median age of 71 years), HA and CAA caused the majority of cases (n = 670, 73%); non-CSVD etiologies made up the remaining quarter (n = 252, 27%). Individuals with HA- and CAA-related ICH exhibited a higher prevalence of predisposing factors than those with non-CSVD etiologies. This includes advanced age (median age: 71 vs. 75 vs. 63 years, p < 0.001), antithrombotic medication usage (33 vs. 37 vs. 19%, p < 0.001), prevalence of vascular risk factors (70 vs. 67 vs. 50%, p < 0.001), and advanced CSVD pathology on MRI (80 vs. 89 vs. 51%, p > 0.001). However, in particular, half of non-CSVD ICH patients were either aged over 60 years, presented with vascular risk factors, or had advanced CSVD on MRI. Conclusion Risk factors for spontaneous ICH are less common in non-CSVD ICH etiologies than in HA- and CAA-related ICH, but are still frequent. Future studies should incorporate these factors, in addition to the main pathology, to stratify an individual's risk of bleeding.
Collapse
Affiliation(s)
- Philipp Arndt
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - Christian Chahem
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Michael Luchtmann
- Department of Neurosurgery, Paracelsus-Klinik, Zwickau, Germany
- Department of Neurosurgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan-Niklas Kuschel
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniel Behme
- Department of Neuroradiology, Otto-von-Guericke University, Magdeburg, Germany
| | - Malte Pfister
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Jens Neumann
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Michael Görtler
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Marc Dörner
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
- Department of Consultation-Liaison-Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Marc Pawlitzki
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Robin Jansen
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sven G. Meuth
- Department of Neurology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Stefan Vielhaber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| | - Solveig Henneicke
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
| | - Stefanie Schreiber
- Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE) within the Helmholtz Association, Magdeburg, Germany
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany
| |
Collapse
|
19
|
Wallace C, Smirl JD, Adhikari SP, Jones KE, Rieger M, Rothlander K, van Donkelaar P. Neurovascular coupling is altered in women who have a history of brain injury from intimate partner violence: a preliminary study. Front Glob Womens Health 2024; 5:1344880. [PMID: 38495125 PMCID: PMC10940333 DOI: 10.3389/fgwh.2024.1344880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/22/2024] [Indexed: 03/19/2024] Open
Abstract
Introduction Intimate partner violence (IPV) is a global health crisis with 30% of women over the age of 15 experiencing at least one event in their lifetime. Brain injury (BI) due to head impacts and/or strangulation is a common but understudied part of this experience. Previous research has shown BI from other injury mechanisms can disrupt neurovascular coupling (NVC). To gain further insight into whether similar changes occur in this population, we assessed NVC responses in women with a history of IPV-BI. Methods NVC responses were measured for the middle and posterior cerebral arteries (MCA, PCA) using transcranial Doppler ultrasound while participants performed a complex visual search task. The lifetime history of previous exposure to IPV-BI was captured using the Brain Injury Severity Assessment (BISA) along with measures of post-traumatic stress disorder (PTSD), anxiety, depression, substance use, and demographic information. Initial analyses of NVC metrics were completed comparing participants who scored low vs. high on the BISA or did or did not experience non-fatal strangulation followed by a stepwise multiple regression to examine the impact of PTSD, anxiety, and depression on the relationship between the NVC metrics and IPV-BI. Results Baseline and peak cerebral blood velocity were higher and the percentage increase was lower in the PCA in the low compared to the high BISA group whereas no differences between the groups were apparent in the MCA. In addition, those participants who had been strangled had a lower initial slope and area under the curve in the PCA than those who had not experienced strangulation. Finally, the stepwise multiple regression demonstrated the percentage increase in the PCA was significantly related to the BISA score and both depression and anxiety significantly contributed to different components of the NVC response. Conclusions This preliminary study demonstrated that a lifetime history of IPV-BI leads to subtle but significant disruptions to NVC responses which are modulated by comorbid depression and anxiety. Future studies should examine cerebrovascular function at the acute and subacute stages after IPV episodes to shed additional light on this experience and its outcomes.
Collapse
Affiliation(s)
- Colin Wallace
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
- Department of Kinesiology, Okanagan College, Penticton, BC, Canada
| | - Jonathan D. Smirl
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
- Sport Injury Prevention Research Centre, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Cerebrovascular Concussion Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
- Integrated Concussion Research Program, University of Calgary, Calgary, AB, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute of Alberta, University of Calgary, Calgary, AB, Canada
| | - Shambhu P. Adhikari
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - K. Elisabeth Jones
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Matt Rieger
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
- Faculty of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Krystal Rothlander
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| | - Paul van Donkelaar
- School of Health and Exercise Sciences, University of British Columbia, Kelowna, BC, Canada
| |
Collapse
|
20
|
Jaafari O, Salih S, Alkatheeri A, Alshehri M, Al-Shammari M, Maeni M, Alqahtani A, Alomaim W, Hasaneen M. Appropriate incorporation of susceptibility-weighted magnetic resonance imaging into routine imaging protocols for accurate diagnosis of traumatic brain injuries: a systematic review. J Med Life 2024; 17:273-280. [PMID: 39044937 PMCID: PMC11262612 DOI: 10.25122/jml-2023-0487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/12/2024] [Indexed: 07/25/2024] Open
Abstract
Traumatic brain injury (TBI) results from physical or traumatic injuries to the brain's surrounding bony structures and associated tissues, which can lead to various sequelae, including simple concussion, acute epidural hematoma, parenchymal contusions, subarachnoid hemorrhage, diffuse axonal injury, and chronic traumatic encephalopathy. Susceptibility-weighted imaging (SWI) has enhanced the accuracy of neuroimaging for these injuries. SWI is based on 3D gradient echo magnetic resonance imaging (MRI) with long echo times and flow compensation. Owing to its sensitivity to deoxyhemoglobin, hemosiderin, iron, and calcium, SWI is extremely informative and superior to conventional MRI for the diagnosis and follow-up of patients with acute, subacute, and prolonged hemorrhage. This systematic review aimed to evaluate and summarize the published articles that report SWI results for the evaluation of TBI and to determine correlations between clinical status and SWI results. Consequently, our analysis also aimed to identify the appropriate MRI sequences to use in the assessment of patients with TBI. We searched the Medline and Embase online electronic databases for relevant papers published from 2012 onwards. We found that SWI had higher sensitivity than gradient echo MRI in detecting and characterizing microbleeds in TBIs and was able to differentiate diamagnetic calcifications from paramagnetic microhemorrhages. However, it is important that future research not only continues to evaluate the utility of SWI in TBIs but also attempts to overcome the limitations of the studies described in this review, which should help validate the conclusions and recommendations from our analysis.
Collapse
Affiliation(s)
- Osama Jaafari
- Radiology Department, Royal Commission Medical Center, King Fahad, Al-Nakheel, Yanbu, Saudi Arabia
| | - Suliman Salih
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Ajnas Alkatheeri
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Muhamed Alshehri
- Department of Radiology and Medical Imaging, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Majedh Al-Shammari
- Department of Radiological Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Mousa Maeni
- Radiology Department, Royal Commission Medical Center, King Fahad, Al-Nakheel, Yanbu, Saudi Arabia
| | - Abdullah Alqahtani
- Radiology Department, Royal Commission Medical Center, King Fahad, Al-Nakheel, Yanbu, Saudi Arabia
| | - Wijdan Alomaim
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Al Ain, United Arab Emirates
| | - Mohamed Hasaneen
- Department of Radiography and Medical Imaging, Fatima College of Health Sciences, Al Ain, United Arab Emirates
| |
Collapse
|
21
|
Ali HT, Sula I, AbuHamdia A, Elejla SA, Elrefaey A, Hamdar H, Elfil M. Nervous System Response to Neurotrauma: A Narrative Review of Cerebrovascular and Cellular Changes After Neurotrauma. J Mol Neurosci 2024; 74:22. [PMID: 38367075 PMCID: PMC10874332 DOI: 10.1007/s12031-024-02193-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/22/2024] [Indexed: 02/19/2024]
Abstract
Neurotrauma is a significant cause of morbidity and mortality worldwide. For instance, traumatic brain injury (TBI) causes more than 30% of all injury-related deaths in the USA annually. The underlying cause and clinical sequela vary among cases. Patients are liable to both acute and chronic changes in the nervous system after such a type of injury. Cerebrovascular disruption has the most common and serious effect in such cases because cerebrovascular autoregulation, which is one of the main determinants of cerebral perfusion pressure, can be effaced in brain injuries even in the absence of evident vascular injury. Disruption of the blood-brain barrier regulatory function may also ensue whether due to direct injury to its structure or metabolic changes. Furthermore, the autonomic nervous system (ANS) can be affected leading to sympathetic hyperactivity in many patients. On a cellular scale, the neuroinflammatory cascade medicated by the glial cells gets triggered in response to TBI. Nevertheless, cellular and molecular reactions involved in cerebrovascular repair are not fully understood yet. Most studies were done on animals with many drawbacks in interpreting results. Therefore, future studies including human subjects are necessarily needed. This review will be of relevance to clinicians and researchers interested in understanding the underlying mechanisms in neurotrauma cases and the development of proper therapies as well as those with a general interest in the neurotrauma field.
Collapse
Affiliation(s)
| | - Idris Sula
- College of Medicine, Sulaiman Al Rajhi University, Al Bukayriyah, Al Qassim, Saudi Arabia
| | - Abrar AbuHamdia
- Department of Medical Laboratory Science, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus, Palestine
| | | | | | - Hiba Hamdar
- Medical Learning Skills Academy, Beirut, Lebanon
- Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Mohamed Elfil
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
22
|
Liang Y, Wang Y, Sun C, Xiang Y, Deng Y. Deferoxamine reduces endothelial ferroptosis and protects cerebrovascular function after experimental traumatic brain injury. Brain Res Bull 2024; 207:110878. [PMID: 38218407 DOI: 10.1016/j.brainresbull.2024.110878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 12/20/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Cerebrovascular dysfunction resulting from traumatic brain injury (TBI) significantly contributes to poor patient outcomes. Recent studies revealed the involvement of iron metabolism in neuronal survival, yet its effect on vasculature remains unclear. This study aims to explore the impact of endothelial ferroptosis on cerebrovascular function in TBI. A Controlled Cortical Impact (CCI) model was established in mice, resulting in a significant increase in iron-related proteins such as TfR1, FPN1, and FTH, as well as oxidative stress biomarker 4HNE. This was accompanied by a decline in expression of the ferroptosis inhibitor GPX4. Moreover, Perls' staining and nonhemin iron content assay showed iron overload in brain microvascular endothelial cells (BMECs) and the ipsilateral cortex. Immunofluorescence staining revealed more FTH-positive cerebral endothelial cells, consistent with impaired perfusion vessel density and cerebral blood flow. As a specific iron chelator, deferoxamine (DFO) treatment inhibited such ferroptotic proteins expression and the accumulation of lipid-reactive oxygen species following CCI, enhancing glutathione peroxidase (GPx) activity. DFO treatment significantly reduced iron deposition in BMECs and brain tissue, and increased density of the cerebral capillaries as well. Consequently, DFO treatment led to improvements in cerebral blood flow (as measured by laser speckle imaging) and behavioral performance (as measured by the neurological severity scores, rotarod test, and Morris water maze test). Taken together, our results indicated that TBI induces remarkable iron disorder and endothelial ferroptosis, and DFO treatment may help maintain iron homeostasis and protect vascular function. This may provide a novel therapeutic strategy to prevent cerebrovascular dysfunction following TBI.
Collapse
Affiliation(s)
- Yidan Liang
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Yanglingxi Wang
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Chao Sun
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Yi Xiang
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China
| | - Yongbing Deng
- Department of Neurosurgery, Chongqing University Central Hospital, Chongqing Emergency Medical Center, Chongqing, China; Chongqing Key Laboratory of Emergency Medicine, Chongqing, China.
| |
Collapse
|
23
|
Agoston DV. Traumatic Brain Injury in the Long-COVID Era. Neurotrauma Rep 2024; 5:81-94. [PMID: 38463416 PMCID: PMC10923549 DOI: 10.1089/neur.2023.0067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Major determinants of the biological background or reserve, such as age, biological sex, comorbidities (diabetes, hypertension, obesity, etc.), and medications (e.g., anticoagulants), are known to affect outcome after traumatic brain injury (TBI). With the unparalleled data richness of coronavirus disease 2019 (COVID-19; ∼375,000 and counting!) as well as the chronic form, long-COVID, also called post-acute sequelae SARS-CoV-2 infection (PASC), publications (∼30,000 and counting) covering virtually every aspect of the diseases, pathomechanisms, biomarkers, disease phases, symptomatology, etc., have provided a unique opportunity to better understand and appreciate the holistic nature of diseases, interconnectivity between organ systems, and importance of biological background in modifying disease trajectories and affecting outcomes. Such a holistic approach is badly needed to better understand TBI-induced conditions in their totality. Here, I briefly review what is known about long-COVID/PASC, its underlying-suspected-pathologies, the pathobiological changes induced by TBI, in other words, the TBI endophenotypes, discuss the intersection of long-COVID/PASC and TBI-induced pathobiologies, and how by considering some of the known factors affecting the person's biological background and the inclusion of mechanistic molecular biomarkers can help to improve the clinical management of TBI patients.
Collapse
Affiliation(s)
- Denes V. Agoston
- Department of Anatomy, Physiology, and Genetics, School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| |
Collapse
|
24
|
Kilgore MO, Hubbard WB. Effects of Low-Level Blast on Neurovascular Health and Cerebral Blood Flow: Current Findings and Future Opportunities in Neuroimaging. Int J Mol Sci 2024; 25:642. [PMID: 38203813 PMCID: PMC10779081 DOI: 10.3390/ijms25010642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/20/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Low-level blast (LLB) exposure can lead to alterations in neurological health, cerebral vasculature, and cerebral blood flow (CBF). The development of cognitive issues and behavioral abnormalities after LLB, or subconcussive blast exposure, is insidious due to the lack of acute symptoms. One major hallmark of LLB exposure is the initiation of neurovascular damage followed by the development of neurovascular dysfunction. Preclinical studies of LLB exposure demonstrate impairment to cerebral vasculature and the blood-brain barrier (BBB) at both early and long-term stages following LLB. Neuroimaging techniques, such as arterial spin labeling (ASL) using magnetic resonance imaging (MRI), have been utilized in clinical investigations to understand brain perfusion and CBF changes in response to cumulative LLB exposure. In this review, we summarize neuroimaging techniques that can further our understanding of the underlying mechanisms of blast-related neurotrauma, specifically after LLB. Neuroimaging related to cerebrovascular function can contribute to improved diagnostic and therapeutic strategies for LLB. As these same imaging modalities can capture the effects of LLB exposure in animal models, neuroimaging can serve as a gap-bridging diagnostic tool that permits a more extensive exploration of potential relationships between blast-induced changes in CBF and neurovascular health. Future research directions are suggested, including investigating chronic LLB effects on cerebral perfusion, exploring mechanisms of dysautoregulation after LLB, and measuring cerebrovascular reactivity (CVR) in preclinical LLB models.
Collapse
Affiliation(s)
- Madison O. Kilgore
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA;
| | - W. Brad Hubbard
- Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY 40536, USA;
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
- Lexington Veterans’ Affairs Healthcare System, Lexington, KY 40502, USA
| |
Collapse
|
25
|
Jeleff A, Suh N, Schranc Á, Diaper J, Bendjelid K, Schiffer E. New Noninvasive Method for the Assessment of Central Venous Oxygen Saturations in Critically Ill Patients. J Cardiothorac Vasc Anesth 2024; 38:170-174. [PMID: 37827917 DOI: 10.1053/j.jvca.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 10/14/2023]
Abstract
OBJECTIVES To compare noninvasive external jugular vein oxygen saturations (SjvO2) and central venous oxygen saturation (ScvO2) from a blood sample in patients admitted to the intensive care unit. DESIGN A prospective, comparative, monocentric clinical trial design was used. SETTING The study was performed in the Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospitals of Geneva (Switzerland). PARTICIPANTS A total of 79 patients were enrolled; patients with confirmed COVID-19 infection requiring invasive mechanical ventilation (patients with COVID-19, n = 36) and patients after liver transplantation (posttransplant patients, n = 43). INTERVENTIONS Simultaneous measurement of SjvO2 by near-infrared spectroscopy and ScvO2 from central venous blood samples using a blood gas analyzer in stable hemodynamic conditions. MEASUREMENTS AND MAIN RESULTS A strong linear correlation was evidenced in both the COVID-19 and posttransplant patient groups between the 2 modalities. The Bland-Altman analysis showed low bias in accordance with low percentage error in both groups (0.57% and 8.09% for patients with COVID-19; 0.00% and 13.72% for posttransplant patients). CONCLUSIONS Central venous oxygen saturation can be estimated reasonably by the continuous noninvasive measurement of SjvO2 using near-infrared spectroscopy.
Collapse
Affiliation(s)
- Alexandre Jeleff
- Division of Anaesthesiology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Noémie Suh
- Division of Intensive Care, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Álmos Schranc
- Unit for Anaesthesiological Investigations, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, Geneva, Switzerland.
| | - John Diaper
- Division of Anaesthesiology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospitals of Geneva, Geneva, Switzerland; Unit for Anaesthesiological Investigations, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University of Geneva, Geneva, Switzerland
| | - Karim Bendjelid
- Division of Intensive Care, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospitals of Geneva, Geneva, Switzerland
| | - Eduardo Schiffer
- Division of Anaesthesiology, Department of Anaesthesiology, Pharmacology, Intensive Care and Emergency Medicine, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
26
|
Thomas R, Lynch CE, Debad J, Campbell C, Chidomere O, Kilianski J, Ding K, Madden C, Sandsmark DK, Diaz-Arrastia R, Gatson JW. Plasma von Willebrand Factor Is Elevated Hyperacutely After Mild Traumatic Brain Injury. Neurotrauma Rep 2023; 4:655-662. [PMID: 37908322 PMCID: PMC10615084 DOI: 10.1089/neur.2023.0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2023] Open
Abstract
Each year in the United States, ∼2.7 million persons seek medical attention for traumatic brain injury (TBI), of which ∼85% are characterized as being mild brain injuries. Many different cell types in the brain are affected in these heterogeneous injuries, including neurons, glia, and the brain vasculature. Efforts to identify biomarkers that reflect the injury of these different cell types have been a focus of ongoing investigation. We hypothesized that von Willebrand factor (vWF) is a sensitive biomarker for acute traumatic vascular injury and correlates with symptom severity post-TBI. To address this, blood was collected from professional boxing athletes (n = 17) before and within 30 min after competition. Plasma levels of vWF and neuron-specific enolase were measured using the Meso Scale Discovery, LLC. (MSD) electrochemiluminescence array-based multi-plex format (MSD, Gaithersburg, MD). Additional symptom and outcome data from boxers and patients, such as the Rivermead symptom scores (Rivermead Post Concussion Symptoms Questionnaire [RPQ-3]), were collected. We found that, subsequent to boxing bouts, there was a 1.8-fold increase in vWF levels within 30 min of injury (p < 0.0009). Moreover, fold-change in vWF correlates moderately (r = 0.51; p = 0.03) with the number of head blows. We also found a positive correlation (r = 0.69; p = 0.002) between fold-change in vWF and self-reported post-concussive symptoms, measured by the RPQ-3. The receiver operating curve analysis of vWF plasma levels and RPQ-3 scoring yielded a sensitivity of 94.12% and a specificity of 76.5% with an area under the curve of 83% for boxers after a fight compared to the pre-bout baseline. This study suggests that vWF is a potential blood biomarker measurable in the hyperacute period after blunt mild TBI. This biomarker may prove to be useful in diagnosing and monitoring traumatic vascular injury.
Collapse
Affiliation(s)
- Rachel Thomas
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Cillian E. Lynch
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Jeff Debad
- Meso Scale Diagnostics, LLC, Rockville, Maryland, USA
| | | | - Onyinyechi Chidomere
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joseph Kilianski
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kan Ding
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Madden
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Danielle K. Sandsmark
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Joshua W. Gatson
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- General Dynamics Information Technology, Falls Church, Virginia, USA
| |
Collapse
|
27
|
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
|
28
|
Langri DS, Sunar U. Non-Invasive Continuous Optical Monitoring of Cerebral Blood Flow after Traumatic Brain Injury in Mice Using Fiber Camera-Based Speckle Contrast Optical Spectroscopy. Brain Sci 2023; 13:1365. [PMID: 37891734 PMCID: PMC10605647 DOI: 10.3390/brainsci13101365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 10/29/2023] Open
Abstract
Neurocritical care focuses on monitoring cerebral blood flow (CBF) to prevent secondary brain injuries before damage becomes irreversible. Thus, there is a critical unmet need for continuous neuromonitoring methods to quantify CBF within the vulnerable cortex continuously and non-invasively. Animal models and imaging biomarkers can provide valuable insights into the mechanisms and kinetics of head injury, as well as insights for potential treatment strategies. For this purpose, we implemented an optical technique for continuous monitoring of blood flow changes after a closed head injury in a mouse model, which is based on laser speckle contrast imaging and a fiber camera-based approach. Our results indicate a significant decrease (~10%, p-value < 0.05) in blood flow within 30 min of a closed head injury. Furthermore, the low-frequency oscillation analysis also indicated much lower power in the trauma group compared to the control group. Overall, blood flow has the potential to be a biomarker for head injuries in the early phase of a trauma, and the system is useful for continuous monitoring with the potential for clinical translation.
Collapse
Affiliation(s)
- Dharminder S. Langri
- Department of Biomedical Engineering, Wright State University, Dayton, OH 45435, USA;
| | - Ulas Sunar
- Department of Biomedical Engineering, Stony Brook University, New York, NY 11794, USA
| |
Collapse
|
29
|
Knight LS, Knight TA. Making the case for prophylactic use of betaine to promote brain health in young (15-24 year old) athletes at risk for concussion. Front Neurosci 2023; 17:1214976. [PMID: 37811321 PMCID: PMC10556504 DOI: 10.3389/fnins.2023.1214976] [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: 05/01/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Betaine supplementation in the context of human nutrition, athletic performance, and clinical therapy demonstrate that the osmolyte and methyl donor, betaine, is cytoprotective and beneficial to human health. These studies also demonstrate that betaine supplementation in healthy humans is straight-forward with no reported adverse effects. Here, we explore betaine uptake in the central nervous system (CNS) and contribute to evidence that betaine may be uniquely protective to the brain. We specifically describe the therapeutic potential of betaine and explore the potential implications of betaine on inhibition mediated by GABA and glycine neurotransmission. The influence of betaine on neurophysiology complement betaine's role as an osmolyte and metabolite and is consistent with clinical evidence of betaine-mediated improvements to cognitive function (reported in elderly populations) and its anti-convulsant properties. Betaine's therapeutic potential in neurological disorders including epilepsy and neurodegenerative diseases combined with benefits of betaine supplementation on athletic performance support the unique application of betaine as a prophylaxis to concussion. As an example, we identify young athletes (15-24 years old), especially females, for prophylactic betaine supplementation to promote brain health and resilience in a cohort at high risk for concussion and for developing Alzheimer's disease.
Collapse
Affiliation(s)
| | - Thomas A. Knight
- Biology Department, Whitman College, Walla Walla, WA, United States
| |
Collapse
|
30
|
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
|
31
|
Zhao ZA, Yan L, Wen J, Satyanarayanan SK, Yu F, Lu J, Liu YU, Su H. Cellular and molecular mechanisms in vascular repair after traumatic brain injury: a narrative review. BURNS & TRAUMA 2023; 11:tkad033. [PMID: 37675267 PMCID: PMC10478165 DOI: 10.1093/burnst/tkad033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/01/2023] [Accepted: 05/26/2023] [Indexed: 09/08/2023]
Abstract
Traumatic brain injury (TBI) disrupts normal brain function and is associated with high morbidity and fatality rates. TBI is characterized as mild, moderate or severe depending on its severity. The damage may be transient and limited to the dura matter, with only subtle changes in cerebral parenchyma, or life-threatening with obvious focal contusions, hematomas and edema. Blood vessels are often injured in TBI. Even in mild TBI, dysfunctional cerebral vascular repair may result in prolonged symptoms and poor outcomes. Various distinct types of cells participate in vascular repair after TBI. A better understanding of the cellular response and function in vascular repair can facilitate the development of new therapeutic strategies. In this review, we analyzed the mechanism of cerebrovascular impairment and the repercussions following various forms of TBI. We then discussed the role of distinct cell types in the repair of meningeal and parenchyma vasculature following TBI, including endothelial cells, endothelial progenitor cells, pericytes, glial cells (astrocytes and microglia), neurons, myeloid cells (macrophages and monocytes) and meningeal lymphatic endothelial cells. Finally, possible treatment techniques targeting these unique cell types for vascular repair after TBI are discussed.
Collapse
Affiliation(s)
- Zi-Ai Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
- Department of Neurology, General Hospital of Northern Theater Command, 83# Wen-Hua Road, Shenyang 110840, China
| | - Lingli Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jing Wen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Senthil Kumaran Satyanarayanan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Feng Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Jiahong Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Yong U Liu
- Laboratory of Neuroimmunology in Health and Disease Institute, Guangzhou First People’s Hospital School of Medicine, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou 511400, China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| |
Collapse
|
32
|
Karimpoor M, Georgiadis M, Zhao MY, Goubran M, Moein Taghavi H, Mills BD, Tran D, Mouchawar N, Sami S, Wintermark M, Grant G, Camarillo DB, Moseley ME, Zaharchuk G, Zeineh MM. Longitudinal Alterations of Cerebral Blood Flow in High-Contact Sports. Ann Neurol 2023; 94:457-469. [PMID: 37306544 DOI: 10.1002/ana.26718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Repetitive head trauma is common in high-contact sports. Cerebral blood flow (CBF) can measure changes in brain perfusion that could indicate injury. Longitudinal studies with a control group are necessary to account for interindividual and developmental effects. We investigated whether exposure to head impacts causes longitudinal CBF changes. METHODS We prospectively studied 63 American football (high-contact cohort) and 34 volleyball (low-contact controls) male collegiate athletes, tracking CBF using 3D pseudocontinuous arterial spin labeling magnetic resonance imaging for up to 4 years. Regional relative CBF (rCBF, normalized to cerebellar CBF) was computed after co-registering to T1-weighted images. A linear mixed effects model assessed the relationship of rCBF to sport, time, and their interaction. Within football players, we modeled rCBF against position-based head impact risk and baseline Standardized Concussion Assessment Tool score. Additionally, we evaluated early (1-5 days) and delayed (3-6 months) post-concussion rCBF changes (in-study concussion). RESULTS Supratentorial gray matter rCBF declined in football compared with volleyball (sport-time interaction p = 0.012), with a strong effect in the parietal lobe (p = 0.002). Football players with higher position-based impact-risk had lower occipital rCBF over time (interaction p = 0.005), whereas players with lower baseline Standardized Concussion Assessment Tool score (worse performance) had relatively decreased rCBF in the cingulate-insula over time (interaction effect p = 0.007). Both cohorts showed a left-right rCBF asymmetry that decreased over time. Football players with an in-study concussion showed an early increase in occipital lobe rCBF (p = 0.0166). INTERPRETATION These results suggest head impacts may result in an early increase in rCBF, but cumulatively a long-term decrease in rCBF. ANN NEUROL 2023;94:457-469.
Collapse
Affiliation(s)
| | | | - Moss Y Zhao
- Department of Radiology, Stanford University, Stanford, CA
| | - Maged Goubran
- Department of Medical Biophysics, University of Toronto, Toronto, Canada
- Physical Sciences Platform & Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | | | - Brian D Mills
- Department of Radiology, Stanford University, Stanford, CA
| | - Dean Tran
- Department of Radiology, Stanford University, Stanford, CA
| | | | - Sohrab Sami
- Department of Radiology, Stanford University, Stanford, CA
| | - Max Wintermark
- Department of Radiology, Stanford University, Stanford, CA
| | - Gerald Grant
- Department of Neurosurgery, Stanford University, Stanford, CA
| | | | | | - Greg Zaharchuk
- Department of Radiology, Stanford University, Stanford, CA
| | | |
Collapse
|
33
|
Acosta CH, Clemons GA, Citadin CT, Carr WC, Udo MSB, Tesic V, Sanicola HW, Freelin AH, Toms JB, Jordan JD, Guthikonda B, Rodgers KM, Wu CYC, Lee RHC, Lin HW. PRMT7 can prevent neurovascular uncoupling, blood-brain barrier permeability, and mitochondrial dysfunction in repetitive and mild traumatic brain injury. Exp Neurol 2023; 366:114445. [PMID: 37196697 PMCID: PMC10960645 DOI: 10.1016/j.expneurol.2023.114445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/03/2023] [Accepted: 05/12/2023] [Indexed: 05/19/2023]
Abstract
Mild traumatic brain injury (TBI) comprises the largest percentage of TBI-related injuries, with pathophysiological and functional deficits that persist in a subset of TBI patients. In our three-hit paradigm of repetitive and mild traumatic brain injury (rmTBI), we observed neurovascular uncoupling via decreased red blood cell velocity, microvessel diameter, and leukocyte rolling velocity 3 days post-rmTBI via intra-vital two-photon laser scanning microscopy. Furthermore, our data suggest increased blood-brain barrier (BBB) permeability (leakage), with corresponding decrease in junctional protein expression post-rmTBI. Mitochondrial oxygen consumption rates (measured via Seahorse XFe24) were also altered 3 days post-rmTBI, along with disrupted mitochondrial dynamics of fission and fusion. Overall, these pathophysiological findings correlated with decreased protein arginine methyltransferase 7 (PRMT7) protein levels and activity post-rmTBI. Here, we increased PRMT7 levels in vivo to assess the role of the neurovasculature and mitochondria post-rmTBI. In vivo overexpression of PRMT7 using a neuronal specific AAV vector led to restoration of neurovascular coupling, prevented BBB leakage, and promoted mitochondrial respiration, altogether to suggest a protective and functional role of PRMT7 in rmTBI.
Collapse
Affiliation(s)
- Christina H Acosta
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Garrett A Clemons
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Cristiane T Citadin
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - William C Carr
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Mariana Sayuri Berto Udo
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Vesna Tesic
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Henry W Sanicola
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America; Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Anne H Freelin
- Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Jamie B Toms
- Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - J Dedrick Jordan
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Bharat Guthikonda
- Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Krista M Rodgers
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Celeste Yin-Chieh Wu
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Reggie Hui-Chao Lee
- Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America
| | - Hung Wen Lin
- Department of Cellular Biology & Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America; Department of Neurology, Louisiana State University Health Sciences Center, Shreveport, LA, United States of America.
| |
Collapse
|
34
|
Schneider AL, Peltz CB, Li Y, Bahorik A, Gardner RC, Yaffe K. Traumatic Brain Injury and Long-Term Risk of Stroke Among US Military Veterans. Stroke 2023; 54:2059-2068. [PMID: 37334708 PMCID: PMC10527414 DOI: 10.1161/strokeaha.123.042360] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 06/05/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) is associated with significant morbidity, but the association of TBI with long-term stroke risk in diverse populations remains less clear. Our objective was to examine the long-term associations of TBI with stroke and to investigate potential differences by age, sex, race and ethnicity, and time since TBI diagnosis. METHODS Retrospective cohort study of US military veterans aged 18+ years receiving healthcare in the Veterans Health Administration system between October 1, 2002 and September 30, 2019. Veterans with TBI were matched 1:1 to veterans without TBI on age, sex, race and ethnicity, and index date, yielding 306 796 veterans with TBI and 306 796 veterans without TBI included in the study. In primary analyses, Fine-Gray proportional hazards models adjusted for sociodemographics and medical/psychiatric comorbidities were used to estimate the association between TBI and stroke risk, accounting for the competing risk of mortality. RESULTS Participants were a mean age of 50 years, 9% were female, and 25% were of non-White race and ethnicity. Overall, 4.7% of veterans developed a stroke over a median follow-up of 5.2 years. Veterans with TBI had 1.69 times (95% CI, 1.64-1.73) increased risk of any stroke (ischemic or hemorrhagic) compared to veterans without TBI. This increased risk was highest in the first-year post-TBI diagnosis (hazard ratio [HR], 2.16 [95% CI, 2.03-2.29]) but remained elevated for 10+ years. Similar patterns were observed for secondary outcomes, with associations of TBI with hemorrhagic stroke (HR, 3.92 [95% CI, 3.59-4.29]) being stronger than with ischemic stroke (HR, 1.56 [95% CI, 1.52-1.61]). Veterans with both mild (HR, 1.47 [95% CI, 1.43-1.52]) and moderate/severe/penetrating injury (HR, 2.02 [95% CI, 1.96-2.09]) had increased risk of stroke compared to veterans without TBI. Associations of TBI with stroke were stronger among older compared to younger individuals (P interaction-by-age<0.001) and were weaker among Black veterans compared to other race and ethnicities (P interaction-by-race<0.001). CONCLUSIONS Veterans with prior TBI are at increased long-term risk for stroke, suggesting they may be an important population to target for primary stroke prevention measures.
Collapse
Affiliation(s)
- Andrea L.C. Schneider
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine at the University of Pennsylvania
| | | | - Yixia Li
- San Francisco Veterans Affairs Health System
| | | | - Raquel C. Gardner
- San Francisco Veterans Affairs Health System
- Department of Neurology, University of California San Francisco
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Kristine Yaffe
- San Francisco Veterans Affairs Health System
- Department of Neurology, University of California San Francisco
- Department of Epidemiology and Biostatistics, University of California San Francisco
- Department of Psychiatry, University of California San Francisco
| |
Collapse
|
35
|
Yang D, Wang W, Yuan Z, Liang Y. Information-Rich Multi-Functional OCT for Adult Zebrafish Intra- and Extracranial Imaging. Bioengineering (Basel) 2023; 10:856. [PMID: 37508883 PMCID: PMC10375992 DOI: 10.3390/bioengineering10070856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
The zebrafish serves as a valuable animal model for both intra- and extracranial research, particularly in relation to the brain and skull. To effectively investigate the development and regeneration of adult zebrafish, a versatile in vivo imaging technique capable of showing both intra- and extracranial conditions is essential. In this paper, we utilized a high-resolution multi-functional optical coherence tomography (OCT) to obtain rich intra- and extracranial imaging outcomes of adult zebrafish, encompassing pigmentation distribution, tissue-specific information, cranial vascular imaging, and the monitoring of traumatic brain injury (TBI). Notably, it is the first that the channels through the zebrafish cranial suture, which may have a crucial function in maintaining the patency of the cranial sutures, have been observed. Rich imaging results demonstrated that a high-resolution multi-functional OCT system can provide a wealth of novel and interpretable biological information for intra- and extracranial studies of adult zebrafish.
Collapse
Affiliation(s)
- Di Yang
- Tianjin Key Laboratory of Micro-Scale Optical Information Science and Technology, Institute of Modern Optics, Nankai University, Tianjin 300350, China
| | - Weike Wang
- Tianjin Key Laboratory of Micro-Scale Optical Information Science and Technology, Institute of Modern Optics, Nankai University, Tianjin 300350, China
| | - Zhuoqun Yuan
- Tianjin Key Laboratory of Micro-Scale Optical Information Science and Technology, Institute of Modern Optics, Nankai University, Tianjin 300350, China
| | - Yanmei Liang
- Tianjin Key Laboratory of Micro-Scale Optical Information Science and Technology, Institute of Modern Optics, Nankai University, Tianjin 300350, China
| |
Collapse
|
36
|
Kamali A, Dieckhaus L, Peters EC, Preszler CA, Witte RS, Pires PW, Hutchinson EB, Laksari K. Ultrasound, photoacoustic, and magnetic resonance imaging to study hyperacute pathophysiology of traumatic and vascular brain injury. J Neuroimaging 2023; 33:534-546. [PMID: 37183044 PMCID: PMC10525021 DOI: 10.1111/jon.13115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/29/2023] [Accepted: 05/02/2023] [Indexed: 05/16/2023] Open
Abstract
BACKGROUND AND PURPOSE Cerebrovascular dynamics and pathomechanisms that evolve in the minutes and hours following traumatic vascular injury in the brain remain largely unknown. We investigated the pathophysiology evolution in mice within the first 3 hours after closed-head traumatic brain injury (TBI) and subarachnoid hemorrhage (SAH), two significant traumatic vascular injuries. METHODS We took a multimodal imaging approach using photoacoustic imaging, color Doppler ultrasound, and MRI to track injury outcomes using a variety of metrics. RESULTS Brain oxygenation and velocity-weighted volume of blood flow (VVF) values significantly decreased from baseline to 15 minutes after both TBI and SAH. TBI resulted in 19.2% and 41.0% ipsilateral oxygenation and VVF reductions 15 minutes postinjury, while SAH resulted in 43.9% and 85.0% ipsilateral oxygenation and VVF reduction (p < .001). We found partial recovery of oxygenation from 15 minutes to 3 hours after injury for TBI but not SAH. Hemorrhage, edema, reduced perfusion, and altered diffusivity were evident from MRI scans acquired 90-150 minutes after injury in both injury models, although the spatial distribution was mostly focal for TBI and diffuse for SAH. CONCLUSIONS The results reveal that the cerebral oxygenation deficits immediately following injuries are reversible for TBI and irreversible for SAH. Our findings can inform future studies on mitigating these early responses to improve long-term recovery.
Collapse
Affiliation(s)
- Ali Kamali
- Department of Biomedical Engineering, University of Arizona College of Engineering, Tucson, AZ
| | - Laurel Dieckhaus
- Department of Biomedical Engineering, University of Arizona College of Engineering, Tucson, AZ
| | - Emily C. Peters
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ
| | - Collin A. Preszler
- Department of Biomedical Engineering, University of Arizona College of Engineering, Tucson, AZ
| | - Russel S. Witte
- Department of Biomedical Engineering, University of Arizona College of Engineering, Tucson, AZ
- Department of Medical Imaging, University of Arizona College of Medicine, Tucson, AZ
- College of Optical Sciences, University of Arizona, Tucson, AZ
| | - Paulo W. Pires
- Department of Physiology, University of Arizona College of Medicine, Tucson, AZ
| | - Elizabeth B. Hutchinson
- Department of Biomedical Engineering, University of Arizona College of Engineering, Tucson, AZ
| | - Kaveh Laksari
- Department of Biomedical Engineering, University of Arizona College of Engineering, Tucson, AZ
- Department of Aerospace and Mechanical Engineering, University of Arizona College of Engineering, Tucson, AZ
| |
Collapse
|
37
|
Choi BR, Johnson KR, Maric D, McGavern DB. Monocyte-derived IL-6 programs microglia to rebuild damaged brain vasculature. Nat Immunol 2023; 24:1110-1123. [PMID: 37248420 PMCID: PMC11531796 DOI: 10.1038/s41590-023-01521-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/25/2023] [Indexed: 05/31/2023]
Abstract
Cerebrovascular injury (CVI) is a common pathology caused by infections, injury, stroke, neurodegeneration and autoimmune disease. Rapid resolution of a CVI requires a coordinated innate immune response. In the present study, we sought mechanistic insights into how central nervous system-infiltrating monocytes program resident microglia to mediate angiogenesis and cerebrovascular repair after an intracerebral hemorrhage. In the penumbrae of human stroke brain lesions, we identified a subpopulation of microglia that express vascular endothelial growth factor A. These cells, termed 'repair-associated microglia' (RAMs), were also observed in a rodent model of CVI and coexpressed interleukin (IL)-6Ra. Cerebrovascular repair did not occur in IL-6 knockouts or in mice lacking microglial IL-6Ra expression and single-cell transcriptomic analyses revealed faulty RAM programming in the absence of IL-6 signaling. Infiltrating CCR2+ monocytes were the primary source of IL-6 after a CVI and were required to endow microglia with proliferative and proangiogenic properties. Faulty RAM programming in the absence of IL-6 or inflammatory monocytes resulted in poor cerebrovascular repair, neuronal destruction and sustained neurological deficits that were all restored via exogenous IL-6 administration. These data provide a molecular and cellular basis for how monocytes instruct microglia to repair damaged brain vasculature and promote functional recovery after injury.
Collapse
Affiliation(s)
- Bo-Ran Choi
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kory R Johnson
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dragan Maric
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
38
|
Liu P, Hu B, Kartchner L, Joshi P, Xu C, Jiang D. Dependence of resting-state-based cerebrovascular reactivity (CVR) mapping on spatial resolution. FRONTIERS IN NEUROIMAGING 2023; 2:1205459. [PMID: 37554643 PMCID: PMC10406303 DOI: 10.3389/fnimg.2023.1205459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/12/2023] [Indexed: 08/10/2023]
Abstract
Cerebrovascular reactivity (CVR) is typically assessed with a carbon dioxide (CO2) stimulus combined with BOLD fMRI. Recently, resting-state (RS) BOLD fMRI has been shown capable of generating CVR maps, providing a potential for broader CVR applications in neuroimaging studies. However, prior RS-CVR studies have primarily been performed at a spatial resolution of 3-4 mm voxel sizes. It remains unknown whether RS-CVR can also be obtained at high-resolution without major degradation in image quality. In this study, we investigated RS-CVR mapping based on resting-state BOLD MRI across a range of spatial resolutions in a group of healthy subjects, in an effort to examine the feasibility of RS-CVR measurement at high resolution. Comparing the results of RS-CVR with the maps obtained by the conventional CO2-inhalation method, our results suggested that good CVR map quality can be obtained at a voxel size as small as 2 mm isotropic. Our results also showed that, RS-CVR maps revealed resolution-dependent sensitivity. However, even at a high resolution of 2 mm isotropic voxel size, the voxel-wise sensitivity is still greater than that of typical task-evoked fMRI. Scan duration affected the sensitivity of RS-CVR mapping, but had no significant effect on its accuracy. These findings suggest that RS-CVR mapping can be applied at a similar resolution as state-of-the-art fMRI studies, which will broaden the use of CVR mapping in basic science and clinical applications including retrospective analysis of previously collected fMRI data.
Collapse
Affiliation(s)
- Peiying Liu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Beini Hu
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Lincoln Kartchner
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Parimal Joshi
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Cuimei Xu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dengrong Jiang
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
39
|
Deshetty UM, Periyasamy P. Potential Biomarkers in Experimental Animal Models for Traumatic Brain Injury. J Clin Med 2023; 12:3923. [PMID: 37373618 DOI: 10.3390/jcm12123923] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Traumatic brain injury (TBI) is a complex and multifaceted disorder that has become a significant public health concern worldwide due to its contribution to mortality and morbidity. This condition encompasses a spectrum of injuries, including axonal damage, contusions, edema, and hemorrhage. Unfortunately, specific effective therapeutic interventions to improve patient outcomes following TBI are currently lacking. Various experimental animal models have been developed to mimic TBI and evaluate potential therapeutic agents to address this issue. These models are designed to recapitulate different biomarkers and mechanisms involved in TBI. However, due to the heterogeneous nature of clinical TBI, no single experimental animal model can effectively mimic all aspects of human TBI. Accurate emulation of clinical TBI mechanisms is also tricky due to ethical considerations. Therefore, the continued study of TBI mechanisms and biomarkers, of the duration and severity of brain injury, treatment strategies, and animal model optimization is necessary. This review focuses on the pathophysiology of TBI, available experimental TBI animal models, and the range of biomarkers and detection methods for TBI. Overall, this review highlights the need for further research to improve patient outcomes and reduce the global burden of TBI.
Collapse
Affiliation(s)
- Uma Maheswari Deshetty
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
40
|
Penn C, Katnik C, Cuevas J, Mohapatra SS, Mohapatra S. Multispectral optoacoustic tomography (MSOT): Monitoring neurovascular changes in a mouse repetitive traumatic brain injury model. J Neurosci Methods 2023; 393:109876. [PMID: 37150303 PMCID: PMC10388337 DOI: 10.1016/j.jneumeth.2023.109876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Evidence suggests that mild TBI injuries, which comprise > 75% of all TBIs, can cause chronic post-concussive symptoms, especially when experienced repetitively (rTBI). rTBI is a major cause of cognitive deficit in athletes and military personnel and is associated with neurovascular changes. Current methods to monitor neurovascular changes in detail are prohibitively expensive and invasive for patients with mild injuries. NEW METHOD We evaluated the potential of multispectral optoacoustic tomography (MSOT) to monitor neurovascular changes and assess therapeutic strategies in a mouse model of rTBI. Mice were subjected to rTBI or sham via controlled cortical impact and administered pioglitazone (PG) or vehicle. Oxygenated and deoxygenated hemoglobin were monitored using MSOT. Indocyanine green clearance was imaged via MSOT to evaluate blood-brain-barrier (BBB) integrity. RESULTS Mice subjected to rTBI show a transient increase in oxygenated/total hemoglobin ratio which can be mitigated by PG administration. rTBI mice also show BBB disruption shortly after injury and reduction of oxygenated/total hemoglobin in the chronic stage, neither of which were affected by PG intervention. COMPARISON WITH EXISTING METHODS MSOT imaging has the potential as a noninvasive in vivo imaging method to monitor neurovascular changes and assess therapeutics in mouse models of rTBI. In comparison to standard methods of tracking inflammation and BBB disruption, MSOT can be used multiple times throughout the course of injury without the need for surgery. Thus, MSOT is especially useful in research of rTBI models for screening therapeutics, and with further technological improvements may be extended for use in rTBI patients.
Collapse
Affiliation(s)
- Courtney Penn
- James A. Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; Department of Molecular Medicine, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd., Tampa, FL 33612, USA
| | - Chris Katnik
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd., Tampa, FL 33612, USA
| | - Javier Cuevas
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd., Tampa, FL 33612, USA
| | - Shyam S Mohapatra
- James A. Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; Department of Internal Medicine, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd., Tampa, FL 33612, USA
| | - Subhra Mohapatra
- James A. Haley Veterans Hospital, 13000 Bruce B Downs Blvd, Tampa, FL 33612, USA; Department of Molecular Medicine, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd., Tampa, FL 33612, USA.
| |
Collapse
|
41
|
Ware JB, Sandsmark DK. Imaging Approach to Concussion. Neuroimaging Clin N Am 2023; 33:261-269. [PMID: 36965944 DOI: 10.1016/j.nic.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
The acute and long-term neurobiological sequelae of concussion (mild traumatic brain injury [mTBI]) and sub-concussive head trauma have become increasingly apparent in recent decades in part due to neuroimaging research. Although imaging has an established role in the clinical management of mTBI for the identification of intracranial lesions warranting urgent interventions, MR imaging is increasingly employed for the detection of post-traumatic sequelae which carry important prognostic significance. As neuroimaging research continues to elucidate the pathophysiology of TBI underlying prolonged recovery and the development of persistent post-concussive symptoms, there is a strong motivation to translate these techniques into clinical use for improved diagnosis and therapeutic monitoring.
Collapse
Affiliation(s)
- Jeffrey B Ware
- Department of Radiology, Neuroradiology Division, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| | - Danielle K Sandsmark
- Department of Neurology, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Anderl WJ, Pearson N, Converse MI, Yu SM, Monson KL. Strain-induced collagen denaturation is rate dependent in failure of cerebral arteries. Acta Biomater 2023; 164:282-292. [PMID: 37116635 DOI: 10.1016/j.actbio.2023.04.032] [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: 10/29/2022] [Revised: 04/06/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023]
Abstract
While soft tissues are commonly damaged by mechanical loading, the manifestation of this damage at the microstructural level is not fully understood. Specifically, while rate-induced stiffening has been previously observed in cerebral arteries, associated changes in microstructural damage patterns following high-rate loading are largely undefined. In this study, we stretched porcine middle cerebral arteries to failure at 0.01 and >150 s-1, both axially and circumferentially, followed by probing for denatured tropocollagen using collagen hybridizing peptide (CHP). We found that collagen fibrils aligned with the loading direction experienced less denaturation following failure tests at high than low rates. Others have demonstrated similar rate dependence in tropocollagen denaturation during soft tissue failure, but this is the first study to quantify this behavior using CHP and to report it for cerebral arteries. These findings may have significant implications for traumatic brain injury and intracranial balloon angioplasty. We additionally observed possible tropocollagen denaturation in vessel layers primarily composed of fibrils transversely aligned to the loading axis. To our knowledge, this is the first observation of collagen denaturation due to transverse loading, but further research is needed to confirm this finding. STATEMENT OF SIGNIFICANCE: Previous work shows that collagen hybridizing peptide (CHP) can be used to identify collagen molecule unfolding and denaturation in mechanically overloaded soft tissues, including the cerebral arteries. But experiments have not explored collagen damage at rates relevant to traumatic brain injury. In this work, we quantified collagen damage in cerebral arteries stretched to failure at both high and low rates. We found that the collagen molecule is less damaged at high than at low rates, suggesting that damage mechanisms of either the collagen molecule or other elements of the collagen superstructure are rate dependent. This work implies that arteries failed at high rates, such as in traumatic brain injury, will have different molecular-level damage patterns than arteries failed at low rates. Consequently, improved understanding of damage characteristics may be expanded in the future to better inform clinically relevant cases of collagen damage such as angioplasty and injury healing.
Collapse
Affiliation(s)
| | - Noah Pearson
- DepSSSartment of Mechanical Engineering, University of Utah
| | | | - S Michael Yu
- Department of Biomedical Engineering, University of Utah; Department of Molecular Pharmaceutics, University of Utah
| | - Kenneth L Monson
- DepSSSartment of Mechanical Engineering, University of Utah; Department of Biomedical Engineering, University of Utah.
| |
Collapse
|
43
|
Hepschke JL, Laws E, Bin Saliman NH, Juncu S, Courtie E, Belli A, Blanch RJ. Modifications in Macular Perfusion and Neuronal Loss After Acute Traumatic Brain Injury. Invest Ophthalmol Vis Sci 2023; 64:35. [PMID: 37115535 PMCID: PMC10150830 DOI: 10.1167/iovs.64.4.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 03/15/2023] [Indexed: 04/29/2023] Open
Abstract
Purpose Traumatic brain injury (TBI) causes structural damage and functional impairment in the visual system, often with retinal ganglion cell (RGC) degeneration occurring without visual symptoms. RGC degeneration is associated with reduced retinal blood-flow, however, it is not known whether reductions in perfusion precede or are secondary to neurodegeneration. Methods We conducted a prospective observational single-center case series. Patients were included if they were admitted to the hospital after acute TBI and underwent ophthalmic clinical examination, including optical coherence tomography (OCT) and OCT angiography (OCTA) acutely and at follow-up. Ganglion cell layer thickness (GCL) thickness, vascular density in the superficial vascular plexus (SVP), and intermediate capillary plexus (ICP) were quantified. Results Twenty-one patients aged 20 to 65 years (mean = 38 years) including 16 men and 5 women were examined less than 14 days after moderate to severe TBI, and again after 2 to 6 months. Macular structure and perfusion were normal at baseline in all patients. Visual function was abnormal at baseline in three patients and subsequent neurodegeneration and loss of perfusion corresponded to baseline visual function abnormalities. Nine patients (43%) had reduced macular GCL thickness at follow up. Perfusion in the SVP strongly associated with local GCL thickness. The strongest association of the SVP metrics was the sum of vessel density (P < 0.0001). Conclusions In cases of reduced visual function after TBI, macular perfusion remained normal until reductions in GCL thickness occurred, indicating that perfusion changes were secondary to local GCL loss.
Collapse
Affiliation(s)
- Jenny L Hepschke
- Ophthalmology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Elinor Laws
- Ophthalmology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Noor H Bin Saliman
- Centre for Optometry Studies, Faculty of Health Sciences, Universiti Teknologi MARA Cawangan Selangor, Bandar Puncak Alam Selangor, Malaysia
| | - Stefana Juncu
- Department of Psychology, University of Portsmouth, Portsmouth, United Kingdom
| | - Ella Courtie
- Ophthalmology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Antonio Belli
- Neurosurgery Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Richard J Blanch
- Ophthalmology Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- NIHR Surgical Reconstruction and Microbiology Centre, University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
- Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, United Kingdom
| |
Collapse
|
44
|
Cowdrick KR, Urner T, Sathialingam E, Fang Z, Quadri A, Turrentine K, Yup Lee S, Buckley EM. Agreement in cerebrovascular reactivity assessed with diffuse correlation spectroscopy across experimental paradigms improves with short separation regression. NEUROPHOTONICS 2023; 10:025002. [PMID: 37034012 PMCID: PMC10079775 DOI: 10.1117/1.nph.10.2.025002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/07/2023] [Indexed: 06/19/2023]
Abstract
Significance Cerebrovascular reactivity (CVR), i.e., the ability of cerebral vasculature to dilate or constrict in response to vasoactive stimuli, is a biomarker of vascular health. Exogenous administration of inhaled carbon dioxide, i.e., hypercapnia (HC), remains the "gold-standard" intervention to assess CVR. More tolerable paradigms that enable CVR quantification when HC is difficult/contraindicated have been proposed. However, because these paradigms feature mechanistic differences in action, an assessment of agreement of these more tolerable paradigms to HC is needed. Aim We aim to determine the agreement of CVR assessed during HC, breath-hold (BH), and resting state (RS) paradigms. Approach Healthy adults were subject to HC, BH, and RS paradigms. End tidal carbon dioxide (EtCO2) and cerebral blood flow (CBF, assessed with diffuse correlation spectroscopy) were monitored continuously. CVR (%/mmHg) was quantified via linear regression of CBF versus EtCO2 or via a general linear model (GLM) that was used to minimize the influence of systemic and extracerebral signal contributions. Results Strong agreement ( CCC ≥ 0.69 ; R ≥ 0.76 ) among CVR paradigms was demonstrated when utilizing a GLM to regress out systemic/extracerebral signal contributions. Linear regression alone showed poor agreement across paradigms ( CCC ≤ 0.35 ; R ≤ 0.45 ). Conclusions More tolerable experimental paradigms coupled with regression of systemic/extracerebral signal contributions may offer a viable alternative to HC for assessing CVR.
Collapse
Affiliation(s)
- Kyle R. Cowdrick
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Tara Urner
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Eashani Sathialingam
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Zhou Fang
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
| | - Ayesha Quadri
- Children’s Healthcare of Atlanta and Emory University School of Medicine, Department of Pediatrics, Atlanta, Georgia, United States
- Morehouse School of Medicine, Atlanta, Georgia, United States
| | - Katherine Turrentine
- Children’s Healthcare of Atlanta and Emory University School of Medicine, Department of Pediatrics, Atlanta, Georgia, United States
| | - Seung Yup Lee
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Kennesaw State University, Department of Electrical and Computer Engineering, Marietta, Georgia, United States
| | - Erin M. Buckley
- Georgia Institute of Technology and Emory University, Wallace H. Coulter Department of Biomedical Engineering, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta and Emory University School of Medicine, Department of Pediatrics, Atlanta, Georgia, United States
- Children’s Healthcare of Atlanta, Children’s Research Scholar, Atlanta, Georgia, United States
| |
Collapse
|
45
|
Johnstone DM, Mitrofanis J, Stone J. The brain's weakness in the face of trauma: How head trauma causes the destruction of the brain. Front Neurosci 2023; 17:1141568. [PMID: 36950132 PMCID: PMC10026135 DOI: 10.3389/fnins.2023.1141568] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 02/15/2023] [Indexed: 03/06/2023] Open
Abstract
Of all our organs, the brain is perhaps the best protected from trauma. The skull has evolved to enclose it and, within the skull, the brain floats in a protective bath of cerebrospinal fluid. It is becoming evident, however, that head trauma experienced in young adult life can cause a dementia that appears decades later. The level of trauma that induces such destruction is still being assessed but includes levels well below that which cracks the skull or causes unconsciousness or concussion. Clinically this damage appears as dementia, in people who played body-contact sports in their youth or have survived accidents or the blasts of combat; and appears also, we argue, in old age, without a history of head trauma. The dementias have been given different names, including dementia pugilistica (affecting boxers), chronic traumatic encephalopathy (following certain sports, particularly football), traumatic brain injury (following accidents, combat) and Alzheimer's (following decades of life). They share common features of clinical presentation and neuropathology, and this conceptual analysis seeks to identify features common to these forms of brain injury and to identify where in the brain the damage common to them occurs; and how it occurs, despite the protection provided by the skull and cerebrospinal fluid. The analysis suggests that the brain's weak point in the face of trauma is its capillary bed, which is torn by the shock of trauma. This identification in turn allows discussion of ways of delaying, avoiding and even treating these trauma-induced degenerations.
Collapse
Affiliation(s)
- Daniel M. Johnstone
- School of Biomedical Sciences and Pharmacy, University of Newcastle and School of Medical Sciences, The University of Sydney, Darlington, NSW, Australia
| | - John Mitrofanis
- Fonds de Dotation Clinatec, Université Grenoble Alpes, France and Institute of Ophthalmology, University College London, London, United Kingdom
| | - Jonathan Stone
- Honorary Associate, Centenary Institute and University of Sydney, Darlington, NSW, Australia
| |
Collapse
|
46
|
Wang Y, Bartels HM, Nelson LD. A Systematic Review of ASL Perfusion MRI in Mild TBI. Neuropsychol Rev 2023; 33:160-191. [PMID: 32808244 PMCID: PMC7889778 DOI: 10.1007/s11065-020-09451-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 08/06/2020] [Indexed: 01/06/2023]
Abstract
Mild traumatic brain injury (mTBI) is a major public health concern. Cerebrovascular alterations play a significant role in the evolution of injury sequelae and in the process of post-traumatic brain repair. Arterial spin labeling (ASL) is an advanced perfusion magnetic resonance imaging technique that permits noninvasive quantification of cerebral blood flow (CBF). This is the first systematic review of ASL research findings in patients with mTBI. Our approach followed the American Academy of Neurology (AAN) and PRISMA guidelines. We searched Ovid/MEDLINE, Web of Science, Scopus, and the Cochrane Index for relevant articles published as of February 20, 2020. Full-text results were combined into Rayyan software for further evaluation. Data extraction, including risk of bias ratings, was performed using American Academy of Neurology's four-tiered classification scheme. Twenty-three articles met inclusion criteria comprising data on up to 566 mTBI patients and 654 control subjects. Of the 23 studies, 18 reported some type of regional CBF abnormality in mTBI patients at rest or during a cognitive task, with more findings of decreased than increased CBF. The evidence supports the conclusion that mTBI likely causes ASL-derived CBF anomalies. However, synthesis of findings was challenging due to substantial methodological variations across studies and few studies with low risk of bias. Thus, larger-scale prospective cohort studies are needed to more definitively chart the course of CBF changes in humans after mTBI and to understand how individual difference factors contribute to post-injury CBF changes.
Collapse
Affiliation(s)
- Yang Wang
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Hannah M Bartels
- Department of Neurosurgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| | - Lindsay D Nelson
- Department of Neurosurgery, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI, 53226, USA
| |
Collapse
|
47
|
Gaggi NL, Ware JB, Dolui S, Brennan D, Torrellas J, Wang Z, Whyte J, Diaz-Arrastia R, Kim JJ. Temporal dynamics of cerebral blood flow during the first year after moderate-severe traumatic brain injury: A longitudinal perfusion MRI study. Neuroimage Clin 2023; 37:103344. [PMID: 36804686 PMCID: PMC9969322 DOI: 10.1016/j.nicl.2023.103344] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/13/2023]
Abstract
Traumatic brain injury (TBI) is associated with alterations in cerebral blood flow (CBF), which may underlie functional disability and precipitate TBI-induced neurodegeneration. Although it is known that chronic moderate-severe TBI (msTBI) causes decreases in CBF, the temporal dynamics during the early chronic phase of TBI remain unknown. Using arterial spin labeled (ASL) perfusion magnetic resonance imaging (MRI), we examined longitudinal CBF changes in 29 patients with msTBI at 3, 6, and 12 months post-injury in comparison to 35 demographically-matched healthy controls (HC). We investigated the difference between the two groups and the within-subject time effect in the TBI patients using whole-brain voxel-wise analysis. Mean CBF in gray matter (GM) was lower in the TBI group compared to HC at 6 and 12 months post-injury. Within the TBI group, we identified widespread regional decreases in CBF from 3 to 6 months post-injury. In contrast, there were no regions with decreasing CBF from 6 to 12 months post-injury, indicating stabilization of hypoperfusion. There was instead a small area of increase in CBF observed in the right precuneus. These CBF changes were not accompanied by cortical atrophy. The change in CBF was correlated with change in executive function from 3 to 6 months post-injury in TBI patients, suggesting functional relevance of CBF measures. Understanding the time course of TBI-induced hypoperfusion and its relationship with cognitive improvement could provide an optimal treatment window to benefit long-term outcome.
Collapse
Affiliation(s)
- Naomi L Gaggi
- City University of New York (CUNY) School of Medicine, Townsend Harris Hall, 160 Convent Avenue, Convent Avenue, New York, NY 10031, United States; City University of New York (CUNY) Graduate Center, 365 5(th) Avenue, New York, NY 10016, United States.
| | - Jeffrey B Ware
- University of Pennsylvania, Perelman School of Medicine, 3400 Civic Center Boulevard, Philadelphia, PA 19104, United States.
| | - Sudipto Dolui
- University of Pennsylvania, Perelman School of Medicine, 3400 Civic Center Boulevard, Philadelphia, PA 19104, United States.
| | - Daniel Brennan
- City University of New York (CUNY) School of Medicine, Townsend Harris Hall, 160 Convent Avenue, Convent Avenue, New York, NY 10031, United States; City University of New York (CUNY) Graduate Center, 365 5(th) Avenue, New York, NY 10016, United States.
| | - Julia Torrellas
- City University of New York (CUNY) School of Medicine, Townsend Harris Hall, 160 Convent Avenue, Convent Avenue, New York, NY 10031, United States.
| | - Ze Wang
- University of Maryland School of Medicine, 655 W Baltimore St. S, Baltimore, MD 21201, United States.
| | - John Whyte
- Moss Rehabilitation Research Institute, 50 Township Line Road, Elkins Park, PA 19027, United States.
| | - Ramon Diaz-Arrastia
- University of Pennsylvania, Perelman School of Medicine, 3400 Civic Center Boulevard, Philadelphia, PA 19104, United States.
| | - Junghoon J Kim
- City University of New York (CUNY) School of Medicine, Townsend Harris Hall, 160 Convent Avenue, Convent Avenue, New York, NY 10031, United States; City University of New York (CUNY) Graduate Center, 365 5(th) Avenue, New York, NY 10016, United States.
| |
Collapse
|
48
|
Cerebrovascular injuries in traumatic brain injury. Clin Neurol Neurosurg 2022; 223:107479. [DOI: 10.1016/j.clineuro.2022.107479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/22/2022] [Accepted: 10/13/2022] [Indexed: 11/19/2022]
|
49
|
Irimia A, Ngo V, Chaudhari NN, Zhang F, Joshi SH, Penkova AN, O'Donnell LJ, Sheikh-Bahaei N, Zheng X, Chui HC. White matter degradation near cerebral microbleeds is associated with cognitive change after mild traumatic brain injury. Neurobiol Aging 2022; 120:68-80. [PMID: 36116396 PMCID: PMC9759713 DOI: 10.1016/j.neurobiolaging.2022.08.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 08/17/2022] [Accepted: 08/20/2022] [Indexed: 11/28/2022]
Abstract
To explore how cerebral microbleeds (CMBs) accompanying mild traumatic brain injury (mTBI) reflect white matter (WM) degradation and cognitive decline, magnetic resonance images were acquired from 62 mTBI adults (imaged ∼7 days and ∼6 months post-injury) and 203 matched healthy controls. On average, mTBI participants had a count of 2.7 ± 2.6 traumatic CMBs in WM, located 6.1 ± 4.4 mm from cortex. At ∼6-month follow-up, 97% of CMBs were associated with significant reductions (34% ± 11%, q < 0.05) in the fractional anisotropy of WM streamlines within ∼1 cm of CMB locations. Male sex and older age were significant risk factors for larger reductions (q < 0.05). For CMBs in the corpus callosum, cingulum bundle, inferior and middle longitudinal fasciculi, fractional anisotropy changes were significantly and positively associated with changes in cognitive functions mediated by these structures (q < 0.05). Our findings distinguish traumatic from non-traumatic CMBs by virtue of surrounding WM alterations and challenge the assumption that traumatic CMBs are neurocognitively silent. Thus, mTBI with CMB findings can be described as a clinical endophenotype warranting longitudinal cognitive assessment.
Collapse
Affiliation(s)
- Andrei Irimia
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Denney Research Center, Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA.
| | - Van Ngo
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Nikhil N Chaudhari
- Ethel Percy Andrus Gerontology Center, Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Fan Zhang
- Laboratory of Mathematics in Imaging, Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shantanu H Joshi
- Ahmanson Lovelace Brain Mapping Center, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Anita N Penkova
- Department of Aerospace and Mechanical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Lauren J O'Donnell
- Laboratory of Mathematics in Imaging, Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nasim Sheikh-Bahaei
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Xiaoyu Zheng
- Department of Materials Science & Engineering, University of California, Berkeley, CA, USA
| | - Helena C Chui
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
50
|
Liu P, Baker Z, Li Y, Li Y, Xu J, Park DC, Welch BG, Pinho M, Pillai JJ, Hillis AE, Mori S, Lu H. CVR-MRICloud: An online processing tool for CO2-inhalation and resting-state cerebrovascular reactivity (CVR) MRI data. PLoS One 2022; 17:e0274220. [PMID: 36170233 PMCID: PMC9518872 DOI: 10.1371/journal.pone.0274220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
Cerebrovascular Reactivity (CVR) provides an assessment of the brain's vascular reserve and has been postulated to be a sensitive marker in cerebrovascular diseases. MRI-based CVR measurement typically employs alterations in arterial carbon dioxide (CO2) level while continuously acquiring Blood-Oxygenation-Level-Dependent (BOLD) images. CO2-inhalation and resting-state methods are two commonly used approaches for CVR MRI. However, processing of CVR MRI data often requires special expertise and may become an obstacle in broad utilization of this promising technique. The aim of this work was to develop CVR-MRICloud, a cloud-based CVR processing pipeline, to enable automated processing of CVR MRI data. The CVR-MRICloud consists of several major steps including extraction of end-tidal CO2 (EtCO2) curve from raw CO2 recording, alignment of EtCO2 curve with BOLD time course, computation of CVR value on a whole-brain, regional, and voxel-wise basis. The pipeline also includes standard BOLD image processing steps such as motion correction, registration between functional and anatomic images, and transformation of the CVR images to canonical space. This paper describes these algorithms and demonstrates the performance of the CVR-MRICloud in lifespan healthy subjects and patients with clinical conditions such as stroke, brain tumor, and Moyamoya disease. CVR-MRICloud has potential to be used as a data processing tool for a variety of basic science and clinical applications.
Collapse
Affiliation(s)
- Peiying Liu
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Zachary Baker
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yue Li
- AnatomyWorks, LLC, Baltimore, Maryland, United States of America
| | - Yang Li
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Jiadi Xu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States of America
| | - Denise C. Park
- Center for Vital Longevity, School of Behavioral and Brain Sciences, University of Texas at Dallas, Dallas, Texas, United States of America
| | - Babu G. Welch
- Department of Neurological Surgery, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Marco Pinho
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Jay J. Pillai
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Argye E. Hillis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Susumu Mori
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States of America
| | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- F.M. Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, United States of America
| |
Collapse
|