51
|
Toniolo S, Di Lorenzo F, Bernardini S, Mercuri NB, Sancesario GM. Blood-Brain Barrier Dysfunction and Aβ42/40 Ratio Dose-Dependent Modulation with the ApoE Genotype within the ATN Framework. Int J Mol Sci 2023; 24:12151. [PMID: 37569528 PMCID: PMC10418506 DOI: 10.3390/ijms241512151] [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] [Received: 06/08/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 08/13/2023] Open
Abstract
The definition of Alzheimer's disease (AD) now considers the presence of the markers of amyloid (A), tau deposition (T), and neurodegeneration (N) essential for diagnosis. AD patients have been reported to have increased blood-brain barrier (BBB) dysfunction, but that has not been tested within the ATN framework so far. As the field is moving towards the use of blood-based biomarkers, the relationship between BBB disruption and AD-specific biomarkers requires considerable attention. Moreover, other factors have been previously implicated in modulating BBB permeability, including age, gender, and ApoE status. A total of 172 cognitively impaired individuals underwent cerebrospinal fluid (CSF) analysis for AD biomarkers, and data on BBB dysfunction, demographics, and ApoE status were collected. Our data showed that there was no difference in BBB dysfunction across different ATN subtypes, and that BBB damage was not correlated with cognitive impairment. However, patients with BBB disruption, if measured with a high Qalb, had low Aβ40 levels. ApoE status did not affect BBB function but had a dose-dependent effect on the Aβ42/40 ratio. These results might highlight the importance of understanding dynamic changes across the BBB in future studies in patients with AD.
Collapse
Affiliation(s)
- Sofia Toniolo
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford OX1 3AZ, UK
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
| | - Francesco Di Lorenzo
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
- Non-Invasive Brain Simulation Unit, IRCSS Santa Lucia Foundation, 00179 Rome, Italy
| | - Sergio Bernardini
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
| | - Giulia Maria Sancesario
- Department of Systems Medicine, University of Rome ‘Tor Vergata’, 00133 Rome, Italy (G.M.S.)
- Biobank Unit, IRCSS Santa Lucia Foundation, 00179 Rome, Italy
| |
Collapse
|
52
|
Abstract
Pericytes are specialized cells located in close proximity to endothelial cells within the microvasculature. They play a crucial role in regulating blood flow, stabilizing vessel walls, and maintaining the integrity of the blood-brain barrier. The loss of pericytes has been associated with the development and progression of various diseases, such as diabetes, Alzheimer's disease, sepsis, stroke, and traumatic brain injury. This review examines the detection of pericyte loss in different diseases, explores the methods employed to assess pericyte coverage, and elucidates the potential mechanisms contributing to pericyte loss in these pathological conditions. Additionally, current therapeutic strategies targeting pericytes are discussed, along with potential future interventions aimed at preserving pericyte function and promoting disease mitigation.
Collapse
Affiliation(s)
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC 29425, USA;
| |
Collapse
|
53
|
Mekli K, Lophatananon A, Maharani A, Nazroo JY, Muir KR. Association between an inflammatory biomarker score and future dementia diagnosis in the population-based UK Biobank cohort of 500,000 people. PLoS One 2023; 18:e0288045. [PMID: 37467176 DOI: 10.1371/journal.pone.0288045] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
This study was designed to investigate the relationship between a systematic inflammatory biomarker measure, concurrent and later cognitive performance, and future dementia risk. The literature has reported the potential involvement of inflammation in cognitive performance as well as Alzheimer's Disease, but not consistently. We used a population-based cohort of 500,000 people in the UK and assessed the association between a composite inflammatory biomarker and cognitive performance measures across five domains measured concurrently and 4-13 years later, taking advantage of the large sample size. We also assessed the same biomarker's association with dementia diagnosis 3-11 years later in the initially dementia-free sample. We report small but significant associations between elevated biomarker levels and worsened cognitive performance at baseline for four cognitive tasks (OR = 1.204, p<0.001 for Prospective memory, β = -0.366, p<0.001 for Fluid intelligence, β = 8.819, p<0.001 for Reaction time, and β = -0.224, p<0.001 for Numeric memory), comparing the highest quartile of the biomarker to the lowest. We also found that for one measure (Pairs matching) higher biomarker levels were associated with fewer errors, i.e. better performance (β = -0.096, p<0.001). We also report that the 4th quartiles of the baseline biomarker levels were significantly associated with cognitive task scores assessed years later on the p< = 0.002 level, except for the Pair matching test, for which none of the quartiles remained a significant predictor. Finally, the highest biomarker quartile was significantly associated with increased dementia risk compared to the lowest quartile (HR = 1.349, p<0.001). A case-only analysis to assess disease subtype heterogeneity suggested probable differences in the association with the highest biomarker quartile between vascular dementia and Alzheimer disease subtypes (OR = 1.483, p = 0.055). Our results indicate that systemic inflammation may play a small but significant part in dementia pathophysiology, especially in vascular dementia.
Collapse
Affiliation(s)
- Krisztina Mekli
- Cathie Marsh Institute and Sociology, The University of Manchester, Manchester, United Kingdom
| | - Artitaya Lophatananon
- Division of Population Health, Health Services Research and Primary Care, The University of Manchester, Manchester, United Kingdom
| | - Asri Maharani
- Department of Nursing, Manchester Metropolitan University, Manchester, United Kingdom
| | - James Y Nazroo
- Cathie Marsh Institute and Sociology, The University of Manchester, Manchester, United Kingdom
| | - Kenneth R Muir
- Division of Population Health, Health Services Research and Primary Care, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
54
|
Musaeus CS, Gleerup HS, Hasselbalch SG, Waldemar G, Simonsen AH. Progression of Blood-Brain Barrier Leakage in Patients with Alzheimer's Disease as Measured with the Cerebrospinal Fluid/Plasma Albumin Ratio Over Time. J Alzheimers Dis Rep 2023; 7:535-541. [PMID: 37313491 PMCID: PMC10259070 DOI: 10.3233/adr-230016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/23/2023] [Indexed: 06/15/2023] Open
Abstract
Background Studies have found a disruption of the blood-brain barrier (BBB) in patients with Alzheimer's disease (AD), but there is little evidence of the changes in the BBB over time. The cerebrospinal fluid's (CSF) protein concentration can be used as an indirect measurement for the permeability of the BBB using the CSF/plasma albumin quotient (Q-Alb) or total CSF protein. Objective In the current study, we wanted to investigate the changes in Q-Alb in patients with AD over time. Methods A total of 16 patients diagnosed with AD, who had at least two lumbar punctures performed, were included in the current study. Results The difference in Q-Alb over time did not show a significant change. However, Q-Alb increased over time if the time interval was > 1 year between the measurements. No significant associations between Q-Alb and age, Mini-Mental State Examination, or AD biomarkers were found. Conclusion The increase in Q-Alb suggests that there is an increased leakage through the BBB, which may become more prominent as the disease progresses. This may be a sign of progressive underlying vascular pathology, even in patients with AD without major vascular lesions. More studies are needed to further understand the role of BBB integrity in patients with AD over time and the association with the progression of the disease.
Collapse
Affiliation(s)
- Christian Sandøe Musaeus
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Helena Sophia Gleerup
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| | - Steen Gregers Hasselbalch
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Gunhild Waldemar
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anja Hviid Simonsen
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital – Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
55
|
Li Z, Jiang Y, Long C, Peng Q, Yue R. The gut microbiota-astrocyte axis: Implications for type 2 diabetic cognitive dysfunction. CNS Neurosci Ther 2023; 29 Suppl 1:59-73. [PMID: 36601656 PMCID: PMC10314112 DOI: 10.1111/cns.14077] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/20/2022] [Accepted: 12/18/2022] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Diabetic cognitive dysfunction (DCD) is one of the most insidious complications of type 2 diabetes mellitus, which can seriously affect the ability to self-monitoring of blood glucose and the quality of life in the elderly. Previous pathological studies of cognitive dysfunction have focused on neuronal dysfunction, characterized by extracellular beta-amyloid deposition and intracellular tau hyperphosphorylation. In recent years, astrocytes have been recognized as a potential therapeutic target for cognitive dysfunction and important participants in the central control of metabolism. The disorder of gut microbiota and their metabolites have been linked to a series of metabolic diseases such as diabetes mellitus. The imbalance of intestinal flora has the effect of promoting the occurrence and deterioration of several diabetes-related complications. Gut microbes and their metabolites can drive astrocyte activation. AIMS We reviewed the pathological progress of DCD related to the "gut microbiota-astrocyte" axis in terms of peripheral and central inflammation, intestinal and blood-brain barrier (BBB) dysfunction, systemic and brain energy metabolism disorders to deepen the pathological research progress of DCD and explore the potential therapeutic targets. CONCLUSION "Gut microbiota-astrocyte" axis, unique bidirectional crosstalk in the brain-gut axis, mediates the intermediate pathological process of neurocognitive dysfunction secondary to metabolic disorders in diabetes mellitus.
Collapse
Affiliation(s)
- Zi‐Han Li
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ya‐Yi Jiang
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Cai‐Yi Long
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Qian Peng
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| | - Ren‐Song Yue
- Hospital of Chengdu University of Traditional Chinese MedicineChengduChina
| |
Collapse
|
56
|
Dai C, Tan C, Zhao L, Liang Y, Liu G, Liu H, Zhong Y, Liu Z, Mo L, Liu X, Chen L. Glucose Metabolism Impairment in Parkinson's Disease. Brain Res Bull 2023; 199:110672. [PMID: 37210012 DOI: 10.1016/j.brainresbull.2023.110672] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 04/19/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
Impairments in systematic and regional glucose metabolism exist in patients with Parkinson's disease (PD) at every stage of the disease course, and such impairments are associated with the incidence, progression, and special phenotypes of PD, which affect each physiological process of glucose metabolism including glucose uptake, glycolysis, tricarboxylic acid cycle, oxidative phosphorylation, and pentose phosphate shunt pathway. These impairments may be attributed to various mechanisms, such as insulin resistance, oxidative stress, abnormal glycated modification, blood-brain-barrier dysfunction, and hyperglycemia-induced damages. These mechanisms could subsequently cause excessive methylglyoxal and reactive oxygen species production, neuroinflammation, abnormal aggregation of protein, mitochondrial dysfunction, and decreased dopamine, and finally result in energy supply insufficiency, neurotransmitter dysregulation, aggregation and phosphorylation of α-synuclein, and dopaminergic neuron loss. This review discusses the glucose metabolism impairment in PD and its pathophysiological mechanisms, and briefly summarized the currently-available therapies targeting glucose metabolism impairment in PD, including glucagon-likepeptide-1 (GLP-1) receptor agonists and dual GLP-1/gastric inhibitory peptide receptor agonists, metformin, and thiazoledinediones.
Collapse
Affiliation(s)
- Chengcheng Dai
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Changhong Tan
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Lili Zhao
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Yi Liang
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Guohui Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Hang Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Yuke Zhong
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Zhihui Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Lijuan Mo
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Xi Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| | - Lifen Chen
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, 74 Linjiang Road, Yuzhong District, Chongqing, 400010, China.
| |
Collapse
|
57
|
Hillmer L, Erhardt EB, Caprihan A, Adair JC, Knoefel JE, Prestopnik J, Thompson J, Hobson S, Rosenberg GA. Blood-brain barrier disruption measured by albumin index correlates with inflammatory fluid biomarkers. J Cereb Blood Flow Metab 2023; 43:712-721. [PMID: 36522849 PMCID: PMC10108191 DOI: 10.1177/0271678x221146127] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022]
Abstract
Blood-brain barrier (BBB) permeability can be measured by the ratio of albumin in cerebrospinal fluid (CSF) and blood and by dynamic contrast-enhanced MRI (DCEMRI). Albumin is a large molecule measured in CSF and blood to form the albumin index (Qalb), which is a global measure of BBB permeability, while the smaller Gadolinium molecule measures regional transfer (Ktrans); few studies have directly compared them in the same patients. We used both methods as part of a study of mechanisms of white matter injury in patients with different forms of dementia. In addition, we also measured biomarkers for inflammation, including proteases, angiogenic growth factors, and cytokines, and correlated them with the BBB results. We found that there was no correlation between Qalb and Ktrans. The Qalb was associated with the matrix metalloproteinases (MMP-2, MMP-3, and MMP-10), the angiogenic factors (VEGF-C and PlGF), and the cytokines (IL-6, IL-8 and TNF-α). On the other hand, Ktrans was associated with the diffusion measures, mean free water and PSMD, which indicate white matter injury. Our results show that the Qalb and Ktrans measure different aspects of BBB permeability, with albumin being a measure of inflammatory BBB opening and Ktrans indicating white matter injury.
Collapse
Affiliation(s)
- Laura Hillmer
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Erik B Erhardt
- Department of Mathematics and
Statistics, University of New Mexico, Albuquerque, New Mexico
| | | | - John C Adair
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
- Department of Neurology, University
of New Mexico, Albuquerque, New Mexico
| | - Janice E Knoefel
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
- Department of Neurology, University
of New Mexico, Albuquerque, New Mexico
| | - Jill Prestopnik
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Jeffrey Thompson
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Sasha Hobson
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
| | - Gary A Rosenberg
- Center for Memory and Aging,
University of New Mexico, Albuquerque, New Mexico
- Department of Neurology, University
of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
58
|
Custodia A, Aramburu-Núñez M, Rodríguez-Arrizabalaga M, Pías-Peleteiro JM, Vázquez-Vázquez L, Camino-Castiñeiras J, Aldrey JM, Castillo J, Ouro A, Sobrino T, Romaus-Sanjurjo D. Biomarkers Assessing Endothelial Dysfunction in Alzheimer's Disease. Cells 2023; 12:cells12060962. [PMID: 36980302 PMCID: PMC10047803 DOI: 10.3390/cells12060962] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023] Open
Abstract
Alzheimer's disease (AD) is the most common degenerative disorder in the elderly in developed countries. Currently, growing evidence is pointing at endothelial dysfunction as a key player in the cognitive decline course of AD. As a main component of the blood-brain barrier (BBB), the dysfunction of endothelial cells driven by vascular risk factors associated with AD allows the passage of toxic substances to the cerebral parenchyma, producing chronic hypoperfusion that eventually causes an inflammatory and neurotoxic response. In this process, the levels of several biomarkers are disrupted, such as an increase in adhesion molecules that allow the passage of leukocytes to the cerebral parenchyma, increasing the permeability of the BBB; moreover, other vascular players, including endothelin-1, also mediate artery inflammation. As a consequence of the disruption of the BBB, a progressive neuroinflammatory response is produced that, added to the astrogliosis, eventually triggers neuronal degeneration (possibly responsible for cognitive deterioration). Recently, new molecules have been proposed as early biomarkers for endothelial dysfunction that can constitute new therapeutic targets as well as early diagnostic and prognostic markers for AD.
Collapse
Affiliation(s)
- Antía Custodia
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marta Aramburu-Núñez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mariña Rodríguez-Arrizabalaga
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Juan Manuel Pías-Peleteiro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Laura Vázquez-Vázquez
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Camino-Castiñeiras
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Manuel Aldrey
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Castillo
- Neuroimaging and Biotechnology Laboratory (NOBEL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
| | - Alberto Ouro
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Tomás Sobrino
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Daniel Romaus-Sanjurjo
- NeuroAging Group (NEURAL), Clinical Neurosciences Research Laboratory (LINC), Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
59
|
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: 2.0] [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
|
60
|
Samir M, Abdelkader RM, Boushehri MS, Mansour S, Lamprecht A, Tammam SN. Enhancement of mitochondrial function using NO releasing nanoparticles; a potential approach for therapy of Alzheimer's disease. Eur J Pharm Biopharm 2023; 184:16-24. [PMID: 36640916 DOI: 10.1016/j.ejpb.2023.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
Alzheimer's disease (AD) is the most common type of dementia. Increasing evidence is showing the important role of mitochondrial dysfunction in AD. Mitochondria based oxidative stress, decrease in respiratory chain activity and ATP production are all associated with AD, hence indicating that the enhancement of mitochondrial function and biogenesis present a promising therapeutic approach for AD. Nitric oxide (NO) is an initiator of mitochondrial biogenesis. However, its gaseous nature and very short half-life limit the realization of its therapeutic potential. Additionally, its uncontrolled in-vivo distribution results in generalized vasodilation, hypotension among other off-target effects. Diazeniumdiolates (NONOates) are NO donors that release NO in physiological temperature and pH. Their encapsulation within a hydrophobic matrix carrier system could control the release of NO, and at the same time enable its delivery to the brain. In this work, PAPANONOate (PN) a NO donor was encapsulated in small (92 ± 7 nm) poly (lactic-co-glycolic acid) (PLGA) NPs. These NPs did not induce hemolysis upon intravenous administration and were able to accumulate in the brains of lipopolysaccharides (LPS) induced neurodegeneration mouse models. The encapsulation of PN within a hydrophobic PLGA matrix enabled the sustained release of NO from NPs (≈ 3 folds slower relative to free PN) and successfully delivered PN to brain. As a result, PN-NPs but not free PN resulted in an enhancement in memory and cognition in animals with neurodegeneration as determined by the Y-maze test. The enhancement in cognition was a result of increased mitochondria function as indicated by the increased production of ATP and Cytochrome C oxidase enzyme activity.
Collapse
Affiliation(s)
- Mirna Samir
- Department of Pharmaceutical Technology, German University in Cairo (GUC), Egypt
| | - Reham M Abdelkader
- Department of Pharmacology, Toxicology and German University in Cairo (GUC), Egypt
| | - Maryam Shetab Boushehri
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Samar Mansour
- Department of Pharmaceutical Technology, German University in Cairo (GUC), Egypt; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Egypt
| | - Alf Lamprecht
- Department of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Germany; Laboratory of Pharmaceutical Engineering (EA4267), University of Franche-Comté, Besançon, France
| | - Salma N Tammam
- Department of Pharmaceutical Technology, German University in Cairo (GUC), Egypt.
| |
Collapse
|
61
|
Roseborough AD, Zhu Y, Zhao L, Laviolette SR, Pasternak SH, Whitehead SN. Fibrinogen primes the microglial NLRP3 inflammasome and propagates pro-inflammatory signaling via extracellular vesicles: Implications for blood-brain barrier dysfunction. Neurobiol Dis 2023; 177:106001. [PMID: 36646389 DOI: 10.1016/j.nbd.2023.106001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023] Open
Abstract
The brain's response to acute injury is characterized by increased permeability of the blood-brain barrier (BBB) and pro-inflammatory microglia signaling, both of which have been linked to poor cognitive outcomes and neurological disease. The damaged BBB has increased leakiness, allowing serum proteins like fibrinogen into the brain, which interacts with local cells in a deleterious manner. At the same time, in response to injury, microglia demonstrate increased NLRP3 inflammasome activity and heightened release of pro-inflammatory cytokines. The relationship between increased fibrinogen uptake and microglial inflammasome signaling in the injured brain has not been well described. In this work, we investigate fibrinogen mediated NLRP3 inflammasome priming of BV-2 cells and primary adult microglia and propose a role for extracellular vesicles (EVs) as propagators of this interaction. Following exposure to fibrinogen microglia significantly upregulate transcription of IL-1β, IL-6, NLRP3 and other pro-inflammatory cytokines which was sustained by repeated fibrinogen exposure. Inhibition of fibrinogen mediated NLRP3 signaling was achieved at the transcriptional and assembly level using cannabidiol (CBD) and the NLRP3 inhibitor MCC950, respectively. EVs released following NLRP3 priming carry IL-1β, IL-18 mRNA and fibrinogen, propagate inflammatory signaling and can be detected in the circulation following BBB disruption in a preclinical stroke model. In conclusion, the interplay between fibrinogen extravasation, microglial NLRP3 signaling, and EV release can perpetuate chronic pro-inflammatory signaling and represents a novel method of inflammatory propagation.
Collapse
Affiliation(s)
- A D Roseborough
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - Y Zhu
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - L Zhao
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - S R Laviolette
- Addictions Research Group, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada; Department of Psychiatry, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - S H Pasternak
- Department of Clinical Neurological Sciences, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada; Robarts Research Institute, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada
| | - S N Whitehead
- Vulnerable Brain Laboratory, Department of Anatomy and Cell Biology, The Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
62
|
Giacopuzzi Grigoli E, Solca F, Milone I, Aiello EN, Dubini A, Ratti A, Torresani E, Poletti B, Ticozzi N, Ciusani E, Silani V, Verde F. Cerebrospinal fluid/serum albumin quotient (Q-Alb) is not increased in Alzheimer's disease compared to neurological disease controls: a retrospective study on 276 patients. Neurol Sci 2023; 44:709-713. [PMID: 36441343 DOI: 10.1007/s10072-022-06530-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND The cerebrospinal fluid (CSF)/serum albumin quotient (Q-Alb) is a marker of the blood-CSF barrier (BCSFB) and possibly of the blood-brain barrier (BBB). The latter is known to be altered in Alzheimer's disease (AD) based on neuropathological and neuroimaging studies. Following investigations performed on clinically diagnosed cohorts, we aimed at comparing Q-Alb in cognitively impaired patients with neurochemical demonstration of AD pathophysiology and neurological disease controls (NDCs). METHODS We evaluated N = 144 AD patients (MCI, N = 43; AD dementia - ADD, N = 101) and N = 132 NDCs. AD patients were all A + according to the A/T/N framework and were neurochemically classified based on T and N parameters. RESULTS Q-Alb did not significantly differ between AD patients and NDCs. Moreover, it was not associated with disease stage (MCI vs. ADD), MMSE score, or CSF AD biomarkers. DISCUSSION Our study indicates that BCSFB dysfunction is not a specific feature of AD. When interpreting Q-Alb as a marker of the BBB, the lack of difference from NDCs might be due to BBB dysfunction widely occurring in other neurological, non-degenerative, conditions or - more probably - to low sensitivity of this biochemical parameter towards subtle BBB alterations causing leakage of molecules smaller than albumin. Furthermore, Q-Alb is not associated with the degree of global cognitive deterioration in AD, nor with CSF AD neurochemical biomarkers.
Collapse
Affiliation(s)
- Eleonora Giacopuzzi Grigoli
- Neurology Residency Program, Università Degli Studi Di Milano, Milan, Italy.,Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Federica Solca
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Ilaria Milone
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Edoardo Nicolò Aiello
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Ph.D. Program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Antonella Dubini
- Department of Laboratory Medicine, Laboratory of Clinical Chemistry, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Medical Biotechnology and Translational Medicine, Università Degli Studi Di Milano, Milan, Italy
| | - Erminio Torresani
- Department of Laboratory Medicine, Laboratory of Clinical Chemistry, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Barbara Poletti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Nicola Ticozzi
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università Degli Studi Di Milano, Milan, Italy
| | - Emilio Ciusani
- Laboratory of Neurological Biochemistry and Neuropharmacology, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Vincenzo Silani
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università Degli Studi Di Milano, Milan, Italy
| | - Federico Verde
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy. .,Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università Degli Studi Di Milano, Milan, Italy.
| |
Collapse
|
63
|
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
|
64
|
Gan J, Yang X, Zhang G, Li X, Liu S, Zhang W, Ji Y. Alzheimer's disease pathology: pathways between chronic vascular risk factors and blood-brain barrier dysfunction in a cohort of patients with different types of dementia. Front Aging Neurosci 2023; 15:1088140. [PMID: 37213537 PMCID: PMC10194826 DOI: 10.3389/fnagi.2023.1088140] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 04/10/2023] [Indexed: 05/23/2023] Open
Abstract
Background Blood brain barrier (BBB) breakdown is considered a potential mechanism of dementia. The Alzheimer's disease (AD) biomarkers and vascular factors are also associated with BBB permeability. Objective In the present study, the combination effects of neuropathological biomarkers of AD and chronic vascular risk factors for BBB were investigated. Methods The cerebrospinal fluid (CSF)/serum albumin ratio (Qalb), an indicator of BBB permeability, was measured in a total of 95 hospitalized dementia patients. The demographics, clinical information, and laboratory tests were collected from the inpatient records. The CSF neuropathological biomarkers of AD and apolipoprotein E (APOE) genotype were also collected. The mediation analysis model was used to calculate the associations among neuropathological biomarkers of AD (mediator), the Qalb, and chronic vascular risk factors. Results Three types of dementia, AD (n = 52), Lewy body dementia (LBD, n = 19), and frontotemporal lobar degeneration (n = 24), were included with a mean Qalb of 7.18 (± 4.36). The Qalb was significantly higher in dementia patients with type 2 diabetes mellitus (T2DM, p = 0.004) but did not differ based on the presence of APOE ε4 allele, CMBs, or amyloid/tau/neurodegeneration (ATN) framework. The Qalb was negatively associated with the levels of Aβ1-42 (B = -20.775, p = 0.009) and Aβ1-40 (B = -305.417, p = 0.005) and positively associated with the presence of T2DM (B = 3.382, p < 0.001) and the levels of glycosylated hemoglobin (GHb, B = 1.163, p < 0.001) and fasting blood glucose (FBG, B = 1.443, p < 0.001). GHb is a direct chronic vascular risk factor for higher Qalb (total effect B = 1.135, 95% CI: 0.611-1.659, p < 0.001). Ratios of Aβ1-42/Aβ1-40 or t-tau/Aβ1-42 were mediators of the association between the Qalb and GHb; the direct effect of GHb on the Qalb was 1.178 (95% CI: 0.662-1.694, p < 0.001). Conclusion Glucose exposure can directly or indirectly affect BBB integrity through Aβ and tau, indicating glucose affects BBB breakdown and glucose stability plays an important role in dementia protection and management.
Collapse
Affiliation(s)
- Jinghuan Gan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xia Yang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guili Zhang
- Department of Cognitive Disorder, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xudong Li
- Department of Cognitive Disorder, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shuai Liu
- Department of Neurology, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
| | - Wei Zhang
- Department of Cognitive Disorder, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yong Ji
- Department of Neurology, Tianjin Dementia Institute, Tianjin Key Laboratory of Cerebrovascular and Neurodegenerative Diseases, Tianjin Huanhu Hospital, Tianjin, China
- *Correspondence: Yong Ji
| |
Collapse
|
65
|
Khan SR, Yaqub A, Ikram MK, van Hagen PM, Peeters RP, Dalm VASH, Chaker L, Ikram MA. The association of serum immunoglobulins with cognition and dementia: the Rotterdam Study. J Neurol 2023; 270:423-432. [PMID: 36123443 DOI: 10.1007/s00415-022-11374-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/06/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Chronic inflammation is involved in the pathophysiology of dementia, but the association of serum immunoglobulins with dementia has been understudied and longitudinal data are currently lacking. We investigated the association of serum immunoglobulin (Ig) A, G, and M with cognition and dementia in a population-based cohort. METHODS This study was embedded in the Rotterdam Study. Participants with information on serum immunoglobulin levels, measured between 1997 and 2009, were followed for incident dementia until 2016. Assessment of cognitive function and dementia was performed according to validated tests and clinical criteria respectively. We studied the association between serum immunoglobulins with prevalent and incident dementia using logistic regression and Cox proportional hazards regression analyses respectively. We performed linear regression analyses to quantify the cross-sectional association of serum immunoglobulins with global cognition as well as separate cognitive tests. Analyses were adjusted for age, sex, lifestyle, and cardiovascular factors. RESULTS We included 8768 participants (median age of 62.2 years, 57% women, median follow-up 10.7 years). Overall, none of the immunoglobulins was associated with prevalent or incident dementia. Higher IgG levels were associated with lower scores of global cognition (adjusted standardized mean difference - 0.04; 95% confidence interval:- 0.06; - 0.02) and separate cognitive tests. CONCLUSION In middle-aged and older individuals from the general population, serum Igs were not associated with prevalent or incident dementia, which may imply that serum Igs are not involved in the pathophysiology of dementia. Although higher IgG levels were associated with worse cognitive function, studies with longitudinal data should exclude reverse causation.
Collapse
Affiliation(s)
- Samer R Khan
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Division of Allergy and Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Amber Yaqub
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - M Kamran Ikram
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Department of Neurology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - P Martin van Hagen
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robin P Peeters
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Virgil A S H Dalm
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Layal Chaker
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.,Division of Endocrinology, Department of Internal Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center Rotterdam, PO Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
66
|
Dong H, Guo L, Yang H, Zhu W, Liu F, Xie Y, Zhang Y, Xue K, Li Q, Liang M, Zhang N, Qin W. Association between gray matter atrophy, cerebral hypoperfusion, and cognitive impairment in Alzheimer's disease. Front Aging Neurosci 2023; 15:1129051. [PMID: 37091519 PMCID: PMC10117777 DOI: 10.3389/fnagi.2023.1129051] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/15/2023] [Indexed: 04/25/2023] Open
Abstract
Background Alzheimer's disease (AD) is one of the most severe neurodegenerative diseases leading to dementia in the elderly. Cerebral atrophy and hypoperfusion are two important pathophysiological characteristics. However, it is still unknown about the area-specific causal pathways between regional gray matter atrophy, cerebral hypoperfusion, and cognitive impairment in AD patients. Method Forty-two qualified AD patients and 49 healthy controls (HC) were recruited in this study. First, we explored voxel-wise inter-group differences in gray matter volume (GMV) and arterial spin labeling (ASL) -derived cerebral blood flow (CBF). Then we explored the voxel-wise associations between GMV and Mini-Mental State Examination (MMSE) score, GMV and CBF, and CBF and MMSE to identify brain targets contributing to cognitive impairment in AD patients. Finally, a mediation analysis was applied to test the causal pathways among atrophied GMV, hypoperfusion, and cognitive impairment in AD. Results Voxel-wise permutation test identified that the left middle temporal gyrus (MTG) had both decreased GMV and CBF in the AD. Moreover, the GMV of this region was positively correlated with MMSE and its CBF, and CBF of this region was also positively correlated with MMSE in AD (p < 0.05, corrected). Finally, mediation analysis revealed that gray matter atrophy of left MTG drives cognitive impairment of AD via the mediation of CBF (proportion of mediation = 55.82%, β = 0.242, 95% confidence interval by bias-corrected and accelerated bootstrap: 0.082 to 0.530). Conclusion Our findings indicated suggested that left MTG is an important hub linking gray matter atrophy, hypoperfusion, and cognitive impairment for AD, and might be a potential treatment target for AD.
Collapse
Affiliation(s)
- Haoyang Dong
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Lining Guo
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Hailei Yang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenshuang Zhu
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Fang Liu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingying Xie
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Yu Zhang
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Kaizhong Xue
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Li
- Technical College for the Deaf, Tianjin University of Technology, Tianjin, China
| | - Meng Liang
- School of Medical Imaging, Tianjin Medical University, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Nan Zhang,
| | - Wen Qin
- Department of Radiology and Tianjin Key Laboratory of Functional Imaging, Tianjin Medical University General Hospital, Tianjin, China
- Wen Qin,
| |
Collapse
|
67
|
Kantor AB, Akassoglou K, Stavenhagen JB. Fibrin-Targeting Immunotherapy for Dementia. J Prev Alzheimers Dis 2023; 10:647-660. [PMID: 37874085 PMCID: PMC11227370 DOI: 10.14283/jpad.2023.105] [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: 10/25/2023]
Abstract
Blood-brain barrier (BBB) disruption is an early event in the development of Alzheimer's disease. It precedes extracellular deposition of amyloid-β in senile plaques and blood vessel walls, the intracellular accumulation of neurofibrillary tangles containing phosphorylated tau protein, microglial activation, and neuronal cell death. BBB disruption allows the coagulation protein fibrinogen to leak from the blood into the brain, where it is converted by thrombin cleavage into fibrin and deposits in the parenchyma and CNS vessels. Fibrinogen cleavage by thrombin exposes a cryptic epitope termed P2 which can bind CD11b and CD11c on microglia, macrophages and dendritic cells and trigger an inflammatory response toxic to neurons. Indeed, genetic and pharmacological evidence demonstrates a causal role for fibrin in innate immune cell activation and the development of neurodegenerative diseases. The P2 inflammatory epitope is spatially and compositionally distinct from the coagulation epitope on fibrin. Mouse monoclonal antibody 5B8, which targets the P2 epitope without interfering with the clotting process, has been shown to reduce neurodegeneration and neuroinflammation in animal models of Alzheimer's disease and multiple sclerosis. The selectivity and efficacy of this anti-human fibrin-P2 antibody in animal models supports the development of a monoclonal antibody drug targeting fibrin P2 for the treatment of neurodegenerative diseases. THN391 is a humanized, affinity-matured antibody which has a 100-fold greater affinity for fibrin P2 and improved development properties compared to the parental 5B8 antibody. It is currently in a Phase 1 clinical trial.
Collapse
Affiliation(s)
- A B Kantor
- Jeffrey Stavenhagen, PhD, Therini Bio, Inc, Sacramento, CA, USA,
| | | | | |
Collapse
|
68
|
Chong JR, Hilal S, Ashton NJ, Karikari TK, Reilhac A, Vrooman H, Schöll M, Zetterberg H, Blennow K, Chen CP, Lai MKP. Brain atrophy and white matter hyperintensities are independently associated with plasma neurofilament light chain in an Asian cohort of cognitively impaired patients with concomitant cerebral small vessel disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2023; 15:e12396. [PMID: 36994314 PMCID: PMC10040495 DOI: 10.1002/dad2.12396] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 03/28/2023]
Abstract
Introduction Plasma neurofilament light chain (NfL) is a potential biomarker for neurodegeneration in Alzheimer's disease (AD), ischemic stroke, and non-dementia cohorts with cerebral small vessel disease (CSVD). However, studies of AD in populations with high prevalence of concomitant CSVD to evaluate associations of brain atrophy, CSVD, and amyloid beta (Aβ) burden on plasma NfL are lacking. Methods Associations were tested between plasma NfL and brain Aβ, medial temporal lobe atrophy (MTA) as well as neuroimaging features of CSVD, including white matter hyperintensities (WMH), lacunes, and cerebral microbleeds. Results We found that participants with either MTA (defined as MTA score ≥2; neurodegeneration [N]+WMH-) or WMH (cut-off for log-transformed WMH volume at 50th percentile; N-WMH+) manifested increased plasma NfL levels. Participants with both pathologies (N+WMH+) showed the highest NfL compared to N+WMH-, N-WMH+, and N-WMH- individuals. Discussion Plasma NfL has potential utility in stratifying individual and combined contributions of AD pathology and CSVD to cognitive impairment.
Collapse
Affiliation(s)
- Joyce R. Chong
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| | - Saima Hilal
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
- Saw Swee Hock School of Public HealthNational University of Singapore and National University Health SystemKent RidgeSingapore
| | - Nicholas J. Ashton
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- King's College LondonInstitute of PsychiatryPsychology and NeuroscienceMaurice Wohl Institute Clinical Neuroscience InstituteLondonUK
- NIHR Biomedical Research Centre for Mental Health and Biomedical Research Unit for Dementia at South London and Maudsley NHS FoundationLondonUK
| | - Thomas K. Karikari
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of PsychiatryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Anthonin Reilhac
- Clinical Imaging Research CentreYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
| | - Henri Vrooman
- Department of Radiology and Nuclear MedicineErasmus Medical CenterRotterdamthe Netherlands
| | - Michael Schöll
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Wallenberg Centre for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
| | - Henrik Zetterberg
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Department of Neurodegenerative DiseaseUCL Queen Square Institute of NeurologyLondonUK
- UK Dementia Research Institute at UCLLondonUK
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
- Hong Kong Center for Neurodegenerative Diseasesthe Hong Kong University of Science and TechnologyHong Kong Science ParkShatinNew TerritoriesHong Kong SARChina
| | - Kaj Blennow
- Department of Psychiatry and NeurochemistryInstitute of Neuroscience and Physiologythe Sahlgrenska Academy at the University of GothenburgGothenburgSweden
- Clinical Neurochemistry LaboratorySahlgrenska University HospitalGothenburgSweden
| | - Christopher P. Chen
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
- Department of Psychological MedicineYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
| | - Mitchell K. P. Lai
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeKent RidgeSingapore
- Memory, Aging and Cognition CentreNational University Health SystemsKent RidgeSingapore
| |
Collapse
|
69
|
Cabral DF, Bigliassi M, Cattaneo G, Rundek T, Pascual-Leone A, Cahalin LP, Gomes-Osman J. Exploring the interplay between mechanisms of neuroplasticity and cardiovascular health in aging adults: A multiple linear regression analysis study. Auton Neurosci 2022; 242:103023. [PMID: 36087362 PMCID: PMC11012134 DOI: 10.1016/j.autneu.2022.103023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 06/13/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Neuroplasticity and cardiovascular health behavior are critically important factors for optimal brain health. OBJECTIVE To assess the association between the efficacy of the mechanisms of neuroplasticity and metrics of cardiovascular heath in sedentary aging adults. METHODS We included thirty sedentary individuals (age = 60.6 ± 3.8 y; 63 % female). All underwent assessments of neuroplasticity, measured by the change in amplitude of motor evoked potentials elicited by single-pulse Transcranial Magnetic Stimulation (TMS) at baseline and following intermittent Theta-Burst (iTBS) at regular intervals. Cardiovascular health measures were derived from the Incremental Shuttle Walking Test and included Heart Rate Recovery (HRR) at 1-min/2-min after test cessation. We also collected plasma levels of brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), and c-reactive protein. RESULTS We revealed moderate but significant relationships between TMS-iTBS neuroplasticity, and the predictors of cardiovascular health (|r| = 0.38 to 0.53, p < .05). HRR1 was the best predictor of neuroplasticity (β = 0.019, p = .002). The best fit model (Likelihood ratio = 5.83, p = .016) of the association between neuroplasticity and HRR1 (β = 0.043, p = .002) was selected when controlling for demographics and health status. VEGF and BDNF plasma levels augmented the association between neuroplasticity and HRR1. CONCLUSIONS Our findings build on existing data demonstrating that TMS may provide insight into neuroplasticity and the role cardiovascular health have on its mechanisms. These implications serve as theoretical framework for future longitudinal and interventional studies aiming to improve cardiovascular and brain health. HRR1 is a potential prognostic measure of cardiovascular health and a surrogate marker of brain health in aging adults.
Collapse
Affiliation(s)
- Danylo F Cabral
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, USA.
| | - Marcelo Bigliassi
- Department of Teaching and Learning, Florida International University, Miami, FL, USA
| | - Gabriele Cattaneo
- Institut Guttmann, Institut Universitari de Neurorehabilitació, Badalona, Spain; Department of Medicine, Universitat Autónoma de Barcelona, Bellaterra, Spain
| | - Tatjana Rundek
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA; Evelyn F. McKnight Brain Institute, University of Miami, Miami, FL, USA
| | - Alvaro Pascual-Leone
- Institut Guttmann, Institut Universitari de Neurorehabilitació, Badalona, Spain; Hinda and Arthur Marcus Institute for Aging Research and Deanna and Sidney Wolk Center for Memory Health, Hebrew SeniorLife, Boston, MA, USA; Department of Neurology, Harvard Medical School, Boston, MA, USA
| | - Lawrence P Cahalin
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, USA
| | - Joyce Gomes-Osman
- Department of Physical Therapy, University of Miami Miller School of Medicine, Coral Gables, FL, USA; Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
70
|
Wen S, Elias PM, Wakefield JS, Mauro TM, Man MQ. The link between cutaneous inflammation and cognitive impairment. J Eur Acad Dermatol Venereol 2022; 36:1705-1712. [PMID: 35748522 PMCID: PMC9481668 DOI: 10.1111/jdv.18360] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/30/2022] [Indexed: 12/01/2022]
Abstract
Cognitive impairment is a symptom of neurological disorders, including dementia and Alzheimer's disease; and mild cognitive impairment can be a precursor of both disorders. Aged humans and animal models with other systemic disorders, such as cardiovascular diseases and diabetes, display a higher incidence of cognitive decline. Epidemiological studies have shown that the incidence of cognitive impairment also is higher in subjects with certain inflammatory skin disorders, including psoriasis and chronic eczematous dermatitis. Chronologically aged individuals exhibit increased cutaneous inflammation and elevated circulating cytokine levels, linked to alterations in epidermal function, which itself can induce cutaneous inflammation. Conversely, strategies that improve epidermal function can lower cytokine levels in both the skin and circulation. Thus, it seems likely that epidermal dysfunction could contribute, at least in part, to the development of chronic low-grade inflammation, also termed 'inflammaging', in the elderly. The evidence of cognitive impairment in patients with inflammatory dermatoses suggests a link between cutaneous inflammation and cognitive impairment. Because of the pathogenic role of epidermal dysfunction in ageing-associated cutaneous inflammation, improvements in epidermal function could be an alternative approach for mitigation of the ageing-associated decline in cognitive function.
Collapse
Affiliation(s)
- S Wen
- Dermatology Hospital, Southern Medical University, Guangdong, China
| | - P M Elias
- Dermatology Service, Veterans Affairs Medical Center San Francisco, San Francisco, California, USA
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - J S Wakefield
- Dermatology Service, Veterans Affairs Medical Center San Francisco, San Francisco, California, USA
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - T M Mauro
- Dermatology Service, Veterans Affairs Medical Center San Francisco, San Francisco, California, USA
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| | - M-Q Man
- Dermatology Hospital, Southern Medical University, Guangdong, China
- Dermatology Service, Veterans Affairs Medical Center San Francisco, San Francisco, California, USA
- Department of Dermatology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
71
|
Zheng H, Wang T, Shi C, Fan L, Su Y, Fan Y, Li X, Yang J, Mao C, Xu Y. Increased PRR14 and VCAM-1 level in serum of patients with Parkinson's disease. Front Neurol 2022; 13:993940. [PMID: 36247752 PMCID: PMC9561935 DOI: 10.3389/fneur.2022.993940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022] Open
Abstract
Background Regarding the complexity of Parkinson's disease (PD), the identification of reliable biomarkers is of great significance for improving the accuracy of diagnosis and monitoring disease progression. Recently, some studies suggested that serum proline-rich protein 14 (PRR14), vascular cell adhesion molecule-1 (VCAM-1), and soluble CD163 (sCD163) factors may be associated with PD, even as potential biomarkers. However, the role of these serum factors is still unclear. Objectives This study aimed to explore the alterations of serum PRR14, VCAM-1, and sCD163 levels during PD progression, and their association with disease-related variables of PD. Methods We performed the assessment of scale tests and the detection of serum samples in patients with PD (n = 100) and healthy controls (HCs, n = 100). Furthermore, we investigated the association between serum factors and sex, cognitive impairments, H&Y (Hohn and Yahr), age at onset (AAO), and other variables in patients with PD. Results Patients with PD exhibited increased PRR14 and VCAM-1 serum levels compared with HCs. No significant differences were found in serum levels of sCD163. Subgroup analysis uncovered increased VCAM-1 in the female and male subgroups (PD and HCs). Among patients with PD, decreased PRR14 and increased VCAM-1 were associated with severer cognitive impairments and severer PD (H&Y), respectively. Bivariate correlation analysis revealed that there was a positive correlation between VCAM-1 and AAO. Conclusions Increased serum levels of PRR14 and VCAM-1 suggest that inflammation and defective autophagy may play vital roles in the pathogenesis of PD. However, the potential mechanisms remain to be elucidated.
Collapse
Affiliation(s)
- Huimin Zheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Tai Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Department of Neurology, Nanyang Central Hospital, Nanyang, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Liyuan Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yun Su
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Yu Fan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Xinwei Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
| | - Chengyuan Mao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- The Academy of Medical Sciences of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- *Correspondence: Chengyuan Mao
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- Institute of Neuroscience, Zhengzhou University, Zhengzhou, China
- Yuming Xu
| |
Collapse
|
72
|
Britton R, Liu AT, Rege SV, Adams JM, Akrapongpisak L, Le D, Alcantara-Lee R, Estrada RA, Ray R, Ahadi S, Gallager I, Yang CF, Minami SS, Braithwaite SP, Czirr E, Campbell MK. Molecular and histological correlates of cognitive decline across age in male C57BL/6J mice. Brain Behav 2022; 12:e2736. [PMID: 35971662 PMCID: PMC9480918 DOI: 10.1002/brb3.2736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 07/01/2022] [Accepted: 07/20/2022] [Indexed: 11/09/2022] Open
Abstract
INTRODUCTION Increasing age is the number one risk factor for developing cognitive decline and neurodegenerative disease. Aged humans and mice exhibit numerous molecular changes that contribute to a decline in cognitive function and increased risk of developing age-associated diseases. Here, we characterize multiple age-associated changes in male C57BL/6J mice to understand the translational utility of mouse aging. METHODS Male C57BL/6J mice from various ages between 2 and 24 months of age were used to assess behavioral, as well as, histological and molecular changes across three modalities: neuronal, microgliosis/neuroinflammation, and the neurovascular unit (NVU). Additionally, a cohort of 4- and 22-month-old mice was used to assess blood-brain barrier (BBB) breakdown. Mice in this cohort were treated with a high, acute dose of lipopolysaccharide (LPS, 10 mg/kg) or saline control 6 h prior to sacrifice followed by tail vein injection of 0.4 kDa sodium fluorescein (100 mg/kg) 2 h later. RESULTS Aged mice showed a decline in cognitive and motor abilities alongside decreased neurogenesis, proliferation, and synapse density. Further, neuroinflammation and circulating proinflammatory cytokines were increased in aged mice. Additionally, we found changes at the BBB, including increased T cell infiltration in multiple brain regions and an exacerbation in BBB leakiness following chemical insult with age. There were also a number of readouts that were unchanged with age and have limited utility as markers of aging in male C57BL/6J mice. CONCLUSIONS Here we propose that these changes may be used as molecular and histological readouts that correspond to aging-related behavioral decline. These comprehensive findings, in the context of the published literature, are an important resource toward deepening our understanding of normal aging and provide an important tool for studying aging in mice.
Collapse
Affiliation(s)
| | - Angela T Liu
- Alkahest, Inc., San Carlos, California, USA.,Coda Biotherapeutics, South San Francisco, California, USA
| | | | | | - Lily Akrapongpisak
- Alkahest, Inc., San Carlos, California, USA.,University of Queensland, Herston, Queensland, Australia
| | - David Le
- Alkahest, Inc., San Carlos, California, USA.,Fountain Therapeutics, South San Francisco, California, USA
| | | | | | - Rebecca Ray
- Alkahest, Inc., San Carlos, California, USA.,202 Chives Way, Walnut Creek, California, USA
| | - Sara Ahadi
- Alkahest, Inc., San Carlos, California, USA
| | | | | | | | | | - Eva Czirr
- Alkahest, Inc., San Carlos, California, USA.,Confluence Therapeutics, South San Francisco, California, USA
| | | |
Collapse
|
73
|
Miedema SSM, Mol MO, Koopmans FTW, Hondius DC, van Nierop P, Menden K, de Veij Mestdagh CF, van Rooij J, Ganz AB, Paliukhovich I, Melhem S, Li KW, Holstege H, Rizzu P, van Kesteren RE, van Swieten JC, Heutink P, Smit AB. Distinct cell type-specific protein signatures in GRN and MAPT genetic subtypes of frontotemporal dementia. Acta Neuropathol Commun 2022; 10:100. [PMID: 35799292 PMCID: PMC9261008 DOI: 10.1186/s40478-022-01387-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/22/2022] [Indexed: 11/16/2022] Open
Abstract
Frontotemporal dementia is characterized by progressive atrophy of frontal and/or temporal cortices at an early age of onset. The disorder shows considerable clinical, pathological, and genetic heterogeneity. Here we investigated the proteomic signatures of frontal and temporal cortex from brains with frontotemporal dementia due to GRN and MAPT mutations to identify the key cell types and molecular pathways in their pathophysiology. We compared patients with mutations in the GRN gene (n = 9) or with mutations in the MAPT gene (n = 13) with non-demented controls (n = 11). Using quantitative proteomic analysis on laser-dissected tissues we identified brain region-specific protein signatures for both genetic subtypes. Using published single cell RNA expression data resources we deduced the involvement of major brain cell types in driving these different protein signatures. Subsequent gene ontology analysis identified distinct genetic subtype- and cell type-specific biological processes. For the GRN subtype, we observed a distinct role for immune processes related to endothelial cells and for mitochondrial dysregulation in neurons. For the MAPT subtype, we observed distinct involvement of dysregulated RNA processing, oligodendrocyte dysfunction, and axonal impairments. Comparison with an in-house protein signature of Alzheimer’s disease brains indicated that the observed alterations in RNA processing and oligodendrocyte function are distinct for the frontotemporal dementia MAPT subtype. Taken together, our results indicate the involvement of different brain cell types and biological mechanisms in genetic subtypes of frontotemporal dementia. Furthermore, we demonstrate that comparison of proteomic profiles of different disease entities can separate general neurodegenerative processes from disease-specific pathways, which may aid the development of disease subtype-specific treatment strategies.
Collapse
Affiliation(s)
- Suzanne S M Miedema
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands.
| | - Merel O Mol
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Frank T W Koopmans
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - David C Hondius
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Pim van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Kevin Menden
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany
| | - Christina F de Veij Mestdagh
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands.,Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, the Netherlands.,Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Jeroen van Rooij
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andrea B Ganz
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands.,Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Iryna Paliukhovich
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Shamiram Melhem
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Ka Wan Li
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - Henne Holstege
- Alzheimer Center, Department of Neurology, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Genetics, Amsterdam Neuroscience, VU University Medical Center, Amsterdam, The Netherlands
| | - Patrizia Rizzu
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany
| | - Ronald E van Kesteren
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| | - John C van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter Heutink
- German Center for Neurodegenerative Diseases (DZNE)-Tübingen, Tübingen, Germany
| | - August B Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, W&N Building, C314. De Boelelaan 1105, 1081 HV, Amsterdam, The Netherlands
| |
Collapse
|
74
|
Zhao Y, Haney MJ, Fallon JK, Rodriguez M, Swain CJ, Arzt CJ, Smith PC, Loop MS, Harrison EB, El-Hage N, Batrakova EV. Using Extracellular Vesicles Released by GDNF-Transfected Macrophages for Therapy of Parkinson Disease. Cells 2022; 11:1933. [PMID: 35741061 PMCID: PMC9222008 DOI: 10.3390/cells11121933] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 02/04/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that facilitate transport of proteins, lipids, and genetic material, playing important roles in intracellular communication. They have remarkable potential as non-toxic and non-immunogenic nanocarriers for drug delivery to unreachable organs and tissues, in particular, the central nervous system (CNS). Herein, we developed a novel platform based on macrophage-derived EVs to treat Parkinson disease (PD). Specifically, we evaluated the therapeutic potential of EVs secreted by autologous macrophages that were transfected ex vivo to express glial-cell-line-derived neurotrophic factor (GDNF). EV-GDNF were collected from conditioned media of GDNF-transfected macrophages and characterized for GDNF content, size, charge, and expression of EV-specific proteins. The data revealed that, along with the encoded neurotrophic factor, EVs released by pre-transfected macrophages carry GDNF-encoding DNA. Four-month-old transgenic Parkin Q311(X)A mice were treated with EV-GDNF via intranasal administration, and the effect of this therapeutic intervention on locomotor functions was assessed over a year. Significant improvements in mobility, increases in neuronal survival, and decreases in neuroinflammation were found in PD mice treated with EV-GDNF. No offsite toxicity caused by EV-GDNF administration was detected. Overall, an EV-based approach can provide a versatile and potent therapeutic intervention for PD.
Collapse
Affiliation(s)
- Yuling Zhao
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (M.J.H.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Matthew J. Haney
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (M.J.H.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - John K. Fallon
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Myosotys Rodriguez
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (N.E.-H.)
| | - Carson J. Swain
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Camryn J. Arzt
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Philip C. Smith
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Matthew Shane Loop
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Emily B. Harrison
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Nazira El-Hage
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (N.E.-H.)
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (M.J.H.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| |
Collapse
|
75
|
Jeong JH, Lee DH, Song J. HMGB1 signaling pathway in diabetes-related dementia: Blood-brain barrier breakdown, brain insulin resistance, and Aβ accumulation. Biomed Pharmacother 2022; 150:112933. [PMID: 35413600 DOI: 10.1016/j.biopha.2022.112933] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 11/28/2022] Open
Abstract
Diabetes contributes to the onset of various diseases, including cancer and cardiovascular and neurodegenerative diseases. Recent studies have highlighted the similarities and relationship between diabetes and dementia as an important issue for treating diabetes-related cognitive deficits. Diabetes-related dementia exhibits several features, including blood-brain barrier disruption, brain insulin resistance, and Aβ over-accumulation. High-mobility group box1 (HMGB1) is a protein known to regulate gene transcription and cellular mechanisms by binding to DNA or chromatin via receptor for advanced glycation end-products (RAGE) and toll-like receptor 4 (TLR4). Recent studies have demonstrated that the interplay between HMGB1, RAGE, and TLR4 can impact both neuropathology and diabetic alterations. Herein, we review the recent research regarding the roles of HMGB1-RAGE-TLR4 axis in diabetes-related dementia from several perspectives and emphasize the importance of the influence of HMGB1 in diabetes-related dementia.
Collapse
Affiliation(s)
- Jae-Ho Jeong
- Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| | - Dong Hoon Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Medical School, and Chonnam National University Hwasun Hospital, Hwasun 58128, Jeollanam-do, Republic of Korea.
| | - Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun 58128, Jeollanam-do, Republic of Korea.
| |
Collapse
|
76
|
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: 64] [Impact Index Per Article: 21.3] [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
|
77
|
Swanson RL, Acharya NK, Cifu DX. Cerebral Microvascular Pathology Is a Common Endophenotype Between Traumatic Brain Injury, Cardiovascular Disease, and Dementia: A Hypothesis and Review. Cureus 2022; 14:e25318. [PMID: 35774720 PMCID: PMC9236636 DOI: 10.7759/cureus.25318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 11/05/2022] Open
Abstract
Traumatic brain injury (TBI) exposure has been associated with an increased risk of age-related cognitive decline or dementia in multiple epidemiological studies. Current therapeutic interventions in the field of Brain Injury Medicine focus largely on episodic symptom management during the chronic phase of TBI recovery, rather than targeting specific underlying pathological processes. This approach may be especially problematic for secondary age-related cognitive decline or dementia following TBI exposure. Although there are likely multiple pathophysiological mechanisms involved, a growing body of literature demonstrates that cerebral microvascular pathology is a common endophenotype across the spectrum of TBI severity. Similarly, a combination of pre-clinical and clinical research over the past two decades has implicated cerebral microvascular pathology in the initiation and progression of multiple neurodegenerative diseases, including Alzheimer’s disease and other dementias. We hypothesize that cerebral microvascular pathology is a common endophenotype between TBI, cardiovascular disease (CVD), and dementia, which can be targeted through modifiable cardiovascular risk factor reductions during the chronic phase of TBI recovery. We posit that our hypothesis is supported by the currently available scientific literature, as detailed in our review.
Collapse
|
78
|
de Oliveira TQ, de Moura AC, Feistauer V, Damiani R, Braga MF, Almeida S, Guedes RP, Giovenardi M. Caloric restriction in mice improves short-term recognition memory and modifies the neuroinflammatory response in the hippocampus of male adult offspring. Behav Brain Res 2022; 425:113838. [PMID: 35283195 DOI: 10.1016/j.bbr.2022.113838] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/20/2022]
Abstract
Restrictive diets (RD) can influence the inflammatory phenotype of dams and their offspring. Thus, this study aimed to evaluate the effects of caloric restriction on the neuroinflammatory profile in the hippocampus and the short-term recognition memory of male offspring from RD-fed dams. Mice dams received standard diet ad libitum (CONT) or restrictive diet (RD; 30% reduction of CONT consumption) during pregnancy and lactation. Male pups were weaned at 21 days and randomly divided into two groups that received CONT or RD; groups were named according to maternal/offspring diets: CONT/CONT, CONT/RD, RD/CONT, and RD/RD. At 90 days old, short-term memory was assessed by the object recognition test (ORT); the inflammatory state of the hippocampus was analyzed by gene expression of sirtuin-1 (Sirt1) and inflammasome Nlrp3; and by protein expression of toll-like receptor-4 (TLR-4) and zonula occludens-1 (ZO-1). Our results showed an improvement in short-term memory in RD-fed offspring. The expression of Sirt1 was higher in RD/CONT compared to CONT/CONT and decreased in RD/RD compared to CONT/RD. Nlrp3 gene expression showed an offspring effect, being decreased in RD-fed mice. TLR-4 expression was higher in RD/CONT compared to CONT/CONT, similarly to ZO-1 expression. However, ZO-1 also showed a maternal diet effect and increased expression in the offspring of RD dams. Our findings demonstrate that caloric restriction improved short-term recognition memory. However, a restrictive diet should be applied with caution; depending on the offspring's diet, it may not benefit the neuroinflammatory phenotype or cognition.
Collapse
Affiliation(s)
- Tharcila Quadros de Oliveira
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Ana Carolina de Moura
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Vanessa Feistauer
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Roberto Damiani
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Matheus Filipe Braga
- Acadêmico do Curso de Biomedicina, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul CEP 90050-170, Brazil
| | - Silvana Almeida
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil
| | - Márcia Giovenardi
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Rua Sarmento Leite, 245, Porto Alegre, Rio Grande do Sul, CEP 90050-170, Brazil.
| |
Collapse
|
79
|
Bai T, Yu S, Feng J. Advances in the Role of Endothelial Cells in Cerebral Small Vessel Disease. Front Neurol 2022; 13:861714. [PMID: 35481273 PMCID: PMC9035937 DOI: 10.3389/fneur.2022.861714] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) poses a serious socio-economic burden due to its high prevalence and severe impact on the quality of life of elderly patients. Pathological changes in CSVD mainly influence small cerebral arteries, microarteries, capillaries, and small veins, which are usually caused by multiple vascular risk factors. CSVD is often identified on brain magnetic resonance imaging (MRI) by recent small subcortical infarcts, white matter hyperintensities, lacune, cerebral microbleeds (CMBs), enlarged perivascular spaces (ePVSs), and brain atrophy. Endothelial cell (EC) dysfunction is earlier than clinical symptoms. Immune activation, inflammation, and oxidative stress may be potential mechanisms of EC injury. ECs of the blood–brain–barrier (BBB) are the most important part of the neurovascular unit (NVU) that ensures constant blood flow to the brain. Impaired cerebral vascular autoregulation and disrupted BBB cause cumulative brain damage. This review will focus on the role of EC injury in CSVD. Furthermore, several specific biomarkers will be discussed, which may be useful for us to assess the endothelial dysfunction and explore new therapeutic directions.
Collapse
|
80
|
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: 62] [Impact Index Per Article: 20.7] [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
|
81
|
Ahmad R, Chowdhury K, Kumar S, Irfan M, Reddy GS, Akter F, Jahan D, Haque M. Diabetes Mellitus: A Path to Amnesia, Personality, and Behavior Change. BIOLOGY 2022; 11:biology11030382. [PMID: 35336756 PMCID: PMC8945557 DOI: 10.3390/biology11030382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Diabetes Mellitus (DM) is a metabolic disorder resulting from a disturbance of insulin secretion, action, or both. Hyperglycemia and overproduction of superoxide induce the development and progression of chronic complications of DM. The impact of DM and its complication on the central nervous system (CNS) such as dementia and Alzheimer’s Disease (AD) still remain obscure. In dementia, there is a gradual decline in cognitive function. The incidence of dementia increases with age, and patient become socially, physically, and mentally more vulnerable and dependent. The symptoms often emerge decades after the onset of pathophysiology, thus impairing early therapeutic intervention. Most diabetic subjects who develop dementia are above the age of 65, but diabetes may also cause an increased risk of developing dementia before 65 years. Vascular dementia is the second most common form of dementia after AD. Type 2 DM (T2DM) increases the incidence of vascular dementia (since its covers the vascular system) and AD. The functional and structural integrity of the CNS is altered in T2DM due to increased synthesis of Aβ. Additionally, hyperphosphorylation of Tau protein also results from dysregulation of various signaling cascades in T2DM, thereby causing neuronal damage and AD. There is the prospect for development of a therapy that may help prevent or halt the progress of dementia resulting from T2DM. Abstract Type 2 diabetes mellitus is increasingly being associated with cognition dysfunction. Dementia, including vascular dementia and Alzheimer’s Disease, is being recognized as comorbidities of this metabolic disorder. The progressive hallmarks of this cognitive dysfunction include mild impairment of cognition and cognitive decline. Dementia and mild impairment of cognition appear primarily in older patients. Studies on risk factors, neuropathology, and brain imaging have provided important suggestions for mechanisms that lie behind the development of dementia. It is a significant challenge to understand the disease processes related to diabetes that affect the brain and lead to dementia development. The connection between diabetes mellitus and dysfunction of cognition has been observed in many human and animal studies that have noted that mechanisms related to diabetes mellitus are possibly responsible for aggravating cognitive dysfunction. This article attempts to narrate the possible association between Type 2 diabetes and dementia, reviewing studies that have noted this association in vascular dementia and Alzheimer’s Disease and helping to explain the potential mechanisms behind the disease process. A Google search for “Diabetes Mellitus and Dementia” was carried out. Search was also done for “Diabetes Mellitus”, “Vascular Dementia”, and “Alzheimer’s Disease”. The literature search was done using Google Scholar, Pubmed, Embase, ScienceDirect, and MEDLINE. Keeping in mind the increasing rate of Diabetes Mellitus, it is important to establish the Type 2 diabetes’ effect on the brain and diseases of neurodegeneration. This narrative review aims to build awareness regarding the different types of dementia and their relationship with diabetes.
Collapse
Affiliation(s)
- Rahnuma Ahmad
- Department of Physiology, Medical College for Women and Hospital, Dhaka 1230, Bangladesh;
| | - Kona Chowdhury
- Department of Pediatrics, Gonoshasthaya Samaj Vittik Medical College and Hospital, Dhaka 1344, Bangladesh;
| | - Santosh Kumar
- Department of Periodontology and Implantology, Karnavati School of Dentistry, Karnavati University, 907/A, Uvarsad Gandhinagar, Gujarat 382422, India;
| | - Mohammed Irfan
- Department of Forensics, Federal University of Pelotas, Pelotas 96020-010, RS, Brazil;
| | - Govindool Sharaschandra Reddy
- Department of Periodontics and Endodontics, School of Dental Medicine, University at Buffalo, Buffalo, NY 14214, USA;
| | - Farhana Akter
- Department of Endocrinology, Chittagong Medical College, Chattogram 4203, Bangladesh;
| | - Dilshad Jahan
- Department of Hematology, Asgar Ali Hospital, 111/1/A Distillery Road, Gandaria Beside Dhupkhola, Dhaka 1204, Bangladesh;
| | - Mainul Haque
- Unit of Pharmacology, Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kem Perdana Sungai Besi, Kuala Lumpur 57000, Malaysia
- Correspondence: or
| |
Collapse
|
82
|
Reese CF, Chinnakkannu P, Tourkina E, Hoffman S, Kuppuswamy D. Multiple subregions within the caveolin-1 scaffolding domain inhibit fibrosis, microvascular leakage, and monocyte migration. PLoS One 2022; 17:e0264413. [PMID: 35213624 PMCID: PMC8880820 DOI: 10.1371/journal.pone.0264413] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/10/2022] [Indexed: 12/27/2022] Open
Abstract
The caveolin-1 scaffolding domain (CSD, amino acids 82-101 of caveolin-1) has been shown to suppress bleomycin-induced lung and skin fibrosis and angiotensin II (AngII)-induced myocardial fibrosis. To identify active subregions within CSD, we split its sequence into three slightly overlapping 8-amino acid subregions (82-89, 88-95, and 94-101). Interestingly, all three peptides showed activity. In bleomycin-treated mice, all three subregions suppressed the pathological effects on lung and skin tissue morphology. In addition, while bone marrow monocytes isolated from bleomycin-treated mice showed greatly enhanced migration in vitro toward CXCL12, treatment in vivo with CSD and its subregions almost completely suppressed this enhanced migration. In AngII-induced heart failure, both 82-89 and 88-95 significantly suppressed fibrosis (both Col I and HSP47 levels), microvascular leakage, and heart weight/ body weight ratio (HW/BW) while improving ventricular function. In contrast, while 94-101 suppressed the increase in Col I, it did not improve the other parameters. The idea that all three subregions can be active depending on the assay was further supported by experiments studying the in vitro migration of human monocytes in which all three subregions were extremely active. These studies are very novel in that it has been suggested that there is only one active region within CSD that is centered on amino acids 90-92. In contrast, we demonstrate here the presence of other active regions within CSD.
Collapse
Affiliation(s)
- Charles F. Reese
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Panneerselvam Chinnakkannu
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Elena Tourkina
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Stanley Hoffman
- Division of Rheumatology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| | - Dhandapani Kuppuswamy
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina, Unites States of America
| |
Collapse
|
83
|
Elevated Cerebrospinal Fluid Anti-CD4 Autoantibody Levels in HIV Associate with Neuroinflammation. Microbiol Spectr 2022; 10:e0197521. [PMID: 34985329 PMCID: PMC8729763 DOI: 10.1128/spectrum.01975-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The mechanisms of persistent central nervous system (CNS) inflammation in people with HIV (PWH) despite effective antiretroviral therapy (ART) are not fully understood. We have recently shown that plasma anti-CD4 IgGs contribute to poor CD4+ T cell recovery during suppressive ART via antibody-mediated cytotoxicity (ADCC) against CD4+ T cells, and that plasma anti-CD4 IgG levels are associated with worse cognitive performance and specific brain area atrophy. However, the role of anti-CD4 IgGs in neuroinflammation remains unclear. In the current study, plasma and cerebrospinal fluid (CSF) samples from 31 ART-naive and 26 treated, virologically suppressed PWH, along with 16 HIV-seronegative controls, were evaluated for CSF levels of anti-CD4 IgG, white blood cell (WBC) counts, soluble biomarkers of neuroinflammation, and neurofilament light chain (NfL). We found that 37% of the PWH exhibited elevated CSF anti-CD4 IgG levels, but few or none of the PWH were observed with elevated CSF anti-CD4 IgM, anti-CD8 IgG, or anti-double-strand DNA IgG. CSF anti-CD4 IgG levels in PWH were directly correlated with neuroinflammation (WBC counts, neopterin, and markers of myeloid cell activation), but not with CSF NfL levels. Using cells from one immune nonresponder to ART, we generated a pathogenic anti-CD4 monoclonal IgG (JF19) presenting with ADCC activity; JF19 induced the production of soluble CD14 (sCD14) and interleukin-8 (IL-8) in human primary monocyte-derived macrophages via CD4 binding in vitro. This study demonstrates for the first time that elevated CSF anti-CD4 IgG levels present in a subgroup of PWH which may play a role in neuroinflammation in HIV. IMPORTANCE This study reports that an autoantibody presents in the CNS of HIV patients and that its levels in the CSF correlate with some markers of neuroinflammation.
Collapse
|
84
|
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: 85] [Impact Index Per Article: 28.3] [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
|
85
|
Stringer MS, Heye AK, Armitage PA, Chappell F, Valdés Hernández MDC, Makin SDJ, Sakka E, Thrippleton MJ, Wardlaw JM. Tracer kinetic assessment of blood-brain barrier leakage and blood volume in cerebral small vessel disease: Associations with disease burden and vascular risk factors. Neuroimage Clin 2022; 32:102883. [PMID: 34911189 PMCID: PMC8607271 DOI: 10.1016/j.nicl.2021.102883] [Citation(s) in RCA: 3] [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: 10/20/2021] [Accepted: 11/16/2021] [Indexed: 12/01/2022]
Abstract
Permeability surface area (PS) was higher, even in normal appearing tissue. PS was higher in patients with more white matter hyperintensities. Tissue damage affecting vascular surface area may affect how we interpret tracer kinetic results.
Subtle blood–brain barrier (BBB) permeability increases have been shown in small vessel disease (SVD) using various analysis methods. Following recent consensus recommendations, we used Patlak tracer kinetic analysis, considered optimal in low permeability states, to quantify permeability-surface area product (PS), a BBB leakage estimate, and blood plasma volume (vP) in 201 patients with SVD who underwent dynamic contrast-enhanced MRI scans. We ran multivariable regression models with a quantitative or qualitative metric of white matter hyperintensity (WMH) severity, demographic and vascular risk factors. PS increased with WMH severity in grey (B = 0.15, Confidence Interval (CI): [0.001,0.299], p = 0.049) and normal-appearing white matter (B = 0.015, CI: [−0.008,0.308], p = 0.062). Patients with more severe WMH had lower vP in WMH (B = -0.088, CI: [−0.138,-0.039], p < 0.001), but higher vP in normal-appearing white matter (B = 0.031, CI: [−0.004,0.065], p = 0.082). PS and vP were lower at older ages in WMH, grey and white matter. We conclude higher PS in normal-appearing tissue with more severe WMH suggests impaired BBB integrity beyond visible lesions indicating that the microvasculature is compromised in normal-appearing white matter and WMH. BBB dysfunction is an important mechanism in SVD, but associations with clinical variables are complex and underlying damage affecting vascular surface area may alter interpretation of tracer kinetic results.
Collapse
Affiliation(s)
- Michael S Stringer
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK DRI at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Anna K Heye
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; Edinburgh Clinical Trials Unit, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Paul A Armitage
- Academic Unit of Radiology, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Royal Hallamshire Hospital, Sheffield, UK
| | - Francesca Chappell
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK DRI at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Maria Del C Valdés Hernández
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK DRI at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
| | | | - Eleni Sakka
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK DRI at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
| | - Michael J Thrippleton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK DRI at the University of Edinburgh, University of Edinburgh, Edinburgh, UK.
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK DRI at the University of Edinburgh, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
86
|
Liu L, Wang N, Kalionis B, Xia S, He Q. HMGB1 plays an important role in pyroptosis induced blood brain barrier breakdown in diabetes-associated cognitive decline. J Neuroimmunol 2022; 362:577763. [PMID: 34844084 DOI: 10.1016/j.jneuroim.2021.577763] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 12/13/2022]
Abstract
Diabetes mellitus increases the risk of dementia, and evidence suggests hyperglycemia is a key contributor to neurodegeneration. However, our understanding of diabetes-associated cognitive decline, an important complication of diabetes mellitus, is lacking and the underlying mechanism is unclear. Blood brain barrier (BBB) breakdown is a possible cause of dementia in diabetes mellitus and Alzheimer's disease. Accumulating evidence shows BBB dysfunction caused by hyperglycemia contributes to cognitive decline. A specific type of inflammatory programmed cell death, called pyroptosis, has potential as a therapeutic target for BBB-associated diseases. Potential inducers of pyroptosis include inflammasomes such as NLRP3, whose activation relies on damage-associated molecular patterns. High mobility group box 1 (HMGB1) is a highly conserved, ubiquitous protein found in most cell types, and acts as a damage-associated molecular pattern when released from the nucleus. We propose that HMGB1 influences vascular inflammation by activating the NLRP3 inflammasome and thereby initiating pyroptosis in vascular cells. Moreover, HMGB1 plays a pivotal role in the pathogenesis of diabetes mellitus and diabetic complications. Here, we review the role of HMGB1 in BBB dysfunction induced by hyperglycemia and propose that HMGB1 is a promising therapeutic target for countering diabetes-associated cognitive decline.
Collapse
Affiliation(s)
- Lumei Liu
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Neng Wang
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Bill Kalionis
- Pregnancy Research Centre, Department of Maternal-Fetal Medicine, Royal Women's Hospital, Parkville, Australia; University of Melbourne, Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, Australia
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, PR China.
| | - Qinghu He
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China; Hunan University of Medicine, Huaihua, PR China.
| |
Collapse
|
87
|
Sheikh MH, Errede M, d'Amati A, Khan NQ, Fanti S, Loiola RA, McArthur S, Purvis GSD, O'Riordan CE, Ferorelli D, Dell'Erba A, Kieswich J, Reutelingsperger C, Maiorano E, Yaqoob M, Thiemermann C, Baragetti A, Catapano AL, Norata GD, Marelli-Berg F, Virgintino D, Solito E. Impact of metabolic disorders on the structural, functional, and immunological integrity of the blood-brain barrier: Therapeutic avenues. FASEB J 2022; 36:e22107. [PMID: 34939700 DOI: 10.1096/fj.202101297r] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/04/2021] [Accepted: 12/03/2021] [Indexed: 12/23/2022]
Abstract
Mounting evidence has linked the metabolic disease to neurovascular disorders and cognitive decline. Using a murine model of a high-fat high-sugar diet mimicking obesity-induced type 2 diabetes mellitus (T2DM) in humans, we show that pro-inflammatory mediators and altered immune responses damage the blood-brain barrier (BBB) structure, triggering a proinflammatory metabolic phenotype. We find that disruption to tight junctions and basal lamina due to loss of control in the production of matrix metalloproteinases (MMPs) and their inhibitors (TIMPs) causes BBB impairment. Together the disruption to the structural and functional integrity of the BBB results in enhanced transmigration of leukocytes across the BBB that could contribute to an initiation of a neuroinflammatory response through activation of microglia. Using a humanized in vitro model of the BBB and T2DM patient post-mortem brains, we show the translatable applicability of our results. We find a leaky BBB phenotype in T2DM patients can be attributed to a loss of junctional proteins through changes in inflammatory mediators and MMP/TIMP levels, resulting in increased leukocyte extravasation into the brain parenchyma. We further investigated therapeutic avenues to reduce and restore the BBB damage caused by HFHS-feeding. Pharmacological treatment with recombinant annexin A1 (hrANXA1) or reversion from a high-fat high-sugar diet to a control chow diet (dietary intervention), attenuated T2DM development, reduced inflammation, and restored BBB integrity in the animals. Given the rising incidence of diabetes worldwide, understanding metabolic-disease-associated brain microvessel damage is vital and the proposed therapeutic avenues could help alleviate the burden of these diseases.
Collapse
Affiliation(s)
- Madeeha H Sheikh
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mariella Errede
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari School of Medicine, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari School of Medicine, Bari, Italy.,Department of Emergency and Organ Transplantation, Section of Anatomic Pathology, University of Bari, Bari, Italy
| | - Noorafza Q Khan
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Silvia Fanti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rodrigo A Loiola
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Laboratoire de la Barrière Hémato-Encéphalique, Faculty Jean Perrin, EA 2465, Université d'Artois, Arras, France
| | - Simon McArthur
- Institute of Dentistry, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth S D Purvis
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Caroline E O'Riordan
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Davide Ferorelli
- Department of Interdisciplinary Medicine, Section of Legal Medicine, University of Bari, Bari, Italy
| | - Alessandro Dell'Erba
- Department of Interdisciplinary Medicine, Section of Legal Medicine, University of Bari, Bari, Italy
| | - Julius Kieswich
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Chis Reutelingsperger
- Cardiovascular Research Institute, Maastricht University, Maastricht, The Netherlands
| | - Eugenio Maiorano
- Department of Emergency and Organ Transplantation, Section of Anatomic Pathology, University of Bari, Bari, Italy
| | - Magdi Yaqoob
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Andrea Baragetti
- Department of Pharmacological and Biomolecular Sciences, Milan University, Milan, Italy.,IRCCS Multimedica, Sesto San Giovanni, Italy
| | - Alberico Luigi Catapano
- Department of Pharmacological and Biomolecular Sciences, Milan University, Milan, Italy.,IRCCS Multimedica, Sesto San Giovanni, Italy
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, Milan University, Milan, Italy.,IRCCS Multimedica, Sesto San Giovanni, Italy.,S.I.S.A. Centre for the Study of Atherosclerosis-Bassini Hospital, Cinisello Balsamo, Italy
| | - Federica Marelli-Berg
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari School of Medicine, Bari, Italy
| | - Egle Solito
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Department of Medicina Molecolare e Biotecnologie Mediche, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
88
|
Busse S, von Hoff F, Michler E, Hartig R, Bogerts B, Busse M. Altered expression of costimulatory molecules in dementias. Eur Arch Psychiatry Clin Neurosci 2022; 272:807-815. [PMID: 34427746 PMCID: PMC9279221 DOI: 10.1007/s00406-021-01297-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 07/04/2021] [Indexed: 01/08/2023]
Abstract
Although the expression of co-stimulatory molecules plays an important role in the immune system, only little is known about their regulation in dementias. Therefore, we determined the expression of CD28, ICOS (CD278) and CTLA-4 (CD152) by CD4 + and CD8 + T cells in the peripheral blood of patients with mild cognitive impairment (MCI; N = 19), Alzheimer's disease (AD; N = 51), vascular dementia (VD; N = 21) and frontotemporal dementia (FTD; N = 6) at the point in time of diagnosis compared to 19 non-demented elderly persons. The expression of CD28 and ICOS by CD4 + and CD8 + T cells was not changed in AD, FTD or VD patients. The expression of the negative regulator CTLA-4 was increased by CD4 + T cells from AD and FTD patients and by CD8 + T cells from VD patients. The classification of the AD patients according to the severity of the disorder showed stage-dependent alterations of CD28, ICOS and CTLA-4 expression. In AD patients, the correlation analysis showed an association between the decline in CD28 + T cells and the increase in CTLA-4 + T cells with cognitive decline, measured by the mini-mental state examination (MMSE), tau proteins and Amyloid-β, important AD biomarkers in cerebrospinal fluid (CSF). In FTD patients, a positive association between Q Albumin, a marker for blood-CSF-barrier function, and CD28 and a negative correlation between Q Albumin and ICOS expression were determined. Our data suggest a dysregulated balance between the expression of negative and positive co-stimulatory molecules by T cells in AD patients, which might contribute to chronic inflammation observed in dementia.
Collapse
Affiliation(s)
- Stefan Busse
- Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Franz von Hoff
- Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Enrico Michler
- Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Roland Hartig
- Institute of Immunology, University of Magdeburg, Magdeburg, Germany
| | - Bernhard Bogerts
- Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Mandy Busse
- Department of Psychiatry, University of Magdeburg, Leipziger Str. 44, 39120, Magdeburg, Germany.
- Experimental Obstetrics and Gynecology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
89
|
Frank CJ, McNay EC. Breakdown of the blood-brain barrier: A mediator of increased Alzheimer's risk in patients with metabolic disorders? J Neuroendocrinol 2022; 34:e13074. [PMID: 34904299 PMCID: PMC8791015 DOI: 10.1111/jne.13074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 11/12/2021] [Accepted: 11/26/2021] [Indexed: 01/03/2023]
Abstract
Metabolic disorders (MDs), including type 1 and 2 diabetes and chronic obesity, are among the faster growing diseases globally and are a primary risk factor for Alzheimer's disease (AD). The term "type-3 diabetes" has been proposed for AD due to the interrelated cellular, metabolic, and immune features shared by diabetes, insulin resistance (IR), and the cognitive impairment and neurodegeneration found in AD. Patients with MDs and/or AD commonly exhibit altered glucose homeostasis and IR; systemic chronic inflammation encompassing all of the periphery, blood-brain barrier (BBB), and central nervous system; pathological vascular remodeling; and increased BBB permeability that allows transfusion of neurotoxic molecules from the blood to the brain. This review summarizes the components of the BBB, mechanisms through which MDs alter BBB permeability via immune and metabolic pathways, the contribution of BBB dysfunction to the manifestation and progression of AD, and current avenues of therapeutic research that address BBB permeability. In addition, issues with the translational applicability of current animal models of AD regarding BBB dysfunction and proposals for future directions of research that address the relationship between MDs, BBB dysfunction, and AD are discussed.
Collapse
Affiliation(s)
- Corey J Frank
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| | - Ewan C McNay
- Behavioral Neuroscience, University at Albany, SUNY, Albany, NY, USA
| |
Collapse
|
90
|
Raut S, Patel R, Pervaiz I, Al-Ahmad AJ. Abeta Peptides Disrupt the Barrier Integrity and Glucose Metabolism of Human Induced Pluripotent Stem Cell-Derived Brain Microvascular Endothelial Cells. Neurotoxicology 2022; 89:110-120. [DOI: 10.1016/j.neuro.2022.01.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022]
|
91
|
Lin L, Wu Y, Chen Z, Huang L, Wang L, Liu L. Severe Hypoglycemia Contributing to Cognitive Dysfunction in Diabetic Mice Is Associated With Pericyte and Blood-Brain Barrier Dysfunction. Front Aging Neurosci 2021; 13:775244. [PMID: 34899278 PMCID: PMC8662820 DOI: 10.3389/fnagi.2021.775244] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Severe hypoglycemia can cause cognitive impairment in diabetic patients, but the underlying molecular mechanism remains unclear. Objective: To assess the effect of severe hypoglycemia on cognitive function in diabetic mice to clarify the relationship between the mechanism and dysfunction of pericytes and the blood–brain barrier (BBB). Method: We established type 1 diabetes mellitus in 80 male C57BL/6J mice by intraperitoneal injection of streptozotocin (150 mg/kg). Further intraperitoneal injection of short-acting insulin induced severe hypoglycemia. The mice were divided into normal, diabetes, and diabetic + severe hypoglycemia groups, and their blood glucose and general weight index were examined. Pericyte and BBB morphology and function were detected by histological and western blot analyses, BBB permeability was detected by Evans blue staining, and cognitive function was detected with the Morris water maze. Results: Severe hypoglycemia aggravated the histological damage, BBB damage, brain edema, and pericyte loss in the diabetic mice. It also reduced the expression of the BBB tight junction proteins occludin and claudin-5, the expression of the pericyte-specific markers PDGFR-β (platelet-derived growth factor receptor-β) and α-SMA, and increased the expression of the inflammatory factor MMP9. At the same time, diabetic mice with severe hypoglycemia had significantly reduced cognitive function. Conclusion: Severe hypoglycemia leads to cognitive dysfunction in diabetic mice, and its possible mechanism is related to pericyte dysfunction and BBB destruction.
Collapse
Affiliation(s)
- Lu Lin
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yubin Wu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhou Chen
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Lishan Huang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Lijing Wang
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Libin Liu
- Department of Endocrinology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
92
|
Linton AE, Weekman EM, Wilcock DM. Pathologic sequelae of vascular cognitive impairment and dementia sheds light on potential targets for intervention. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100030. [PMID: 36324710 PMCID: PMC9616287 DOI: 10.1016/j.cccb.2021.100030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/11/2021] [Accepted: 10/08/2021] [Indexed: 11/30/2022]
Abstract
Vascular contributions to cognitive impairment and dementia (VCID) is one of the leading causes of dementia along with Alzheimer's disease (AD) and, importantly, VCID often manifests as a comorbidity of AD(Vemuri and Knopman 2016; Schneider and Bennett 2010)(Vemuri and Knopman 2016; Schneider and Bennett 2010). Despite its common clinical manifestation, the mechanisms underlying VCID disease progression remains elusive. In this review, existing knowledge is used to propose a novel hypothesis linking well-established risk factors of VCID with the distinct neurodegenerative cascades of neuroinflammation and chronic hypoperfusion. It is hypothesized that these two synergistic signaling cascades coalesce to initiate aberrant angiogenesis and induce blood brain barrier breakdown trough a mechanism mediated by vascular growth factors and matrix metalloproteinases respectively. Finally, this review concludes by highlighting several potential therapeutic interventions along this neurodegenerative sequalae providing diverse opportunities for future translational study.
Collapse
Affiliation(s)
- Alexandria E. Linton
- University of Kentucky, College of Medicine, Sanders-Brown Center on Aging, Department of Physiology, Lexington KY 40536, USA
| | - Erica M. Weekman
- University of Kentucky, College of Medicine, Sanders-Brown Center on Aging, Department of Physiology, Lexington KY 40536, USA
| | - Donna M. Wilcock
- University of Kentucky, College of Medicine, Sanders-Brown Center on Aging, Department of Physiology, Lexington KY 40536, USA
| |
Collapse
|
93
|
Abstract
Leptin for over 25 years has been a central theme in the study of appetite, obesity, and starvation. As the major site of leptin production is peripheral, and the site of action of greatest interest is the hypothalamus, how leptin accesses the central nervous system (CNS) and crosses the blood-brain barrier (BBB) has been of great interest. We review here the ongoing research that addresses fundamental questions such as the sites of leptin resistances in obesity and other conditions, the causes of resistances and their relations to one another, the three barrier sites of entry into the CNS, why recent studies using suprapharmacological doses cannot address these questions but give insight into nonsaturable entry of leptin into the CNS, and how that might be useful in using leptin therapeutically. The current status of the controversy of whether the short form of the leptin receptor acts as the BBB leptin transporter and how obesity may transform leptin transport is reviewed. Review of these and other topics summarizes in a new appreciation of what leptin may have actually evolved to do and what physiological role leptin resistance may play. © 2021 American Physiological Society. Compr Physiol 11:1-19, 2021.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
94
|
Evans LE, Taylor JL, Smith CJ, Pritchard HAT, Greenstein AS, Allan SM. Cardiovascular co-morbidities, inflammation and cerebral small vessel disease. Cardiovasc Res 2021; 117:2575-2588. [PMID: 34499123 DOI: 10.1093/cvr/cvab284] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Cerebral small vessel disease (cSVD) is the most common cause of vascular cognitive impairment and affects all levels of the brain's vasculature. Features include diverse structural and functional changes affecting small arteries and capillaries that lead to a decline in cerebral perfusion. Due to an aging population, incidence of cerebral small vessel disease (cSVD) is continually rising. Despite its prevalence and its ability to cause multiple debilitating illnesses, such as stroke and dementia, there are currently no therapeutic strategies for the treatment of cSVD. In the healthy brain, interactions between neuronal, vascular and inflammatory cells are required for normal functioning. When these interactions are disturbed, chronic pathological inflammation can ensue. The interplay between cSVD and inflammation has attracted much recent interest and this review discusses chronic cardiovascular diseases, particularly hypertension, and explores how the associated inflammation may impact on the structure and function of the small arteries of the brain in cSVD. Molecular approaches in animal studies are linked to clinical outcomes in patients and novel hypotheses regarding inflammation and cSVD are proposed that will hopefully stimulate further discussion and study in this important area.
Collapse
Affiliation(s)
- Lowri E Evans
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Jade L Taylor
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Craig J Smith
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Manchester Centre for Clinical Neurosciences, Manchester Academic Health Science Centre, Salford Royal Hospital, Manchester Academic Health Sciences Centre (MAHSC)
| | - Harry A T Pritchard
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Adam S Greenstein
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom.,Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK
| | - Stuart M Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance & University of Manchester, Manchester, UK.,Division of Neuroscience and Experimental Psychology, The University of Manchester, Manchester, UK
| |
Collapse
|
95
|
Axenhus M, Bogdanovic N. Confusion, cognitive impairment, and spinal cord compression caused by plasmacytoma: a case report. BMC Neurol 2021; 21:303. [PMID: 34362322 PMCID: PMC8342271 DOI: 10.1186/s12883-021-02332-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 06/29/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Plasmacytomas are rare tumors comprised of neoplastic monoclonal plasma cells and can be found anywhere in the body. Plasmacytomas that involve the nervous system can give rise to diffuse symptoms depending on their location. Patients with confusion or dementia might be difficult to neurologically assess in an acute setting and the subtle symptoms of neurological pathology caused by rare malignancies might go undiagnosed. CASE PRESENTATION The patient is an 80 year old man presenting to the ER with walking difficulties, pain, and confusion. He underwent neurological evaluation for dementia and was eventually diagnosed with possible Alzheimer's disease and a malignant plasmacytoma causing spinal cord compression. His CSF sample showed normal amyloid rate and very low Aβ. Following rehabilitation and oncological treatment, his walking ability and confusion improved. CONCLUSION This case is unique as we demonstrate that spinal cord compression by plasmacytoma can lead to abnormal CSF levels of several known pathology markers for Alzheimer's disease and neuronal damage. We suggest that highly divergent amyloid CSF levels could be indicative of spinal pathologies affecting CSF circulation. We also suggest closer assessment of elderly confusion patients in ER settings by consultants specialized in neurological disorders.
Collapse
Affiliation(s)
- Michael Axenhus
- Department of Neurobiology, Care Sciences and Society, Section of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institute, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Nenad Bogdanovic
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Karolinska Institute, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
96
|
Li X, Yin Q, Han X, Zhang H, Wang F, Ma J, Zhuang P, Zhang Y. Dynamic expression of vascular endothelial growth factor (VEGF) and platelet-derived growth factor receptor beta (PDGFRβ) in diabetic brain contributes to cognitive dysfunction. Brain Res Bull 2021; 175:99-106. [PMID: 34303767 DOI: 10.1016/j.brainresbull.2021.07.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/18/2021] [Accepted: 07/19/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Cognitive dysfunction is increasingly recognized as an important complication of diabetes mellitus (DM). Accumulating evidence indicates that the abnormality of cerebrovascular structure and function plays an essential role in diabetic cognitive impairment (DCI), however, changes in cerebrovascular factors have been blurred during the development of diabetes. OBJECTIVE To evaluate the changes in the structure and function of cerebrovascular in DCI mice and to investigate the changes of cerebral angiogenesis and stability factors during the development of DM. METHODS Diabetes was induced by feeding with high-fat diet combined with intraperitoneal injection of streptozotocin (STZ,120 mg/kg). Cognitive function was evaluated at different stages of DM, cerebral neovascularization, blood-brain barrier (BBB) permeability and hippocampal neurons were measured of DCI mice, and the expression of vascular endothelial growth factor (VEGF) and platelet-derived growth factor receptor β (PDGFRβ) in hippocampus was detected during the development of DM. RESULTS With the progress of diabetes, the learning and memory ability of mice gradually decreased, and DCI mice showed neuronal degeneration, increased BBB permeability and pathological cerebral neovascularization. Moreover, the expression of VEGF in the hippocampus increased first and then decreased at DM+8week, PDGFRβ decreased continuously with the development of diabetes. CONCLUSIONS Our results demonstrate that DCI may be attributed to the dynamic expression of VEGF/PDGFRβ in diabetic hippocampus, and pathological cerebral neovascularization, increased BBB permeability and neuronal degeneration are the key links.
Collapse
Affiliation(s)
- Xueli Li
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Qingsheng Yin
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xu Han
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Hanyu Zhang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fang Wang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jing Ma
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Pengwei Zhuang
- Chinese Materia Medica College, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yanjun Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
97
|
Sharma S, Brown CE. Microvascular basis of cognitive impairment in type 1 diabetes. Pharmacol Ther 2021; 229:107929. [PMID: 34171341 DOI: 10.1016/j.pharmthera.2021.107929] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/23/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
The complex computations of the brain require a constant supply of blood flow to meet its immense metabolic needs. Perturbations in blood supply, even in the smallest vascular networks, can have a profound effect on neuronal function and cognition. Type 1 diabetes is a prevalent and insidious metabolic disorder that progressively and heterogeneously disrupts vascular signalling and function in the brain. As a result, it is associated with an array of adverse vascular changes such as impaired regulation of vascular tone, pathological neovascularization and vasoregression, capillary plugging and blood brain barrier disruption. In this review, we highlight the link between microvascular dysfunction and cognitive impairment that is commonly associated with type 1 diabetes, with the aim of synthesizing current knowledge in this field.
Collapse
Affiliation(s)
- Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
98
|
Lin Z, Sur S, Liu P, Li Y, Jiang D, Hou X, Darrow J, Pillai JJ, Yasar S, Rosenberg P, Albert M, Moghekar A, Lu H. Blood-Brain Barrier Breakdown in Relationship to Alzheimer and Vascular Disease. Ann Neurol 2021; 90:227-238. [PMID: 34041783 PMCID: PMC8805295 DOI: 10.1002/ana.26134] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/04/2021] [Accepted: 05/15/2021] [Indexed: 02/06/2023]
Abstract
Objective: Blood–brain barrier (BBB) breakdown has been suggested to be an early biomarker in human cognitive impairment. However, the relationship between BBB breakdown and brain pathology, most commonly Alzheimer disease (AD) and vascular disease, is still poorly understood. The present study measured human BBB function in mild cognitive impairment (MCI) patients on 2 molecular scales, specifically BBB’s permeability to water and albumin molecules. Methods: Fifty-five elderly participants were enrolled, including 33 MCI patients and 22 controls. BBB permeability to water was measured with a new magnetic resonance imaging technique, water extraction with phase contrast arterial spin tagging. BBB permeability to albumin was determined using cerebrospinal fluid (CSF)/serum albumin ratio. Cognitive performance was assessed by domain-specific composite scores. AD pathology (including CSF Aβ and ptau) and vascular risk factors were examined. Results: Compared to cognitively normal subjects, BBB in MCI patients manifested an increased permeability to small molecules such as water but was no more permeable to large molecules such as albumin. BBB permeability to water was found to be related to AD markers of CSF Aβ and ptau. On the other hand, BBB permeability to albumin was found to be related to vascular risk factors, especially hypercholesterolemia, but was not related to AD pathology. BBB permeability to small molecules, but not to large molecules, was found to be predictive of cognitive function. Interpretation: These findings provide early evidence that BBB breakdown is related to both AD and vascular risks, but their effects can be differentiated by spatial scales. BBB permeability to small molecules has a greater impact on cognitive performance.
Collapse
Affiliation(s)
- Zixuan Lin
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sandeepa Sur
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Peiying Liu
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Yang Li
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Dengrong Jiang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Xirui Hou
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jacqueline Darrow
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jay J Pillai
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sevil Yasar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Paul Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Abhay Moghekar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Hanzhang Lu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD.,F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, MD
| |
Collapse
|
99
|
Hansson O. Biomarkers for neurodegenerative diseases. Nat Med 2021; 27:954-963. [PMID: 34083813 DOI: 10.1038/s41591-021-01382-x] [Citation(s) in RCA: 562] [Impact Index Per Article: 140.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022]
Abstract
Biomarkers for neurodegenerative diseases are needed to improve the diagnostic workup in the clinic but also to facilitate the development and monitoring of effective disease-modifying therapies. Positron emission tomography methods detecting amyloid-β and tau pathology in Alzheimer's disease have been increasingly used to improve the design of clinical trials and observational studies. In recent years, easily accessible and cost-effective blood-based biomarkers detecting the same Alzheimer's disease pathologies have been developed, which might revolutionize the diagnostic workup of Alzheimer's disease globally. Relevant biomarkers for α-synuclein pathology in Parkinson's disease are also emerging, as well as blood-based markers of general neurodegeneration and glial activation. This review presents an overview of the latest advances in the field of biomarkers for neurodegenerative diseases. Future directions are discussed regarding implementation of novel biomarkers in clinical practice and trials.
Collapse
Affiliation(s)
- Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden. .,Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
100
|
Martins-Filho RK, Zotin MC, Rodrigues G, Pontes-Neto O. Biomarkers Related to Endothelial Dysfunction and Vascular Cognitive Impairment: A Systematic Review. Dement Geriatr Cogn Disord 2021; 49:365-374. [PMID: 33045717 DOI: 10.1159/000510053] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/07/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION The damage in the endothelium and the neurovascular unit appears to play a key role in the pathogenesis of vascular cognitive impairment (VCI). Although there have been many advances in understanding the physiopathology of this disease, several questions remain unanswered. The association with other degenerative diseases and the heterogeneity of its clinical spectrum establish a diagnostic problem, compromising a better comprehension of the pathology and halting the development of effective treatments. The investigation of biomarkers is an important movement to the development of novel explicative models and treatment targets involved in VCI. METHODS We searched MEDLINE considering the original research based on VCI biomarkers in the past 20 years, following prespecified selection criteria, data extraction, and qualitative synthesis. RESULTS We reviewed 42 articles: 16 investigated plasma markers, 17 analyzed neuropathological markers, 4 studied CSF markers, 4 evaluated neuroimaging markers (ultrasound and MRI), and 1 used peripheral Doppler perfusion imaging. CONCLUSIONS The biomarkers in these studies suggest an intrinsic relationship between endothelial dysfunction and VCI. Nonetheless, there is still a need for identification of a distinctive set of markers that can integrate the clinical approach of VCI, improve diagnostic accuracy, and support the discovery of alternative therapies.
Collapse
Affiliation(s)
- Rui Kleber Martins-Filho
- Department of Neurosciences and Behavioural Sciences, Hospital das Clínicas - Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil,
| | - Maria Clara Zotin
- Department of Internal Medicine, Radiology Division, Hospital das Clínicas - Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Guilherme Rodrigues
- Department of Neurosciences and Behavioural Sciences, Hospital das Clínicas - Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Octavio Pontes-Neto
- Department of Neurosciences and Behavioural Sciences, Hospital das Clínicas - Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|