1
|
Lacoste B, Prat A, Freitas-Andrade M, Gu C. The Blood-Brain Barrier: Composition, Properties, and Roles in Brain Health. Cold Spring Harb Perspect Biol 2025; 17:a041422. [PMID: 38951020 PMCID: PMC12047665 DOI: 10.1101/cshperspect.a041422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Blood vessels are critical to deliver oxygen and nutrients to tissues and organs throughout the body. The blood vessels that vascularize the central nervous system (CNS) possess unique properties, termed the blood-brain barrier (BBB), which allow these vessels to tightly regulate the movement of ions, molecules, and cells between the blood and the brain. This precise control of CNS homeostasis allows for proper neuronal function and protects the neural tissue from toxins and pathogens, and alterations of this barrier are important components of the pathogenesis and progression of various neurological diseases. The physiological barrier is coordinated by a series of physical, transport, and metabolic properties possessed by the brain endothelial cells (ECs) that form the walls of the blood vessels. These properties are regulated by interactions between different vascular, perivascular, immune, and neural cells. Understanding how these cell populations interact to regulate barrier properties is essential for understanding how the brain functions in both health and disease contexts.
Collapse
Affiliation(s)
- Baptiste Lacoste
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
- University of Ottawa Brain and Mind Research Institute, Ottawa, Ontario K1H 8M5, Canada
| | - Alexandre Prat
- Department of Neuroscience, Université de Montréal, Montréal, Québec H2X 0A9, Canada
| | - Moises Freitas-Andrade
- Ottawa Hospital Research Institute, Neuroscience Program, Ottawa, Ontario K1H 8M5, Canada
| | - Chenghua Gu
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
2
|
Rust R, Sagare AP, Zhang M, Zlokovic BV, Kisler K. The blood-brain barrier as a treatment target for neurodegenerative disorders. Expert Opin Drug Deliv 2025; 22:673-692. [PMID: 40096820 DOI: 10.1080/17425247.2025.2480654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/14/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
INTRODUCTION The blood-brain barrier (BBB) is a vascular endothelial membrane which restricts entry of toxins, cells, and microorganisms into the brain. At the same time, the BBB supplies the brain with nutrients, key substrates for DNA and RNA synthesis, and regulatory molecules, and removes metabolic waste products from brain to blood. BBB breakdown and/or dysfunction have been shown in neurogenerative disorders including Alzheimer's disease (AD). Current data suggests that these BBB changes may initiate and/or contribute to neuronal, synaptic, and cognitive dysfunction, and possibly other aspects of neurodegenerative processes. AREAS COVERED We first briefly review recent studies uncovering molecular composition of brain microvasculature and examine the BBB as a possible therapeutic target in neurodegenerative disorders with a focus on AD. Current strategies aimed at protecting and/or restoring altered BBB functions are considered. The relevance of BBB-directed approaches to improve neuronal and synaptic function, and to slow progression of neurodegenerative processes are also discussed. Lastly, we review recent advancements in drug delivery across the BBB. EXPERT OPINION BBB breakdown and/or dysfunction can significantly affect neuronal and synaptic function and neurodegenerative processes. More attention should focus on therapeutics to preserve or restore BBB functions when considering treatments of neurodegenerative diseases and AD.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay P Sagare
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mingzi Zhang
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience and the Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
3
|
Sun B, Li L, Harris OA, Luo J. Blood-brain barrier disruption: a pervasive driver and mechanistic link between traumatic brain injury and Alzheimer's disease. Transl Neurodegener 2025; 14:16. [PMID: 40140960 PMCID: PMC11938631 DOI: 10.1186/s40035-025-00478-5] [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: 09/11/2024] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Traumatic brain injury (TBI) has emerged as a significant risk factor for Alzheimer's disease (AD), a complex and devastating neurodegenerative disorder characterized by progressive cognitive decline and memory loss. Both conditions share a common feature: blood‒brain barrier (BBB) dysfunction, which is believed to play a pivotal role in linking TBI to the development of AD. This review delves into the intricate relationship between TBI and AD, with a focus on BBB dysfunction and its critical role in disease mechanisms and therapeutic development. We first present recent evidence from epidemiological studies highlighting the increased incidence of AD among individuals with a history of TBI, as well as pathological and animal model studies that demonstrate how TBI can accelerate AD-like pathology. Next, we explore the mechanisms by which BBB dysfunction may mediate TBI-induced AD pathology. Finally, we investigate the shared molecular pathways associated with BBB dysfunction in both TBI and AD conditions and discuss the latest findings on how targeting these pathways and employing regenerative approaches, such as stem cell therapy and pharmacological interventions, can enhance BBB function and mitigate neurodegeneration.
Collapse
Affiliation(s)
- Bryan Sun
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Lulin Li
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Odette A Harris
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Jian Luo
- Palo Alto Veterans Institute for Research, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
- Polytrauma System of Care, VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA.
| |
Collapse
|
4
|
Géraudie A, De Rossi P, Canney M, Carpentier A, Delatour B. Effects of blood-brain barrier opening using ultrasound on tauopathies: A systematic review. J Control Release 2025; 379:1029-1044. [PMID: 39875073 DOI: 10.1016/j.jconrel.2025.01.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
Blood-brain barrier opening with ultrasound can potentiate drug efficacy in the treatment of brain pathologies and also provides therapeutic effects on its own. It is an innovative tool to transiently, repeatedly and safely open the barrier, with studies showing beneficial effects in both preclinical models for Alzheimer's disease and recent clinical studies. The first preclinical and clinical work has mainly shown a decrease in amyloid burden in mice models and in patients. However, Alzheimer's disease pathology also encompasses tauopathy, which is closely related to cognitive decline, making it a crucial therapeutic target. The effects of blood-brain barrier opening with ultrasound have been rarely assessed on tau and are still unclear. METHODS This systematic review, conducted through searches using Pubmed, Embase, Web of Science and Cochrane Central databases, extracted results of 15 studies reporting effects of blood-brain barrier opening using ultrasound on tau proteins. RESULTS This review of the literature indicates that blood-brain barrier opening using ultrasound can decrease the extent of the tau pathology or potentialize the effect of a therapeutic drug. However, selected studies report paradoxically that blood-brain barrier opening can increase tau pathology burden and induce brain damage. DISCUSSION Apparent discrepancies between reports could originate from the variability in protocols or analytical methods that may impact the effects of blood-brain barrier opening with ultrasound on tauopathies, glial populations, tissue integrity and functional outcomes. CONCLUSION This calls for a better standardization effort combined with improved methodologies allowing between-studies comparisons, and for further understanding of the effects of blood-brain barrier opening on tau pathology as an essential prerequisite before translation to clinic.
Collapse
Affiliation(s)
- Amandine Géraudie
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France.
| | | | | | - Alexandre Carpentier
- Department of Neurosurgery, Sorbonne University, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France; Faculty of Medicine, Sorbonne University, GRC 23, Brain Machine Interface, APHP, La Pitié-Salpêtrière Hospital, 75013 Paris, France; Advanced Surgical Research Technology Lab, Sorbonne University, 75013 Paris, France
| | - Benoît Delatour
- Paris Brain Institute, ICM, Inserm U1127, CNRS UMR 7225, Sorbonne University, 75013 Paris, France
| |
Collapse
|
5
|
Guzmán‐Hernández R, Fossati S. Fibrillar tau alters cerebral endothelial cell metabolism, vascular inflammatory activation, and barrier function in vitro and in vivo. Alzheimers Dement 2025; 21:e70077. [PMID: 40110691 PMCID: PMC11923556 DOI: 10.1002/alz.70077] [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: 10/21/2024] [Revised: 02/10/2025] [Accepted: 02/10/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION The presence of tau aggregates in and around the brain vasculature in Alzheimer's disease (AD) and tauopathies suggests its possible pathogenicity to cerebral endothelial cells (ECs). METHODS We used an in vitro model of the blood-brain barrier (BBB) to understand the mechanisms of fibrillar tau-mediated cerebral EC and BBB pathology, confirming our findings in 3-month-old P301S mice brains and extracted microvessels. RESULTS Protofibrillar and fibrillar tau species induce endothelial barrier permeability through an increase in glycolysis, which activates ECs toward a pro-inflammatory phenotype, inducing loss of junction protein expression and localization. The Warburg-like metabolic shift toward glycolysis and increased vascular pathological phenotypes are also present in young P301S mice. DISCUSSION In sum, our work reveals that fibrillar tau species, by enhancing endothelial glycolytic metabolism, promote vascular inflammatory phenotypes and loss of BBB function, highlighting the importance of addressing and targeting early tau-mediated neurovascular damage in AD and tauopathies. HIGHLIGHTS We improve the understanding of the mechanisms of vascular pathology in tauopathies. Fibrillar tau mediates vascular metabolic changes, inflammation, and blood-brain barrier (BBB) dysfunction. These events are replicated at early stages in a tauopathy mouse model. Inhibiting altered glycolysis reduces BBB permeability and endothelial activation.
Collapse
Affiliation(s)
- Roberto Guzmán‐Hernández
- Alzheimer's Center at TempleDepartment of Neural SciencesTemple University Lewis Katz School of MedicinePhiladelphiaPennsylvaniaUSA
| | - Silvia Fossati
- Alzheimer's Center at TempleDepartment of Neural SciencesTemple University Lewis Katz School of MedicinePhiladelphiaPennsylvaniaUSA
| |
Collapse
|
6
|
Wen T, Meng L, Liu H, Zhang Q, Dai L, Huang L, Dan L, Zhu K, Luo J, Zhang Z. Fibrinogen-tau Aggregates Exacerbate Tau Pathology and Memory Deficits in Alzheimer's Disease Model Mice. Neurosci Bull 2025:10.1007/s12264-025-01366-8. [PMID: 39971888 DOI: 10.1007/s12264-025-01366-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/30/2024] [Indexed: 02/21/2025] Open
Abstract
Vascular damage plays a significant role in the onset and progression of Alzheimer's disease (AD). However, the precise molecular mechanisms underlying the induction of neuronal injury by vascular damage remain unclear. The present study aimed to examine the impact of fibrinogen (Fg) on tau pathology. The results showed that Fg deposits in the brains of tau P301S transgenic mice interact with tau, enhancing the cytotoxicity of pathological tau aggregates and promoting tau phosphorylation and aggregation. Notably, Fg-modified tau fibrils caused enhanced neuronal apoptosis and synaptic damage compared to unmodified fibrils. Furthermore, intrahippocampal injection of Fg-modified tau fibrils worsened the tau pathology, neuroinflammation, synaptic damage, neuronal apoptosis, and cognitive dysfunction in tau P301S mice compared to controls. The present study provides compelling evidence linking Fg and tau, thereby connecting cerebrovascular damage to tau pathology in AD. Consequently, inhibiting Fg-mediated tau pathology could potentially impede the progression of AD.
Collapse
Affiliation(s)
- Tingting Wen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lanxia Meng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Han Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qian Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liqin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liang Dan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Kedong Zhu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiaying Luo
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhaohui Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
7
|
Denkinger M, Baker S, Harrison TM, Chadwick T, Jagust WJ. Cross-sectional and longitudinal relationships among blood-brain barrier disruption, Alzheimer's disease biomarkers, and cognition in cognitively normal older adults. Neurobiol Aging 2025; 146:15-23. [PMID: 39571410 DOI: 10.1016/j.neurobiolaging.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/09/2024] [Accepted: 11/05/2024] [Indexed: 12/15/2024]
Abstract
Blood-brain barrier disruption (BBBd) occurs in aging, particularly in regions vulnerable to Alzheimer's disease (AD) pathology. However, its relationship to pathological protein accumulation, neurodegeneration, and cognitive impairment in normal aging is unclear. We used dynamic contrast-enhanced MRI (DCE-MRI) and positron emission tomography (PET) imaging in cognitively normal older adults to explore how BBBd correlates with brain atrophy and cognitive function, and whether these relationships are influenced by Aβ or tau. We found that greater BBBd in the hippocampus (HC) and an averaged BBBd-susceptible ROI were linked to worse episodic memory, with interactions between BBBd and atrophy influencing this relationship, independent of Aβ and tau. However, there were no significant relationships between BBBd and non-memory cognitive performance. In participants with longitudinal AD biomarker and cognitive data acquired prior to DCE-MRI, faster longitudinal entorhinal cortex (EC) tau accumulation and episodic memory decline were associated with greater HC BBBd, independent of global Aβ changes and regional atrophy. These findings enhance our understanding of the complex relationships between AD biomarkers, cognitive decline, and BBBd.
Collapse
Affiliation(s)
- Marisa Denkinger
- Department of Neuroscience, University of California, Berkeley, CA, United States.
| | - Suzanne Baker
- Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Theresa M Harrison
- Department of Neuroscience, University of California, Berkeley, CA, United States
| | - Trevor Chadwick
- Department of Neuroscience, University of California, Berkeley, CA, United States
| | - William J Jagust
- Department of Neuroscience, University of California, Berkeley, CA, United States; Lawrence Berkeley National Laboratory, Berkeley, CA, United States.
| |
Collapse
|
8
|
Huang C, Wei Z, Zheng N, Yan J, Zhang J, Ye X, Zhao W. The interaction between dysfunction of vasculature and tauopathy in Alzheimer's disease and related dementias. Alzheimers Dement 2025; 21:e14618. [PMID: 39998958 PMCID: PMC11854360 DOI: 10.1002/alz.14618] [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: 08/12/2024] [Revised: 01/01/2025] [Accepted: 01/12/2025] [Indexed: 02/27/2025]
Abstract
Tauopathy is one of the pathological features of Alzheimer's disease and related dementias (ADRD). At present, there have been many studies on the formation, deposition, and intercellular transmission of tau in neurons and immune cells. The vasculature is an important component of the central nervous system. This review discusses the interaction between vasculature and tau in detail from three aspects. (1) The vascular risk factors (VRFs) discussed in this review include diabetes mellitus (DM), abnormal blood pressure (BP), and hypercholesterolemia. (2) In ADRD pathology, the hyperphosphorylation and deposition of tau interact with disrupted vasculature, such as different cells (endothelial cells, smooth muscular cells, and pericytes), the blood-brain barrier (BBB), and the cerebral lymphatic system. (3) The functions of vasculature are regulated by various signaling transductions. Endothelial nitric oxide synthase/nitric oxide, calcium signaling, Rho/Rho-associated coiled-coil containing Kinase, and receptors for advanced glycation end products are discussed in this review. Our findings indicate that the prevention and treatment of vascular health may be a potential target for ADRD combination therapy. HIGHLIGHTS: Persistent VRFs increase early disruption of vascular mechanisms and are strongly associated with tau pathology in ADRD. Cell dysfunction in the vasculature causes BBB leakage and drainage incapacity of the cerebral lymphatic system, which interacts with tau pathology. Signaling molecules in the vasculature regulate vasodilation and contraction, angiogenesis, and CBF. Abnormal signaling transduction is related to tau hyperphosphorylation and deposition.
Collapse
Affiliation(s)
- Chuyao Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Zhenwen Wei
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Ningxiang Zheng
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jingsi Yan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Jiayu Zhang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Xinyi Ye
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Wei Zhao
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| |
Collapse
|
9
|
Olofsson HE, Englund E. Increased frontocortical microvascular raspberry density in frontotemporal lobar degeneration compared to Lewy body disease and control cases: a neuropathological study. FREE NEUROPATHOLOGY 2025; 6:7. [PMID: 40052111 PMCID: PMC11884261 DOI: 10.17879/freeneuropathology-2025-6178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 02/20/2025] [Indexed: 03/09/2025]
Abstract
Background: Brain raspberries are histologically defined microvascular entities that are highly prevalent in the neocortex. Increased cortical raspberry density occurs in vascular dementia, but also with advancing age. Here, we examined the raspberry density in two neurodegenerative diseases, wherein vascular alterations distinct from conventional vascular risk factors have been indicated: frontotemporal lobar degeneration (FTLD) and Lewy body disease (LBD). Methods: This retrospective study included 283 clinically autopsied individuals: 105 control cases without neurodegenerative disease, 98 FTLD cases (mainly FTLD-tau and FTLD-TDP), and 80 LBD cases (mainly neocortical). The raspberry density was quantified on haematoxylin-eosin-stained tissue sections from the frontal cortex, and the frontocortical atrophy was ranked 0-3. Results: There was a higher raspberry density in the FTLD group compared to both other groups (P ≤ 0.001; Games-Howell post hoc test). The difference between the FTLD and LBD groups remained significant in multiple linear regression models that included age, sex, and either brain weight (P = 0.034) or cortical atrophy (P = 0.012). The difference between the FTLD and control groups remained significant when including age, sex, and brain weight in the model (P = 0.004), while a trend towards significance was demonstrated when including age, sex, and cortical atrophy (P = 0.054). Further analyses of the FTLD group revealed a trend towards a positive correlation between raspberry density and cortical atrophy (P = 0.062; Spearman rank correlation). Comparisons of FTLD subgroups were inconclusive. Conclusion: The frontocortical raspberry density is increased in FTLD. An examination of the raspberry density in relation to a quantitative measure of cortical atrophy is motivated to validate the results. Future studies are needed to determine whether increased raspberry density in FTLD could function as a marker for more widespread vascular alterations, and to elucidate the relation between microvascular alterations and neurodegenerative disease.
Collapse
Affiliation(s)
- Henric Ek Olofsson
- Division of Pathology, Department of Clinical Sciences
Lund, Lund University, Lund, Sweden
| | - Elisabet Englund
- Division of Pathology, Department of Clinical Sciences
Lund, Lund University, Lund, Sweden
| |
Collapse
|
10
|
Reas ET, Solders SK, Tsiknia A, Triebswetter C, Shen Q, Rivera CS, Andrews MJ, Alderson-Myers A, Brewer JB. APOE 𝜀4-related blood-brain barrier breakdown is associated with microstructural abnormalities. Alzheimers Dement 2024; 20:8615-8624. [PMID: 39411970 DOI: 10.1002/alz.14302] [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: 06/25/2024] [Revised: 08/23/2024] [Accepted: 09/10/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION Blood-brain barrier (BBB) dysfunction occurs in Alzheimer's disease (AD). Yet, the stage at which it appears along the AD time course and whether it contributes to neurodegeneration remain unclear. METHODS Older adults (61 to 90 years) from cognitively normal (CN) to mildly cognitively impaired (CI), enriched for APOE 𝜀4 and amyloid positivity, underwent dynamic contrast-enhanced (DCE) magnetic resonance imaging (MRI) and diffusion MRI to measure BBB permeability and brain microstructure. Analysis of variance compared BBB permeability according to cognitive status, amyloid beta (Aβ), and APOE4. Linear regressions assessed associations of BBB permeability with brain microstructure and interactions with Aβ and APOE4. RESULTS BBB permeability was elevated for APOE4 carriers across the cortical gray matter, with the strongest differences among CN amyloid-negative individuals. Associations between entorhinal BBB permeability and microstructure interacted with Aβ and APOE4, with the strongest relationships in amyloid-positive individuals and APOE4 carriers. DISCUSSION APOE4 may drive widespread BBB dysfunction in preclinical AD, which may contribute to neurodegenerative changes early along the AD cascade. HIGHLIGHTS Gray matter blood-brain barrier (BBB) permeability is elevated for APOE4 carriers. APOE4-related BBB breakdown appears in the absence of cognitive decline or amyloid. BBB leakage correlates with entorhinal cortex microstructural injury. Associations with microstructure are strongest for amyloid-positive APOE4 carriers.
Collapse
Affiliation(s)
- Emilie T Reas
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Seraphina K Solders
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Amaryllis Tsiknia
- Imaging Genetics Center, USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Marina Del Rey, California, USA
| | | | - Qian Shen
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Charlotte S Rivera
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - Murray J Andrews
- Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Austin Alderson-Myers
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
| | - James B Brewer
- Department of Neurosciences, University of California, San Diego, La Jolla, California, USA
- Department of Radiology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
11
|
Hossen F, Geng X, Sun GY, Yao X, Lee JC. Oligomeric Amyloid-β and Tau Alter Cell Adhesion Properties and Induce Inflammatory Responses in Cerebral Endothelial Cells Through the RhoA/ROCK Pathway. Mol Neurobiol 2024; 61:8759-8776. [PMID: 38561558 PMCID: PMC11445398 DOI: 10.1007/s12035-024-04138-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 03/19/2024] [Indexed: 04/04/2024]
Abstract
Dysfunction of cerebral endothelial cells (CECs) has been implicated in the pathology of Alzheimer's disease (AD). Despite evidence showing cytotoxic effects of oligomeric amyloid-β (oAβ) and Tau (oTau) in the central nervous system, their direct effects on CECs have not been fully investigated. In this study, we examined the direct effects of oAβ, oTau, and their combination on cell adhesion properties and inflammatory responses in CECs. We found that both oAβ and oTau increased cell stiffness, as well as the p-selectin/Sialyl-LewisX (sLeX) bonding-mediated membrane tether force and probability of adhesion in CECs. Consistent with these biomechanical alterations, treatments with oAβ or oTau also increased actin polymerization and the expression of p-selectin at the cell surface. These toxic oligomeric peptides also triggered inflammatory responses, including upregulations of p-NF-kB p65, IL-1β, and TNF-α. In addition, they rapidly activated the RhoA/ROCK pathway. These biochemical and biomechanical changes were further enhanced by the treatment with the combination of oAβ and oTau, which were significantly suppressed by Fasudil, a specific inhibitor for the RhoA/ROCK pathway. In conclusion, our data suggest that oAβ, oTau, and their combination triggered subcellular mechanical alterations and inflammatory responses in CECs through the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Faruk Hossen
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Xue Geng
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Grace Y Sun
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Xincheng Yao
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - James C Lee
- Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA.
| |
Collapse
|
12
|
Naranjo O, Osborne OM, Torices S, Schmidlin S, Tiburcio D, Park M, Toborek M. Microvessel isolation protocol for RNA visualization and profiling. Sci Rep 2024; 14:25558. [PMID: 39462110 PMCID: PMC11513094 DOI: 10.1038/s41598-024-77501-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/23/2024] [Indexed: 10/28/2024] Open
Abstract
Disruptions in pericyte and endothelial cell communication can compromise the integrity of the blood-brain barrier (BBB), leading to neurovascular dysfunction and the development of neurological disorders. However, the evaluation of microvessel RNAs has been limited to tissue homogenates, with spatial visualization only available for protein targets. The aim of the present study is the development of an innovative microvessel isolation technique that is RNA-friendly for the purpose of coupling with in situ hybridization RNAscope analysis. RNA-friendly microvessel isolation combined with RNAscope analysis enables the visualization of cell-specific RNA within the spatial and histological context of the BBB. Using this approach, we have gained valuable insights into the structural and functional differences associated with the microvessels of 5xFAD mice, a mouse model of Alzheimer's disease (AD). RNAscope analysis revealed a decrease in pericytes from microvessels isolated from 5xFAD mice in comparison to wild-type mice. Additionally, the microvessels of 5xFAD mice exhibited an increase in TYRO protein tyrosine kinase binding protein (TYROBP) mRNA expression. These findings significantly advance our understanding of neurovascular interactions and hold great promise for guiding the development of targeted therapeutic interventions. This innovative approach enables visualization of cell RNA while preserving the spatial and histological context of the BBB, shedding light on the mechanisms underlying neurovascular unit communication.
Collapse
Affiliation(s)
- Oandy Naranjo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Olivia M Osborne
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Silvia Torices
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sarah Schmidlin
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Destiny Tiburcio
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Minseon Park
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
- Institute of Physiotherapy and Health Sciences, the Blood-Brain Barrier Research Center, The Jerzy Kukuczka Academy of Physical Education, Katowice, Poland.
| |
Collapse
|
13
|
Marques D, Moura-Louro D, Silva IP, Matos S, Santos CND, Figueira I. Unlocking the potential of low-molecular-weight (Poly)phenol metabolites: Protectors at the blood-brain barrier frontier. Neurochem Int 2024; 179:105836. [PMID: 39151552 DOI: 10.1016/j.neuint.2024.105836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Neurodegenerative diseases (NDDs) are an increasing group of chronic and progressive neurological disorders that ultimately lead to neuronal cell failure and death. Despite all efforts throughout decades, their burden on individuals and society still casts one of the most massive socioeconomic problems worldwide. The neuronal failure observed in NDDs results from an intricacy of events, mirroring disease complexity, ranging from protein aggregation, oxidative stress, (neuro)inflammation, and even blood-brain barrier (BBB) dysfunction, ultimately leading to cognitive and motor symptoms in patients. As a result of such complex pathobiology, to date, there are still no effective treatments to treat/halt NDDs progression. Fortunately, interest in the bioavailable low molecular weight (LMW) phenolic metabolites derived from the metabolism of dietary (poly)phenols has been rising due to their multitargeted potential in attenuating multiple NDDs hallmarks. Even if not highly BBB permeant, their relatively high concentrations in the bloodstream arising from the intake of (poly)phenol-rich diets make them ideal candidates to act within the vasculature and particularly at the level of BBB. In this review, we highlight the most recent - though still scarce - studies demonstrating LMW phenolic metabolites' ability to modulate BBB homeostasis, including the improvement of tight and adherens junctional proteins, as well as their power to decrease pro-inflammatory cytokine secretion and oxidative stress levels in vitro and in vivo. Specific BBB-permeant LMW phenolic metabolites, such as simple phenolic sulfates, have been emerging as strong BBB properties boosters, pleiotropic compounds capable of improving cell fitness under oxidative and pro-inflammatory conditions. Nevertheless, further studies should be pursued to obtain a holistic overview of the promising role of LMW phenolic metabolites in NDDs prevention and management to fully harness their true therapeutic potential.
Collapse
Affiliation(s)
- Daniela Marques
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Diogo Moura-Louro
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Inês P Silva
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Sara Matos
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal
| | - Cláudia Nunes Dos Santos
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade NOVA de Lisboa, Avenida da República, Oeiras, Portugal; iBET, Instituto de Biologia Experimental e Tecnológica, Avenida da República, Apartado 12, Oeiras, Portugal
| | - Inês Figueira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, Lisboa, Portugal.
| |
Collapse
|
14
|
Han J, Wang Y, Wei P, Lu D, Shan Y. Unveiling the hidden connection: the blood-brain barrier's role in epilepsy. Front Neurol 2024; 15:1413023. [PMID: 39206290 PMCID: PMC11349696 DOI: 10.3389/fneur.2024.1413023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Epilepsy is characterized by abnormal synchronous electrical activity of neurons in the brain. The blood-brain barrier, which is mainly composed of endothelial cells, pericytes, astrocytes and other cell types and is formed by connections between a variety of cells, is the key physiological structure connecting the blood and brain tissue and is critical for maintaining the microenvironment in the brain. Physiologically, the blood-brain barrier controls the microenvironment in the brain mainly by regulating the passage of various substances. Disruption of the blood-brain barrier and increased leakage of specific substances, which ultimately leading to weakened cell junctions and abnormal regulation of ion concentrations, have been observed during the development and progression of epilepsy in both clinical studies and animal models. In addition, disruption of the blood-brain barrier increases drug resistance through interference with drug trafficking mechanisms. The changes in the blood-brain barrier in epilepsy mainly affect molecular pathways associated with angiogenesis, inflammation, and oxidative stress. Further research on biomarkers is a promising direction for the development of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Yongzhi Shan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Wise A, Li J, Yamakawa M, Loureiro J, Peterson B, Worringer K, Sivasankaran R, Palma JA, Mitic L, Heuer HW, Lario-Lago A, Staffaroni AM, Clark A, Taylor J, Ljubenkov PA, Vandevrede L, Grinberg LT, Spina S, Seeley WW, Miller BL, Boeve BF, Dickerson BC, Grossman M, Litvan I, Pantelyat A, Tartaglia MC, Zhang Z, Wills AMA, Rexach J, Rojas JC, Boxer AL. CSF Proteomics in Patients With Progressive Supranuclear Palsy. Neurology 2024; 103:e209585. [PMID: 38959435 PMCID: PMC11226322 DOI: 10.1212/wnl.0000000000209585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 05/15/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Identification of fluid biomarkers for progressive supranuclear palsy (PSP) is critical to enhance therapeutic development. We implemented unbiased DNA aptamer (SOMAmer) proteomics to identify novel CSF PSP biomarkers. METHODS This is a cross-sectional study in original (18 clinically diagnosed PSP-Richardson syndrome [PSP-RS], 28 cognitively healthy controls]), validation (23 PSP-RS, 26 healthy controls), and neuropathology-confirmed (21 PSP, 52 non-PSP frontotemporal lobar degeneration) cohorts. Participants were recruited through the University of California, San Francisco, and the 4-Repeat Neuroimaging Initiative. The original and neuropathology cohorts were analyzed with the SomaScan platform version 3.0 (5026-plex) and the validation cohort with version 4.1 (7595-plex). Clinical severity was measured with the PSP Rating Scale (PSPRS). CSF proteomic data were analyzed to identify differentially expressed targets, implicated biological pathways using enrichment and weighted consensus gene coexpression analyses, diagnostic value of top targets with receiver-operating characteristic curves, and associations with disease severity with linear regressions. RESULTS A total of 136 participants were included (median age 70.6 ± 8 years, 68 [50%] women). One hundred fifty-five of 5,026 (3.1%), 959 of 7,595 (12.6%), and 321 of 5,026 (6.3%) SOMAmers were differentially expressed in PSP compared with controls in original, validation, and neuropathology-confirmed cohorts, with most of the SOMAmers showing reduced signal (83.1%, 95.1%, and 73.2%, respectively). Three coexpression modules were associated with PSP across cohorts: (1) synaptic function/JAK-STAT (β = -0.044, corrected p = 0.002), (2) vesicle cytoskeletal trafficking (β = 0.039, p = 0.007), and (3) cytokine-cytokine receptor interaction (β = -0.032, p = 0.035) pathways. Axon guidance was the top dysregulated pathway in PSP in original (strength = 1.71, p < 0.001), validation (strength = 0.84, p < 0.001), and neuropathology-confirmed (strength = 0.78, p < 0.001) cohorts. A panel of axon guidance pathway proteins discriminated between PSP and controls in original (area under the curve [AUC] = 0.924), validation (AUC = 0.815), and neuropathology-confirmed (AUC = 0.932) cohorts. Two inflammatory proteins, galectin-10 and cytotoxic T lymphocyte-associated protein-4, correlated with PSPRS scores across cohorts. DISCUSSION Axon guidance pathway proteins and several other molecular pathways are downregulated in PSP, compared with controls. Proteins in these pathways may be useful targets for biomarker or therapeutic development.
Collapse
Affiliation(s)
- Amy Wise
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jingyao Li
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Mai Yamakawa
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Joseph Loureiro
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Brant Peterson
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Kathleen Worringer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Rajeev Sivasankaran
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jose-Alberto Palma
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Laura Mitic
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Hilary W Heuer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Argentina Lario-Lago
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Adam M Staffaroni
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Annie Clark
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jack Taylor
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Peter A Ljubenkov
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Lawren Vandevrede
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Lea T Grinberg
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Salvatore Spina
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - William W Seeley
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bruce L Miller
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bradley F Boeve
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Bradford C Dickerson
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Murray Grossman
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Irene Litvan
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Alexander Pantelyat
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Maria Carmela Tartaglia
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Zihan Zhang
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Anne-Marie A Wills
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Jessica Rexach
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Julio C Rojas
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| | - Adam L Boxer
- From the Weill Institute for Neurosciences (A.W., L.M., H.W.H., A.L.-L., A.M.S., A.C., J.T., P.A.L., L.V., L.T.G., S.S., W.W.S., B.L.M., J.C.R., A.L.B.), Department of Neurology, Memory and Aging Center, University of California, San Francisco; Novartis Institutes for Biomedical Research, Inc. (J. Li, J. Loureiro, B.P., K.W., R.S., J.-A.P.), Cambridge, MA; Department of Neurology (M.Y., J.R.), University of California, Los Angeles; The Bluefield Project to Cure FTD (L.M.); Department of Neurology (B.F.B.), Mayo Clinic, Rochester, MN; Department of Neurology (B.C.D., A.-M.A.W.), Massachusetts General Hospital and Harvard Medical School, Boston; Department of Neurology (M.G.), University of Pennsylvania, Philadelphia; Department of Neurology (I.L.), University of California, San Diego; Department of Neurology (A.P.), Johns Hopkins University, Baltimore, MD; Department of Neurology (M.C.T.), University of Toronto, Ontario, Canada; and Departments of Mathematics and Statistics (Z.Z.), University of California, Los Angeles
| |
Collapse
|
16
|
Rather MA, Khan A, Jahan S, Siddiqui AJ, Wang L. Influence of Tau on Neurotoxicity and Cerebral Vasculature Impairment Associated with Alzheimer's Disease. Neuroscience 2024; 552:1-13. [PMID: 38871021 DOI: 10.1016/j.neuroscience.2024.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/15/2024]
Abstract
Alzheimer's disease is a fatal chronic neurodegenerative condition marked by a gradual decline in cognitive abilities and impaired vascular function within the central nervous system. This affliction initiates its insidious progression with the accumulation of two aberrant protein entities including Aβ plaques and neurofibrillary tangles. These chronic elements target distinct brain regions, steadily erasing the functionality of the hippocampus and triggering the erosion of memory and neuronal integrity. Several assumptions are anticipated for AD as genetic alterations, the occurrence of Aβ plaques, altered processing of amyloid precursor protein, mitochondrial damage, and discrepancy of neurotropic factors. In addition to Aβ oligomers, the deposition of tau hyper-phosphorylates also plays an indispensable part in AD etiology. The brain comprises a complex network of capillaries that is crucial for maintaining proper function. Tau is expressed in cerebral blood vessels, where it helps to regulate blood flow and sustain the blood-brain barrier's integrity. In AD, tau pathology can disrupt cerebral blood supply and deteriorate the BBB, leading to neuronal neurodegeneration. Neuroinflammation, deficits in the microvasculature and endothelial functions, and Aβ deposition are characteristically detected in the initial phases of AD. These variations trigger neuronal malfunction and cognitive impairment. Intracellular tau accumulation in microglia and astrocytes triggers deleterious effects on the integrity of endothelium and cerebral blood supply resulting in further advancement of the ailment and cerebral instability. In this review, we will discuss the impact of tau on neurovascular impairment, mitochondrial dysfunction, oxidative stress, and the role of hyperphosphorylated tau in neuron excitotoxicity and inflammation.
Collapse
Affiliation(s)
- Mashoque Ahmad Rather
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States.
| | - Andleeb Khan
- Department of Biosciences, Faculty of Science, Integral University, Lucknow, 226026, India
| | - Sadaf Jahan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, Saudi Arabia
| | - Arif Jamal Siddiqui
- Department of Biology, College of Science, University of Hail, Hail City, Saudi Arabia
| | - Lianchun Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, United States
| |
Collapse
|
17
|
Prajapati SK, Pathak A, Samaiya PK. Alzheimer's disease: from early pathogenesis to novel therapeutic approaches. Metab Brain Dis 2024; 39:1231-1254. [PMID: 39046584 DOI: 10.1007/s11011-024-01389-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/15/2024] [Indexed: 07/25/2024]
Abstract
The mainstay behind Alzheimer's disease (AD) remains unknown due to the elusive pathophysiology of the disease. Beta-amyloid and phosphorylated Tau is still widely incorporated in various research studies while studying AD. However, they are not sufficient. Therefore, many scientists and researchers have dug into AD studies to deliver many innovations in this field. Many novel biomarkers, such as phosphoglycerate-dehydrogenase, clusterin, microRNA, and a new peptide ratio (Aβ37/Aβ42) in cerebral-spinal fluid, plasma glial-fibrillary-acidic-protein, and lipid peroxidation biomarkers, are mushrooming. They are helping scientists find breakthroughs and substantiating their research on the early detection of AD. Neurovascular unit dysfunction in AD is a significant discovery that can help us understand the relationship between neuronal activity and cerebral blood flow. These new biomarkers are promising and can take these AD studies to another level. There have also been big steps forward in diagnosing and finding AD. One example is self-administered-gerocognitive-examination, which is less expensive and better at finding AD early on than mini-mental-state-examination. Quantum brain sensors and electrochemical biosensors are innovations in the detection field that must be explored and incorporated into the studies. Finally, novel innovations in AD studies like nanotheranostics are the future of AD treatment, which can not only diagnose and detect AD but also offer treatment. Non-pharmacological strategies to treat AD have also yielded interesting results. Our literature review spans from 1957 to 2022, capturing research and trends in the field over six decades. This review article is an update not only on the recent advances in the search for credible biomarkers but also on the newer detection techniques and therapeutic approaches targeting AD.
Collapse
Affiliation(s)
- Santosh Kumar Prajapati
- Bhavdiya Institute of Pharmaceutical Sciences and Research, Ayodhya, UP, India
- Department of Neurosurgery and Brain Repair, University of South Florida, Tampa, FL, 33613, USA
| | - Arjit Pathak
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India
| | - Puneet K Samaiya
- Department of Pharmacy Shri G.S. Institute of Technology and Science, Indore, 452003, Madhya Pradesh, India.
| |
Collapse
|
18
|
Kong Y, Cao L, Wang J, Zhuang J, Xie F, Zuo C, Huang Q, Shi K, Rominger A, Li M, Wu P, Guan Y, Ni R. In vivo reactive astrocyte imaging using [ 18F]SMBT-1 in tauopathy and familial Alzheimer's disease mouse models: A multi-tracer study. J Neurol Sci 2024; 462:123079. [PMID: 38878650 DOI: 10.1016/j.jns.2024.123079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 05/13/2024] [Accepted: 06/03/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Reactive astrocytes play an important role in the development of Alzheimer's disease and primary tauopathies. Here, we aimed to investigate the relationships between reactive astrocytes. Microgliosis and glucose metabolism with Tau and amyloid beta pathology by using multi-tracer imaging in widely used tauopathy and familial Alzheimer's disease mouse models. RESULTS Positron emission tomography imaging using [18F]PM-PBB3 (tau), [18F]florbetapir (amyloid-beta), [18F]SMBT-1 (monoamine oxidase-B), [18F]DPA-714 (translocator protein) and [18F]fluorodeoxyglucose was carried out in 3- and 7-month-old rTg4510 tau mice, 5 × FAD familial Alzheimer's disease mice and wild-type mice. Immunofluorescence staining was performed to validate the pathological distribution in the mouse brain after in vivo imaging. We found increased regional levels of [18F]PM-PBB3, [18F]SMBT-1, and [18F]DPA-714 and hypoglucose metabolism in the brains of 7-month-old rTg4510 mice compared to age-matched wild-type mice. Increased [18F]SMBT-1 uptake was observed in the brains of 3, 7-month-old 5 × FAD mice, with elevated regional [18F]florbetapir and [18F]DPA-714 uptakes in the brains of 7-month-old 5 × FAD mice, compared to age-matched wild-type mice. Positive correlations were shown between [18F]SMBT-1 and [18F]PM-PBB3, [18F]DPA-714 and [18F]PM-PBB3 in rTg4510 mice, and between [18F]florbetapir and [18F]DPA-714 SUVRs in 5 × FAD mice. CONCLUSION In summary, these findings provide in vivo evidence that reactive astrocytes, microglial activation, and cerebral hypoglucose metabolism are associated with tau and amyloid pathology development in animal models of tauopathy and familial Alzheimer's disease.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Cao
- PET Center, Huashan Hospital, Fudan University, Shanghai, China; Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Jiao Wang
- Lab of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Junyi Zhuang
- Lab of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Qi Huang
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Kuangyu Shi
- Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Axel Rominger
- Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Ming Li
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Wu
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China.
| | - Ruiqing Ni
- Inst. Regenerative Medicine, University of Zurich, Zurich, Switzerland; Dept. Nuclear Medicine, Bern University Hospital, Bern, Switzerland; Inst. Biomedical Engineering, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Teng CY, Kao NJ, Nguyen NTK, Lin CI, Cross TWL, Lin SH. Effects of xylo-oligosaccharide on gut microbiota, brain protein expression, and lipid profile induced by high-fat diet. J Nutr Biochem 2024; 129:109640. [PMID: 38583497 DOI: 10.1016/j.jnutbio.2024.109640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/31/2024] [Accepted: 04/03/2024] [Indexed: 04/09/2024]
Abstract
Midlife overweight and obesity are risk factors of cognitive decline and Alzheimer' s disease (AD) in late life. In addition to increasing risk of obesity and cognitive dysfunction, diets rich in fats also contributes to an imbalance of gut microbiota. Xylo-oligosaccharides (XOS) are a kind of prebiotic with several biological advantages, and can selectively promote the growth of beneficial microorganisms in the gut. To explore whether XOS can alleviate cognitive decline induced by high-fat diet (HFD) through improving gut microbiota composition, mice were fed with normal control or 60% HFD for 9 weeks to induce obesity. After that, mice were supplemented with XOS (30 g or 60 g/kg-diet) or without, respectively, for 12 weeks. The results showed that XOS inhibited weight gain, decreased epidydimal fat weight, and improved fasting blood sugar and blood lipids in mice. Additionally, XOS elevated spatial learning and memory function, decreased amyloid plaques accumulation, increased brain-derived neurotrophic factor levels, and improved neuroinflammation status in hippocampus. Changes in glycerolipids metabolism-associated lipid compounds caused by HFD in hippocampus were reversed after XOS intervention. On the other hand, after XOS intervention, increase in immune-mediated bacteria, Faecalibacterium was observed. In conclusion, XOS improved gut dysbiosis and ameliorated spatial learning and memory dysfunction caused by HFD by decreasing cognitive decline-associated biomarkers and changing lipid composition in hippocampus.
Collapse
Affiliation(s)
- Chu-Yun Teng
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan
| | - Ning-Jo Kao
- Department of Nutrition and Health Sciences, Kainan University, Taoyuan, Taiwan
| | - Ngan Thi Kim Nguyen
- Program of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ching-I Lin
- Department of Nutrition and Health Sciences, Chang-Gung University of Science and Technology, Taoyuan, Taiwan
| | - Tzu-Wen L Cross
- Departmen of Nutrition Science, Purdue University, West Lafayette, Indiana, USA
| | - Shyh-Hsiang Lin
- School of Nutrition and Health Sciences, Taipei Medical University, Taipei, Taiwan; School of Food Safety, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
20
|
Ritson M, Wheeler-Jones CPD, Stolp HB. Endothelial dysfunction in neurodegenerative disease: Is endothelial inflammation an overlooked druggable target? J Neuroimmunol 2024; 391:578363. [PMID: 38728929 DOI: 10.1016/j.jneuroim.2024.578363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/29/2024] [Accepted: 05/02/2024] [Indexed: 05/12/2024]
Abstract
Neurological diseases with a neurodegenerative component have been associated with alterations in the cerebrovasculature. At the anatomical level, these are centred around changes in cerebral blood flow and vessel organisation. At the molecular level, there is extensive expression of cellular adhesion molecules and increased release of pro-inflammatory mediators. Together, these has been found to negatively impact blood-brain barrier integrity. Systemic inflammation has been found to accelerate and exacerbate endothelial dysfunction, neuroinflammation and degeneration. Here, we review the role of cerebrovasculature dysfunction in neurodegenerative disease and discuss the potential contribution of intermittent pro-inflammatory systemic disease in causing endothelial pathology, highlighting a possible mechanism that may allow broad-spectrum therapeutic targeting in the future.
Collapse
Affiliation(s)
- Megan Ritson
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK
| | | | - Helen B Stolp
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London NW1 0TU, UK.
| |
Collapse
|
21
|
Denkinger M, Baker S, Inglis B, Kobayashi S, Juarez A, Mason S, Jagust W. Associations between regional blood-brain barrier permeability, aging, and Alzheimer's disease biomarkers in cognitively normal older adults. PLoS One 2024; 19:e0299764. [PMID: 38837947 PMCID: PMC11152304 DOI: 10.1371/journal.pone.0299764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/05/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Increased blood-brain barrier permeability (BBBp) has been hypothesized as a feature of aging that may lead to the development of Alzheimer's disease (AD). We sought to identify the brain regions most vulnerable to greater BBBp during aging and examine their regional relationship with neuroimaging biomarkers of AD. METHODS We studied 31 cognitively normal older adults (OA) and 10 young adults (YA) from the Berkeley Aging Cohort Study (BACS). Both OA and YA received dynamic contrast-enhanced MRI (DCE-MRI) to quantify Ktrans values, as a measure of BBBp, in 37 brain regions across the cortex. The OA also received Pittsburgh compound B (PiB)-PET to create distribution volume ratios (DVR) images and flortaucipir (FTP)- PET to create partial volume corrected standardized uptake volume ratios (SUVR) images. Repeated measures ANOVA assessed the brain regions where OA showed greater BBBp than YA. In OA, Ktrans values were compared based on sex, Aβ positivity status, and APOE4 carrier status within a composite region across the areas susceptible to aging. We used linear models and sparse canonical correlation analysis (SCCA) to examine the relationship between Ktrans and AD biomarkers. RESULTS OA showed greater BBBp than YA predominately in the temporal lobe, with some involvement of parietal, occipital and frontal lobes. Within an averaged ROI of affected regions, there was no difference in Ktrans values based on sex or Aβ positivity, but OA who were APOE4 carriers had significantly higher Ktrans values. There was no direct relationship between averaged Ktrans and global Aβ pathology, but there was a trend for an Ab status by tau interaction on Ktrans in this region. SCCA showed increased Ktrans was associated with increased PiB DVR, mainly in temporal and parietal brain regions. There was not a significant relationship between Ktrans and FTP SUVR. DISCUSSION Our findings indicate that the BBB shows regional vulnerability during normal aging that overlaps considerably with the pattern of AD pathology. Greater BBBp in brain regions affected in aging is related to APOE genotype and may also be related to the pathological accumulation of Aβ.
Collapse
Affiliation(s)
- Marisa Denkinger
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Suzanne Baker
- Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| | - Ben Inglis
- Henry H. Wheeler Jr. Brain Imaging Center, University of California, Berkeley, Berkeley, California, United States of America
| | - Sarah Kobayashi
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Alexis Juarez
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - Suzanne Mason
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
| | - William Jagust
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, United States of America
- Lawrence Berkeley National Laboratory, Berkeley, California, United States of America
| |
Collapse
|
22
|
Oliveri D, Moschetti G, Griego A, Scarpa E. Endothelial cellular senescence and tau accumulation: An interplay full of opportunities? IBRAIN 2024; 10:225-230. [PMID: 38915948 PMCID: PMC11193862 DOI: 10.1002/ibra.12154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 06/26/2024]
Abstract
Recent research has shown that tau protein can be passed to neighboring cells, leading to cellular senescence in the endothelial cells present in the central nervous system (CNS). This discovery could potentially open new doors for testing novel therapeutic compounds that specifically target senescent cells (senolytics) or for identifying new biomarkers that can enable early detection of tauopathies and dementia.
Collapse
Affiliation(s)
- Doriana Oliveri
- Department of Pharmaceutical Sciences (DISFARM)University of MilanMilanItaly
- Infection Dynamics Laboratory‐National Institute of Molecular Genetics (INGM)MilanItaly
| | - Giorgia Moschetti
- Department of Pharmaceutical Sciences (DISFARM)University of MilanMilanItaly
- Infection Dynamics Laboratory‐National Institute of Molecular Genetics (INGM)MilanItaly
| | - Anna Griego
- Department of Pharmaceutical Sciences (DISFARM)University of MilanMilanItaly
- Infection Dynamics Laboratory‐National Institute of Molecular Genetics (INGM)MilanItaly
| | - Edoardo Scarpa
- Department of Pharmaceutical Sciences (DISFARM)University of MilanMilanItaly
- Infection Dynamics Laboratory‐National Institute of Molecular Genetics (INGM)MilanItaly
| |
Collapse
|
23
|
Li J, Long Q, Ding H, Wang Y, Luo D, Li Z, Zhang W. Progress in the Treatment of Central Nervous System Diseases Based on Nanosized Traditional Chinese Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308677. [PMID: 38419366 PMCID: PMC11040388 DOI: 10.1002/advs.202308677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 02/07/2024] [Indexed: 03/02/2024]
Abstract
Traditional Chinese Medicine (TCM) is widely used in clinical practice to treat diseases related to central nervous system (CNS) damage. However, the blood-brain barrier (BBB) constitutes a significant impediment to the effective delivery of TCM, thus substantially diminishing its efficacy. Advances in nanotechnology and its applications in TCM (also known as nano-TCM) can deliver active ingredients or components of TCM across the BBB to the targeted brain region. This review provides an overview of the physiological and pathological mechanisms of the BBB and systematically classifies the common TCM used to treat CNS diseases and types of nanocarriers that effectively deliver TCM to the brain. Additionally, drug delivery strategies for nano-TCMs that utilize in vivo physiological properties or in vitro devices to bypass or cross the BBB are discussed. This review further focuses on the application of nano-TCMs in the treatment of various CNS diseases. Finally, this article anticipates a design strategy for nano-TCMs with higher delivery efficiency and probes their application potential in treating a wider range of CNS diseases.
Collapse
Affiliation(s)
- Jing Li
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Qingyin Long
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| | - Huang Ding
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| | - Yang Wang
- Institute of Integrative MedicineDepartment of Integrated Traditional Chinese and Western MedicineXiangya HospitalCentral South University ChangshaChangsha410008China
| | - Dan Luo
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Zhou Li
- Beijing Institute of Nanoenergy and NanosystemsChinese Academy of SciencesBeijing101400China
| | - Wei Zhang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio‐Cerebral Diseases, School of Integrated Chinese and Western MedicineHunan University of Chinese MedicineChangshaHunan410208China
| |
Collapse
|
24
|
Zeng J, Liao Z, Yang H, Wang Q, Wu Z, Hua F, Zhou Z. T cell infiltration mediates neurodegeneration and cognitive decline in Alzheimer's disease. Neurobiol Dis 2024; 193:106461. [PMID: 38437992 DOI: 10.1016/j.nbd.2024.106461] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/06/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder with pathological features of β-amyloid (Aβ) and hyperphosphorylated tau protein accumulation in the brain, often accompanied by cognitive decline. So far, our understanding of the extent and role of adaptive immune responses in AD has been quite limited. T cells, as essential members of the adaptive immune system, exhibit quantitative and functional abnormalities in the brains of AD patients. Dysfunction of the blood-brain barrier (BBB) in AD is considered one of the factors leading to T cell infiltration. Moreover, the degree of neuronal loss in AD is correlated with the quantity of T cells. We first describe the differentiation and subset functions of peripheral T cells in AD patients and provide an overview of the key findings related to BBB dysfunction and how T cells infiltrate the brain parenchyma through the BBB. Furthermore, we emphasize the risk factors associated with AD, including Aβ, Tau protein, microglial cells, apolipoprotein E (ApoE), and neuroinflammation. We discuss their regulation of T cell activation and proliferation, as well as the connection between T cells, neurodegeneration, and cognitive decline. Understanding the innate immune response is crucial for providing comprehensive personalized therapeutic strategies for AD.
Collapse
Affiliation(s)
- Junjian Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Zhiqiang Liao
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Hanqin Yang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Qiong Wang
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Zhiyong Wu
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China
| | - Fuzhou Hua
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China.
| | - Zhidong Zhou
- Department of Anesthesiology, the Second Affiliated Hospital of Nanchang University, 330006 Nanchang, Jiangxi, China; Key Laboratory of Anesthesiology of Jiangxi Province, 1# Minde Road, 330006 Nanchang City, Jiangxi Province, China.
| |
Collapse
|
25
|
Kim YA, Mellen M, Kizil C, Santa-Maria I. Mechanisms linking cerebrovascular dysfunction and tauopathy: Adding a layer of epiregulatory complexity. Br J Pharmacol 2024; 181:879-895. [PMID: 37926507 DOI: 10.1111/bph.16280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 09/11/2023] [Accepted: 09/30/2023] [Indexed: 11/07/2023] Open
Abstract
Intracellular accumulation of hyperphosphorylated misfolded tau proteins are found in many neurodegenerative tauopathies, including Alzheimer's disease (AD). Tau pathology can impact cerebrovascular physiology and function through multiple mechanisms. In vitro and in vivo studies have shown that alterations in the blood-brain barrier (BBB) integrity and function can result in synaptic abnormalities and neuronal damage. In the present review, we will summarize how tau proteostasis dysregulation contributes to vascular dysfunction and, conversely, we will examine the factors and pathways leading to tau pathological alterations triggered by cerebrovascular dysfunction. Finally, we will highlight the role epigenetic and epitranscriptomic factors play in regulating the integrity of the cerebrovascular system and the progression of tauopathy including a few observartions on potential therapeutic interventions. LINKED ARTICLES: This article is part of a themed issue From Alzheimer's Disease to Vascular Dementia: Different Roads Leading to Cognitive Decline. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.6/issuetoc.
Collapse
Affiliation(s)
- Yoon A Kim
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Marian Mellen
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain
| | - Caghan Kizil
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, New York, USA
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcon, Madrid, Spain
| |
Collapse
|
26
|
Wu YC, Bogale TA, Koistinaho J, Pizzi M, Rolova T, Bellucci A. The contribution of β-amyloid, Tau and α-synuclein to blood-brain barrier damage in neurodegenerative disorders. Acta Neuropathol 2024; 147:39. [PMID: 38347288 PMCID: PMC10861401 DOI: 10.1007/s00401-024-02696-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Accepted: 01/23/2024] [Indexed: 02/15/2024]
Abstract
Central nervous system (CNS) accumulation of fibrillary deposits made of Amyloid β (Aβ), hyperphosphorylated Tau or α-synuclein (α-syn), present either alone or in the form of mixed pathology, characterizes the most common neurodegenerative diseases (NDDs) as well as the aging brain. Compelling evidence supports that acute neurological disorders, such as traumatic brain injury (TBI) and stroke, are also accompanied by increased deposition of toxic Aβ, Tau and α-syn species. While the contribution of these pathological proteins to neurodegeneration has been experimentally ascertained, the cellular and molecular mechanisms driving Aβ, Tau and α-syn-related brain damage remain to be fully clarified. In the last few years, studies have shown that Aβ, Tau and α-syn may contribute to neurodegeneration also by inducing and/or promoting blood-brain barrier (BBB) disruption. These pathological proteins can affect BBB integrity either directly by affecting key BBB components such as pericytes and endothelial cells (ECs) or indirectly, by promoting brain macrophages activation and dysfunction. Here, we summarize and critically discuss key findings showing how Aβ, Tau and α-syn can contribute to BBB damage in most common NDDs, TBI and stroke. We also highlight the need for a deeper characterization of the role of these pathological proteins in the activation and dysfunction of brain macrophages, pericytes and ECs to improve diagnosis and treatment of acute and chronic neurological disorders.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Tizibt Ashine Bogale
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy
- Department of Acute Brain and Cardiovascular Injury, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jari Koistinaho
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Marina Pizzi
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy
| | - Taisia Rolova
- Neuroscience Center, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arianna Bellucci
- Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, 25123, Brescia, BS, Italy.
| |
Collapse
|
27
|
Jiang Z, Wang J, Qin Y, Liu S, Luo B, Bai F, Wei H, Zhang S, Wei J, Ding G, Ma L, He S, Chen R, Sun Y, Chen Y, Wang L, Xu H, Wang X, Chen G, Lei W. A nonhuman primate model with Alzheimer's disease-like pathology induced by hippocampal overexpression of human tau. Alzheimers Res Ther 2024; 16:22. [PMID: 38281031 PMCID: PMC10821564 DOI: 10.1186/s13195-024-01392-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/15/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the most burdening diseases of the century with no disease-modifying treatment at this time. Nonhuman primates (NHPs) share genetic, anatomical, and physiological similarities with humans, making them ideal model animals for investigating the pathogenesis of AD and potential therapies. However, the use of NHPs in AD research has been hindered by the paucity of AD monkey models due to their long generation time, ethical considerations, and technical challenges in genetically modifying monkeys. METHODS Here, we developed an AD-like NHP model by overexpressing human tau in the bilateral hippocampi of adult rhesus macaque monkeys. We evaluated the pathological features of these monkeys with immunostaining, Nissl staining, cerebrospinal fluid (CSF) analysis, magnetic resonance imaging (MRI), positron emission tomography (PET), and behavioural tests. RESULTS We demonstrated that after hippocampal overexpression of tau protein, these monkeys displayed multiple pathological features of AD, including 3-repeat (3R)/4-repeat (4R) tau accumulation, tau hyperphosphorylation, tau propagation, neuronal loss, hippocampal atrophy, neuroinflammation, Aβ clearance deficits, blood vessel damage, and cognitive decline. More interestingly, the accumulation of both 3R and 4R tau is specific to NHPs but not found in adult rodents. CONCLUSIONS This work establishes a tau-induced AD-like NHP model with many key pathological and behavioural features of AD. In addition, our model may potentially become one of the AD NHP models adopted by researchers worldwide since it can be generated within 2 ~ 3 months through a single injection of AAVs into the monkey brains. Hence, our model NHPs may facilitate mechanistic studies and therapeutic treatments for AD.
Collapse
Affiliation(s)
- Zhouquan Jiang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Jing Wang
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yongpeng Qin
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shanggong Liu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Bin Luo
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Fan Bai
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Huiyi Wei
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shaojuan Zhang
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Junjie Wei
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Guoyu Ding
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Long Ma
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Shu He
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Rongjie Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Ying Sun
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Yi Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Lu Wang
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Hao Xu
- Department of Nuclear Medicine and PET/CT-MRI Centre, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Xiangyu Wang
- Department of Neurosurgery, the First Affiliated Hospital, Jinan University, Guangzhou, 510630, Guangdong, China
| | - Gong Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China.
| | - Wenliang Lei
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
28
|
Xiao X, Han Y, Li Q, Zheng D, Cheng CY, Ni Y. Exploring the evolving function of soluble intercellular adhesion molecule-1 in junction dynamics during spermatogenesis. Front Endocrinol (Lausanne) 2024; 14:1281812. [PMID: 38260159 PMCID: PMC10801026 DOI: 10.3389/fendo.2023.1281812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/15/2023] [Indexed: 01/24/2024] Open
Abstract
Intercellular adhesion molecule-1 (ICAM-1) is a transmembrane glycoprotein expressed on immune, endothelial, and epithelial cells. Its ectodomain can be proteolytically cleaved to release a circulating soluble form called sICAM-1. Clinical studies demonstrate sICAM-1 is upregulated in various diseases and associated with disease severity. Research has identified sICAM-1 as a regulator of the blood-testis barrier (BTB) and spermatogenesis. Overexpression of sICAM-1 weakened the BTB in vitro and in vivo, downregulated junction proteins including N-cadherin, γ-catenin, and connexin 43, and caused germ cell loss. This contrasts with barrier-strengthening effects of membrane-bound ICAM-1. sICAM-1 may act as a molecular switch enabling germ cells to open BTB and Sertoli-germ cell adhesion for transport across the seminiferous epithelium. While the mechanism remains unclear, reduced SRC family kinase (SFK) signaling was observed following sICAM-1 overexpression. SRC promotes BTB protein endocytosis and degradation, influences cytoskeletal dynamics, and affects cell polarity. As sICAM-1 overexpression phenocopies SRC inhibition, SRC may operate downstream of sICAM-1 in regulating BTB dynamics and spermatogenesis. Investigating sICAM-1's structure-function regions and downstream targets will elucidate the molecular mechanisms of junction disruption. This knowledge could enable strategies targeting sICAM-1/SRC to modulate BTB permeability and treat male infertility or diseases involving endothelial/epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Xiang Xiao
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
- Engineering Research Center of Novel Vaccine of Zhejiang Province, Hangzhou Medical College, Hangzhou, China
| | - Yating Han
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Qin Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou, China
| | - Dongwang Zheng
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| | - C. Yan Cheng
- Department of Urology and Andrology, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ya Ni
- Center for Reproductive Health, School of Pharmaceutical Sciences, Hangzhou Medical College (Zhejiang Academy of Medical Sciences), Hangzhou, China
| |
Collapse
|
29
|
Nystuen KL, McNamee SM, Akula M, Holton KM, DeAngelis MM, Haider NB. Alzheimer's Disease: Models and Molecular Mechanisms Informing Disease and Treatments. Bioengineering (Basel) 2024; 11:45. [PMID: 38247923 PMCID: PMC10813760 DOI: 10.3390/bioengineering11010045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/15/2023] [Accepted: 12/22/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's Disease (AD) is a complex neurodegenerative disease resulting in progressive loss of memory, language and motor abilities caused by cortical and hippocampal degeneration. This review captures the landscape of understanding of AD pathology, diagnostics, and current therapies. Two major mechanisms direct AD pathology: (1) accumulation of amyloid β (Aβ) plaque and (2) tau-derived neurofibrillary tangles (NFT). The most common variants in the Aβ pathway in APP, PSEN1, and PSEN2 are largely responsible for early-onset AD (EOAD), while MAPT, APOE, TREM2 and ABCA7 have a modifying effect on late-onset AD (LOAD). More recent studies implicate chaperone proteins and Aβ degrading proteins in AD. Several tests, such as cognitive function, brain imaging, and cerebral spinal fluid (CSF) and blood tests, are used for AD diagnosis. Additionally, several biomarkers seem to have a unique AD specific combination of expression and could potentially be used in improved, less invasive diagnostics. In addition to genetic perturbations, environmental influences, such as altered gut microbiome signatures, affect AD. Effective AD treatments have been challenging to develop. Currently, there are several FDA approved drugs (cholinesterase inhibitors, Aß-targeting antibodies and an NMDA antagonist) that could mitigate AD rate of decline and symptoms of distress.
Collapse
Affiliation(s)
- Kaden L. Nystuen
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Shannon M. McNamee
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Monica Akula
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Kristina M. Holton
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Margaret M. DeAngelis
- Department of Ophthalmology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Neena B. Haider
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
30
|
Conn KA, Borsom EM, Cope EK. Implications of microbe-derived ɣ-aminobutyric acid (GABA) in gut and brain barrier integrity and GABAergic signaling in Alzheimer's disease. Gut Microbes 2024; 16:2371950. [PMID: 39008552 PMCID: PMC11253888 DOI: 10.1080/19490976.2024.2371950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/19/2024] [Indexed: 07/17/2024] Open
Abstract
The gut microbial ecosystem communicates bidirectionally with the brain in what is known as the gut-microbiome-brain axis. Bidirectional signaling occurs through several pathways including signaling via the vagus nerve, circulation of microbial metabolites, and immune activation. Alterations in the gut microbiota are implicated in Alzheimer's disease (AD), a progressive neurodegenerative disease. Perturbations in gut microbial communities may affect pathways within the gut-microbiome-brain axis through altered production of microbial metabolites including ɣ-aminobutyric acid (GABA), the primary inhibitory mammalian neurotransmitter. GABA has been shown to act on gut integrity through modulation of gut mucins and tight junction proteins and may be involved in vagus nerve signal inhibition. The GABAergic signaling pathway has been shown to be dysregulated in AD, and may be responsive to interventions. Gut microbial production of GABA is of recent interest in neurological disorders, including AD. Bacteroides and Lactic Acid Bacteria (LAB), including Lactobacillus, are predominant producers of GABA. This review highlights how temporal alterations in gut microbial communities associated with AD may affect the GABAergic signaling pathway, intestinal barrier integrity, and AD-associated inflammation.
Collapse
Affiliation(s)
- Kathryn A. Conn
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| | - Emily M. Borsom
- Center for Data-Driven Discovery for Biology, Allen Institute, Seattle, WA, USA
| | - Emily K. Cope
- Center for Applied Microbiome Sciences, The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, AZ, USA
- Department of Biological Sciences, Northern Arizona University, Flagstaff, AZ, USA
| |
Collapse
|
31
|
Asamu MO, Oladipo OO, Abayomi OA, Adebayo AA. Alzheimer's disease: The role of T lymphocytes in neuroinflammation and neurodegeneration. Brain Res 2023; 1821:148589. [PMID: 37734576 DOI: 10.1016/j.brainres.2023.148589] [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: 09/03/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
Alzheimer's disease, the leading cause of progressive cognitive decline globally, has been reported to be enhanced by neuroinflammation. Brain-resident innate immune cells and adaptive immune cells work together to produce neuroinflammation. Studies over the past decade have established the neuroimmune axis present in Alzheimer's disease; the crosstalk between adaptive and innate immune cells within and outside the brain is crucial to the onset and progression of Alzheimer's disease. Although the role of the adaptive immune system in Alzheimer's disease is not fully understood, it has been hypothesized that the brain's immune homeostasis is significantly disrupted, which greatly contributes to neuroinflammation. Brain-infiltrating T cells possess proinflammatory phenotypes and activities that directly contribute to neuroinflammation. The pro-inflammatory activities of the adaptive immune system in Alzheimer's disease are characterized by the upregulation of effector T cell activities and the downregulation of regulatory T cell activities in the brain, blood, and cerebrospinal fluid. In this review, we discuss the major impact of T lymphocytes on the pathogenesis and progression of Alzheimer's disease. Understanding the role and mechanism of action of T cells in Alzheimer's disease would significantly contribute to the identification of novel biomarkers for diagnosing and monitoring the progression of the disease. This knowledge could also be crucial to the development of immunotherapies for Alzheimer's disease.
Collapse
Affiliation(s)
- Moses O Asamu
- Department of Anatomy, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| | - Oladapo O Oladipo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria.
| | - Oluseun A Abayomi
- College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; Olabisi Onabanjo University Teaching Hospital (OOUTH), Sagamu, Ogun State, Nigeria
| | - Afeez A Adebayo
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria; College of Health Sciences, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
| |
Collapse
|
32
|
Alexander C, Parsaee A, Vasefi M. Polyherbal and Multimodal Treatments: Kaempferol- and Quercetin-Rich Herbs Alleviate Symptoms of Alzheimer's Disease. BIOLOGY 2023; 12:1453. [PMID: 37998052 PMCID: PMC10669725 DOI: 10.3390/biology12111453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023]
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder impairing cognition and memory in the elderly. This disorder has a complex etiology, including senile plaque and neurofibrillary tangle formation, neuroinflammation, oxidative stress, and damaged neuroplasticity. Current treatment options are limited, so alternative treatments such as herbal medicine could suppress symptoms while slowing cognitive decline. We followed PRISMA guidelines to identify potential herbal treatments, their associated medicinal phytochemicals, and the potential mechanisms of these treatments. Common herbs, including Ginkgo biloba, Camellia sinensis, Glycyrrhiza uralensis, Cyperus rotundus, and Buplerum falcatum, produced promising pre-clinical results. These herbs are rich in kaempferol and quercetin, flavonoids with a polyphenolic structure that facilitate multiple mechanisms of action. These mechanisms include the inhibition of Aβ plaque formation, a reduction in tau hyperphosphorylation, the suppression of oxidative stress, and the modulation of BDNF and PI3K/AKT pathways. Using pre-clinical findings from quercetin research and the comparatively limited data on kaempferol, we proposed that kaempferol ameliorates the neuroinflammatory state, maintains proper cellular function, and restores pro-neuroplastic signaling. In this review, we discuss the anti-AD mechanisms of quercetin and kaempferol and their limitations, and we suggest a potential alternative treatment for AD. Our findings lead us to conclude that a polyherbal kaempferol- and quercetin-rich cocktail could treat AD-related brain damage.
Collapse
Affiliation(s)
- Claire Alexander
- Department of Biology, Lamar University, Beaumont, TX 77705, USA
| | - Ali Parsaee
- Biological Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Maryam Vasefi
- Department of Biology, Lamar University, Beaumont, TX 77705, USA
| |
Collapse
|
33
|
Afsar A, Chen M, Xuan Z, Zhang L. A glance through the effects of CD4 + T cells, CD8 + T cells, and cytokines on Alzheimer's disease. Comput Struct Biotechnol J 2023; 21:5662-5675. [PMID: 38053545 PMCID: PMC10694609 DOI: 10.1016/j.csbj.2023.10.058] [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: 07/31/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 12/07/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia. Unfortunately, despite numerous studies, an effective treatment for AD has not yet been established. There is remarkable evidence indicating that the innate immune mechanism and adaptive immune response play significant roles in the pathogenesis of AD. Several studies have reported changes in CD8+ and CD4+ T cells in AD patients. This mini-review article discusses the potential contribution of CD4+ and CD8+ T cells reactivity to amyloid β (Aβ) protein in individuals with AD. Moreover, this mini-review examines the potential associations between T cells, heme oxygenase (HO), and impaired mitochondria in the context of AD. While current mathematical models of AD have not extensively addressed the inclusion of CD4+ and CD8+ T cells, there exist models that can be extended to consider AD as an autoimmune disease involving these T cell types. Additionally, the mini-review covers recent research that has investigated the utilization of machine learning models, considering the impact of CD4+ and CD8+ T cells.
Collapse
Affiliation(s)
- Atefeh Afsar
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Min Chen
- Department of Mathematical Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Zhenyu Xuan
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| | - Li Zhang
- Department of Biological Sciences, University of Texas at Dallas, Richardson, TX, USA
| |
Collapse
|
34
|
Moon Y, Jeon HJ, Han SH, Min-Young N, Kim HJ, Kwon KJ, Moon WJ, Kim SH. Blood-brain barrier breakdown is linked to tau pathology and neuronal injury in a differential manner according to amyloid deposition. J Cereb Blood Flow Metab 2023; 43:1813-1825. [PMID: 37283062 PMCID: PMC10676138 DOI: 10.1177/0271678x231180035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 06/08/2023]
Abstract
The blood-brain barrier (BBB) breakdown has been suggested as an early marker for Alzheimer's disease (AD); yet the relationship between BBB breakdown and AD-specific biomarkers based on the amyloid/tau/neurodegeneration framework is not clear. This study investigated the relationship between BBB permeability, AD-specific biomarkers, and cognition in patients with cognitive impairment. In this prospective study, we enrolled 62 participants with mild cognitive impairment or dementia between January 2019 and October 2020. All participants were assessed through cognitive tests, amyloid positron emission tomography (PET), dynamic contrast-enhanced magnetic resonance imaging (MRI) for BBB permeability (Ktrans), cerebrospinal fluid studies for Aβ42/40 ratio, phosphorylated-tau Thr181 protein (p-tau), total tau protein (t-tau), and structural MRI for neurodegeneration. In amyloid PET (+) group, higher cortical Ktrans was associated with lower Aβ40 (r = -0.529 p = 0.003), higher Aβ42/40 ratio (r = 0.533, p = 0.003), lower p-tau (r = -0.452, p = 0.014) and lower hippocampal volume (r = -0.438, p = 0.017). In contrast, cortical Ktrans was positively related to t-tau level. (r = 0.489, p = 0.004) in amyloid PET (-) group. Our results suggest that BBB permeability is related to AD-specific biomarkers, but the relationship can vary by the presence of Aβ plaque accumulation.
Collapse
Affiliation(s)
- Yeonsil Moon
- Department of Neurology, Konkuk University Medical Center, Seoul, Republic of Korea
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
| | - Hong Jun Jeon
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
- Department of Psychiatry, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Seol-Heui Han
- Department of Neurology, Konkuk University Medical Center, Seoul, Republic of Korea
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
| | - Noh Min-Young
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hee-Jin Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Kyoung Ja Kwon
- Center for Geriatric Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Won-Jin Moon
- Research Institute of Medical Science, Konkuk University of Medicine, Seoul, Republic of Korea
- Department of Radiology, Konkuk University Medical Center, Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| |
Collapse
|
35
|
Liang J, Liu B, Dong X, Wang Y, Cai W, Zhang N, Zhang H. Decoding the role of gut microbiota in Alzheimer's pathogenesis and envisioning future therapeutic avenues. Front Neurosci 2023; 17:1242254. [PMID: 37790586 PMCID: PMC10544353 DOI: 10.3389/fnins.2023.1242254] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/04/2023] [Indexed: 10/05/2023] Open
Abstract
Alzheimer's disease (AD) emerges as a perturbing neurodegenerative malady, with a profound comprehension of its underlying pathogenic mechanisms continuing to evade our intellectual grasp. Within the intricate tapestry of human health and affliction, the enteric microbial consortium, ensconced within the milieu of the human gastrointestinal tract, assumes a role of cardinal significance. Recent epochs have borne witness to investigations that posit marked divergences in the composition of the gut microbiota between individuals grappling with AD and those favored by robust health. The composite vicissitudes in the configuration of the enteric microbial assembly are posited to choreograph a participatory role in the inception and progression of AD, facilitated by the intricate conduit acknowledged as the gut-brain axis. Notwithstanding, the precise nature of this interlaced relationship remains enshrouded within the recesses of obscurity, poised for an exhaustive revelation. This review embarks upon the endeavor to focalize meticulously upon the mechanistic sway exerted by the enteric microbiota upon AD, plunging profoundly into the execution of interventions that govern the milieu of enteric microorganisms. In doing so, it bestows relevance upon the therapeutic stratagems that form the bedrock of AD's management, all whilst casting a prospective gaze into the horizon of medical advancements.
Collapse
Affiliation(s)
- Junyi Liang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Bin Liu
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Xiaohong Dong
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Yueyang Wang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Wenhui Cai
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Ning Zhang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang, China
| | - Hong Zhang
- Heilongjiang Jiamusi Central Hospital, Jiamusi, Heilongjiang, China
| |
Collapse
|
36
|
Aquilani R, Cotta Ramusino M, Maestri R, Iadarola P, Boselli M, Perini G, Boschi F, Dossena M, Bellini A, Buonocore D, Doria E, Costa A, Verri M. Several dementia subtypes and mild cognitive impairment share brain reduction of neurotransmitter precursor amino acids, impaired energy metabolism, and lipid hyperoxidation. Front Aging Neurosci 2023; 15:1237469. [PMID: 37655338 PMCID: PMC10466813 DOI: 10.3389/fnagi.2023.1237469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
Objective Dementias and mild cognitive impairment (MCI) are associated with variously combined changes in the neurotransmitter system and signaling, from neurotransmitter synthesis to synaptic binding. The study tested the hypothesis that different dementia subtypes and MCI may share similar reductions of brain availability in amino acid precursors (AAPs) of neurotransmitter synthesis and concomitant similar impairment in energy production and increase of oxidative stress, i.e., two important metabolic alterations that impact neurotransmission. Materials and methods Sixty-five demented patients (Alzheimer's disease, AD, n = 44; frontotemporal disease, FTD, n = 13; vascular disease, VaD, n = 8), 10 subjects with MCI and 15 control subjects (CTRL) were recruited for this study. Cerebrospinal fluid (CSF) and plasma levels of AAPs, energy substrates (lactate, pyruvate), and an oxidative stress marker (malondialdehyde, MDA) were measured in all participants. Results Demented patients and subjects with MCI were similar for age, anthropometric parameters, biohumoral variables, insulin resistance (HOMA index model), and CSF neuropathology markers. Compared to age-matched CTRL, both demented patients and MCI subjects showed low CSF AAP tyrosine (precursor of dopamine and catecholamines), tryptophan (precursor of serotonin), methionine (precursor of acetylcholine) limited to AD and FTD, and phenylalanine (an essential amino acid largely used for protein synthesis) (p = 0.03 to <0.0001). No significant differences were found among dementia subtypes or between each dementia subtype and MCI subjects. In addition, demented patients and MCI subjects, compared to CTRL, had similar increases in CSF and plasma levels of pyruvate (CSF: p = 0.023 to <0.0001; plasma: p < 0.002 to <0.0001) and MDA (CSF: p < 0.035 to 0.002; plasma: p < 0.0001). Only in AD patients was the CSF level of lactate higher than in CTRL (p = 0.003). Lactate/pyruvate ratios were lower in all experimental groups than in CTRL. Conclusion AD, FTD, and VaD dementia patients and MCI subjects may share similar deficits in AAPs, partly in energy substrates, and similar increases in oxidative stress. These metabolic alterations may be due to AAP overconsumption following high brain protein turnover (leading to phenylalanine reductions), altered mitochondrial structure and function, and an excess of free radical production. All these metabolic alterations may have a negative impact on synaptic plasticity and activity.
Collapse
Affiliation(s)
- Roberto Aquilani
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Matteo Cotta Ramusino
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Roberto Maestri
- Department of Biomedical Engineering of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Paolo Iadarola
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Mirella Boselli
- Neurorehabilitation Unit of the Montescano Institute, Istituti Clinici Scientifici Maugeri IRCCS, Montescano, Italy
| | - Giulia Perini
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Dementia Research Center, IRCCS Mondino Foundation, Pavia, Italy
| | - Federica Boschi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maurizia Dossena
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Anna Bellini
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Daniela Buonocore
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Enrico Doria
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Alfredo Costa
- Unit of Behavioral Neurology and Center for Cognitive Disorders and Dementia, IRCCS C. Mondino Foundation, Pavia, Italy
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Manuela Verri
- Department of Biology and Biotechnology, “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
37
|
Emmerson JT, Do Carmo S, Liu Y, Shalhoub A, Liu A, Bonomo Q, Malcolm JC, Breuillaud L, Cuello AC. Progressive human-like tauopathy with downstream neurodegeneration and neurovascular compromise in a transgenic rat model. Neurobiol Dis 2023; 184:106227. [PMID: 37454780 DOI: 10.1016/j.nbd.2023.106227] [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: 05/02/2023] [Revised: 06/27/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023] Open
Abstract
Tauopathies, including frontotemporal dementia (FTD) and Alzheimer's disease (AD), clinically present with progressive cognitive decline and the deposition of neurofibrillary tangles (NFTs) in the brain. Neurovascular compromise is also prevalent in AD and FTD however the relationship between tau and the neurovascular unit is less understood relative to other degenerative phenotypes. Current animal models confer the ability to recapitulate aspects of the CNS tauopathies, however, existing models either display overaggressive phenotypes, or do not develop neuronal loss or genuine neurofibrillary lesions. In this report, we communicate the longitudinal characterization of brain tauopathy in a novel transgenic rat model, coded McGill-R955-hTau. The model expresses the longest isoform of human P301S tau. Homozygous R955-hTau rats displayed a robust, progressive accumulation of mutated human tau leading to the detection of tau hyperphosphorylation and cognitive deficits accelerating from 14 months of age. This model features extensive tau hyperphosphorylation with endogenous tau recruitment, authentic neurofibrillary lesions, and tau-associated neuronal loss, ventricular dilation, decreased brain volume, and gliosis in aged rats. Further, we demonstrate how neurovascular integrity becomes compromised at aged life stages using a combination of electron microscopy, injection of the tracer horseradish peroxidase and immunohistochemical approaches.
Collapse
Affiliation(s)
- Joshua T Emmerson
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - Sonia Do Carmo
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - Yingying Liu
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - Ali Shalhoub
- Department of Biochemistry, McGill University, Montreal H3A 0C7, Canada
| | - Ai Liu
- Integrated Program in Neuroscience, McGill University, Montreal H3A 1A1, Canada
| | - Quentin Bonomo
- Integrated Program in Neuroscience, McGill University, Montreal H3A 1A1, Canada
| | - Janice C Malcolm
- Department of Anatomy and Cell Biology, McGill University, Montreal H3A 0C7, Canada
| | - Lionel Breuillaud
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - A Claudio Cuello
- Department of Pharmacology & Therapeutics, McGill University, Montreal H3G1Y6, Canada; Integrated Program in Neuroscience, McGill University, Montreal H3A 1A1, Canada; Department of Pharmacology, Oxford University, Oxford OX13QT, UK.
| |
Collapse
|
38
|
Johnson AM, Lukens JR. The innate immune response in tauopathies. Eur J Immunol 2023; 53:e2250266. [PMID: 36932726 PMCID: PMC10247424 DOI: 10.1002/eji.202250266] [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: 01/06/2023] [Revised: 02/23/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023]
Abstract
Tauopathies, which include frontotemporal dementia, Alzheimer's disease, and chronic traumatic encephalopathy, are a class of neurological disorders resulting from pathogenic tau aggregates. These aggregates disrupt neuronal health and function leading to the cognitive and physical decline of tauopathy patients. Genome-wide association studies and clinical evidence have brought to light the large role of the immune system in inducing and driving tau-mediated pathology. More specifically, innate immune genes are found to harbor tauopathy risk alleles, and innate immune pathways are upregulated throughout the course of disease. Experimental evidence has expanded on these findings by describing pivotal roles for the innate immune system in the regulation of tau kinases and tau aggregates. In this review, we summarize the literature implicating innate immune pathways as drivers of tauopathy.
Collapse
Affiliation(s)
- Alexis M. Johnson
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA
- BIG Training Graduate Program, UVA, Charlottesville, VA 22908, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA
- BIG Training Graduate Program, UVA, Charlottesville, VA 22908, USA
| |
Collapse
|
39
|
Stevenson M, Varghese R, Hebron ML, Liu X, Ratliff N, Smith A, Turner RS, Moussa C. Inhibition of discoidin domain receptor (DDR)-1 with nilotinib alters CSF miRNAs and is associated with reduced inflammation and vascular fibrosis in Alzheimer's disease. J Neuroinflammation 2023; 20:116. [PMID: 37194065 PMCID: PMC10186647 DOI: 10.1186/s12974-023-02802-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023] Open
Abstract
Discoidin Domain Receptor (DDR)-1 is activated by collagen. Nilotinib is a tyrosine kinase inhibitor that is FDA-approved for leukemia and potently inhibits DDR-1. Individuals diagnosed with mild-moderate Alzheimer's disease (AD) treated with nilotinib (versus placebo) for 12 months showed reduction of amyloid plaque and cerebrospinal fluid (CSF) amyloid, and attenuation of hippocampal volume loss. However, the mechanisms are unclear. Here, we explored unbiased next generation whole genome miRNA sequencing from AD patients CSF and miRNAs were matched with their corresponding mRNAs using gene ontology. Changes in CSF miRNAs were confirmed via measurement of CSF DDR1 activity and plasma levels of AD biomarkers. Approximately 1050 miRNAs are detected in the CSF but only 17 miRNAs are specifically altered between baseline and 12-month treatment with nilotinib versus placebo. Treatment with nilotinib significantly reduces collagen and DDR1 gene expression (upregulated in AD brain), in association with inhibition of CSF DDR1. Pro-inflammatory cytokines, including interleukins and chemokines are reduced along with caspase-3 gene expression. Specific genes that indicate vascular fibrosis, e.g., collagen, Transforming Growth Factors (TGFs) and Tissue Inhibitors of Metalloproteases (TIMPs) are altered by DDR1 inhibition with nilotinib. Specific changes in vesicular transport, including the neurotransmitters dopamine and acetylcholine, and autophagy genes, including ATGs, indicate facilitation of autophagic flux and cellular trafficking. Inhibition of DDR1 with nilotinib may be a safe and effective adjunct treatment strategy involving an oral drug that enters the CNS and adequately engages its target. DDR1 inhibition with nilotinib exhibits multi-modal effects not only on amyloid and tau clearance but also on anti-inflammatory markers that may reduce cerebrovascular fibrosis.
Collapse
Affiliation(s)
- Max Stevenson
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Rency Varghese
- Genomics and Epigenomics Shared Resource, Department of Oncology, Georgetown University Medical Center, Building D, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Nick Ratliff
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Amelia Smith
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - R Scott Turner
- Memory Disorders Program, Department of Neurology, Georgetown University Medical Center, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
40
|
Hussong SA, Banh AQ, Van Skike CE, Dorigatti AO, Hernandez SF, Hart MJ, Ferran B, Makhlouf H, Gaczynska M, Osmulski PA, McAllen SA, Dineley KT, Ungvari Z, Perez VI, Kayed R, Galvan V. Soluble pathogenic tau enters brain vascular endothelial cells and drives cellular senescence and brain microvascular dysfunction in a mouse model of tauopathy. Nat Commun 2023; 14:2367. [PMID: 37185259 PMCID: PMC10126555 DOI: 10.1038/s41467-023-37840-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 03/31/2023] [Indexed: 05/17/2023] Open
Abstract
Vascular mechanisms of Alzheimer's disease (AD) may constitute a therapeutically addressable biological pathway underlying dementia. We previously demonstrated that soluble pathogenic forms of tau (tau oligomers) accumulate in brain microvasculature of AD and other tauopathies, including prominently in microvascular endothelial cells. Here we show that soluble pathogenic tau accumulates in brain microvascular endothelial cells of P301S(PS19) mice modeling tauopathy and drives AD-like brain microvascular deficits. Microvascular impairments in P301S(PS19) mice were partially negated by selective removal of pathogenic soluble tau aggregates from brain. We found that similar to trans-neuronal transmission of pathogenic forms of tau, soluble tau aggregates are internalized by brain microvascular endothelial cells in a heparin-sensitive manner and induce microtubule destabilization, block endothelial nitric oxide synthase (eNOS) activation, and potently induce endothelial cell senescence that was recapitulated in vivo in microvasculature of P301S(PS19) mice. Our studies suggest that soluble pathogenic tau aggregates mediate AD-like brain microvascular deficits in a mouse model of tauopathy, which may arise from endothelial cell senescence and eNOS dysfunction triggered by internalization of soluble tau aggregates.
Collapse
Affiliation(s)
- Stacy A Hussong
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Oklahoma City Veterans Health Care System, 921 NE 13th Street, Oklahoma City, OK, 73104, USA
| | - Andy Q Banh
- South Texas Medical Scientist Training Program, University of Texas Health San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Candice E Van Skike
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Angela O Dorigatti
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Stephen F Hernandez
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
| | - Matthew J Hart
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Therapeutic Science, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Beatriz Ferran
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Haneen Makhlouf
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
| | - Maria Gaczynska
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Pawel A Osmulski
- Barshop Institute for Longevity and Aging Studies, University of Texas Health San Antonio, 4939 Charles Katz Drive, San Antonio, TX, 78229, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX, 78229, USA
| | - Salome A McAllen
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Kelly T Dineley
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Zoltan Ungvari
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, 800 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, H-1085 Budapest, Üllői út 26, Budapest, Hungary
| | | | - Rakez Kayed
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Mitchell Center for Neurodegenerative Disease, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
- Sealy Center for Vaccine Development, University of Texas Medical Branch at Galveston, 301 University Blvd, Galveston, TX, 77555, USA
| | - Veronica Galvan
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA.
- Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, 940 Stanton L Young Blvd, Oklahoma City, OK, 73104, USA.
- Oklahoma City Veterans Health Care System, 921 NE 13th Street, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
41
|
Afsar A, Chacon Castro MDC, Soladogun AS, Zhang L. Recent Development in the Understanding of Molecular and Cellular Mechanisms Underlying the Etiopathogenesis of Alzheimer's Disease. Int J Mol Sci 2023; 24:7258. [PMID: 37108421 PMCID: PMC10138573 DOI: 10.3390/ijms24087258] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that leads to dementia and patient death. AD is characterized by intracellular neurofibrillary tangles, extracellular amyloid beta (Aβ) plaque deposition, and neurodegeneration. Diverse alterations have been associated with AD progression, including genetic mutations, neuroinflammation, blood-brain barrier (BBB) impairment, mitochondrial dysfunction, oxidative stress, and metal ion imbalance.Additionally, recent studies have shown an association between altered heme metabolism and AD. Unfortunately, decades of research and drug development have not produced any effective treatments for AD. Therefore, understanding the cellular and molecular mechanisms underlying AD pathology and identifying potential therapeutic targets are crucial for AD drug development. This review discusses the most common alterations associated with AD and promising therapeutic targets for AD drug discovery. Furthermore, it highlights the role of heme in AD development and summarizes mathematical models of AD, including a stochastic mathematical model of AD and mathematical models of the effect of Aβ on AD. We also summarize the potential treatment strategies that these models can offer in clinical trials.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA
| |
Collapse
|
42
|
Xu X, Xu H, Zhang Z. Cerebral amyloid angiopathy-related cardiac injury: Focus on cardiac cell death. Front Cell Dev Biol 2023; 11:1156970. [PMID: 36910141 PMCID: PMC9998697 DOI: 10.3389/fcell.2023.1156970] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/16/2023] [Indexed: 03/14/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a kind of disease in which amyloid β (Aβ) and other amyloid protein deposits in the cerebral cortex and the small blood vessels of the brain, causing cerebrovascular and brain parenchymal damage. CAA patients are often accompanied by cardiac injury, involving Aβ, tau and transthyroxine amyloid (ATTR). Aβ is the main injury factor of CAA, which can accelerate the formation of coronary artery atherosclerosis, aortic valve osteogenesis calcification and cardiomyocytes basophilic degeneration. In the early stage of CAA (pre-stroke), the accompanying locus coeruleus (LC) amyloidosis, vasculitis and circulating Aβ will induce first hit to the heart. When the CAA progresses to an advanced stage and causes a cerebral hemorrhage, the hemorrhage leads to autonomic nervous function disturbance, catecholamine surges, and systemic inflammation reaction, which can deal the second hit to the heart. Based on the brain-heart axis, CAA and its associated cardiac injury can create a vicious cycle that accelerates the progression of each other.
Collapse
Affiliation(s)
- Xiaofang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huikang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of the Diagnosis and Treatment for Severe Trauma and Burn of Zhejiang Province, Hangzhou, China.,Zhejiang Province Clinical Research Center for Emergency and Critical care medicine, Hangzhou, China
| |
Collapse
|
43
|
Green R, Mayilsamy K, McGill AR, Martinez TE, Chandran B, Blair LJ, Bickford PC, Mohapatra SS, Mohapatra S. SARS-CoV-2 infection increases the gene expression profile for Alzheimer's disease risk. Mol Ther Methods Clin Dev 2022; 27:217-229. [PMID: 36187720 PMCID: PMC9508696 DOI: 10.1016/j.omtm.2022.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/21/2022] [Indexed: 02/02/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has caused over 600,000,000 infections globally thus far. Up to 30% of individuals with mild to severe disease develop long COVID, exhibiting diverse neurologic symptoms including dementias. However, there is a paucity of knowledge of molecular brain markers and whether these can precipitate the onset of Alzheimer's disease (AD). Herein, we report the brain gene expression profiles of severe COVID-19 patients showing increased expression of innate immune response genes and genes implicated in AD pathogenesis. The use of a mouse-adapted strain of SARS-CoV-2 (MA10) in an aged mouse model shows evidence of viral neurotropism, prolonged viral infection, increased expression of tau aggregator FKBP51, interferon-inducible gene Ifi204, and complement genes C4 and C5AR1. Brain histopathology shows AD signatures including increased tau-phosphorylation, tau-oligomerization, and α-synuclein expression in aged MA10 infected mice. The results of gene expression profiling of SARS-CoV-2-infected and AD brains and studies in the MA10 aged mouse model taken together, for the first time provide evidence suggesting that SARS-CoV-2 infection alters expression of genes in the brain associated with the development of AD. Future studies of common molecular markers in SARS-CoV-2 infection and AD could be useful for developing novel therapies targeting AD.
Collapse
Affiliation(s)
- Ryan Green
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Karthick Mayilsamy
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Andrew R. McGill
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Taylor E. Martinez
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Bala Chandran
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Laura J. Blair
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- Byrd Alzheimer’s Research Institute, University of South Florida, Tampa, FL 33613, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Paula C. Bickford
- Center of Excellence for Aging and Brain Repair, Departments of Neurosurgery and Brain Repair, and Molecular Pharmacology and Physiology, Morsani College of Medicine, Tampa, FL 33613, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Shyam S. Mohapatra
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| |
Collapse
|
44
|
Abdul Y, Karakaya E, Chandran R, Jamil S, Ergul A. Endothelin A receptors contribute to senescence of brain microvascular endothelial cells. Can J Physiol Pharmacol 2022; 100:1087-1096. [PMID: 36384316 PMCID: PMC10052805 DOI: 10.1139/cjpp-2022-0071] [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: 11/18/2022]
Abstract
Cellular senescence plays a pivotal role in the aging and progression of neurodegenerative diseases, including vascular cognitive impairment and dementia (VCID). In postmortem brains from individuals with VCID, endothelin-1 (ET-1) levels closely correlate with blood barrier breakdown and cerebral hypoperfusion. Brain microvascular endothelial cells (BMVECs), previously thought to have exclusively endothelin B receptors, also possess endothelin A (ETA) receptors; however, the functional significance of this receptor in BMVECs is not known. We hypothesize that ETA receptors mediate BMVEC senescence. Serum-starved human BMVECs (HBEC5i) were incubated with ET-1 (1 µmol/L) in the presence/absence of ETA receptor antagonist BQ-123 (20 µmol/L). Cells were collected for Western blot and quantitative real-time PCR analyses. Treatment of ET-1 increased protein expression of ETA receptor, while it was prevented by the ETA receptor antagonist. ET-1 increased p21, p16, p53, LIF1 and cyclin D1 protein levels, and β-galactosidase accumulation, which were prevented in the presence of ETA blockade. While there was no change in tight junction proteins, ET-1 decreased adherent junction protein vascular endothelial cadherin (VE-cadherin) levels. In conclusion, ET-1 upregulates ETA receptors in BMVECs in an autocrine manner and triggers the activation of senescence. These in vitro findings need to be further studied in vivo to establish the role of ETA receptors in the progression of endothelial senescence in VCID.
Collapse
Affiliation(s)
- Yasir Abdul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Eda Karakaya
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Raghavendar Chandran
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Sarah Jamil
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| | - Adviye Ergul
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC
| |
Collapse
|
45
|
Weigand AJ, Hamlin AM, Breton J, Clark AL. Cerebral blood flow, tau imaging, and memory associations in cognitively unimpaired older adults. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2022; 3:100153. [PMID: 36353072 PMCID: PMC9637859 DOI: 10.1016/j.cccb.2022.100153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 10/11/2022] [Accepted: 10/15/2022] [Indexed: 06/16/2023]
Abstract
Objective Cerebral blood flow (CBF) has been independently linked to cognitive impairment and traditional Alzheimer's disease (AD) pathology (e.g., amyloid-beta [Aβ], tau) in older adults. However, less is known about the possible interactive effects of CBF, Aβ, and tau on memory performance. The present study examined whether CBF moderates the effect of Aβ and tau on objective and subjective memory within cognitively unimpaired (CU) older adults. Methods Participants included 54 predominately white CU older adults from the Alzheimer's Disease Neuroimaging Initiative. Multiple linear regression models examined meta-temporal CBF associations with (1) meta-temporal tau PET adjusting for cortical Aβ PET and (2) and cortical Aβ PET adjusting for tau PET. The CBF and tau meta region was an average of 5 distinct temporal lobe regions. CBF interactions with Aβ or tau PET on memory performance were also examined. Covariates for all models included age, sex, education, pulse pressure, APOE-ε4 positivity, and imaging acquisition date differences. Results CBF was significantly negatively associated with tau PET (t = -2.16, p = .04) but not Aβ PET (t = 0.98, p = .33). Results revealed a CBF by tau PET interaction such that there was a stronger effect of tau PET on objective (t = 2.51, p = .02) and subjective (t = -2.67, p = .01) memory outcomes among individuals with lower levels of CBF. Conclusions Cerebrovascular and tau pathologies may interact to influence cognitive performance. This study highlights the need for future vascular risk interventions, which could offer a scalable and cost-effective method for AD prevention.
Collapse
Affiliation(s)
- Alexandra J. Weigand
- San Diego State University/University of California San Diego Joint Doctoral Program in Clinical Psychology, United States
| | - Abbey M. Hamlin
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, 108 East Dean Keeton, SEA 3.234, Austin, TX 78712, United States
| | - Jordana Breton
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, 108 East Dean Keeton, SEA 3.234, Austin, TX 78712, United States
| | - Alexandra L. Clark
- Department of Psychology, College of Liberal Arts, University of Texas at Austin, 108 East Dean Keeton, SEA 3.234, Austin, TX 78712, United States
| |
Collapse
|
46
|
Pintér P, Alpár A. The Role of Extracellular Matrix in Human Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms231911085. [PMID: 36232390 PMCID: PMC9569603 DOI: 10.3390/ijms231911085] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The dense neuropil of the central nervous system leaves only limited space for extracellular substances free. The advent of immunohistochemistry, soon followed by advanced diagnostic tools, enabled us to explore the biochemical heterogeneity and compartmentalization of the brain extracellular matrix in exploratory and clinical research alike. The composition of the extracellular matrix is critical to shape neuronal function; changes in its assembly trigger or reflect brain/spinal cord malfunction. In this study, we focus on extracellular matrix changes in neurodegenerative disorders. We summarize its phenotypic appearance and biochemical characteristics, as well as the major enzymes which regulate and remodel matrix establishment in disease. The specifically built basement membrane of the central nervous system, perineuronal nets and perisynaptic axonal coats can protect neurons from toxic agents, and biochemical analysis revealed how the individual glycosaminoglycan and proteoglycan components interact with these molecules. Depending on the site, type and progress of the disease, select matrix components can either proactively trigger the formation of disease-specific harmful products, or reactively accumulate, likely to reduce tissue breakdown and neuronal loss. We review the diagnostic use and the increasing importance of medical screening of extracellular matrix components, especially enzymes, which informs us about disease status and, better yet, allows us to forecast illness.
Collapse
Affiliation(s)
- Panka Pintér
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
| | - Alán Alpár
- Department of Anatomy, Semmelweis University, 1113 Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Hungarian Academy of Sciences, 1051 Budapest, Hungary
- Correspondence:
| |
Collapse
|
47
|
Schreiner TG, Creangă-Murariu I, Tamba BI, Lucanu N, Popescu BO. In Vitro Modeling of the Blood–Brain Barrier for the Study of Physiological Conditions and Alzheimer’s Disease. Biomolecules 2022; 12:biom12081136. [PMID: 36009030 PMCID: PMC9405874 DOI: 10.3390/biom12081136] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/09/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022] Open
Abstract
The blood–brain barrier (BBB) is an essential structure for the maintenance of brain homeostasis. Alterations to the BBB are linked with a myriad of pathological conditions and play a significant role in the onset and evolution of neurodegenerative diseases, including Alzheimer’s disease. Thus, a deeper understanding of the BBB’s structure and function is mandatory for a better knowledge of neurodegenerative disorders and the development of effective therapies. Because studying the BBB in vivo imposes overwhelming difficulties, the in vitro approach remains the main possible way of research. With many in vitro BBB models having been developed over the last years, the main aim of this review is to systematically present the most relevant designs used in neurological research. In the first part of the article, the physiological and structural–functional parameters of the human BBB are detailed. Subsequently, available BBB models are presented in a comparative approach, highlighting their advantages and limitations. Finally, the new perspectives related to the study of Alzheimer’s disease with the help of novel devices that mimic the in vivo human BBB milieu gives the paper significant originality.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Neurology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
- Correspondence:
| | - Ioana Creangă-Murariu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str., No. 16, 700155 Iasi, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, Universitatii Str., No. 16, 700155 Iasi, Romania
| | - Nicolae Lucanu
- Department of Applied Electronics and Intelligent Systems, Faculty of Electronics, Telecommunications and Information Technology, Gheorghe Asachi Technical University of Iasi, 21-23 Professor Dimitrie Mangeron Blvd., 700050 Iasi, Romania
| | - Bogdan Ovidiu Popescu
- Faculty of Medicine, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Neurology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Laboratory of Cell Biology, Neurosciences and Experimental Myology, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
48
|
Salwierz P, Davenport C, Sumra V, Iulita MF, Ferretti MT, Tartaglia MC. Sex and gender differences in dementia. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 164:179-233. [PMID: 36038204 DOI: 10.1016/bs.irn.2022.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The dementia landscape has undergone a striking paradigm shift. The advances in understanding of neurodegeneration and proteinopathies has changed our approach to patients with cognitive impairment. Firstly, it has recently been shown that the various proteinopathies that are the cause of the dementia begin to build up long before the appearance of any obvious symptoms. This has cemented the idea that there is an urgency in diagnosis as it occurs very late in the pathophysiology of these diseases. Secondly, that accurate diagnosis is required to deliver targeted therapies, that is precision medicine. With this latter point, the realization that various factors of a person need to be considered as they may impact the presentation and progression of disease has risen to the forefront. Two of these factors aside from race and age are biological sex and gender (social construct), as both can have tremendous impact on manifestation of disease. This chapter will cover what is known and remains to be known on the interaction of sex and gender with some of the major causes of dementia.
Collapse
Affiliation(s)
- Patrick Salwierz
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Carly Davenport
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - Vishaal Sumra
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada
| | - M Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain; Women's Brain Project, Guntershausen, Switzerland
| | | | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, Canada; Memory Clinic, Krembil Brain Institute, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
49
|
Zhu N, Wei M, Yuan L, He X, Chen C, Ji A, Zhang G. Claudin-5 relieves cognitive decline in Alzheimer's disease mice through suppression of inhibitory GABAergic neurotransmission. Aging (Albany NY) 2022; 14:3554-3568. [PMID: 35471411 PMCID: PMC9085235 DOI: 10.18632/aging.204029] [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: 12/07/2021] [Accepted: 03/08/2022] [Indexed: 11/25/2022]
Abstract
Alzheimer’s disease (AD) is characterized by progressive cognitive decline, which is considered as the most common form of dementia in the elderly. Recently, it is suggested that impaired cerebrovascular function may precede the onset of AD. Claudin-5, which is the most enriched tight junction protein, has been reported to prevent the passage of damaging material at the blood-brain barrier. However, whether claudin-5 impacts AD has no direct evidence. We found a decrease level of claudin-5 in the hippocampus of AD and elder mice. And intravenous injection of claudin-5 improved learning and memory ability in these mice, while knockout of the protein led to impaired learning and memory and long-term potentiation in adult control mice. Furthermore, the effects of claudin-5 are mediated by suppressing inhibitory GABAergic neurotransmission. Our results suggest benefit effects of claudin-5 on learning and memory, which may provide a new treatment strategy for AD.
Collapse
Affiliation(s)
- Ning Zhu
- General Practice Center, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, China.,Department of Pharmacy, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, China
| | - Meidan Wei
- Department of Pharmacy, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Linguang Yuan
- College of Basic Medicine, Changsha Medical University, Changsha 410219, China
| | - Xiaodan He
- General Practice Center, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, China
| | - Chunli Chen
- Department of Pharmacy, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, China
| | - Aimin Ji
- Department of Pharmacy, The Seventh Affiliated Hospital, Southern Medical University, Foshan 528244, China
| | - Guozeng Zhang
- Institute of Nursing and Health, School of Nursing and Health, Henan University, Kaifeng 475004, China
| |
Collapse
|
50
|
Kindler D, Maschio C, Ni R, Zerbi V, Razansky D, Klohs J. Arterial spin labeling demonstrates preserved regional cerebral blood flow in the P301L mouse model of tauopathy. J Cereb Blood Flow Metab 2022; 42:686-693. [PMID: 34822744 PMCID: PMC8943618 DOI: 10.1177/0271678x211062274] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
There is growing evidence for the vascular contribution to cognitive impairment and dementia in Alzheimer's disease (AD) and other neurodegenerative diseases. While perfusion deficits have been observed in patients with Alzheimer's disease and tauopaties, little is known about the role of tau in vascular dysfunction. In the present study, regional cerebral blood (rCBF) was characterized in P301L mice with arterial spin labeling. No differences in rCBF in P301L mice compared to their age-matched non-transgenic littermates at mid (10-12 months of age) and advanced (19-21 months of age) disease stages. This was concomitant with preservation of cortical brain structure as assessed with structural T2-weighted magnetic resonance imaging. These results show that hypoperfusion and neurodegeneration are not a phenotype of P301L mice. More studies are thus needed to understand the relationship of tau, neurodegeneration and vascular dysfunction and its modulators in AD and primary tauopathies.
Collapse
Affiliation(s)
- Diana Kindler
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland
| | - Cinzia Maschio
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| | - Ruiqing Ni
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| | - Valerio Zerbi
- Zurich Neuroscience Center (ZNZ), Zurich, Switzerland.,Neural Control of Movement Lab, Department of Health Sciences and Technology, ETH Zurich, 27219ETH Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, 27219ETH Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), Zurich, Switzerland
| |
Collapse
|