1
|
Mohammadi S, Dolatshahi M, Rahmani F, Raji CA. Altered Clearance in Alzheimer's Disease and Cerebral Amyloid Angiopathy. Neuroimaging Clin N Am 2025; 35:277-286. [PMID: 40210383 DOI: 10.1016/j.nic.2024.12.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
With our focus on Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA), the authors herein provided a comprehensive overview of impaired amyloid beta and tau clearance pathways observed through advanced neuroimaging techniques such as dynamic contrast-enhanced MR imaging, arterial spin labeling, phase contrast MR imaging, PET, and functional MR imaging. The findings suggest the role of impaired degradation clearance, blood-brain barrier clearance, perivascular clearance, glymphatic system clearance, and cerebrospinal fluid dynamics in AD and CAA pathogenesis.
Collapse
Affiliation(s)
- Soheil Mohammadi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mahsa Dolatshahi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Farzaneh Rahmani
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Cyrus A Raji
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, Saint Louis, MO, USA; Department of Neurology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
2
|
Cogswell PM, Burkett BJ, Johnson DR, Pillai JJ. Altered Clearance and Amyloid-Related Imaging Abnormalities. Neuroimaging Clin N Am 2025; 35:267-275. [PMID: 40210382 DOI: 10.1016/j.nic.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Abstract
Amyloid-related imaging abnormalities (ARIA) are parenchymal edema/effusion and hemorrhage that occur as side effects of immunotherapies targeting beta-amyloid for patients with Alzheimer's disease (AD). The mechanism of ARIA remains unclear but is thought to be due to loss of vascular integrity secondary to overwhelming perivascular clearance pathways, worsening of cerebral amyloid angiopathy (CAA), removal of amyloid from the vessel wall, and initiation of a local immune response. Further imaging and neuropathological studies may help further inform the mechanism of ARIA and its relationship to underlying AD and CAA pathology.
Collapse
Affiliation(s)
| | | | | | - Jay J Pillai
- Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Yang H, Guo Y, Lu J, Hassan H, Cao A, Yang Y, Yassin MM, Zaman A, Zeng X, Miao X, Chen Z, Huang G, Han T, Qiu H, Luo Y, Kang Y. Optimizing acute ischemic stroke outcome prediction by integrating radiomics features of DSC-PWI and perfusion parameter maps. Front Neurol 2025; 16:1528812. [PMID: 40191591 PMCID: PMC11968395 DOI: 10.3389/fneur.2025.1528812] [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: 11/15/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Accurate prediction of the prognostic outcomes for patients with ischemic stroke can contribute to personalized treatment decisions and improve life-saving outcomes. This study focuses on the performance of critical moments DSC-PWI in the prognostic prediction of acute ischemic stroke (AIS). It aims to integrate this with perfusion parameters to enhance prediction accuracy. Methods Firstly, The radiomics technique employed to extract DSC-PWI features of critical moments and perfusion parameter features. Following this, a T-test and Lasso algorithm was used to select features associated with the prognosis. Subsequently, machine learning techniques were applied to predict the predictive outcomes of AIS patients. Results The experimental results showed that DSC-PWI sequences at three critical time points-the first moment after contrast injection, the moment of minimum mean time intensity, and the last moment, collectively referred to as 3PWI, had better prognostic prediction than a single perfusion parameter, achieving an optimal model AUC of 0.863. The performance improved by 23.9, 19.6, 6, and 24% compared with CBV, CBF, MTT, and Tmax parameters. The best prognostic prediction for AIS was obtained by integrating the radiomic features from both 3PWI and perfusion parameters, resulting in the highest AUC of 0.915. Discussion Integrating the radiomics features of DSC-PWI sequences of three critical scan time points with those from perfusion parameters can further improve the accuracy of prognostic prediction for AIS patients. This approach may provide new insights into the prognostic evaluation of AIS and provide clinicians with valuable support in making treatment decisions.
Collapse
Affiliation(s)
- Huihui Yang
- Country School of Applied Technology, Shenzhen University, Shenzhen, China
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| | - Yingwei Guo
- School of Electrical and Information Engineering, Northeast Petroleum University, Daqing, China
| | - Jiaxi Lu
- Country School of Applied Technology, Shenzhen University, Shenzhen, China
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| | - Haseeb Hassan
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| | - Anbo Cao
- Country School of Applied Technology, Shenzhen University, Shenzhen, China
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| | - Yingjian Yang
- Department of Radiological Research and Development, Shenzhen Lanmage Medical Technology Co., Ltd, Shenzhen, China
| | - Mazen M. Yassin
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Asim Zaman
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Xueqiang Zeng
- Country School of Applied Technology, Shenzhen University, Shenzhen, China
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| | - Xiaoqiang Miao
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Ziran Chen
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Guangtao Huang
- Country School of Applied Technology, Shenzhen University, Shenzhen, China
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| | - Taiyu Han
- Country School of Applied Technology, Shenzhen University, Shenzhen, China
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
| | - Huiling Qiu
- College of Pharmacy, Shenzhen Technology University, Shenzhen, China
| | - Yu Luo
- Department of Radiology, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yan Kang
- Country School of Applied Technology, Shenzhen University, Shenzhen, China
- College of Health Science and Environmental Engineering, Shenzhen Technology University, Shenzhen, China
- School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
- Engineering Research Centre of Medical Imaging and Intelligent Analysis, Ministry of Education, Shenyang, China
| |
Collapse
|
4
|
Wen C, Gan JH, Liu S, Lu H, Wang LC, Wu H, Shi ZH, Ji Y. Enlarged perivascular spaces correlate with blood-brain barrier leakage and cognitive impairment in Alzheimer's disease. J Alzheimers Dis 2025; 104:382-392. [PMID: 39924914 DOI: 10.1177/13872877251317220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
BackgroundThe clinical significance of enlarged perivascular spaces (EPVS) in Alzheimer' s disease (AD) was ambiguous.ObjectiveTo investigate whether EPVS contribute to blood-brain barrier (BBB) leakage and cognition in AD.MethodsThe study included a total of 64 participants (26 healthy controls and 38 patients with AD). The evaluation of EPVS and BBB permeability was performed in specific anatomical locations: the centrum semiovale (CSO), basal ganglia, and hippocampus. The EPVS ratings were performed according to Potter's instructions. BBB permeability was evaluated using dynamic contrast-enhanced-MRI. The relationship between EPVS and global cognition (Mini-Mental State Examination and Montreal Cognitive Assessment), cognitive subdomains, and BBB permeability were examined in both groups. Finally, the relationship between CSO BBB permeability and cognition in AD patients was investigated.ResultsHigh-grade CSO EPVS was found associated with AD (OR: 3.40, 95% CI: 1.11-11.90, p = 0.04). In the AD group, a significant correlation was observed between high-grade CSO EPVS and lower MMSE score (r = -0.36, p = 0.03) and verbal fluency (r = -0.44, p = 0.01). High-grade CSO EPVS positively correlated with BBB leakage (r = 0.58, p < 0.001). The BBB permeability of CSO negatively correlated with verbal fluency (r = -0.52, p < 0.001) and attention (r = -0.40, p = 0.01).ConclusionsHigh-grade CSO EPVS is related to BBB leakage, which contributes to cognitive impairment in AD patients, especially verbal frequency. CSO EPVS can function as a convenient AD marker for intervention and therapy.
Collapse
Affiliation(s)
- Chen Wen
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jing-Huan Gan
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Hao Lu
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Li-Chen Wang
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin, China
| | - Hao Wu
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Zhi-Hong Shi
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| | - Yong Ji
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, China National Clinical Research Center for Neurological Diseases, Beijing, China
- Department of Neurology, Tianjin Huanhu Hospital, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin, China
| |
Collapse
|
5
|
Kizilçay AO, Tütüncü B, Koçarslan M, Gözel MA. Effects of 1800 MHz and 2100 MHz mobile phone radiation on the blood-brain barrier of New Zealand rabbits. Med Biol Eng Comput 2025; 63:915-932. [PMID: 39548043 DOI: 10.1007/s11517-024-03238-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
In this study, the impact of mobile phone radiation on blood-brain barrier (BBB) permeability was investigated. A total of 21 New Zealand rabbits were used for the experiments, divided into three groups, each consisting of 7 rabbits. One group served as the control, while the other two were exposed to electromagnetic radiation at frequencies of 1800 MHz with a distance of 14.5 cm and 2100 MHz with a distance of 17 cm, maintaining a constant power intensity of 15 dBm, for a duration equivalent to the current average daily conversation time of 38 min. The exposure was conducted under non-thermal conditions, with RF radiation levels approximately ten times lower than normal values. Evans blue (EB) dye was used as a marker to assess BBB permeability. EB binds to plasma proteins, and its presence in brain tissue indicates a disruption in BBB integrity, allowing for a quantitative evaluation of radiation-induced permeability changes. Left and right brain tissue samples were analyzed using trichloroacetic acid (TCA) and phosphate-buffered solution (PBS) solutions to measure EB amounts at 620 nm via spectrophotometry. After the experiments, BBB tissue samples were collected from the right and left brains of all rabbits in the three groups and subjected to a series of medical procedures. Samples from Group 1 were compared with those from Group 2 and Group 3 using statistical methods to determine if there were any significant differences. As a result, it was found that there was no statistically significant difference in the BBB of rabbits exposed to 1800 MHz radiation, whereas there was a statistically significant difference at a 95% confidence level in the BBB of rabbits exposed to 2100 MHz radiation. A decrease in EB values was observed upon the arithmetic examination of the BBB.
Collapse
Affiliation(s)
- Abdullah Oğuz Kizilçay
- Department of Computer Engineering, Zonguldak Bülent Ecevit University, Zonguldak, Turkey
| | - Bilal Tütüncü
- Energy Institute, Istanbul Technical University, Istanbul, Turkey.
| | - Mehmet Koçarslan
- Department of Sterilization, Dursun Odabaş Medical Faculty, Van Yüzüncü Yıl University, Van, Turkey
| | - Mahmut Ahmet Gözel
- Department of Electrical and Electronics Engineering, Süleyman Demirel University, Isparta, Turkey
| |
Collapse
|
6
|
Padrela BE, Slivka M, Sneve MH, Garrido PF, Dijsselhof MBJ, Hageman T, Geier O, Grydeland H, Mahroo A, Kuijer JPA, Konstandin S, Eickel K, Barkhof F, Günther M, Walhovd KB, Fjell AM, Mutsaerts HJMM, Petr J. Blood-brain barrier water permeability across the adult lifespan: A multi-echo ASL study. Neurobiol Aging 2025; 147:176-186. [PMID: 39798256 DOI: 10.1016/j.neurobiolaging.2024.12.012] [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: 08/13/2024] [Revised: 11/28/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
An emerging biomarker of blood-brain barrier (BBB) permeability is the time of exchange (Tex) of water from the blood to tissue, as measured by multi-echo arterial spin labeling (ASL) MRI. This new non-invasive sequence, already tested in mice, has recently been adapted to humans and optimized for clinical scanning time. In this study, we studied the normal variability of Tex over age and sex, which needs to be established as a reference for studying changes in neurological disease. We evaluated Tex, cerebral blood flow (CBF) and arterial transit time (ATT) in 209 healthy adults between 26 and 87 years, over age and sex, using general linear models in gray matter, white matter, and regionally in cerebral lobes. After QC, 194 participants were included in the main analysis, and the results demonstrated that both gray matter (GM) and white matter (WM) BBB permeability was higher with higher age (Tex lower by 0.47 ms per year in GM [p < 0.05], and by 0.49 ms in WM, for females; no significant for males), with the largest Tex difference in the frontal lobes (0.64 ms decrease per year, p = 0.011, population average). CBF was lower with higher age in the GM (-0.71 mL/min/100g per year, p < 0.001, for females; -0.31 mL/min/100g per year, p < 0.05, for males). When correcting Tex models for CBF and ATT, effect of age on Tex disappears in the GM, but not in the WM (β=-0.28, p = 0.08). The CBF findings of this study are in line with previous studies, demonstrating the validity of the new sequence. The BBB water permeability variation over age and sex described in this study provides a reference for future BBB research.
Collapse
Affiliation(s)
- Beatriz E Padrela
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands.
| | - Maksim Slivka
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Markus H Sneve
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Pablo F Garrido
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physics and Computational Radiology, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Mathijs B J Dijsselhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Tamara Hageman
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands
| | - Oliver Geier
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Department of Physics and Computational Radiology, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Håkon Grydeland
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway
| | - Amnah Mahroo
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Joost P A Kuijer
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Simon Konstandin
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; Bremerhaven University of Applied Sciences, Bremerhaven, Germany
| | - Klaus Eickel
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; Bremerhaven University of Applied Sciences, Bremerhaven, Germany
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands; Queen Square Institute of Neurology and Centre for Medical Image Computing (CMIC), University College London, London, UK
| | - Matthias Günther
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany; mediri GmbH, Heidelberg, Germany; Faculty 1 - Physics / Electrical Engineering, University Bremen, Bremen, Germany
| | - Kristine B Walhovd
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Computational Radiology and Artificial Intelligence, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Anders M Fjell
- Center for Lifespan Changes in Brain and Cognition, Department of Psychology, University of Oslo, Norway; Computational Radiology and Artificial Intelligence, Clinics of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Henk J M M Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Amsterdam Neuroscience, Brain Imaging, Amsterdam, the Netherlands
| | - Jan Petr
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Amsterdam University Medical Center, Location VUmc, Amsterdam, the Netherlands; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| |
Collapse
|
7
|
Digiovanni S, Lorenzati M, Bianciotto OT, Godel M, Fontana S, Akman M, Costamagna C, Couraud PO, Buffo A, Kopecka J, Riganti C, Salaroglio IC. Blood-brain barrier permeability increases with the differentiation of glioblastoma cells in vitro. Fluids Barriers CNS 2024; 21:89. [PMID: 39487455 PMCID: PMC11529439 DOI: 10.1186/s12987-024-00590-0] [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: 12/31/2023] [Accepted: 10/24/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is an aggressive tumor, difficult to treat pharmacologically because of the blood-brain barrier (BBB), which is rich in ATP-binding cassette (ABC) transporters and tight junction (TJ) proteins. The BBB is disrupted within GBM bulk, but it is competent in brain-adjacent-to-tumor areas, where eventual GBM foci can trigger tumor relapse. How GBM cells influence the permeability of BBB is poorly investigated. METHODS To clarify this point, we co-cultured human BBB models with 3 patient-derived GBM cells, after separating from each tumor the stem cell/neurosphere (SC/NS) and the differentiated/adherent cell (AC) components. Also, we set up cultures of BBB cells with the conditioned medium of NS or AC, enriched or depleted of IL-6. Extracellular cytokines were measured by protein arrays and ELISA. The intracellular signaling in BBB cells was measured by immunoblotting, in the presence of STAT3 pharmacological inhibitor or specific PROTAC. The competence of BBB was evaluated by permeability assays and TEER measurement. RESULTS The presence of GBM cells or their conditioned medium increased the permeability to doxorubicin, mitoxantrone and dextran-70, decreased TEER, down-regulated ABC transporters and TJ proteins at the transcriptional level. These effects were higher with AC or their medium than with NS. The secretome analysis identified IL-6 as significantly more produced by AC than by NS. Notably, AC-conditioned medium treated with an IL-6 neutralizing antibody reduced the BBB permeability to NS levels, while NS-conditioned medium enriched with IL-6 increased BBB permeability to AC levels. Mechanistically, IL-6 released by AC GBM cells activated STAT3 in BBB cells. In turn, STAT3 down-regulated ABC transporter and TJ expression, increased permeability and decreased TEER. The same effects were obtained in BBB cells treated with STA-21, a pharmacological inhibitor of STAT3, or with a PROTAC targeting STAT3. CONCLUSIONS Our work demonstrates for the first time that the degree of GBM differentiation influences BBB permeability. The crosstalk between GBM cells that release IL-6 and BBB cells that respond by activating STAT3, controls the expression of ABC transporters and TJ proteins on BBB. These results may pave the way for novel therapeutic tools to tune BBB permeability and improve drug delivery to GBM.
Collapse
Affiliation(s)
- Sabrina Digiovanni
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Martina Lorenzati
- Department of Neuroscience Rita Levi Montalcini, Institute Cavalieri Ottolenghi, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Olga Teresa Bianciotto
- Department of Neuroscience Rita Levi Montalcini, Institute Cavalieri Ottolenghi, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Martina Godel
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Simona Fontana
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Muhlis Akman
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Costanzo Costamagna
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | | | - Annalisa Buffo
- Department of Neuroscience Rita Levi Montalcini, Institute Cavalieri Ottolenghi, Regione Gonzole 10, Orbassano, 10043, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy.
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy.
| | - Iris Chiara Salaroglio
- Department of Oncology, University of Torino, piazza Nizza 44, Torino, 10126, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, piazza Nizza 44, Torino, 10126, Italy
| |
Collapse
|
8
|
Joseph CR. Assessing Mild Traumatic Brain Injury-Associated Blood-Brain Barrier (BBB) Damage and Restoration Using Late-Phase Perfusion Analysis by 3D ASL MRI: Implications for Predicting Progressive Brain Injury in a Focused Review. Int J Mol Sci 2024; 25:11522. [PMID: 39519073 PMCID: PMC11547134 DOI: 10.3390/ijms252111522] [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: 08/08/2024] [Revised: 10/09/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Mild traumatic brain injury (mTBI) is a common occurrence around the world, associated with a variety of blunt force and torsion injuries affecting all age groups. Most never reach medical attention, and the identification of acute injury and later clearance to return to usual activities is relegated to clinical evaluation-particularly in sports injuries. Advanced structural imaging is rarely performed due to the usual absence of associated acute anatomic/hemorrhagic changes. This review targets physiologic imaging techniques available to identify subtle blood-brain barrier dysfunction and white matter tract shear injury and their association with chronic traumatic encephalopathy. These techniques provide needed objective measures to assure recovery from injury in those patients with persistent cognitive/emotional symptoms and in the face of repetitive mTBI.
Collapse
Affiliation(s)
- Charles R Joseph
- Department of Neurology and Internal Medicine, College of Osteopathic Medicine, Liberty University, Lynchburg, VA 24502, USA
| |
Collapse
|
9
|
Soyer A, Goutal S, Leterrier S, Marie S, Larrat B, Selingue E, Winkeler A, Sarazin M, Bottlaender M, Tournier N. [ 18F]2-fluoro-2-deoxy-sorbitol ([ 18F]FDS) PET imaging repurposed for quantitative estimation of blood-brain barrier permeability in a rat model of Alzheimer's disease. ANNALES PHARMACEUTIQUES FRANÇAISES 2024; 82:822-829. [PMID: 38657857 DOI: 10.1016/j.pharma.2024.04.004] [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: 03/04/2024] [Revised: 04/05/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Numerous studies suggest that blood-brain barrier (BBB) dysfunction may contribute to the progression of Alzheimer's disease (AD). Clinically available neuroimaging methods are needed for quantitative "scoring" of BBB permeability in AD patients. [18F]2-fluoro-2-deoxy-sorbitol ([18F]FDS), which can be easily obtained from simple chemical reduction of commercial [18F]2-fluoro-2-deoxy-glucose ([18F]FDG), was investigated as a small-molecule marker of BBB permeability, in a pre-clinical model of AD using in vivo PET imaging. Chemical reduction of [18F]FDG to [18F]FDS was obtained with a 100% conversion yield. Dynamic PET acquisitions were performed in the APP/PS1 rat model of AD (TgF344-AD, n=3) compared with age-matched littermates (WT, n=4). The brain uptake of [18F]FDS was determined in selected brain regions, delineated from a coregistered rat brain template. The brain uptake of [18F]FDS in the brain regions of AD rats versus WT rats was compared using a 2-way ANOVA. The uptake of [18F]FDS was significantly higher in the whole brain of AD rats, as compared with WT rats (P<0.001), suggesting increased BBB permeability. Enhanced brain uptake of [18F]FDS in AD rats was significantly different across brain regions (P<0.001). Minimum difference was observed in the amygdala (+89.0±7.6%, P<0.001) and maximum difference was observed in the midbrain (+177.8±29.2%, P<0.001). [18F]FDS, initially proposed as radio-pharmaceutical to estimate renal filtration using PET imaging, can be repurposed for non-invasive and quantitative determination of BBB permeability in vivo. Making the best with the quantitative properties of PET imaging, it was possible to estimate the extent of enhanced BBB permeability in a rat model of AD.
Collapse
Affiliation(s)
- Amélie Soyer
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Sébastien Goutal
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Sarah Leterrier
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Solène Marie
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Benoit Larrat
- Centre d'études de Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Paris-Saclay University, 91191 Gif-sur-Yvette, France
| | - Erwan Selingue
- Centre d'études de Saclay, CEA, CNRS, NeuroSpin/BAOBAB, Paris-Saclay University, 91191 Gif-sur-Yvette, France
| | - Alexandra Winkeler
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Marie Sarazin
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Michel Bottlaender
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France
| | - Nicolas Tournier
- Service hospitalier Frédéric-Joliot, laboratoire d'imagerie biomédicale multimodale (BioMaps), Inserm, CNRS, CEA, université Paris-Saclay, 91401 Orsay, France.
| |
Collapse
|
10
|
Leterrier S, Goutal S, Hugon G, Goislard M, Saba W, Hosten B, Specklin S, Winkeler A, Tournier N. Imaging quantitative changes in blood-brain barrier permeability using [ 18F]2-fluoro-2-deoxy-sorbitol ([ 18F]FDS) PET in relation to glial cell recruitment in a mouse model of endotoxemia. J Cereb Blood Flow Metab 2024; 44:1117-1127. [PMID: 38441006 PMCID: PMC11179610 DOI: 10.1177/0271678x241236755] [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: 09/15/2023] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/06/2024]
Abstract
The quantitative relationship between the disruption of the blood-brain barrier (BBB) and the recruitment of glial cells was explored in a mouse model of endotoxemia. [18F]2-Fluoro-2-deoxy-sorbitol ([18F]FDS) PET imaging was used as a paracellular marker for quantitative monitoring of BBB permeability after i.v injection of increasing doses of lipopolysaccharide (LPS) or vehicle (saline, n = 5). The brain distribution of [18F]FDS (VT, mL.cm-3) was estimated using kinetic modeling. LPS dose-dependently increased the brain VT of [18F]FDS after injection of LPS 4 mg/kg (5.2 ± 2.4-fold, n = 4, p < 0.01) or 5 mg/kg (9.0 ± 9.1-fold, n = 4, p < 0.01) but not 3 mg/kg (p > 0.05, n = 7). In 12 individuals belonging to the different groups, changes in BBB permeability were compared with expression of markers of astrocyte (GFAP) and microglial cell (CD11b) using ex vivo immunohistochemistry. Increased expression of CD11b and GFAP expression was observed in mice injected with 3 mg/kg of LPS, which did not increase with higher LPS doses. Quantitative [18F]FDS PET imaging can capture different levels of BBB permeability in vivo. A biphasic effect was observed with the lowest dose of LPS that triggered neuroinflammation without disruptive changes in BBB permeability, and higher LPS doses that increased BBB permeability without additional recruitment of glial cells.
Collapse
Affiliation(s)
- Sarah Leterrier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Sébastien Goutal
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Gaëlle Hugon
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Maud Goislard
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Wadad Saba
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Benoit Hosten
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Simon Specklin
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Alexandra Winkeler
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| | - Nicolas Tournier
- Laboratoire d’Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Université Paris-Saclay, Inserm, CNRS, Orsay, France
| |
Collapse
|
11
|
Reiter JT, Schulte F, Bauer T, David B, Endler C, Isaak A, Schuch F, Bitzer F, Witt JA, Hattingen E, Deichmann R, Attenberger U, Becker AJ, Helmstaedter C, Radbruch A, Surges R, Friedman A, Rüber T. Evidence for interictal blood-brain barrier dysfunction in people with epilepsy. Epilepsia 2024; 65:1462-1474. [PMID: 38436479 DOI: 10.1111/epi.17929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 02/11/2024] [Accepted: 02/12/2024] [Indexed: 03/05/2024]
Abstract
OBJECTIVE Interictal blood-brain barrier dysfunction in chronic epilepsy has been demonstrated in animal models and pathological specimens. Ictal blood-brain barrier dysfunction has been shown in humans in vivo using an experimental quantitative magnetic resonance imaging (MRI) protocol. Here, we hypothesized that interictal blood-brain barrier dysfunction is also present in people with drug-resistant epilepsy. METHODS Thirty-nine people (21 females, mean age at MRI ± SD = 30 ± 8 years) with drug-resistant epilepsy were prospectively recruited and underwent interictal T1-relaxometry before and after administration of a paramagnetic contrast agent. Likewise, quantitative T1 was acquired in 29 people without epilepsy (12 females, age at MRI = 48 ± 18 years). Quantitative T1 difference maps were calculated and served as a surrogate imaging marker for blood-brain barrier dysfunction. Values of quantitative T1 difference maps inside hemispheres ipsilateral to the presumed seizure onset zone were then compared, on a voxelwise level and within presumed seizure onset zones, to the contralateral side of people with epilepsy and to people without epilepsy. RESULTS Compared to the contralateral side, ipsilateral T1 difference values were significantly higher in white matter (corrected p < .05), gray matter (uncorrected p < .05), and presumed seizure onset zones (p = .04) in people with epilepsy. Compared to people without epilepsy, significantly higher T1 difference values were found in the anatomical vicinity of presumed seizure onset zones (p = .004). A subgroup of people with hippocampal sclerosis demonstrated significantly higher T1 difference values in the ipsilateral hippocampus and in regions strongly interconnected with the hippocampus compared to people without epilepsy (corrected p < .01). Finally, z-scores reflecting the deviation of T1 difference values within the presumed seizure onset zone were associated with verbal memory performance (p = .02) in people with temporal lobe epilepsy. SIGNIFICANCE Our results indicate a blood-brain barrier dysfunction in drug-resistant epilepsy that is detectable interictally in vivo, anatomically related to the presumed seizure onset zone, and associated with cognitive deficits.
Collapse
Affiliation(s)
- Johannes T Reiter
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Freya Schulte
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Tobias Bauer
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | - Bastian David
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Christoph Endler
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Alexander Isaak
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Fabiane Schuch
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Felix Bitzer
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| | | | - Elke Hattingen
- Institute of Neuroradiology, University Hospital and Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ralf Deichmann
- Brain Imaging Center, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ulrike Attenberger
- Department of Diagnostic and Interventional Radiology, University Hospital Bonn, Bonn, Germany
| | - Albert J Becker
- Department of Neuropathology, University Hospital Bonn, Bonn, Germany
| | | | | | - Rainer Surges
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
| | - Alon Friedman
- Department of Medical Neuroscience, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia, Canada
- Departments of Cognitive and Brain Sciences, Physiology, and Cell Biology, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Theodor Rüber
- Department of Epileptology, University Hospital Bonn, Bonn, Germany
- Department of Neuroradiology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
12
|
Hosten B, Goutal S, Leterrier S, Corvo C, Breuil L, Barret O, Specklin S, Truillet C, Tournier N. Brain delivery enabled by transient blood-brain barrier disruption induced by regadenoson: a PET imaging study. Expert Opin Drug Deliv 2024; 21:797-807. [PMID: 38881261 DOI: 10.1080/17425247.2024.2369765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024]
Abstract
BACKGROUND Regadenoson, an agonist of adenosine A2 receptors, enables transient blood-brain barrier (BBB) disruption. The relevance of regadenoson as a pharmacological strategy for brain delivery was investigated using in vivo PET imaging in rats. RESEARCH DESIGN AND METHODS Kinetic modeling of brain PET data was performed to estimate the impact of regadenoson (0.05 mg.kg-1, i.v.) on BBB permeation compared with control rats (n = 4-6 per group). Three radiolabeled compounds of different sizes, which do not cross the intact BBB, were tested. RESULTS Regadenoson significantly increased the BBB penetration (+116 ± 13%, p < 0.001) of [18F]2-deoxy-2-fluoro-D-sorbitol ([18F]FDS, MW = 183 Da), a small-molecule marker of BBB permeability. The magnitude of the effect was different across brain regions, with a maximum increase in the striatum. Recovery of BBB integrity was observed 30 min after regadenoson injection. Regadenoson also increased the brain penetration (+72 ± 45%, p < 0.05) of a radiolabeled nanoparticle [89Zr]AGuIX (MW = 9 kDa). However, the brain kinetics of a monoclonal antibody ([89Zr]mAb, MW = 150 kDa) remained unchanged (p > 0.05). CONCLUSIONS PET imaging showed the features and limitations of BBB disruption induced by regadenoson in terms of extent, regional distribution, and reversibility. Nevertheless, regadenoson enables the brain delivery of small molecules or nanoparticles in rats.
Collapse
Affiliation(s)
- Benoit Hosten
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
- INSERM UMR1144, Université Paris Cité, Paris, France
| | - Sébastien Goutal
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Sarah Leterrier
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Cassandre Corvo
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Louise Breuil
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
- INSERM UMR1144, Université Paris Cité, Paris, France
| | - Olivier Barret
- CEA, CNRS, Université Paris-Saclay, MIRCen, Laboratoire des Maladies Neurodégénératives, Fontenay-Aux-Roses, France
| | - Simon Specklin
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Charles Truillet
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| | - Nicolas Tournier
- Université Paris-Saclay, Inserm, CNRS, CEA, Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Service Hospitalier Frédéric Joliot, Orsay, France
| |
Collapse
|
13
|
Zapata-Acevedo JF, Losada-Barragán M, Osma JF, Cruz JC, Reiber A, Petry KG, Caillard A, Sauldubois A, Llamosa Pérez D, Morillo Zárate AJ, Muñoz SB, Daza Moreno A, Silva RV, Infante-Duarte C, Chamorro-Coral W, González-Reyes RE, Vargas-Sánchez K. Specific nanoprobe design for MRI: Targeting laminin in the blood-brain barrier to follow alteration due to neuroinflammation. PLoS One 2024; 19:e0302031. [PMID: 38603692 PMCID: PMC11008835 DOI: 10.1371/journal.pone.0302031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
Chronic neuroinflammation is characterized by increased blood-brain barrier (BBB) permeability, leading to molecular changes in the central nervous system that can be explored with biomarkers of active neuroinflammatory processes. Magnetic resonance imaging (MRI) has contributed to detecting lesions and permeability of the BBB. Ultra-small superparamagnetic particles of iron oxide (USPIO) are used as contrast agents to improve MRI observations. Therefore, we validate the interaction of peptide-88 with laminin, vectorized on USPIO, to explore BBB molecular alterations occurring during neuroinflammation as a potential tool for use in MRI. The specific labeling of NPS-P88 was verified in endothelial cells (hCMEC/D3) and astrocytes (T98G) under inflammation induced by interleukin 1β (IL-1β) for 3 and 24 hours. IL-1β for 3 hours in hCMEC/D3 cells increased their co-localization with NPS-P88, compared with controls. At 24 hours, no significant differences were observed between groups. In T98G cells, NPS-P88 showed similar nonspecific labeling among treatments. These results indicate that NPS-P88 has a higher affinity towards brain endothelial cells than astrocytes under inflammation. This affinity decreases over time with reduced laminin expression. In vivo results suggest that following a 30-minute post-injection, there is an increased presence of NPS-P88 in the blood and brain, diminishing over time. Lastly, EAE animals displayed a significant accumulation of NPS-P88 in MRI, primarily in the cortex, attributed to inflammation and disruption of the BBB. Altogether, these results revealed NPS-P88 as a biomarker to evaluate changes in the BBB due to neuroinflammation by MRI in biological models targeting laminin.
Collapse
Affiliation(s)
- Juan F. Zapata-Acevedo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Mónica Losada-Barragán
- Grupo de Biología Celular y Funcional e Ingeniería de Biomoleculas, Departamento de Biología, Universidad Antonio Nariño, Bogotá, Colombia
| | - Johann F. Osma
- Department of Electrical and Electronic Engineering, Universidad de los Andes, Bogotá, Colombia
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá, Colombia
| | - Andreas Reiber
- Chemistry Department, Grupo La Quimica en la interfase inorgánica-orgánica QUINORG, Universidad de los Andes, Bogotá, Colombia
| | - Klaus G. Petry
- CNRS UMR 5536 Centre de Resonance Magnétique des Systemes Biologiques and INSERM U1049 Neuroinflammation, University of Bordeaux, Bordeaux, France
| | | | | | - Daniel Llamosa Pérez
- Facultad de Ciencias, Grupo Investigación fundamental y aplicada en Materiales, Universidad Antonio Nariño, Bogotá, Colombia
| | | | | | - Agustín Daza Moreno
- Oficial de Protección Radiológica, Fundación Santa Fé de Bogotá, Bogotá, Colombia
| | - Rafaela V. Silva
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Infante-Duarte
- Experimental and Clinical Research Center, a Cooperation between the Max Delbrück Center for Molecular Medicine in the Helmholtz Association and Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - William Chamorro-Coral
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| | - Rodrigo E. González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencia Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Karina Vargas-Sánchez
- Laboratorio de Neurofisiología Celular, Grupo de Neurociencia Traslacional, Facultad de Medicina, Universidad de los Andes, Bogotá, Colombia
| |
Collapse
|
14
|
Joseph CR. Progressive Age-Associated Blood-Brain Barrier Leak/Dysfunction-Nexus of Neurodegenerative Disease Using MRI Markers to Identify Preclinical Disease and Potential New Targets for Future Treatments. Diagnostics (Basel) 2024; 14:726. [PMID: 38611639 PMCID: PMC11011559 DOI: 10.3390/diagnostics14070726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
This review article focuses on the upstream pertinent pathophysiology leading to neurodegenerative disease. Specifically, the nexus appears to be blood-brain barrier (BBB) leakiness resulting in a two-prong inflammatory disease spectrum damaging the microvasculature and corrupting protein synthesis and degradation with accumulating misfolded toxic proteins. The suboptimal results of removing misfolded proteins mean a new approach to disease in the preclinical state is required aimed at other targets. Validated noninvasive imaging and serologic biomarkers of early preclinical disease implemented in the high-risk patient cohort along with periodic surveillance once effective treatments are developed will be required. This review discusses the physiology and pathophysiology of the BBB, new MRI imaging techniques identifying the leak, and altered fluid dynamic effects in the preclinical state. The risk factors for disease development, preventative measures, and potential treatment targets are also discussed.
Collapse
Affiliation(s)
- Charles R Joseph
- Neurology and Internal Medicine, College of Osteopathic Medicine, Liberty University, Lynchburg, VA 24502, USA
| |
Collapse
|
15
|
Joseph CR, Lim JK, Grohol BN, Zivcevska M, Lencke J, Rich ED, Arrasmith CJ, Dorman IS, Clark BW, Love K, Ferry B, Rolfs ME. Identifying delay in glymphatic clearance of labeled protons post-acute head trauma utilizing 3D ASL MRI (arterial spin labeling): a pilot study. Sci Rep 2024; 14:6188. [PMID: 38485759 PMCID: PMC10940642 DOI: 10.1038/s41598-024-56236-6] [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: 11/02/2023] [Accepted: 03/04/2024] [Indexed: 03/18/2024] Open
Abstract
This study correlated mild traumatic brain injury (mTBI) cognitive changes with ASL-MRI glymphatic clearance rates (GCRs) and recovery with GCR improvement. mTBI disrupts the blood brain barrier (BBB), reducing capillary mean transit time and GCRs. mTBI is clinically diagnosed utilizing history/examination findings with no physiologic biomarkers. 3D TGSE (turbo-gradient spin-echo) pulsed arterial spin-labeling 3T MRI with 7 long inversion times (TIs) assessed the signal clearance of labeled protons 2800-4000 ms postlabeling in bifrontal, bitemporal, and biparietal regions within 7 days of mTBI and once clinically cleared to resume activities. The Sport Concussion Assessment Tool Version 5 (SKAT5) and Brief Oculomotor/Vestibular Assessment evaluated injured athletes' cognitive function prior to MRIs. The pilot study demonstrated significant GCRs improvement (95% CI - 0.06 to - 0.03 acute phase; to CI-recovery CI 0.0772 to - 0.0497; P < 0.001 in frontal lobes; and parietal lobes (95% CI - 0.0584 to - 0.0251 acute; CI - 0.0727 to - 0.0392 recovery; P = 0.024) in 9 mTBI athletes (8 female, 1 male). Six age/activity-matched controls (4 females, 2 males) were also compared. mTBI disrupts the BBB, reducing GCR measured using the 3D ASL MRI technique. ASL MRI is a potential noninvasive biomarker of mTBI and subsequent recovery.
Collapse
Affiliation(s)
- Charles R Joseph
- Liberty University College of Osteopathic Medicine, Lynchburg, USA.
| | - Jubin Kang Lim
- Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Bryce N Grohol
- Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Marija Zivcevska
- Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Joshua Lencke
- Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Ethan Dean Rich
- Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | | | | | | | - Kim Love
- K. R. Love Quantitative Consulting and Collaboration, Athens, USA
| | - Ben Ferry
- Liberty University College of Osteopathic Medicine, Lynchburg, USA
| | - Mark E Rolfs
- Liberty University College of Osteopathic Medicine, Lynchburg, USA
| |
Collapse
|
16
|
van der Knaap N, Ariës MJH, van der Horst ICC, Jansen JFA. On the merits and potential of advanced neuroimaging techniques in COVID-19: A scoping review. Neuroimage Clin 2024; 42:103589. [PMID: 38461701 PMCID: PMC10938171 DOI: 10.1016/j.nicl.2024.103589] [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: 01/09/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/12/2024]
Abstract
Many Coronavirus Disease 2019 (COVID-19) patients are suffering from long-term neuropsychological sequelae. These patients may benefit from a better understanding of the underlying neuropathophysiological mechanisms and identification of potential biomarkers and treatment targets. Structural clinical neuroimaging techniques have limited ability to visualize subtle cerebral abnormalities and to investigate brain function. This scoping review assesses the merits and potential of advanced neuroimaging techniques in COVID-19 using literature including advanced neuroimaging or postmortem analyses in adult COVID-19 patients published from the start of the pandemic until December 2023. Findings were summarized according to distinct categories of reported cerebral abnormalities revealed by different imaging techniques. Although no unified COVID-19-specific pattern could be subtracted, a broad range of cerebral abnormalities were revealed by advanced neuroimaging (likely attributable to hypoxic, vascular, and inflammatory pathology), even in absence of structural clinical imaging findings. These abnormalities are validated by postmortem examinations. This scoping review emphasizes the added value of advanced neuroimaging compared to structural clinical imaging and highlights implications for brain functioning and long-term consequences in COVID-19.
Collapse
Affiliation(s)
- Noa van der Knaap
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Research Institute of Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Marcel J H Ariës
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Research Institute of Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Iwan C C van der Horst
- Department of Intensive Care Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands; Research Institute of Mental Health & Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
17
|
Choi S, Lake S, Harrison DM. Evaluation of the Blood-Brain Barrier, Demyelination, and Neurodegeneration in Paramagnetic Rim Lesions in Multiple Sclerosis on 7 Tesla MRI. J Magn Reson Imaging 2024; 59:941-951. [PMID: 37276054 PMCID: PMC10754232 DOI: 10.1002/jmri.28847] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Paramagnetic rim lesions (PRLs) are associated with chronic inflammation in multiple sclerosis (MS). 7-Tesla (7T) magnetic resonance imaging (MRI) can evaluate the integrity of the blood-brain barrier (BBB) in addition to the tissue myelination status and cell loss. PURPOSE To use MRI metrics to investigate underlying physiology and clinical importance of PRLs. STUDY TYPE Prospective. SUBJECTS Thirty-six participants (mean-age 47, 23 females, 13 males) of mixed MS subtypes. FIELD STRENGTH/SEQUENCE 7T, MP2RAGE, MULTI-ECHO 3D-GRE, FLAIR. ASSESSMENT Lesion heterogeneity; longitudinal changes in lesion counts; comparison of T1, R2*, and χ; association between baseline lesion types and disease progression (2-3 annual MRI visits with additional years of annual clinical follow-up). STATISTICAL TESTS Two-sample t-test, Wilcoxon Rank-Sum test, Pearson's chi-square test, two-group comparison with linear-mixed-effect model, mixed-effect ANOVA, logistic regression. P-values <0.05 were considered significant. RESULTS A total of 58.3% of participants had at least one PRL at baseline. Higher male proportion in PRL+ group was found. Average change in PRL count was 0.20 (SD = 2.82) for PRLs and 0.00 (SD = 0.82) for mottled lesions. Mean and median pre-/post-contrast T1 were longer in PRL+ than in PRL-. No differences in mean χ were seen for lesions grouped by PRL (P = 0.310, pre-contrast; 0.086, post-contrast) or PRL/M presence (P = 0.234, pre-contrast; 0.163, post-contrast). Median χ were less negative in PRL+ and PRL/M+ than in PRL- and PRL/M-. Mean and median pre-/post-contrast R2* were slower in PRL+ compared to PRL-. Mean and median pre-/post-contrast R2* were slower in PRL/M+ than in PRL/M-. PRL presence at baseline was associated with confirmed EDSS Plus progression (OR 3.75 [1.22-7.59]) and PRL/M+ at baseline with confirmed EDSS Plus progression (OR 3.63 [1.14-7.43]). DATA CONCLUSION Evidence of BBB breakdown in PRLs was not seen. Quantitative metrics confirmed prior results suggesting greater demyelination, cell loss, and possibly disruption of tissue anisotropy in PRLs. EVIDENCE LEVEL 2 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Seongjin Choi
- Department of Neurology, University of Maryland School of Medicine, Baltimore Maryland
| | - Sarah Lake
- Hasbro Children’s Hospital, Brown University
| | - Daniel M. Harrison
- Department of Neurology, University of Maryland School of Medicine, Baltimore Maryland
- Department of Neurology, Baltimore Veterans Affairs Medical Center, Baltimore, Maryland
| |
Collapse
|
18
|
Doron O, Patel AB, Hawryluk GWJ. Neurovascular Interventions for Neurotrauma: From Treatment of Injured Vessels to Treatment of the Injured Brain? Oper Neurosurg (Hagerstown) 2024; 26:247-255. [PMID: 37976141 DOI: 10.1227/ons.0000000000000980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 09/17/2023] [Indexed: 11/19/2023] Open
Abstract
Traumatic brain injury is often associated with a direct or secondary neurovascular pathology. In this review, we present recent advancements in endovascular neurosurgery that enable accurate and effective vessel reconstruction with emphasis on its role in early diagnosis, the expanding use of flow diversion in pseudoaneurysms, and traumatic arteriovenous fistulas. In addition, future directions in which catheter-based interventions could potentially affect traumatic brain injury are described: targeting blood brain barrier integrity using the advantages of intra-arterial drug delivery of blood brain barrier stabilizers to prevent secondary brain edema, exploring the impact of endovascular venous access as a means to modulate venous outflow in an attempt to reduce intracranial pressure and augment brain perfusion, applying selective intra-arterial hypothermia as a neuroprotection method mitigating some of the risks conferred by systemic cooling, trans-vessel wall delivery of regenerative therapy agents, and shifting attention using multimodal neuromonitoring to post-traumatic vasospasm to further characterize the role it plays in secondary brain injury. Thus, we believe that the potential of endovascular tools can be expanded because they enable access to the "highways" governing perfusion and flow and call for further research focused on exploring these routes because it may contribute to novel endovascular approaches currently used for treating injured vessels, harnessing them for treatment of the injured brain.
Collapse
Affiliation(s)
- Omer Doron
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston , Massachusetts , USA
- Department of Biomedical Engineering, The Aldar and Iby Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv , Israel
| | - Aman B Patel
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston , Massachusetts , USA
| | - Gregory W J Hawryluk
- Department of Neurosurgery, Akron General Neuroscience Institute, Cleveland Clinic, Akron , Ohio , USA
| |
Collapse
|
19
|
Voorter PHM, van Dinther M, Jansen WJ, Postma AA, Staals J, Jansen JFA, van Oostenbrugge RJ, van der Thiel MM, Backes WH. Blood-Brain Barrier Disruption and Perivascular Spaces in Small Vessel Disease and Neurodegenerative Diseases: A Review on MRI Methods and Insights. J Magn Reson Imaging 2024; 59:397-411. [PMID: 37658640 DOI: 10.1002/jmri.28989] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 09/03/2023] Open
Abstract
Perivascular spaces (PVS) and blood-brain barrier (BBB) disruption are two key features of cerebral small vessel disease (cSVD) and neurodegenerative diseases that have been linked to cognitive impairment and are involved in the cerebral waste clearance system. Magnetic resonance imaging (MRI) offers the possibility to study these pathophysiological processes noninvasively in vivo. This educational review provides an overview of the MRI techniques used to assess PVS functionality and BBB disruption. MRI-visible PVS can be scored on structural images by either (subjectively) counting or (automatically) delineating the PVS. We highlight emerging (diffusion) techniques to measure proxies of perivascular fluid and its movement, which may provide a more comprehensive understanding of the role of PVS in diseases. For the measurement of BBB disruption, we explain the most commonly used MRI technique, dynamic contrast-enhanced (DCE) MRI, as well as a more recently developed technique based on arterial spin labeling (ASL). DCE MRI and ASL are thought to measure complementary characteristics of the BBB. Furthermore, we describe clinical studies that have utilized these MRI techniques in cSVD and neurodegenerative diseases, particularly Alzheimer's disease (AD). These studies demonstrate the role of PVS and BBB dysfunction in these diseases and provide insight into the large overlap, but also into the differences between cSVD and AD. Overall, MRI techniques may provide valuable insights into the pathophysiological mechanisms underlying these diseases and have the potential to be used as markers for disease progression and treatment response. LEVEL OF EVIDENCE: 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Paulien H M Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Maud van Dinther
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Willemijn J Jansen
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Julie Staals
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Robert J van Oostenbrugge
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
- Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Merel M van der Thiel
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- Department of Psychiatry and Neuropsychology, Alzheimer Center Limburg, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
- School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
20
|
Abdelmoaty MM, Lu E, Kadry R, Foster EG, Bhattarai S, Mosley RL, Gendelman HE. Clinical biomarkers for Lewy body diseases. Cell Biosci 2023; 13:209. [PMID: 37964309 PMCID: PMC10644566 DOI: 10.1186/s13578-023-01152-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023] Open
Abstract
Synucleinopathies are a group of neurodegenerative disorders characterized by pathologic aggregates of neural and glial α-synuclein (α-syn) in the form of Lewy bodies (LBs), Lewy neurites, and cytoplasmic inclusions in both neurons and glia. Two major classes of synucleinopathies are LB disease and multiple system atrophy. LB diseases include Parkinson's disease (PD), PD with dementia, and dementia with LBs. All are increasing in prevalence. Effective diagnostics, disease-modifying therapies, and therapeutic monitoring are urgently needed. Diagnostics capable of differentiating LB diseases are based on signs and symptoms which might overlap. To date, no specific diagnostic test exists despite disease-specific pathologies. Diagnostics are aided by brain imaging and cerebrospinal fluid evaluations, but more accessible biomarkers remain in need. Mechanisms of α-syn evolution to pathologic oligomers and insoluble fibrils can provide one of a spectrum of biomarkers to link complex neural pathways to effective therapies. With these in mind, we review promising biomarkers linked to effective disease-modifying interventions.
Collapse
Affiliation(s)
- Mai M Abdelmoaty
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Eugene Lu
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rana Kadry
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Emma G Foster
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Shaurav Bhattarai
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - R Lee Mosley
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
21
|
Palmer JM, Huentelman M, Ryan L. More than just risk for Alzheimer's disease: APOE ε4's impact on the aging brain. Trends Neurosci 2023; 46:750-763. [PMID: 37460334 DOI: 10.1016/j.tins.2023.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/16/2023] [Accepted: 06/12/2023] [Indexed: 08/18/2023]
Abstract
The apolipoprotein ε4 (APOE ε4) allele is most commonly associated with increased risk for late-onset Alzheimer's disease (AD). However, recent longitudinal studies suggest that these risks are overestimated; most ε4 carriers will not develop dementia in their lifetime. In this article, we review new evidence regarding the impact of APOE ε4 on cognition among healthy older adults. We discuss emerging work from animal models suggesting that ε4 impacts brain structure and function in multiple ways that may lead to age-related cognitive impairment, independent from AD pathology. We discuss the importance of taking an individualized approach in future studies by incorporating biomarkers and neuroimaging methods that may better disentangle the phenotypic influences of APOE ε4 on the aging brain from prodromal AD pathology.
Collapse
Affiliation(s)
- Justin M Palmer
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| | - Matthew Huentelman
- Translational Genomics Research Institute, Phoenix, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA
| | - Lee Ryan
- The University of Arizona, Tucson, AZ, USA; Arizona Alzheimer's Consortium, Phoenix, AZ, USA.
| |
Collapse
|
22
|
Tap L, Vernooij MW, Wolters F, van den Berg E, Mattace-Raso FUS. New horizons in cognitive and functional impairment as a consequence of cerebral small vessel disease. Age Ageing 2023; 52:afad148. [PMID: 37585592 PMCID: PMC10431695 DOI: 10.1093/ageing/afad148] [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: 06/06/2023] [Indexed: 08/18/2023] Open
Abstract
Cerebral small vessel disease (cSVD) is a frequent finding in imaging of the brain in older adults, especially in the concomitance of cardiovascular disease risk factors. Despite the well-established link between cSVD and (vascular) cognitive impairment (VCI), it remains uncertain how and when these vascular alterations lead to cognitive decline. The extent of acknowledged markers of cSVD is at best modestly associated with the severity of clinical symptoms, but technological advances increasingly allow to identify and quantify the extent and perhaps also the functional impact of cSVD more accurately. This will facilitate a more accurate diagnosis of VCI, against the backdrop of concomitant other neurodegenerative pathology, and help to identify persons with the greatest risk of cognitive and functional deterioration. In this study, we discuss how better assessment of cSVD using refined neuropsychological and comprehensive geriatric assessment as well as modern image analysis techniques may improve diagnosis and possibly the prognosis of VCI. Finally, we discuss new avenues in the treatment of cSVD and outline how these contemporary insights into cSVD can contribute to optimise screening and treatment strategies in older adults with cognitive impairment and multimorbidity.
Collapse
Affiliation(s)
- Lisanne Tap
- Department of Internal Medicine, Section of Geriatric Medicine and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Meike W Vernooij
- Department of Radiology and Nuclear Medicine and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Frank Wolters
- Department of Epidemiology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Esther van den Berg
- Department of Neurology and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Francesco U S Mattace-Raso
- Department of Internal Medicine, Section of Geriatric Medicine and Alzheimer Center Erasmus MC, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
23
|
Pizarro-Galleguillos BM, Kunert L, Brüggemann N, Prasuhn J. Neuroinflammation and Mitochondrial Dysfunction in Parkinson's Disease: Connecting Neuroimaging with Pathophysiology. Antioxidants (Basel) 2023; 12:1411. [PMID: 37507950 PMCID: PMC10375976 DOI: 10.3390/antiox12071411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
There is a pressing need for disease-modifying therapies in patients suffering from neurodegenerative diseases, including Parkinson's disease (PD). However, these disorders face unique challenges in clinical trial designs to assess the neuroprotective properties of potential drug candidates. One of these challenges relates to the often unknown individual disease mechanisms that would, however, be relevant for targeted treatment strategies. Neuroinflammation and mitochondrial dysfunction are two proposed pathophysiological hallmarks and are considered to be highly interconnected in PD. Innovative neuroimaging methods can potentially help to gain deeper insights into one's predominant disease mechanisms, can facilitate patient stratification in clinical trials, and could potentially map treatment responses. This review aims to highlight the role of neuroinflammation and mitochondrial dysfunction in patients with PD (PwPD). We will specifically introduce different neuroimaging modalities, their respective technical hurdles and challenges, and their implementation into clinical practice. We will gather preliminary evidence for their potential use in PD research and discuss opportunities for future clinical trials.
Collapse
Affiliation(s)
- Benjamin Matís Pizarro-Galleguillos
- Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, 23562 Lübeck, Germany
- Center for Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Liesa Kunert
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, 23562 Lübeck, Germany
- Center for Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Norbert Brüggemann
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
- Center for Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
| | - Jannik Prasuhn
- Department of Neurology, University Medical Center Schleswig-Holstein, Campus Lübeck, 23562 Lübeck, Germany
- Institute of Neurogenetics, University of Lübeck, 23562 Lübeck, Germany
- Center for Brain, Behavior, and Metabolism, University of Lübeck, 23562 Lübeck, Germany
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21287, USA
| |
Collapse
|
24
|
Elschot EP, Backes WH, de Jong JJA, Drenthen GS, Wong SM, Staals J, Postma AA, Rouhl RPW, van Oostenbrugge RJ, Jansen JFA. Assessment of the clinical feasibility of detecting subtle blood-brain barrier leakage in cerebral small vessel disease using dynamic susceptibility contrast MRI. Magn Reson Imaging 2023; 102:55-61. [PMID: 37137345 DOI: 10.1016/j.mri.2023.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
PURPOSE Cerebral small vessel disease (cSVD) involves several pathologies affecting the small vessels, including blood-brain barrier (BBB) impairment. Dynamic susceptibility contrast (DSC) MRI is sensitive to both blood perfusion and BBB leakage, and correction methods may be crucial for obtaining reliable perfusion measures. These methods might also be applicable to detect BBB leakage itself. This study investigated to what extent DSC-MRI can measure subtle BBB leakage in a clinical feasibility setting. METHODS In vivo DCE and DSC data were collected from fifteen cSVD patients (71 (±10) years, 6F/9M) and twelve elderly controls (71 (±10) years, 4F/8M). DSC-derived leakage fractions were obtained using the Boxerman-Schmainda-Weisskoff method (K2). K2 was compared with the DCE-derived leakage rate Ki, obtained from Patlak analysis. Subsequently, differences were assessed between white matter hyperintensities (WMH), cortical gray matter (CGM), and normal-appearing white matter (NAWM). Additionally, computer simulations were performed to assess the sensitivity of DSC-MRI to BBB leakage. RESULTS K2 showed significant differences between tissue regions (P < 0.001 for CGM-NAWM and CGM-WMH, and P = 0.001 for NAWM-WMH). Conversely, according to the computer simulations the DSC sensitivity was insufficient to measure subtle BBB leakage, as the K2 values were below the derived limit of quantification (4∙10-3 min-1). As expected, Ki was elevated in the WMH compared to CGM and NAWM (P < 0.001). CONCLUSIONS Although clinical DSC-MRI seems capable to detect subtle BBB leakage differences between WMH and normal-appearing brain tissue it is not recommended. K2 as a direct measure for subtle BBB leakage remains ambiguous as its signal effects are due to mixed T1- and T2∗-weighting. Further research is warranted to better disentangle perfusion from leakage effects.
Collapse
Affiliation(s)
- Elles P Elschot
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Joost J A de Jong
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Gerhard S Drenthen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Sau May Wong
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Rob P W Rouhl
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; Department of Neurology, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; Academic Center for Epileptology Kempenhaeghe/MUMC+, Heeze and Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; Department of Neurology, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Cardiovascular Diseases (CARIM), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center+, P. Debyelaan 25, Maastricht, the Netherlands; School for Mental Health and Neuroscience (MHeNs), Maastricht University, Minderbroedersberg 4-6, Maastricht, the Netherlands; Department of Electrical Engineering, Eindhoven University of Technology, De Rondom 70, Eindhoven, the Netherlands; Academic Center for Epileptology Kempenhaeghe/MUMC+, Heeze and Maastricht, the Netherlands.
| |
Collapse
|
25
|
Moyaert P, Padrela BE, Morgan CA, Petr J, Versijpt J, Barkhof F, Jurkiewicz MT, Shao X, Oyeniran O, Manson T, Wang DJJ, Günther M, Achten E, Mutsaerts HJMM, Anazodo UC. Imaging blood-brain barrier dysfunction: A state-of-the-art review from a clinical perspective. Front Aging Neurosci 2023; 15:1132077. [PMID: 37139088 PMCID: PMC10150073 DOI: 10.3389/fnagi.2023.1132077] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/15/2023] [Indexed: 05/05/2023] Open
Abstract
The blood-brain barrier (BBB) consists of specialized cells that tightly regulate the in- and outflow of molecules from the blood to brain parenchyma, protecting the brain's microenvironment. If one of the BBB components starts to fail, its dysfunction can lead to a cascade of neuroinflammatory events leading to neuronal dysfunction and degeneration. Preliminary imaging findings suggest that BBB dysfunction could serve as an early diagnostic and prognostic biomarker for a number of neurological diseases. This review aims to provide clinicians with an overview of the emerging field of BBB imaging in humans by answering three key questions: (1. Disease) In which diseases could BBB imaging be useful? (2. Device) What are currently available imaging methods for evaluating BBB integrity? And (3. Distribution) what is the potential of BBB imaging in different environments, particularly in resource limited settings? We conclude that further advances are needed, such as the validation, standardization and implementation of readily available, low-cost and non-contrast BBB imaging techniques, for BBB imaging to be a useful clinical biomarker in both resource-limited and well-resourced settings.
Collapse
Affiliation(s)
- Paulien Moyaert
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
- Lawson Health Research Institute, London, ON, Canada
- Department of Neurology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- *Correspondence: Paulien Moyaert,
| | - Beatriz E. Padrela
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Catherine A. Morgan
- School of Psychology and Centre for Brain Research, The University of Auckland, Auckland, New Zealand
- Centre for Advanced MRI, Auckland UniServices Limited, Auckland, New Zealand
| | - Jan Petr
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Jan Versijpt
- Department of Neurology, Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, United Kingdom
| | | | - Xingfeng Shao
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Olujide Oyeniran
- Lawson Health Research Institute, London, ON, Canada
- Department of Medical Biophysics, Western University, London, ON, Canada
| | - Tabitha Manson
- Centre for Advanced MRI, Auckland UniServices Limited, Auckland, New Zealand
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Danny J. J. Wang
- Laboratory of FMRI Technology (LOFT), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Matthias Günther
- Fraunhofer Institute for Digital Medicine, University of Bremen, Bremen, Germany
| | - Eric Achten
- Department of Medical Imaging, Ghent University Hospital, Ghent, Belgium
| | - Henk J. M. M. Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Amsterdam Neuroscience, Brain Imaging, Amsterdam, Netherlands
| | - Udunna C. Anazodo
- Lawson Health Research Institute, London, ON, Canada
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
26
|
Lai QL, Cai MT, Zheng Y, Fang GL, Du BQ, Shen CH, Wang JJ, Weng QJ, Zhang YX. Evaluation of CSF albumin quotient in neuronal surface antibody-associated autoimmune encephalitis. Fluids Barriers CNS 2022; 19:93. [DOI: 10.1186/s12987-022-00392-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
Abstract
Background and Objectives
Disruption of brain barriers is considered to be involved in the pathogenesis of neuronal surface antibody-associated autoimmune encephalitis (NSAE), but few studies have focused on their relationship. We aimed to explore the association between the integrity of brain barriers and clinical and paraclinical characteristics in patients with NSAE.
Methods
This retrospective study consecutively recruited patients with NSAE. The cerebrospinal fluid (CSF) / serum albumin quotient (Qalb) was used to evaluate the function of brain barriers. The data on demographic information, clinical manifestations, magnetic resonance imaging (MRI), CSF findings and prognosis were collected and analyzed.
Results
Of the 93 patients included, 33 (35.5%) patients were assigned to the elevated Qalb group and 60 (64.5%) patients to the normal Qalb group. Males and prodromal symptoms were more common in elevated Qalb group (both P < 0.05). The CSF white blood cell, protein, immunoglobulin G and albumin were significantly higher in elevated Qalb group (all P < 0.05). Patients with elevated Qalb were more likely to have brain lesions on MRI (60.6% versus 33.3%, P = 0.011). The modified Rankin Scale (mRS) scores at discharge and at last follow-up were significantly higher in patients with elevated Qalb than those with normal Qalb (both P < 0.05). After univariate and multivariate analyses, Qalb elevation (adjusted odds ratio = 3.96, 95% confidence interval = 1.15–13.59, P = 0.029) was demonstrated as the only independent risk factor for a poor prognosis.
Discussion
Males, prodromal symptoms, brain lesions on MRI, CSF pleocytosis, and elevated CSF protein were more common in NSAE patients with increased Qalb. Qalb elevation was an independent prognostic indicator for a poor prognosis in NSAE.
Collapse
|
27
|
Xu W, Bai Q, Dong Q, Guo M, Cui M. Blood–Brain Barrier Dysfunction and the Potential Mechanisms in Chronic Cerebral Hypoperfusion Induced Cognitive Impairment. Front Cell Neurosci 2022; 16:870674. [PMID: 35783093 PMCID: PMC9243657 DOI: 10.3389/fncel.2022.870674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Chronic cerebral hypoperfusion (CCH) is a major cause of vascular cognitive impairment and dementia (VCID). Although the underlying mechanisms have not been fully elucidated, the emerging data suggest that blood–brain barrier (BBB) dysfunction is one of the pivotal pathological changes in CCH. BBB dysfunction appears early in CCH, contributing to the deterioration of white matter and the development of cognitive impairment. In this review, we summarize the latest experimental and clinical evidence implicating BBB disruption as a major cause of VCID. We discuss the mechanisms of BBB dysfunction in CCH, focusing on the cell interactions within the BBB, as well as the potential role of APOE genotype. In summary, we provide novel insights into the pathophysiological mechanisms underlying BBB dysfunction and the potential clinical benefits of therapeutic interventions targeting BBB in CCH.
Collapse
Affiliation(s)
- WenQing Xu
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qingke Bai
- Department of Neurology, Pudong People’s Hospital, Shanghai, China
| | - Qiang Dong
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- Min Guo,
| | - Mei Cui
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
- *Correspondence: Mei Cui,
| |
Collapse
|
28
|
van Dinther M, Voorter PH, Jansen JF, Jones EA, van Oostenbrugge RJ, Staals J, Backes WH. Assessment of microvascular rarefaction in human brain disorders using physiological magnetic resonance imaging. J Cereb Blood Flow Metab 2022; 42:718-737. [PMID: 35078344 PMCID: PMC9014687 DOI: 10.1177/0271678x221076557] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cerebral microvascular rarefaction, the reduction in number of functional or structural small blood vessels in the brain, is thought to play an important role in the early stages of microvascular related brain disorders. A better understanding of its underlying pathophysiological mechanisms, and methods to measure microvascular density in the human brain are needed to develop biomarkers for early diagnosis and to identify targets for disease modifying treatments. Therefore, we provide an overview of the assumed main pathophysiological processes underlying cerebral microvascular rarefaction and the evidence for rarefaction in several microvascular related brain disorders. A number of advanced physiological MRI techniques can be used to measure the pathological alterations associated with microvascular rarefaction. Although more research is needed to explore and validate these MRI techniques in microvascular rarefaction in brain disorders, they provide a set of promising future tools to assess various features relevant for rarefaction, such as cerebral blood flow and volume, vessel density and radius and blood-brain barrier leakage.
Collapse
Affiliation(s)
- Maud van Dinther
- Department of Neurology, Maastricht University Medical Center, The Netherlands.,CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands
| | - Paulien Hm Voorter
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, The Netherlands.,MHeNs - School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Jacobus Fa Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, The Netherlands.,MHeNs - School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | | | - Robert J van Oostenbrugge
- Department of Neurology, Maastricht University Medical Center, The Netherlands.,CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands.,MHeNs - School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| | - Julie Staals
- Department of Neurology, Maastricht University Medical Center, The Netherlands.,CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands
| | - Walter H Backes
- CARIM - School for Cardiovascular Diseases, Maastricht University, The Netherlands.,Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, The Netherlands.,MHeNs - School for Mental Health and Neuroscience, Maastricht University, The Netherlands
| |
Collapse
|
29
|
Hugon G, Goutal S, Dauba A, Breuil L, Larrat B, Winkeler A, Novell A, Tournier N. [ 18F]2-Fluoro-2-deoxy-sorbitol PET Imaging for Quantitative Monitoring of Enhanced Blood-Brain Barrier Permeability Induced by Focused Ultrasound. Pharmaceutics 2021; 13:pharmaceutics13111752. [PMID: 34834167 PMCID: PMC8621256 DOI: 10.3390/pharmaceutics13111752] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/10/2021] [Accepted: 10/14/2021] [Indexed: 01/18/2023] Open
Abstract
Focused ultrasound in combination with microbubbles (FUS) provides an effective means to locally enhance the delivery of therapeutics to the brain. Translational and quantitative imaging techniques are needed to noninvasively monitor and optimize the impact of FUS on blood-brain barrier (BBB) permeability in vivo. Positron-emission tomography (PET) imaging using [18F]2-fluoro-2-deoxy-sorbitol ([18F]FDS) was evaluated as a small-molecule (paracellular) marker of blood-brain barrier (BBB) integrity. [18F]FDS was straightforwardly produced from chemical reduction of commercial [18F]2-deoxy-2-fluoro-D-glucose. [18F]FDS and the invasive BBB integrity marker Evan’s blue (EB) were i.v. injected in mice after an optimized FUS protocol designed to generate controlled hemispheric BBB disruption. Quantitative determination of the impact of FUS on the BBB permeability was determined using kinetic modeling. A 2.2 ± 0.5-fold higher PET signal (n = 5; p < 0.01) was obtained in the sonicated hemisphere and colocalized with EB staining observed post mortem. FUS significantly increased the blood-to-brain distribution of [18F]FDS by 2.4 ± 0.8-fold (VT; p < 0.01). Low variability (=10.1%) of VT values in the sonicated hemisphere suggests reproducibility of the estimation of BBB permeability and FUS method. [18F]FDS PET provides a readily available, sensitive and reproducible marker of BBB permeability to noninvasively monitor the extent of BBB disruption induced by FUS in vivo.
Collapse
Affiliation(s)
- Gaëlle Hugon
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Sébastien Goutal
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Ambre Dauba
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Louise Breuil
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Benoit Larrat
- CNRS, CEA, DRF/JOLIOT/NEUROSPIN/BAOBAB, Université Paris-Saclay, 91191 Gif-sur-Yvette, France;
| | - Alexandra Winkeler
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Anthony Novell
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
| | - Nicolas Tournier
- CEA, CNRS, Inserm, BioMaps, Université Paris-Saclay, 91401 Orsay, France; (G.H.); (S.G.); (A.D.); (L.B.); (A.W.); (A.N.)
- Correspondence:
| |
Collapse
|
30
|
Empana JP, Boutouyrie P, Lemogne C, Jouven X, van Sloten TT. Microvascular Contribution to Late-Onset Depression: Mechanisms, Current Evidence, Association With Other Brain Diseases, and Therapeutic Perspectives. Biol Psychiatry 2021; 90:214-225. [PMID: 34325805 DOI: 10.1016/j.biopsych.2021.04.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022]
Abstract
Depression is common in older individuals and is associated with high disability and mortality. A major problem is treatment resistance: >50% of older patients do not respond to current antidepressants. Therefore, new effective interventions for prevention and treatment of depression in older individuals need to be developed, which requires a better understanding of the mechanisms underlying depression. The pathophysiology of depression is multifactorial and complex. Microvascular dysfunction may be an early and targetable mechanism in the development of depression, notably depression that initiates in late life (late-onset depression). Late-onset depression commonly co-occurs with other diseases or syndromes that may share a microvascular origin, including apathy, cognitive impairment, dementia, and stroke. Together, these disabilities may all be part of one large phenotype resulting from global cerebral microvascular dysfunction. In this review, we discuss the pathophysiology of microvascular dysfunction-related late-onset depression, summarize recent epidemiological evidence on the association between cerebral microvascular dysfunction and depression, and indicate potential drivers of cerebral microvascular dysfunction. We also propose the hypothesis that depression may be a manifestation of a larger phenotype of cerebral microvascular dysfunction, highlight potential therapeutic targets and interventions, and give directions for future research.
Collapse
Affiliation(s)
- Jean-Philippe Empana
- Université de Paris, INSERM, U970, Paris Cardiovascular Research Center, Paris, France
| | - Pierre Boutouyrie
- Université de Paris, INSERM, U970, Paris Cardiovascular Research Center, Paris, France
| | - Cédric Lemogne
- Université de Paris, AP-HP, Hôpital Hôtel-Dieu, DMU Psychiatrie et Addictologie, Service de Psychiatrie de l'adulte, INSERM, Institut de Psychiatrie et Neurosciences de Paris, UMR_S1266, Paris, France
| | - Xavier Jouven
- Université de Paris, INSERM, U970, Paris Cardiovascular Research Center, Paris, France
| | - Thomas T van Sloten
- Université de Paris, INSERM, U970, Paris Cardiovascular Research Center, Paris, France; School for Cardiovascular Diseases Maastricht and Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, the Netherlands.
| |
Collapse
|
31
|
Hansson O. Biomarkers for neurodegenerative diseases. Nat Med 2021; 27:954-963. [PMID: 34083813 DOI: 10.1038/s41591-021-01382-x] [Citation(s) in RCA: 532] [Impact Index Per Article: 133.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022]
Abstract
Biomarkers for neurodegenerative diseases are needed to improve the diagnostic workup in the clinic but also to facilitate the development and monitoring of effective disease-modifying therapies. Positron emission tomography methods detecting amyloid-β and tau pathology in Alzheimer's disease have been increasingly used to improve the design of clinical trials and observational studies. In recent years, easily accessible and cost-effective blood-based biomarkers detecting the same Alzheimer's disease pathologies have been developed, which might revolutionize the diagnostic workup of Alzheimer's disease globally. Relevant biomarkers for α-synuclein pathology in Parkinson's disease are also emerging, as well as blood-based markers of general neurodegeneration and glial activation. This review presents an overview of the latest advances in the field of biomarkers for neurodegenerative diseases. Future directions are discussed regarding implementation of novel biomarkers in clinical practice and trials.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|