1
|
Kadam R, Gupta M, Lazarov O, Prabhakar BS. Brain-immune interactions: implication for cognitive impairments in Alzheimer's disease and autoimmune disorders. J Leukoc Biol 2024; 116:1269-1290. [PMID: 38869088 DOI: 10.1093/jleuko/qiae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Progressive memory loss and cognitive dysfunction, encompassing deficits in learning, memory, problem solving, spatial reasoning, and verbal expression, are characteristics of Alzheimer's disease and related dementia. A wealth of studies has described multiple roles of the immune system in the development or exacerbation of dementia. Individuals with autoimmune disorders can also develop cognitive dysfunction, a phenomenon termed "autoimmune dementia." Together, these findings underscore the pivotal role of the neuroimmune axis in both Alzheimer's disease and related dementia and autoimmune dementia. The dynamic interplay between adaptive and innate immunity, both in and outside the brain, significantly affects the etiology and progression of these conditions. Multidisciplinary research shows that cognitive dysfunction arises from a bidirectional relationship between the nervous and immune systems, though the specific mechanisms that drive cognitive impairments are not fully understood. Intriguingly, this reciprocal regulation occurs at multiple levels, where neuronal signals can modulate immune responses, and immune system-related processes can influence neuronal viability and function. In this review, we consider the implications of autoimmune responses in various autoimmune disorders and Alzheimer's disease and explore their effects on brain function. We also discuss the diverse cellular and molecular crosstalk between the brain and the immune system, as they may shed light on potential triggers of peripheral inflammation, their effect on the integrity of the blood-brain barrier, and brain function. Additionally, we assess challenges and possibilities associated with developing immune-based therapies for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Rashmi Kadam
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| |
Collapse
|
2
|
Rather MA, Khan A, Jahan S, Siddiqui AJ, Wang L. Influence of Tau on Neurotoxicity and Cerebral Vasculature Impairment Associated with Alzheimer's Disease. Neuroscience 2024; 552:1-13. [PMID: 38871021 DOI: 10.1016/j.neuroscience.2024.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Alzheimer's disease is a fatal chronic neurodegenerative condition marked by a gradual decline in cognitive abilities and impaired vascular function within the central nervous system. This affliction initiates its insidious progression with the accumulation of two aberrant protein entities including Aβ plaques and neurofibrillary tangles. These chronic elements target distinct brain regions, steadily erasing the functionality of the hippocampus and triggering the erosion of memory and neuronal integrity. Several assumptions are anticipated for AD as genetic alterations, the occurrence of Aβ plaques, altered processing of amyloid precursor protein, mitochondrial damage, and discrepancy of neurotropic factors. In addition to Aβ oligomers, the deposition of tau hyper-phosphorylates also plays an indispensable part in AD etiology. The brain comprises a complex network of capillaries that is crucial for maintaining proper function. Tau is expressed in cerebral blood vessels, where it helps to regulate blood flow and sustain the blood-brain barrier's integrity. In AD, tau pathology can disrupt cerebral blood supply and deteriorate the BBB, leading to neuronal neurodegeneration. Neuroinflammation, deficits in the microvasculature and endothelial functions, and Aβ deposition are characteristically detected in the initial phases of AD. These variations trigger neuronal malfunction and cognitive impairment. Intracellular tau accumulation in microglia and astrocytes triggers deleterious effects on the integrity of endothelium and cerebral blood supply resulting in further advancement of the ailment and cerebral instability. In this review, we will discuss the impact of tau on neurovascular impairment, mitochondrial dysfunction, oxidative stress, and the role of hyperphosphorylated tau in neuron excitotoxicity and inflammation.
Collapse
Affiliation(s)
- Mashoque Ahmad Rather
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States.
| | - Andleeb Khan
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, 226026, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail City, Saudi Arabia
| | - Lianchun Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States
| |
Collapse
|
3
|
Chen T, Dai Y, Hu C, Lin Z, Wang S, Yang J, Zeng L, Li S, Li W. Cellular and molecular mechanisms of the blood-brain barrier dysfunction in neurodegenerative diseases. Fluids Barriers CNS 2024; 21:60. [PMID: 39030617 PMCID: PMC11264766 DOI: 10.1186/s12987-024-00557-1] [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: 03/17/2024] [Accepted: 06/20/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Maintaining the structural and functional integrity of the blood-brain barrier (BBB) is vital for neuronal equilibrium and optimal brain function. Disruptions to BBB performance are implicated in the pathology of neurodegenerative diseases. MAIN BODY Early indicators of multiple neurodegenerative disorders in humans and animal models include impaired BBB stability, regional cerebral blood flow shortfalls, and vascular inflammation associated with BBB dysfunction. Understanding the cellular and molecular mechanisms of BBB dysfunction in brain disorders is crucial for elucidating the sustenance of neural computations under pathological conditions and for developing treatments for these diseases. This paper initially explores the cellular and molecular definition of the BBB, along with the signaling pathways regulating BBB stability, cerebral blood flow, and vascular inflammation. Subsequently, we review current insights into BBB dynamics in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. The paper concludes by proposing a unified mechanism whereby BBB dysfunction contributes to neurodegenerative disorders, highlights potential BBB-focused therapeutic strategies and targets, and outlines lessons learned and future research directions. CONCLUSIONS BBB breakdown significantly impacts the development and progression of neurodegenerative diseases, and unraveling the cellular and molecular mechanisms underlying BBB dysfunction is vital to elucidate how neural computations are sustained under pathological conditions and to devise therapeutic approaches.
Collapse
Affiliation(s)
- Tongli Chen
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Yan Dai
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Chenghao Hu
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Zihao Lin
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Shengzhe Wang
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jing Yang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Linghui Zeng
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Shanshan Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| | - Weiyun Li
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China.
- Institute of Brain and Cognitive Science, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
4
|
Bae J, Qayyum S, Zhang J, Das A, Reyes I, Aronowitz E, Stavarache MA, Kaplitt MG, Masurkar A, Kim SG. Feasibility of measuring blood-brain barrier permeability using ultra-short echo time radial magnetic resonance imaging. J Neuroimaging 2024; 34:320-328. [PMID: 38616297 PMCID: PMC11090723 DOI: 10.1111/jon.13199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The purpose of this study is to evaluate the feasibility of using 3-dimensional (3D) ultra-short echo time (UTE) radial imaging method for measurement of the permeability of the blood-brain barrier (BBB) to gadolinium-based contrast agent. In this study, we propose to use the golden-angle radial sparse parallel (GRASP) method with 3D center-out trajectories for UTE, hence named as 3D UTE-GRASP. We first examined the feasibility of using 3D UTE-GRASP dynamic contrast-enhanced (DCE)-magnetic resonance imaging (MRI) for differentiating subtle BBB disruptions induced by focused ultrasound (FUS). Then, we examined the BBB permeability changes in Alzheimer's disease (AD) pathology using Alzheimer's disease transgenic mice (5xFAD) at different ages. METHODS For FUS experiments, we used four Sprague Dawley rats at similar ages where we compared BBB permeability of each rat receiving the FUS sonication with different acoustic power (0.4-1.0 MPa). For AD transgenic mice experiments, we included three 5xFAD mice (6, 12, and 16 months old) and three wild-type mice (4, 8, and 12 months old). RESULTS The result from FUS experiments showed a progressive increase in BBB permeability with increase of acoustic power (p < .05), demonstrating the sensitivity of DCE-MRI method for detecting subtle changes in BBB disruption. Our AD transgenic mice experiments suggest an early BBB disruption in 5xFAD mice, which is further impaired with aging. CONCLUSION The results in this study substantiate the feasibility of using the proposed 3D UTE-GRASP method for detecting subtle BBB permeability changes expected in neurodegenerative diseases, such as AD.
Collapse
Affiliation(s)
- Jonghyun Bae
- Vilcek Institute of Graduate Biomedical Science, New York University School of Medicine
- Center for Biomedical Imaging, Radiology, New York University School of Medicine
- Center for Advanced Imaging Innovation and Research, Radiology, New York University School of Medicine
- Department of Radiology, Weill Cornell Medical College
| | - Sawwal Qayyum
- Department of Radiology, Weill Cornell Medical College
| | - Jin Zhang
- Department of Radiology, Weill Cornell Medical College
| | - Ayesha Das
- Department of Radiology, Weill Cornell Medical College
| | - Isabel Reyes
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine
- Department of Neuroscience & Physiology, New York University School of Medicine
- Neuroscience Institute, New York University School of Medicine
| | | | | | | | - Arjun Masurkar
- Center for Cognitive Neurology, Department of Neurology, New York University School of Medicine
- Department of Neuroscience & Physiology, New York University School of Medicine
- Neuroscience Institute, New York University School of Medicine
| | | |
Collapse
|
5
|
Wheeler KV, Irimia A, Braskie MN. Using Neuroimaging to Study Cerebral Amyloid Angiopathy and Its Relationship to Alzheimer's Disease. J Alzheimers Dis 2024; 97:1479-1502. [PMID: 38306032 DOI: 10.3233/jad-230553] [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] [Indexed: 02/03/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by amyloid-β aggregation in the media and adventitia of the leptomeningeal and cortical blood vessels. CAA is one of the strongest vascular contributors to Alzheimer's disease (AD). It frequently co-occurs in AD patients, but the relationship between CAA and AD is incompletely understood. CAA may drive AD risk through damage to the neurovascular unit and accelerate parenchymal amyloid and tau deposition. Conversely, early AD may also drive CAA through cerebrovascular remodeling that impairs blood vessels from clearing amyloid-β. Sole reliance on autopsy examination to study CAA limits researchers' ability to investigate CAA's natural disease course and the effect of CAA on cognitive decline. Neuroimaging allows for in vivo assessment of brain function and structure and can be leveraged to investigate CAA staging and explore its associations with AD. In this review, we will discuss neuroimaging modalities that can be used to investigate markers associated with CAA that may impact AD vulnerability including hemorrhages and microbleeds, blood-brain barrier permeability disruption, reduced cerebral blood flow, amyloid and tau accumulation, white matter tract disruption, reduced cerebrovascular reactivity, and lowered brain glucose metabolism. We present possible areas for research inquiry to advance biomarker discovery and improve diagnostics.
Collapse
Affiliation(s)
- Koral V Wheeler
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Department of Biomedical Engineering, Corwin D. Denney Research Center, Viterbi School of Engineering, University of Southern California, Los Angeles, CA, USA
| | - Meredith N Braskie
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Marina Del Rey, CA, USA
| |
Collapse
|
6
|
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: 1.0] [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
|
7
|
Lee RL, Funk KE. Imaging blood–brain barrier disruption in neuroinflammation and Alzheimer’s disease. Front Aging Neurosci 2023; 15:1144036. [PMID: 37009464 PMCID: PMC10063921 DOI: 10.3389/fnagi.2023.1144036] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/28/2023] [Indexed: 03/19/2023] Open
Abstract
The blood–brain barrier (BBB) is the neurovascular structure that regulates the passage of cells and molecules to and from the central nervous system (CNS). Alzheimer’s disease (AD) is a neurodegenerative disorder that is associated with gradual breakdown of the BBB, permitting entry of plasma-derived neurotoxins, inflammatory cells, and microbial pathogens into the CNS. BBB permeability can be visualized directly in AD patients using imaging technologies including dynamic contrast-enhanced and arterial spin labeling magnetic resonance imaging, and recent studies employing these techniques have shown that subtle changes in BBB stability occur prior to deposition of the pathological hallmarks of AD, senile plaques, and neurofibrillary tangles. These studies suggest that BBB disruption may be useful as an early diagnostic marker; however, AD is also accompanied by neuroinflammation, which can complicate these analyses. This review will outline the structural and functional changes to the BBB that occur during AD pathogenesis and highlight current imaging technologies that can detect these subtle changes. Advancing these technologies will improve both the diagnosis and treatment of AD and other neurodegenerative diseases.
Collapse
|
8
|
Uchida Y, Kan H, Sakurai K, Oishi K, Matsukawa N. Contributions of blood-brain barrier imaging to neurovascular unit pathophysiology of Alzheimer's disease and related dementias. Front Aging Neurosci 2023; 15:1111448. [PMID: 36861122 PMCID: PMC9969807 DOI: 10.3389/fnagi.2023.1111448] [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: 11/29/2022] [Accepted: 01/26/2023] [Indexed: 02/11/2023] Open
Abstract
The blood-brain barrier (BBB) plays important roles in the maintenance of brain homeostasis. Its main role includes three kinds of functions: (1) to protect the central nervous system from blood-borne toxins and pathogens; (2) to regulate the exchange of substances between the brain parenchyma and capillaries; and (3) to clear metabolic waste and other neurotoxic compounds from the central nervous system into meningeal lymphatics and systemic circulation. Physiologically, the BBB belongs to the glymphatic system and the intramural periarterial drainage pathway, both of which are involved in clearing interstitial solutes such as β-amyloid proteins. Thus, the BBB is believed to contribute to preventing the onset and progression for Alzheimer's disease. Measurements of BBB function are essential toward a better understanding of Alzheimer's pathophysiology to establish novel imaging biomarkers and open new avenues of interventions for Alzheimer's disease and related dementias. The visualization techniques for capillary, cerebrospinal, and interstitial fluid dynamics around the neurovascular unit in living human brains have been enthusiastically developed. The purpose of this review is to summarize recent BBB imaging developments using advanced magnetic resonance imaging technologies in relation to Alzheimer's disease and related dementias. First, we give an overview of the relationship between Alzheimer's pathophysiology and BBB dysfunction. Second, we provide a brief description about the principles of non-contrast agent-based and contrast agent-based BBB imaging methodologies. Third, we summarize previous studies that have reported the findings of each BBB imaging method in individuals with the Alzheimer's disease continuum. Fourth, we introduce a wide range of Alzheimer's pathophysiology in relation to BBB imaging technologies to advance our understanding of the fluid dynamics around the BBB in both clinical and preclinical settings. Finally, we discuss the challenges of BBB imaging techniques and suggest future directions toward clinically useful imaging biomarkers for Alzheimer's disease and related dementias.
Collapse
Affiliation(s)
- Yuto Uchida
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| | - Hirohito Kan
- Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keita Sakurai
- Department of Radiology, National Center for Geriatrics and Gerontology, Ōbu, Aichi, Japan
| | - Kenichi Oishi
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Noriyuki Matsukawa
- Department of Neurology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan,*Correspondence: Yuto Uchida, ; Noriyuki Matsukawa,
| |
Collapse
|
9
|
Abstract
Alzheimer's disease (AD) is a genetically complex and heterogeneous disorder with multifaceted neuropathological features, including β-amyloid plaques, neurofibrillary tangles, and neuroinflammation. Over the past decade, emerging evidence has implicated both beneficial and pathological roles for innate immune genes and immune cells, including peripheral immune cells such as T cells, which can infiltrate the brain and either ameliorate or exacerbate AD neuropathogenesis. These findings support a neuroimmune axis of AD, in which the interplay of adaptive and innate immune systems inside and outside the brain critically impacts the etiology and pathogenesis of AD. In this review, we discuss the complexities of AD neuropathology at the levels of genetics and cellular physiology, highlighting immune signaling pathways and genes associated with AD risk and interactions among both innate and adaptive immune cells in the AD brain. We emphasize the role of peripheral immune cells in AD and the mechanisms by which immune cells, such as T cells and monocytes, influence AD neuropathology, including microglial clearance of amyloid-β peptide, the key component of β-amyloid plaque cores, pro-inflammatory and cytotoxic activity of microglia, astrogliosis, and their interactions with the brain vasculature. Finally, we review the challenges and outlook for establishing immune-based therapies for treating and preventing AD.
Collapse
|
10
|
Kwon YS, Ko JS, Oh SY, Han YT, Jo SA. Oleracone F Alleviates Cognitive Impairment and Neuropathology in APPswe/PSEN1dE9 Mice by Reducing the Expression of Vascular Cell Adhesion Molecule and Leukocyte Adhesion to Brain Vascular Endothelial Cells. Int J Mol Sci 2023; 24:ijms24032056. [PMID: 36768379 PMCID: PMC9916962 DOI: 10.3390/ijms24032056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease and the blood-brain barrier dysfunction has been suggested as a key pathological feature of the disease. Our research group successfully established a synthetic protocol for oleracones, a novel series of flavonoids isolated from the plant extract of Portulaca oleracea L. (PO). PO extract was reported to have anti-inflammatory and antioxidant effects, enhancing cognitive function. Thus, we investigated the effects and mechanism of oleracones on cognition using AD model transgenic mice (Tg; APPswe/PSEN1dE9). Oleracone F treatment significantly improved memory dysfunction in Tg mice. Oleracone F decreased the number, burden, and immunoreactivity of amyloid plaques and amyloid precursor protein (APP) protein levels in the brains of Tg mice compared to wild-type mice. Oleracone F also alleviated inflammation observed in Tg mice brains. In vitro studies in human microvascular endothelial cells (HBMVECs) demonstrated that oleracones D, E, and F blocked the elevations in VCAM-1 protein induced by tumor necrosis factor-α (TNF-α), hindering leukocyte adhesion to HBMVECs. Taken together, our results suggest that oleracones ameliorated cognitive impairment by blocking TNF-α-induced increases in VCAM-1, thereby reducing leukocyte infiltration to the brain and modulating brain inflammation.
Collapse
Affiliation(s)
- Young-Sun Kwon
- Department of Nanobiomedical Science & BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Jin-Sung Ko
- Department of Nanobiomedical Science & BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Se-Young Oh
- Department of Convergence Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul 07985, Republic of Korea
| | - Young Taek Han
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea
| | - Sangmee Ahn Jo
- Department of Nanobiomedical Science & BK21 Four NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea
- Correspondence: ; Tel.: +82-41-550-1433
| |
Collapse
|
11
|
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: 15] [Impact Index Per Article: 15.0] [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
|
12
|
Kang KM, Byun MS, Yi D, Lee KH, Kim M, Ahn H, Jung G, Lee J, Kim YK, Lee Y, Sohn C, Lee DY. Enlarged perivascular spaces are associated with decreased brain tau deposition. CNS Neurosci Ther 2022; 29:577-586. [PMID: 36468423 PMCID: PMC9873511 DOI: 10.1111/cns.14040] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/11/2022] [Accepted: 11/14/2022] [Indexed: 12/10/2022] Open
Abstract
AIMS The aim of this study was to investigate the associations of enlarged perivascular spaces (EPVS) in the basal ganglia (BG) and centrum semiovale (CSO) with beta-amyloid (Aβ) and tau deposition in older adults with a diverse cognitive spectrum. METHODS A total of 163 (68 cognitively normal and 95 cognitively impaired) older participants underwent [11 C] Pittsburgh compound B and [18 F] AV-1451 PET, and MRI. EPVS in the BG and CSO and other small vessel disease markers, such as white matter hyperintensities, lacunes, and deep and lobar microbleeds, were assessed. RESULTS Increased EPVS in the BG showed a significant association with lower cerebral tau deposition, even after controlling for other small vessel disease markers. Further exploratory analyses showed that this association was significant in cognitively impaired, Aβ-positive, or APOE4-positive individuals, but not significant in the cognitively normal, Aβ-negative, or APOE4-negative participants. In contrast to EPVS in the BG, EPVS in the CSO did not have any relationship with cerebral tau deposition. In addition, none of the two types of EPVS were associated with cerebral Aβ deposition. CONCLUSION Brain tau deposition appears to be reduced with increased EPVS in the BG, especially in individuals with cognitive impairment, pathological amyloid burden, or genetic Alzheimer's disease risk.
Collapse
Affiliation(s)
- Koung Mi Kang
- Department of RadiologySeoul National University HospitalSeoulKorea,Department of RadiologySeoul National University College of MedicineSeoulKorea
| | - Min Soo Byun
- Department of NeuropsychiatrySeoul National University College of MedicineSeoulKorea,Department of NeuropsychiatrySeoul National University HospitalSeoulKorea
| | - Dahyun Yi
- Biomedical Research Institute, Seoul National University HospitalSeoulKorea
| | - Kyung Hoon Lee
- Department of RadiologySeoul National University HospitalSeoulKorea
| | - Min Jung Kim
- Department of NeuropsychiatrySeoul National University HospitalSeoulKorea
| | - Hyejin Ahn
- Department of NeuropsychiatrySeoul National University HospitalSeoulKorea
| | - Gijung Jung
- Department of NeuropsychiatrySeoul National University HospitalSeoulKorea
| | - Jun‐Young Lee
- Department of NeuropsychiatrySeoul National University College of MedicineSeoulKorea,Department of NeuropsychiatrySMG‐SNU Boramae Medical CenterSeoulKorea
| | - Yu Kyeong Kim
- Department of Nuclear MedicineSMG‐SNU Boramae Medical CenterSeoulKorea
| | - Yun‐Sang Lee
- Department of Nuclear MedicineSeoul National University College of MedicineSeoulKorea
| | - Chul‐Ho Sohn
- Department of RadiologySeoul National University HospitalSeoulKorea,Department of RadiologySeoul National University College of MedicineSeoulKorea
| | - Dong Young Lee
- Department of NeuropsychiatrySeoul National University College of MedicineSeoulKorea,Department of NeuropsychiatrySeoul National University HospitalSeoulKorea,Institute of Human Behavioral MedicineMedical Research Center Seoul National UniversitySeoulKorea
| | | |
Collapse
|
13
|
Alruwais NM, Rusted JM, Tabet N, Dowell NG. Evidence of emerging BBB changes in mid-age apolipoprotein E epsilon-4 carriers. Brain Behav 2022; 12:e2806. [PMID: 36408825 PMCID: PMC9759141 DOI: 10.1002/brb3.2806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/23/2022] [Accepted: 10/08/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Studies have recognized that the loss of the blood-brain barrier (BBB) integrity is a major structural biomarker where neurodegenerative disease potentially begins. Using a combination of high-quality neuroimaging techniques, we investigated potential subtle differences in BBB permeability in mid-age healthy people, comparing carriers of the apolipoprotein E epsilon-4 (APOEε4) genotype, the biggest risk factor for late onset, non-familial AD (LOAD) with APOEε3 carriers, the population norm. METHODS Forty-one cognitively healthy mid-age participants (42-59) were genotyped and pseudo-randomly selected to participate in the study by a third party. Blind to genotype, all participants had a structural brain scan acquisition including gadolinium-based dynamic contrast-enhanced magnetic resonance imaging acquired using a T1-weighted 3D vibe sequence. A B1 map and T1 map were acquired as part of the multi-parametric mapping acquisition. RESULTS Non-significant, but subtle differences in blood-brain barrier permeability were identified between healthy mid-age APOEε4 and APOEε3 carriers, matched on age, education, and gender. DISCUSSION This study demonstrated a tendency toward BBB permeability in APOEε4 participants emerging from mid-age, with quantitative differences observable on a number of the measures. While the differences did not reach a statistical significance, the results from this study hint at early changes in ε4 carrier BBB that may help identify at-risk populations and facilitate the development of early interventions to change the trajectory of decline.
Collapse
Affiliation(s)
- Nourah M Alruwais
- Health science department, College of Applied Studies and Community Services, King Saud University, Riyadh, Saudi Arabia.,School of Psychology, University of Sussex, Brighton, UK
| | | | - Naji Tabet
- Brighton and Sussex Medical School (BSMS), Brighton, UK
| | | |
Collapse
|
14
|
KALA D, ŠULC V, OLŠEROVÁ A, SVOBODA J, PRYSIAZHNIUK Y, POŠUSTA A, KYNČL M, ŠANDA J, TOMEK A, OTÁHAL J. Evaluation of blood-brain barrier integrity by the analysis of dynamic contrast-enhanced MRI - a comparison of quantitative and semi-quantitative methods. Physiol Res 2022; 71:S259-S275. [PMID: 36647914 PMCID: PMC9906669 DOI: 10.33549/physiolres.934998] [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] [Indexed: 01/26/2023] Open
Abstract
Disruption of the blood-brain barrier (BBB) is a key feature of various brain disorders. To assess its integrity a parametrization of dynamic magnetic resonance imaging (DCE MRI) with a contrast agent (CA) is broadly used. Parametrization can be done quantitatively or semi-quantitatively. Quantitative methods directly describe BBB permeability but exhibit several drawbacks such as high computation demands, reproducibility issues, or low robustness. Semi-quantitative methods are fast to compute, simply mathematically described, and robust, however, they do not describe the status of BBB directly but only as a variation of CA concentration in measured tissue. Our goal was to elucidate differences between five semi-quantitative parameters: maximal intensity (Imax), normalized permeability index (NPI), and difference in DCE values between three timepoints: baseline, 5 min, and 15 min (delta5-0, delta15-0, delta15-5) and two quantitative parameters: transfer constant (Ktrans) and an extravascular fraction (Ve). For the purpose of comparison, we analyzed DCE data of four patients 12-15 days after the stroke with visible CA enhancement. Calculated parameters showed abnormalities spatially corresponding with the ischemic lesion, however, findings in individual parameters morphometrically differed. Ktrans and Ve were highly correlated. Delta5-0 and delta15-0 were prominent in regions with rapid CA enhancement and highly correlated with Ktrans. Abnormalities in delta15-5 and NPI were more homogenous with less variable values, smoother borders, and less detail than Ktrans. Moreover, only delta15-5 and NPI were able to distinguish vessels from extravascular space. Our comparison provides important knowledge for understanding and interpreting parameters derived from DCE MRI by both quantitative and semi-quantitative methods.
Collapse
Affiliation(s)
- David KALA
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic,Faculty of Electrical Engineering, Czech Technical University in Prague, Czech Republic
| | - Vlastimil ŠULC
- Department of Neurology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Anna OLŠEROVÁ
- Department of Neurology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan SVOBODA
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Yeva PRYSIAZHNIUK
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Antonín POŠUSTA
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Martin KYNČL
- Department of Radiology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jan ŠANDA
- Department of Radiology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Aleš TOMEK
- Department of Neurology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Jakub OTÁHAL
- Laboratory of Developmental Epileptology, Institute of Physiology of the Czech Academy of Sciences, Prague, Czech Republic,Department of Pathophysiology, Second Faculty of Medicine, Charles University, Czech Republic
| |
Collapse
|
15
|
Ware JB, Sinha S, Morrison J, Walter AE, Gugger JJ, Schneider ALC, Dabrowski C, Zamore H, Wesley L, Magdamo B, Petrov D, Kim JJ, Diaz-Arrastia R, Sandsmark DK. Dynamic contrast enhanced MRI for characterization of blood-brain-barrier dysfunction after traumatic brain injury. Neuroimage Clin 2022; 36:103236. [PMID: 36274377 PMCID: PMC9668646 DOI: 10.1016/j.nicl.2022.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/30/2022] [Accepted: 10/16/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND AND PURPOSE Dysfunction of the blood-brain-barrier (BBB) is a recognized pathological consequence of traumatic brain injury (TBI) which may play an important role in chronic TBI pathophysiology. We hypothesized that BBB disruption can be detected with dynamic contrast-enhanced (DCE) MRI not only in association with focal traumatic lesions but also in normal-appearing brain tissue of TBI patients, reflecting microscopic microvascular injury. We further hypothesized that BBB integrity would improve but not completely normalize months after TBI. MATERIALS AND METHODS DCE MRI was performed in 40 adult patients a median of 23 days after hospitalized TBI and in 21 healthy controls. DCE data was analyzed using Patlak and linear models, and derived metrics of BBB leakage including the volume transfer constant (Ktrans) and the normalized permeability index (NPI) were compared between groups. BBB metrics were compared with focal lesion distribution as well as with contemporaneous measures of symptomatology and cognitive function in TBI patients. Finally, BBB metrics were examined longitudinally among 18 TBI patients who returned for a second MRI a median of 204 days postinjury. RESULTS TBI patients exhibited higher mean Ktrans (p = 0.0028) and proportion of suprathreshold NPI voxels (p = 0.001) relative to controls. Tissue-based analysis confirmed greatest TBI-related BBB disruption in association with focal lesions, however elevated Ktrans was also observed in perilesional (p = 0.011) and nonlesional (p = 0.044) regions. BBB disruption showed inverse correlation with quality of life (rho = -0.51, corrected p = 0.016). Among the subset of TBI patients who underwent a second MRI several months after the initial evaluation, metrics of BBB disruption did not differ significantly at the group level, though variable longitudinal changes were observed at the individual subject level. CONCLUSIONS This pilot investigation suggests that TBI-related BBB disruption is detectable in the early post-injury period in association with focal and diffuse brain injury.
Collapse
Affiliation(s)
- Jeffrey B Ware
- Division of Neuroradiology, Department of Radiology, Hospital of University of Pennsylvania, Perelman School of Medicine of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| | - Saurabh Sinha
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Justin Morrison
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Alexa E Walter
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - James J Gugger
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Andrea L C Schneider
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA; Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Cian Dabrowski
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Hannah Zamore
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Leroy Wesley
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Brigid Magdamo
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Dmitriy Petrov
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Junghoon J Kim
- Department of Molecular, Cellular, and Biomedical Sciences, CUNY School of Medicine at The City College of New York, Townsend Harris Hall, 160 Convent Avenue, New York, NY 10031, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| | - Danielle K Sandsmark
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104, USA
| |
Collapse
|
16
|
Zhang Y, Ding N, Hao X, Zhao J, Zhao Y, Li Y, Li Z. Manual acupuncture benignly regulates blood-brain barrier disruption and reduces lipopolysaccharide loading and systemic inflammation, possibly by adjusting the gut microbiota. Front Aging Neurosci 2022; 14:1018371. [PMID: 36313024 PMCID: PMC9607933 DOI: 10.3389/fnagi.2022.1018371] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/23/2022] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Blood-brain barrier (BBB) disruption and gut microbiota dysbiosis play crucial roles in Alzheimer's disease (AD). Lipopolysaccharide (LPS) stimulation triggered by gut microbial dysbiosis is an important factor in BBB disruption and systemic inflammation, but the mechanism of acupuncture regulation of BBB disruption via the gut microbiota in AD is not clear. OBJECTIVE The current study evaluated the effect of manual acupuncture (MA) on BBB dysfunction in APP/PS1 mice and examined the mechanism of gut microbiota by acupuncture in AD. METHODS Acupoints were applied to Baihui (GV20), Yintang (GV29), and Zusanli (ST36) in the MA group. Mice in the manual acupuncture plus antibiotics (MAa) group received antibiotics and acupuncture, while mice in the probiotics (P) group received probiotics. Alterations in spatial learning and memory, the gut microbiota, tightly connected structure and permeability of BBB, and the expression of LPS and inflammatory factors in each group were assessed. RESULTS Compared to the normal (N) group, cognitive ability was significantly impaired, the gut microbiota composition was markedly altered, the BBB was significantly disrupted, and the expression of LPS in serum and brain, serum TNF-α, and IL-1β were significantly increased in the AD group (p < 0.01). These changes were inhibited in the MA and P groups (p < 0.01 or p < 0.05), and antibiotics reversed the benign regulatory effects of MA (p < 0.01 or p < 0.05). CONCLUSION Manual acupuncture benignly modulated the gut microbiota and BBB dysfunction, reduced LPS, TNF-α, and IL-1β. These effects were comparable to probiotics. The decrease in LPS load and systemic inflammation may play important roles in the regulation of BBB dysfunction by acupuncture, and the gut microbiota may be a potential target for the benign regulation of BBB disruption by acupuncture.
Collapse
Affiliation(s)
- Yue Zhang
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ning Ding
- Department of Acupuncture, Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin Hao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Zhao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yali Zhao
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yiran Li
- School of International, Beijing University of Chinese Medicine, Beijing, China
| | - Zhigang Li
- School of Acupuncture, Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
17
|
Ganz T, Fainstein N, Ben-Hur T. When the infectious environment meets the AD brain. Mol Neurodegener 2022; 17:53. [PMID: 35986296 PMCID: PMC9388962 DOI: 10.1186/s13024-022-00559-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/08/2022] [Indexed: 11/23/2022] Open
Abstract
Background The Amyloid theory of Alzheimer’s disease (AD) suggests that the deposition of Amyloid β (Aβ) in the brain triggers a chain of events, involving the deposition of phosphorylated Tau and other misfolded proteins, leading to neurodegeneration via neuroinflammation, oxidative stress, and neurovascular factors. The infectious theory linked various infectious agents with the development of AD, raising the possibility that they serve as etiological causes of the disease. Are these theories mutually exclusive, or do they coincide? Main body In this review, we will discuss how the two theories converge. We present a model by which (1) the systemic infectious burden accelerates the development of AD brain pathology via bacterial Amyloids and other pathogen-associated molecular patterns (PAMPs), and (2) the developing AD brain pathology increases its susceptibility to the neurotoxicity of infectious agents -derived PAMPs, which drive neurodegeneration via activated microglia. Conclusions The reciprocal effects of amyloid deposition and systemic infectious burden may lead to a vicious cycle fueling Alzheimer’s disease pathogenesis.
Collapse
|
18
|
Saiyasit N, Butlig EAR, Chaney SD, Traylor MK, Hawley NA, Randall RB, Bobinger HV, Frizell CA, Trimm F, Crook ED, Lin M, Hill BD, Keller JL, Nelson AR. Neurovascular Dysfunction in Diverse Communities With Health Disparities-Contributions to Dementia and Alzheimer's Disease. Front Neurosci 2022; 16:915405. [PMID: 35844216 PMCID: PMC9279126 DOI: 10.3389/fnins.2022.915405] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease and related dementias (ADRD) are an expanding worldwide crisis. In the absence of scientific breakthroughs, the global prevalence of ADRD will continue to increase as more people are living longer. Racial or ethnic minority groups have an increased risk and incidence of ADRD and have often been neglected by the scientific research community. There is mounting evidence that vascular insults in the brain can initiate a series of biological events leading to neurodegeneration, cognitive impairment, and ADRD. We are a group of researchers interested in developing and expanding ADRD research, with an emphasis on vascular contributions to dementia, to serve our local diverse community. Toward this goal, the primary objective of this review was to investigate and better understand health disparities in Alabama and the contributions of the social determinants of health to those disparities, particularly in the context of vascular dysfunction in ADRD. Here, we explain the neurovascular dysfunction associated with Alzheimer's disease (AD) as well as the intrinsic and extrinsic risk factors contributing to dysfunction of the neurovascular unit (NVU). Next, we ascertain ethnoregional health disparities of individuals living in Alabama, as well as relevant vascular risk factors linked to AD. We also discuss current pharmaceutical and non-pharmaceutical treatment options for neurovascular dysfunction, mild cognitive impairment (MCI) and AD, including relevant studies and ongoing clinical trials. Overall, individuals in Alabama are adversely affected by social and structural determinants of health leading to health disparities, driven by rurality, ethnic minority status, and lower socioeconomic status (SES). In general, these communities have limited access to healthcare and healthy food and other amenities resulting in decreased opportunities for early diagnosis of and pharmaceutical treatments for ADRD. Although this review is focused on the current state of health disparities of ADRD patients in Alabama, future studies must include diversity of race, ethnicity, and region to best be able to treat all individuals affected by ADRD.
Collapse
Affiliation(s)
- Napatsorn Saiyasit
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Evan-Angelo R. Butlig
- Department of Neurology, Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, CA, United States
| | - Samantha D. Chaney
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Miranda K. Traylor
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Nanako A. Hawley
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Ryleigh B. Randall
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Hanna V. Bobinger
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Carl A. Frizell
- Department of Physician Assistant Studies, University of South Alabama, Mobile, AL, United States
| | - Franklin Trimm
- College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Errol D. Crook
- Department of Internal Medicine, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Mike Lin
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| | - Benjamin D. Hill
- Department of Psychology, University of South Alabama, Mobile, AL, United States
| | - Joshua L. Keller
- Department of Health, Kinesiology, and Sport, University of South Alabama, Mobile, AL, United States
| | - Amy R. Nelson
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, AL, United States
| |
Collapse
|
19
|
Nehra G, Bauer B, Hartz AMS. Blood-brain barrier leakage in Alzheimer's disease: From discovery to clinical relevance. Pharmacol Ther 2022; 234:108119. [PMID: 35108575 PMCID: PMC9107516 DOI: 10.1016/j.pharmthera.2022.108119] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD brain pathology starts decades before the onset of clinical symptoms. One early pathological hallmark is blood-brain barrier dysfunction characterized by barrier leakage and associated with cognitive decline. In this review, we summarize the existing literature on the extent and clinical relevance of barrier leakage in AD. First, we focus on AD animal models and their susceptibility to barrier leakage based on age and genetic background. Second, we re-examine barrier dysfunction in clinical and postmortem studies, summarize changes that lead to barrier leakage in patients and highlight the clinical relevance of barrier leakage in AD. Third, we summarize signaling mechanisms that link barrier leakage to neurodegeneration and cognitive decline in AD. Finally, we discuss clinical relevance and potential therapeutic strategies and provide future perspectives on investigating barrier leakage in AD. Identifying mechanistic steps underlying barrier leakage has the potential to unravel new targets that can be used to develop novel therapeutic strategies to repair barrier leakage and slow cognitive decline in AD and AD-related dementias.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
20
|
Montagne A, Barnes SR, Nation DA, Kisler K, Toga AW, Zlokovic BV. Imaging subtle leaks in the blood-brain barrier in the aging human brain: potential pitfalls, challenges, and possible solutions. GeroScience 2022; 44:1339-1351. [PMID: 35469116 PMCID: PMC9213625 DOI: 10.1007/s11357-022-00571-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/10/2022] [Indexed: 02/06/2023] Open
Abstract
Recent studies using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) with gadolinium-based contrast agents (GBCA) have demonstrated subtle blood-brain barrier (BBB) leaks in the human brain during normal aging, in individuals with age-related cognitive dysfunction, genetic risk for Alzheimer's disease (AD), mild cognitive impairment, early AD, cerebral small vessel disease (SVD), and other neurodegenerative disorders. In these neurological conditions, the BBB leaks, quantified by the unidirectional BBB GBCA tracer's constant Ktrans maps, are typically orders of magnitude lower than in brain tumors, after stroke and/or during relapsing episodes of multiple sclerosis. This puts extra challenges for the DCE-MRI technique by pushing calculations towards its lower limits of detectability. In addition, presently, there are no standardized multivendor protocols or evidence of repeatability and reproducibility. Nevertheless, subtle BBB leaks may critically contribute to the pathophysiology of cognitive impairment and dementia associated with AD or SVD, and therefore, efforts to improve sensitivity of detection, reliability, and reproducibility are warranted. A larger number of participants scanned by different MR scanners at different clinical sites are sometimes required to detect differences in BBB integrity between control and at-risk groups, which impose additional challenges. Here, we focus on these new challenges and propose some approaches to normalize and harmonize DCE data between different scanners. In brief, we recommend specific regions to be used for the tracer's vascular input function and DCE data processing and how to find and correct negative Ktrans values that are physiologically impossible. We hope this information will prove helpful to new investigators wishing to study subtle BBB damage in neurovascular and neurodegenerative conditions and in the aging human brain.
Collapse
Affiliation(s)
- Axel Montagne
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK.
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Samuel R Barnes
- Department of Radiology, Loma Linda University, Loma Linda, CA, USA.
| | - Daniel A Nation
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
- Department of Psychological Science, University of California Irvine, Irvine, CA, USA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuroimaging, USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
21
|
Kucharz K, Kutuzov N, Zhukov O, Mathiesen Janiurek M, Lauritzen M. Shedding Light on the Blood-Brain Barrier Transport with Two-Photon Microscopy In Vivo. Pharm Res 2022; 39:1457-1468. [PMID: 35578062 DOI: 10.1007/s11095-022-03266-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023]
Abstract
Treatment of brain disorders relies on efficient delivery of therapeutics to the brain, which is hindered by the blood-brain barrier (BBB). The work of Prof. Margareta Hammarlund-Udenaes was instrumental in understanding the principles of drug delivery to the brain and developing new tools to study it. Here, we show how some of the concepts developed in her research can be translated to in vivo 2-photon microscopy (2PM) studies of the BBB. We primarily focus on the methods developed in our laboratory to characterize the paracellular diffusion, adsorptive-mediated transcytosis, and receptor-mediated transcytosis of drug nanocarriers at the microscale, illustrating how 2PM can deepen our understanding of the mechanisms of drug delivery to the brain.
Collapse
Affiliation(s)
- Krzysztof Kucharz
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nikolay Kutuzov
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Oleg Zhukov
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Mathiesen Janiurek
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Lauritzen
- Department of Neuroscience, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark. .,Department of Clinical Neurophysiology, Rigshospitalet, Glostrup, Denmark.
| |
Collapse
|
22
|
Is the Brain Undernourished in Alzheimer's Disease? Nutrients 2022; 14:nu14091872. [PMID: 35565839 PMCID: PMC9102563 DOI: 10.3390/nu14091872] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/22/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023] Open
Abstract
Cerebrospinal fluid (CSF) amino acid (AA) levels and CSF/plasma AA ratios in Alzheimer Disease (AD) in relation to nutritional state are not known. Methods: In 30 fasting patients with AD (46% males, 74.4 ± 8.2 years; 3.4 ± 3.2 years from diagnosis) and nine control (CTRL) matched subjects, CSF and venous blood samples were drawn for AA measurements. Patients were stratified according to nutritional state (Mini Nutritional Assessment, MNA, scores). Results: Total CSF/plasma AA ratios were lower in the AD subpopulations than in NON-AD (p < 0.003 to 0.017. In combined malnourished (16.7%; MNA < 17) and at risk for malnutrition (36.6%, MNA 17−24) groups (CG), compared to CTRL, all essential amino acids (EAAs) and 30% of non-EAAs were lower (p < 0.018 to 0.0001), whereas in normo-nourished ADs (46.7%, MNA > 24) the CSF levels of 10% of EAAs and 25% of NON-EAAs were decreased (p < 0.05 to 0.00021). CG compared to normo-nourished ADs, had lower CSF aspartic acid, glutamic acid and Branched-Chain AA levels (all, p < 0.05 to 0.003). CSF/plasma AA ratios were <1 in NON-AD but even lower in the AD population. Conclusions: Compared to CTRL, ADs had decreased CSF AA Levels and CSF/plasma AA ratios, the degree of which depended on nutritional state.
Collapse
|
23
|
Nelson AR. Peripheral Pathways to Neurovascular Unit Dysfunction, Cognitive Impairment, and Alzheimer’s Disease. Front Aging Neurosci 2022; 14:858429. [PMID: 35517047 PMCID: PMC9062225 DOI: 10.3389/fnagi.2022.858429] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. It was first described more than a century ago, and scientists are acquiring new data and learning novel information about the disease every day. Although there are nuances and details continuously being unraveled, many key players were identified in the early 1900’s by Dr. Oskar Fischer and Dr. Alois Alzheimer, including amyloid-beta (Aβ), tau, vascular abnormalities, gliosis, and a possible role of infections. More recently, there has been growing interest in and appreciation for neurovascular unit dysfunction that occurs early in mild cognitive impairment (MCI) before and independent of Aβ and tau brain accumulation. In the last decade, evidence that Aβ and tau oligomers are antimicrobial peptides generated in response to infection has expanded our knowledge and challenged preconceived notions. The concept that pathogenic germs cause infections generating an innate immune response (e.g., Aβ and tau produced by peripheral organs) that is associated with incident dementia is worthwhile considering in the context of sporadic AD with an unknown root cause. Therefore, the peripheral amyloid hypothesis to cognitive impairment and AD is proposed and remains to be vetted by future research. Meanwhile, humans remain complex variable organisms with individual risk factors that define their immune status, neurovascular function, and neuronal plasticity. In this focused review, the idea that infections and organ dysfunction contribute to Alzheimer’s disease, through the generation of peripheral amyloids and/or neurovascular unit dysfunction will be explored and discussed. Ultimately, many questions remain to be answered and critical areas of future exploration are highlighted.
Collapse
|
24
|
Fisher RA, Miners JS, Love S. Pathological changes within the cerebral vasculature in Alzheimer's disease: New perspectives. Brain Pathol 2022; 32:e13061. [PMID: 35289012 PMCID: PMC9616094 DOI: 10.1111/bpa.13061] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular disease underpins vascular dementia (VaD), but structural and functional changes to the cerebral vasculature contribute to disease pathology and cognitive decline in Alzheimer's disease (AD). In this review, we discuss the contribution of cerebral amyloid angiopathy and non‐amyloid small vessel disease in AD, and the accompanying changes to the density, maintenance and remodelling of vessels (including alterations to the composition and function of the cerebrovascular basement membrane). We consider how abnormalities of the constituent cells of the neurovascular unit – particularly of endothelial cells and pericytes – and impairment of the blood‐brain barrier (BBB) impact on the pathogenesis of AD. We also discuss how changes to the cerebral vasculature are likely to impair Aβ clearance – both intra‐periarteriolar drainage (IPAD) and transport of Aβ peptides across the BBB, and how impaired neurovascular coupling and reduced blood flow in relation to metabolic demand increase amyloidogenic processing of APP and the production of Aβ. We review the vasoactive properties of Aβ peptides themselves, and the probable bi‐directional relationship between vascular dysfunction and Aβ accumulation in AD. Lastly, we discuss recent methodological advances in transcriptomics and imaging that have provided novel insights into vascular changes in AD, and recent advances in assessment of the retina that allow in vivo detection of vascular changes in the early stages of AD.
Collapse
Affiliation(s)
- Robert A Fisher
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - J Scott Miners
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - Seth Love
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| |
Collapse
|
25
|
Vacher M, Porter T, Milicic L, Bourgeat P, Dore V, Villemagne VL, Laws SM, Doecke JD. A Targeted Association Study of Blood-Brain Barrier Gene SNPs and Brain Atrophy. J Alzheimers Dis 2022; 86:1817-1829. [DOI: 10.3233/jad-210644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: The blood-brain barrier (BBB) is formed by a high-density lining of endothelial cells, providing a border between circulating blood and the brain interstitial fluid. This structure plays a key role in protecting the brain microenvironment by restricting passage of certain molecules and circulating pathogens. Objective: To identify associations between brain volumetric changes and a set of 355 BBB-related single nucleotide polymorphisms (SNP). Method: In a population of 721 unrelated individuals, linear mixed effect models were used to assess if specific variants were linked to regional rates of atrophy over a 12-year time span. Four brain regions were investigated, including cortical grey matter, cortical white matter, ventricle, and hippocampus. Further, we also investigated the potential impact of history of hypertension, diabetes, and the incidence of stroke on regional brain volume change. Results: History of hypertension, diabetes, and stroke was not associated with longitudinal brain volume change. However, we identified a series of genetic variants associated with regional brain volume changes. The associations were independent of variation due to the APOEɛ4 allele and were significant post correction for multiple comparisons. Conclusion: This study suggests that key genes involved in the regulation of BBB integrity may be associated with longitudinal changes in specific brain regions. The derived polygenic risk scores indicate that these interactions are multigenic. Further research needs to be conducted to investigate how BBB functions maybe compromised by genetic variation.
Collapse
Affiliation(s)
- Michael Vacher
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Floreat, Western Australia, Australia
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
| | - Tenielle Porter
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia
| | - Lidija Milicic
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
| | - Pierrick Bourgeat
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Herston, Queensland, Australia
| | - Vincent Dore
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Herston, Queensland, Australia
| | - Victor L Villemagne
- Department of Molecular Imaging & Therapy and Centre for PET, Austin Health, Heidelberg, Victoria, Australia
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon M. Laws
- Centre for Precision Health, Edith Cowan University, Joondalup, Western Australia, Australia
- Collaborative Genomics and Translation Group, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia
| | - James D. Doecke
- CSIRO Health and Biosecurity, Australian e-Health Research Centre, Herston, Queensland, Australia
| |
Collapse
|
26
|
Barisano G, Montagne A, Kisler K, Schneider JA, Wardlaw JM, Zlokovic BV. Blood-brain barrier link to human cognitive impairment and Alzheimer's Disease. NATURE CARDIOVASCULAR RESEARCH 2022; 1:108-115. [PMID: 35450117 PMCID: PMC9017393 DOI: 10.1038/s44161-021-00014-4] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/21/2021] [Indexed: 01/18/2023]
Abstract
Vascular dysfunction is frequently seen in disorders associated with cognitive impairment, dementia and Alzheimer's disease (AD). Recent advances in neuroimaging and fluid biomarkers suggest that vascular dysfunction is not an innocent bystander only accompanying neuronal dysfunction. Loss of cerebrovascular integrity, often referred to as breakdown in the blood-brain barrier (BBB), has recently shown to be an early biomarker of human cognitive dysfunction and possibly underlying mechanism of age-related cognitive decline. Damage to the BBB may initiate or further invoke a range of tissue injuries causing synaptic and neuronal dysfunction and cognitive impairment that may contribute to AD. Therefore, better understanding of how vascular dysfunction caused by BBB breakdown interacts with amyloid-β and tau AD biomarkers to confer cognitive impairment may lead to new ways of thinking about pathogenesis, and possibly treatment and prevention of early cognitive impairment, dementia and AD, for which we still do not have effective therapies.
Collapse
Affiliation(s)
- Giuseppe Barisano
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA, USA
- These authors contributed equally: Giuseppe Barisano and Axel Montagne
| | - Axel Montagne
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- These authors contributed equally: Giuseppe Barisano and Axel Montagne
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Julie A. Schneider
- Departments of Pathology and Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
- Rush Alzheimer’s Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Joanna M. Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Berislav V. Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Alzheimer’s Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
27
|
Spampinato SF, Takeshita Y, Obermeier B. An In Vitro Model of the Blood-Brain Barrier to Study Alzheimer's Disease: The Role of β-Amyloid and Its Influence on PBMC Infiltration. Methods Mol Biol 2022; 2492:333-352. [PMID: 35733055 DOI: 10.1007/978-1-0716-2289-6_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The blood-brain barrier (BBB) is a highly specialized structure, constituted by endothelial cells that together with astrocytes and pericytes provide a functional interface between the central nervous system and the periphery. Several pathological conditions may affect its functions, and lately BBB involvement in the pathogenesis of Alzheimer's disease has been demonstrated. Both endothelial cells and astrocytes can be differentially affected during the course of the disease. In vitro BBB models present a powerful tool in evaluating the effects that β-amyloid (Aβ), or other pathogenic stimuli, play on the BBB at cellular level. In vitro BBB models derived from human cell sources are rare and not easily implemented. We generated two conditionally immortalized human cell lines, brain microvascular endothelial cells (TY10), and astrocytes (hAST), that, when co-cultured under appropriate conditions, exhibit BBB-like characteristics. This model allowed us to evaluate the transmigration of peripheral blood mononuclear cells (PBMCs) through the in vitro barrier exposed to Aβ and the role played by astrocytes in the modulation of this phenomenon. We describe here the methodology used in our lab to set up our in vitro model of the BBB and to carry out a PBMC transmigration assay.
Collapse
Affiliation(s)
- Simona Federica Spampinato
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy.
- Departement of Scienza e Tecnologia del Farmaco, Universita' di Turin, Turin, Italy.
| | - Yukio Takeshita
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | |
Collapse
|
28
|
Tao QQ, Lin RR, Chen YH, Wu ZY. Discerning the Role of Blood Brain Barrier Dysfunction in Alzheimer’s Disease. Aging Dis 2022; 13:1391-1404. [PMID: 36186141 PMCID: PMC9466977 DOI: 10.14336/ad.2022.0130-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 01/30/2022] [Indexed: 12/04/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of neurodegenerative disease. The predominant characteristics of AD are the accumulation of amyloid-β (Aβ) and hyperphosphorylated tau in the brain. Blood brain barrier (BBB) dysfunction as one of the causative factors of cognitive impairment is increasingly recognized in the last decades. However, the role of BBB dysfunction in AD pathogenesis is still not fully understood. It remains elusive whether BBB dysfunction is a consequence or causative fact of Aβ pathology, tau pathology, neuroinflammation, or other conditions. In this review, we summarized the major findings of BBB dysfunction in AD and the reciprocal relationships between BBB dysfunction, Aβ pathology, tau pathology, and neuroinflammation. In addition, the implications of BBB dysfunction in AD for delivering therapeutic drugs were presented. Finally, we discussed how to better determine the underlying mechanisms between BBB dysfunction and AD, as well as how to explore new therapies for BBB regulation to treat AD in the future.
Collapse
Affiliation(s)
| | | | | | - Zhi-Ying Wu
- Correspondence should be addressed to: Dr. Zhi-Ying Wu, the Department of Neurology in Second Affiliated Hospital, and Key Laboratory of Medical Neurobiology of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China. E-mail:
| |
Collapse
|
29
|
Nguyen B, Bix G, Yao Y. Basal lamina changes in neurodegenerative disorders. Mol Neurodegener 2021; 16:81. [PMID: 34876200 PMCID: PMC8650282 DOI: 10.1186/s13024-021-00502-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neurodegenerative disorders are a group of age-associated diseases characterized by progressive degeneration of the structure and function of the CNS. Two key pathological features of these disorders are blood-brain barrier (BBB) breakdown and protein aggregation. MAIN BODY The BBB is composed of various cell types and a non-cellular component---the basal lamina (BL). Although how different cells affect the BBB is well studied, the roles of the BL in BBB maintenance and function remain largely unknown. In addition, located in the perivascular space, the BL is also speculated to regulate protein clearance via the meningeal lymphatic/glymphatic system. Recent studies from our laboratory and others have shown that the BL actively regulates BBB integrity and meningeal lymphatic/glymphatic function in both physiological and pathological conditions, suggesting that it may play an important role in the pathogenesis and/or progression of neurodegenerative disorders. In this review, we focus on changes of the BL and its major components during aging and in neurodegenerative disorders, including Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS). First, we introduce the vascular and lymphatic systems in the CNS. Next, we discuss the BL and its major components under homeostatic conditions, and summarize their changes during aging and in AD, PD, and ALS in both rodents and humans. The functional significance of these alterations and potential therapeutic targets are also reviewed. Finally, key challenges in the field and future directions are discussed. CONCLUSIONS Understanding BL changes and the functional significance of these changes in neurodegenerative disorders will fill the gap of knowledge in the field. Our goal is to provide a clear and concise review of the complex relationship between the BL and neurodegenerative disorders to stimulate new hypotheses and further research in this field.
Collapse
Affiliation(s)
- Benjamin Nguyen
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA
| | - Gregory Bix
- Clinical Neuroscience Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA, USA.
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, MDC 8, Tampa, Florida, 33612, USA.
| |
Collapse
|
30
|
Zhao M, Jiang XF, Zhang HQ, Sun JH, Pei H, Ma LN, Cao Y, Li H. Interactions between glial cells and the blood-brain barrier and their role in Alzheimer's disease. Ageing Res Rev 2021; 72:101483. [PMID: 34610479 DOI: 10.1016/j.arr.2021.101483] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 09/14/2021] [Accepted: 09/30/2021] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD), which is an irreversible neurodegenerative disorder characterized by senile plaques and neurofibrillary tangles, is the most common form of dementia worldwide. However, currently, there are no satisfying curative therapies for AD. The blood-brain barrier (BBB) acts as a selective physical barrier and plays protective roles in maintaining brain homeostasis. BBB dysfunction as an upstream or downstream event promotes the onset and progression of AD. Moreover, the pathogenesis of AD caused by BBB injury hasn't been well elucidated. Glial cells, BBB compartments and neurons form a minimal functional unit called the neurovascular unit (NVU). Emerging evidence suggests that glial cells are regulators in maintaining the BBB integrity and neuronal function. Illustrating the regulatory mechanism of glial cells in the BBB assists us in drawing a glial-vascular coupling diagram of AD, which may offer new insight into the pathogenesis of AD and early intervention strategies for AD. This review aims to summarize our current knowledge of glial-BBB interactions and their pathological implications in AD and to provide new therapeutic potentials for future investigations.
Collapse
|
31
|
Chagnot A, Barnes SR, Montagne A. Magnetic Resonance Imaging of Blood-Brain Barrier permeability in Dementia. Neuroscience 2021; 474:14-29. [PMID: 34400249 PMCID: PMC8528227 DOI: 10.1016/j.neuroscience.2021.08.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) and cerebral small vessel disease (cSVD) are the two main causes of dementia with blood-brain barrier (BBB) breakdown being a common contributor. Recent advances in neuroimaging techniques offer new possibilities to understand how the brain functions in health and disease. This includes methods such as dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) which allows the detection of subtle regional changes in the BBB integrity. The purpose of this work is to provide a review on the recent DCE-MRI findings of subtle BBB leakage focusing on cSVD and AD, including both clinical and pre-clinical studies. Despite being widely used and well-established, we also highlight some of the DCE-MRI challenges and pitfalls faced in the context of dementia inherent to the subtle nature of BBB impairment.
Collapse
Affiliation(s)
- Audrey Chagnot
- Normandie Université, UNICAEN, INSERM, UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute Blood and Brain @ Caen-Normandie (BB@C), GIP Cyceron, Caen, France
| | - Samuel R Barnes
- Department of Radiology, Loma Linda University, Loma Linda, CA, USA
| | - Axel Montagne
- UK Dementia Research Institute, Edinburgh Medical School, University of Edinburgh, Edinburgh, UK; Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
32
|
Utilizing 3D Arterial Spin Labeling to Identify Cerebrovascular Leak and Glymphatic Obstruction in Neurodegenerative Disease. Diagnostics (Basel) 2021; 11:diagnostics11101888. [PMID: 34679586 PMCID: PMC8534509 DOI: 10.3390/diagnostics11101888] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/27/2021] [Accepted: 10/04/2021] [Indexed: 01/19/2023] Open
Abstract
New approaches are required to successfully intervene therapeutically in neurodegenerative diseases. Addressing the earliest phases of disease, blood brain barrier (BBB) leak before the accumulation of misfolded proteins has significant potential for success. To do so, however, a reliable, noninvasive and economical test is required. There are two potential methods of identifying the BBB fluid leak that results in the accumulation of normally excluded substances which alter neuropil metabolism, protein synthesis and degradation with buildup of misfolded toxic proteins. The pros and cons of dynamic contrast imaging (DCI or DCE) and 3D TGSE PASL are discussed as potential early identifying methods. The results of prior publications of the 3D ASL technique and an overview of the associated physiologic challenges are discussed. Either method may serve well as reliable physiologic markers as novel therapeutic interventions directed at the vasculopathy of early neurodegenerative disease are developed. They may serve well in addressing other neurologic diseases associated with either vascular leak and/or reduced glymphatic flow.
Collapse
|
33
|
Yan XZ, Lai L, Ao Q, Tian XH, Zhang YH. Interleukin-17A in Alzheimer's disease: recent advances and controversies. Curr Neuropharmacol 2021; 20:372-383. [PMID: 34429057 PMCID: PMC9413786 DOI: 10.2174/1570159x19666210823110004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/03/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease that mainly affects older adults. Although the global burden of AD is increasing year by year, the causes of AD remain largely unknown. Numerous basic and clinical studies have shown that interleukin-17A (IL-17A) may play a significant role in the pathogenesis of AD. A comprehensive assessment of the role of IL-17A in AD would benefit the diagnosis, understanding of etiology and treatment. However, over the past decade, controversies remain regarding the expression level and role of IL-17A in AD. We have incorporated newly published researches and point out that IL-17A expression levels may vary along with the development of AD, exercising different roles at different stages of AD, although much more work remains to be done to support the potential role of IL-17A in AD-related pathology. Here, it is our intention to review the underlying mechanisms of IL-17A in AD and address the current controversies in an effort to clarify the results of existing research and suggest future studies.
Collapse
Affiliation(s)
- Xin-Zhu Yan
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang 110122. China
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT. 0
| | - Qiang Ao
- Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064. China
| | - Xiao-Hong Tian
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang 110122. China
| | - Yan-Hui Zhang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, 77 Puhe Avenue, Shenbei New District, Shenyang 110122. China
| |
Collapse
|
34
|
Liu S, Gao J, Liu K, Zhang HL. Microbiota-gut-brain axis and Alzheimer's disease: Implications of the blood-brain barrier as an intervention target. Mech Ageing Dev 2021; 199:111560. [PMID: 34411603 DOI: 10.1016/j.mad.2021.111560] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/26/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
The microbiota-gut-brain axis has emerged as a focal point of biomedical research. Alterations of gut microbiota are involved in not only various immune/inflammatory disorders but also neurological disorders including Alzheimer's disease (AD). The initial stage of the involvement of gut microbiota in the pathogenesis of AD may be the dysfunction of the blood-brain barrier (BBB). Gut microbiota-derived products in the circulation can worsen the BBB integrity, easily cross the disrupted BBB and enter the brain to promote pathological changes in AD. In this review, we first summarize the current evidence of the associations among gut microbiota, AD, and BBB integrity. We then discuss the mechanism of gut microbiota on BBB dysfunction with a focus on bacteria-derived lipopolysaccharide and exosomal high-mobility group box 1. Novel insights into the modification of the BBB as an intervention approach for AD are highlighted as well.
Collapse
Affiliation(s)
- Shan Liu
- Department of Neurology, First Hospital of Jilin University, Jilin University, Changchun, China
| | - Jiguo Gao
- Department of Neurology, First Hospital of Jilin University, Jilin University, Changchun, China
| | - Kangding Liu
- Department of Neurology, First Hospital of Jilin University, Jilin University, Changchun, China.
| | - Hong-Liang Zhang
- Department of Life Sciences, National Natural Science Foundation of China, Shuangqing Road 83, 100085, Beijing, China.
| |
Collapse
|
35
|
Sharma HS, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Buzoianu AD, Sahib S, Tian ZR, Bryukhovetskiy I, Manzhulo I, Menon PK, Patnaik R, Wiklund L, Sharma A. Alzheimer's disease neuropathology is exacerbated following traumatic brain injury. Neuroprotection by co-administration of nanowired mesenchymal stem cells and cerebrolysin with monoclonal antibodies to amyloid beta peptide. PROGRESS IN BRAIN RESEARCH 2021; 265:1-97. [PMID: 34560919 DOI: 10.1016/bs.pbr.2021.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Military personnel are prone to traumatic brain injury (TBI) that is one of the risk factors in developing Alzheimer's disease (AD) at a later stage. TBI induces breakdown of the blood-brain barrier (BBB) to serum proteins into the brain and leads to extravasation of plasma amyloid beta peptide (ΑβP) into the brain fluid compartments causing AD brain pathology. Thus, there is a need to expand our knowledge on the role of TBI in AD. In addition, exploration of the novel roles of nanomedicine in AD and TBI for neuroprotection is the need of the hour. Since stem cells and neurotrophic factors play important roles in TBI and in AD, it is likely that nanodelivery of these agents exert superior neuroprotection in TBI induced exacerbation of AD brain pathology. In this review, these aspects are examined in details based on our own investigations in the light of current scientific literature in the field. Our observations show that TBI exacerbates AD brain pathology and TiO2 nanowired delivery of mesenchymal stem cells together with cerebrolysin-a balanced composition of several neurotrophic factors and active peptide fragments, and monoclonal antibodies to amyloid beta protein thwarted the development of neuropathology following TBI in AD, not reported earlier.
Collapse
Affiliation(s)
- Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Igor Manzhulo
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
36
|
Price BR, Johnson LA, Norris CM. Reactive astrocytes: The nexus of pathological and clinical hallmarks of Alzheimer's disease. Ageing Res Rev 2021; 68:101335. [PMID: 33812051 PMCID: PMC8168445 DOI: 10.1016/j.arr.2021.101335] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/21/2021] [Accepted: 03/20/2021] [Indexed: 02/06/2023]
Abstract
Astrocyte reactivity is a hallmark of neuroinflammation that arises with Alzheimer’s disease (AD) and nearly every other neurodegenerative condition. While astrocytes certainly contribute to classic inflammatory processes (e.g. cytokine release, waste clearance, and tissue repair), newly emerging technologies for measuring and targeting cell specific activities in the brain have uncovered essential roles for astrocytes in synapse function, brain metabolism, neurovascular coupling, and sleep/wake patterns. In this review, we use a holistic approach to incorporate, and expand upon, classic neuroinflammatory concepts to consider how astrocyte dysfunction/reactivity modulates multiple pathological and clinical hallmarks of AD. Our ever-evolving understanding of astrocyte signaling in neurodegeneration is not only revealing new drug targets and treatments for dementia but is suggesting we reimagine AD pathophysiological mechanisms.
Collapse
Affiliation(s)
- Brittani R Price
- Department of Neuroscience, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA, 02111, USA
| | - Lance A Johnson
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Physiology, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA
| | - Christopher M Norris
- Sanders-Brown Center on Aging, University of Kentucky, 800 S. Limestone St., Lexington, KY, 40356, USA; Department of Pharmacology and Nutritional Sciences, University of Kentucky, College of Medicine, UK Medical Center MN 150, Lexington, KY, 40536, USA.
| |
Collapse
|
37
|
Canjels LPW, Jansen JFA, van den Kerkhof M, Alers RJ, Poser BA, Wiggins CJ, Schiffer VMMM, van de Ven V, Rouhl RPW, Palm WM, van Oostenbrugge RJ, Aldenkamp AP, Ghossein-Doha C, Spaanderman MEA, Backes WH. 7T dynamic contrast-enhanced MRI for the detection of subtle blood-brain barrier leakage. J Neuroimaging 2021; 31:902-911. [PMID: 34161640 PMCID: PMC8519128 DOI: 10.1111/jon.12894] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/01/2022] Open
Abstract
Background and Purpose Dynamic contrast‐enhanced MRI (DCE‐MRI) can be employed to assess the blood–brain barrier (BBB) integrity. Detection of BBB leakage at lower field strengths (≤3T) is cumbersome as the signal is noisy, while leakage can be subtle. Utilizing the increased signal‐to‐noise ratio at higher field strengths, we explored the application of 7T DCE‐MRI for assessing BBB leakage. Methods A dual‐time resolution DCE‐MRI method was implemented at 7T and a slow injection rate (0.3 ml/s) and low dose (3 mmol) served to obtain signal changes linearly related to the gadolinium concentration, that is, minimized for T2* degradation effects. With the Patlak graphical approach, the leakage rate (Ki) and blood plasma volume fraction (vp) were calculated. The method was evaluated in 10 controls, an ischemic stroke patient, and a patient with a transient ischemic attack. Results Ki and vp were significantly higher in gray matter compared to white matter of all participants. These Ki values were higher in both patients compared to the control subjects. Finally, for the lesion identified in the ischemic stroke patient, higher leakage values were observed compared to normal‐appearing tissue. Conclusion We demonstrate how a dual‐time resolution DCE‐MRI protocol at 7T, with administration of half the clinically used contrast agent dose, can be used for assessing subtle BBB leakage. Although the feasibility of DCE‐MRI for assessing the BBB integrity at 3T is well known, we showed that a continuous sampling DCE‐MRI method tailored for 7T is also capable of assessing leakage with a high sensitivity over a range of Ki values.
Collapse
Affiliation(s)
- Lisanne P W Canjels
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,MHENS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,MHENS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Marieke van den Kerkhof
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,MHENS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Robert-Jan Alers
- Department of Gynecology and Obstetrics, Maastricht University Medical Center, Maastricht, the Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Benedikt A Poser
- Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | | | - Veronique M M M Schiffer
- Department of Gynecology and Obstetrics, Maastricht University Medical Center, Maastricht, the Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Vincent van de Ven
- Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Rob P W Rouhl
- MHENS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands.,Academic Center for Epileptology Kempenhaeghe/Maastricht UMC+, Heeze and Maastricht, the Netherlands
| | - W M Palm
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- MHENS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands.,CARIM, School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| | - Albert P Aldenkamp
- MHENS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.,Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands.,Academic Center for Epileptology Kempenhaeghe/Maastricht UMC+, Heeze and Maastricht, the Netherlands
| | - Chahinda Ghossein-Doha
- GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.,CARIM, School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands.,Department of Cardiology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Marc E A Spaanderman
- Department of Gynecology and Obstetrics, Maastricht University Medical Center, Maastricht, the Netherlands.,GROW, School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Walter H Backes
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,MHENS, School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,CARIM, School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
38
|
Sun H, Hu H, Liu C, Sun N, Duan C. Methods used for the measurement of blood-brain barrier integrity. Metab Brain Dis 2021; 36:723-735. [PMID: 33635479 DOI: 10.1007/s11011-021-00694-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/11/2021] [Indexed: 01/12/2023]
Abstract
The blood-brain barrier (BBB) comprises the interface between blood, brain and cerebrospinal fluid. Its primary function, which is mainly carried out by tight junctions, is to stabilize the tightly controlled microenvironment of the brain. To study the development and maintenance of the BBB, as well as various roles their intrinsic mechanisms that play in neurological disorders, suitable measurements are required to demonstrate integrity and functional changes at the interfaces between the blood and brain tissue. Markers and plasma proteins with different molecular weight (MW) are used to measure the permeability of BBB. In addition, the expression changes of tight-junction proteins form the basic structure of BBB, and imaging modalities are available to study the disruption of BBB. In the present review, above mentioned methods are depicted in details, together with the pros and cons as well as the differences between these methods, which maybe benefit research studies focused on the detection of BBB breakdown.
Collapse
Affiliation(s)
- Huixin Sun
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Huiling Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Chuanjie Liu
- Weihai City Key Laboratory of Autoimmunity, Weihai Central Hospital, Weihai, 264400, Shandong Province, China
| | - Nannan Sun
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing, 100005, China.
| | - Chaohui Duan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
39
|
Taler M, Aronovich R, Henry Hornfeld S, Dar S, Sasson E, Weizman A, Hochman E. Regulatory effect of lithium on hippocampal blood-brain barrier integrity in a rat model of depressive-like behavior. Bipolar Disord 2021; 23:55-65. [PMID: 32558151 DOI: 10.1111/bdi.12962] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Recent evidence has associated mood disorders with blood-brain barrier (BBB)/ neurovascular unit (NVU) dysfunction, and reduction in blood vessels coverage by the water channel aquaporin-4 (AQP4) immunoreactive astrocytes. Lithium is an established treatment for mood disorders, yet, its mechanism of action is partially understood. We investigated the effects of lithium on BBB integrity and NVU-related protein expression in chronic mild stress (CMS) rat model of depressive-like behavior. METHODS Male Wistar rats were exposed for 5 weeks to unpredictable mild stressors with daily co-administration of lithium chloride to half of the stressed and unstressed groups. Sucrose preference and open field tests were conducted to validate the depressive-like phenotype, and dynamic contrast-enhanced MRI analysis was utilized to assess BBB integrity in brain regions relevant to the pathophysiology of depression. Hippocampal AQP4 and claudin-5 expression were studied using immunofluorescence, western blot, and enzyme-linked immunosorbent assays. RESULTS Lithium administration to the stressed rats prevented the reductions in sucrose preference and distance traveled in the open field, and normalized the stress-induced hippocampal BBB hyperpermeability, whereas lithium administration to the unstressed rats increased hippocampal BBB permeability. Additionally, lithium treatment attenuated the decrease in hippocampal AQP4 to glial fibrillary acidic protein immunoreactivity ratio in the stressed rats and upregulated hippocampal claudin-5 and BDNF proteins expression. CONCLUSIONS Our findings suggest that lithium administration in a rat CMS model of depressive-like behavior is associated with attenuation of stressed-induced hippocampal BBB/NVU disruption. These protective effects may be relevant to the mode of action of lithium in depression.
Collapse
Affiliation(s)
- Michal Taler
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel
| | - Ramona Aronovich
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel
| | - Shay Henry Hornfeld
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel
| | - Shira Dar
- Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel
| | | | - Abraham Weizman
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel.,Geha Mental Health Center, Petah Tikva, Israel
| | - Eldar Hochman
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel.,Laboratory of Biological Psychiatry, Felsenstein Medical Research Center, Petah Tikva, Israel.,Geha Mental Health Center, Petah Tikva, Israel
| |
Collapse
|
40
|
Wei J, Dai Y, Wen W, Li J, Ye LL, Xu S, Duan DD. Blood-brain barrier integrity is the primary target of alcohol abuse. Chem Biol Interact 2021; 337:109400. [PMID: 33516661 DOI: 10.1016/j.cbi.2021.109400] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/31/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022]
Abstract
The effects of long-term alcohol consumptions on cognitive function remain elusive with contradictory results. Whilst it is widely accepted that long-term intoxication can cause cognitive impairment, moderate drinking can improve cognitive function. In reality, many older people and those with chronic medical conditions are long-term alcohol consumers in Asian countries. Our previous studies have suggested that long-term alcohol consumption can damage blood-brain barrier (BBB) integrity and aggravate cognitive deficit in APPswe/PS1De9 mice, but little is known about the underlying mechanisms, especially whether this consumption can cause cognitive decline via aggravating BBB damage in people who are exposed to the risk factors for cognitive disorders such as aging or inflammation. These questions were addressed in this study. The mouse models of cognitive deficit induced by d-galactose or lipopolysaccharide, the important risk conditions in human on cognitive function, were used to evaluate the effects of long-term alcohol consumption on the BBB integrity. After alcohol administration for 30 days in these models the BBB integrity was significantly destroyed with remarkably increased permeability and down-regulated protein expression of zonula occludens-1, VE-cadherin, occludin, low-density lipoprotein receptor-related protein-1, receptor for advanced glycation end products, major facilitator superfamily domain-containing protein-2a and aquaporin-4, which is the most closely related with the structure and function of BBB integrity. Meanwhile, the level of oxidative stress in d-galactose mice or inflammatory factors in cortex and serum in lipopolysaccharide mice, which might be involved in the cognitive dysfunctions, was significantly amplified. Furthermore, the impaired memory and hippocampal neuron damage induced by d-galactose and lipopolysaccharide were concurrently aggravated. Collectively, our study provided novel and compelling evidence that the structural and functional proteins for BBB integrity may be the primary targets for the detrimental effects of alcohol abuse that lead to cognitive dysfunction and neurological deficits in high risk populations.
Collapse
Affiliation(s)
- Jiangping Wei
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China
| | - Yuan Dai
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China; School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China
| | - Wen Wen
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China
| | - Jin Li
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China; School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China
| | - Lingyu Linda Ye
- Center for Phenomics of Traditional Chinese Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Shijun Xu
- Institute of Material Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, PR China.
| | - Dayue Darrel Duan
- Center for Phenomics of Traditional Chinese Medicine and the Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
41
|
Fang K, Wang Z, Li Z, Wang B, Han G, Cheng Z, Chen Z, Lan C, Zhang Y, Zhao P, Jin X, Liu Y, Bai R. Convolutional neural network for accelerating the computation of the extended Tofts model in dynamic contrast-enhanced magnetic resonance imaging. J Magn Reson Imaging 2021; 53:1898-1910. [PMID: 33382513 DOI: 10.1002/jmri.27495] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 01/09/2023] Open
Abstract
Quantitative physiological parameters can be obtained from nonlinear pharmacokinetic models, such as the extended Tofts (eTofts) model, applied to dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). However, the computation of such nonlinear models is time consuming. The aim of this study was to develop a convolutional neural network (CNN) for accelerating the computation of fitting eTofts model without sacrificing agreement with conventional nonlinear-least-square (NLLS) fitting. This was a retrospective study, which included 13 patients with brain glioma for training (75%) and validation (25%), and 11 patients (three glioma, four brain metastases, and four lymphoma) for testing. CAIPIRINHA-Dixon-TWIST DCE-MRI and double flip angle T1 map acquired at 3 T were used. A CNN with both local pathway and global pathway modules was designed to estimate the eTofts model parameters, the volume transfer constant (Ktrans ), blood volume fraction (vp ), and volume fraction of extracellular extravascular space (ve ), from DCE-MRI data of tumor and normal-appearing voxels. The CNN was trained on mixed dataset consisting of synthetic and patient data. The CNN result and computation speed were compared with NLLS fitting. The robustness to noise variations and generalization to brain metastases and lymphoma data were also evaluated. Statistical tests used were Student's t test on mean absolute error, concordance correlation coefficient (CCC), and normalized root mean squared error. Including global pathway modules in the CNN and training the network with mixed data significantly (p < 0.05) improved the CNN performance. Compared with NLLS fitting, CNN yields an average CCC greater than 0.986 for Ktrans , greater than 0.965 for vp , and greater than 0.948 for ve . The CNN accelerated computation speed approximately 2000 times compared to NLLS, showed robustness to noise (signal-to-noise ratio >34.42 dB), and had no significant (p > 0.21) difference applied to brain metastases and lymphoma data. In conclusion, the proposed CNN to estimate eTofts parameters showed comparable result as NLLS fitting while significantly reducing the computation time. LEVEL OF EVIDENCE: 3 TECHNICAL EFFICACY STAGE: 1.
Collapse
Affiliation(s)
- Ke Fang
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Zejun Wang
- Department of Physical Medicine and Rehabilitation of the Affiliated Sir Run Run Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Zhaoqing Li
- Department of Physical Medicine and Rehabilitation of the Affiliated Sir Run Run Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Bao Wang
- Department of Radiology, Qilu Hospital of Shandong University, Jinan, China
| | - Guangxu Han
- Department of Physical Medicine and Rehabilitation of the Affiliated Sir Run Run Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Zhaowei Cheng
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Zhihong Chen
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Chuanjin Lan
- School of Medicine, Shandong University, Jinan, China
| | - Yi Zhang
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Peng Zhao
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xinyu Jin
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, China
| | - Yingchao Liu
- Department of Neurosurgery, Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ruiliang Bai
- Department of Physical Medicine and Rehabilitation of the Affiliated Sir Run Run Shaw Hospital and Interdisciplinary Institute of Neuroscience and Technology, School of Medicine, Zhejiang University, Hangzhou, China.,Key Laboratory of Biomedical Engineering of Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
42
|
Kontopodis E, Marias K, Manikis GC, Nikiforaki K, Venianaki M, Maris TG, Mastorodemos V, Papadakis GZ, Papadaki E. Extended perfusion protocol for MS lesion quantification. Open Med (Wars) 2020; 15:520-530. [PMID: 33336007 PMCID: PMC7711864 DOI: 10.1515/med-2020-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/09/2020] [Accepted: 03/13/2020] [Indexed: 11/15/2022] Open
Abstract
This study aims to examine a time-extended dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) protocol and report a comparative study with three different pharmacokinetic (PK) models, for accurate determination of subtle blood-brain barrier (BBB) disruption in patients with multiple sclerosis (MS). This time-extended DCE-MRI perfusion protocol, called Snaps, was applied on 24 active demyelinating lesions of 12 MS patients. Statistical analysis was performed for both protocols through three different PK models. The Snaps protocol achieved triple the window time of perfusion observation by extending the magnetic resonance acquisition time by less than 2 min on average for all patients. In addition, the statistical analysis in terms of adj-R 2 goodness of fit demonstrated that the Snaps protocol outperformed the conventional DCE-MRI protocol by detecting 49% more pixels on average. The exclusive pixels identified from the Snaps protocol lie in the low k trans range, potentially reflecting areas with subtle BBB disruption. Finally, the extended Tofts model was found to have the highest fitting accuracy for both analyzed protocols. The previously proposed time-extended DCE protocol, called Snaps, provides additional temporal perfusion information at the expense of a minimal extension of the conventional DCE acquisition time.
Collapse
Affiliation(s)
- Eleftherios Kontopodis
- Foundation for Research and Technology - Hellas, Institute of Computer Science, Computational Bio-Medicine Laboratory, N. Plastira 100, Vassilika Vouton, GR-700 13 Heraklion, Crete, Greece.,Department of Radiology, Medical School, University of Crete, P. O. Box 2208, Heraklion, Crete, Greece
| | - Kostas Marias
- Technological Educational Institute of Crete, Department of Informatics Engineering, Heraklion , Crete, Estavromenos, TK 71410, Greece
| | - Georgios C Manikis
- Foundation for Research and Technology - Hellas, Institute of Computer Science, Computational Bio-Medicine Laboratory, N. Plastira 100, Vassilika Vouton, GR-700 13 Heraklion, Crete, Greece.,Department of Radiology, Medical School, University of Crete, P. O. Box 2208, Heraklion, Crete, Greece
| | - Katerina Nikiforaki
- Foundation for Research and Technology - Hellas, Institute of Computer Science, Computational Bio-Medicine Laboratory, N. Plastira 100, Vassilika Vouton, GR-700 13 Heraklion, Crete, Greece.,Department of Radiology, Medical School, University of Crete, P. O. Box 2208, Heraklion, Crete, Greece
| | - Maria Venianaki
- Science and Technology Park of Crete, Gnosis Data Analysis, N. Plastira 100, Vassilika Vouton, GR-700 13, Heraklion, Greece
| | - Thomas G Maris
- Foundation for Research and Technology - Hellas, Institute of Computer Science, Computational Bio-Medicine Laboratory, N. Plastira 100, Vassilika Vouton, GR-700 13 Heraklion, Crete, Greece.,Department of Radiology, Medical School, University of Crete, P. O. Box 2208, Heraklion, Crete, Greece
| | - Vasileios Mastorodemos
- Department of Neurology, Medical School, University of Crete, P. O. Box 2208, Heraklion, Crete, Greece
| | - Georgios Z Papadakis
- Foundation for Research and Technology - Hellas, Institute of Computer Science, Computational Bio-Medicine Laboratory, N. Plastira 100, Vassilika Vouton, GR-700 13 Heraklion, Crete, Greece.,Department of Radiology, Medical School, University of Crete, P. O. Box 2208, Heraklion, Crete, Greece
| | - Efrosini Papadaki
- Foundation for Research and Technology - Hellas, Institute of Computer Science, Computational Bio-Medicine Laboratory, N. Plastira 100, Vassilika Vouton, GR-700 13 Heraklion, Crete, Greece.,Department of Radiology, Medical School, University of Crete, P. O. Box 2208, Heraklion, Crete, Greece
| |
Collapse
|
43
|
van den Kerkhof M, Voorter PHM, Canjels LPW, de Jong JJA, van Oostenbrugge RJ, Kroon AA, Jansen JFA, Backes WH. Time-efficient measurement of subtle blood-brain barrier leakage using a T 1 mapping MRI protocol at 7 T. Magn Reson Med 2020; 85:2761-2770. [PMID: 33349996 PMCID: PMC7898690 DOI: 10.1002/mrm.28629] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 12/22/2022]
Abstract
Purpose Blood–brain barrier (BBB) disruption is commonly measured with DCE‐MRI using continuous dynamic scanning. For precise measurement of subtle BBB leakage, a long acquisition time (>20 minutes) is required. As extravasation of the contrast agent is slow, discrete sampling at strategic time points might be beneficial, and gains scan time for additional sequences. Here, we aimed to explore the feasibility of a sparsely sampled MRI protocol at 7 T. Methods The scan protocol consisted of a precontrast quantitative T1 measurement, using an MP2RAGE sequence, and after contrast agent injection, a fast‐sampling dynamic gradient‐echo perfusion scan and two postcontrast quantitative T1 measurements were applied. Simulations were conducted to determine the optimal postcontrast sampling time points for measuring subtle BBB leakage. The graphical Patlak approach was used to quantify the leakage rate (Ki) and blood plasma volume (vp) of normal‐appearing white and gray matter. Results The simulations showed that two postcontrast T1 maps are sufficient to detect subtle leakage, and most sensitive when the last T1 map is acquired late, approximately 30 minutes, after contrast agent administration. The in vivo measurements found Ki and vp values in agreement with other studies, and significantly higher values in gray matter compared with white matter (both p = .04). Conclusion The sparsely sampled protocol was demonstrated to be sensitive to quantify subtle BBB leakage, despite using only three T1 maps. Due to the time‐efficiency of this method, it will become more feasible to incorporate BBB leakage measurements in clinical research MRI protocols.
Collapse
Affiliation(s)
- Marieke van den Kerkhof
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Paulien H M Voorter
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Lisanne P W Canjels
- Department of Radiology & 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
| | - Joost J A de Jong
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Robert J van Oostenbrugge
- School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Department of Neurology, Maastricht University Medical Center, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Abraham A Kroon
- Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands.,Department of Internal Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Jacobus F A Jansen
- Department of Radiology & 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
| | - Walter H Backes
- Department of Radiology & Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands.,School for Mental Health and Neuroscience, Maastricht University, Maastricht, the Netherlands.,Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
44
|
Verheggen ICM, de Jong JJA, van Boxtel MPJ, Postma AA, Verhey FRJ, Jansen JFA, Backes WH. Permeability of the windows of the brain: feasibility of dynamic contrast-enhanced MRI of the circumventricular organs. Fluids Barriers CNS 2020; 17:66. [PMID: 33115484 PMCID: PMC7594295 DOI: 10.1186/s12987-020-00228-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 10/17/2020] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Circumventricular organs (CVOs) are small structures without a blood-brain barrier surrounding the brain ventricles that serve homeostasic functions and facilitate communication between the blood, cerebrospinal fluid and brain. Secretory CVOs release peptides and sensory CVOs regulate signal transmission. However, pathogens may enter the brain through the CVOs and trigger neuroinflammation and neurodegeneration. We investigated the feasibility of dynamic contrast-enhanced (DCE) MRI to assess the CVO permeability characteristics in vivo, and expected significant contrast uptake in these regions, due to blood-brain barrier absence. METHODS Twenty healthy, middle-aged to older males underwent brain DCE MRI. Pharmacokinetic modeling was applied to contrast concentration time-courses of CVOs, and in reference to white and gray matter. We investigated whether a significant and positive transfer from blood to brain could be measured in the CVOs, and whether this differed between secretory and sensory CVOs or from normal-appearing brain matter. RESULTS In both the secretory and sensory CVOs, the transfer constants were significantly positive, and all secretory CVOs had significantly higher transfer than each sensory CVO. The transfer constants in both the secretory and sensory CVOs were higher than in the white and gray matter. CONCLUSIONS Current measurements confirm the often-held assumption of highly permeable CVOs, of which the secretory types have the strongest blood-to-brain transfer. The current study suggests that DCE MRI could be a promising technique to further assess the function of the CVOs and how pathogens can potentially enter the brain via these structures. TRIAL REGISTRATION Netherlands Trial Register number: NL6358, date of registration: 2017-03-24.
Collapse
Affiliation(s)
- Inge C M Verheggen
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands.
- Alzheimer Center Limburg, Maastricht, The Netherlands.
| | - Joost J A de Jong
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Martin P J van Boxtel
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Alida A Postma
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Alzheimer Center Limburg, Maastricht, The Netherlands
| | - Jacobus F A Jansen
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Walter H Backes
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- School for Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
45
|
Abstract
The blood-brain barrier (BBB) is the interface between the blood and brain tissue, which regulates the maintenance of homeostasis within the brain. Impaired BBB integrity is increasingly associated with various neurological diseases. To gain a better understanding of the underlying processes involved in BBB breakdown, magnetic resonance imaging (MRI) techniques are highly suitable for noninvasive BBB assessment. Commonly used MRI techniques to assess BBB integrity are dynamic contrast-enhanced and dynamic susceptibility contrast MRI, both relying on leakage of gadolinium-based contrast agents. A number of conceptually different methods exist that target other aspects of the BBB. These alternative techniques make use of endogenous markers, such as water and glucose, as contrast media. A comprehensive overview of currently available MRI techniques to assess the BBB condition is provided from a scientific point of view, including potential applications in disease. Improvements that are required to make these techniques clinically more easily applicable will also be discussed.
Collapse
|
46
|
Montagne A, Huuskonen MT, Rajagopal G, Sweeney MD, Nation DA, Sepehrband F, D'Orazio LM, Harrington MG, Chui HC, Law M, Toga AW, Zlokovic BV. Undetectable gadolinium brain retention in individuals with an age-dependent blood-brain barrier breakdown in the hippocampus and mild cognitive impairment. Alzheimers Dement 2020; 15:1568-1575. [PMID: 31862169 DOI: 10.1016/j.jalz.2019.07.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 07/02/2019] [Accepted: 07/14/2019] [Indexed: 01/02/2023]
Abstract
INTRODUCTION Blood-brain barrier (BBB) breakdown is an early independent biomarker of human cognitive dysfunction, as found using gadolinium (Gd) as a contrast agent. Whether Gd accumulates in brains of individuals with an age-dependent BBB breakdown and/or mild cognitive impairment remains unclear. METHODS We analyzed T1-weighted magnetic resonance imaging (MRI) scans from 52 older participants with BBB breakdown in the hippocampus 19-28 months after either cyclic or linear Gd agent. RESULTS There was no change in T1-weighted signal intensity between the baseline contrast MRI and unenhanced MRI on re-examination in any of the studied 10 brain regions with either Gd agent suggesting undetectable Gd brain retention. DISCUSSION Gd does not accumulate in brains of older individuals with a BBB breakdown in the hippocampus. Thus, Gd agents can be used without risk of brain retention within a ∼2-year follow-up to study BBB in the aging human brain in relation to cognition and/or other pathologies.
Collapse
Affiliation(s)
- Axel Montagne
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mikko T Huuskonen
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Gautham Rajagopal
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Melanie D Sweeney
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Farshid Sepehrband
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lina M D'Orazio
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Helena C Chui
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Meng Law
- Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Alzheimer's Disease Research Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
47
|
Verheggen ICM, de Jong JJA, van Boxtel MPJ, Gronenschild EHBM, Palm WM, Postma AA, Jansen JFA, Verhey FRJ, Backes WH. Increase in blood-brain barrier leakage in healthy, older adults. GeroScience 2020; 42:1183-1193. [PMID: 32601792 PMCID: PMC7394987 DOI: 10.1007/s11357-020-00211-2] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/02/2020] [Indexed: 12/22/2022] Open
Abstract
Blood-brain barrier (BBB) breakdown can disrupt nutrient supply and waste removal, which affects neuronal functioning. Currently, dynamic contrast-enhanced (DCE) MRI is the preferred in-vivo method to quantify BBB leakage. Dedicated DCE MRI studies in normal aging individuals are lacking, which could hamper value estimation and interpretation of leakage rate in pathological conditions. Therefore, we applied DCE MRI to investigate the association between BBB disruption and age in a healthy sample. Fifty-seven cognitively and neurologically healthy, middle-aged to older participants (mean age: 66 years, range: 47-91 years) underwent MRI, including DCE MRI with intravenous injection of a gadolinium-based contrast agent. Pharmacokinetic modeling was applied to contrast concentration time-curves to estimate BBB leakage rate in each voxel. Subsequently, leakage rate was calculated in the white and gray matter, and primary (basic sensory and motor functions), secondary (association areas), and tertiary (higher-order cognition) brain regions. A difference in vulnerability to deterioration was expected between these regions, with especially tertiary regions being affected by age. Higher BBB leakage rate was significantly associated with older age in the white and gray matter, and also in tertiary, but not in primary or secondary brain regions. Even in healthy individuals, BBB disruption was stronger in older persons, which suggests BBB disruption is a normal physiologically aging phenomenon. Age-related increase in BBB disruption occurred especially in brain regions most vulnerable to age-related deterioration, which may indicate that BBB disruption is an underlying mechanism of normal age-related decline.Netherlands Trial Register number: NL6358, date of registration: 2017-03-24.
Collapse
Affiliation(s)
- Inge C M Verheggen
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands.
| | - Joost J A de Jong
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Martin P J van Boxtel
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Ed H B M Gronenschild
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Walter M Palm
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Alida A Postma
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Jacobus F A Jansen
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Frans R J Verhey
- Department of Psychiatry and Neuropsychology, Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Alzheimer Center Limburg, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
| | - Walter H Backes
- School for Mental Health and Neuroscience (MHeNs), Maastricht University, P.O. Box 616, 6200 MD, Maastricht, The Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
48
|
Naganawa S, Ito R, Kato Y, Kawai H, Taoka T, Yoshida T, Maruyama K, Murata K, Körzdörfer G, Pfeuffer J, Nittka M, Sone M. Intracranial Distribution of Intravenously Administered Gadolinium-based Contrast Agent over a Period of 24 Hours: Evaluation with 3D-real IR Imaging and MR Fingerprinting. Magn Reson Med Sci 2020; 20:91-98. [PMID: 32295977 PMCID: PMC7952208 DOI: 10.2463/mrms.mp.2020-0030] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE To evaluate the feasibility for the detection of slight contrast effects after intravenous administration of single dose gadolinium-based contrast agent (IV-SD-GBCA), the time course of the GBCA distribution up to 24 h was examined in various fluid spaces and brain parenchyma using 3D-real IR imaging and MR fingerprinting (MRF). METHODS Twenty-four patients with a suspicion of endolymphatic hydrops were scanned at pre-administration and at 10 min, 4 and 24 h post-IV-SD-GBCA. 3D-real IR images and MRF at the level of the internal auditory canal were obtained. The signal intensity on the 3D-real IR image of the cerebrospinal fluid (CSF) in the cerebellopontine angle cistern (CPA), Sylvian fissure (Syl), lateral ventricle (LV), and cochlear perilymph (CPL) was measured. The T1 and T2 values of cerebellar gray (GM) and white matter (WM) were measured using MRF. Each averaged value at the various time points was compared using an analysis of variance. RESULTS The signal intensity on the 3D-real IR image in each CSF region peaked at 4 h, and was decreased significantly by 24 h (P< 0.05). All patients had a maximum signal intensity at 4 h in the CPA, and Syl. The mean CPL signal intensity peaked at 4 h and decreased significantly by 24 h (P < 0.05). All patients but two had a maximum signal intensity at 4 h. Regarding the T1 value in the cerebellar WM and GM, the T1 value at 10 min post-IV-GBCA was significantly decreased compared to the pre-contrast scan, but no significant difference was observed at the other time points. There was no significant change in T2 in the gray or white matter at any of the time points. CONCLUSION Time course of GBCA after IV-SD-GBCA could be evaluated by 3D-real IR imaging in CSF spaces and in the brain by MRF.
Collapse
Affiliation(s)
- Shinji Naganawa
- Department of Radiology, Nagoya University Graduate School of Medicine
| | - Rintaro Ito
- Department of Radiology, Nagoya University Graduate School of Medicine
| | - Yutaka Kato
- Department of Radiology, Nagoya University Graduate School of Medicine
| | - Hisashi Kawai
- Department of Radiology, Nagoya University Graduate School of Medicine
| | - Toshiaki Taoka
- Department of Radiology, Nagoya University Graduate School of Medicine
| | - Tadao Yoshida
- Department of Otorhinolaryngology, Nagoya University Graduate School of Medicine
| | | | | | | | | | | | - Michihiko Sone
- Department of Otorhinolaryngology, Nagoya University Graduate School of Medicine
| |
Collapse
|
49
|
Hui L, Soliman ML, Geiger NH, Miller NM, Afghah Z, Lakpa KL, Chen X, Geiger JD. Acidifying Endolysosomes Prevented Low-Density Lipoprotein-Induced Amyloidogenesis. J Alzheimers Dis 2020; 67:393-410. [PMID: 30594929 DOI: 10.3233/jad-180941] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cholesterol dyshomeostasis has been linked to the pathogenesis of sporadic Alzheimer's disease (AD). In furthering the understanding of mechanisms by which increased levels of circulating cholesterol augments the risk of developing sporadic AD, others and we have reported that low-density lipoprotein (LDL) enters brain parenchyma by disrupting the blood-brain barrier and that endolysosome de-acidification plays a role in LDL-induced amyloidogenesis in neurons. Here, we tested the hypothesis that endolysosome de-acidification was central to amyloid-β (Aβ) generation and that acidifying endolysosomes protects against LDL-induced increases in Aβ levels in neurons. We demonstrated that LDL, but not HDL, de-acidified endolysosomes and increased intraneuronal and secreted levels of Aβ. ML-SA1, an agonist of endolysosome-resident TRPML1 channels, acidified endolysosomes, and TRPML1 knockdown attenuated ML-SA1-induced endolysosome acidification. ML-SA1 blocked LDL-induced increases in intraneuronal and secreted levels of Aβ as well as Aβ accumulation in endolysosomes, prevented BACE1 accumulation in endolysosomes, and decreased BACE1 activity levels. LDL downregulated TRPML1 protein levels, and TRPML1 knockdown worsens LDL-induced increases in Aβ. Our findings suggest that endolysosome acidification by activating TRPML1 may represent a protective strategy against sporadic AD.
Collapse
Affiliation(s)
- Liang Hui
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Mahmoud L Soliman
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicholas H Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Nicole M Miller
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Zahra Afghah
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Koffi L Lakpa
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
50
|
Klohs J. An Integrated View on Vascular Dysfunction in Alzheimer's Disease. NEURODEGENER DIS 2020; 19:109-127. [PMID: 32062666 DOI: 10.1159/000505625] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/23/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Cerebrovascular disease is a common comorbidity in patients with Alzheimer's disease (AD). It is believed to contribute additively to the cognitive impairment and to lower the threshold for the development of dementia. However, accumulating evidence suggests that dysfunction of the cerebral vasculature and AD neuropathology interact in multiple ways. Vascular processes even proceed AD neuropathology, implicating a causal role in the etiology of AD. Thus, the review aims to provide an integrated view on vascular dysfunction in AD. SUMMARY In AD, the cerebral vasculature undergoes pronounced cellular, morphological and structural changes, which alters regulation of blood flow, vascular fluid dynamics and vessel integrity. Stiffening of central blood vessels lead to transmission of excessive pulsatile energy to the brain microvasculature, causing end-organ damage. Moreover, a dysregulated hemostasis and chronic vascular inflammation further impede vascular function, where its mediators interact synergistically. Changes of the cerebral vasculature are triggered and driven by systemic vascular abnormalities that are part of aging, and which can be accelerated and aggravated by cardiovascular diseases. Key Messages: In AD, the cerebral vasculature is the locus where multiple pathogenic processes converge and contribute to cognitive impairment. Understanding the molecular mechanism and pathophysiology of vascular dysfunction in AD and use of vascular blood-based and imaging biomarker in clinical studies may hold promise for future prevention and therapy of the disease.
Collapse
Affiliation(s)
- Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland, .,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland,
| |
Collapse
|